Abstract
The progressive aging of the global population has prompted considerable interest in the development of safe and effective strategies to promote healthy longevity. Diet constitutes a substantial modifiable factor, with fermented foods emerging as a pivotal domain of research. The health benefits of these foods are largely attributed to viable microorganisms and a rich array of bioactive compounds, such as organic acids, phenolics, and peptides, which are generated during fermentation. This review methodically synthesizes recent advances regarding the anti-aging potential of fermented foods, emphasizing the pivotal molecular and cellular mechanisms involved. Specifically, we elucidate major pathways, including the scavenging of reactive oxygen species, the mitigation of oxidative stress, and the regulation of critical signal transduction networks. Furthermore, this review analyzes the multifaceted benefits of fermented foods, which encompass gut microbiota modulation, immune system regulation, anti-inflammatory responses, and the enhancement of host antioxidant defense systems. By integrating current evidence, this work establishes a theoretical framework to guide the development of innovative fermented food products and strategies targeting healthy aging.
Keywords: Fermented foods, Anti-aging, Active substances, Mechanisms
Graphical abstract
Highlights
-
•
Fermented foods exhibit anti-aging potential through both shared mechanisms (e.g., redox regulation and inflammation control) and product-specific effects shaped by microbial consortia and food matrices.
-
•
Fermentation-derived bioactive compounds, rather than nutrients alone, play a central role in modulating key aging-related pathways, including Nrf2, AMPK, sirtuin signaling, and NF-κB.
-
•
Gut microbiota remodeling emerges as a critical integrative node linking oxidative stress, immune regulation, and metabolic homeostasis during aging.
-
•
Evidence from cellular and animal models highlights mechanistic diversity across fermented dairy, soy, grains, vegetables, and teas, underscoring context-dependent rather than universal effects.
-
•
Translational application of fermented foods for healthy aging requires improved mechanistic resolution, standardized fermentation strategies, and rigorous clinical validation.
1. Introduction
The accelerated aging of the global population has led to a notable increase in the prevalence of health issues among the elderly (Octary et al., 2025). Projections indicate that by 2050, the proportion of the global population aged 60 and above will reach 22 %. The impact of aging and its associated diseases on individuals' health and quality of life has increased exponentially worldwide (Gao et al., 2023). This demographic shift is projected to result in a substantial escalation in healthcare expenditures (Ros and Carrascosa, 2020). Therefore, identifying safe and effective interventions to mitigate the effects of aging and extend the healthy lifespan of humans will have a profound impact on health, the economy, and society (Qin et al., 2026).
Aging is a natural phenomenon involving irreversible degenerative changes at the molecular, cellular, tissue, and organ levels (Fatt et al., 2022). These modifications enhance susceptibility to disease and represent a significant risk factor for a range of chronic illnesses, including cancer, metabolic disorders, cardiovascular disease, and neurodegenerative conditions (Barone et al., 2022; Conrad et al., 2023; Song and Zhang, 2023). In order to develop effective interventions, research has focused on the underlying mechanisms, recently summarized as twelve scientifically validated “Hallmarks of Aging” (Fig. 1). The hallmarks of aging include genomic instability, telomere attrition, epigenetic alterations, and cellular senescence. These hallmarks, when taken together, lead to integrative features such as chronic inflammation and ecological dysregulation(López-Otín et al., 2013).
Fig. 1.
The hallmarks of aging and diseases of aging(López-Otín et al., 2023).
Among the hallmarks of aging, chronic low-grade inflammation, known as inflammaging, and dysbiosis (predominantly manifesting as gut microbiota dysbiosis) have been identified as the primary drivers of unhealthy aging (Yin et al., 2025). Crucially, these two characteristics are highly plastic and can be effectively regulated through dietary and microbial interventions. Although aging is inevitable, research into decelerating the process has persisted (Jothi and Kulka, 2024). Current strategies often focus on pharmacological or dietary interventions (Partridge et al., 2020). However, the potential for negative side effects from anti-aging drugs (Juricic et al., 2022; Soukas et al., 2019) has intensified the search for safer alternatives. Researchers have increasingly focused on identifying natural anti-aging substances within functional foods (Teng et al., 2025), as diet is a fundamental regulator of human metabolism.
Within this field, traditional fermented foods (e.g., yogurt, kefir, kimchi, fermented soy products) are regarded as unique functional foods, precisely because their health benefits are fundamentally driven by microbiology (Iwatani and Yamamoto, 2019). Their potential anti-aging properties are mediated through both viable probiotic microorganisms and fermentation-derived bioactive factors (Orisakwe et al., 2020; Singh et al., 2021). These microbial factors are uniquely positioned to target the gut dysbiosis and inflammaging hallmarks identified as critical in the aging process. While several studies have reported the anti-aging effects of fermented foods (Das et al., 2020), systematic reviews focusing on the anti-aging mechanisms of fermented foods are still scarce. Although the microbial composition of fermented foods has been extensively cataloged, there is currently no comprehensive review that systematically links specific starter cultures (e.g., Lactobacillus plantarum) and their metabolic signatures directly to the molecular regulatory mechanisms of host aging.
This review synthesizes the current evidence on the anti-aging properties of fermented foods through a mechanistic lens. The present study focuses on the synergistic duality of viable microbes and fermentation products, distinguishing this from a generalized perspective. A multi-scale analysis of preclinical and clinical data has been conducted to elucidate key longevity networks, including redox regulation, immunomodulation, metabolic signaling, and gut-host interactions. The review also assesses challenges in safety and translation. The resulting framework is designed to support rational product design and precise nutritional strategies for the aging population.
2. Anti-aging effects of different fermented foods
According to the International Scientific Association for Probiotics and Prebiotics (ISAPP), fermented foods are defined as “foods made through desired microbial growth and enzymatic conversions of food components” (Marco et al., 2021). Fermented foods have been shown to contain functional molecules that have been demonstrated to be beneficial to the human body. These foods achieve the purpose of preventing disease and promoting health by regulating the physiological functions of the human body (Cui et al., 2024; Gonzalez-Gonzalez et al., 2023; Sørensen et al., 2023). There is a wide variety of fermented foods, and common fermented foods include yoghurt, kimchi, soy sauce, tempeh, bread, and a variety of alcoholic products (Zhang et al., 2023). Since Metchnikoff's suggestion (Mackowiak, 2013), that high consumption of lactobacillus-fermented milk may prolong the life expectancy of the general population, in-depth studies have been conducted on the anti-aging effects of fermented foods, and a large number of functional fermented foods with promising have been found (Das et al., 2020). Depending on the raw materials (Fig. 2), fermented foods can be divided into the following categories: fermented soybean products (e.g., bean paste, soy sauce), fermented dairy products (e.g., yogurt, cheese), fermented cereals (e.g., vinegar, yellow wine), and other fermented foods (e.g., kimchi, black tea).
Fig. 2.
Classification of fermented foods.
2.1. Fermented dairy products
Fermented dairy products, such as yogurt, cheese, kefir, and koumiss, are produced through the metabolic activity of starter cultures (e.g., Lactobacillus, Bifidobacterium, and yeasts) on milk substrates(Bintsis, 2024). Beyond extending shelf life and enhancing flavor, fermentation enzymatically breaks down lactose, proteins, and fats into bioactive metabolites, including free amino acids, short-chain fatty acids (SCFAs), and bioactive peptides, thereby enhancing nutritional bioavailability (Rizzoli and Biver, 2024).
As outlined in Table 1, a comprehensive review of both human clinical trials and preclinical studies has demonstrated the potential of fermented dairy to mitigate age-related physiological decline. In human clinical trials, probiotic-fermented dairy has demonstrated significant efficacy in combating immunosenescence. For instance, Dong et al. (2013) reported that the daily consumption of a probiotic drink containing Lactobacillus casei Shirota (1.3 × 1010 CFU) over a period of four weeks significantly enhanced Natural Killer (NK) cell activity and promoted an anti-inflammatory cytokine profile (increased IL-10/IL-12 ratio) in healthy older adults (55–74 years). In addition, observational studies have indicated that the long-term consumption of cheese is associated with enhanced cognitive processing speed in the elderly. Furthermore, interventions incorporating Lactobacillus reuteri fortified dairy have been observed to mitigate bone loss in older women (Nilsson et al., 2018). In animal models, the anti-aging mechanisms of fermented dairy have been frequently linked to the regulation of oxidative stress and the gut-brain axis. Zhang et al. (2017) demonstrated that exopolysaccharides (EPS) isolated from Tibetan kefir significantly upregulated antioxidant enzymes (SOD, GSH-Px) and reduced plasma malondialdehyde (MDA) levels in aging mice. Notably, this antioxidant effect was strongly correlated with the modulation of gut microbiota, specifically the enrichment of beneficial Blautia and Butyricicoccus genera. Furthermore, fermentation-derived peptides play a critical role in neuroprotection. Ano et al. (2018) identified novel tryptophan-tyrosine (GTWY) peptides in fermented whey that improved memory function in aged mice. Mechanistically, these peptides inhibited monoamine oxidase B (MAO-B) activity, thereby preserving hippocampal dopamine levels. Additionally, recent evidence highlights the potential of cheese extracts in longevity. Cardin et al. (2021) found that lipid extracts from raw goat milk cheese extended the lifespan of Caenorhabditis elegans by activating the DAF-16/FOXO signaling pathway, a conserved regulator of longevity.
Table 1.
Anti-aging effects of fermented dairy products.
| Fermented Product | Microorganism & Fermentation Conditions | Key Bioactive Compound | Experimental Model (n/Sex/Age) | Intervention (Dose & Duration) | Key Outcomes & Mechanisms | Refer |
|---|---|---|---|---|---|---|
| Probiotic drink | Lactobacillus casei Shirota | Live probiotic | Healthy older adults (n = 30; 18Female/12Male; 55–74 y) | 130 mL/day (1.3 × 1010 CFU) for 4 weeks | Outcomes: ↑ Innate immunity (NK cell activity); ↓ T-cell activation (CD25 MFI); Shift to anti-inflammatory profile (↑ IL-10/IL-12 ratio). Mechanism: Attenuation of immunosenescence. |
Dong et al. (2013) |
| Naturally fermented Xinjiang yogurt | Lactobacillus plantarum KSFY02 | Live probiotic | Kunming mice (n = 50; 25Female/25Male; 6 weeks) | 1.0 × 109 CFU/kg BW/day for 10 weeks | Outcomes: Preserved organ indices; ↑ SOD; GSH-Px; GSH; ↓ NO; MDA. Mechanism: Activation of Nrf2–HO-1 antioxidant pathway (↑ Nrf2, SOD, CAT, HO-1, ↓ Nos2). |
X. Zhao et al. (2019) |
| Tibetan kefir | Lactobacillus plantarum YW11 (37 °C; 18 h) | Exopolysaccharide (EPS) | ICR mice (n = 48; Male; 8 weeks) | EPS: 20–50 mg/kg/d or Fermented milk: 20 mL/kg/d for 12 weeks | Outcomes: ↑ SOD; GSH-Px; CAT; T-AOC; ↓ MDA; ↑ SCFA production. Mechanism: Microbiota modulation (↓ Flexispira, ↑ Blautia, ↑ Butyricicoccus) linked to ROS scavenging. |
Zhang et al. (2017) |
| Tibet kefir grains | Lactobacillus plantarum MA2 (CGMCC3005; 37 °C; 18 h) | Live probiotic | Male Kunming mice (n = 60; 8 weeks) | 108–1010 CFU/day (oral gavage) for 6 weeks | Outcomes: ↑ Antioxidant capacity (SOD, GSH-Px); ↓ MDA. Mechanism: Sustained stable intestinal colonization (ileum, colon, feces) supporting antioxidant effects. |
Tang et al. (2016) |
| Fermented whey protein | Enzymatic fermentation (Aspergillus melleus or Bacillus stearothermophilus) | Trp-Tyr (GTWY) containing peptides | C57BL/6 mice (n = 24; Male; 22 months) | 1–10 mg/kg for 10 days | Outcomes: Improved working and episodic memory in amnesia/aged mice. Mechanism: Inhibition of MAO-B activity leading to ↑ hippocampal dopamine levels. |
Ano et al. (2018) |
| Dadih (Buffalo milk) | Lactic acid bacteria (Natural fermentation in bamboo) | Bioactive peptides; Live probiotics | Rattus norvegicus (n = 30; Male; 24 months) | 4.5 g/day (once or twice daily) for 42 days | Outcomes: ↓ Renal oxidative damage (MDA); Attenuated renal interstitial fibrosis. Mechanism: Peptide- and probiotic-driven ROS scavenging and lipid peroxidation inhibition. |
Harnavi et al. (2020) |
| Kumiss (Mare milk) | Natural mixed microbiota (LAB and yeasts) | Unsaturated fatty acids; Milk proteins | BALB/c mice (n = 40; Male; 20–25 g) | 20 mL/kg (i.p.; weekly) for 20 weeks | Outcomes: ↓ Oxidative stress (8-OHdG; OSI; TOC); ↑ TAC; GSH. Mechanism: Upregulated SIRT2 and SIRT3 expression (hepatic/colonic) indicating sirtuin-mediated metabolic regulation. |
Gulmez and Atakisi (2020) |
| Camembert cheese | Surface fermentation (Penicillium candidum) | Oleamide | 5xFAD transgenic mice (n = 22; Female; 3 months) | 2 % (w/w) fermented cheese in diet for 3 months | Outcomes: ↓ Cerebral Aβ burden; ↓ Hippocampal inflammation (TNF-α; MIP-1α); ↑ Neurotrophic support (BDNF; GDNF). Mechanism: Oleamide-mediated microglial activation toward anti-inflammatory phenotype (↑ CD36, ↓ CD68). |
Ano et al. (2015) |
| Selenium-enriched yogurt | Limosilactobacillus fermentum CGMCC 17434 (42 °C; 6 h + post-ripening) | Organic selenium; Resveratrol | ICR mice (n = 70; Male; 18–22 g) | 0.1–0.4 mL/day for 8 weeks | Outcomes: ↑ GSH, SOD; ↓ ROS, MDA, PCO; Attenuated systemic/neuroinflammation (↓ IL-1β, IL-6, TNF-α). Mechanism: Combined antioxidant-anti-inflammatory protection of spleen/hippocampus. |
Luo et al. (2025) |
| Raw goat milk cheese | Natural fermentation (Traditional cheese-making) | Lipophilic and hydrophilic cheese extracts | C. elegans (N2 wild-type & mutants) | 0.5–1 % (w/v) extracts | Outcomes: Extended lifespan; Enhanced oxidative stress resistance; ↓ ROS levels. Mechanism: DAF-16 nuclear translocation required (DAF-2/DAF-16 signaling) and p38 MAPK pathway activation. |
Cardin et al. (2021) |
| Eight French raw-milk cheeses | Traditional diverse processing (Goat; cow; ewe) | Bioactive peptides; Phenolic compounds; S-amino acids | C. elegans N2 | 1 % (w/v) cheese fractions for 5 days | Outcomes: Enhanced survival and ↓ ROS accumulation under oxidative stress. Mechanism: Activation of IIS/DAF-16 and p38 MAPK/SKN-1 pathways (↑ ctl-2, sod-3). |
Diet et al. (2024) |
| Buffalo & Cow Milk Cheddar | Traditional ripening (8 °C; up to 6 months) | Water-soluble peptides (WSPs) | Caco-2 human intestinal epithelial cells | 100–300 μg/mL WSP extracts (24 h) | Outcomes: ↑ ABTS scavenging activity; ↓ Intracellular ROS in t-BHP-induced stress. Note: Higher activity observed in buffalo milk cheese (linked to WSP content). |
Huma et al. (2018) |
| Himalayan cheese (Kalari) | L. plantarum; L. casei; L. brevis (37 °C; 24 h fermentation) | Bioactive peptides | MCF-7, HCT-116, IMR-32 cells; Mouse monocytes | 10–50 mg/mL water-soluble extract | Outcomes: Dose-dependent anti-proliferative activity in cancer lines. Mechanism: Immunomodulatory effects (↑ NO production in monocytes) via proteolysis-derived peptides. |
Mushtaq et al. (2019) |
Abbreviations: 8-OHdG, 8-hydroxy-2′-deoxyguanosine; Aβ, amyloid beta; ABTS, 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid); BDNF, brain-derived neurotrophic factor; BW, body weight; CAT, catalase; CD, cluster of differentiation; CFU, colony-forming units; DAF, dauer formation; GDNF, glial cell line-derived neurotrophic factor; GSH, glutathione; GSH-Px, glutathione peroxidase; HO-1, heme oxygenase-1; i.p., intraperitoneal; IIS, insulin/IGF-1 signaling; IL, interleukin; MAPK, mitogen-activated protein kinase; MFI, mean fluorescence intensity; MIP-1α, macrophage inflammatory protein-1 alpha; Nrf2, nuclear factor erythroid 2-related factor 2; NO, nitric oxide; Nos2, nitric oxide synthase 2; OSI, oxidative stress index; PCO, protein carbonyls; ROS, reactive oxygen species; SIRT, sirtuin; SOD, superoxide dismutase; T-AOC/TAC, total antioxidant capacity; t-BHP, tert-butyl hydroperoxide; TNF-α, tumor necrosis factor alpha; TOC, total oxidant capacity; WSPs, water-soluble peptides.
