Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Nutr., 15 December 2025

Sec. Nutrition, Psychology and Brain Health

Volume 12 - 2025 | https://doi.org/10.3389/fnut.2025.1677197

Nutritional strategies against dementia in rural populations

  • 1Departments of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
  • 2Department of Medicine, Sharif Medical and Dental College, Lahore, Pakistan

Dementia is a progressive neurodegenerative disorder that represents a growing challenge to global health, especially in aging populations. The burden of dementia is high in rural communities, where access to healthcare services, nutritional resources, and educational opportunities is significantly limited. A critical knowledge gap exists regarding how rural-specific nutritional disparities mechanistically interact with gut–brain axis dysfunction to influence dementia risk. The gut-brain axis mediates neuroimmune communication, metabolic regulation, and microbial signaling, and nutritional insufficiency is associated with reduced microbial diversity, impaired short-chain fatty acid (SCFAs) production, increased intestinal permeability, and heightened systemic inflammatory activity, all of which contribute to neurodegenerative processes. This review delineates the mechanistic pathways linking malnutrition-induced gut dysregulation to neuronal injury and cognitive decline, with a specific focus on rural populations. We further evaluate the biological actions of key nutraceutical classes, including phytochemicals (curcumin, resveratrol, sulforaphane, ginkgo biloba), vitamins (B12, C, E, and D), and metabolic/dietary interventions (omega-3 fatty acids, ketogenic diet, Mediterranean diet, trehalose). By consolidating preclinical and clinical evidence, we identify the molecular targets through which these nutraceuticals modulate oxidative stress, inflammatory signaling networks, blood-brain barrier (BBB) integrity, and microRNA regulation. In summary, our findings suggest that nutraceutical-based approaches targeting gut-brain axis dysfunction may provide a feasible and biologically informed strategy for mitigating dementia disparities in rural settings.

1 Introduction

Dementia is a major health problem worldwide, involving progressive cognitive decline that interferes with independent daily living (1). Common subtypes include vascular dementia, frontotemporal dementia, Lewy body dementia, mixed dementia, and Alzheimer's disease (AD) (1, 2). The lifetime risk for dementia in individuals over 55 years old in the U.S. is 42%, with elevated risk in females, black adults, and APOE4 gene carriers (3). Studies have shown that rural residents are disproportionately underdiagnosed, diagnosed at advanced stages, and experience a disproportionate mortality burden from dementia-related causes (46). Even without a diagnosis of dementia, older adults in rural populations exhibit reduced cognitive performance along a rural-urban gradient, a disparity that is mechanistically linked to structural socioeconomic determinants such as attenuated educational attainment, a higher burden of chronic health conditions, and persistent socioeconomic stressors, including food and resource insecurity (710).

Epidemiological studies show that rural populations experience a disproportionate prevalence of cognitive impairment and dementia compared with urban areas. In the United States, older adults living in rural settings exhibit accelerated cognitive decline across the rural-urban gradient, in part due to attenuated educational attainment, higher rates of chronic illness, reduced healthcare access, and limited availability of nutritious foods (4, 5). Rural communities also demonstrate elevated levels of food insecurity and poorer dietary quality, both of which contribute to systemic inflammation and metabolic dysfunction (11).

In addition to traditional risk factors (hypertension and obesity), rural living is associated with increased nutritional inequities (810, 12, 13). Furthermore, nutritional disparity in the rural population is further exacerbated by geographic and economic barriers to accessing healthy foods (13, 14). The limited access to grocery stores, high food prices, and the dominance of ultra-processed foods (UPF) collectively reinforce a structural nutritional disadvantage (13). Roughly 2.3 million rural Americans live in “food deserts,” characterized by limited access to fresh fruit and vegetables (11, 15, 16). These environmental and economic constraints contribute to an inequitable risk profile for dementia in these communities.

Given the importance of identifying effective interventions for populations experiencing disproportionate dementia burden, the incorporation of nutraceuticals offers a promising pathway. The central objective of this review article is to provide a comprehensive overview of the current understanding of dementia in rural populations, its underlying pathophysiological mechanisms, and the potential therapeutic benefits of nutraceuticals. Briefly, the first section outlines how rural structural conditions create contextual disparities that increase vulnerability to cognitive decline. The following section examines the gut–brain axis to illustrate how nutritional patterns influence microbial metabolites and immune signaling. Subsequent sections discuss emerging evidence on nutraceuticals, with a detailed description of four potent phytochemicals, vitamins, and dietary/metabolic interventions with documented neuroprotective properties, including findings from human clinical trials.

This review extends beyond previous studies by presenting a unified view of how rural environmental and socioeconomic conditions intersect with dietary patterns and cognitive vulnerability. Unlike prior studies that consider diet or microbiota separately, these determinants are evaluated within the structural context of rural health, marked by food insecurity, limited healthcare access, and environmental exposures, to clarify their contribution to disparities in cognitive outcomes. This perspective underscores the need for approaches that consider both the social context and the biological pathways involved in cognitive decline.

It is important to clarify how rural environmental and nutritional disadvantages translate into measurable biological disruption. Nutritional inadequacies characteristic of rural settings, such as low fiber intake, micronutrient deficiencies, and reliance on ultra-processed foods (UPF), alter gut microbial composition, impair intestinal barrier integrity, and amplify systemic inflammatory signaling. These diet-induced perturbations constitute the mechanistic link between structural socioeconomic disadvantages and neurodegenerative processes, establishing the rationale for examining rural health disparities and their molecular consequences.

2 Rural health disparities and dementia risk

Nutrient deficiencies in rural populations significantly contribute to dementia risk through interconnected biological and social mechanisms. Chronic malnutrition, geographic isolation, and limited access to fresh, nutritious food create a cycle that exacerbates cognitive vulnerability (17). According to the United States Department of Agriculture, rural food deserts (areas with inadequate access to grocery stores) affect over 2.3 million Americans, disproportionately impacting older adults and low-income families (15). Food cost and geographic proximity are the main factors influencing food choices in rural Appalachia (13, 15). Individuals in these areas frequently consume energy-dense but nutrient-poor diets, leading to higher caloric intake and increased risk of chronic disease and neurodegeneration (13). A cross-sectional study revealed increasing reliance on packaged food products, further depleting diet quality (18). These intersecting structural determinants, including rurality, income level, food access, and self-care capacity, collectively intensify nutritional vulnerability and contribute to a disproportionate dementia burden.

Food illiteracy and comorbid health conditions further compound these disparities. Between 2001 and 2018, the average intake of minimally processed foods declined significantly (19). Rural adults consuming low-nutrient-dense diets were found to be twice as likely to be obese and deficient in vitamin B12, and up to 17 times more likely to fall short in essential nutrient intake (20). A randomized controlled trial demonstrated that a 2-week high ultra-processed food (UPF) diet caused substantial increases in energy intake and weight gain (21). Malnutrition represents an additional factor contributing to cognitive vulnerability in rural populations. Insufficient intake of nutrients, including omega-3 fatty acids, B-vitamins, and antioxidant compounds, has been linked to impaired synaptic function, increased oxidative stress, and increased neuroinflammation (2224). Observational studies also show that older adults with nutrient deficiencies or poor diet quality have an elevated risk of cognitive decline and dementia (17, 25).

In the United States, significant rural-urban gradients in dementia burden have been shown, caused by differences in socioeconomic status, educational attainment, and dietary access. Older adults in rural areas show disproportionate prevalence of cognitive impairment and dementia compared with their urban counterparts, partly due to lower educational opportunities, higher poverty rates, and limited access to healthy foods and healthcare services (5, 26, 27).

Evidence suggests a strong correlation between diet quality and the risk of dementia (28). A prospective cohort study linked greater UPF intake with increased dementia incidence, while replacing UPFs with whole or minimally processed foods was associated with a 19% reduction in dementia risk (28). Chronic low-grade systemic inflammation, frequently amplified by the pro-inflammatory Standard American Diet prevalent in rural settings, is a central driver of neurodegenerative processes (29). This inflammatory state activates adipose and immune signaling pathways, which are critical components of the gut-brain axis. These effects are further exacerbated in rural communities, where heightened pollution exposure and lower antioxidant intake reflect environmental and nutritional inequities (30). Healthy Eating Index-2020 (HEI-2020) and the Dietary Inflammatory Index (DII) analyses revealed significant associations between elevated interleukin (IL)-6, IL-1β, lipopolysaccharide binding protein, Toll-like receptor 4 (TLR4), as well as zonula occludens-1 (ZO-1) and cognitive impairment, highlighting inflammation as a key modifiable target (31). Together, these biomarkers indicate a diet-linked inflammatory cascade in which impaired gut-barrier integrity facilitates microbial translocation, upregulates TLR4-mediated signaling, and intensifies peripheral immune activation. This inflammatory activity communicates with the brain, promoting microglial activation and reinforcing the neuroinflammatory processes associated with cognitive decline (31).

The absence of fruits, vegetables, whole grains, and lean proteins limits neuroprotective nutrients crucial for synaptic plasticity and energy metabolism (32). Polyphenols, omega-3 fatty acids, B-complex vitamins, and minerals support neurogenesis, membrane fluidity, and neurotransmission. However, high intake of palmitic acid and saturated fats disrupts the blood-brain barrier (BBB) and promotes β-amyloid accumulation (33). Deficiencies in vitamin B12 and omega-3s, especially docosahexaenoic acid (DHA), an essential component of synaptic membranes, contribute to oxidative stress, impaired signaling, and cognitive decline (34, 35).

Emerging evidence shows a significant global rise in early-onset dementia, emphasizing the impact of nutritional and metabolic factors throughout life. Global Burden of Disease data indicate that the number of individuals under 70 with dementia has been increasing, with continued growth projected through 2050 (36, 37). These findings highlight that nutritional interventions are relevant not only for older adults but also for preventing or slowing premature cognitive decline in younger populations, particularly in rural areas undergoing rapid dietary changes.

It has been shown that the gut-brain axis has a role in cognitive health (38, 39). Disruptions in this bidirectional system, including elevated inflammatory markers and compromised gut barrier integrity, are increasingly recognized as contributors to neurodegenerative processes. Elevated inflammatory markers and compromised gut integrity, reflected by increased intestinal permeability and dysbiosis, have been linked to cognitive impairment (31). The gut-brain connection mediates neuroinflammation through microbial metabolites and immune signaling, indicating that altered gut physiology represents a mechanistic pathway through which rural nutritional inequities contribute to cognitive decline (40). In this context, understanding how gut-brain signaling influences inflammatory and metabolic pathways clarifies why these mechanisms represent key targets for intervention in cognitive decline.

3 The gut-brain axis in cognitive decline

The gut microbiota plays a crucial role in maintaining systemic and neurological health (40). The gut-brain axis is a bidirectional communication network connecting the gut microbiome with the central nervous system through neural, immune, and endocrine pathways (4143). Notably, the vagus nerve detects microbial metabolites, such as short-chain fatty acids (SCFAs), and relays these signals to the brain (44). These microbial metabolites, including amino acids and neurotransmitter precursors, influence both gastrointestinal and brain function. Disruption of the gut microbiota, known as gut dysbiosis, can impair neurotransmitter synthesis, compromise the intestinal barrier, and promote neuroinflammation, contributing to neurodegeneration, leading to cognitive impairment (45). Dysbiosis alters microglial activation, elevates pro-inflammatory cytokines including IL-6, IL-1β, and tumor necrosis factor-alpha (TNF-α), and reduces the production of beneficial microbial metabolites such as SCFAs (46, 47). SCFAs such as acetate, butyrate, and propionate produced by bacterial fermentation of dietary fiber have potent anti-inflammatory effects. Additionally, SCFAs regulate microglial function, suppress Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation, and modulate both intestinal and blood-brain barrier integrity (4850). Experimental models show that SCFA supplementation in Alzheimer's mice improves memory and reduces Aβ plaque formation by upregulating IL-10 and downregulating IL-6 (51, 52).

Several studies demonstrate that dietary patterns modulate gut-brain signaling (5356). Diets rich in fiber, polyphenols, probiotics, and unsaturated fats increase beneficial SCFA-producing genera such as Faecalibacterium and Roseburia, resulting in elevated butyrate and propionate levels that suppress NF-κB activation, regulate microglial homeostasis, and reduce neuroinflammation (57, 58). However, high-fat or ultra-processed diets impair the microbiome, reducing SCFA production, increasing systemic pro-inflammatory cytokine (IL-6 and TNF-α), leading to gut permeability (through the downregulation of Occludin and E-cadherin), and promoting hippocampal neuroinflammation, which accelerates cognitive decline (59, 60). Food insecurity and nutritionally inadequate diets, which disproportionately affect rural communities, further compound these disruptions (61). These diet-induced alterations in gut microbial composition and metabolic activity can be quantitatively assessed through changes in microbial α- and β-diversity, circulating levels of SCFAs, and systemic inflammatory mediators. Together, these indicators provide a mechanistic link between nutritional patterns, microbiota-derived signaling, neuroinflammatory cascades, and the progression of neurodegenerative pathology (60, 62).

Emerging therapeutic approaches, such as probiotic supplementation, prebiotic fiber, and fecal microbiota transplantation (FMT), show promise in delaying cognitive decline (63, 64). In clinical trials, AD patients receiving multispecies probiotics exhibited improved cognition (65, 66). In addition, FMT in transgenic AD mouse models reduced inflammation via the TLR4-Myeloid differentiation factor 88 (MyD88)-NF-κB (TLR4/MyD88/NF-κB) pathway and increased SCFA production (67).

Environmental and social determinants, including pesticide exposure and access to healthcare, significantly impact gut microbiome health. Rural populations, particularly those in agricultural and resource-limited regions, experience disproportionate exposure to environmental toxins and have restricted access to healthcare and nutritious foods, making them especially susceptible to microbiome disruption (68, 69). These factors exacerbate systemic inflammation, reduce the availability of neuroprotective metabolites, and contribute to disease progression. Studies show that rural residents with dementia have shorter survival rates and a disproportionately higher disease burden compared with their urban counterparts (26, 70). However, there is no evidence directly comparing whether malnutrition or pesticide exposure has a greater impact on dementia risk. It has been demonstrated that both factors negatively impact gut-brain axis regulation by increasing oxidative stress, inflammation, and microbiome disruption. In rural areas, where these exposures frequently coexist, their combined effects may further contribute to contextual cognitive vulnerability (68, 69). Addressing microbiome health through targeted dietary strategies and nutraceutical interventions could offer a viable path to support cognitive resilience in these at-risk populations (Figure 1).

Figure 1
Flowchart illustrating the connection between malnutrition, gut microbiota dysfunction, and cognitive decline in rural populations. It shows microbial metabolites affecting the gut-brain axis, leading to immune signaling, neural pathways alteration, and endocrine changes. This results in increased blood-brain permeability causing microglial activation, neuroinflammation, neuronal damage, and cognitive decline. Nutritional interventions include phytochemicals, vitamins, and metabolic modifiers like omega-3 and diets to mitigate these effects. Arrows indicate process flow and direction of impact.

Figure 1. Schematic representation showing the protective effects of nutraceuticals in the dementia-associated gut-brain axis. Nutraceuticals effectively inhibit neuroinflammation and neurodegeneration through their remarkable pharmacological properties. Image proposed by the authors.

Taken together, this section shows a mechanistic pathway that has not been fully explored in previous studies. Rural environmental and socioeconomic determinants influence dietary quality and exposure patterns, altering gut microbial composition, metabolic signaling, and neuroinflammatory activation. Rural living conditions, therefore, represent a biologically relevant context in which disruptions to gut-brain communication arise, linking dietary insufficiency, microbiota imbalance, and immune activation to increased susceptibility to neurodegenerative processes. These mechanistic connections underscore the need for interventions capable of modulating inflammatory, metabolic, and microbiota-derived pathways in populations experiencing disproportionate dementia burden.

4 Nutraceuticals: mechanisms, and neuroprotective potential

Nutraceuticals, derived from foods and medicinal plants, have emerged as promising adjunctive strategies for the prevention and management of dementia (71, 72). Their therapeutic potential stems from the ability to modulate key molecular pathways implicated in neurodegeneration, including the suppression of pro-inflammatory cascades (73, 74). In addition, nutraceuticals influence synaptic plasticity and neuronal survival, which is crucial for memory and learning. Additionally, evidence highlights their role in modulating the expression of specific microRNAs (miRs), small non-coding RNAs that regulate gene networks involved in neuroinflammation (7577). This multimodal activity positions nutraceuticals as attractive, low-cost interventions capable of improving cognitive decline, especially in vulnerable rural populations where conventional therapeutic options may be limited.

4.1 Phytochemicals

Phytochemicals are plant-derived compounds that can target neuroinflammation by reducing systemic inflammation, preserving the BBB, and protecting against gastrointestinal signaling to the brain (70, 78). It can interact with various signaling pathways to inhibit inflammation caused by Aβ peptide accumulation. By inhibiting pro-inflammatory gene expression, polyphenols can decrease neurodegeneration (79, 80). This section aims to analyze how the following phytochemicals- Curcumin, Resveratrol, Sulforaphane, and Ginkgo Biloba can modulate different signaling pathways and miRs to improve memory and cognitive functions. Table 1 summarizes these mechanisms.

Table 1
www.frontiersin.org

Table 1. Effect of phytochemicals on the pathophysiological complications associated with cognitive decline/dementia.

4.1.1 Curcumin

Curcumin is a polyphenol compound extracted from the herb Curcuma Longa Linn (turmeric) (81). Curcumin is the active component in turmeric, which is a spice used in many Middle Eastern and South Asian diets in the Indian subcontinent (82). It is often consumed as an oral capsule, powder form, and it can be effective against a wide variety of chronic inflammatory conditions, such as dementia (83). Despite its therapeutic benefits, a major limitation of curcumin supplementation is its low systemic bioavailability due to rapid metabolism (84). In the context of neurodegeneration, curcumin protects against dementia by scavenging reactive oxygen species (ROS), suppressing pro-inflammatory cytokines, and modulating signaling pathways (85). It improves cognitive function, spatial memory, and decreases AD-associated pathology, such as tubulin associated unit (tau) phosphorylation and Aβ plaques (86).

Through multiple studies, curcumin has exhibited an array of activity against dementia progression (8790). An in vivo study showed that curcumin decreased amyloid levels, plaque formation, and blocked the aggregation of fibrils in the tg2576 AD model (87). Furthermore, Tetrahydrocurcumin, a metabolite of curcumin, has a protective effect against amyloid-induced toxicity, oxidative stress, and mitochondrial dysfunction in primary hippocampal culture (88). Aside from general systemic anti-inflammatory and antioxidant activity, curcumin showed an improvement in spatial memory deficits by regulating the Brain-Derived Neurotrophic Factor (BDNF)-Extracellular Signal-Regulated Kinase (ERK) (BDNF-ERK) signaling pathway (89). Curcumin is a lipophilic compound and can cross the BBB to induce neuroprotection by increasing cyclic adenosine monophosphate (cAMP) and BDNF levels (90). Furthermore, an in vivo study has demonstrated that chronic curcumin infusions were associated with an increase in hippocampal BDNF expression and phosphorylated ERK levels, which was associated with enhanced memory performance. In AD models of amyloid precursor protein (APP) transgenic mice, it was found that curcumin improved memory deficits by modulating product-specific receptor (RAGE)-TLR4-NF-κB (RAGE/TLR4-NF-κB) inflammatory signaling pathway (91). In addition, curcumin reduced Beta-site APP Cleaving Enzyme 1 (BACE1) expression, a key protease in Aβ generation in dementia, through the estrogen receptor β (ERβ) and NF-κB signaling pathway in SH-SY5Y cells (92). Additionally, curcumin also demonstrated neuroprotective capacity in Aβ-Exposed THP-1 Cells by inhibiting the Mitogen-Activated Protein Kinases (MAPKs) and NF-κB signaling pathways (93). It was also seen in brain microglia cells, curcumin markedly suppressed Janus kinase (JAK)-signal transducer and activator of transcription (STAT) inflammatory signaling through activation of Src homology 2 domain-containing tyrosine phosphatase 2 (94). Due to its poor bioavailability and water solubility, many formulations have been constructed to enhance the clinical effects of curcumin (95). One formulation of curcumin and demethoxycurcumin, bisdemethoxycurcumin, improved the innate immunity and transcription of Mannoside Acetylglucosaminyltransferase 3 (MGAT3), a gene impaired in phagocytosis of Aβ plaques and Toll-like receptors in AD patients (96). In a randomized, double-blind, and placebo-controlled clinical trial, it was shown that Biocurmuax significantly improved cognitive function in older adults (97).

It has been shown that curcumin can also modulate miRs expression and activity, which mitigates the progression of neurodegenerative diseases (98). Curcumin inhibited miR-146a expression, an interleukin-1 receptor-associated kinase (IRAK)-1, IRAK-2, and NF-kB signaling pathways regulator in astroglia cells (99). Furthermore, it was found that curcumin decreased inflammatory expression of IL-1β, nitric oxide synthases (NOS), and complement factor H in the temporal lobe in AD by regulating miR-146a levels (100, 101). Studies have shown that miR-miR-26a-5p, miR-203a-3p, and miR-155-5p play a pivotal role in neuroinflammation and apoptosis through modulation of triggering receptor expressed on myeloid cells 1 (TREM-1)-DAP12-NLR family pyrin domain containing 3 (NLP3)-Caspase1-IL1B (TREM- DAP12/NLRP3/CASP1/IL1B-TLR4/NF-κB) signaling pathways, and Tau hyperphosphorylation were downregulated after curcumin administration in microglia cells (102). In addition, a nanoparticle system that co-delivers curcumin and miR-124 decreased the β-secretase, leading to inhibition of Aβ production and associated neurotoxicity (103).