2.2. Fermented soy products
Fermented soy products are foods or condiments with special flavors and shapes made from beans as the main raw material, fermented by micro-organisms and then blended (Park et al., 2019). A broad array of fermented soy products, including soy sauce, tempeh, soy paste, and fermented bean curd (see Fig. 3), have garnered significant consumer appeal worldwide, thereby becoming integral components of global diets. During fermentation, soy products undergo biochemical transformations that break down macromolecules into soluble small molecules and convert indigestible substances into absorbable nutrients (Cao et al., 2019). This process reduces anti-nutritional factors like protease inhibitors, oligosaccharides, and lectins (Miao et al., 2024), while generating beneficial compounds. These include active peptides, unsaturated fatty acids, isoflavone glycosides, γ-aminobutyric acid and various vitamins (Qiao et al., 2022).
Fig. 3.
Process flow chart of the main bean products(Liu et al., 2022).
As outlined in Table 2, emerging evidence from clinical trials and animal models underscores the multifaceted anti-aging properties of fermented soy products. In human clinical trials, fermented soy interventions have demonstrated efficacy in regulating metabolic health and oxidative stress. For instance, Lee et al. (2006) reported that acute consumption of traditional Dark Soy Sauce significantly decreased plasma lipid peroxidation biomarkers (F2-isoprostanes) in healthy adults, attributed to its potent postprandial radical scavenging activity. Furthermore, Jeong et al. (2021) conducted a 12-week intervention with Chungkookjang (a Korean fermented soybean paste) in overweight adults, observing significant reductions in visceral fat and apolipoprotein B, suggesting a potential role in mitigating age-related metabolic dysregulation. In animal models, fermented soy products have been shown to exert anti-aging effects across multiple aging-related domains, including neuroprotection, metabolic regulation, sarcopenia prevention, and lifespan extension. In terms of neuroprotection, Chan et al. (2018) demonstrated that tempeh supplementation attenuated cognitive decline and reduced brain amyloid-β accumulation in senescence-accelerated mice, an effect mechanistically associated with activation of the Nrf2 antioxidant signaling pathway. Beyond cognitive outcomes, fermented soy products have also shown efficacy in mitigating age-related metabolic deterioration. Koh et al. (2023) found that fermented black soybean produced by Bacillus subtilis var. natto prevented sarcopenia and reduced visceral fat accumulation in aged mice, effects that were closely linked to modulation of gut microbiota composition and associated metabolic pathways. At the organismal level, lifespan extension has also been observed, as Ibe et al. (2013) demonstrated that a water-soluble extract of natto significantly prolonged the lifespan of Caenorhabditis elegans and delayed the accumulation of lipofuscin, a hallmark of cellular aging, through activation of stress-response pathways.
Table 2.
Anti-aging effects of fermented soy products.
| Fermented Product | Microorganism & Fermentation Conditions | Key Bioactive Compound | Experimental Model (n/Sex/Age) | Intervention (Dose & Duration) | Key Outcomes & Mechanisms | Reference |
|---|---|---|---|---|---|---|
| Dark soy sauce (DSS) | Traditional fermentation | Antioxidant compounds (Total phenolics) | Healthy adults (n = 24; 12Female/12Male; ∼23 y) | 30 mL DSS (single dose w/meal) | Outcomes: ↓ Lipid peroxidation (Plasma F2-isoprostanes); ↓ Diastolic BP (transient). Mechanism: Rapid postprandial radical scavenging in plasma. |
Lee et al. (2006) |
| Chungkookjang | Mixed Bacillus spp. (Salt-free; 24–72 h) | γ-PGA; Isoflavones; Peptides | Overweight/obese adults (n = 52) | Human: 26 g/d (12 weeks) | Outcomes: ↓ Visceral fat & ApoB. Mechanism: Attenuation of oxidative stress and neuroinflammation. |
Jeong et al. (2021) |
| Novel fermented soybean food | Bacillus spp.; L. delbrueckii subsp. bulgaricus; Hansenula anomala (37 °C; 21 h) | Peptides; Phenolics; Flavonoids | C57BL/6J mice (n = 50; Male; 5 months) | 30–750 mg/kg/d (oral) for 3 weeks | Outcomes: ↑ Hepatic glycogen; ↓ BUN, Lactic acid; ↑ SOD, GSH-Px; ↓ MDA. Mechanism: Activation of Nrf2/ARE pathway (↑ Nq1, Gclc, Gclm). |
Cui et al. (2020) |
| Fermented black soybean & adlay | Bacillus subtilis var. natto (28 °C; 43 h) | Nattokinase; γ-PGA; Phenolics | Aged C57BL/6J mice (n = 56; Male/Female; 48 weeks) | 2 % or 6 % diet for 14 weeks | Outcomes: ↓ Muscle loss (Sarcopenia); ↓ Visceral fat; ↓ Pro-inflammatory cytokines. Mechanism: Microbiota modulation & reduction of hepatic oxidative stress markers. |
Koh et al. (2023) |
| Tempeh | Rhizopus oligosporus (Solid-state; 37 °C; 5–7 d) | Aglycone isoflavones; Polyphenols | SAMP8 (Aging model) & SAMR1 mice (n = 58; Male; 6 months) | 300–900 mg/kg BW/d for 12 weeks | Outcomes: ↑ Cognitive performance; ↓ Brain Aβ accumulation; ↓ Protein carbonyls. Mechanism: Nrf2 signaling activation and modulation of MAPK pathways. |
Chan et al. (2018) |
| Fermented soy beverage | Bifidobacterium pseudocatenulatum INIA P815 (Anaerobic; 37 °C; 24 h) | Aglycone isoflavones (Genistein, Daidzein) | C57BL/6J mice (n = 96; Female; 10 months; Menopause model) | 4 mL/day for 36 days | Outcomes: ↑ Isoflavone bioavailability; ↑ Reproductive health (cyclic mice); ↓ Serum triglycerides. Mechanism: Bio-conversion to aglycones rather than microbiota remodeling. |
Ruiz de la Bastida et al. (2023) |
| Miso soup | Traditional fermentation (High salt) | Isoflavones; Melanoidins; Vitamins | Dahl salt-sensitive rats (n = 32; Male; 4 weeks) | 10 % Miso soup (1.3 % NaCl) for 8 weeks | Outcomes: ↓ Salt-induced hypertension; ↓ Renal/Cardiac structural damage. Mechanism: Protection against salt-induced cardiovascular stress despite Na intake. |
Yoshinaga et al. (2012) |
| Hawaijar | Mixed Bacillus spp. (Natural fermentation; 3–7 d) | Fibrinolytic enzymes; ACE-inhibitory peptides | Sprague–Dawley rats (Male; 6–8 weeks) | 200 mg/kg BW/d for 4 weeks | Outcomes: ↑ Fibrinolytic activity; ↓ ACE activity; Improved lipid oxidation. Mechanism: Bioactive peptide-mediated cardiometabolic regulation. |
Anand Singh et al. (2023) |
| Doenjang | Traditional aging (Long-term) | Aglycone isoflavones; Peptides | C57BL/6J mice (n = 47; Male; 4 weeks) | 14.4 % freeze-dried Doenjang in HFD (11 weeks) | Outcomes: ↓ Neuronal loss; ↑ Neurogenesis; ↓ Aβ & Tau hyperphosphorylation. Mechanism: Inhibition of neuroinflammation and oxidative stress pathways. |
Ko et al. (2019) |
| Co-fermented red bean extract | Bacillus subtilis & Lactobacillus bulgaricus (37 °C; 120 h) | Catechin; Rutin; Ferulic acid | C57BL/6 mice (n = 30; Male; 8 weeks) & Drosophila | Drosophila: 3.3–10 mg/mL (diet); Mice: 0.5 g/kg (15 d) | Outcomes: ↑ Motor function (Mice); ↑ Lifespan (Drosophila); ↑ GPx. Mechanism: Neuroprotection via antioxidant capacity and radical scavenging. |
Chou et al. (2025) |
| Natto | Bacillus subtilis natto T9303 (39 °C; 20h + 4 °C; 24h) | Low-MW fraction (<14 kDa) | Caenorhabditis elegans N2 (Adult) | 0.5–1 mg/mL water extract (22.5–25.5 d) | Outcomes: ↑ Lifespan; ↑ Thermotolerance; ↓ Lipofuscin accumulation. Mechanism: Activation of stress-response pathways (independent of spermidine). |
Ibe et al. (2013) |
| Fermented mung bean | Rhizopus microsporus 5351 (Solid-state; 30 °C; 48 h) | GABA; Free amino acids; Phenolics | MCF-7 cells; Murine splenocytes (Ex vivo) | 2–3 mg/mL (24–72 h) | Outcomes: ↑ Splenocyte proliferation (Immunomodulation); ↑ Cytokines (IL-2, IFN-γ). Mechanism: Cell-cycle arrest and apoptosis in cancer cells; Immune stimulation. |
Ali et al. (2016) |
| Soymilk kefir | Kefir grains (Microbial consortium; 20 °C; 32 h) | Soy peptides; Isoflavones | Salmonella typhimurium TA98 (Mutagenicity assay) | 10 mg/mL freeze-dried powder | Outcomes: ↑ Antimutagenic activity; ↑ Antioxidant capacity. Mechanism: Peptide-mediated radical scavenging (superior to unfermented milk). |
Liu et al. (2005) |
Abbreviations: ACE, angiotensin-converting enzyme; ApoB, apolipoprotein B; ARE, antioxidant response element; BP, blood pressure; BUN, blood urea nitrogen; d, day(s); DSS, dark soy sauce; GABA, gamma-aminobutyric acid; Gclc, glutamate-cysteine ligase catalytic subunit; Gclm, glutamate-cysteine ligase modifier subunit; h, hour(s); HFD, high-fat diet; IFN-γ, interferon gamma; kDa, kilodalton; MW, molecular weight; Nq1, NAD(P)H:quinone oxidoreductase 1; SAMP8, senescence-accelerated mouse prone 8; SAMR1, senescence-accelerated mouse resistant 1; y, years; γ-PGA, poly-gamma-glutamic acid.
2.3. Fermented cereal products
Cereal grains are considered staple foods on a global scale (Abdel-Aal, 2023), yet their nutritional potential is frequently constrained by the presence of anti-nutritional factors such as phytates and tannins (Samtiya et al., 2020). Fermentation, a process of biochemical breakdown, has been shown to yield a number of benefits for human health. These benefits include the production of beneficial compounds, a reduction in anti-nutrient content, and an enhancement of bioavailability, thereby improving nutritional value (Melini et al., 2019; T. Zhao et al., 2019, Zhao et al., 2019). Fermented cereal foods constitute a significant component of global food cultures, exhibiting a long historical presence and a diverse array of varieties (Petrova and Petrov, 2020). The primary fermented cereal foods encompass sweet pasta sauces, rice vinegar, rice wine, bread, and similar products. Fermented cereal products have emerged as a viable solution to address the mounting demand for healthy, nutritious, and low-fat or plant-based dietary patterns. These products are garnering increasing recognition as a pivotal category within the expanding functional food landscape (Tsafrakidou et al., 2020).
As outlined in Table 3, contemporary research, predominantly grounded in animal models and mechanistic studies, underscores the distinct anti-aging consequences of fermented cereal products in neuroprotection, metabolic regulation, and cellular longevity. In animal models, fermented cereal products have shown pronounced efficacy in alleviating neurodegenerative and metabolic aging phenotypes. Supplementation with Yellow Rice Wine significantly improved memory performance and fatigue resistance in D-galactose–induced aging mice (Liu et al., 2020), an effect mechanistically associated with the suppression of hippocampal neuronal apoptosis via regulation of the Bax/Bcl-2 ratio and Caspase-3 signaling. Similarly, Red Mold Rice fermented by Monascus purpureus reduced cerebral amyloid-β accumulation in hyperlipidemic rats by shifting amyloid precursor protein processing toward the non-amyloidogenic pathway (Lee et al., 2010). Beyond neuroprotection, fermented cereal products also exert beneficial metabolic and gut-related effects. Fermented wheat germ co-fermented with Lactobacillus plantarum and Saccharomyces cerevisiae enhanced learning and memory while increasing hepatic antioxidant enzyme activities (SOD and GSH-Px) in aging mice, with these effects closely linked to gut microbiota remodeling, suggesting involvement of the gut–brain axis (Y. Zhao et al., 2021). Concurrently, black vinegar extracts have been shown to ameliorate hyperlipidemia and systemic inflammation in hamsters fed a high-fat diet. This effect is attributed to a reduction in pro-inflammatory cytokines (TNF-α and IL-1β) and an enhancement of hepatoprotective antioxidant defenses (Chou et al., 2015). In mechanistic and lifespan models, specific fermentation metabolites have been identified as key drivers of longevity. Barathikannan et al. (2023) observed that fermented brown rice extracts extended the lifespan of Caenorhabditis elegans and reduced lipid accumulation by downregulating fatty acid desaturase genes. Furthermore, Luti et al. (2020) identified low-molecular-weight peptides (<3 kDa) in sourdough bread that exerted potent anti-inflammatory effects in macrophages by inhibiting the NF-κB signaling pathway.
Table 3.
Anti-aging effects of fermented cereal products.
| Fermented Product | Microorganism & Fermentation Conditions | Key Bioactive Compound(s) | Experimental Model (n/Sex/Age) | Intervention (Dose & Duration) | Key Outcomes & Mechanisms | Reference |
|---|---|---|---|---|---|---|
| Fermented Wheat Germ | Lactobacillus plantarum + Baker's yeast; (28–37 °C; 18 h) | Ferulic acid; BCAAs; Peptides | BALB/c mice (n = 18; 6 weeks) | 20 mg/kg/day for 6 weeks | Outcomes: ↑ Learning/memory; ↑ SOD, GSH-Px; ↓ MDA; ↑ Hepatic glycogen. Mechanism: Modulation of gut microbiota composition linked to metabolic regulation. |
Y. Zhao et al. (2021) |
| Chinese Rice Wine | Mixed yeast; fungi; and LAB (Traditional starter) | Amino acids; GABA; Polysaccharides | Male Kunming mice (n = 40; Male; 12 weeks) | 0.15–0.40 mL/day for 15–56 days | Outcomes: ↑ Hepatic glycogen; ↓ Blood lactic acid/BUN; ↑ SOD, GSH-Px, CAT; ↓ MDA. Mechanism: Enhanced antioxidant defense and energy metabolism. |
Zhao et al. (2018) |
| Yellow Rice Wine | Traditional Qu (Semi-solid fermentation) | Functional oligosaccharides; Phenolics | Kunming mice (n = 120; Male; 12 weeks) | 4–12 mL/kg BW/day for 6 weeks | Outcomes: ↓ Fatigue resistance; ↑ Memory performance; ↑ Antioxidant enzymes. Mechanism: Suppression of hippocampal neuronal apoptosis via Bax/Bcl-2 and Caspase-3 signaling. |
Liu et al. (2020) |
| Red Mold Rice | Monascus purpureus NTU 568 (Solid-state) | Monacolin K; Secondary metabolites | Wistar rats (n = 49; 280–320g) | 151–755 mg/kg/day for 28 days | Outcomes: ↑ Memory; ↓ Brain cholesterol; ↓ Aβ burden. Mechanism: Shift of APP processing toward non-amyloidogenic sAPPα pathway via inhibition of ApoE/β-secretase. |
Lee et al. (2010) |
| Black Vinegar | Traditional surface fermentation | Amino acids; Minerals; Polyphenols | Golden Syrian hamsters (High-fat diet; n = 32; Male; 5 weeks) | 0.13–0.26 g/kg BW/day for 9 weeks | Outcomes: ↓ Serum/Hepatic lipids; ↓ Inflammatory cytokines (TNF-α, IL-1β); ↑ Fecal lipid excretion. Mechanism: Combined hepatoprotective and antioxidant regulation. |
Chou et al. (2015) |
| Rice Wine (Cheong-ju) | Koji and Yeast saccharification | Glycerol derivatives; Amino acids | Hairless mice (n = 15; Female; 6 weeks) | Topical application (2 %, daily) for 2 months | Outcomes: ↓ UV-induced oxidative stress; ↑ Skin barrier integrity; ↓ MMP-1 expression. Mechanism: Enhanced extracellular matrix synthesis (↑ Type I procollagen). |
Seo et al. (2009) |
| Tapuy (Philippine Rice Wine) | Indigenous Bubod starter | Phenolic compounds | Caenorhabditis elegans (Wild-type N2; L4 stage) | 5000–50,000 μg/mL in growth medium | Outcomes: Modest lifespan extension (+6.1 % at high dose). Mechanism: Dissociation between antioxidant capacity and longevity effects (non-antioxidant pathway). |
Chua et al. (2024) |
| Fermented Brown Rice | Pediococcus acidilactici MNL5 (Solid-state; 37 °C; 24h) | Ferulic acid | C. elegans (N2 & daf-2 mutants) | 1 mg/mL extract | Outcomes: ↑ Lifespan (Wild-type & daf-2); ↓ Lipid accumulation. Mechanism: Downregulation of fatty acid desaturase genes (fat-4, fat-5, fat-6). |
Barathikannan et al. (2023) |
| Tempeh-like Chenopodium | Rhizopus microsporus var. Oligosporus(35 °C; 4 d) | Glycine-rich peptide (GRP) | C. elegans (N2) & Human fibroblasts | 50–100 μg/mL (GRP) | Outcomes: ↑ Lifespan; ↑ Stress resistance; ↓ Cellular senescence. Mechanism: Activation of Nrf2-dependent pathway and upregulation of skn-1 (ortholog of Nrf2). |
Hsieh et al. (2025) |
| Sourdough Bread | Selected Lactobacillus spp. (30 °C; 18h) | Low-MW peptides (<3 kDa) | RAW 264.7 murine macrophages | 0.01 mg/mL peptides (1 h pretreatment) | Outcomes: ↓ Intracellular ROS; ↓ iNOS and COX-2 expression. Mechanism: Inhibition of NF-κB signaling via stabilization of IκB. |
Luti et al. (2020) |
| Fermented Rice Bran | Issatchenkia orientalis (35 °C; 5 d) | Ferulic acid derivatives | 3T3-L1 adipocytes | 10–50 μg/mL extract | Outcomes: ↓ Oxidative stress-induced ROS; ↑ PPAR-γ, Adiponectin. Mechanism: Regulation of adipogenesis and inflammation-related gene expression. |
Kim and Han (2011) |
| Commercial Beers | Saccharomyces cerevisiae + Saccharomyces carlsbergensis (3–20 °C; 4–12 weeks) | Catechin; Caffeic acid | Isolated thoracic aorta rings from male Wistar rats (10–12 weeks) | Cumulative concentrations | Outcomes: ↑ Endothelium-dependent vasorelaxation; ↑ Antioxidant capacity. Mechanism: Nitric oxide (NO) signaling pathway modulation. |
Oliveira Neto et al. (2017) |
| Fermented Buckwheat Flour | L. rhamnosus; S. thermophilus; R. oligosporus | Phenolic compounds | In vitro simulated digestion model | 10 g freeze-dried product | Outcomes: ↑ Bioaccessibility of phenolics; ↑ Anti-AGEs activity. Mechanism: Digestion-dependent release of functional phenolics. |
Zieliński et al. (2020) |
| Boza | Mixed yeast and LAB (Traditional) | ACE-inhibitory peptides (<10 kDa) | In vitro simulated digestion model | N/A | Outcomes: Strong ACE-inhibitory activity. Mechanism: Production of bioactive low-MW peptides during fermentation. |
Kancabaş and Karakaya (2013) |
| Shanxi Aged Vinegar | Traditional solid-state fermentation | Melanoidins; Polyphenols | In vitro antioxidant assays | N/A | Outcomes: High antioxidant activity (ABTS, FRAP). Mechanism: Correlation between aging time and accumulation of melanoidins/phenolics. |
Xia et al. (2018) |
Abbreviations: AGEs, advanced glycation end-products; ApoE, apolipoprotein E; APP, amyloid precursor protein; Bax, Bcl-2-associated X protein; BCAAs, branched-chain amino acids; Bcl-2, B-cell lymphoma 2; COX-2, cyclooxygenase-2; daf-2, abnormal dauer formation-2 (insulin/IGF-1 receptor homolog); FRAP, ferric reducing antioxidant power; GRP, glycine-rich peptide; IκB, inhibitor of nuclear factor kappa B; iNOS, inducible nitric oxide synthase; MMP-1, matrix metalloproteinase-1; NF-κB, nuclear factor kappa B; PPAR-γ, peroxisome proliferator-activated receptor gamma; sAPPα, soluble amyloid precursor protein alpha; UV, ultraviolet.
2.4. Other fermented products
Fermented foods have been consumed globally for millennia, exhibiting remarkable diversity in terms of raw materials, microbial communities, and processing strategies (Rul et al., 2022). In addition to the extensive research conducted on dairy, soy, and cereal-based products (Lindner and Bernini, 2022), a mounting body of evidence suggests that fermented vegetables, fruits, teas, medicinal herbs, and animal-derived products may also offer anti-aging benefits through conserved molecular mechanisms (Das et al., 2020; Zhang et al., 2022). The primary anti-aging value of these products is attributable to the microbial biotransformation of substrate-specific phytonutrients. Fermentation is a process that has been shown to effectively convert conjugated phenolic compounds into bioactive aglycones and synthesize novel metabolites. These metabolites include theabrownin and microbial exopolysaccharides. The result of this process is an enhancement of the bioavailability and functional potency of the compounds compared to their unfermented counterparts (Rajendran et al., 2023; Yuan et al., 2023, 2024).
As outlined in Table 4, a wide array of these fermented products has exhibited substantial anti-aging potential, encompassing effects on skin health and metabolic regulation. In human clinical trials, fermentation-derived metabolites have demonstrated potential in mitigating skin aging. Chan et al. (2021) conducted a randomized trial involving a fermented vegetable-fruit drink (fermented by Saccharomyces cerevisiae and Streptococcus thermophilus). After 8 weeks of supplementation, healthy adults exhibited significantly increased systemic antioxidant capacity (SOD, CAT) and improved skin elasticity, accompanied by a reduction in wrinkles and spots. In the context of animal models, Lei et al. (2022) demonstrated that Pu-erh tea, which is abundant in theabrownin polyphenol, exhibited a positive effect on cognitive functions, specifically learning and memory, in mice subjected to D-galactose-induced aging. This neuroprotective effect was achieved by modulating the gut–liver–brain axis, specifically through the enrichment of Akkermansia muciniphila. Similarly, Cardoso et al. (2021) found that Kombucha (fermented black/green tea) alleviated hepatic steatosis and insulin resistance in rats by regulating lipid metabolism genes (e.g., SREBP-1c, CPT-1). Logozzi et al. (2020) provided compelling evidence that a Fermented Papaya Preparation (FPP) modulated redox balance in aging mice. It is important to note that this intervention led to the induction of a “pro-longevity” molecular signature, as evidenced by an increase in telomere length and telomerase activity in bone marrow and ovarian cells. Wang et al. (2022) reported that fermented Lycium barbarum (Wolfberry) polysaccharides significantly extended the lifespan of Caenorhabditis elegans and enhanced stress resistance via the activation of the DAF-16/FOXO pathway, independent of insulin signaling. In vitro investigations have elucidated the cellular mechanisms that are unique to fermented vegetables and animal products. Kim et al. (Kim et al., 2011) investigated the properties of kimchi, and specifically its extracts from “optimally ripened” kimchi (pH 4.2). They found that these extracts extended the replicative lifespan of human fibroblasts and suppressed stress-induced premature senescence by inhibiting NF-κB signaling. In addition, Park et al. (2018) demonstrated that fermented fish oil provided superior photoprotection in comparison with unfermented DHA/EPA. This superiority was evidenced by the effective inhibition of UVB-induced apoptosis in keratinocytes through MAPK pathway modulation.
Table 4.
Anti-aging effects of other fermented products.
| Fermented Product | Microorganism & Fermentation Conditions | Key Bioactive Compound(s) | Experimental Model (n/Sex/Age) | Intervention (Dose & Duration) | Key Outcomes & Mechanisms | Reference |
|---|---|---|---|---|---|---|
| Fermented Veg-Fruit Drink | S. cerevisiae + S. thermophilus TCI125 (35 °C; 10 d) | Polyphenols; SOD-like enzymes | Healthy humans (n = 30; 13Male/17Female; 35–55 y) | 50 mL/day for 8 weeks | Outcomes: ↑ Systemic antioxidant status (SOD, CAT); ↑ Skin elasticity/moisture; ↓ Wrinkles/Spots. Mechanism: Enhanced collagen synthesis and systemic redox defense. |
Chan et al. (2021) |
| Malus micromalus Makino fruit wine | Yeast-mediated fruit fermentation | Polysaccharides (MWP-2); Phenolics | Kunming mice (n = 60; half male, half female; 6 weeks) | 50–150 mg/kg BW/day for 4 weeks | Outcomes: ↓ Brain lipid peroxidation; ↓ Neuronal apoptosis; ↑ SOD, GPx, CAT. Mechanism: Polysaccharide-mediated redox regulation and neuroprotection. |
Hui et al. (2019) |
| Pu-erh Tea | Microbial solid-state fermentation | Theabrownin (TB) | C57BL/6 mice (n = 40; Male; 5–6 weeks) | 1.2 g/kg BW (TB) for 8 weeks | Outcomes: ↑ Learning/memory; ↓ Systemic inflammation (IL-6, TNF-α); ↑ Intestinal barrier. Mechanism: Gut-liver-brain axis remodeling (↑ Akkermansia, ↓ E. coli). |
Lei et al. (2022) |
| Fermented Papaya | Yeast fermentation (10 months) | Antioxidants (Ascorbic acid) | C57BL/6J mice (n = 40; Female; 4 weeks) | 6 mg/mouse/day (water) long-term | Outcomes: ↑ Telomere length (Bone marrow/Ovaries); ↑ Telomerase activity; ↓ ROS. Mechanism: Time-dependent redox balance and telomere maintenance. |
Logozzi et al. (2020) |
| Kombucha (Green/Black Tea) | Symbiotic Culture of Bacteria and Yeast (25 °C; 10 d) | Catechins; Phenolic acids | Wistar rats (n = 32; Male; 45–50 d) | 30 % (v/v) drink for 10 weeks | Outcomes: ↓ Insulin resistance; ↓ Hepatic steatosis; ↑ SOD, CAT; ↓ NLR (Inflammation). Mechanism: Coordinated regulation of lipid metabolism genes (↓ SREBP1c, ↑ CPT-1). |
Cardoso et al. (2021) |
| Haihong Fruit Wine | Commercial fruit wine (∼10 % v/v alcohol) | Polyphenols; Flavonoids; Triterpenoids; Phytosterols | Kunming mice (n = 60; 30Male/30Female; 20–25 g) | 0.075–0.30 mL/10 gBW/day for 42 days | Outcomes: ↑ Systemic antioxidant defense (SOD, CAT, GPx); ↓ Lipid peroxidation (MDA) in liver and brain. Mechanism: Polyphenol-mediated ROS scavenging and membrane protection. |
Hui et al. (2021) |
| Black Tea Extract | Commercial CTC-grade | Theaflavins | Wistar rats (n = 48; Male; Young to Old) | 1 mL/100g BW/day for 30 days | Outcomes: ↑ Plasma FRAP; ↑ Erythrocyte GSH; ↓ MDA/AOPPs. Mechanism: Theaflavin-mediated mitigation of age-related oxidative stress. |
Kumar and Rizvi (2017) |
| Fermented Green Tea | Aspergillus niger (30 °C; 4 d) | Catechins; Proanthocyanidins; L-theanine | Caenorhabditis elegans (N2 wild-type) | 10 % extract in medium | Outcomes: ↑ Lifespan; ↑ Locomotion; ↑ Mitochondrial function. Mechanism: Activation of DAF-16, HSF-1, and p53 signaling pathways. |
Liu et al. (2025) |
| Probiotic Fermented Ginseng | lactic acid bacteria and yeast | Ginsenoside metabolites | Caenorhabditis elegans (Wild-type N2; L4 stage) | 0–160 μg/mL | Outcomes: ↑ Lifespan; ↑ Thermal/Oxidative tolerance; ↓ Apoptosis. Mechanism: IIS pathway modulation (↓ daf-2, ↑ skn-1, ↑ sod-3). |
Xu et al. (2023) |
| Dragon Fruit-Kiwi Beverage | S. cerevisiae Y1 + L. mesenteroides + S. thermophilus (30–37 °C; 9–15 d) | GABA; Phenolics; Riboflavin | Caenorhabditis elegans (N2 wild-type) | 1.56 % (v/v) in medium | Outcomes: ↑ Lifespan (∼18 %); ↑ SOD; ↓ MDA. Mechanism: GABA/Phenolic-mediated redox regulation. |
Tang et al. (2022) |
| Fermented Lycium barbarum (Wolfberry) | Rice wine yeast (28 °C; 36 h) | Lycium barbarum polysaccharide (LBP-Y) | Caenorhabditis elegans N2 (Synchronized; ∼50/group) | 0.1–1.0 mg/mL (Lifespan); 1.0 mg/mL (Stress assays) | Outcomes: ↑ Lifespan under normal/oxidative/heat stress; ↑ Radical scavenging & T-AOC. Mechanism: Activation of stress-response genes (↑ daf-16, sod-3, hsp-16.2) independent of daf-2 signaling. |
Wang et al. (2022) |
| Fermented Purple Sweet Potato | Weissella confusa NJLY1 (37 °C; 24 h) | Anthocyanins; Phenolic acids | Caenorhabditis elegans (N2; >30 worms) | 80 μg/mL anthocyanins | Outcomes: ↑ Lifespan vs. non-fermented; ↓ Lipofuscin. Mechanism: Activation of DAF-16/FOXO, SKN-1/Nrf2, and SIR-2.1 pathways. |
(J. Zhao et al., 2021) |
| Kimchi (Optimally Ripened) | Traditional lactic acid fermentation at 5 °C to pH 4.2 | Organic acids; Phytochemical metabolites | WI-38 human fibroblasts (Aging models) | 100 μg/mL extract (24 h pre-treat) | Outcomes: ↑ Replicative lifespan; ↓ Lipid peroxidation; ↑ Cell viability. Mechanism: Suppression of NF-κB activation (↓ nuclear NF-κB, iNOS, COX-2). |
Kim et al. (2011) |
| Solar Salts Kimchi | Traditional lactic acid bacteria fermentation | Phenolics; Flavonoids | RAW 264.7 macrophages | 2.5 mg/mL (24 h) | Outcomes: ↑ Antioxidant capacity (FRAP); ↓ Inflammatory cytokines (IL-1β, IL-6, TNF-α). Mechanism: Stronger anti-inflammatory effects in optimally ripened variants. |
Yun et al. (2023) |
| Fermented Lespedeza cuneata | Lactobacillus pentosus (37 °C; 5 d) | Aglycone flavonoids (Kaempferol, Quercetin) | Hs68 human fibroblasts | 100–400 μg/mL (48 h) | Outcomes: ↑ Elastase inhibition; ↑ Type I procollagen; ↓ UV-induced MMP-1. Mechanism: β-glucosidase-mediated conversion to bioactive aglycones. | Seong et al. (2017) |
| Jeju Citrus Vinegar | S. cerevisiae + A. pasteurianus (25–35 °C; 22 d) | Flavonoids (Hesperidin) | 3T3-L1 adipocytes; WI-38 fibroblasts | 1/100 dilution (24 h–8 days) | Outcomes: ↓ Lipid accumulation; ↓ Senescence-associated genes (p21, p53). Mechanism: Downregulation of adipogenic regulators (PPARγ, C/EBPα). |
Yun et al. (2021) |
| Fermented Meat (Slavonian Kulen) | Lactiplantibacillus plantarum 1K(37 °C; 48 h) | Metabolites <2 kDa | Human PBMCs (Male donor) | Metabolites (10 ng/mL) (4 h) | Outcomes: ↓ TNF-α secretion (∼70 %); High DPPH scavenging. Mechanism: Immunomodulation by small microbial metabolites without cytotoxicity. |
Kostelac et al. (2022) |
| Fermented Fish Oil | L. plantarum + S. cerevisiae (30 °C; 15 d → 18–20 °C; 50 d) | EPA; DHA; Fatty acids | HaCaT keratinocytes; HR-1 hairless mice (n = 20; Male, 7 weeks) | In vitro: 10–50 μg/mL, 24 h; In vivo: topical 2 or 20 mg/mL, 2 weeks | Outcomes: ↓ UVB-induced apoptosis (↑ Bcl-2/Bax); ↓ MAPK signaling. Mechanism: Synergistic photoprotection greater than DHA alone. |
Park et al. (2018) |
Abbreviations: AOPPs, advanced oxidation protein products; C/EBPα, CCAAT/enhancer-binding protein alpha; CPT-1, carnitine palmitoyltransferase-1; CTC, crush, tear, curl (tea processing grade); DHA, docosahexaenoic acid; DPPH, 2,2-diphenyl-1-picrylhydrazyl; EPA, eicosapentaenoic acid; FOXO, forkhead box O; HSF-1, heat shock factor 1; hsp, heat shock protein; NLR, neutrophil-to-lymphocyte ratio; PBMCs, peripheral blood mononuclear cells; SREBP1c, sterol regulatory element-binding protein 1c; TB, theabrownin; UVB, ultraviolet B.