4.1.2 Resveratrol

Resveratrol (RSV) is a polyphenol produced by plants in response to external stressors (104). It is available in plants, fruits, grape skin, red wine, peanuts, and berries (81). Despite its therapeutic properties, it has poor bioavailability due to rapid hepatic metabolism and poor water solubility (105). To overcome these limitations, RSV can be incorporated into nanoparticles, liposomes, and polymeric particles to improve the available concentration (106). RSV has been shown to exhibit a wide range of clinically relevant properties, including antioxidant, anti-inflammatory, and anti-proliferative effects (107109). These characteristics suggest that RSV is a potential candidate to decrease the onset of neurodegeneration and improve cognitive dysfunction.

Multiple studies show the effects of RSV on improving the progression of dementia through the modulation of different signaling pathways (110, 111). An in vivo study showed that resveratrol protected rats from Aβ-induced neurotoxicity by downregulating inducible nitric oxide synthase (iNOS), heme oxygenase-1 (HO-1) expression, as well as decreasing the levels of lipid peroxidation, leading to improvements in spatial memory (112). RSV also demonstrated a neuroprotective effect against β-amyloid-induced oxidative damage and memory-associated proteins in H19-7 hippocampal neuronal cells via modulation of antioxidant enzyme levels, such as superoxide dismutase, catalase, and glutathione reductase (113). Furthermore, in vivo and in vitro studies have also shown that RSV decreased the levels of glial fibrillary acidic protein, molecules related to inflammation and apoptosis, including TNF-α, IL-1β, NLRP3, cleaved caspase-3, and B-cell lymphoma 2 (Bcl-2), apoptosis regulator (Bax), as well as increased the levels of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IkBα) and Bcl-2, attenuating the cognitive impairment (110, 114, 115). Additionally, RSV mitigated lipopolysaccharide (LPS)- and amyloid-beta (Aβ)-mediated microglial inflammation by inhibiting the TLR4/NF-κB/STAT signaling cascade (116). In an in vitro model, using human neural stem cells, RSV showed a neuroprotective effect against Aβ-induced inflammation and oxidative stress via activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK)-dependent pathways (117, 118). In addition, RSV suppressed β-amyloid-induced neurotoxicity and neuronal apoptosis via modulation of Sirtuin-1 (SIRT1)-Rho-associated coiled-coil containing protein kinase 1(ROCK1) signaling pathway in PC12 cells (119). Furthermore, RSV showed a neuroprotective effect in SH-SY5Y and HEK293 cells overexpressing APP, increasing SIRT1 activity and decreasing levels of CD147- a protein that promotes Aβ formation (109). In a randomized, double-blind, and placebo-controlled clinical trial, it was shown that RSV significantly improved cognitive function by significantly decreasing the cerebrospinal fluid Aβ40 biomarker, indicating that there are possible effects on anti-Aβ aggregation (120). Specifically, clinical trials of RSV have shown efficacy in improving cognitive impairments through modulation of pro-inflammatory cytokines and molecules related to neurodegeneration, including IL-10, IL-4, IL-12P40, IL-12P70, Fibroblast Growth Factor-2 (FGF2), regulated on activation, normal T-cell expressed and secreted (RANTES), and metalloproteinase (MMP)- 9 (110, 111). Alongside altering the levels of pro-inflammatory cytokines, RSV treatment also increased levels of serum SIRT1, improving the cognitive impairments in AD patients (121).

It has been shown that RSV can also modulate miRs expression (122). In an in vitro model, using human neural cells in primary culture, RSV showed a neuroprotective effect by upregulating miR-146a, which has an anti-inflammatory response in the pathogenesis of AD by decreasing levels of IL-1β and Aβ42 (123). RSV also improved learning and memory in normally aged mice, suppressing the miR-134- and miR124-cAMP response element-binding protein (CREB)-BDNF pathway (124). Additionally, RSV increased the expression of miR-2, which enhanced brain insulin signaling and resulted in decreased memory impairment (124). Similarly, RSV attenuated diabetic adipose tissue-derived extracellular vesicles-induced hippocampal Ferroptosis and cognitive dysfunction via inhibiting miR-9-3p/solute carrier family 7-member 11 (SLC7A11) axis (125).

4.1.3 Sulforaphane

Another nutritional additive is Sulforaphane (SFN), which is obtained from cruciferous vegetables, such as broccoli, cauliflower, and brussels sprouts (126). It is converted from the precursor glucoraphanin into the isothiocyanate SFN by the enzyme myrosinase (127). The bioavailability is dependent on the form of supplementation and whether it was co-delivered with the myrionase enzyme (128). SFN is permeable to the blood-brain barrier, decreasing neuroinflammation and neurotoxicity (129). SFN can modulate inflammation, oxidative stress, and apoptosis to improve the outcomes of dementia (130).

SFN has exhibited a unique potential role in managing dementia development by modulating different signaling pathways (131134). In an LPS-activated microglia model in vitro, SFN has been shown to reduce the levels of inflammatory mediators [iNOS, cyclooxygenase-2 (COX-2), NO, and prostaglandin E2 (PGE2)] and proinflammatory cytokines (TNF-α, IL-6, and IL-1β), through c-Jun N-terminal kinase (JNK)/activator protein-1 (AP-1)/NF-κB inhibition and nuclear factor erythroid 2-related factor 2 (Nrf2)/HO-1 activation (131). SFN has also demonstrated beneficial effects in modulating anti-inflammatory cytokines, IL-4 and IL-10, in a methylglyoxal-induced SH-SY5Y cytotoxicity model (132). In addition, SFN protected immature hippocampal neurons against death caused by exposure to hemin or oxygen and glucose deprivation through modulation of Nrf2-dependent cytoprotective genes, including NAD(P)H quinone oxidoreductase 1 (NQO1), HO-1, and glutamate-cysteine ligase modifier subunit (GCLM), which is involved in glutathione biosynthesis (133). Furthermore, a study using primary mouse brain microvascular endothelial cells has also shown that SFN protects the brain vascular system and effectively reduces ischemic cognitive deficits by increasing the adenosine triphosphate (ATP) production, expression, and activity of glucose transporter-1, and glycolysis, as well as anti-oxidative stress responses and redox signaling (135). Similarly, SFN showed a neuroprotective effect in a vascular-induced cognitively impaired model in rats through the upregulation of the Nrf2 signaling pathway, improving neuronal damage and cognition (134). Moreover, SFN was able to increase the expression of glutamate-cysteine ligase, a key enzyme in neutralizing ROS (136). Additionally, SFN improved brain metabolic stress, neuroinflammation, and cognitive impairment in rats, increasing the levels of glutathione (137). Furthermore, an in vivo study using AD PS1V97L transgenic mice model has demonstrated that SFN inhibited the production of amyloid-β oligomer and improved spatial learning and memory (138). In addition, a computational study showed that SFN is a potential candidate with potent and selective BACE1 inhibitory properties that play an important role in AD prevention (139).

Studies have shown that SFN modulates neuroinflammation by regulating a variety of miRs (140143). In an in vitro model, using microglia, SFN showed a neuroprotective effect by inhibiting the NLRP3 inflammasome by suppressing the inflammatory miR-155 and increasing the anti-inflammatory miR-223, leading to Nrf2 signaling pathway modulation (140). It has been shown that SFN also modulates miR-17-5p, miR-16-5p, and miR-26b-5p, key miRs in neuroinflammation and the apoptosis process (141). It has been shown that in microglia of AD patients, miR-17-5p is upregulated, suppressing amyloid-β degradation (142). Similarly, miR-16-5p is upregulated in the brain tissue of 5xFAD mice, inducing neuronal cell apoptosis through direct targeting and suppression of Bcl-2 (143). In addition, miR-26b-5p expresses signals that increase Aβ levels and tau hyperphosphorylation, contributing to neurodegeneration (144).

4.1.4 Ginkgo biloba

Ginkgo biloba, also known as the maidenhair tree, has leaves, seeds, and an exocarp that played an important role in traditional Chinese herbal medicine. Ingredients in the leaves include flavonoids, terpenoids, and phenolic acids (145). EGb761 is a derivative of the phytochemical Ginkgo Biloba extract (GbE) (146). It is often consumed in various forms, including tablets, guttate pills, and in teas (147). The bioavailability of the extract depends on the form that is consumed, with tablets having the highest concentrations (148). The extract can have neuroprotective effects in neurodegenerative diseases, such as dementia (149, 150). EGb 761 extract can improve hippocampal neurogenesis, decrease Aβ aggregation, and enhance neuronal plasticity (151). Overall, ginkgo biloba is anti-inflammatory, anti-oxidative, and has a stimulatory effect on neurotransmitters (152).

Ginkgo Biloba has therapeutic effects in decreasing dementia by modulating various signaling pathways involved in oxidative stress and inflammation, leading to decreased tau phosphorylation associated with Aβ deposits (149, 153). An in vivo study using AD Tg2576 mice has demonstrated that long-term EGb761 supplementation decreased APP levels, a key protein in the AD pathogenesis (154). Furthermore, EGb761 supplementation improved dopaminergic transmission in the prefrontal cortex, improving cognitive function (155). It has been shown that mice supplemented with Ginkgo Biloba increased the superoxide dismutase and catalase levels in the hippocampus, striatum, and substantia nigra, improving cognitive decline in rats (156). Additionally, EGb761 improved cognitive function and regulated inflammatory responses in the APP/presenilin-1(PS1) mouse through downregulating pro-inflammatory cytokines and iNOS and upregulating anti-inflammatory cytokines and Arginase-1 (Arg-1) (157). In a randomized, double-blind, and placebo-controlled clinical trial, it was shown that Ginkgo biloba significantly improved cognitive function by modulating the cholinergic synaptic transmission by impacting the activity of acetylcholine receptors (158). However, studies have shown no significant reduction in the incidence or progression of dementia among older adults receiving EGb761 supplementation (159). Similarly, a meta-analysis reported mixed results, with benefits varying by dose, duration, and study design (160). These discrepancies highlight the need for standardized protocols and long-term randomized trials to clarify the true efficacy of Ginkgo biloba in the prevention of dementia.

There is limited research on the impact Ginkgo Biloba has on miR levels in the treatment of dementia. However, the treatment with EGb761 improved cognitive dysfunction and was associated with a decrease in levels of miR-1-3p/miR-206-3p in scopolamine-induced memory deficits (161).

4.2 Vitamins

Vitamins are essential macronutrients that are sorted into the fat-soluble (A, D, E, K) and the water-soluble (B-complex and C), which are all pertinent for the various roles they play in human physiology. It contributes to the understanding of how individuals can utilize dietary and vitamin-based supplements to help with recovery and overall wellbeing (162). This section aims to analyze how the following vitamins- Vitamin B12, Vitamin D, Vitamin C, and Vitamin E can modulate different signaling pathways and miRs to improve memory and cognitive functions. Table 2 summarizes these mechanisms.

Table 2
www.frontiersin.org

Table 2. Effect of vitamins on the pathophysiological complications associated with cognitive decline/dementia.

4.2.1 B12—Cobalamin

Vitamin B12, or cobalamin, is classified as a water-soluble vitamin (163). It is primarily sourced from animal-based foods such as poultry, red meats, fish, and dairy products (164). Vitamin B12 absorption is a complex process that has various factors influencing its bioavailability and storage (165). The active circulating forms of vitamin B12 in the body include methylcobalamin and adenosylcobalamin, while cyanocobalamin is a common supplement form (165, 166). Vitamin B12 plays a critical role in the development and aging of the brain through DNA synthesis, fatty acid metabolism, and the maintenance of the nervous system (167, 168). Suboptimal baseline serum vitamin B12 levels and a vitamin B12 deficiency have both been associated with a faster rate of cognitive decline and increased risk of dementia in AD patients (169).

It has been shown that B12 hypovitaminosis is linked to cognitive dysfunction and Alzheimer's disease gray matter atrophy (170). It has been shown that Vitamin B12 increases glial migration and synapse formation through modulation of leukocyte-common antigen-related receptor-type tyrosine-protein phosphatase (PTP-3/LAR PRTP) expression, improving neurological damage, maintaining the sensory neuron function (167). Additionally, vitamin B12 also demonstrated neuroprotective effects in hydrogen peroxide-induced apoptosis in SH-SY5Y cells by up-regulation of poly pyrimidine tract binding protein 1 (PTBP1) (171). Furthermore, an in vivo study has demonstrated that vitamin B12 deficiency leads to persistent brain defects by reducing transcription factor Stat3 signaling pathway, a key pathway for neuron survival (172). In addition, vitamin B12 shortages also affected upstream kinases of the Stat3 signaling pathway (phospho-Erk1/2, phospho-Src, phospho-JNK, and phospho-p38 as well as downstream target gene products (Bcl-2 and Bcl-xL), thus promoting apoptosis (172). Furthermore, in a scopolamine-induced AD model, it was found that vitamin B12 decreased hippocampal inflammation [Postsynaptic density protein 95 (PSD-95) and neurexin 1 and neuroligin] and apoptosis markers (COX-2 and activated caspase-3) and preserved pre- and post-synaptic proteins and possibly synaptic integrity in the hippocampus (173). Clinical trials for vitamin B12 are still in progress; however, it has been shown that low vitamin B12 levels are linked to cognitive impairment and dementia (174). Similarly, in another study, low vitamin B12 levels were associated with cognitive decline and AD development (175). Furthermore, lower vitamin B12 levels were associated with cognitive impairment by hypermethylation of redox-related genes NUDT15 and TXNRD1 (176). However, it has been shown that vitamin B supplementation has no significant improvements in memory or executive function among older adults (177).

Notable miRs are correlated with vitamin B12 and their impact on neurodegeneration, cognitive decline, and AD development (178). Studies have shown that B12 and other B vitamins increased the expression of different miRs, including miR-34a-5p, miR-128-3p, miR-181a-5p, and miR-204-5p, miRs relating to NFKB1, ATF3, and NR3CI expression, key genes associated with cognitive function (178). Additionally, in response to vitamin B12 deprivation, miR-124 increased, leading to the suppression of Stat3 signaling, which is associated with hippocampal neuron differentiation (172).

4.2.2 Vitamins C

Vitamin C, or ascorbic acid, is water-soluble and crucial for human physiology (179). Vitamin C is obtained rapidly through diet through foods such as citrus fruits and cruciferous vegetables (180). Its homeostasis is regulated by intestinal absorption, transport to tissues, renal uptake, and urine excretion involving the Sodium-Dependent Vitamin C Transporters (181). Remarkably, vitamin C accumulates in the brain at millimolar intracellular concentrations and higher levels in cerebrospinal fluid compared to plasma (182). These elevated brain concentrations underscore their significance in antioxidant defense, neuromodulation across glutamatergic, dopaminergic, cholinergic, and gamma-aminobutyric acid (GABA)ergic systems, and protection against oxidative neuronal injury (183). Importantly, vitamin C deficiency correlates with increased risk of cognitive decline and dementia, while normalizing levels has shown potential cognitive and neuroprotective benefits (184).

Multiple studies have shown that vitamin C has exhibited an array of activity against cognitive decline and dementia progression (185188). It has been shown that vitamin C protects SH-SY5Y cells from apoptosis and death induced by Aβ, through modulation of oxidative stress (185). Similarly, vitamin C showed a neuroprotective effect in human neural progenitor cells through the downregulation of WNT/β-catenin/ROS signaling pathway, improving cellular damage (189). In addition, vitamin C significantly protected neurons from N-methyl-D-aspartic acid (NMDA)- or glutamate-induced injury and significantly reduced cell death, proposing its role in preventing excitotoxicity in cerebral cortical neuron cells (190). Furthermore, vitamin C restores behavioral deficits and decreased Aβ oligomerization, brain oxidative stress, ratio of soluble AB42 to AB40, increased synaptophysin levels, and phosphorylation of tau at SER3 in a mouse model of AD (191). An in vivo study has demonstrated that vitamin B12 deficiency in the brain impairs cognition, increases amyloid accumulation and deposition, as well as oxidative stress in the APP/PS1 mouse model (186). Similarly, in another study, low vitamin C levels were associated with an increase of oxidative stress, neurodegeneration, cognitive decline, and AD development in APP/PS1 mouse model (187). Additionally, vitamin C deficiency decreased dopamine and its metabolites 3,4-dihydroxyphenyl-acetic acid (DOPAC), 3-methoxytyramine (3-MT), and homovanillic acid (HVA) release in APP/PSEN1 mice, leading to cognitive impairment (188). Moreover, vitamin C supplementation was able to increase glutamate levels, reduce neuronal apoptosis in the hippocampal CA1, pro-apoptotic Bax, and increase the levels of Bcl-2 (192). Clinical evidence further supports the potential role of vitamin C in cognitive function. In a double-blind, placebo-controlled trial, healthy young adults supplemented with vitamin C showed improved reaction time and attention scores, which were positively correlated with plasma vitamin C levels (193). In elderly individuals, long-term vitamin C supplementation improved mood, reduced memory-related errors, and enhanced global cognition, particularly in those with lower baseline cognitive performance (194). Furthermore, evidence regarding vitamin C and cognition is also mixed. Although some studies demonstrate reduced oxidative stress and improved memory with higher plasma vitamin C levels, interventional trials show minimal or no improvement in cognitive scores (195, 196).

There is limited research on the role of vitamin C on miR levels in the treatment of cognitive decline and dementia. However, it has been shown that vitamin C can regenerate antioxidants and reduce the production of inflammatory signaling molecules such as IL-6 and TNF-α, through modulation the expression of miR-155 (197). By reducing oxidative damage, antioxidants help preserve brain cells, reduce inflammation, and protect against cellular changes that could lead to dementias, memory deficits, and cognitive decline (198). More studies need to be done on this therapeutic target and the miRs involved, as there are molecular pathways with vitamin C and the progression of neurological disorders that have not been fully uncovered yet (199).

4.2.3 Vitamin E

Vitamin E, a lipid-soluble antioxidant, exists in eight isoforms, among which α-tocopherol is the most biologically active in humans (200). Dietary sources such as nuts, seeds, vegetable oils, and green leafy vegetables provide vitamin E in highly bioavailable forms, often showing greater absorption efficiency than synthetic supplements (201). Upon ingestion, vitamin E is released in the small intestine, where it is emulsified by bile salts and incorporated into mixed micelles, facilitating its uptake via membrane transporters such as SR-BI and CD36 (202). Once absorbed, α-tocopherol is transported in plasma predominantly bound to lipoproteins, including very low-density lipoprotein and high-density lipoprotein, which mediate its systemic distribution and uptake by peripheral tissues (202).

Vitamin E has exhibited a crucial role in managing dementia development by modulating different signaling pathways. It has been described that vitamin E protected cultured cortical neurons from oxidative stress-induced cell death through the activation of MAPK-phosphatidylinositol 3-kinase (PI3K), decreasing the Bax/Bcl-2 ratio and apoptosis (203). Furthermore, an in vitro study using hippocampal HT22 cells and rat cerebellar granule neurons demonstrated that vitamin E protected neurons effectively against oxidative cell death by inducing the activity of the redox-sensitive transcription factor NF-kB (204). In addition, vitamin E prevents neuronal cell death by repressing COX-2 activity in HT22 cells (205). Additionally, the antioxidant and free radical scavenger effect of vitamin E inhibited Aβ-induced cell death in PC12 cells (206). In an LPS-activated microglia model in vitro, vitamin E has been shown to reduce the levels of inflammatory mediators, including TNF-α, IL-1α, and NO, by inhibition of p38 MAPK- NF-kB (207). Furthermore, an in vivo study showed that vitamin E supplementation improved oxidative stress, cognitive functions, in aged mice by upregulation of SIRT1, Nrf2, and Calstabin2, as well as reducing the oxidative stress, attenuating cognitive decline (208). Similarly, vitamin E supplementation prevented spatial learning deficits and dendritic alterations in aged apolipoprotein E-deficient mice, decreasing lipid peroxidation and increasing glutathione levels (209). In addition, vitamin E protects against oxidative damage and learning disability after mild traumatic brain injury in rats, increasing hippocampal levels of BDNF, Calcium/calmodulin-dependent protein kinase II (CaMKII), CREB, synapsin I, duperoxide dismutase, and Sir2 (210). Despite promising antioxidant and neuroprotective properties, clinical outcomes with vitamin E remain inconsistent. Randomized controlled trials and meta-analyses have shown no significant cognitive benefit or disease progression delay in Alzheimer's disease patients receiving vitamin E supplementation (211, 212). Variability in dosage, disease stage, and study design may explain these conflicting results, underscoring the need for well-controlled longitudinal studies.

It has been shown that vitamin E can also modulate miRs expression (213). It has been shown that miR-122 was positively and significantly correlated with inflammatory molecules [granulocyte-macrophage colony-stimulating factor (GM-CSF), interferon alpha-2 (INF-α2), IL-1α, IL-8, and macrophage inflammatory protein-1 beta (MIP-1β)] as well as with vitamin E (214). Additionally, logistic regression analysis showed that vitamin E serum levels were associated with a higher AD probability and partially mediated by miR-122 (215).

4.2.4 Vitamin D

Vitamin D is another fat-soluble vitamin that is mainly obtained through sunlight exposure, dietary sources, and supplements (216). The major dietary sources of vitamin D are fatty fish (e.g., salmon, mackerel), fortified foods (e.g., milk, cereals), and supplements (vitamin D2 and D3) are primary sources (217). The presence of dietary fats, which enhances bioavailability, however, the breakdown of the dietary products in the upper gastrointestinal system, mainly in the duodenum, allows for this vitamin to be released (218). Evidence has linked adequate vitamin D levels to a reduced risk of cognitive decline and dementia, suggesting a potential neuroprotective effect (219).

It is well-known that vitamin D receptors have been found in neurons and glial cells, with their highest expression in the hippocampus and thalamus (220). It has been shown that Vitamin D downregulates the L-type voltage-sensitive calcium channels (LVSCC), LVSCC-A1C and LVSCC-A1D, and upregulates nerve growth factors, leading to a neuroprotection of cortical neurons (220). Furthermore, an in vitro study showed that vitamin D increases the Aβ phagocytosis by AD macrophages (221). Additionally, vitamin D supplementation ameliorated cognitive impairment and altered neurodegenerative (Aβ1-40, Aβ1-42, p-Tau, and Neprilysin) and inflammatory markers (IL-6, IL-1β, TNF-α, and NF-κβ) in the scopolamine-induced rat model (222). An in vivo study has demonstrated that vitamin D improves neural stem cell proliferation and oligodendrocyte differentiation by increasing the expression of important neurotrophic factors including neurotrophin-3 (NT-3), BDNF, glial cell line-derived neurotrophic factor (GDNF) and ciliary neurotrophic factor (CNTF) (223). Furthermore, in a randomized, double-blind, and placebo-controlled clinical trial, it was shown that vitamin D supplementation significantly improved the levels of Aβ-42, APP, and BACE1 in elderly patients with AD (224). Additionally, another study showed that low vitamin D concentration is associated with mild cognitive impairment status in the older population (225). However, studies have shown no significant reduction in the incidence or progression of dementia among older adults receiving vitamin D supplementation (226, 227). Thus, the role of vitamin D in the prevention of dementia may depend on baseline deficiency and population characteristics.

There is limited research on the role of vitamin D on miR levels in the treatment of cognitive decline and dementia. However, it has been shown that vitamin D upregulates miR-9-3p, which is related to neuroinflammation and memory deficits (228).

4.3 Metabolic modifiers/dietary interventions

The following section reviews the cognitive benefits of omega-3 fatty acids, ketogenic diets, the Mediterranean diet, and trehalose by consolidating findings from various studies. Each of these dietary modifications conveys neuroprotective effects either by preventing amyloid plaque accumulation, antioxidant properties, anti-inflammatory signaling, epigenetic modifications, or microRNA expression. Table 3 summarizes these mechanisms.

Table 3
www.frontiersin.org

Table 3. Effect of metabolic modifiers/dietary interventions on the pathophysiological complications associated with dementia.

4.3.1 Omega-3 fatty acids

Omega-3 fatty acids, or n-3 fatty acids (n-3 FAs), are essential fatty acids with a double bond between the third and fourth carbon atoms from the methyl group, or omega (229). The polyunsaturated n-3 FAs (n-3 PUFAs) of particular interest for their health benefits in humans are α-linoleic acid (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), with ALA acting as a precursor molecule for EPA and DHA (230). As n-3 PUFAs are essential, they must be consumed from various dietary sources such as seeds, vegetable oils, green leafy vegetables, fish oils, fish meat, beef, and lamb (230, 231). Sources such as chia seeds or flaxseed oil are rich in ALA, which requires further modification in the body after absorption before use; however, fish oil, fish meat, and marine mammal blubber are rich in EPA and DHA, which require no modification (231). Given the limited rate of conversion from ALA to DHA and EPA, recommendations to increase dietary intake of fish and fish oil, as well as supplementation with formulations containing EPA, DHA, and triglycerides, have become increasingly popular (232, 233). Several studies have shown that those with an increased consumption of omega-3 fatty acids are at a decreased risk for developing cognitive decline or dementia (232). Increases in consumption of DHA and EPA have been shown to lower the risk of cognitive decline by around 8%−9.9% per 0.1 g/d (234). This is likely due to the anti-inflammatory and neuroprotective properties that combat the chronic neuroinflammation and amyloid plaque buildup associated with dementia and Alzheimer's disease progression, namely Alzheimer's disease (235).

Multiple studies have revealed that Omega-3 fatty acids show promising effects in mitigating dementia progression through a wide variety of signaling pathways (236238). EPA and DHA have shown promise in suppressing the inflammatory responses of LPS-stimulated mouse microglia through the SIRT1 pathway-mediated inhibition of NF-κB signaling, decreasing the levels of pro-inflammatory cytokines IL-6, TNF,α as well as the key enzyme in the cellular metabolism, nicotinamide phosphoribosyl transferase (NAMPT) (236). In addition, DHA has also been shown to bind to the G protein-coupled receptor (GPR)120, suppressing microglia reactivity and neuroinflammation (237, 238). Furthermore, neuroprotectin D1, a stereoselective mediator derived from DHA has a protective effect against Aβ42 production by modulating key proteins involved in the processing of β-amyloid precursor protein, including peroxisome proliferator-activated receptor gamma (PPARγ)-dependent expression of BACE1, disintegrin and metalloproteinase 10 (ADAM10), and soluble amyloid precursor protein alpha (sAPPα), in 3xTg-AD mouse models and human neuronal-glial cells in primary culture (239). EPA and DHA also exhibit antiapoptotic effects in human neural cells and primary hippocampal neurons, by increasing the expression of Bcl-2, Bcl-xl, and Bcl-2-related gene expressed in fetal liver (Bfl-1) and decreasing the expression of Bcl-2-associated x Bax proteins through neuroprotectin D1 (NPD1)-mediated suppression of Aβ42 and suppression of caspase-3 and caspase-9 (240, 241). In addition, EPA and DHA suppressed the formation of tangles in AD through the attenuation of phosphorylated glycogen synthase kinase 3 (p-GSK3β) and p-Tau expression (241). Furthermore, it has also been shown that DHA leads to the activation of the Akt-mTOR-S6K signaling pathway, promoting upregulation of Tau and collapsin response mediator protein 2 (CMPR2), two important axon-related proteins in cortical neurons (242). In addition, when phosphorylated by GSK3 in primary hippocampal neurons and SH-SY5Y neuroblastoma cells, the ability of CMPR2 to assist with axonal growth is decreased. The ability of DHA to reduce the expression of GSK3 and increase the expression of CMPR2 may help to explain some of the cognitive benefits of DHA (243).

While the data is limited, studies have shown that the neuroprotective effects of n-3 PUFAs may act through modifications of miR expression (244246). Supplementation with n-3 FAs has been shown to alleviate neuroinflammation in mouse models by upregulating miR-107 targeting piezo-type mechanosensitive ion channel component 1 (PIEZO1)/NFκB p65 signaling pathway (244). Elevation of miR-29a-3p expression as a result of treatment with EPA in murine BV-2 cells lowered the activity of nod-like receptor pyrin-containing 3 (NLRP3) inflammasomes by activating autophagy through direct interaction with MAPK8 (245). Analysis of gene expression affected by foods rich in n-3 PUFAs also shows changes in the expression of miR-17-5p, which evidence suggests could influence the regulation of APP in in vitro models (246, 247).

4.3.2 Ketogenic diet

The classic ketogenic diet is a low-carbohydrate, high-fat diet with varying levels of protein designed to induce ketosis in the body for different health benefits (248). Ketosis occurs because the main source of energy for the body has become fatty acids, which are converted to acetyl-CoA in the liver via β-oxidation followed by synthesis with acetoacetyl-CoA to form 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA), the cleavage of HMG-CoA to acetyl-CoA and acetoacetate (ACA), and the eventual reduction of acetoacetate to β-hydroxybutyrate (β-HB) (249). When the levels of ketone bodies in circulation are high enough, the ketone bodies ACA and β-HB are transported across the blood-brain barrier using monocarboxylate transporters, where they are converted back to acetyl-CoA and modified in the tricarboxylic acid cycle without the use of ATP (250). The brain's usage of ketone bodies as energy, when paired with a calorie deficit, provides several neuroprotective benefits, such as a reduction in the damage and dysfunction of neurons, an increase in neuroprotective factors, an improvement in mitochondrial function, and a decrease in neuroinflammation (251).

Ketogenic diets have been reported to have protective roles in dementia progression; however, the neuroprotective effects are not yet fully understood. It has been shown that one of the mechanisms related to Ketogenic diets is related to decrease in oxidative stress levels (252). In an in vitro study in neocortical neurons, ketone bodies significantly inhibited oxidative stress through mitochondria modulation, a key pathway that contributes to vascular dementia and AD progression (253). Similarly, in another study, Ketone bodies were able to increase the rate of reduced nicotinamide adenine dinucleotide (NADH) oxidation without altering glutathione levels, protecting against glutamate toxicity and ultimately, neuronal death (254). In addition, while ketone bodies showed a reduction in the amount of reactive ROS produced by neuronal mitochondria, specifically in complex I, these molecules also increased the expression of uncoupling proteins (UCP)-2, UCP-4, and UCP-5 as well, further decreasing the level of ROS, leading to neuroprotective effects in the hippocampus (255). Another study, which evaluated the effects of β-HB treatment on mice, showed that β-HB also acts as a histone deacetylase inhibitor, suppressing oxidative stress by modulating the expression of forkhead box O3a (FOXO3A) and metallothionein 2 (MT2) (256). Additionally, insulin sensitivity, which is impaired in AD development, is also affected by ketone bodies through modulation in the expression of protein kinase A (PKA), ERK1/2, Ras/Raf, PI3K, and JNK in neurons and astrocytes from C57Bl6/N mice (257, 258). The ability of ketone bodies to affect insulin sensitivity, as well as act as an alternate energy source to glucose, may help ameliorate the increased cerebral amyloid burden that correlates with the high-glycemic state seen in Alzheimer's disease (259, 260). Additionally, comparative studies of the brains of AD patients and cognitively normal aged adults showed impaired uptake of glucose, but not ketone bodies, further supporting the role of ketone bodies as an alternative energy source for the brain (261). Clinical trials have also shown improvements in daily functioning, quality of life, and memory for Alzheimer's patients consuming a ketogenic diet (262, 263).

There is limited research on the impact of Ketogenic diets on miR levels in dementia progression.

4.3.4 Mediterranean diet

The Mediterranean diet (MD) refers to the typical dietary patterns of those living in parts of Greece and Italy in the 1960s; the diet is characterized by unrefined plant foods, olive oil, eggs, fish, poultry, small amounts of red meat, and moderate red wine intake (264). The diet has long been studied for its health benefits, including reducing the risk of dementia and AD development (265, 266). Some of the potential health benefits are due to the high amount of omega-3 fatty acids consumed in the diet, the benefits of which have been addressed in this review (246). The diet is also rich in polyphenols and B, C, D, and E vitamins, whose neuroprotective effects have also been reviewed (246). Other beneficial effects of the MD come from the consumption of magnesium-rich foods, which show cognitive benefits through alterations of the calcium: magnesium (Ca:Mg) ratio (267). Overall, the nutrients consumed from the classic MD confer antioxidant effects, anti-inflammatory effects, and inhibition of Aβ deposition, reducing the risk of dementia and AD development (268, 269).

The MD is rich in calcium and magnesium (267, 270). Lowering the Ca:Mg ratio by Mg supplementation improves cognitive function, partially through 5-mC modifications of two CpG sites in the APOE gene (271). In addition, magnesium itself directly inhibits NMDA receptors, a receptor that plays a critical role in Alzheimer's pathology, through glutamate excitotoxicity (272). In another study, magnesium was able to modulate the cleavage of Aβ precursor protein to the non-amyloidogenic pathway through activation of the ERK1/2 and PPARγ signaling pathways in APPswe/PS1DE9 transgenic mice (mouse model of AD), improving cognitive decline (273, 274). In addition, magnesium ameliorated cognitive deficit by attenuating hippocampal neurogenesis impairment in an APPswe/PS1dE9 mouse model of AD (275).

Comparisons of gene regulation and expression in the MD show upregulation of three miRs: miR-17-5p, miR-335-5p, and miR-93-5p (246). Analysis of gene expressions affected by foods rich in n-3 PUFAs also shows changes in the expression of miR-17-5p, which evidence suggests could affect the regulation of APP in in vitro models (246, 247). miR-335-5p is inversely correlated with the expression of JNK3, a protein associated with Aβ accumulation (276). The role of miR-93-5p in dementia has not yet been fully elucidated, but studies show that it is upregulated in the brain tissue of patients with AD (277279).

4.3.5 Trehalose

Trehalose is a disaccharide comprised of two glucose molecules, commonly found in plants, bacteria, and fungi (280). Trehalose is useful in cryoprotection and protein stabilization and has been found to modulate metabolism in cancer cells, has shown influence over several signaling pathways, and its neuroprotective effects are still under investigation (281). Studies have shown that trehalose is capable of acting as an autophagy activator, inhibiting the accumulation of proteins, and influencing different pathways involved in oxidative stress, protein folding, and neuroinflammation regulation (282).

It has been shown that trehalose has a protective effect on dementia and AD development (283285). An in vitro study showed that trehalose upregulates progranulin, a secreted growth factor important for neuronal survival and lysosome homeostasis, improving dementia in human cells H4, N2a, and SH-SY5Y (283). Furthermore, an in vivo study demonstrated that trehalose improved spatial learning and memory, levels of synaptophysin (synaptic vesicle protein), and doublecortin (marker of neurogenesis) in the hippocampus and cortex in the Tg2576 Alzheimer's mouse model (284). Similarly, trehalose significantly restored memory, reduced neuroinflammation, and preserved neuronal density in the hippocampus and frontal cortex in the Aβ-induced murine model of AD (285).

Studies have shown that Trehalose modulates neuroinflammation by regulating a variety of miRs (286288). The expression of miR-132, as well as the expression of SIRT1, is upregulated by trehalose in the hippocampus, reducing neuroinflammation (286). miR-132 also directly inhibits phosphatase and tensin homolog deleted on chromosome 10 (PTEN), forkhead transcription factor O3a (FOXO3a), and histone acetyltransferase p300 (P300), which are pro-apoptotic proteins of the Akt pathway (289). miR-181c is upregulated by trehalose and alleviates the progression of AD via targeted downregulation of NLRP1 (287, 290). Additionally, miR-539-5p is upregulated by trehalose, also contributing to neuroprotection (288). miR-539-5p inhibits Aβ accumulation, tau phosphorylation, oxidative stress, as well as apoptosis by directly targeting APP, Caveolin 1, and GSK-3β. The neuroprotective effects are conveyed via regulation of the PI3K/Akt/GSK-3β pathways as well (291).

5 Translational and clinical perspectives

As beneficial as the covered nutraceutical interventions may be for alleviating dementia symptoms and progression, there are many barriers to consider when implementing nutraceutical-based strategies in rural settings. Those living in rural areas face persistent structural challenges, including limited affordability, reduced proximity to healthy food sources, insufficient nutritional education, and entrenched stigmas or misconceptions surrounding health programs.

A significant contributor to rural nutritional disparity is the prevalence of food deserts, where individuals consume lower-quality diets compared to their urban counterparts (13). The Supplemental Nutrition Assistance Program (SNAP) is a program designed to assist low-income households with the acquisition of food; however, it does not adequately address rural-specific constraints (292). Research indicates that even SNAP recipients in rural Appalachia experience disproportionately high food insecurity relative to non-recipients, underscoring systemic gaps in reach and effectiveness (292). Grocery stores that accept SNAP benefits are also less likely to have diverse food options, and rural farmers' markets are less likely to accept SNAP benefits (293). Transportation barriers, administrative complexity, and reduced program accessibility (294, 295). Educational efforts, specifically those targeting schools, can play a vital role in shaping long-term health behaviors. Poor childhood dietary habits are associated with chronic conditions in adulthood (296, 297). Structured school lunch initiatives requiring fruit and vegetable intake have shown the capability to reduce childhood obesity, which in turn reduces the risk of obesity in adulthood, a major risk factor for poor health outcomes like dementia (298, 299). Beyond SNAP, community-based interventions that modify both education and food environments have proven effective (300). In addition to improving SNAP coverage for rural contexts, expanding educational outreach and increasing farmers' market participation may strengthen program impact (293, 300). These initiatives underscore the importance of local engagement and multilevel interventions in promoting healthier dietary behaviors in underserved rural regions.

This review encompasses several dietary components and modifications as interventions for dementia and its associated complications, particularly in rural populations. However, effective dietary strategies to reduce dementia risk must go beyond generalized recommendations. While lifestyle modifications are associated with lower dementia risk in older populations, isolated supplements or single modifications can only partially counteract genetic susceptibility and environmental exposures (301). The most effective treatments for dementia are multimodal and personalized to account for the patient's genetic predispositions and environmental factors (302, 303). Only 12.2% of Americans are metabolically healthy, with rural elderly populations disproportionately affected by metabolic syndrome (303, 304). In order to properly implement dietary modifications to mitigate dementia risk, symptomology, and progression in elderly rural populations, the modifications must be multifactorial, personalized, and economically accessible through public health policies and community-based initiatives.

The novelty of our review is in linking population-level determinants with molecular mechanisms to propose a unified model of dementia risk. This model positions nutraceutical strategies as part of a broader approach to address health challenges in rural populations. Connecting social factors such as food insecurity and environmental toxin exposure to microbiota-driven neuroinflammation, it supports the development of targeted interventions at different levels, including public health actions, community programs, and molecular therapies.

6 Conclusions

In summary, this review describes the pathogenic progression of dementia through the gut-brain axis and the importance of nutraceuticals for its possible mitigation (Figure 2). As presented, dementia is a multifactorial disorder influenced by genetic, environmental, and lifestyle factors, many of which are disproportionately concentrated in rural areas. We emphasized that rural populations are vulnerable to cognitive impairment due to nutritional inadequacies, limited access to preventive healthcare, and adverse social determinants of health. Additionally, we showed that the gut-brain axis has emerged as a central mediator linking these factors to neuroinflammation and cognitive decline. Furthermore, the nutraceuticals presented have demonstrated efficacy in preclinical and clinical models by attenuating oxidative stress, modulating inflammatory responses, and regulating miRs involved in neuronal survival. We believe that the studies consolidated in this review illustrate the therapeutic potential of specific nutraceuticals in mitigating cognitive impairment in rural populations through modulation of the gut-brain axis. However, translating these findings into clinical and public health practice remains challenging. Economic constraints, geographical barriers, and inequitable access to healthcare services continue to limit implementation—particularly in rural settings with insufficient infrastructure. Successful integration of precision nutrition into dementia prevention and management will require coordinated interdisciplinary efforts, evidence-based clinical guidelines, and policy-level support. Importantly, the strategies discussed here align with the United Nations Sustainable Development Goals (SDGs), particularly SDG 3 (“Good Health and WellBeing”) and SDG 10 (“Reduced Inequalities”), by promoting equitable access to nutrition-based interventions and reducing disparities in cognitive health outcomes. Addressing these barriers is essential to ensure that nutraceutical-based strategies are accessible, scalable, and effective across diverse populations. Future work should focus on integrating precision nutrition into rural healthcare frameworks, strengthening the gut–brain axis as a therapeutic target, and supporting long-term dietary interventions to promote cognitive resilience in vulnerable communities.

Figure 2
Flowchart illustrating the impact of reduced healthcare and malnutrition in rural populations, leading to gut-brain axis dysregulation, neuroinflammation, neuronal damage, and cognitive impairment. Nutritional interventions like phytochemicals, vitamins, and metabolic modifiers are suggested to address these issues.

Figure 2. Schematic representation showing the nutritional strategies against dementia in rural populations. Image proposed by the authors.