3. Mechanism of the anti-aging effects of fermented foods
The pursuit of healthy aging has led to a heightened interest in natural, safe, and effective dietary interventions. Fermented foods, defined as food matrices that have undergone a process of transformation by the enzymatic activity of microorganisms that are beneficial to health, have emerged as a potent functional food category, a notion that is supported by a growing body of evidence (Dimidi et al., 2019; Ibrahim et al., 2023) The anti-aging efficacy of these foods stems from a fundamental bioconversion process involving the degradation of anti-nutritional factors (e.g., flatulence-causing oligosaccharides) (Mollakhalili-Meybodi et al., 2022) and the simultaneous biosynthesis of health-promoting metabolites, such as lactic acid, bioactive peptides, and enhanced phenolic compounds (Gopikrishna et al., 2021). Beyond improving nutritional bioavailability, fermented foods exhibit pleiotropic biological activities, including antioxidant, antimicrobial, and immunomodulatory effects(Hasegawa and Bolling, 2023). Moreover, they play a pivotal role in systemic metabolic regulation, including the reduction of cholesterol and the modulation of the gut microbiota to prevent chronic age-related diseases (Thumu and Halami, 2020). The aforementioned benefits are attributable to the synergistic action of viable probiotics and their functional metabolites(Mahamud et al., 2025). As demonstrated in Fig. 4, the underlying molecular mechanisms are intricate and interconnected, primarily involving the mitigation of oxidative stress, the regulation of apoptotic and autophagic pathways, the enhancement of immune function, and the restoration of gut dysbiosis.
Fig. 4.
Anti-aging mechanisms of fermented foods.
3.1. Mitigation of oxidative stress
Oxidative stress, defined as an imbalance between the production of reactive oxygen species (ROS) and the ability of cells to maintain their structural and functional integrity, is a major contributing factor to the aging process and the development of age-related metabolic abnormalities (Luo et al., 2021). While endogenous antioxidants have been shown to counteract this damage, their capacity declines with age. Fermented foods have therefore received significant attention for their potential to mitigate oxidative stress through bioactive compounds derived from fermentation. The fermentation process has been shown to enrich foods with hydrolyzed antioxidant peptides, free amino acids, and free polyphenols. These bioactive compounds function through two distinct mechanisms: (1) Direct Scavenging: These enzymes have been observed to facilitate the chelation of metal ions, engage in the direct scavenging of free radicals through proton or hydrogen donation, and inactivate singlet oxygen (Chen et al., 2023). (2) Enzymatic Regulation: These substances have been shown to modulate the host's endogenous defense systems by stimulating the secretion of key antioxidant enzymes, including SOD, CAT, HO-1, and GSH-Px (Zheng et al., 2023). The mechanisms in question are illustrated by specific fermented products. For instance, in vitro and animal studies suggest that kimchi retards the progression of aging by diminishing the production of free radicals and augmenting antioxidant enzyme activity (Kim et al., 2011). Furthermore, the fermentation of grains (e.g., buckwheat, barley) has been demonstrated to markedly augment phenolic and flavonoid content, consequently enhancing DPPH radical scavenging capacity and FRAP activity in comparison to unfermented controls (Bhanja Dey and Kuhad, 2014).
3.2. Regulation of apoptosis
Apoptosis, a fundamental physiological process, is dysregulated in cases of accelerated tissue aging. Fermented foods have been demonstrated to exert a protective effect against age-related cellular loss by modulating apoptotic signaling pathways. Bioactive components that are enriched during the fermentation process have been shown to safeguard the intracellular environment by restoring the balance between pro-apoptotic and anti-apoptotic factors (Calabrò et al., 2024). In animal models, fermentation-derived metabolites have been demonstrated to directly target the Bcl-2 and Caspase families. Specifically, interventions with fermented products have been shown to downregulate the expression of the pro-apoptotic protein Bax and the executioner protease Caspase-3, while simultaneously upregulating the anti-apoptotic protein Bcl-2. This shift has been demonstrated to impede the cleavage of cellular substrates and to inhibit the initiation of programmed cell death (Q. Y. Gong et al., 2022; Xu et al., 2019). Furthermore, fermentation bioactives have been demonstrated to play a critical role in the SIRT1-p53 axis. In contrast to the age-related decline in sirtuin activity, fermentation-derived polyphenols and peptides have been shown to activate SIRT1, which subsequently deacetylates and inhibits the oncogenic protein p53, thereby decreasing apoptotic susceptibility and extending cellular lifespan (Chen et al., 2017; Ou and Schumacher, 2018). The validation of this mechanism in vivo has been demonstrated by studies that have shown that probiotics, active peptides, and resveratrol, which are enriched in fermented foods, significantly downregulated the mRNA expression of p53, NF-κB, and Caspase-3 in the brain tissue of aged mice. Concurrently, these interventions have been shown to enhance the expression of neuroprotective factors, such as Nrf2 and Bcl-2, thereby effectively mitigating neuronal apoptosis and preserving cognitive function (Y. Wang et al., 2020; Xia et al., 2020).
3.3. Activation of autophagy
Autophagy is a critical cellular renewal process where cells degrade damaged organelles and misfolded proteins to maintain homeostasis. The decline of autophagic capacity is a hallmark of aging, leading to the accumulation of cellular debris, mitochondrial dysfunction, and chronic inflammation (Deretic, 2021). Fermented foods have been shown to intervene in this process primarily by modulating nutrient-sensing signaling pathways. In both animal and cellular models, the anti-aging effects of fermented foods have been demonstrated to be closely linked to the activation of AMPK (adenosine monophosphate-activated protein kinase) (Hofer et al., 2022). AMPK functions as a central energy sensor that, upon activation, triggers autophagy to restore energy homeostasis. A body of research has indicated that specific probiotics and bioactive metabolites isolated from fermented foods can significantly increase AMPK phosphorylation levels (Hor et al., 2019; Jang et al., 2019). This activation promotes autophagic flux, thereby reducing oxidative stress and enhancing metabolic resilience during aging (Wang et al., 2019). Conversely, the mTOR (mammalian target of rapamycin) pathway, which functions as a negative regulator of autophagy, is often hyperactive in aging tissues. Mechanistic studies suggest that fermentation-derived polyphenols can effectively inhibit mTOR signaling (Liu and Sabatini, 2020). The suppression of mTOR and the concomitant activation of AMPK by these bioactive compounds induce a pro-autophagic state, thereby facilitating cellular rejuvenation (R. Gong et al., 2022). Moreover, this regulatory framework frequently intersects with the insulin/IGF-1 signaling (IIS) pathway(Kim and Lee, 2019) and the Sirtuin pathway (Jiang et al., 2025). Collectively, improving autophagic capacity through fermented food interventions represents a vital strategy for maintaining cellular metabolic balance and ameliorating pathological aging.
3.4. Enhanced immunity
Immunosenescence, defined as the gradual decline of both adaptive and innate immunity, plays a substantial role in the development of age-related diseases. Fermented foods have been demonstrated to offer a nutritional strategy to counteract this decline by modulating immune cell activity and cytokine profiles (Salminen et al., 2019; Sanyal and Haldar, 2025). In animal models, probiotics isolated from fermented foods (e.g., Lactobacillus plantarum, Lactobacillus helveticus, and Lactobacillus pentosus) have demonstrated potent immunomodulatory properties. The efficacy of these strains in enhancing the phagocytic activity of macrophages and restoring the homeostatic balance of T lymphocytes has been demonstrated, as evidenced by the increase in the numbers of naïve CD4+ and CD8+ T cells (J. Wang et al., 2020; You et al., 2020). Mechanistically, these interventions promote the expression of immunostimulatory cytokines (e.g., NO, IL-2, IL-6) while simultaneously suppressing pro-inflammatory mediators like TNF-α, thereby enhancing resistance to pathogens (Agrawal et al., 2017; Wastyk et al., 2021; You et al., 2020). Beyond viable bacteria, bioactive metabolites generated during fermentation play a crucial role. As stated by Badr et al. (2012), an increase in the proliferative capacity of lymphocytes and monocytes in response to antigenic stimuli was observed following the administration of fermented whey protein concentrates. In addition, fermentation-derived polyphenols and polysaccharides have been shown to facilitate the polarization of macrophages from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, and to promote the differentiation of immune-tolerant dendritic cells. These cellular shifts effectively mitigate chronic low-grade inflammation and decelerate organismal aging (Sun et al., 2022; Wang et al., 2024).
3.5. Relieve inflammation
“Inflammaging,” a term denoting the chronic, sterile, low-grade inflammation associated with the aging process, has been identified as a central driver of organismal degeneration (Picca and Marzetti, 2023). Fermented foods have been demonstrated to intervene in this process by reducing pro-inflammatory cytokines and reinforcing mucosal barriers to prevent systemic immune activation. In human clinical trials, evidence has emerged to support the systemic anti-inflammatory potential of fermented interventions. A meta-analysis by Gui et al. (Gui et al., 2020; Varsha et al., 2022) confirmed that the ingestion of fermented foods significantly enhances NK cell activity, suggesting that this dietary intervention may bolster immune surveillance, thereby preventing chronic inflammatory accumulation. Moreover, the maintenance of epithelial barrier integrity is paramount. As Polo et al. (2023) noted, the incorporation of fermented matrices aids in preserving tight junctions, thereby mitigating the systemic influx of endotoxins (“leaky gut”) that often precipitates inflammation in aged populations. In cellular and animal models, specific fermentation-derived metabolites have been demonstrated to modulate intracellular signaling pathways. As demonstrated by Gabriele et al. (2023), extracts from fermented dough exerted anti-inflammatory effects by activating the Nrf2/HO-1 signaling pathway, effectively coupling antioxidant defense with the suppression of inflammatory mediators. In a similar manner, the consumption of fermented soy products has been demonstrated to impede the generation of pro-inflammatory cytokines, such as MIF and IL-1β, and to diminish the infiltration of inflammatory cells. This effect is achieved through the repression of oxidative stress markers (Das et al., 2022). The bioactive compounds that are enriched during fermentation, including lovastatin and theabrownine in fermented tea, also play a mechanistic role. Deng et al. (2021) reported that these molecules significantly reduce TNF-α expression and promote the apoptosis of pro-inflammatory macrophages, thereby disrupting the vicious cycle of inflammation and senescence.
3.6. Regulate gut microbiota
Aging is frequently accompanied by gut dysbiosis, characterized by a decrease in microbial diversity, a reduction in beneficial genera, and an enrichment of pro-inflammatory pathobionts. This imbalance disrupts intestinal barrier function and accelerates systemic aging (Chao et al., 2025). Fermented foods intervene in this process through a dual mechanism involving the colonization of viable probiotics and the supply of postbiotic nutrients (Funk et al., 2020; Liu et al., 2021; Schluter et al., 2020). In animal models, specific probiotic strains isolated from fermented foods have demonstrated the capacity to reverse age-related dysbiosis. As demonstrated by Bu et al. (2023), the oral administration of Lactobacillus plantarum YRL45 to mice resulted in a substantial remodeling of the gut microbiota composition. Specifically, the intervention led to an enrichment of beneficial taxa, including Akkermansia muciniphila and Mycobacterium anisopliae, while concomitantly suppressing the proliferation of potentially harmful Clostridium spp. This microbial shift was accompanied by an increase in the production of SCFAs (short-chain fatty acids), specifically acetic and propionic acid. These SCFAs have been shown to enhance the phagocytic activity of macrophages and thereby promote resistance to intestinal inflammation (Brasiel & Potente Dutra Luquetti, 2025). Beyond viable bacteria, the fermentation matrix itself, which is abundant in vitamins and exopolysaccharides, plays a critical role in restoring homeostasis. According to Zhang et al. (2017), the consumption of Tibetan kefir in mice resulted in enhanced evenness and diversity of the intestinal flora. The ecological restoration demonstrated efficacy in reducing fecal nitrogen oxide levels and alleviating intestinal oxidative stress. Furthermore, fermentation-derived SCFAs have been shown to reinforce the intestinal mucosal layer and regulate glucose and lipid metabolism, thereby mitigating insulin resistance and metabolic disorders associated with aging (Fantini et al., 2023; Verediano et al., 2021).
3.7. Integrated network and synergistic mechanisms
Despite being presented as discrete biological processes in Sections 3.1–3.6, these mechanisms function within a highly interconnected regulatory network rather than independently. Aging is increasingly recognized as a multifactorial process driven by reciprocal interactions among oxidative stress, chronic low-grade inflammation, and metabolic dysregulation (López-Otín et al., 2023). Consequently, the anti-aging effects of fermented foods are more plausibly attributed to their coordinated modulation of these interdependent signaling pathways, rather than the regulation of any single mechanism alone, as schematically summarized in Fig. 5.
Fig. 5.
Anti-aging mechanism and related signaling pathways of fermented food.
The modulation of the gut ecosystem appears to function as a pivotal upstream regulatory event. The enrichment of beneficial microbial taxa, such as Lactobacillus and Akkermansia, in conjunction with the enhanced production of SCFAs, contributes to the reinforcement of intestinal epithelial barrier integrity. This, in turn, limits the translocation of bacterial endotoxins, particularly lipopolysaccharide, into systemic circulation. This effect attenuates a major initiating factor of chronic low-grade inflammation and, indirectly, reduces peripheral oxidative stress (Franceschi et al., 2018; Vaiserman et al., 2017). Research has demonstrated that bioactive compounds derived from fermented foods possess the capacity to disrupt the self-amplifying cycle between oxidative stress and inflammation. At the mechanistic level, direct scavenging of ROS, in conjunction with the activation of Nrf2-dependent antioxidant defenses, results in the suppression of downstream activation of the pro-inflammatory NF-κB signaling pathway. Concurrently, the attenuation of inflammatory cytokine production has been shown to reduce intracellular ROS generation, thereby disrupting the feed-forward loop that drives cumulative cellular damage during aging (Gao et al., 2022). Metabolic signaling also serves as a critical interface linking redox homeostasis to cellular quality control mechanisms. Fermentation-derived bioactive compounds, including polyphenols and bioactive peptides, have been demonstrated to modulate central energy sensors such as AMPK and SIRT1. The activation of these pathways has been shown to suppress mTOR signaling, thereby promoting autophagy-mediated clearance of damaged organelles. Concurrently, this process limits premature apoptosis through SIRT1-dependent deacetylation of p53 (Yang et al., 2025).
In summary, the evidence suggests that the consumption of fermented foods may offer benefits that contribute to a healthier, longer life. These effects appear to be the result of a coordinated, multi-target mode of action, encompassing modulation of the gut ecosystem to limit systemic inflammatory triggers, attenuation of the reciprocal amplification between oxidative stress and inflammation, and regulation of cellular longevity pathways via key metabolic sensors.
4. Current challenges and limitations
A mounting body of evidence suggests that the consumption of fermented foods may offer benefits that promote healthy aging. However, it is imperative to address the challenges and safety concerns associated with this practice to ensure that any potential benefits are not outweighed by unintended risks. Of particular concern are the presence of antinutritional factors and fermentation-associated byproducts. Fermentation has been demonstrated to be an effective method for reducing antinutritional components, such as phytates, trypsin inhibitors, and tannins, that are present in raw legumes and cereals (Sharma, 2021). However, the efficiency of antinutritional factor degradation is contingent upon the microbial strains employed, the nature of the substrate, and the specific fermentation conditions. Suboptimal or insufficiently controlled fermentation processes may therefore lead to the persistence of residual antinutritional components, which can compromise mineral bioavailability and protein digestibility. This is a particularly salient issue for older adults with compromised gastrointestinal function (Salim et al., 2023). Furthermore, spontaneous or uncontrolled fermentation has been shown to increase the risk of contamination by undesirable microorganisms, potentially leading to the formation of mycotoxins (e.g., aflatoxins) or excessive accumulation of biogenic amines such as histamine and tyramine (Saha Turna et al., 2024). Elevated concentrations of these amines, frequently reported in aged cheeses and certain fermented soy products, have been linked to adverse physiological responses, including headaches, elevated blood pressure, and pseudo-allergic reactions. Such effects may pose additional concerns for older individuals, particularly those with pre-existing cardiovascular susceptibility.
The high sodium content of many traditional fermented foods represents a significant limitation. A considerable proportion of products, including pickled vegetables (e.g., kimchi and sauerkraut) and fermented condiments such as soy sauce, depend on elevated salt concentrations to ensure microbial stability and product safety during fermentation and storage (Fitsum et al., 2025). Excessive sodium intake is a well-recognized risk factor for hypertension, cardiovascular disease, and gastric disorders. In the context of aging populations, the prolonged ingestion of high-salt fermented foods has the potential to mitigate their anticipated health benefits, thereby augmenting the cardiovascular strain. Consequently, the development and validation of low-salt or salt-reduced fermentation strategies have emerged as a significant technological and nutritional priority for the sustainable application of fermented foods in healthy aging.
The limited standardization and reproducibility of fermented foods constitute additional barriers to their translational application as anti-aging interventions. It is well established that traditional fermentation processes are inherently variable, often exhibiting pronounced batch-to-batch differences in microbial composition and metabolite profiles. This variability can lead to inconsistent bioactive properties (Tamang et al., 2017). This variability leads to inconsistent concentrations of bioactive compounds, which complicates the definition of standardized intake levels, the establishment of clear dose–response relationships, and the reproducibility of reported health outcomes. Collectively, these limitations hinder the incorporation of fermented foods into evidence-based nutritional guidelines and clinical recommendations (Hernández-Velázquez et al., 2024).
5. Conclusion and prospect
The deliberate extension of health span has long been a central objective in biomedical and nutritional research. In the context of a rapidly aging global population, there has been an increasing focus on food-based strategies, with fermented foods emerging as a particularly promising category. The evidence synthesized in this review indicates that the consumption of fermented foods may exert anti-aging effects primarily through the attenuation of oxidative stress, modulation of inflammatory responses and immune function, preservation of cellular homeostasis, and regulation of gut microbiota composition. The beneficial effects of this process are largely attributed to the synergistic actions of viable microorganisms and fermentation-derived bioactive compounds, including organic acids, phenolic metabolites, polysaccharides, and bioactive peptides.
Despite these encouraging findings, the extant evidence base remains heterogeneous, and a comprehensive understanding of how fermented foods influence aging processes is still incomplete. In light of the strengths and limitations identified in this review, the following research priorities are proposed for future investigation:
-
(1)
Mechanistic Elucidation via Multi-Omics. Future research should prioritize causal mechanistic validation by integrating genetic manipulation with multi-omics approaches to clarify how fermentation-derived components regulate key longevity-related pathways, particularly Nrf2-driven antioxidant defense, AMPK-mediated metabolic control, and sirtuin signaling (SIRT1/SIRT3).
-
(2)
Deciphering interactions within complex food matrices. Fermented foods are intricate matrices in which peptides, phenolics, and microbes interact. It is imperative to methodically examine whether these components manifest synergistic or antagonistic effects on aging endpoints through component fractionation and recombination experiments, as opposed to studying isolated compounds in a vacuum.
-
(3)
Strengthening translational and clinical evidence. These trials must incorporate validated aging biomarkers, such as oxidative stress indices, inflammatory cytokines, and cognitive functional endpoints, while accounting for inter-individual variability in metabolism.
-
(4)
Cautious advancement toward personalized nutrition. Despite the nascent state of personalized nutrition research, future studies may integrate dietary interventions with microbiota and metabolic profiling to identify response patterns. This integration has the potential to inform stratified rather than universal dietary recommendations.
-
(5)
Evidence-guided innovation in fermentation technology. The potential of synthetic ecology advancements should be exploited to enhance the efficacy of starter cultures. The objective is to enhance product consistency and safety by mitigating risk factors (e.g., biogenic amines, excessive salt) while maximizing the bioavailability of anti-aging bioactives.
In summary, the consumption of fermented foods is associated with numerous health benefits, including the potential to support healthy aging. To realize this potential, a shift toward mechanism-driven research, rigorous clinical validation, and evidence-based technological innovation is necessary. This will establish a solid scientific foundation for the rational development of fermented food–based strategies for healthy aging.
Data availability statement
Data sharing not applicable to this article as no datasets were generated or analysed during the current study.
CRediT authorship contribution statement
Hongyu Mu: Writing–original draft. Yeyan Yao: Methodology. Yongqiang Gong: Writing–review & editing. Tao Yang: Supervision.
Funding
Postgraduate Scientific Research and Innovation Project, Central South University of Forestry and Technology (2025CX01110).
Declaration of competing interest
The authors declare that they have no conflicts of interest to disclose. All authors have approved the final version of the manuscript for submission.
Handling Editor: Professor Alejandro G.Marangoni
Contributor Information
Hongyu Mu, Email: muhongyu0710@163.com.
Yeyan Yao, Email: 15207796469@163.com.
Yongqiang Gong, Email: 1039517015@qq.com.
Tao Yang, Email: yangtao@csuft.edu.cn.
References
- Abdel-Aal E.M. Legumes and cereals: physicochemical characterization, technical innovation and nutritional challenges. Foods. 2023;13(1) doi: 10.3390/foods13010005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agrawal A., Agrawal S., Gupta S. Role of dendritic cells in inflammation and loss of tolerance in the elderly. Front. Immunol. 2017;8:896. doi: 10.3389/fimmu.2017.00896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ali N.M., Yeap S.K., Yusof H.M., Beh B.K., Ho W.Y., Koh S.P., Abdullah M.P., Alitheen N.B., Long K. Comparison of free amino acids, antioxidants, soluble phenolic acids, cytotoxicity and immunomodulation of fermented mung bean and soybean. J. Sci. Food Agric. 2016;96(5):1648–1658. doi: 10.1002/jsfa.7267. [DOI] [PubMed] [Google Scholar]
- Anand Singh T., Nongthombam G., Goksen G., Singh H.B., Rajauria G., Kumar Sarangi P. Hawaijar - an ethnic vegan fermented soybean food of Manipur, India: a comprehensive review. Food Res. Int. 2023;170 doi: 10.1016/j.foodres.2023.112983. [DOI] [PubMed] [Google Scholar]
- Ano Y., Ayabe T., Kutsukake T., Ohya R., Takaichi Y., Uchida S., Yamada K., Uchida K., Takashima A., Nakayama H. Novel lactopeptides in fermented dairy products improve memory function and cognitive decline. Neurobiol. Aging. 2018;72:23–31. doi: 10.1016/j.neurobiolaging.2018.07.016. [DOI] [PubMed] [Google Scholar]
- Ano Y., Ozawa M., Kutsukake T., Sugiyama S., Uchida K., Yoshida A., Nakayama H. Preventive effects of a fermented dairy product against Alzheimer's disease and identification of a novel oleamide with enhanced microglial phagocytosis and anti-inflammatory activity. PLoS One. 2015;10(3) doi: 10.1371/journal.pone.0118512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Badr G., Ebaid H., Mohany M., Abuelsaad A.S. Modulation of immune cell proliferation and chemotaxis towards CC chemokine ligand (CCL)-21 and CXC chemokine ligand (CXCL)-12 in undenatured whey protein-treated mice. J. Nutr. Biochem. 2012;23(12):1640–1646. doi: 10.1016/j.jnutbio.2011.11.006. [DOI] [PubMed] [Google Scholar]
- Barathikannan K., Tyagi A., Shan L., Kim N.-H., Lee D.-S., Park J.-S., Chelliah R., Oh D.-H. Antiobesity and antioxidative effect of fermented brown rice using in vitro with in vivo Caenorhabditis elegans model. Life. 2023;13(2):374. doi: 10.3390/life13020374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barone M., D'Amico F., Rampelli S., Brigidi P., Turroni S. Age-related diseases, therapies and gut microbiome: a new frontier for healthy aging. Mech. Ageing Dev. 2022;206 doi: 10.1016/j.mad.2022.111711. [DOI] [PubMed] [Google Scholar]
- Bhanja Dey T., Kuhad R.C. Upgrading the antioxidant potential of cereals by their fungal fermentation under solid-state cultivation conditions. Lett. Appl. Microbiol. 2014;59(5):493–499. doi: 10.1111/lam.12300. [DOI] [PubMed] [Google Scholar]
- Bintsis T. Dairy fermentation 2.0. Fermentation. 2024;10(9):456. doi: 10.3390/fermentation10090456. [DOI] [Google Scholar]
- Brasiel P.G.A., Potente Dutra Luquetti S.C. Effects of probiotics supplementation on short-chain fatty acids: a systematic review of randomized controlled trials. Nutr. Rev. 2025 doi: 10.1093/nutrit/nuaf047. [DOI] [PubMed] [Google Scholar]