Author contributions

BG: Conceptualization, Project administration, Supervision, Visualization, Writing – original draft, Writing – review & editing. NG: Writing – original draft. CY: Writing – original draft. AK: Writing – original draft. AlN: Writing – original draft. SP: Writing – original draft. RP: Writing – original draft. AsN: Conceptualization, Project administration, Supervision, Visualization, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Gen AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Ornish D, Madison C, Kivipelto M, Kemp C, McCulloch CE, Galasko D, et al. Effects of intensive lifestyle changes on the progression of mild cognitive impairment or early dementia due to Alzheimer's disease: a randomized, controlled clinical trial. Alzheimers Res Ther. (2024) 16:122. doi: 10.1186/s13195-024-01482-z

PubMed Abstract | Crossref Full Text | Google Scholar

2. Schneider JA. Neuropathology of dementia disorders. Continuum. (2022) 28:834–51. doi: 10.1212/CON.0000000000001137

PubMed Abstract | Crossref Full Text | Google Scholar

3. Fang M, Hu J, Weiss J, Knopman DS, Albert M, Windham BG, et al. Lifetime risk and projected burden of dementia. Nat Med. (2025) 31:772–6. doi: 10.1038/s41591-024-03340-9

PubMed Abstract | Crossref Full Text | Google Scholar

4. Rahman M, White EM, Mills C, Thomas KS, Jutkowitz E. Rural-urban differences in diagnostic incidence and prevalence of Alzheimer's disease and related dementias. Alzheimers Dement. (2021) 17:1213–30. doi: 10.1002/alz.12285

PubMed Abstract | Crossref Full Text | Google Scholar

5. Abner EL, Jicha GA, Christian WJ, Schreurs BG. Rural-urban differences in alzheimer's disease and related disorders diagnostic prevalence in Kentucky and West Virginia. J Rural Health. (2016) 32:314–20. doi: 10.1111/jrh.12155

PubMed Abstract | Crossref Full Text | Google Scholar

6. Kramer M, Cutty M, Knox S, Alekseyenko AV, Mollalo A. Rural-urban disparities of Alzheimer's disease and related dementias: a scoping review. Alzheimers Dement. (2025) 11:e70047. doi: 10.1002/trc2.70047

PubMed Abstract | Crossref Full Text | Google Scholar

7. Saenz JL, Downer B, Garcia MA, Wong R. Cognition and context: rural-urban differences in cognitive aging among older Mexican adults. J Aging Health. (2018) 30:965–86. doi: 10.1177/0898264317703560

PubMed Abstract | Crossref Full Text | Google Scholar

8. NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in hypertension prevalence and progress in treatment and control from 1990 to 2019: a pooled analysis of 1201 population-representative studies with 104 million participants. Lancet. (2021) 398:957–80. doi: 10.1016/S0140-6736(21)01330-1

Crossref Full Text | Google Scholar

9. Samanic CM, Barbour KE, Liu Y, Wang Y, Fang J, Lu H, et al. Prevalence of self-reported hypertension and antihypertensive medication use by county and rural-urban classification - United States, 2017. MMWR Morb Mortal Wkly Rep. (2020) 69:533–9. doi: 10.15585/mmwr.mm6918a1

PubMed Abstract | Crossref Full Text | Google Scholar

10. Dintica CS, Yaffe K. Epidemiology and risk factors for dementia. Psychiatr Clin North Am. (2022) 45:677–89. doi: 10.1016/j.psc.2022.07.011

PubMed Abstract | Crossref Full Text | Google Scholar

11. Byker Shanks C, Andress L, Hardison-Moody A, Jilcott Pitts S, Patton-Lopez M, Prewitt TE, et al. Food insecurity in the rural United States: an examination of struggles and coping mechanisms to feed a family among households with a low-income. Nutrients. (2022) 14:5250. doi: 10.3390/nu14245250

PubMed Abstract | Crossref Full Text | Google Scholar

12. NCD Risk Factor Collaboration (NCD-RisC). Rising rural body-mass index is the main driver of the global obesity epidemic in adults. Nature. (2019) 569:260–4. doi: 10.1038/s41586-019-1171-x

Crossref Full Text | Google Scholar

13. Lacko A, Ng SW, Popkin B. Urban vs. rural socioeconomic differences in the nutritional quality of household packaged food purchases by store type. Int J Environ Res Public Health. (2020) 17:7637. doi: 10.3390/ijerph17207637

PubMed Abstract | Crossref Full Text | Google Scholar

14. Choudhury S, Bi AZ, Medina-Lara A, Morrish N, Veettil PC. The rural food environment and its association with diet, nutrition status, and health outcomes in low-income and middle-income countries (LMICs): a systematic review. BMC Public Health. (2025) 25:994. doi: 10.1186/s12889-025-22098-4

PubMed Abstract | Crossref Full Text | Google Scholar

15. Miller WC, Rogalla D, Spencer D, Zia N, Griffith BN, Heinsberg HB. Community adaptations to an impending food desert in rural Appalachia, USA. Rural Remote Health. (2016) 16:3901. doi: 10.22605/RRH3901

PubMed Abstract | Crossref Full Text | Google Scholar

16. Jin H, Lu Y. Evaluating consumer nutrition environment in food deserts and food swamps. Int J Environ Res Public Health. (2021) 18:2675. doi: 10.3390/ijerph18052675

PubMed Abstract | Crossref Full Text | Google Scholar

17. Feng L, Chu Z, Quan X, Zhang Y, Yuan W, Yao Y, et al. Malnutrition is positively associated with cognitive decline in centenarians and oldest-old adults: a cross-sectional study. EClinicalMedicine. (2022) 47:101336. doi: 10.1016/j.eclinm.2022.101336

PubMed Abstract | Crossref Full Text | Google Scholar

18. Koziatek CA, Motola HL, Holden KS, Hubert-Simon J, Wise N, Prabu A, et al. Key sociodemographic factors and food packaging attributes associated with poor diet quality among rural Americans: a cross-sectional survey study. BMJ Open. (2025) 15:e099774. doi: 10.1136/bmjopen-2025-099774

PubMed Abstract | Crossref Full Text | Google Scholar

19. Juul F, Parekh N, Martinez-Steele E, Monteiro CA, Chang VW. Ultra-processed food consumption among US adults from 2001 to 2018. Am J Clin Nutr. (2022) 115:211–21. doi: 10.1093/ajcn/nqab305

PubMed Abstract | Crossref Full Text | Google Scholar

20. Beal T, Manohar S, Miachon L, Fanzo J. Nutrient-dense foods and diverse diets are important for ensuring adequate nutrition across the life course. Proc Natl Acad Sci USA. (2024) 121:e2319007121. doi: 10.1073/pnas.2319007121

PubMed Abstract | Crossref Full Text | Google Scholar

21. Levy RB, Barata MF, Leite MA, Andrade GC. How and why ultra-processed foods harm human health. Proc Nutr Soc. (2024) 83:1–8. doi: 10.1017/S0029665123003567

PubMed Abstract | Crossref Full Text | Google Scholar

22. Kim C, Schilder N, Adolphus K, Berry A, Musillo C, Dye L, et al. The dynamic influence of nutrition on prolonged cognitive healthspan across the life course: a perspective review. Neurosci Appl. (2024) 3:104072. doi: 10.1016/j.nsa.2024.104072

PubMed Abstract | Crossref Full Text | Google Scholar

23. Park H, Ni M, Le Y. Neuroinflammation and nutrition in Alzheimer's disease. Front Neurol. (2025) 16:1622571. doi: 10.3389/fneur.2025.1622571

PubMed Abstract | Crossref Full Text | Google Scholar

24. Samieri C, Yassine HN. Melo van Lent D, Lefevre-Arbogast S, van de Rest O, Bowman GL, et al. Personalized nutrition for dementia prevention. Alzheimers Dement. (2022) 18:1424–37. doi: 10.1002/alz.12486

Crossref Full Text | Google Scholar

25. Jung SE, Kim S, Bishop A, Hermann J. Poor nutritional status among low-income older adults: examining the interconnection between self-care capacity, food insecurity, and depression. J Acad Nutr Diet. (2019) 119:1687–94. doi: 10.1016/j.jand.2018.04.009

PubMed Abstract | Crossref Full Text | Google Scholar

26. Rahman M, White EM, Thomas KS, Jutkowitz E. Assessment of rural-urban differences in health care use and survival among medicare beneficiaries with alzheimer disease and related dementia. JAMA Netw Open. (2020) 3:e2022111. doi: 10.1001/jamanetworkopen.2020.22111

PubMed Abstract | Crossref Full Text | Google Scholar

27. Kosar CM, Loomer L, Ferdows NB, Trivedi AN, Panagiotou OA, Rahman M. Assessment of rural-urban differences in postacute care utilization and outcomes among older US adults. JAMA Netw Open. (2020) 3:e1918738. doi: 10.1001/jamanetworkopen.2019.18738

PubMed Abstract | Crossref Full Text | Google Scholar

28. Li H, Li S, Yang H, Zhang Y, Zhang S, Ma Y, et al. Association of ultraprocessed food consumption with risk of dementia: a prospective cohort study. Neurology. (2022) 99:e1056–e66. doi: 10.1212/WNL.0000000000200871

PubMed Abstract | Crossref Full Text | Google Scholar

29. Totsch SK, Meir RY, Quinn TL, Lopez SA, Gower BA, Sorge RE. Effects of a Standard American Diet and an anti-inflammatory diet in male and female mice. Eur J Pain. (2018) 22:1203–13. doi: 10.1002/ejp.1207

PubMed Abstract | Crossref Full Text | Google Scholar

30. Clemente-Suarez VJ, Beltran-Velasco AI, Redondo-Florez L, Martin-Rodriguez A, Tornero-Aguilera JF. Global impacts of western diet and its effects on metabolism and health: a narrative review. Nutrients. (2023) 15:2749. doi: 10.3390/nu15122749

PubMed Abstract | Crossref Full Text | Google Scholar

31. Arikawa AY, Waterman A, Mishra SP, Labyak C, Williams C, Chaudhari DS, et al. Cognitive impairment is associated with alterations in diet quality and inflammatory biomarkers in older adults: a cross-sectional analysis of data collected from the Microbiome in Aging Gut and Brain (MiaGB) consortium cohort. J Nutr. (2025) 155:3048–3056. doi: 10.1016/j.tjnut.2025.06.026. [Epub ahead of print].

PubMed Abstract | Crossref Full Text | Google Scholar

32. Fado R, Molins A, Rojas R, Casals N. Feeding the brain: effect of nutrients on cognition, synaptic function, and AMPA Receptors. Nutrients. (2022) 14:4137. doi: 10.3390/nu14194137

PubMed Abstract | Crossref Full Text | Google Scholar

33. Vesga-Jimenez DJ, Martin C, Barreto GE, Aristizabal-Pachon AF, Pinzon A, Gonzalez J. Fatty acids: an insight into the pathogenesis of neurodegenerative diseases and therapeutic potential. Int J Mol Sci. (2022) 23:2577. doi: 10.3390/ijms23052577

PubMed Abstract | Crossref Full Text | Google Scholar

34. Plascencia-Villa G, Perry G. Roles of oxidative stress in synaptic dysfunction and neuronal cell death in Alzheimer's disease. Antioxidants. (2023) 12:1628. doi: 10.3390/antiox12081628

PubMed Abstract | Crossref Full Text | Google Scholar

35. Gil Martinez V, Avedillo Salas A, Santander Ballestin S. Vitamin supplementation and dementia: a systematic review. Nutrients. (2022) 14:1033. doi: 10.3390/nu14051033

PubMed Abstract | Crossref Full Text | Google Scholar

36. Feng S, Wang T, Su Y, Yan J, Wang Y, Zhang Z, et al. Global burden, risk factors, and projections of early-onset dementia: Insights from the Global Burden of Disease Study 2021. Ageing Res Rev. (2025) 104:102644. doi: 10.1016/j.arr.2024.102644

PubMed Abstract | Crossref Full Text | Google Scholar

37. Zhang Z, Han S, Zhu H, Wang Q, Cheng S, Han Y, et al. Global, regional, and national burden of early-onset Alzheimer's disease and other dementias in young adults aged 40-64 years, 1990-2021: a population-based study. Eur J Neurol. (2025) 32:e70116. doi: 10.1111/ene.70116

PubMed Abstract | Crossref Full Text | Google Scholar

38. Hameed M, Noor F, Hussain H, Khan RG, Khattak Haroon Ur Rashid S, Haroon Ur Rashid S, et al. Gut-brain axis: investigating the effects of gut health on cognitive functioning in adults. Cureus. (2024) 16:e64286. doi: 10.7759/cureus.64286

PubMed Abstract | Crossref Full Text | Google Scholar

39. Trisal A, Singh I, Garg G, Jorwal K, Singh AK. Gut-brain axis and brain health: modulating neuroinflammation, cognitive decline, and neurodegeneration. 3 Biotech. (2025) 15:25. doi: 10.1007/s13205-024-04187-0

PubMed Abstract | Crossref Full Text | Google Scholar

40. Tiwari P, Dwivedi R, Bansal M, Tripathi M, Dada R. Role of gut microbiota in neurological disorders and its therapeutic significance. J Clin Med. (2023) 12:1650. doi: 10.3390/jcm12041650

PubMed Abstract | Crossref Full Text | Google Scholar

41. Zhang R, Ding N, Feng X, Liao W. The gut microbiome, immune modulation, and cognitive decline: insights on the gut-brain axis. Front Immunol. (2025) 16:1529958. doi: 10.3389/fimmu.2025.1529958

PubMed Abstract | Crossref Full Text | Google Scholar

42. Khan MT, Zohair M, Khan A, Kashif A, Mumtaz S, Muskan F. From gut to brain: the roles of intestinal microbiota, immune system, and hormones in intestinal physiology and gut-brain-axis. Mol Cell Endocrinol. (2025) 607:112599. doi: 10.1016/j.mce.2025.112599

PubMed Abstract | Crossref Full Text | Google Scholar

43. Kasarello K, Cudnoch-Jedrzejewska A, Czarzasta K. Communication of gut microbiota and brain via immune and neuroendocrine signaling. Front Microbiol. (2023) 14:1118529. doi: 10.3389/fmicb.2023.1118529

PubMed Abstract | Crossref Full Text | Google Scholar

44. Stolzer I, Scherer E, Suss P, Rothhammer V, Winner B, Neurath MF, et al. Impact of microbiome-brain communication on neuroinflammation and neurodegeneration. Int J Mol Sci. (2023) 24:14925. doi: 10.3390/ijms241914925

PubMed Abstract | Crossref Full Text | Google Scholar

45. Zheng Y, Bonfili L, Wei T, Eleuteri AM. Understanding the gut-brain axis and its therapeutic implications for neurodegenerative disorders. Nutrients. (2023) 15:4631. doi: 10.3390/nu15214631

PubMed Abstract | Crossref Full Text | Google Scholar

46. Solanki R, Karande A, Ranganathan P. Emerging role of gut microbiota dysbiosis in neuroinflammation and neurodegeneration. Front Neurol. (2023) 14:1149618. doi: 10.3389/fneur.2023.1149618

PubMed Abstract | Crossref Full Text | Google Scholar

47. Tarawneh R, Penhos E. The gut microbiome and Alzheimer's disease: complex and bidirectional interactions. Neurosci Biobehav Rev. (2022) 141:104814. doi: 10.1016/j.neubiorev.2022.104814

PubMed Abstract | Crossref Full Text | Google Scholar

48. Mou Y, Du Y, Zhou L, Yue J, Hu X, Liu Y, et al. Gut microbiota interact with the brain through systemic chronic inflammation: implications on neuroinflammation, neurodegeneration, and aging. Front Immunol. (2022) 13:796288. doi: 10.3389/fimmu.2022.796288

PubMed Abstract | Crossref Full Text | Google Scholar

49. Fock E, Parnova R. Mechanisms of blood-brain barrier protection by microbiota-derived short-chain fatty acids. Cells. (2023) 12:657. doi: 10.3390/cells12040657

PubMed Abstract | Crossref Full Text | Google Scholar

50. Park J, Wang Q, Wu Q, Mao-Draayer Y, Kim CH. Bidirectional regulatory potentials of short-chain fatty acids and their G-protein-coupled receptors in autoimmune neuroinflammation. Sci Rep. (2019) 9:8837. doi: 10.1038/s41598-019-45311-y

PubMed Abstract | Crossref Full Text | Google Scholar

51. Liu Q, Xi Y, Wang Q, Liu J, Li P, Meng X, et al. Mannan oligosaccharide attenuates cognitive and behavioral disorders in the 5xFAD Alzheimer's disease mouse model via regulating the gut microbiota-brain axis. Brain Behav Immun. (2021) 95:330–43. doi: 10.1016/j.bbi.2021.04.005

PubMed Abstract | Crossref Full Text | Google Scholar

52. Sun J, Xu J, Ling Y, Wang F, Gong T, Yang C, et al. Fecal microbiota transplantation alleviated Alzheimer's disease-like pathogenesis in APP/PS1 transgenic mice. Transl Psychiatry. (2019) 9:189. doi: 10.1038/s41398-019-0525-3

PubMed Abstract | Crossref Full Text | Google Scholar

53. Berding K, Vlckova K, Marx W, Schellekens H, Stanton C, Clarke G, et al. Diet and the microbiota-gut-brain axis: sowing the seeds of good mental health. Adv Nutr. (2021) 12:1239–85. doi: 10.1093/advances/nmaa181

PubMed Abstract | Crossref Full Text | Google Scholar

54. Liow YJ, Sarkar A, Carmody RN. Industrialized diets modulate host eating behavior via the microbiome-gut-brain axis. Trends Endocrinol Metab. (2025). doi: 10.1016/j.tem.2025.06.002

PubMed Abstract | Crossref Full Text | Google Scholar

55. Horn J, Mayer DE, Chen S, Mayer EA. Role of diet and its effects on the gut microbiome in the pathophysiology of mental disorders. Transl Psychiatry. (2022) 12:164. doi: 10.1038/s41398-022-01922-0

PubMed Abstract | Crossref Full Text | Google Scholar

56. Zhang R, Zhang M, Wang P. The intricate interplay between dietary habits and cognitive function: insights from the gut-brain axis. Front Nutr. (2025) 12:1539355. doi: 10.3389/fnut.2025.1539355

PubMed Abstract | Crossref Full Text | Google Scholar

57. Wiatrak B, Balon K, Jawien P, Bednarz D, Jeskowiak I, Szelag A. The role of the microbiota-gut-brain axis in the development of Alzheimer's disease. Int J Mol Sci. (2022) 23:4862. doi: 10.3390/ijms23094862

PubMed Abstract | Crossref Full Text | Google Scholar

58. Townsend R, Fairley A, Gregory S, Ritchie C, Stevenson E, Shannon OM. Nutrition for dementia prevention: a state of the art update for clinicians. Age Ageing. (2024) 53:ii30–ii8. doi: 10.1093/ageing/afae030

PubMed Abstract | Crossref Full Text | Google Scholar

59. Menezes C, Campolim CM, Triana A, Oliveira KM, Gama SCL, Xavier FG, et al. Effects of ultra-processed diets on adiposity, gut barrier integrity, inflammation, and microbiota in male and female mice. Nutrients. (2025) 17:3116. doi: 10.3390/nu17193116

PubMed Abstract | Crossref Full Text | Google Scholar

60. Chen Q, Shi J, Yu G, Xie H, Yu S, Xu J, et al. Gut microbiota dysbiosis in patients with Alzheimer's disease and correlation with multiple cognitive domains. Front Aging Neurosci. (2024) 16:1478557. doi: 10.3389/fnagi.2024.1478557

PubMed Abstract | Crossref Full Text | Google Scholar

61. Qian H, Khadka A, Martinez SM, Singh S, Brenowitz WD, Zeki Al Hazzouri A, et al. Food insecurity, memory, and dementia among us adults aged 50 years and older. JAMA Netw Open. (2023) 6:e2344186. doi: 10.1001/jamanetworkopen.2023.44186

PubMed Abstract | Crossref Full Text | Google Scholar

62. Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. (2016) 165:1332–45. doi: 10.1016/j.cell.2016.05.041

PubMed Abstract | Crossref Full Text | Google Scholar

63. Horwat P, Mariowska A, Szymanska A, Dzieciatkowska M, Pierudzka W. Multimodal interventions targeting gut microbiota and microbial metabolites in cognitive impairment. Cureus. (2025) 17:e85688. doi: 10.7759/cureus.85688

PubMed Abstract | Crossref Full Text | Google Scholar

64. Zhang T, Gao G, Kwok LY, Sun Z. Gut microbiome-targeted therapies for Alzheimer's disease. Gut Microbes. (2023) 15:2271613. doi: 10.1080/19490976.2023.2271613

PubMed Abstract | Crossref Full Text | Google Scholar

65. Assad A, Johnson SL. 3rd, Reichard RA. Probiotic supplementation improves the clinical measures of cognition in patients with mild cognitive impairment. Cureus. (2025) 17:e89302. doi: 10.7759/cureus.89302

Crossref Full Text | Google Scholar

66. Hsu YC, Huang YY, Tsai SY, Kuo YW, Lin JH, Ho HH, et al. Efficacy of probiotic supplements on brain-derived neurotrophic factor, inflammatory biomarkers, oxidative stress and cognitive function in patients with Alzheimer's dementia: a 12-week randomized, double-blind active-controlled study. Nutrients. (2023) 16:16. doi: 10.3390/nu16010016

PubMed Abstract | Crossref Full Text | Google Scholar

67. Li X, Ding Q, Wan X, Wu Q, Ye S, Lou Y. Fecal microbiota transplantation attenuates Alzheimer's disease symptoms in APP/PS1 transgenic mice via inhibition of the TLR4-MyD88-NF-kappaB signaling pathway-mediated inflammation. Behav Brain Funct. (2025) 21:2. doi: 10.1186/s12993-024-00265-8

Crossref Full Text | Google Scholar

68. Zhang K, Paul K, Jacobs JP, Cockburn MG, Bronstein JM, Del Rosario I, et al. Ambient long-term exposure to organophosphorus pesticides and the human gut microbiome: an observational study. Environ Health. (2024) 23:41. doi: 10.1186/s12940-024-01078-y

PubMed Abstract | Crossref Full Text | Google Scholar

69. Wiese LAK, Gibson A, Guest MA, Nelson AR, Weaver R, Gupta A, et al. Global rural health disparities in Alzheimer's disease and related dementias: state of the science. Alzheimers Dement. (2023) 19:4204–25. doi: 10.1002/alz.13104

PubMed Abstract | Crossref Full Text | Google Scholar

70. Dintica CS, Bahorik A, Xia F, Kind A, Yaffe K. Dementia risk and disadvantaged neighborhoods. JAMA Neurol. (2023) 80:903–9. doi: 10.1001/jamaneurol.2023.2120

PubMed Abstract | Crossref Full Text | Google Scholar

71. AlAli M, Alqubaisy M, Aljaafari MN, AlAli AO, Baqais L, Molouki A, et al. Nutraceuticals: transformation of conventional foods into health promoters/disease preventers and safety considerations. Molecules. (2021) 26:2540. doi: 10.3390/molecules26092540

PubMed Abstract | Crossref Full Text | Google Scholar

72. Sachdeva V, Roy A, Bharadvaja N. Current prospects of nutraceuticals: a review. Curr Pharm Biotechnol. (2020) 21:884–96. doi: 10.2174/1389201021666200130113441