- Bu Y., Liu Y., Zhang T., Liu Y., Zhang Z., Yi H. Bacteriocin-producing Lactiplantibacillus plantarum YRL45 enhances intestinal immunity and regulates gut microbiota in mice. Nutrients. 2023;15(15) doi: 10.3390/nu15153437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calabrò A., Accardi G., Aiello A., Caruso C., Galimberti D., Candore G. Senotherapeutics to counteract senescent cells are prominent topics in the context of anti-ageing strategies. Int. J. Mol. Sci. 2024;25(3) doi: 10.3390/ijms25031792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao Z.H., Green-Johnson J.M., Buckley N.D., Lin Q.Y. Bioactivity of soy-based fermented foods: a review. Biotechnol. Adv. 2019;37(1):223–238. doi: 10.1016/j.biotechadv.2018.12.001. [DOI] [PubMed] [Google Scholar]
- Cardin G., Poupet C., Bonnet M., Veisseire P., Ripoche I., Chalard P., Chauder A., Saunier E., Priam J., Bornes S., Rios L. A mechanistic study of the antiaging effect of raw-milk cheese extracts. Nutrients. 2021;13(3) doi: 10.3390/nu13030897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cardoso R.R., Moreira L.d.P.D., de Campos Costa M.A., Toledo R.C.L., Grancieri M., Nascimento T.P.d., Ferreira M.S.L., da Matta S.L.P., Eller M.R., Duarte Martino H.S., de Barros F.A.R. Kombuchas from green and black teas reduce oxidative stress, liver steatosis and inflammation, and improve glucose metabolism in Wistar rats fed a high-fat high-fructose diet [10.1039/D1FO02106K] Food Funct. 2021;12(21):10813–10827. doi: 10.1039/D1FO02106K. [DOI] [PubMed] [Google Scholar]
- Chan L., Tseng Y., Liu C., Liang C.J. Anti-oxidant and anti-aging activities of fermented vegetable-fruit drink. J. Food Nutr. Res. 2021;9:240–250. doi: 10.12691/jfnr-9-5-1. [DOI] [Google Scholar]
- Chan Y.-C., Lee I.-T., Wang M.-F., Yeh W.-C., Liang B.-C.J. Tempeh attenuates cognitive deficit, antioxidant imbalance, and amyloid β of senescence-accelerated mice by modulating Nrf2 expression via MAPK pathway. J. Funct.Foods. 2018;50:112–119. doi: 10.1016/j.jff.2018.09.023. [DOI] [Google Scholar]
- Chao J., Coleman R.A., Keating D.J., Martin A.M. Gut microbiome regulation of gut hormone secretion. Endocrinology. 2025;166(4) doi: 10.1210/endocr/bqaf004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen J., Wang A., Chen Q. SirT3 and p53 deacetylation in aging and cancer. J. Cell. Physiol. 2017;232(9):2308–2311. doi: 10.1002/jcp.25669. [DOI] [PubMed] [Google Scholar]
- Chen Y., Liu C., Yang F., Chen H., Yang C., Fan Z., Xiao Y., Xiao D. UPLC–QQQ–MS/MS-based widely targeted metabolomic analysis, antioxidant and α-glucosidase inhibitory activities of mulberry leaves processed by solid-state fermentation. LWT. 2023;188 doi: 10.1016/j.lwt.2023.115351. [DOI] [Google Scholar]
- Chou C.H., Liu C.W., Yang D.J., Wu Y.H., Chen Y.C. Amino acid, mineral, and polyphenolic profiles of black vinegar, and its lipid lowering and antioxidant effects in vivo. Food Chem. 2015;168:63–69. doi: 10.1016/j.foodchem.2014.07.035. [DOI] [PubMed] [Google Scholar]
- Chou S.-T., Chen Y.-H., Chen Y.-H., Tsai Y.C., Chung Y.-C., Li J.-P. Safety and antioxidant assessments of BLR-E50, 50% ethanolic extract from red beans co-fermented by Bacillus subtilis and Lactobacillus bulgaricus. Food Chem. Toxicol. 2025;197 doi: 10.1016/j.fct.2025.115261. [DOI] [PubMed] [Google Scholar]
- Chua S.P.L., Umayat L.D., Medina P.M.B. Phenolic-rich lees from Philippine rice wine (tapuy) increases Caenorhabditis elegans lifespan in spite of having low antioxidant activity. Food Product. Proc. Nutrit. 2024;6(1):1. doi: 10.1186/s43014-023-00181-w. [DOI] [Google Scholar]
- Conrad N., Misra S., Verbakel J.Y., Verbeke G., Molenberghs G., Taylor P.N., Mason J., Sattar N., McMurray J.J.V., McInnes I.B., Khunti K., Cambridge G. Incidence, prevalence, and co-occurrence of autoimmune disorders over time and by age, sex, and socioeconomic status: a population-based cohort study of 22 million individuals in the UK. Lancet. 2023;401(10391):1878–1890. doi: 10.1016/s0140-6736(23)00457-9. [DOI] [PubMed] [Google Scholar]
- Cui J., Xia P., Zhang L., Hu Y., Xie Q., Xiang H. A novel fermented soybean, inoculated with selected Bacillus, Lactobacillus and Hansenula strains, showed strong antioxidant and anti-fatigue potential activity. Food Chem. 2020;333 doi: 10.1016/j.foodchem.2020.127527. [DOI] [PubMed] [Google Scholar]
- Cui R., Zhang C., Pan Z.H., Hu T.G., Wu H. Probiotic-fermented edible herbs as functional foods: a review of current status, challenges, and strategies. Compr. Rev. Food Sci. Food Saf. 2024;23(2) doi: 10.1111/1541-4337.13305. [DOI] [PubMed] [Google Scholar]
- Das D., Sarkar S., Borsingh Wann S., Kalita J., Manna P. Current perspectives on the anti-inflammatory potential of fermented soy foods. Food Res. Int. 2022;152 doi: 10.1016/j.foodres.2021.110922. [DOI] [PubMed] [Google Scholar]
- Das G., Paramithiotis S., Sundaram Sivamaruthi B., Wijaya C.H., Suharta S., Sanlier N., Shin H.S., Patra J.K. Traditional fermented foods with anti-aging effect: a concentric review. Food Res. Int. 2020;134 doi: 10.1016/j.foodres.2020.109269. [DOI] [PubMed] [Google Scholar]
- Deng X., Hou Y., Zhou H., Li Y., Xue Z., Xue X., Huang G., Huang K., He X., Xu W. Hypolipidemic, anti-inflammatory, and anti-atherosclerotic effects of tea before and after microbial fermentation. Food Sci. Nutr. 2021;9(2):1160–1170. doi: 10.1002/fsn3.2096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deretic V. Autophagy in inflammation, infection, and immunometabolism. Immunity. 2021;54(3):437–453. doi: 10.1016/j.immuni.2021.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diet A., Poix C., Bonnet M., Coelho C., Ripoche I., Decombat C., Priam J., Saunier E., Chalard P., Bornes S., Caldefie-Chezet F., Rios L. Exploring the impact of French raw-milk cheeses on oxidative process using Caenorhabditis elegans and human leukocyte models. Nutrients. 2024;16(12):1862. doi: 10.3390/nu16121862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dimidi E., Cox S.R., Rossi M., Whelan K. Fermented foods: definitions and characteristics, impact on the gut microbiota and effects on gastrointestinal health and disease. Nutrients. 2019;11(8) doi: 10.3390/nu11081806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong H., Rowland I., Thomas L.V., Yaqoob P. Immunomodulatory effects of a probiotic drink containing Lactobacillus casei Shirota in healthy older volunteers. Eur. J. Nutr. 2013;52(8):1853–1863. doi: 10.1007/s00394-012-0487-1. [DOI] [PubMed] [Google Scholar]
- Fantini C., Corinaldesi C., Lenzi A., Migliaccio S., Crescioli C. Vitamin D as a shield against aging. Int. J. Mol. Sci. 2023;24(5) doi: 10.3390/ijms24054546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fatt M.P., Tran L.M., Vetere G., Storer M.A., Simonetta J.V., Miller F.D., Frankland P.W., Kaplan D.R. Restoration of hippocampal neural precursor function by ablation of senescent cells in the aging stem cell niche. Stem Cell Rep. 2022;17(2):259–275. doi: 10.1016/j.stemcr.2021.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fitsum S., Gebreyohannes G., Sbhatu D.B. Bioactive compounds in fermented foods: health benefits, safety, and future perspectives. Appl. Food Res. 2025;5(2) doi: 10.1016/j.afres.2025.101097. [DOI] [Google Scholar]
- Franceschi C., Garagnani P., Parini P., Giuliani C., Santoro A. Inflammaging: a new immune–metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 2018;14(10):576–590. doi: 10.1038/s41574-018-0059-4. [DOI] [PubMed] [Google Scholar]
- Funk M.C., Zhou J., Boutros M. Ageing, metabolism and the intestine. EMBO Rep. 2020;21(7) doi: 10.15252/embr.202050047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gabriele M., Arouna N., Árvay J., Longo V., Pucci L. Sourdough fermentation improves the antioxidant, antihypertensive, and anti-inflammatory properties of triticum dicoccum. Int. J. Mol. Sci. 2023;24(7) doi: 10.3390/ijms24076283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao L., Liu X., Luo X., Lou X., Li P., Li X., Liu X. Antiaging effects of dietary supplements and natural products. Front. Pharmacol. 2023;14 doi: 10.3389/fphar.2023.1192714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao W., Guo L., Yang Y., Wang Y., Xia S., Gong H., Zhang B.-K., Yan M. Dissecting the crosstalk between Nrf2 and NF-κB response pathways in drug-induced toxicity. Front. Cell Dev. Biol. 2022;9:2021. doi: 10.3389/fcell.2021.809952. [Review] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong Q.Y., Cai L., Jing Y., Wang W., Yang D.X., Chen S.W., Tian H.L. Urolithin A alleviates blood-brain barrier disruption and attenuates neuronal apoptosis following traumatic brain injury in mice. Neural Regen. Res. 2022;17(9):2007–2013. doi: 10.4103/1673-5374.335163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong R., Wang D., Abbas G., Li S., Liu Q., Cui M., Zhang X.E. A switch-on molecular biosensor for detection of caspase-3 and imaging of apoptosis of cells. Sci. China Life Sci. 2022;65(3):540–549. doi: 10.1007/s11427-021-1986-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez-Gonzalez C.R., Melgar-Lalanne G., Oliveira D.L., Al-Habsi N. Editorial: fermentation and enzymatic processes for the production of functional food [Editorial] Front. Sustain. Food Syst. 2023;7 doi: 10.3389/fsufs.2023.1270631. [DOI] [Google Scholar]
- Gopikrishna T., Suresh Kumar H.K., Perumal K., Elangovan E. Impact of Bacillus in fermented soybean foods on human health. Ann. Microbiol. 2021;71(1):30. doi: 10.1186/s13213-021-01641-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gui Q., Wang A., Zhao X., Huang S., Tan Z., Xiao C., Yang Y. Effects of probiotic supplementation on natural killer cell function in healthy elderly individuals: a meta-analysis of randomized controlled trials. Eur. J. Clin. Nutr. 2020;74(12):1630–1637. doi: 10.1038/s41430-020-0670-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gulmez C., Atakisi O. Kumiss supplementation reduces oxidative stress and activates Sirtuin deacetylases by regulating antioxidant system. Nutr. Cancer. 2020;72(3):495–503. doi: 10.1080/01635581.2019.1635628. [DOI] [PubMed] [Google Scholar]
- Harnavi H., Yanwirasti Y., Bambang P., Endang Purwati R. The effect of giving dadih on Malondialdehyde levels and renal interstitial fibrosis at aging kidney. Open Access Maced. J. Med. Sci. 2020;8(A) doi: 10.3889/OAMJMS.2020.4285. [DOI] [Google Scholar]
- Hasegawa Y., Bolling B.W.J. Yogurt consumption for improving immune health. Curr. Opin. Food Sci. 2023;51 doi: 10.1016/j.cofs.2023.101017. [DOI] [Google Scholar]
- Hernández-Velázquez R., Flörl L., Lavrinienko A., Sebechlebská Z., Merk L., Greppi A., Bokulich N.A. The future is fermented: microbial biodiversity of fermented foods is a critical resource for food innovation and human health. Trends Food Sci. Technol. 2024;150 doi: 10.1016/j.tifs.2024.104569. [DOI] [Google Scholar]
- Hofer S.J., Carmona-Gutierrez D., Mueller M.I., Madeo F. The ups and downs of caloric restriction and fasting: from molecular effects to clinical application. EMBO Mol. Med. 2022;14(1) doi: 10.15252/emmm.202114418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hor Y.Y., Ooi C.H., Khoo B.Y., Choi S.B., Seeni A., Shamsuddin S., Oon C.E., Ong K.L., Jeong W.S., Liong M.T. Lactobacillus strains alleviated aging symptoms and aging-induced metabolic disorders in aged rats. J. Med. Food. 2019;22(1):1–13. doi: 10.1089/jmf.2018.4229. [DOI] [PubMed] [Google Scholar]
- Hsieh C.-C., Lin H.-W., Huang H.-C., Khumsupan D., Shen S.-C., Lin S.-P., Hsieh C.-W., Tsai T.-Y., Jantama S.S., Kuo H.-C., Cheng K.-C. Peptide from tempeh-like fermented Chenopodium formosanum counters senescence while enhancing antioxidant ability in non-replicative aging. LWT. 2025;222 doi: 10.1016/j.lwt.2025.117641. [DOI] [Google Scholar]
- Hui Y., Jun-Li H., Chuang W. Anti-oxidation and anti-aging activity of polysaccharide from Malus micromalus Makino fruit wine. Int. J. Biol. Macromol. 2019;121:1203–1212. doi: 10.1016/j.ijbiomac.2018.10.096. [DOI] [PubMed] [Google Scholar]
- Hui Y., Wen S., Lihong W., Chuang W., Chaoyun W. Molecular structures of nonvolatile components in the Haihong fruit wine and their free radical scavenging effect. Food Chem. 2021;353 doi: 10.1016/j.foodchem.2021.129298. [DOI] [PubMed] [Google Scholar]
- Huma N., Rafiq S., Sameen A., Pasha I., Khan M.I. Antioxidant potential of buffalo and cow milk Cheddar cheeses to tackle human colon adenocarcinoma (Caco-2) cells. Asian-Australas. J. Anim. Sci. 2018;31(2):287–292. doi: 10.5713/ajas.17.0031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ibe S., Kumada K., Yoshida K., Otobe K.J.B., biotechnology, & biochemistry Natto (fermented soybean) extract extends the adult lifespan of Caenorhabditis elegans. Biosci. Biotechnol. Biochem. 2013;77(2):392–394. doi: 10.1271/bbb.120726. [DOI] [PubMed] [Google Scholar]
- Ibrahim S.A., Yeboah P.J., Ayivi R.D., Eddin A.S., Wijemanna N.D., Paidari S., Bakhshayesh R.V.J., Technology A review and comparative perspective on health benefits of probiotic and fermented foods. Int. J. Food Sci. Technol. 2023;58(10):4948–4964. doi: 10.1111/ijfs.16619. [DOI] [Google Scholar]
- Iwatani S., Yamamoto N. Functional food products in Japan: a review. Food Sci. Hum. Wellness. 2019;8(2):96–101. doi: 10.1016/j.fshw.2019.03.011. [DOI] [Google Scholar]
- Jang H.M., Han S.K., Kim J.K., Oh S.J., Jang H.B., Kim D.H. Lactobacillus sakei alleviates high-fat-diet-induced obesity and anxiety in mice by inducing AMPK activation and SIRT1 expression and inhibiting Gut Microbiota-mediated NF-κB activation. Mol. Nutr. Food Res. 2019;63(6) doi: 10.1002/mnfr.201800978. [DOI] [PubMed] [Google Scholar]
- Jeong D.Y., Ryu M.S., Yang H.J., Park S. γ-PGA-Rich Chungkookjang, short-term fermented soybeans: prevents memory impairment by modulating brain insulin sensitivity, neuro-inflammation, and the gut-microbiome-brain axis. Foods. 2021;10(2) doi: 10.3390/foods10020221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang H., Liu Y., Yu Y., Yan Y. Sirtuin 1 suppresses hydrogen peroxide-induced senescence and promotes viability and migration in lens epithelial cells by inhibiting Forkhead Box protein O1/Toll-Like receptor 4 pathway. J. Biochem. Mol. Toxicol. 2025;39(2) doi: 10.1002/jbt.70150. [DOI] [PubMed] [Google Scholar]