PubMed Abstract | Crossref Full Text | Google Scholar

73. Alba G, Dakhaoui H, Santa-Maria C, Palomares F, Cejudo-Guillen M, Geniz I, et al. Nutraceuticals as potential therapeutic modulators in immunometabolism. Nutrients. (2023) 15:411. doi: 10.3390/nu15020411

PubMed Abstract | Crossref Full Text | Google Scholar

74. Jeyabalan JB, Pathak S, Mariappan E, Mohanakumar KP, Dhanasekaran M. Validating the nutraceutical and neuroprotective pharmacodynamics of flavones. Neurochem Int. (2024) 180:105829. doi: 10.1016/j.neuint.2024.105829

PubMed Abstract | Crossref Full Text | Google Scholar

75. Desjarlais M, Wirth M, Lahaie I, Ruknudin P, Hardy P, Rivard A, et al. Nutraceutical targeting of inflammation-modulating microRNAs in severe forms of COVID-19: a novel approach to prevent the cytokine storm. Front Pharmacol. (2020) 11:602999. doi: 10.3389/fphar.2020.602999

PubMed Abstract | Crossref Full Text | Google Scholar

76. Alvarez M, Trent E, Goncalves BS, Pereira DG, Puri R, Frazier NA, et al. Cognitive dysfunction associated with COVID-19: Prognostic role of circulating biomarkers and microRNAs. Front Aging Neurosci. (2022) 14:1020092. doi: 10.3389/fnagi.2022.1020092

PubMed Abstract | Crossref Full Text | Google Scholar

77. Ghosh S, Kumar V, Mukherjee H, Lahiri D, Roy P. Nutraceutical regulation of miRNAs involved in neurodegenerative diseases and brain cancers. Heliyon. (2021) 7:e07262. doi: 10.1016/j.heliyon.2021.e07262

PubMed Abstract | Crossref Full Text | Google Scholar

78. Jaberi KR, Alamdari-Palangi V, Savardashtaki A, Vatankhah P, Jamialahmadi T, Tajbakhsh A, et al. Modulatory effects of phytochemicals on gut-brain axis: therapeutic implication. Curr Dev Nutr. (2024) 8:103785. doi: 10.1016/j.cdnut.2024.103785

PubMed Abstract | Crossref Full Text | Google Scholar

79. Sandur SK, Ichikawa H, Pandey MK, Kunnumakkara AB, Sung B, Sethi G, et al. Role of pro-oxidants and antioxidants in the anti-inflammatory and apoptotic effects of curcumin (diferuloylmethane). Free Radic Biol Med. (2007) 43:568–80. doi: 10.1016/j.freeradbiomed.2007.05.009

PubMed Abstract | Crossref Full Text | Google Scholar

80. Jalouli M, Rahman MA, Biswas P, Rahman H, Harrath AH, Lee IS, et al. Targeting natural antioxidant polyphenols to protect neuroinflammation and neurodegenerative diseases: a comprehensive review. Front Pharmacol. (2025) 16:1492517. doi: 10.3389/fphar.2025.1492517

PubMed Abstract | Crossref Full Text | Google Scholar

81. de Souza Goncalves B, Sangani D, Nayyar A, Puri R, Irtiza M, Nayyar A, et al. COVID-19-associated sepsis: potential role of phytochemicals as functional foods and nutraceuticals. Int J Mol Sci. (2024) 25:8781. doi: 10.3390/ijms25158481

PubMed Abstract | Crossref Full Text | Google Scholar

82. El-Saadony MT, Saad AM, Mohammed DM, Alkafaas SS, Ghosh S, Negm SH, et al. Curcumin, an active component of turmeric: biological activities, nutritional aspects, immunological, bioavailability, and human health benefits - a comprehensive review. Front Immunol. (2025) 16:1603018. doi: 10.3389/fimmu.2025.1603018

PubMed Abstract | Crossref Full Text | Google Scholar

83. Tsai IC, Hsu CW, Chang CH, Tseng PT, Chang KV. The effect of curcumin differs on individual cognitive domains across different patient populations: a systematic review and meta-analysis. Pharmaceuticals. (2021) 14:1235. doi: 10.3390/ph14121235

PubMed Abstract | Crossref Full Text | Google Scholar

84. Darmonkow A, Rowe ZEM, Harding SV. Advancements in curcuminoid formulations: an update on bioavailability enhancement strategies curcuminoid bioavailability and formulations. Open Life Sci. (2025) 20:20251112. doi: 10.1515/biol-2025-1112

PubMed Abstract | Crossref Full Text | Google Scholar

85. Azzini E, Pena-Corona SI, Hernandez-Parra H, Chandran D, Saleena LAK, Sawikr Y, et al. Neuroprotective and anti-inflammatory effects of curcumin in Alzheimer's disease: targeting neuroinflammation strategies. Phytother Res. (2024) 38:3169–89. doi: 10.1002/ptr.8200

PubMed Abstract | Crossref Full Text | Google Scholar

86. Shao S, Ye X, Su W, Wang Y. Curcumin alleviates Alzheimer's disease by inhibiting inflammatory response, oxidative stress and activating the AMPK pathway. J Chem Neuroanat. (2023) 134:102363. doi: 10.1016/j.jchemneu.2023.102363

PubMed Abstract | Crossref Full Text | Google Scholar

87. Yang F, Lim GP, Begum AN, Ubeda OJ, Simmons MR, Ambegaokar SS, et al. Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J Biol Chem. (2005) 280:5892–901. doi: 10.1074/jbc.M404751200

PubMed Abstract | Crossref Full Text | Google Scholar

88. Mishra S, Mishra M, Seth P, Sharma SK. Tetrahydrocurcumin confers protection against amyloid beta-induced toxicity. Neuroreport. (2011) 22:23–7. doi: 10.1097/WNR.0b013e328341e141

Crossref Full Text | Google Scholar

89. Zhang L, Fang Y, Xu Y, Lian Y, Xie N, Wu T, et al. Curcumin improves amyloid beta-peptide (1-42) induced spatial memory deficits through BDNF-ERK signaling pathway. PLoS ONE. (2015) 10:e0131525. doi: 10.1371/journal.pone.0131525

Crossref Full Text | Google Scholar

90. Gomez-Pinilla F, Nguyen TT. Natural mood foods: the actions of polyphenols against psychiatric and cognitive disorders. Nutr Neurosci. (2012) 15:127–33. doi: 10.1179/1476830511Y.0000000035

PubMed Abstract | Crossref Full Text | Google Scholar

91. Han Y, Chen R, Lin Q, Liu Y, Ge W, Cao H, et al. Curcumin improves memory deficits by inhibiting HMGB1-RAGE/TLR4-NF-kappaB signalling pathway in APPswe/PS1dE9 transgenic mice hippocampus. J Cell Mol Med. (2021) 25:8947–56. doi: 10.1111/jcmm.16855

Crossref Full Text | Google Scholar

92. Huang P, Zheng N, Zhou HB, Huang J. Curcumin inhibits BACE1 expression through the interaction between ERbeta and NFkappaB signaling pathway in SH-SY5Y cells. Mol Cell Biochem. (2020) 463:161–73. doi: 10.1007/s11010-019-03638-0

Crossref Full Text | Google Scholar

93. Maugeri A, Russo C, Patane GT, Barreca D, Mandalari G, Navarra M. The inhibition of mitogen-activated protein kinases (MAPKs) and NF-kappaB underlies the neuroprotective capacity of a cinnamon/curcumin/turmeric spice blend in abeta-exposed THP-1 cells. Molecules. (2023) 28:7949. doi: 10.3390/molecules28247949

Crossref Full Text | Google Scholar

94. Kim HY, Park EJ, Joe EH, Jou I. Curcumin suppresses Janus kinase-STAT inflammatory signaling through activation of Src homology 2 domain-containing tyrosine phosphatase 2 in brain microglia. J Immunol. (2003) 171:6072–9. doi: 10.4049/jimmunol.171.11.6072

PubMed Abstract | Crossref Full Text | Google Scholar

95. Anand P, Kunnumakkara AB, Newman RA, Aggarwal BB. Bioavailability of curcumin: problems and promises. Mol Pharm. (2007) 4:807–18. doi: 10.1021/mp700113r

PubMed Abstract | Crossref Full Text | Google Scholar

96. Fiala M, Liu PT, Espinosa-Jeffrey A, Rosenthal MJ, Bernard G, Ringman JM, et al. Innate immunity and transcription of MGAT-III and Toll-like receptors in Alzheimer's disease patients are improved by bisdemethoxycurcumin. Proc Natl Acad Sci USA. (2007) 104:12849–54. doi: 10.1073/pnas.0701267104

PubMed Abstract | Crossref Full Text | Google Scholar

97. Rainey-Smith SR, Brown BM, Sohrabi HR, Shah T, Goozee KG, Gupta VB, et al. Curcumin and cognition: a randomised, placebo-controlled, double-blind study of community-dwelling older adults. Br J Nutr. (2016) 115:2106–13. doi: 10.1017/S0007114516001203

PubMed Abstract | Crossref Full Text | Google Scholar

98. Wang X, Zhang S, Li Y, Zhang Y. The regulation of miRNAs using curcumin and other polyphenols during the prevention and treatment of Alzheimer's disease. Hum Mol Genet. (2025) 34:117–27. doi: 10.1093/hmg/ddae154

PubMed Abstract | Crossref Full Text | Google Scholar

99. Cui JG Li YY, Zhao Y, Bhattacharjee S, Lukiw WJ. Differential regulation of interleukin-1 receptor-associated kinase-1 (IRAK-1) and IRAK-2 by microRNA-146a and NF-kappaB in stressed human astroglial cells and in Alzheimer disease. J Biol Chem. (2010) 285:38951–60. doi: 10.1074/jbc.M110.178848

PubMed Abstract | Crossref Full Text | Google Scholar

100. Gong J, Sun D. Study on the mechanism of curcumin to reduce the inflammatory response of temporal lobe in Alzheimer's disease by regulating miR-146a. Minerva Med. (2022) 113:109–18. doi: 10.23736/S0026-4806.20.06463-0

PubMed Abstract | Crossref Full Text | Google Scholar

101. Li YY, Cui JG, Hill JM, Bhattacharjee S, Zhao Y, Lukiw WJ. Increased expression of miRNA-146a in Alzheimer's disease transgenic mouse models. Neurosci Lett. (2011) 487:94–8. doi: 10.1016/j.neulet.2010.09.079

PubMed Abstract | Crossref Full Text | Google Scholar

102. Nguyen HD, Jo WH, Hoang NHM, Kim MS. Curcumin-attenuated TREM-1/DAP12/NLRP3/Caspase-1/IL1B, TLR4/NF-kappaB pathways, and tau hyperphosphorylation induced by 1,2-Diacetyl Benzene: an in vitro and in silico study. Neurotox Res. (2022) 40:1272–91. doi: 10.1007/s12640-022-00535-1

Crossref Full Text | Google Scholar

103. Song X, Wang C, Ding Q, Li P, Sun S, Wei W, et al. Modulation of beta secretase and neuroinflammation by biomimetic nanodelivery system for Alzheimer's disease therapy. J Control Release. (2025) 378:735–49. doi: 10.1016/j.jconrel.2024.12.060

Crossref Full Text | Google Scholar

104. Samec D, Karalija E, Sola I, Vujcic Bok V, Salopek-Sondi B. The role of polyphenols in abiotic stress response: the influence of molecular structure. Plants. (2021) 10:118. doi: 10.3390/plants10010118

PubMed Abstract | Crossref Full Text | Google Scholar

105. Salla M, Karaki N, El Kaderi B, Ayoub AJ, Younes S, Abou Chahla MN, et al. Enhancing the bioavailability of resveratrol: combine it, derivatize it, or encapsulate it? Pharmaceutics. (2024) 16:569. doi: 10.3390/pharmaceutics16040569

PubMed Abstract | Crossref Full Text | Google Scholar

106. Bohara RA, Tabassum N, Singh MP, Gigli G, Ragusa A, Leporatti S. Recent overview of resveratrol's beneficial effects and its nano-delivery systems. Molecules. (2022) 27:5154. doi: 10.3390/molecules27165154

PubMed Abstract | Crossref Full Text | Google Scholar

107. Chen X, Zhang J, Yin N, Wele P, Li F, Dave S, et al. Resveratrol in disease prevention and health promotion: a role of the gut microbiome. Crit Rev Food Sci Nutr. (2024) 64:5878–95. doi: 10.1080/10408398.2022.2159921

PubMed Abstract | Crossref Full Text | Google Scholar

108. Xiang Z, Krainc D. Pharmacological upregulation of PGC1alpha in oligodendrocytes: implications for Huntington's disease. J Huntingtons Dis. (2013) 2:101–5. doi: 10.3233/JHD-130046

Crossref Full Text | Google Scholar

109. Xie J, Li X, Zhou Y, Wu J, Tan Y, Ma X, et al. Resveratrol abrogates hypoxia-induced up-regulation of exosomal amyloid-beta partially by inhibiting CD147. Neurochem Res. (2019) 44:1113–26. doi: 10.1007/s11064-019-02742-3

Crossref Full Text | Google Scholar

110. Li XM, Zhou MT, Wang XM Ji MH, Zhou ZQ, Yang JJ. Resveratrol pretreatment attenuates the isoflurane-induced cognitive impairment through its anti-inflammation and -apoptosis actions in aged mice. J Mol Neurosci. (2014) 52:286–93. doi: 10.1007/s12031-013-0141-2

PubMed Abstract | Crossref Full Text | Google Scholar

111. Moussa C, Hebron M, Huang X, Ahn J, Rissman RA, Aisen PS, et al. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer's disease. J Neuroinflammation. (2017) 14:1. doi: 10.1186/s12974-016-0779-0

PubMed Abstract | Crossref Full Text | Google Scholar

112. Huang TC, Lu KT, Wo YY, Wu YJ, Yang YL. Resveratrol protects rats from Abeta-induced neurotoxicity by the reduction of iNOS expression and lipid peroxidation. PLoS ONE. (2011) 6:e29102. doi: 10.1371/journal.pone.0029102

Crossref Full Text | Google Scholar

113. Rege SD, Geetha T, Broderick TL, Babu JR. Resveratrol protects beta amyloid-induced oxidative damage and memory associated proteins in H19-7 hippocampal neuronal cells. Curr Alzheimer Res. (2015) 12:147–56. doi: 10.2174/1567205012666150204130009

Crossref Full Text | Google Scholar

114. Cheng X, Wang Q, Li N, Zhao H. Effects of resveratrol on hippocampal astrocytes and expression of TNF-alpha in Alzheimer's disease model rate. Wei Sheng Yan Jiu. (2015) 44:610–4.

Google Scholar

115. Sanchez-Fidalgo S, Cardeno A, Villegas I, Talero E. de la Lastra CA. Dietary supplementation of resveratrol attenuates chronic colonic inflammation in mice. Eur J Pharmacol. (2010) 633:78–84. doi: 10.1016/j.ejphar.2010.01.025

Crossref Full Text | Google Scholar

116. Capiralla H, Vingtdeux V, Zhao H, Sankowski R, Al-Abed Y, Davies P, et al. Resveratrol mitigates lipopolysaccharide- and Abeta-mediated microglial inflammation by inhibiting the TLR4/NF-kappaB/STAT signaling cascade. J Neurochem. (2012) 120:461–72. doi: 10.1111/j.1471-4159.2011.07594.x

Crossref Full Text | Google Scholar

117. Chiang MC, Nicol CJ, Cheng YC. Resveratrol activation of AMPK-dependent pathways is neuroprotective in human neural stem cells against amyloid-beta-induced inflammation and oxidative stress. Neurochem Int. (2018) 115:1–10. doi: 10.1016/j.neuint.2017.10.002

PubMed Abstract | Crossref Full Text | Google Scholar

118. Vingtdeux V, Giliberto L, Zhao H, Chandakkar P, Wu Q, Simon JE, et al. AMP-activated protein kinase signaling activation by resveratrol modulates amyloid-beta peptide metabolism. J Biol Chem. (2010) 285:9100–13. doi: 10.1074/jbc.M109.060061

PubMed Abstract | Crossref Full Text | Google Scholar

119. Feng X, Liang N, Zhu D, Gao Q, Peng L, Dong H, et al. Resveratrol inhibits beta-amyloid-induced neuronal apoptosis through regulation of SIRT1-ROCK1 signaling pathway. PLoS ONE. (2013) 8:e59888. doi: 10.1371/journal.pone.0059888

Crossref Full Text | Google Scholar

120. Turner RS, Thomas RG, Craft S, van Dyck CH, Mintzer J, Reynolds BA, et al. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology. (2015) 85:1383–91. doi: 10.1212/WNL.0000000000002035

PubMed Abstract | Crossref Full Text | Google Scholar

121. Kumar R, Chaterjee P, Sharma PK, Singh AK, Gupta A, Gill K, et al. Sirtuin1: a promising serum protein marker for early detection of Alzheimer's disease. PLoS One. (2013) 8:e61560. doi: 10.1371/journal.pone.0061560

PubMed Abstract | Crossref Full Text | Google Scholar

122. Kou X, Chen N. Resveratrol as a natural autophagy regulator for prevention and treatment of Alzheimer's disease. Nutrients. (2017) 9:927. doi: 10.3390/nu9090927

PubMed Abstract | Crossref Full Text | Google Scholar

123. Lukiw WJ, Zhao Y, Cui JG. An NF-kappaB-sensitive micro RNA-146a-mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J Biol Chem. (2008) 283:31315–22. doi: 10.1074/jbc.M805371200

PubMed Abstract | Crossref Full Text | Google Scholar

124. Zhao YN Li WF, Li F, Zhang Z, Dai YD, Xu AL, et al. Resveratrol improves learning and memory in normally aged mice through microRNA-CREB pathway. Biochem Biophys Res Commun. (2013) 435:597–602. doi: 10.1016/j.bbrc.2013.05.025

PubMed Abstract | Crossref Full Text | Google Scholar

125. Hu Y, Xu J, Wang J, Wang J, Li Y, Chen W, et al. Resveratrol alleviates diabetic adipose tissue-derived extracellular vesicles-induced hippocampal ferroptosis and cognitive dysfunction via inhibiting miR-9-3p/SLC7A11 axis. Mol Neurobiol. (2025) 62:12307–30. doi: 10.1007/s12035-025-04922-5

PubMed Abstract | Crossref Full Text | Google Scholar

126. Kim J. Pre-clinical neuroprotective evidences and plausible mechanisms of sulforaphane in Alzheimer's disease. Int J Mol Sci. (2021) 22:2929. doi: 10.3390/ijms22062929

PubMed Abstract | Crossref Full Text | Google Scholar

127. Schepici G, Bramanti P, Mazzon E. Efficacy of sulforaphane in neurodegenerative diseases. Int J Mol Sci. (2020) 21:8637. doi: 10.3390/ijms21228637

PubMed Abstract | Crossref Full Text | Google Scholar

128. Fahey JW, Holtzclaw WD, Wehage SL, Wade KL, Stephenson KK, Talalay P. Sulforaphane bioavailability from glucoraphanin-rich broccoli: control by active endogenous myrosinase. PLoS ONE. (2015) 10:e0140963. doi: 10.1371/journal.pone.0140963

PubMed Abstract | Crossref Full Text | Google Scholar

129. Fahey JW, Liu H, Batt H, Panjwani AA, Tsuji P. Sulforaphane and brain health: from pathways of action to effects on specific disorders. Nutrients. (2025) 17:1353. doi: 10.3390/nu17081353

PubMed Abstract | Crossref Full Text | Google Scholar

130. Zhang Y, Zhang W, Zhao Y, Peng R, Zhang Z, Xu Z, et al. Bioactive sulforaphane from cruciferous vegetables: advances in biosynthesis, metabolism, bioavailability, delivery, health benefits, and applications. Crit Rev Food Sci Nutr. (2025) 65:3027–47. doi: 10.1080/10408398.2024.2354937

PubMed Abstract | Crossref Full Text | Google Scholar

131. Subedi L, Lee JH, Yumnam S, Ji E, Kim SY. Anti-inflammatory effect of sulforaphane on LPS-activated microglia potentially through JNK/AP-1/NF-kappaB inhibition and Nrf2/HO-1 activation. Cells. (2019) 8:194. doi: 10.3390/cells8020194

Crossref Full Text | Google Scholar

132. Angeloni C, Malaguti M, Rizzo B, Barbalace MC, Fabbri D, Hrelia S. Neuroprotective effect of sulforaphane against methylglyoxal cytotoxicity. Chem Res Toxicol. (2015) 28:1234–45. doi: 10.1021/acs.chemrestox.5b00067

PubMed Abstract | Crossref Full Text | Google Scholar

133. Soane L, Li Dai W, Fiskum G, Bambrick LL. Sulforaphane protects immature hippocampal neurons against death caused by exposure to hemin or to oxygen and glucose deprivation. J Neurosci Res. (2010) 88:1355–63. doi: 10.1002/jnr.22307

PubMed Abstract | Crossref Full Text | Google Scholar

134. Mao L, Yang T, Li X, Lei X, Sun Y, Zhao Y, et al. Protective effects of sulforaphane in experimental vascular cognitive impairment: contribution of the Nrf2 pathway. J Cereb Blood Flow Metab. (2019) 39:352–66. doi: 10.1177/0271678X18764083

PubMed Abstract | Crossref Full Text | Google Scholar

135. Sajja RK, Kaisar MA, Vijay V, Desai VG, Prasad S, Cucullo L. In vitro modulation of redox and metabolism interplay at the brain vascular endothelium: genomic and proteomic profiles of sulforaphane activity. Sci Rep. (2018) 8:12708. doi: 10.1038/s41598-018-31137-7

PubMed Abstract | Crossref Full Text | Google Scholar

136. Sun Y, Yang T, Mao L, Zhang F. Sulforaphane protects against brain diseases: roles of cytoprotective enzymes. Austin J Cerebrovasc Dis Stroke. (2017) 4:1054. doi: 10.26420/austinjcerebrovascdisstroke.2017.1054

PubMed Abstract | Crossref Full Text | Google Scholar

137. Gonzalez-Fraguela ME, Blanco L, Fernandez CI, Lorigados L, Serrano T, Fernandez JL. Glutathione depletion: starting point of brain metabolic stress, neuroinflammation and cognitive impairment in rats. Brain Res Bull. (2018) 137:120–31. doi: 10.1016/j.brainresbull.2017.11.015

PubMed Abstract | Crossref Full Text | Google Scholar

138. Hou TT, Yang HY, Wang W, Wu QQ, Tian YR, Jia JP. Sulforaphane inhibits the generation of amyloid-beta oligomer and promotes spatial learning and memory in Alzheimer's disease (PS1V97L) transgenic mice. J Alzheimers Dis. (2018) 62:1803–13. doi: 10.3233/JAD-171110

Crossref Full Text | Google Scholar

139. Youn K, Yoon JH, Lee N, Lim G, Lee J, Sang S, et al. Discovery of sulforaphane as a potent BACE1 inhibitor based on kinetics and computational studies. Nutrients. (2020) 12:3026. doi: 10.3390/nu12103026

PubMed Abstract | Crossref Full Text | Google Scholar

140. Tufekci KU, Ercan I, Isci KB, Olcum M, Tastan B, Gonul CP, et al. Sulforaphane inhibits NLRP3 inflammasome activation in microglia through Nrf2-mediated miRNA alteration. Immunol Lett. (2021) 233:20–30. doi: 10.1016/j.imlet.2021.03.004

PubMed Abstract | Crossref Full Text | Google Scholar

141. Vu GH, Nguyen HD. Molecular mechanisms of sulforaphane in Alzheimer's disease: insights from an in-silico study. In Silico Pharmacol. (2024) 12:96. doi: 10.1007/s40203-024-00267-4

PubMed Abstract | Crossref Full Text | Google Scholar

142. Estfanous S, Daily KP, Eltobgy M, Deems NP, Anne MNK, Krause K, et al. Elevated expression of MiR-17 in microglia of Alzheimer's disease patients abrogates autophagy-mediated amyloid-beta degradation. Front Immunol. (2021) 12:705581. doi: 10.3389/fimmu.2021.705581

Crossref Full Text | Google Scholar

143. Kim YJ, Kim SH, Park Y, Park J, Lee JH, Kim BC, et al. miR-16-5p is upregulated by amyloid beta deposition in Alzheimer's disease models and induces neuronal cell apoptosis through direct targeting and suppression of BCL-2. Exp Gerontol. (2020) 136:110954. doi: 10.1016/j.exger.2020.110954

Crossref Full Text | Google Scholar

144. Denk J, Oberhauser F, Kornhuber J, Wiltfang J, Fassbender K, Schroeter ML, et al. Specific serum and CSF microRNA profiles distinguish sporadic behavioural variant of frontotemporal dementia compared with Alzheimer patients and cognitively healthy controls. PLoS ONE. (2018) 13:e0197329. doi: 10.1371/journal.pone.0197329

PubMed Abstract | Crossref Full Text | Google Scholar

145. Liu Y, Xin H, Zhang Y, Che F, Shen N, Cui Y. Leaves, seeds and exocarp of Ginkgo biloba L. (Ginkgoaceae): a Comprehensive Review of Traditional Uses, phytochemistry, pharmacology, resource utilization and toxicity. J Ethnopharmacol. (2022) 298:115645. doi: 10.1016/j.jep.2022.115645

Crossref Full Text | Google Scholar

146. Kulic Z, Lehner MD, Dietz GPH. Ginkgo biloba leaf extract EGb 761((R)) as a paragon of the product by process concept. Front Pharmacol. (2022) 13:1007746. doi: 10.3389/fphar.2022.1007746

Crossref Full Text | Google Scholar

147. Su X, Shi R, Hu H, Hu L, Wei Q, Guan Y, et al. Medicinal values and potential risks evaluation of ginkgo biloba leaf extract (GBE) drinks made from the leaves in autumn as dietary supplements. Molecules. (2022) 27:7479. doi: 10.3390/molecules27217479

PubMed Abstract | Crossref Full Text | Google Scholar

148. Rusak G, Vujcic Bok V, Sola I, Niksa E, Males Z. Effect of protein, carbohydrate, and oil on phytochemical bioaccessibility and bioactivities of the Ginkgo biloba L. leaf formulations after in vitro digestion. Molecules. (2024) 29:5300. doi: 10.3390/molecules29225300

PubMed Abstract | Crossref Full Text | Google Scholar

149. Morato X, Marquie M, Tartari JP, Lafuente A, Abdelnour C, Alegret M, et al. A randomized, open-label clinical trial in mild cognitive impairment with EGb 761 examining blood markers of inflammation and oxidative stress. Sci Rep. (2023) 13:5406. doi: 10.1038/s41598-023-32515-6

PubMed Abstract | Crossref Full Text | Google Scholar

150. Noor ET, Das R, Lami MS, Chakraborty AJ, Mitra S, Tallei TE, et al. Ginkgo biloba: a treasure of functional phytochemicals with multimedicinal applications. Evid Based Complement Alternat Med. (2022) 2022:8288818. doi: 10.1155/2022/8288818

PubMed Abstract | Crossref Full Text | Google Scholar

151. Morato X, Tartari JP, Pytel V, Boada M. Pharmacodynamic and clinical effects of Ginkgo biloba extract EGb 761 and its phytochemical components in Alzheimer's disease. J Alzheimers Dis. (2024) 101:S285–S98. doi: 10.3233/JAD-231372

PubMed Abstract | Crossref Full Text | Google Scholar

152. Liu Q, Wang J, Gu Z, Ouyang T, Gao H, Kan H, et al. Comprehensive exploration of the neuroprotective mechanisms of Ginkgo biloba leaves in treating neurological disorders. Am J Chin Med. (2024) 52:1053–86. doi: 10.1142/S0192415X24500435

PubMed Abstract | Crossref Full Text | Google Scholar

153. Yoshikawa T, Naito Y, Kondo M. Ginkgo biloba leaf extract: review of biological actions and clinical applications. Antioxid Redox Signal. (1999) 1:469–80. doi: 10.1089/ars.1999.1.4-469

PubMed Abstract | Crossref Full Text | Google Scholar

154. Augustin S, Rimbach G, Augustin K, Schliebs R, Wolffram S, Cermak R. Effect of a short- and long-term treatment with Ginkgo biloba extract on amyloid precursor protein levels in a transgenic mouse model relevant to Alzheimer's disease. Arch Biochem Biophys. (2009) 481:177–82. doi: 10.1016/j.abb.2008.10.032

PubMed Abstract | Crossref Full Text | Google Scholar

155. Yoshitake T, Yoshitake S, Kehr J. The Ginkgo biloba extract EGb 761(R) and its main constituent flavonoids and ginkgolides increase extracellular dopamine levels in the rat prefrontal cortex. Br J Pharmacol. (2010) 159:659–68. doi: 10.1111/j.1476-5381.2009.00580.x

PubMed Abstract | Crossref Full Text | Google Scholar

156. Bridi R, Crossetti FP, Steffen VM, Henriques AT. The antioxidant activity of standardized extract of Ginkgo biloba (EGb 761) in rats. Phytother Res. (2001) 15:449–51. doi: 10.1002/ptr.814

PubMed Abstract | Crossref Full Text | Google Scholar

157. Wan W, Zhang C, Danielsen M, Li Q, Chen W, Chan Y, et al. EGb761 improves cognitive function and regulates inflammatory responses in the APP/PS1 mouse. Exp Gerontol. (2016) 81:92–100. doi: 10.1016/j.exger.2016.05.007

PubMed Abstract | Crossref Full Text | Google Scholar

158. Mazza M, Capuano A, Bria P, Mazza S. Ginkgo biloba and donepezil: a comparison in the treatment of Alzheimer's dementia in a randomized placebo-controlled double-blind study. Eur J Neurol. (2006) 13:981–5. doi: 10.1111/j.1468-1331.2006.01409.x

PubMed Abstract | Crossref Full Text | Google Scholar

159. DeKosky ST, Williamson JD, Fitzpatrick AL, Kronmal RA, Ives DG, Saxton JA, et al. Ginkgo biloba for prevention of dementia: a randomized controlled trial. JAMA. (2008) 300:2253–62. doi: 10.1001/jama.2008.683

PubMed Abstract | Crossref Full Text | Google Scholar

160. Birks J, Grimley Evans J. Ginkgo biloba for cognitive impairment and dementia. Cochrane Database Syst Rev. 2009:CD003120. doi: 10.1002/14651858.CD003120.pub3

PubMed Abstract | Crossref Full Text | Google Scholar

161. Zhang GJ, Zheng D, Yu H, Luo XP, Wu W. Ginkgo biloba extract ameliorates scopolamine-induced memory deficits via rescuing synaptic damage. Curr Med Sci. (2022) 42:474–82. doi: 10.1007/s11596-022-2582-8

PubMed Abstract | Crossref Full Text | Google Scholar

162. Chimakurthy AK, Lingam S, Pasya SKR, Copeland BJ. A systematic review of dietary supplements in Alzheimer's disease. Cureus. (2023) 15:e33982. doi: 10.7759/cureus.33982

PubMed Abstract | Crossref Full Text | Google Scholar

163. Mucha P, Kus F, Cysewski D, Smolenski RT, Tomczyk M. Vitamin B metabolism: a network of multi-protein mediated processes. Int J Mol Sci. (2024) 25:8021. doi: 10.20944/preprints202406.1262.v1

Crossref Full Text | Google Scholar

164. Pardo-Cabello AJ, Manzano-Gamero V, Puche-Canas E. Vitamin B12: for more than just the treatment of megaloblastic anemia? Rev Clin Esp. (2023) 223:114–9. doi: 10.1016/j.rceng.2022.11.004

PubMed Abstract | Crossref Full Text | Google Scholar

165. Temova Rakusa Z, Roskar R, Hickey N, Geremia S. Vitamin B in foods, food supplements, and medicines-a review of its role and properties with a focus on its stability. Molecules. (2022) 28:240. doi: 10.3390/molecules28010240

Crossref Full Text | Google Scholar

166. Alruwaili M, Basri R, AlRuwaili R, Albarrak AM, Ali NH. Neurological implications of vitamin B12 deficiency in diet: a systematic review and meta-analysis. Healthcare. (2023) 11:958. doi: 10.3390/healthcare11070958

PubMed Abstract | Crossref Full Text | Google Scholar

167. Zhang A, Ackley BD, Yan D. Vitamin B12 regulates glial migration and synapse formation through isoform-specific control of PTP-3/LAR PRTP expression. Cell Rep. (2020) 30:3981–8. e3. doi: 10.1016/j.celrep.2020.02.113

PubMed Abstract | Crossref Full Text | Google Scholar

168. Calderon-Ospina CA, Nava-Mesa MO, B. Vitamins in the nervous system: Current knowledge of the biochemical modes of action and synergies of thiamine, pyridoxine, and cobalamin. CNS Neurosci Ther. (2020) 26:5–13. doi: 10.1111/cns.13207

Crossref Full Text | Google Scholar

169. Beaudry-Richard A, Abdelhak A, Saloner R, Sacco S, Montes SC, Oertel FC, et al. Vitamin B12 Levels association with functional and structural biomarkers of central nervous system injury in older adults. Ann Neurol. (2025) 97:1190–204. doi: 10.1002/ana.27200

PubMed Abstract | Crossref Full Text | Google Scholar

170. Lauer AA, Grimm HS, Apel B, Golobrodska N, Kruse L, Ratanski E, et al. Mechanistic Link between vitamin B12 and Alzheimer's disease. Biomolecules. (2022) 12:129. doi: 10.3390/biom12010129

PubMed Abstract | Crossref Full Text | Google Scholar

171. Zhong L, Zhou J, Chen X, Lou Y, Liu D, Zou X, et al. Quantitative proteomics study of the neuroprotective effects of B12 on hydrogen peroxide-induced apoptosis in SH-SY5Y cells. Sci Rep. (2016) 6:22635. doi: 10.1038/srep22635

PubMed Abstract | Crossref Full Text | Google Scholar

172. Kerek R, Geoffroy A, Bison A, Martin N, Akchiche N, Pourie G, et al. Early methyl donor deficiency may induce persistent brain defects by reducing Stat3 signaling targeted by miR-124. Cell Death Dis. (2013) 4:e755. doi: 10.1038/cddis.2013.278

PubMed Abstract | Crossref Full Text | Google Scholar

173. Mehrdad J, Leila E, Emsehgol N. The effect of vitamin B12 on synaptic plasticity of hippocampus in Alzheimer's disease model rats. Int J Neurosci. (2023) 133:654–9. doi: 10.1080/00207454.2021.1962863

PubMed Abstract | Crossref Full Text | Google Scholar

174. Jatoi S, Hafeez A, Riaz SU, Ali A, Ghauri MI, Zehra M. Low vitamin B12 levels: an underestimated cause of minimal cognitive impairment and dementia. Cureus. (2020) 12:e6976. doi: 10.7759/cureus.6976

PubMed Abstract | Crossref Full Text | Google Scholar

175. Cho HS, Huang LK, Lee YT, Chan L, Hong CT. Suboptimal baseline serum vitamin B12 is associated with cognitive decline in people with Alzheimer's disease undergoing cholinesterase inhibitor treatment. Front Neurol. (2018) 9:325. doi: 10.3389/fneur.2018.00325

PubMed Abstract | Crossref Full Text | Google Scholar

176. An Y, Feng L, Zhang X, Wang Y, Wang Y, Tao L, et al. Dietary intakes and biomarker patterns of folate, vitamin B, and vitamin B can be associated with cognitive impairment by hypermethylation of redox-related genes NUDT15 and TXNRD1. Clin Epigenetics. (2019) 11:139. doi: 10.1186/s13148-019-0741-y

Crossref Full Text | Google Scholar

177. Aisen PS, Schneider LS, Sano M, Diaz-Arrastia R, van Dyck CH, Weiner MF, et al. High-dose B vitamin supplementation and cognitive decline in Alzheimer disease: a randomized controlled trial. JAMA. (2008) 300:1774–83. doi: 10.1001/jama.300.15.1774

PubMed Abstract | Crossref Full Text | Google Scholar

178. Nguyen HD, Kim MS. The role of mixed B vitamin intakes on cognitive performance: Modeling, genes and miRNAs involved. J Psychiatr Res. (2022) 152:38–56. doi: 10.1016/j.jpsychires.2022.06.006

PubMed Abstract | Crossref Full Text | Google Scholar

179. Cerullo G, Negro M, Parimbelli M, Pecoraro M, Perna S, Liguori G, et al. The long history of vitamin C: from prevention of the common cold to potential aid in the treatment of COVID-19. Front Immunol. (2020) 11:574029. doi: 10.3389/fimmu.2020.574029

PubMed Abstract | Crossref Full Text | Google Scholar

180. Dosedel M, Jirkovsky E, Macakova K, Krcmova LK, Javorska L, Pourova J, et al. Vitamin C-sources, physiological role, kinetics, deficiency, use, toxicity, and determination. Nutrients. (2021) 13:615. doi: 10.3390/nu13020615

PubMed Abstract | Crossref Full Text | Google Scholar

181. Lykkesfeldt J, Carr AC, Tveden-Nyborg P. The pharmacology of vitamin C. Pharmacol Rev. (2025) 77:100043. doi: 10.1016/j.pharmr.2025.100043

Crossref Full Text | Google Scholar

182. Travica N, Ried K, Hudson I, Sali A, Scholey A, Pipingas A. The contribution of plasma and brain vitamin C on age and gender-related cognitive differences: a mini-review of the literature. Front Integr Neurosci. (2020) 14:47. doi: 10.3389/fnint.2020.00047

PubMed Abstract | Crossref Full Text | Google Scholar

183. Zylinska L, Lisek M, Guo F, Boczek T. Vitamin C modes of action in calcium-involved signaling in the brain. Antioxidants (Basel). (2023) 12. doi: 10.3390/antiox12020231

PubMed Abstract | Crossref Full Text | Google Scholar

184. Sharma Y, Popescu A, Horwood C, Hakendorf P, Thompson C. Relationship between vitamin C deficiency and cognitive impairment in older hospitalised patients: a cross-sectional study. Antioxidants. (2022) 11:463. doi: 10.3390/antiox11030463

PubMed Abstract | Crossref Full Text | Google Scholar

185. Huang J, May JM. Ascorbic acid protects SH-SY5Y neuroblastoma cells from apoptosis and death induced by beta-amyloid. Brain Res. (2006) 1097:52–8. doi: 10.1016/j.brainres.2006.04.047

PubMed Abstract | Crossref Full Text | Google Scholar

186. Dixit S, Bernardo A, Walker JM, Kennard JA, Kim GY, Kessler ES, et al. Vitamin C deficiency in the brain impairs cognition, increases amyloid accumulation and deposition, and oxidative stress in APP/PSEN1 and normally aging mice. ACS Chem Neurosci. (2015) 6:570–81. doi: 10.1021/cn500308h

PubMed Abstract | Crossref Full Text | Google Scholar

187. Harrison FE, May JM, McDonald MP. Vitamin C deficiency increases basal exploratory activity but decreases scopolamine-induced activity in APP/PSEN1 transgenic mice. Pharmacol Biochem Behav. (2010) 94:543–52. doi: 10.1016/j.pbb.2009.11.009

PubMed Abstract | Crossref Full Text | Google Scholar

188. Consoli DC, Brady LJ, Bowman AB, Calipari ES, Harrison FE. Ascorbate deficiency decreases dopamine release in gulo(-/-) and APP/PSEN1 mice. J Neurochem. (2021) 157:656–65. doi: 10.1111/jnc.15151

PubMed Abstract | Crossref Full Text | Google Scholar

189. Rharass T, Lantow M, Gbankoto A, Weiss DG, Panakova D, Lucas S. Ascorbic acid alters cell fate commitment of human neural progenitors in a WNT/beta-catenin/ROS signaling dependent manner. J Biomed Sci. (2017) 24:78. doi: 10.1186/s12929-017-0385-1

Crossref Full Text | Google Scholar

190. Majewska MD, Bell JA. Ascorbic acid protects neurons from injury induced by glutamate and NMDA. Neuroreport. (1990) 1:194–6. doi: 10.1097/00001756-199011000-00004

PubMed Abstract | Crossref Full Text | Google Scholar

191. Murakami K, Murata N, Ozawa Y, Kinoshita N, Irie K, Shirasawa T, et al. Vitamin C restores behavioral deficits and amyloid-beta oligomerization without affecting plaque formation in a mouse model of Alzheimer's disease. J Alzheimers Dis. (2011) 26:7–18. doi: 10.3233/JAD-2011-101971

Crossref Full Text | Google Scholar

192. Shah SA, Yoon GH, Kim HO, Kim MO. Vitamin C neuroprotection against dose-dependent glutamate-induced neurodegeneration in the postnatal brain. Neurochem Res. (2015) 40:875–84. doi: 10.1007/s11064-015-1540-2

PubMed Abstract | Crossref Full Text | Google Scholar

193. Sim M, Hong S, Jung S, Kim JS, Goo YT, Chun WY, et al. Vitamin C supplementation promotes mental vitality in healthy young adults: results from a cross-sectional analysis and a randomized, double-blind, placebo-controlled trial. Eur J Nutr. (2022) 61:447–59. doi: 10.1007/s00394-021-02656-3

PubMed Abstract | Crossref Full Text | Google Scholar

194. Smith AP, Clark RE, Nutt DJ, Haller J, Hayward SG, Perry K. Vitamin C, mood and cognitive functioning in the elderly. Nutr Neurosci. (1999) 2:249–56. doi: 10.1080/1028415X.1999.11747281

PubMed Abstract | Crossref Full Text | Google Scholar

195. Travica N, Ried K, Sali A, Hudson I, Scholey A, Pipingas A. Plasma vitamin C concentrations and cognitive function: a cross-sectional study. Front Aging Neurosci. (2019) 11:72. doi: 10.3389/fnagi.2019.00072

PubMed Abstract | Crossref Full Text | Google Scholar

196. Travica N, Ried K, Sali A, Scholey A, Hudson I, Pipingas A. Vitamin C status and cognitive function: a systematic review. Nutrients. (2017) 9:960. doi: 10.3390/nu9090960

PubMed Abstract | Crossref Full Text | Google Scholar

197. Kim SM, Lim SM, Yoo JA, Woo MJ, Cho KH. Consumption of high-dose vitamin C (1250 mg per day) enhances functional and structural properties of serum lipoprotein to improve anti-oxidant, anti-atherosclerotic, and anti-aging effects via regulation of anti-inflammatory microRNA. Food Funct. (2015) 6:3604–12. doi: 10.1039/C5FO00738K

PubMed Abstract | Crossref Full Text | Google Scholar

198. Uttara B, Singh AV, Zamboni P, Mahajan RT. Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol. (2009) 7:65–74. doi: 10.2174/157015909787602823