- Jothi D., Kulka L.A.M. Strategies for modeling aging and age-related diseases. NPJ Aging. 2024;10(1):32. doi: 10.1038/s41514-024-00161-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Juricic P., Lu Y.X., Leech T., Drews L.F., Paulitz J., Lu J., Nespital T., Azami S., Regan J.C., Funk E., Fröhlich J., Grönke S., Partridge L. Long-lasting geroprotection from brief rapamycin treatment in early adulthood by persistently increased intestinal autophagy. Nat. Aging. 2022;2(9):824–836. doi: 10.1038/s43587-022-00278-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kancabaş A., Karakaya S. Angiotensin-converting enzyme (ACE)-inhibitory activity of boza, a traditional fermented beverage. J. Sci. Food Agric. 2013;93(3):641–645. doi: 10.1002/jsfa.5883. [DOI] [PubMed] [Google Scholar]
- Kim B., Park K.Y., Kim H.Y., Ahn S.C., Cho E.J.J.F.S., Biotechnology Anti-aging effects and mechanisms of kimchi during fermentation under stress-induced premature senescence cellular system. Food Sci. Biotechnol. 2011;20:643–649. doi: 10.1007/s10068-011-0091-9. [DOI] [Google Scholar]
- Kim D., Han G.D. Ameliorating effects of fermented rice bran extract on oxidative stress induced by high glucose and hydrogen peroxide in 3T3-L1 adipocytes. Plant Foods Hum. Nutr. 2011;66(3):285–290. doi: 10.1007/s11130-011-0243-3. [DOI] [PubMed] [Google Scholar]
- Kim S.S., Lee C.K. Growth signaling and longevity in mouse models. BMB Rep. 2019;52(1):70–85. doi: 10.5483/BMBRep.2019.52.1.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ko J.W., Chung Y.S., Kwak C.S., Kwon Y.H. Doenjang, A Korean traditional fermented soybean paste, ameliorates neuroinflammation and neurodegeneration in mice fed a high-fat diet. Nutrients. 2019;11(8) doi: 10.3390/nu11081702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koh Y.C., Kuo L.H., Chang Y.Y., Tung Y.C., Lo Y.C., Pan M.H. Modulatory effect of fermented Black soybean and adlay on Gut microbiota contributes to healthy aging. Mol. Nutr. Food Res. 2023;67(5) doi: 10.1002/mnfr.202200700. [DOI] [PubMed] [Google Scholar]
- Kostelac D., Gerić M., Gajski G., Frece J. Probiotic bacteria isolated from fermented meat displays high antioxidant and anti-inflammatory potential. Mutagenesis. 2022;38(1):58–63. doi: 10.1093/mutage/geac023. [DOI] [PubMed] [Google Scholar]
- Kumar D., Rizvi S.I. Black tea supplementation augments redox balance in rats: relevance to aging. Arch. Physiol. Biochem. 2017;123(4):212–218. doi: 10.1080/13813455.2017.1302963. [DOI] [PubMed] [Google Scholar]
- Lee C.L., Kuo T.F., Wu C.L., Wang J.J., Pan T.M. Red mold rice promotes neuroprotective sAPPalpha secretion instead of Alzheimer's risk factors and amyloid beta expression in hyperlipidemic Abeta40-infused rats. J. Agric. Food Chem. 2010;58(4):2230–2238. doi: 10.1021/jf904027y. [DOI] [PubMed] [Google Scholar]
- Lee C.Y., Isaac H.B., Wang H., Huang S.H., Long L.H., Jenner A.M., Kelly R.P., Halliwell B. Cautions in the use of biomarkers of oxidative damage; the vascular and antioxidant effects of dark soy sauce in humans. Biochem. Biophys. Res. Commun. 2006;344(3):906–911. doi: 10.1016/j.bbrc.2006.03.217. [DOI] [PubMed] [Google Scholar]
- Lei S., Zhang Z., Xie G., Zhao C., Miao Y., Chen D., Zhang G., Liu H., Peng C., Hou Y., Gong J. Theabrownin modulates the gut microbiome and serum metabolome in aging mice induced by D-galactose. J. Funct.Foods. 2022;89 doi: 10.1016/j.jff.2022.104941. [DOI] [Google Scholar]
- Lindner J.D., Bernini V. New insights into food fermentation. Foods. 2022;11(3) doi: 10.3390/foods11030283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J.R., Chen M.J., Lin C.W. Antimutagenic and antioxidant properties of milk-kefir and soymilk-kefir. J. Agric. Food Chem. 2005;53(7):2467–2474. doi: 10.1021/jf048934k. [DOI] [PubMed] [Google Scholar]
- Liu L., Chen X., Hao L., Zhang G., Jin Z., Li C., Yang Y., Rao J., Chen B. Traditional fermented soybean products: processing, flavor formation, nutritional and biological activities. Crit. Rev. Food Sci. Nutr. 2022;62(7):1971–1989. doi: 10.1080/10408398.2020.1848792. [DOI] [PubMed] [Google Scholar]
- Liu R., Fu Z., Zhang F., Mao Q., Luan C., Han X., Xue J., Wang D., Qin S., Hao F.J.F.S., Wellness H. Effect of yellow rice wine on anti-aging ability in aged mice induced by d-galactose. Food Sci. Hum. Wellness. 2020;9(2):184–191. doi: 10.1016/j.fshw.2020.02.003. [DOI] [Google Scholar]
- Liu X., Chen Y., Zhang S., Dong L. Gut microbiota-mediated immunomodulation in tumor. J. Exp. Clin. Cancer Res. 2021;40(1):221. doi: 10.1186/s13046-021-01983-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y., Zhang X., Liu X., Li R., Yang X., Liao Z., Fang X., Wang J. Enhancing the anti-aging potential of green tea extracts through liquid-state fermentation with Aspergillus niger RAF106. Foods. 2025;14(20):3548. doi: 10.3390/foods14203548. https://www.mdpi.com/2304-8158/14/20/3548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu G.Y., Sabatini D.M. mTOR at the nexus of nutrition, growth, ageing and disease. Nat. Rev. Mol. Cell Biol. 2020;21(4):183–203. doi: 10.1038/s41580-019-0199-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logozzi M., Di Raimo R., Mizzoni D., Andreotti M., Spada M., Macchia D., Fais S. Beneficial effects of Fermented Papaya Preparation (FPP®) supplementation on redox balance and aging in a mouse model. Antioxidants. 2020;9(2):144. doi: 10.3390/antiox9020144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- López-Otín C., Blasco M.A., Partridge L., Serrano M., Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186(2):243–278. doi: 10.1016/j.cell.2022.11.001. [DOI] [PubMed] [Google Scholar]
- López-Otín C., Blasco M.A., Partridge L., Serrano M., Kroemer G.J.C. The hallmarks of aging. Cell. 2013;153(6):1194–1217. doi: 10.1016/j.cell.2013.05.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo J., Si H., Jia Z., Liu D. Dietary anti-aging polyphenols and potential mechanisms. Antioxidants. 2021;10(2) doi: 10.3390/antiox10020283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo J., Tian Z., Yuan W., Peng X., Zhou H., Shen Q., Luo Y., Guo Y., Shi Z., Jiang X., Chen B., Pan D., Fan X. Anti-aging effect of Limosilactobacillus fermentum CGMCC 17434 in mice fed with fermented selenium-enriched yogurt. Food Biosci. 2025;68 doi: 10.1016/j.fbio.2025.106582. [DOI] [Google Scholar]
- Luti S., Mazzoli L., Ramazzotti M., Galli V., Venturi M., Marino G., Lehmann M., Guerrini S., Granchi L., Paoli P., Pazzagli L. Antioxidant and anti-inflammatory properties of sourdoughs containing selected Lactobacilli strains are retained in breads. Food Chem. 2020;322 doi: 10.1016/j.foodchem.2020.126710. [DOI] [PubMed] [Google Scholar]
- Mackowiak P.A. Recycling metchnikoff: probiotics, the intestinal microbiome and the quest for long life. Front. Public Health. 2013;1:52. doi: 10.3389/fpubh.2013.00052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahamud A., Tanvir I.A., Kabir M.E., Samonty I., Chowdhury M.A.H., Rahman M.A. Gerobiotics: exploring the potential and limitations of repurposing probiotics in addressing aging hallmarks and chronic diseases. Probiotics Antimicrob. Proteins. 2025 doi: 10.1007/s12602-025-10501-w. [DOI] [PubMed] [Google Scholar]
- Marco M.L., Sanders M.E., Gänzle M., Arrieta M.C., Cotter P.D., De Vuyst L., Hill C., Holzapfel W., Lebeer S., Merenstein D., Reid G., Wolfe B.E., Hutkins R. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on fermented foods. Nat. Rev. Gastroenterol. Hepatol. 2021;18(3):196–208. doi: 10.1038/s41575-020-00390-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melini F., Melini V., Luziatelli F., Ficca A.G., Ruzzi M. Health-promoting components in fermented foods: an up-to-date systematic review. Nutrients. 2019;11(5):1189. doi: 10.3390/nu11051189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miao X., Niu H., Sun M., Dong X., Hua M., Su Y., Wang J., Li D. A comparative study on the nutritional composition, protein structure and effects on gut microbiota of 5 fermented soybean products (FSPs) Food Res. Int. 2024;183 doi: 10.1016/j.foodres.2024.114199. [DOI] [PubMed] [Google Scholar]
- Mollakhalili-Meybodi N., Arab M., Zare L. Harmful compounds of soy milk: characterization and reduction strategies. J. Food Sci. Technol. 2022;59(10):3723–3732. doi: 10.1007/s13197-021-05249-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mushtaq M., Gani A., Masoodi F.A. Himalayan cheese (Kalari/Kradi) fermented with different probiotic strains: in vitro investigation of nutraceutical properties. LWT--Food Sci. Technol. 2019;104:53–60. doi: 10.1016/j.lwt.2019.01.024. [DOI] [Google Scholar]
- Nilsson A.G., Sundh D., Bäckhed F., Lorentzon M. Lactobacillus reuteri reduces bone loss in older women with low bone mineral density: a randomized, placebo-controlled, double-blind, clinical trial. J. Intern. Med. 2018;284(3):307–317. doi: 10.1111/joim.12805. [DOI] [PubMed] [Google Scholar]
- Octary T., Sung C.M., Chen R., Chiang K.J., Lee C.K., Arifin H., Banda K.J., Chou K.R. Global prevalence, incidence, and factors influencing falls in older adults with dementia: implications for nursing and healthy aging. J. Nurs. Scholarsh. 2025 doi: 10.1111/jnu.70012. [DOI] [PubMed] [Google Scholar]
- Oliveira Neto J.R., de Oliveira T.S., Ghedini P.C., Vaz B.G., Gil E.d.S. Antioxidant and vasodilatory activity of commercial beers. J. Funct.Foods. 2017;34:130–138. doi: 10.1016/j.jff.2017.04.019. [DOI] [Google Scholar]
- Orisakwe O.E., Amadi C.N., Frazzoli C., Dokubo A. Nigerian foods of probiotics relevance and chronic metal exposure: a systematic review. Environ. Sci. Pollut. Control Ser. 2020;27(16):19285–19297. doi: 10.1007/s11356-020-08537-2. [DOI] [PubMed] [Google Scholar]
- Ou H.L., Schumacher B. DNA damage responses and p53 in the aging process. Blood. 2018;131(5):488–495. doi: 10.1182/blood-2017-07-746396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park J.E., Hyun Y.J., Piao M.J., Kang K.A., Ryu Y.S., Shilnikova K., Zhen A.X., Ahn M.J., Ahn Y.S., Koh Y.S., Kang H.K., Hyun J.W. Mackerel-derived fermented fish oil protects skin against UVB-induced cellular damage by inhibiting oxidative stress. J. Funct.Foods. 2018;46:147–158. doi: 10.1016/j.jff.2018.04.057. [DOI] [Google Scholar]
- Park Y.K., Lee J.H., Mah J.H. Occurrence and reduction of biogenic amines in traditional Asian fermented soybean foods: a review. Food Chem. 2019;278:1–9. doi: 10.1016/j.foodchem.2018.11.045. [DOI] [PubMed] [Google Scholar]
- Partridge L., Fuentealba M., Kennedy B.K. The quest to slow ageing through drug discovery. Nat. Rev. Drug Discov. 2020;19(8):513–532. doi: 10.1038/s41573-020-0067-7. [DOI] [PubMed] [Google Scholar]
- Petrova P., Petrov K. Lactic acid fermentation of cereals and pseudocereals: ancient nutritional biotechnologies with modern applications. Nutrients. 2020;12(4) doi: 10.3390/nu12041118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Picca A., Marzetti E. Advancements in anti-aging treatment development. Int. J. Mol. Sci. 2023;24(10) doi: 10.3390/ijms24108515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polo A., Tlais A.Z.A., Filannino P., Da Ros A., Arora K., Cantatore V., Vincentini O., Nicolodi A., Nicolodi R., Gobbetti M., Di Cagno R. Novel fermented ice cream formulations with improved antiradical and anti-inflammatory features. Fermentation. 2023;9(2):117. doi: 10.3390/fermentation9020117. [DOI] [Google Scholar]
- Qiao Y., Zhang K., Zhang Z., Zhang C., Sun Y., Feng Z. Fermented soybean foods: a review of their functional components, mechanism of action and factors influencing their health benefits. Food Res. Int. 2022;158 doi: 10.1016/j.foodres.2022.111575. [DOI] [PubMed] [Google Scholar]
- Qin X., Xu W., Qiu L., Li Y., Liu R., Zhu L., Wang X. Plant-derived bioactive peptides in anti-aging applications: from molecular mechanisms to advanced delivery platforms. Trends Food Sci. Technol. 2026;167 doi: 10.1016/j.tifs.2025.105419. [DOI] [Google Scholar]
- Rajendran S., Silcock P., Bremer P. Flavour volatiles of fermented vegetable and fruit substrates: a review. Molecules. 2023;28(7) doi: 10.3390/molecules28073236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rizzoli R., Biver E. Role of fermented dairy products in the health benefits of a mediterranean diet. Aging Clin. Exp. Res. 2024;36(1):75. doi: 10.1007/s40520-024-02721-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ros M., Carrascosa J.M. Current nutritional and pharmacological anti-aging interventions. Biochim. Biophys. Acta Mol. Basis Dis. 2020;1866(3) doi: 10.1016/j.bbadis.2019.165612. [DOI] [PubMed] [Google Scholar]
- Ruiz de la Bastida A., Langa S., Peirotén Á., Fernández-Gonzalez R., Sánchez-Jiménez A., Maroto M., Antonio Curiel J., Guillamon E., Arqués J.L., Gutiérrez-Adán A., Landete J.M. Effect of fermented soy beverage in aged female mice model. Food Res. Int. 2023;169 doi: 10.1016/j.foodres.2023.112745. [DOI] [PubMed] [Google Scholar]
- Rul F., Béra-Maillet C., Champomier-Vergès M.C., El-Mecherfi K.E., Foligné B., Michalski M.C., Milenkovic D., Savary-Auzeloux I. Underlying evidence for the health benefits of fermented foods in humans. Food Funct. 2022;13(9):4804–4824. doi: 10.1039/d1fo03989j. [DOI] [PubMed] [Google Scholar]
- Saha Turna N., Chung R., McIntyre L. A review of biogenic amines in fermented foods: occurrence and health effects. Heliyon. 2024;10(2) doi: 10.1016/j.heliyon.2024.e24501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salim R., Nehvi I.B., Mir R.A., Tyagi A., Ali S., Bhat O.M. A review on anti-nutritional factors: unraveling the natural gateways to human health. Front. Nutr. 2023;10:2023. doi: 10.3389/fnut.2023.1215873. [Review] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salminen A., Kaarniranta K., Kauppinen A. Immunosenescence: the potential role of myeloid-derived suppressor cells (MDSC) in age-related immune deficiency. Cell. Mol. Life Sci. 2019;76(10):1901–1918. doi: 10.1007/s00018-019-03048-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samtiya M., Aluko R.E., Dhewa T. Plant food anti-nutritional factors and their reduction strategies: an overview. Food Product. Proc. Nutrit. 2020;2(1):6. doi: 10.1186/s43014-020-0020-5. [DOI] [Google Scholar]