PubMed Abstract | Crossref Full Text | Google Scholar

199. Hamid M, Mansoor S, Amber S, Zahid S. A quantitative meta-analysis of vitamin C in the pathophysiology of Alzheimer's disease. Front Aging Neurosci. (2022) 14:970263. doi: 10.3389/fnagi.2022.970263

PubMed Abstract | Crossref Full Text | Google Scholar

200. Kaye AD, Thomassen AS, Mashaw SA, MacDonald EM, Waguespack A, Hickey L, et al. Vitamin E (alpha-Tocopherol): emerging clinical role and adverse risks of supplementation in adults. Cureus. (2025) 17:e78679. doi: 10.7759/cureus.78679

Crossref Full Text | Google Scholar

201. Mohd Zaffarin AS, Ng SF, Ng MH, Hassan H, Alias E. Pharmacology and pharmacokinetics of vitamin E: nanoformulations to enhance bioavailability. Int J Nanomedicine. (2020) 15:9961–74. doi: 10.2147/IJN.S276355

PubMed Abstract | Crossref Full Text | Google Scholar

202. Kiyose C. Absorption, transportation, and distribution of vitamin E homologs. Free Radic Biol Med. (2021) 177:226–37. doi: 10.1016/j.freeradbiomed.2021.10.016

PubMed Abstract | Crossref Full Text | Google Scholar

203. Numakawa Y, Numakawa T, Matsumoto T, Yagasaki Y, Kumamaru E, Kunugi H, et al. Vitamin E protected cultured cortical neurons from oxidative stress-induced cell death through the activation of mitogen-activated protein kinase and phosphatidylinositol 3-kinase. J Neurochem. (2006) 97:1191–202. doi: 10.1111/j.1471-4159.2006.03827.x

PubMed Abstract | Crossref Full Text | Google Scholar

204. Behl C. Vitamin E protects neurons against oxidative cell death in vitro more effectively than 17-beta estradiol and induces the activity of the transcription factor NF-kappaB. J Neural Transm. (2000) 107:393–407. doi: 10.1007/s007020070082

PubMed Abstract | Crossref Full Text | Google Scholar

205. Ezaki Y, Nishihara E, Shibata Y, Matsuo T, Kitagawa N, Nagata I, et al. Vitamin E prevents the neuronal cell death by repressing cyclooxygenase-2 activity. Neuroreport. (2005) 16:1163–7. doi: 10.1097/00001756-200508010-00006

PubMed Abstract | Crossref Full Text | Google Scholar

206. Behl C, Davis J, Cole GM, Schubert D. Vitamin E protects nerve cells from amyloid beta protein toxicity. Biochem Biophys Res Commun. (1992) 186:944–50. doi: 10.1016/0006-291X(92)90837-B

PubMed Abstract | Crossref Full Text | Google Scholar

207. Li Y, Liu L, Barger SW, Mrak RE, Griffin WS. Vitamin E suppression of microglial activation is neuroprotective. J Neurosci Res. (2001) 66:163–70. doi: 10.1002/jnr.1208

PubMed Abstract | Crossref Full Text | Google Scholar

208. Molavi Vasei F, Zamanian MY, Golmohammadi M, Mahmoodi M, Khademalhosseini M, Tavakoli T, et al. The impact of vitamin E supplementation on oxidative stress, cognitive functions, and aging-related gene expression in aged mice. Food Sci Nutr. (2024) 12:9834–45. doi: 10.1002/fsn3.4548

PubMed Abstract | Crossref Full Text | Google Scholar

209. Veinbergs I, Mallory M, Sagara Y, Masliah E. Vitamin E supplementation prevents spatial learning deficits and dendritic alterations in aged apolipoprotein E-deficient mice. Eur J Neurosci. (2000) 12:4541–6. doi: 10.1046/j.1460-9568.2000.01308.x

PubMed Abstract | Crossref Full Text | Google Scholar

210. Aiguo W, Zhe Y, Gomez-Pinilla F. Vitamin E protects against oxidative damage and learning disability after mild traumatic brain injury in rats. Neurorehabil Neural Repair. (2010) 24:290–8. doi: 10.1177/1545968309348318

PubMed Abstract | Crossref Full Text | Google Scholar

211. Dysken MW, Sano M, Asthana S, Vertrees JE, Pallaki M, Llorente M, et al. Effect of vitamin E and memantine on functional decline in Alzheimer disease: the TEAM-AD VA cooperative randomized trial. JAMA. (2014) 311:33–44. doi: 10.1001/jama.2013.282834

PubMed Abstract | Crossref Full Text | Google Scholar

212. Wang W, Li J, Zhang H, Wang X, Zhang X. Effects of vitamin E supplementation on the risk and progression of AD: a systematic review and meta-analysis. Nutr Neurosci. (2021) 24:13–22. doi: 10.1080/1028415X.2019.1585506

PubMed Abstract | Crossref Full Text | Google Scholar

213. Farina N, Isaac MG, Clark AR, Rusted J, Tabet N. Vitamin E for Alzheimer's dementia and mild cognitive impairment. Cochrane Database Syst Rev. (2012) 11:CD002854. doi: 10.1002/14651858.CD002854.pub3

PubMed Abstract | Crossref Full Text | Google Scholar

214. Boccardi V, Poli G, Cecchetti R, Bastiani P, Scamosci M, Febo M, et al. miRNAs and Alzheimer's disease: exploring the role of inflammation and vitamin E in an old-age population. Nutrients. (2023) 15:634. doi: 10.3390/nu15030634

PubMed Abstract | Crossref Full Text | Google Scholar

215. De Biase L, Marsili P, Salerni E, D'Aroma A, D'Aurizio C, D'Andrea F, et al. [Electrocardiographic aspects of phases of relapse in patients with multiple sclerosis]. Cardiologia. (1987) 32:753–8.

Google Scholar

216. Nagaria TD, Shinde RK, Shukla S, Acharya S, Acharya N, Jogdand SD. The Sunlight-Vitamin D Connection: Implications for Patient Outcomes in the Surgical Intensive Care Unit. Cureus. (2023) 15:e46819. doi: 10.7759/cureus.46819

PubMed Abstract | Crossref Full Text | Google Scholar

217. Dominguez LJ, Farruggia M, Veronese N, Barbagallo M. Vitamin D sources, Metabolism, and deficiency: available compounds and guidelines for its treatment. Metabolites. (2021) 11:255. doi: 10.3390/metabo11040255

PubMed Abstract | Crossref Full Text | Google Scholar

218. Pasidi E, Vareltzis P. Vitamin D bioaccessibility from supplements and foods-gastric pH Effect using a static in vitro gastrointestinal model. Molecules. (2024) 29:1153. doi: 10.20944/preprints202402.1468.v1

Crossref Full Text | Google Scholar

219. Kang JH, Vyas CM, Okereke OI, Ogata S, Albert M, Lee IM, et al. Effect of vitamin D on cognitive decline: results from two ancillary studies of the VITAL randomized trial. Sci Rep. (2021) 11:23253. doi: 10.1038/s41598-021-02485-8

PubMed Abstract | Crossref Full Text | Google Scholar

220. Littlejohns TJ, Henley WE, Lang IA, Annweiler C, Beauchet O, Chaves PH, et al. Vitamin D and the risk of dementia and Alzheimer disease. Neurology. (2014) 83:920–8. doi: 10.1212/WNL.0000000000000755

PubMed Abstract | Crossref Full Text | Google Scholar

221. Mizwicki MT, Menegaz D, Zhang J, Barrientos-Duran A, Tse S, Cashman JR, et al. Genomic and nongenomic signaling induced by 1alpha,25(OH)2-vitamin D3 promotes the recovery of amyloid-beta phagocytosis by Alzheimer's disease macrophages. J Alzheimers Dis. (2012) 29:51–62. doi: 10.3233/JAD-2012-110560

Crossref Full Text | Google Scholar

222. Patel P, Shah J. Vitamin D supplementation ameliorates cognitive impairment and alters neurodegenerative and inflammatory markers in scopolamine induced rat model. Metab Brain Dis. (2022) 37:2653–67. doi: 10.1007/s11011-022-01086-2

Crossref Full Text | Google Scholar

223. Shirazi HA, Rasouli J, Ciric B, Rostami A, Zhang GX. 1,25-Dihydroxyvitamin D3 enhances neural stem cell proliferation and oligodendrocyte differentiation. Exp Mol Pathol. (2015) 98:240–5. doi: 10.1016/j.yexmp.2015.02.004

PubMed Abstract | Crossref Full Text | Google Scholar

224. Jia J, Hu J, Huo X, Miao R, Zhang Y, Ma F. Effects of vitamin D supplementation on cognitive function and blood Abeta-related biomarkers in older adults with Alzheimer's disease: a randomised, double-blind, placebo-controlled trial. J Neurol Neurosurg Psychiatry. (2019) 90:1347–52. doi: 10.1136/jnnp-2018-320199

Crossref Full Text | Google Scholar

225. Annweiler C, Fantino B, Schott AM, Krolak-Salmon P, Allali G, Beauchet O. Vitamin D insufficiency and mild cognitive impairment: cross-sectional association. Eur J Neurol. (2012) 19:1023–9. doi: 10.1111/j.1468-1331.2012.03675.x

PubMed Abstract | Crossref Full Text | Google Scholar

226. Du Y, Liang F, Zhang L, Liu J, Dou H. Vitamin D supplement for prevention of Alzheimer's disease: a systematic review and meta-analysis. Am J Ther. (2020) 28:e638–e48. doi: 10.1097/MJT.0000000000001302

PubMed Abstract | Crossref Full Text | Google Scholar

227. Chakkera M, Ravi N, Ramaraju R, Vats A, Nair AR, Bandhu AK, et al. The efficacy of vitamin D supplementation in patients with Alzheimer's disease in preventing cognitive decline: a systematic review. Cureus. (2022) 14:e31710. doi: 10.7759/cureus.31710

PubMed Abstract | Crossref Full Text | Google Scholar

228. Sinn N, Milte CM, Street SJ, Buckley JD, Coates AM, Petkov J, et al. Effects of n-3 fatty acids, EPA v. DHA, on depressive symptoms, quality of life, memory and executive function in older adults with mild cognitive impairment: a 6-month randomised controlled trial. Br J Nutr. (2012) 107:1682–93. doi: 10.1017/S0007114511004788

PubMed Abstract | Crossref Full Text | Google Scholar

229. Dighriri IM, Alsubaie AM, Hakami FM, Hamithi DM, Alshekh MM, Khobrani FA, et al. Effects of omega-3 polyunsaturated fatty acids on brain functions: a systematic review. Cureus. (2022) 14:e30091. doi: 10.7759/cureus.30091

PubMed Abstract | Crossref Full Text | Google Scholar

230. Van Dael P. Role of n-3 long-chain polyunsaturated fatty acids in human nutrition and health: review of recent studies and recommendations. Nutr Res Pract. (2021) 15:137–59. doi: 10.4162/nrp.2021.15.2.137

PubMed Abstract | Crossref Full Text | Google Scholar

231. Mariamenatu AH, Abdu EM. Overconsumption of Omega-6 polyunsaturated fatty acids (PUFAs) versus deficiency of omega-3 PUFAs in modern-day diets: the disturbing factor for their “balanced antagonistic metabolic functions” in the human body. J Lipids. (2021) 2021:8848161. doi: 10.1155/2021/8848161

PubMed Abstract | Crossref Full Text | Google Scholar

232. Shahidi F, Ambigaipalan P. Omega-3 polyunsaturated fatty acids and their health benefits. Annu Rev Food Sci Technol. (2018) 9:345–81. doi: 10.1146/annurev-food-111317-095850

PubMed Abstract | Crossref Full Text | Google Scholar

233. Dempsey M, Rockwell MS, Wentz LM. The influence of dietary and supplemental omega-3 fatty acids on the omega-3 index: a scoping review. Front Nutr. (2023) 10:1072653. doi: 10.3389/fnut.2023.1072653

PubMed Abstract | Crossref Full Text | Google Scholar

234. Wei BZ Li L, Dong CW, Tan CC. Alzheimer's Disease Neuroimaging I, Xu W. The relationship of omega-3 fatty acids with dementia and cognitive decline: evidence from prospective cohort studies of supplementation, dietary intake, and blood markers. Am J Clin Nutr. (2023) 117:1096–109. doi: 10.1016/j.ajcnut.2023.04.001

Crossref Full Text | Google Scholar

235. Mett J. The impact of medium chain and polyunsaturated omega-3-fatty acids on amyloid-beta deposition, oxidative stress and metabolic dysfunction associated with Alzheimer's disease. Antioxidants. (2021) 10:1991. doi: 10.3390/antiox10121991

Crossref Full Text | Google Scholar

236. Inoue T, Tanaka M, Masuda S, Ohue-Kitano R, Yamakage H, Muranaka K, et al. Omega-3 polyunsaturated fatty acids suppress the inflammatory responses of lipopolysaccharide-stimulated mouse microglia by activating SIRT1 pathways. Biochim Biophys Acta Mol Cell Biol Lipids. (2017) 1862:552–60. doi: 10.1016/j.bbalip.2017.02.010

PubMed Abstract | Crossref Full Text | Google Scholar

237. Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, et al. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell. (2010) 142:687–98. doi: 10.1016/j.cell.2010.07.041

PubMed Abstract | Crossref Full Text | Google Scholar

238. Nakajima S, Demers G, Machuca-Parra AI, Pour ZD, Bairamian D, Bouyakdan K, et al. Central activation of the fatty acid sensor GPR120 suppresses microglia reactivity and alleviates sickness- and anxiety-like behaviors. J Neuroinflammation. (2023) 20:302. doi: 10.1186/s12974-023-02978-5

PubMed Abstract | Crossref Full Text | Google Scholar

239. Zhao Y, Calon F, Julien C, Winkler JW, Petasis NA, Lukiw WJ, et al. Docosahexaenoic acid-derived neuroprotectin D1 induces neuronal survival via secretase- and PPARgamma-mediated mechanisms in Alzheimer's disease models. PLoS ONE. (2011) 6:e15816. doi: 10.1371/journal.pone.0015816

Crossref Full Text | Google Scholar

240. Lukiw WJ, Cui JG, Marcheselli VL, Bodker M, Botkjaer A, Gotlinger K, et al. A role for docosahexaenoic acid-derived neuroprotectin D1 in neural cell survival and Alzheimer disease. J Clin Invest. (2005) 115:2774–83. doi: 10.1172/JCI25420

PubMed Abstract | Crossref Full Text | Google Scholar

241. Wang YW, Li Q, Li XY, Zhao YC, Wang CC, Xue CH, et al. A Comparative study about the neuroprotective effects of DHA-enriched phosphatidylserine and EPA-enriched phosphatidylserine against oxidative damage in primary hippocampal neurons. Mar Drugs. (2023) 21:410. doi: 10.3390/md21070410

PubMed Abstract | Crossref Full Text | Google Scholar

242. Mita T, Mayanagi T, Ichijo H, Fukumoto K, Otsuka K, Sakai A, et al. Docosahexaenoic acid promotes axon outgrowth by translational regulation of tau and collapsin response mediator protein 2 expression. J Biol Chem. (2016) 291:4955–65. doi: 10.1074/jbc.M115.693499

PubMed Abstract | Crossref Full Text | Google Scholar

243. Cole AR, Knebel A, Morrice NA, Robertson LA, Irving AJ, Connolly CN, et al. GSK-3 phosphorylation of the Alzheimer epitope within collapsin response mediator proteins regulates axon elongation in primary neurons. J Biol Chem. (2004) 279:50176–80. doi: 10.1074/jbc.C400412200

PubMed Abstract | Crossref Full Text | Google Scholar

244. Liu H, Zhou L, Yi P, Zhan F, Zhou L, Dong Y, et al. ω3-PUFA alleviates neuroinflammation by upregulating miR-107 targeting PIEZO1/NFkappaB p65. Int Immunopharmacol. (2024) 132:111996. doi: 10.1016/j.intimp.2024.111996

Crossref Full Text | Google Scholar

245. Pan JP, Xie JL, Huang LY, Wu QZ, Tang DF, Jin Q, et al. miR-29a-3p promotes the regulatory role of eicosapentaenoic acid in the NLRP3 inflammasome and autophagy in microglial cells. Kaohsiung J Med Sci. (2023) 39:565–75. doi: 10.1002/kjm2.12670

PubMed Abstract | Crossref Full Text | Google Scholar

246. Bottero V, Potashkin JA. A comparison of gene expression changes in the blood of individuals consuming diets supplemented with olives, nuts or long-chain omega-3 fatty acids. Nutrients. (2020) 12:3765. doi: 10.3390/nu12123765

PubMed Abstract | Crossref Full Text | Google Scholar

247. Hebert SS, Horre K, Nicolai L, Bergmans B, Papadopoulou AS, Delacourte A, et al. MicroRNA regulation of Alzheimer's Amyloid precursor protein expression. Neurobiol Dis. (2009) 33:422–8. doi: 10.1016/j.nbd.2008.11.009

PubMed Abstract | Crossref Full Text | Google Scholar

248. McGaugh E, Barthel B. A review of ketogenic diet and lifestyle. Mo Med. (2022) 119:84–8.

PubMed Abstract | Google Scholar

249. Yang H, Shan W, Zhu F, Wu J, Wang Q. Ketone bodies in neurological diseases: focus on neuroprotection and underlying mechanisms. Front Neurol. (2019) 10:585. doi: 10.3389/fneur.2019.00585

PubMed Abstract | Crossref Full Text | Google Scholar

250. Jensen NJ, Wodschow HZ, Nilsson M, Rungby J. Effects of ketone bodies on brain metabolism and function in neurodegenerative diseases. Int J Mol Sci. (2020) 21:8767. doi: 10.3390/ijms21228767

PubMed Abstract | Crossref Full Text | Google Scholar

251. Dynka D, Kowalcze K, Paziewska A. The role of ketogenic diet in the treatment of neurological diseases. Nutrients. (2022) 14:5003. doi: 10.3390/nu14235003

PubMed Abstract | Crossref Full Text | Google Scholar

252. Tao Y, Leng SX, Zhang H. Ketogenic diet: an effective treatment approach for neurodegenerative diseases. Curr Neuropharmacol. (2022) 20:2303–19. doi: 10.2174/1570159X20666220830102628

PubMed Abstract | Crossref Full Text | Google Scholar

253. Kim DY, Davis LM, Sullivan PG, Maalouf M, Simeone TA, van Brederode J, et al. Ketone bodies are protective against oxidative stress in neocortical neurons. J Neurochem. (2007) 101:1316–26. doi: 10.1111/j.1471-4159.2007.04483.x

PubMed Abstract | Crossref Full Text | Google Scholar

254. Maalouf M, Sullivan PG, Davis L, Kim DY, Rho JM. Ketones inhibit mitochondrial production of reactive oxygen species production following glutamate excitotoxicity by increasing NADH oxidation. Neuroscience. (2007) 145:256–64. doi: 10.1016/j.neuroscience.2006.11.065

PubMed Abstract | Crossref Full Text | Google Scholar

255. Sullivan PG, Rippy NA, Dorenbos K, Concepcion RC, Agarwal AK, Rho JM. The ketogenic diet increases mitochondrial uncoupling protein levels and activity. Ann Neurol. (2004) 55:576–80. doi: 10.1002/ana.20062

PubMed Abstract | Crossref Full Text | Google Scholar

256. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, et al. Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. (2013) 339:211–4. doi: 10.1126/science.1227166

Crossref Full Text | Google Scholar

257. Yoon JH, Hwang J, Son SU, Choi J, You SW, Park H, et al. How can insulin resistance cause Alzheimer's disease? Int J Mol Sci. (2023) 24:3506. doi: 10.3390/ijms24043506

PubMed Abstract | Crossref Full Text | Google Scholar

258. Koppel SJ, Pei D, Wilkins HM, Weidling IW, Wang X, Menta BW, et al. A ketogenic diet differentially affects neuron and astrocyte transcription. J Neurochem. (2021) 157:1930–45. doi: 10.1111/jnc.15313

PubMed Abstract | Crossref Full Text | Google Scholar

259. Hersant H, Grossberg G. The ketogenic diet and Alzheimer's disease. J Nutr Health Aging. (2022) 26:606–14. doi: 10.1007/s12603-022-1807-7

PubMed Abstract | Crossref Full Text | Google Scholar

260. Taylor MK, Sullivan DK, Swerdlow RH, Vidoni ED, Morris JK, Mahnken JD, et al. A high-glycemic diet is associated with cerebral amyloid burden in cognitively normal older adults. Am J Clin Nutr. (2017) 106:1463–70. doi: 10.3945/ajcn.117.162263

PubMed Abstract | Crossref Full Text | Google Scholar

261. Castellano CA, Nugent S, Paquet N, Tremblay S, Bocti C, Lacombe G, et al. Lower brain 18F-fluorodeoxyglucose uptake but normal 11C-acetoacetate metabolism in mild Alzheimer's disease dementia. J Alzheimers Dis. (2015) 43:1343–53. doi: 10.3233/JAD-141074

PubMed Abstract | Crossref Full Text | Google Scholar

262. Phillips MCL, Deprez LM, Mortimer GMN, Murtagh DKJ, McCoy S, Mylchreest R, et al. Randomized crossover trial of a modified ketogenic diet in Alzheimer's disease. Alzheimers Res Ther. (2021) 13:51. doi: 10.1186/s13195-021-00783-x

PubMed Abstract | Crossref Full Text | Google Scholar

263. Ota M, Matsuo J, Ishida I, Takano H, Yokoi Y, Hori H, et al. Effects of a medium-chain triglyceride-based ketogenic formula on cognitive function in patients with mild-to-moderate Alzheimer's disease. Neurosci Lett. (2019) 690:232–6. doi: 10.1016/j.neulet.2018.10.048

PubMed Abstract | Crossref Full Text | Google Scholar

264. Guasch-Ferre M, Willett WC. The mediterranean diet and health: a comprehensive overview. J Intern Med. (2021) 290:549–66. doi: 10.1111/joim.13333

PubMed Abstract | Crossref Full Text | Google Scholar

265. Nucci D, Sommariva A, Degoni LM, Gallo G, Mancarella M, Natarelli F, et al. Association between mediterranean diet and dementia and Alzheimer disease: a systematic review with meta-analysis. Aging Clin Exp Res. (2024) 36:77. doi: 10.1007/s40520-024-02718-6

PubMed Abstract | Crossref Full Text | Google Scholar

266. Dominguez LJ, Di Bella G, Veronese N, Barbagallo M. Impact of mediterranean diet on chronic non-communicable diseases and longevity. Nutrients. (2021) 13:2028. doi: 10.3390/nu13062028

PubMed Abstract | Crossref Full Text | Google Scholar

267. Stefaniak O, Dobrzynska M, Drzymala-Czyz S, Przyslawski J. Diet in the prevention of Alzheimer's disease: current knowledge and future research requirements. Nutrients. (2022) 14:4564. doi: 10.3390/nu14214564

PubMed Abstract | Crossref Full Text | Google Scholar

268. Chen H, Dhana K, Huang Y, Huang L, Tao Y, Liu X, et al. Association of the mediterranean dietary approaches to stop hypertension intervention for neurodegenerative delay (MIND) diet with the risk of dementia. JAMA Psychiatry. (2023) 80:630–8. doi: 10.1001/jamapsychiatry.2023.0800

PubMed Abstract | Crossref Full Text | Google Scholar

269. Liu X, Morris MC, Dhana K, Ventrelle J, Johnson K, Bishop L, et al. Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) study: Rationale, design and baseline characteristics of a randomized control trial of the MIND diet on cognitive decline. Contemp Clin Trials. (2021) 102:106270. doi: 10.1016/j.cct.2021.106270

PubMed Abstract | Crossref Full Text | Google Scholar

270. Castro-Barquero S, Larroya M, Crispi F, Estruch R, Nakaki A, Paules C, et al. Diet quality and nutrient density in pregnant women according to adherence to Mediterranean diet. Front Public Health. (2023) 11:1144942. doi: 10.3389/fpubh.2023.1144942

PubMed Abstract | Crossref Full Text | Google Scholar

271. Zhu X, Borenstein AR, Zheng Y, Zhang W, Seidner DL, Ness R, et al. Ca:Mg ratio, APOE cytosine modifications, and cognitive function: results from a randomized trial. J Alzheimers Dis. (2020) 75:85–98. doi: 10.3233/JAD-191223

PubMed Abstract | Crossref Full Text | Google Scholar

272. Alam AB, Lutsey PL, Gottesman RF, Tin A, Alonso A. Low serum magnesium is associated with incident dementia in the ARIC-NCS cohort. Nutrients. (2020) 12:3074. doi: 10.3390/nu12103074

PubMed Abstract | Crossref Full Text | Google Scholar

273. Yu J, Sun M, Chen Z, Lu J, Liu Y, Zhou L, et al. Magnesium modulates amyloid-beta protein precursor trafficking and processing. J Alzheimers Dis. (2010) 20:1091–106. doi: 10.3233/JAD-2010-091444

PubMed Abstract | Crossref Full Text | Google Scholar

274. Yu X, Guan PP, Guo JW, Wang Y, Cao LL, Xu GB, et al. By suppressing the expression of anterior pharynx-defective-1alpha and−1beta and inhibiting the aggregation of beta-amyloid protein, magnesium ions inhibit the cognitive decline of amyloid precursor protein/presenilin 1 transgenic mice. FASEB J. (2015) 29:5044–58. doi: 10.1096/fj.15-275578

Crossref Full Text | Google Scholar

275. Xiong Y, Yang Y, Ruan Y, Ou W, Hu Z, Li W, et al. Magnesium-L-threonate ameliorates cognitive deficit by attenuating adult hippocampal neurogenesis impairment in a mouse model of Alzheimer's disease. Exp Neurobiol. (2025) 34:53–62. doi: 10.5607/en24030

PubMed Abstract | Crossref Full Text | Google Scholar

276. Wang D, Fei Z, Luo S, Wang H. MiR-335-5p Inhibits beta-Amyloid (Abeta) accumulation to attenuate cognitive deficits through targeting c-jun-N-terminal kinase 3 in Alzheimer's disease. Curr Neurovasc Res. (2020) 17:93–101. doi: 10.2174/1567202617666200128141938

Crossref Full Text | Google Scholar

277. Li F, Xie XY, Sui XF, Wang P, Chen Z, Zhang JB. Profile of pathogenic proteins and MicroRNAs in plasma-derived extracellular vesicles in Alzheimer's disease: a pilot study. Neuroscience. (2020) 432:240–6. doi: 10.1016/j.neuroscience.2020.02.044

PubMed Abstract | Crossref Full Text | Google Scholar

278. Alamro H, Thafar MA, Albaradei S, Gojobori T, Essack M, Gao X. Exploiting machine learning models to identify novel Alzheimer's disease biomarkers and potential targets. Sci Rep. (2023) 13:4979. doi: 10.1038/s41598-023-30904-5

PubMed Abstract | Crossref Full Text | Google Scholar

279. Yin P, Xue Y, Wang T, Zhong D, Li G. The therapeutic targets of fingolimod (FTY720) are involved in pathological processes in the frontal cortex of Alzheimer's disease patients: a network pharmacology study. Front Aging Neurosci. (2021) 13:609679. doi: 10.3389/fnagi.2021.609679

PubMed Abstract | Crossref Full Text | Google Scholar

280. Chen A, Gibney PA. Dietary trehalose as a bioactive nutrient. Nutrients. (2023) 15:1393. doi: 10.3390/nu15061393

PubMed Abstract | Crossref Full Text | Google Scholar

281. Sharma E, Shruti PS, Singh S, Singh T, Kaur P, Jodha B, et al. Trehalose and its diverse biological potential. Curr Protein Pept Sci. (2023) 24:503–17. doi: 10.2174/1389203724666230606154719

PubMed Abstract | Crossref Full Text | Google Scholar

282. Sevriev B, Dimitrova S, Kehayova G, Dragomanova S. Trehalose: neuroprotective effects and mechanisms-an updated review. Neurosci. (2024) 5:429–44. doi: 10.3390/neurosci5040032

PubMed Abstract | Crossref Full Text | Google Scholar

283. Holler CJ, Taylor G, McEachin ZT, Deng Q, Watkins WJ, Hudson K, et al. Trehalose upregulates progranulin expression in human and mouse models of GRN haploinsufficiency: a novel therapeutic lead to treat frontotemporal dementia. Mol Neurodegener. (2016) 11:46. doi: 10.1186/s13024-016-0114-3

PubMed Abstract | Crossref Full Text | Google Scholar

284. Portbury SD, Hare DJ, Sgambelloni C, Perronnes K, Portbury AJ, Finkelstein DI, et al. Trehalose improves cognition in the transgenic Tg2576 mouse model of Alzheimer's disease. J Alzheimers Dis. (2017) 60:549–60. doi: 10.3233/JAD-170322

PubMed Abstract | Crossref Full Text | Google Scholar

285. Pupyshev AB, Akopyan AA, Tenditnik MV, Ovsyukova MV, Dubrovina NI, Belichenko VM, et al. Alimentary treatment with trehalose in a pharmacological model of Alzheimer's disease in mice: effects of different dosages and treatment regimens. Pharmaceutics. (2024) 16:813. doi: 10.3390/pharmaceutics16060813

PubMed Abstract | Crossref Full Text | Google Scholar

286. Shafiei B, Afgar A, Nematollahi MH, Shabani M, Nazari-Robati M. Effect of trehalose on miR-132 and SIRT1 in the hippocampus of aged rats. Neurosci Lett. (2023) 813:137418. doi: 10.1016/j.neulet.2023.137418

PubMed Abstract | Crossref Full Text | Google Scholar

287. Shafiei B, Shabani M, Afgar A, Rajizadeh MA, Nazari-Robati M. Trehalose attenuates learning and memory impairments in aged rats via overexpression of miR-181c. Neurochem Res. (2022) 47:3309–17. doi: 10.1007/s11064-022-03687-w

PubMed Abstract | Crossref Full Text | Google Scholar

288. Radbakhsh S, Abrego-Guandique DM, Bacchetti T, Aghaee-Bakhtiari SH, Mahmoudi A, Manteghi AA, et al. Direct hybridization and bioinformatics analysis of circulating microRNAs in patients with Alzheimer's disease under intravenous trehalose treatment. Brain Res. (2025) 1857:149607. doi: 10.1016/j.brainres.2025.149607

PubMed Abstract | Crossref Full Text | Google Scholar

289. Wong HK, Veremeyko T, Patel N, Lemere CA, Walsh DM, Esau C, et al. De-repression of FOXO3a death axis by microRNA-132 and−212 causes neuronal apoptosis in Alzheimer's disease. Hum Mol Genet. (2013) 22:3077–92. doi: 10.1093/hmg/ddt164

Crossref Full Text | Google Scholar

290. Wang T, Long Q, Hu Y, Yang Y, Li X, Wei H. miR-181c-5p suppresses neuronal pyroptosis via NLRP1 in Alzheimer's disease. Behav Brain Res. (2023) 447:114387. doi: 10.1016/j.bbr.2023.114387

PubMed Abstract | Crossref Full Text | Google Scholar

291. Jiang Y, Zhang Y, Su L. MiR-539-5p decreases amyloid beta-protein production, hyperphosphorylation of Tau and memory impairment by regulating PI3K/Akt/GSK-3beta pathways in APP/PS1 double transgenic mice. Neurotox Res. (2020) 38:524–35. doi: 10.1007/s12640-020-00217-w

Crossref Full Text | Google Scholar

292. DeWitt E, Gillespie R, Norman-Burgdolf H, Cardarelli KM, Slone S, Gustafson A. Rural SNAP participants and food insecurity: how can communities leverage resources to meet the growing food insecurity status of rural and low-income residents? Int J Environ Res Public Health. (2020) 17:6037. doi: 10.3390/ijerph17176037

PubMed Abstract | Crossref Full Text | Google Scholar

293. Franck KL, Jarvandi S, Johnson K, Elizer A, Middleton S, Sammons L. Working with rural producers to expand EBT in farmers' markets: a case study in Hardeman County, Tennessee. Health Promot Pract. (2023) 24(1_suppl):125S−7S. doi: 10.1177/15248399221115707

PubMed Abstract | Crossref Full Text | Google Scholar

294. Ma X, Sharpe PA, Bell BA, Liu J, White K, Liese AD. Food acquisition and shopping patterns among residents of low-income and low-access communities in South Carolina. J Acad Nutr Diet. (2018) 118:1844–54. doi: 10.1016/j.jand.2018.04.017

PubMed Abstract | Crossref Full Text | Google Scholar

295. Purkait T, Dev DA, Koziol N, Saltzman JA, Smith J, Franzen-Castle L. Urban-rural disparities in perceived benefits and challenges for supplemental nutrition assistance program (SNAP) participation. Am J Health Promot. 2025:8901171251352703. doi: 10.1177/08901171251352703

PubMed Abstract | Crossref Full Text | Google Scholar

296. Emery RL, Yoon C, Mason SM, Neumark-Sztainer D. Childhood maltreatment and disordered eating attitudes and behaviors in adult men and women:Findings from project EAT. Appetite. (2021) 163:105224. doi: 10.1016/j.appet.2021.105224

PubMed Abstract | Crossref Full Text | Google Scholar

297. Penha SDC, Norde MM, Carioca AAF. Childhood eating practices are relevant to ultra-processed food consumption in adulthood: results from the Nutritionists' Health Study. J Dev Orig Health Dis. (2022) 13:583–92. doi: 10.1017/S2040174421000696

PubMed Abstract | Crossref Full Text | Google Scholar

298. Localio AM, Knox MA, Basu A, Lindman T, Walkinshaw LP, Jones-Smith JC. Universal free school meals policy and childhood obesity. Pediatrics. (2024) 153:e2023063749. doi: 10.1542/peds.2023-063749

PubMed Abstract | Crossref Full Text | Google Scholar

299. Wong Zhang DE, Tran V, Vinh A, Dinh QN, Drummond GR, Sobey CG, et al. Pathophysiological links between obesity and dementia. Neuromolecular Med. (2023) 25:451–6. doi: 10.1007/s12017-023-08746-1

PubMed Abstract | Crossref Full Text | Google Scholar

300. Rivera RL, Dunne J, Maulding MK, Wang Q, Savaiano DA, Nickols-Richardson SM, et al. Exploring the association of urban or rural county status and environmental, nutrition- and lifestyle-related resources with the efficacy of SNAP-Ed (Supplemental Nutrition Assistance Program-Education) to improve food security. Public Health Nutr. (2018) 21:957–66. doi: 10.1017/S1368980017003391

PubMed Abstract | Crossref Full Text | Google Scholar

301. Chen H, Deng Y, Li X, Ding A, Wang J, Sun A, et al. Factors associated with dementia risk reduction lifestyle in mild cognitive impairment: a cross-sectional study of individuals and their family caregivers. BMC Neurol. (2025) 25:169. doi: 10.1186/s12883-025-04183-8

PubMed Abstract | Crossref Full Text | Google Scholar

302. Ross MK, Raji C, Lokken KL, Bredesen DE, Roach JC, Funk CC, et al. Case study: a precision medicine approach to multifactorial dementia and Alzheimer's disease. J Alzheimers Dis Parkinsonism. (2021) 11(Suppl 5):018.

PubMed Abstract | Google Scholar

303. Sandison H, Callan NGL, Rao RV, Phipps J, Bradley R. Observed improvement in cognition during a personalized lifestyle intervention in people with cognitive decline. J Alzheimers Dis. (2023) 94:993–1004. doi: 10.3233/JAD-230004

PubMed Abstract | Crossref Full Text | Google Scholar

304. Hirode G, Wong RJ. Trends in the prevalence of metabolic syndrome in the United States, 2011-2016. JAMA. (2020) 323:2526–8. doi: 10.1001/jama.2020.4501

PubMed Abstract | Crossref Full Text | Google Scholar

305. Voulgaropoulou SD, van Amelsvoort T, Prickaerts J, Vingerhoets C. The effect of curcumin on cognition in Alzheimer's disease and healthy aging: a systematic review of pre-clinical and clinical studies. Brain Res. (2019) 1725:146476. doi: 10.1016/j.brainres.2019.146476

PubMed Abstract | Crossref Full Text | Google Scholar

306. Esmaealzadeh N, Miri MS, Mavaddat H, Peyrovinasab A, Ghasemi Zargar S, Sirous Kabiri S, et al. The regulating effect of curcumin on NF-kappaB pathway in neurodegenerative diseases: a review of the underlying mechanisms. Inflammopharmacology. (2024) 32:2125–51. doi: 10.1007/s10787-024-01492-1

Crossref Full Text | Google Scholar

307. Koushki M, Amiri-Dashatan N, Ahmadi N, Abbaszadeh HA, Rezaei-Tavirani M. Resveratrol: a miraculous natural compound for diseases treatment. Food Sci Nutr. (2018) 6:2473–90. doi: 10.1002/fsn3.855

PubMed Abstract | Crossref Full Text | Google Scholar

308. El-Sayed NS, Elatrebi S, Said R, Ibrahim HF, Omar EM. Potential mechanisms underlying the association between type II diabetes mellitus and cognitive dysfunction in rats: a link between miRNA-21 and Resveratrol's neuroprotective action. Metab Brain Dis. (2022) 37:2375–88. doi: 10.1007/s11011-022-01035-z

PubMed Abstract | Crossref Full Text | Google Scholar

309. Isobe C, Abe T, Terayama Y. Homocysteine may contribute to pathogenesis of RNA damage in brains with Alzheimer's disease. Neurodegener Dis. (2009) 6:252–7. doi: 10.1159/000262443

PubMed Abstract | Crossref Full Text | Google Scholar

310. Rizvi S, Raza ST, Ahmed F, Ahmad A, Abbas S, Mahdi F. The role of vitamin e in human health and some diseases. Sultan Qaboos Univ Med J. (2014) 14:e157–65. doi: 10.18295/2075-0528.1566

PubMed Abstract | Crossref Full Text | Google Scholar

311. Gezen-Ak D, Dursun E, Yilmazer S. The effects of vitamin D receptor silencing on the expression of LVSCC-A1C and LVSCC-A1D and the release of NGF in cortical neurons. PLoS ONE. (2011) 6:e17553. doi: 10.1371/journal.pone.0017553

PubMed Abstract | Crossref Full Text | Google Scholar

312. Rey C, Nadjar A, Buaud B, Vaysse C, Aubert A, Pallet V, et al. Resolvin D1 and E1 promote resolution of inflammation in microglial cells in vitro. Brain Behav Immun. (2016) 55:249–59. doi: 10.1016/j.bbi.2015.12.013

PubMed Abstract | Crossref Full Text | Google Scholar

313. Taganov KD, Boldin MP, Chang KJ, Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA. (2006) 103:12481–6. doi: 10.1073/pnas.0605298103

PubMed Abstract | Crossref Full Text | Google Scholar

314. Sheedy FJ, Palsson-McDermott E, Hennessy EJ, Martin C, O'Leary JJ, Ruan Q, et al. Negative regulation of TLR4 via targeting of the proinflammatory tumor suppressor PDCD4 by the microRNA miR-21. Nat Immunol. (2010) 11:141–7. doi: 10.1038/ni.1828

PubMed Abstract | Crossref Full Text | Google Scholar

315. Maucher D, Schmidt B, Kuhlmann K, Schumann J. Polyunsaturated fatty acids of both the omega-3 and the omega-6 family abrogate the cytokine-induced upregulation of miR-29a-3p by endothelial cells. Molecules. (2020) 25:4466. doi: 10.3390/molecules25194466

PubMed Abstract | Crossref Full Text | Google Scholar

316. Scoyni F, Giudice L, Vaananen MA, Downes N, Korhonen P, Choo XY, et al. Alzheimer's disease-induced phagocytic microglia express a specific profile of coding and non-coding RNAs. Alzheimers Dement. (2024) 20:954–74. doi: 10.1002/alz.13502

PubMed Abstract | Crossref Full Text | Google Scholar

317. Mei Z, Liu J, Schroeder JP, Weinshenker D, Duong DM, Seyfried NT, et al. Lowering hippocampal miR-29a expression slows cognitive decline and reduces beta-amyloid deposition in 5xFAD mice. Mol Neurobiol. (2024) 61:3343–56. doi: 10.1007/s12035-023-03791-0

PubMed Abstract | Crossref Full Text | Google Scholar

318. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. (2015) 21:263–9. doi: 10.1038/nm.3804

Crossref Full Text | Google Scholar

319. Zhang J, Li X, Ren Y, Zhao Y, Xing A, Jiang C, et al. Intermittent fasting alleviates the increase of lipoprotein lipase expression in brain of a mouse model of Alzheimer's disease: possibly mediated by beta-hydroxybutyrate. Front Cell Neurosci. (2018) 12:1. doi: 10.3389/fncel.2018.00001

Crossref Full Text | Google Scholar

320. Yamada K, Takizawa S, Ohgaku Y, Asami T, Furuya K, Yamamoto K, et al. MicroRNA 16-5p is upregulated in calorie-restricted mice and modulates inflammatory cytokines of macrophages. Gene. (2020) 725:144191. doi: 10.1016/j.gene.2019.144191

PubMed Abstract | Crossref Full Text | Google Scholar

321. Zhang N, Li WW, Lv CM, Gao YW, Liu XL, Zhao L. miR-16-5p and miR-19b-3p prevent amyloid beta-induced injury by targeting BACE1 in SH-SY5Y cells. Neuroreport. (2020) 31:205–12. doi: 10.1097/WNR.0000000000001379

Crossref Full Text | Google Scholar

322. Liao W, Jiang M, Li M, Jin C, Xiao S, Fan S, et al. magnesium elevation promotes neuronal differentiation while suppressing glial differentiation of primary cultured adult mouse neural progenitor cells through ERK/CREB activation. Front Neurosci. (2017) 11:87. doi: 10.3389/fnins.2017.00087

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: dementia, cognitive decline, gut-brain axis, rural population, nutraceuticals

Citation: Goncalves BS, Gannavaram NL, Yates C, Kandula A, Nayyar A, Pillai SS, Puri RK and Nayyar A (2025) Nutritional strategies against dementia in rural populations. Front. Nutr. 12:1677197. doi: 10.3389/fnut.2025.1677197

Received: 01 August 2025; Revised: 18 November 2025; Accepted: 24 November 2025;
Published: 15 December 2025.

Edited by:

Clarissa Fantin Cavarsan, Wesleyan University Libraries, United States

Reviewed by:

Shixing Feng, International Academy of Food and Phytomedicine Homology, France
Afreen Saif, King George Medical University, India

Copyright © 2025 Goncalves, Gannavaram, Yates, Kandula, Nayyar, Pillai, Puri and Nayyar. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Bruno S. Goncalves, ZGVzb3V6YWdvbmNhQG1hcnNoYWxsLmVkdQ==; Asma Nayyar, bmF5eWFyQG1hcnNoYWxsLmVkdQ==

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.