- Sanyal K., Haldar J. Fungal pathogens: life cycle, infection, host immunity, and drug discovery. J. Med. Chem. 2025;68(8):7852–7853. doi: 10.1021/acs.jmedchem.5c00983. [DOI] [PubMed] [Google Scholar]
- Schluter J., Peled J.U., Taylor B.P., Markey K.A., Smith M., Taur Y., Niehus R., Staffas A., Dai A., Fontana E., Amoretti L.A., Wright R.J., Morjaria S., Fenelus M., Pessin M.S., Chao N.J., Lew M., Bohannon L., Bush A.…Xavier J.B. The gut microbiota is associated with immune cell dynamics in humans. Nature. 2020;588(7837):303–307. doi: 10.1038/s41586-020-2971-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seo M.Y., Chung S.Y., Choi W.K., Seo Y.K., Jung S.H., Park J.M., Seo M.J., Park J.K., Kim J.W., Park C.S. Anti-aging effect of rice wine in cultured human fibroblasts and keratinocytes. J. Biosci. Bioeng. 2009;107(3):266–271. doi: 10.1016/j.jbiosc.2008.11.016. [DOI] [PubMed] [Google Scholar]
- Seong J.S., Xuan S.H., Park S.H., Lee K.S., Park Y.M., Park S.N. Antioxidative and antiaging activities and component analysis of Lespedeza cuneata G. Don extracts fermented with Lactobacillus pentosus. J. Microbiol. Biotechnol. 2017;27(11):1961–1970. doi: 10.4014/jmb.1706.06028. [DOI] [PubMed] [Google Scholar]
- Sharma A. A review on traditional technology and safety challenges with regard to antinutrients in legume foods. J. Food Sci. Technol. 2021;58(8):2863–2883. doi: 10.1007/s13197-020-04883-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh J.P., Singh B., Kaur A.J.N., Food, Ageing, D. i., & Longevity Nutraceuticals and functional foods in aging and aging-associated diseases. Healthy Ageing Longevity. 2021:221–238. doi: 10.1007/978-3-030-83017-5_12. [DOI] [Google Scholar]
- Song L., Zhang S. Anti-aging activity and modes of action of compounds from natural food sources. Biomolecules. 2023;13(11) doi: 10.3390/biom13111600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soukas A.A., Hao H., Wu L. Metformin as anti-aging therapy: is it for everyone? Trends Endocrinol. Metabol. 2019;30(10):745–755. doi: 10.1016/j.tem.2019.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sørensen H.M., Rochfort K.D., Maye S., MacLeod G., Loscher C., Brabazon D., Freeland B. Bioactive ingredients from dairy-based lactic acid bacterial fermentations for functional food production and their health effects. Nutrients. 2023;15(22) doi: 10.3390/nu15224754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun Z.Q., Liu J.F., Luo W., Wong C.H., So K.F., Hu Y., Chiu K. Lycium barbarum extract promotes M2 polarization and reduces oligomeric amyloid-β-induced inflammatory reactions in microglial cells. Neural Regen. Res. 2022;17(1):203–209. doi: 10.4103/1673-5374.314325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tamang J.P., Holzapfel W.H., Shin D.H., Felis G.E. Editorial: microbiology of ethnic fermented foods and alcoholic beverages of the world [Editorial] Front. Microbiol. 2017;8:2017. doi: 10.3389/fmicb.2017.01377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang W., Xing Z., Hu W., Li C., Wang J., Wang Y.J.A.M., Biotechnology Antioxidative effects in vivo and colonization of Lactobacillus plantarum MA2 in the murine intestinal tract. Appl. Microbiol. Biotechnol. 2016;100:7193–7202. doi: 10.1007/s00253-016-7581-x. [DOI] [PubMed] [Google Scholar]
- Tang Z., Zhao Z., Chen S., Lin W., Wang Q., Shen N., Qin Y., Xiao Y., Chen H., Chen H., Bu T., Li Q., Yao H., Yuan M. Dragon fruit-kiwi fermented beverage: in vitro digestion, untargeted metabolome analysis and anti-aging activity in Caenorhabditis elegans. Front. Nutr. 2022;9 doi: 10.3389/fnut.2022.1052818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teng H., Xiao H., Li X., Huang J., Zhang B., Zeng M. Recent advances in the anti-aging effects of natural polysaccharides: sources, structural characterization, action mechanisms and structure-activity relationships. Trends Food Sci. Technol. 2025;160 doi: 10.1016/j.tifs.2025.105000. [DOI] [Google Scholar]
- Thumu S.C.R., Halami P.M. In vivo safety assessment of Lactobacillus fermentum strains, evaluation of their cholesterol-lowering ability and intestinal microbial modulation. J. Sci. Food Agric. 2020;100(2):705–713. doi: 10.1002/jsfa.10071. [DOI] [PubMed] [Google Scholar]
- Tsafrakidou P., Michaelidou A.M., C G.B. Fermented cereal-based products: nutritional aspects, possible impact on Gut Microbiota and health implications. Foods. 2020;9(6) doi: 10.3390/foods9060734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vaiserman A.M., Koliada A.K., Marotta F. Gut microbiota: a player in aging and a target for anti-aging intervention. Ageing Res. Rev. 2017;35:36–45. doi: 10.1016/j.arr.2017.01.001. [DOI] [PubMed] [Google Scholar]
- Varsha K.K., Narisetty V., Brar K.K., Madhavan A., Alphy M.P., Sindhu R., Awasthi M.K., Varjani S., Binod P. Bioactive metabolites in functional and fermented foods and their role as immunity booster and anti-viral innate mechanisms. J. Food Sci. Technol. 2022;60(9):1–10. doi: 10.1007/s13197-022-05528-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verediano T.A., Stampini Duarte Martino H., Dias Paes M.C., Tako E. Effects of anthocyanin on intestinal health: a systematic review. Nutrients. 2021;13(4) doi: 10.3390/nu13041331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J., Fang X., Wu T., Fang L., Liu C., Min W. In vitro immunomodulatory effects of acidic exopolysaccharide produced by Lactobacillus planetarium JLAU103 on RAW264.7 macrophages. Int. J. Biol. Macromol. 2020;156:1308–1315. doi: 10.1016/j.ijbiomac.2019.11.169. [DOI] [PubMed] [Google Scholar]
- Wang X., Hu K., Chen Y., Lai J., Zhang M., Li J., Li Q., Zhao N., Liu S. Effect of Lactiplantibacillus plantarum fermentation on the physicochemical, antioxidant activity and immunomodulatory ability of polysaccharides from Lvjian okra. Int. J. Biol. Macromol. 2024;257(Pt 1) doi: 10.1016/j.ijbiomac.2023.128649. [DOI] [PubMed] [Google Scholar]
- Wang Y., Cui X., Lin Q., Cai J., Tang L., Liang Y. Active peptide KF-8 from rice Bran attenuates oxidative stress in a mouse model of aging induced by d-Galactose. J. Agric. Food Chem. 2020;68(44):12271–12283. doi: 10.1021/acs.jafc.0c04358. [DOI] [PubMed] [Google Scholar]
- Wang Z., Chen Z., Jiang Z., Luo P., Liu L., Huang Y., Wang H., Wang Y., Long L., Tan X., Liu D., Jin T., Wang Y., Wang Y., Liao F., Zhang C., Chen L., Gan Y., Liu Y.…Shi C. Cordycepin prevents radiation ulcer by inhibiting cell senescence via NRF2 and AMPK in rodents. Nat. Commun. 2019;10(1):2538. doi: 10.1038/s41467-019-10386-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z., Sun Q., Fang J., Wang C., Wang D., Li M. The anti-aging activity of Lycium barbarum polysaccharide extracted by yeast fermentation: in vivo and in vitro studies. Int. J. Biol. Macromol. 2022;209:2032–2041. doi: 10.1016/j.ijbiomac.2022.04.184. [DOI] [PubMed] [Google Scholar]
- Wastyk H.C., Fragiadakis G.K., Perelman D., Dahan D., Merrill B.D., Yu F.B., Topf M., Gonzalez C.G., Van Treuren W., Han S., Robinson J.L., Elias J.E., Sonnenburg E.D., Gardner C.D., Sonnenburg J.L. Gut-microbiota-targeted diets modulate human immune status. Cell. 2021;184(16):4137–4153.e4114. doi: 10.1016/j.cell.2021.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia C., Cao X., Cui L., Liu H., Wang S., Chen T.J. Anti-aging effect of the combination of Bifidobacterium longum and B. animalis in a d-galactose-treated mice. J. Funct.Foods. 2020;69 doi: 10.1016/j.jff.2020.103938. [DOI] [Google Scholar]
- Xia T., Yao J., Zhang J., Duan W., Zhang B., Xie X., Xia M., Song J., Zheng Y., Wang M. Evaluation of nutritional compositions, bioactive compounds, and antioxidant activities of Shanxi aged vinegars during the aging process. J. Food Sci. Technol. 2018;83(10):2638–2644. doi: 10.1111/1750-3841.14356. [DOI] [PubMed] [Google Scholar]
- Xu H.Y., Li Q.C., Zhou W.J., Zhang H.B., Chen Z.X., Peng N., Gong S.Y., Liu B., Zeng F. Anti-oxidative and anti-aging effects of probiotic fermented ginseng by modulating Gut Microbiota and metabolites in Caenorhabditis elegans. Plant Foods Hum. Nutr. 2023;78(2):320–328. doi: 10.1007/s11130-023-01055-9. [DOI] [PubMed] [Google Scholar]
- Xu Z., Zhang K., Wang Q., Zheng Y. MicroRNA-124 improves functional recovery and suppresses Bax-dependent apoptosis in rats following spinal cord injury. Mol. Med. Rep. 2019;19(4):2551–2560. doi: 10.3892/mmr.2019.9904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang J., Sun H., Xu K., Zhang X., Huang M., Jin G., Liu Y., Chen W., Lin S., Shen J., Zhong C.Q., Xu Y., Zhang Q., Liu W., Yang Y., Ou J. Aging-rewired metabolic cues promote autophagy and senescence via DRAM1. Autophagy. 2025;21(12):3142–3164. doi: 10.1080/15548627.2025.2568487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin Q., Qin F., Gan F., Zhao G., Chen R., Wen Y., Hua X., Zeng F., Zhang Y., Xiao Y., Xie W., Tao Y. Colonic aging and colorectal cancer: an unignorable interplay and its translational implications. Biology. 2025;14(7):805. doi: 10.3390/biology14070805. https://www.mdpi.com/2079-7737/14/7/805 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshinaga M., Toda N., Tamura Y., Terakado S., Ueno M., Otsuka K., Numabe A., Kawabata Y., Uehara Y. Japanese traditional miso soup attenuates salt-induced hypertension and its organ damage in Dahl salt-sensitive rats. Nutrition. 2012;28(9):924–931. doi: 10.1016/j.nut.2011.09.010. [DOI] [PubMed] [Google Scholar]
- You X., Li Z., Ma K., Zhang C., Chen X., Wang G., Yang L., Dong M., Rui X., Zhang Q., Li W. Structural characterization and immunomodulatory activity of an exopolysaccharide produced by Lactobacillus helveticus LZ-R-5. Carbohydr. Polym. 2020;235 doi: 10.1016/j.carbpol.2020.115977. [DOI] [PubMed] [Google Scholar]
- You X., Yang L., Zhao X., Ma K., Chen X., Zhang C., Wang G., Dong M., Rui X., Zhang Q., Li W. Isolation, purification, characterization and immunostimulatory activity of an exopolysaccharide produced by Lactobacillus pentosus LZ-R-17 isolated from Tibetan kefir. Int. J. Biol. Macromol. 2020;158:408–419. doi: 10.1016/j.ijbiomac.2020.05.027. [DOI] [PubMed] [Google Scholar]
- Yuan X., Wang T., Sun L., Qiao Z., Pan H., Zhong Y., Zhuang Y. Recent advances of fermented fruits: a review on strains, fermentation strategies, and functional activities. Food Chem. X. 2024;22 doi: 10.1016/j.fochx.2024.101482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan Y., Yang Y., Xiao L., Qu L., Zhang X., Wei Y. Advancing insights into probiotics during vegetable fermentation. Foods. 2023;12(20) doi: 10.3390/foods12203789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yun Y.-R., Choi Y.-J., Kim Y.-S., Chon S.-Y., Lee M.-A., Chung Y.B., Park S.-H., Min S.-G., Yang H.-C., Seo H.-Y. Antioxidant and anti-inflammatory effects of solar salt brined kimchi. Food Sci. Biotechnol. 2023;32(5):679–687. doi: 10.1007/s10068-022-01203-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yun Y.R., Park B.Y., Kim S.H., Jung J.H. Antioxidant, anti-obesity, and anti-aging activities of Jeju citrus blended vinegar. Foods. 2021;10(7) doi: 10.3390/foods10071441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J., Zhao X., Jiang Y., Zhao W., Guo T., Cao Y., Teng J., Hao X., Zhao J., Yang Z. Antioxidant status and gut microbiota change in an aging mouse model as influenced by exopolysaccharide produced by Lactobacillus plantarum YW11 isolated from Tibetan kefir. J. Dairy Sci. 2017;100(8):6025–6041. doi: 10.3168/jds.2016-12480. [DOI] [PubMed] [Google Scholar]
- Zhang K., Zhang T.T., Guo R.R., Ye Q., Zhao H.L., Huang X.H. The regulation of key flavor of traditional fermented food by microbial metabolism: a review. Food Chem. X. 2023;19 doi: 10.1016/j.fochx.2023.100871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang X., Liu L., Luo J., Peng X. Anti-aging potency correlates with metabolites from in vitro fermentation of edible fungal polysaccharides using human fecal intestinal microflora. Food Funct. 2022;13(22):11592–11603. doi: 10.1039/d2fo01951e. [DOI] [PubMed] [Google Scholar]
- Zhao J., Yu J., Zhi Q., Yuan T., Lei X., Zeng K., Ming J. Anti-aging effects of the fermented anthocyanin extracts of purple sweet potato on Caenorhabditis elegans. Food Funct. 2021;12(24):12647–12658. doi: 10.1039/d1fo02671b. [DOI] [PubMed] [Google Scholar]
- Zhao P., Wang J., Zhao W., Ma X., Sun H. Antifatigue and antiaging effects of Chinese rice wine in mice. Food Sci. Nutr. 2018;6(8):2386–2394. doi: 10.1002/fsn3.830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao T., Li X., Zhu R., Ma Z., Liu L., Wang X., Hu X. Effect of natural fermentation on the structure and physicochemical properties of wheat starch. Carbohydr. Polym. 2019;218:163–169. doi: 10.1016/j.carbpol.2019.04.061. [DOI] [PubMed] [Google Scholar]
- Zhao X., Yi R., Zhou X., Mu J., Long X., Pan Y., Song J.L., Park K.Y. Preventive effect of Lactobacillus plantarum KSFY02 isolated from naturally fermented yogurt from Xinjiang, China, on d-galactose-induced oxidative aging in mice. J. Dairy Sci. 2019;102(7):5899–5912. doi: 10.3168/jds.2018-16033. [DOI] [PubMed] [Google Scholar]
- Zhao Y., Liao A.-M., Liu N., Huang J.-H., Lv X., Yang C.-R., Chen W.-J., Hou Y.-C., Ma L.-J., Hui M.J.F.B. Potential anti-aging effects of fermented wheat germ in aging mice. Food Biosci. 2021;42 doi: 10.1016/j.fbio.2021.101182. [DOI] [Google Scholar]
- Zheng H.L., Li M.T., Zhou T., Wang Y.Y., Shang E.X., Hua Y.Q., Duan J.A., Zhu Y. Protective effects of Lycium barbarum L. berry extracts against oxidative stress-induced damage of the retina of aging mouse and ARPE-19 cells. Food Funct. 2023;14(1):399–412. doi: 10.1039/d2fo02788g. [DOI] [PubMed] [Google Scholar]
- Zieliński H., Szawara-Nowak D., Wronkowska M. Bioaccessibility of anti-AGEs activity, antioxidant capacity and phenolics from water biscuits prepared from fermented buckwheat flours. LWT--Food Sci. Technol. 2020;123 doi: 10.1016/j.lwt.2020.109051. [DOI] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing not applicable to this article as no datasets were generated or analysed during the current study.






