Next Article in Journal
Metabolic and Mitochondrial Dysregulations in Diabetic Cardiac Complications
Previous Article in Journal
Glatiramer Acetate Modifies the Immune Profiles of Monocyte-Derived Dendritic Cells In Vitro Without Affecting Their Generation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of the Exposome on Alzheimer’s Disease: The Influence of Nutrition

1
Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy
2
IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(7), 3015; https://doi.org/10.3390/ijms26073015
Submission received: 14 February 2025 / Revised: 18 March 2025 / Accepted: 24 March 2025 / Published: 26 March 2025
(This article belongs to the Special Issue Molecular Research of Alzheimer’s Disease and Nutrition Therapy)

Abstract

:
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline, memory loss, and behavioural changes. While genetic predispositions and pathological processes have been the traditional focus, this review highlights the fundamental role of environmental factors, particularly nutrition, within the exposome framework in modulating the risk and progression of AD. The exposome, which includes the totality of environmental exposures in an individual’s lifetime, provides a comprehensive approach to understanding the complex aetiology of AD. In this review, we explore the impact of dietary factors and cyclic nucleotide pathways (cAMP/cGMP) on AD, emphasizing the potential of dietary interventions as therapeutic strategies. We investigate key aspects of how nutrition affects the accumulation of β-amyloid, the aggregation of tau proteins, and neuroinflammation. We also examine the impact of specific nutrients on cognitive performance and the risk of AD. Additionally, we discuss the potential of nutraceuticals with anti-phosphodiesterase activity and the role of various animal models of AD (such as 5xFAD, 3xTg-AD, Tg2576, and APP/PS1 mice) in demonstrating the effects of dietary interventions on disease onset and progression.

1. Alzheimer’s Disease and the Exposome

1.1. Introduction to AD

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that predominantly affects older adults and is characterized by cognitive decline, memory loss, and behavioural changes [1]. The global burden of AD is astonishing. According to the latest World Alzheimer Report [2], a new case of dementia develops every three seconds. This alarming statistic underscores the urgent need for effective prevention, treatment, and care strategies. The early stage of AD often manifests as mild cognitive impairment (MCI), where memory deficits exceed normal age-related changes [3]. Like many other neurodegenerative disorders, AD is today also considered a multifactorial disease. Among the multiple factors involved in this pathology, the accumulation of amyloid-β (Aβ), a 42-residue peptide whose production and clearance are finely regulated in the brain, plays a role. Research has demonstrated that impaired clearance of Aβ is a significant factor in AD progression [4]. According to the Aβ hypothesis, soluble aggregates of Aβ peptides, particularly dimers and small oligomers, are highly neurotoxic, disrupting neuronal structure and function [5]. Additionally, Aβ peptides contribute to inflammation [6], tau hyperphosphorylation [7], and, ultimately, cell death [8]. A further hallmark of AD is the formation of neurofibrillary tangles, which are primarily composed of tau protein aggregates. These tangles follow a hierarchical pattern of development in specific brain regions and are closely associated with neuronal loss as the disease advances [9]. Although tau aggregation is widely acknowledged as a pivotal neuropathological hallmark of AD [10], the precise mechanisms that underpin its toxicity remain unclear [11,12]. Another potential contributing factor to the development of AD is neuroinflammation, which plays a pivotal role in both the initiation and progression of the disease. Long-term neuroinflammation can cause cell damage by increasing inflammatory cells, producing reactive oxygen species (ROS), and causing significant changes to DNA [13,14]. Therefore, it seems that inflammation may be a central mechanism in the pathogenesis of AD, prompting the question of whether it is a cause or a consequence of neurodegeneration in this disease [15]. The most probable hypothesis is that there exists a bidirectional relationship between inflammation and AD [16]. While recent immunological therapies have been claimed to possess disease modifying properties, challenges remain regarding their efficacy and safety. These treatments target Aβ deposition, but their impact on disease progression is still debated [17,18]. As a matter of fact, both lecanemab and donanemab, the two anti-Aβ antibodies recently approved by FDA for the treatment of AD, showed marginal, though significant, efficacy on cognitive decline and on activities of daily living, with differences between the placebo and drug-treated populations of 0.45 and 0.7 points, respectively, on the CDR-SB (score range 0–18)in the lecanemab (CLARITY) and in the donanemab (TRAILBLAZER-ALZ 2) clinical trials and 2.91–3.25 points on the iADRS (score range 0–144) in the latter case [18]. Such limited differences have raised many concerns in the scientific community on the clinical relevance of these treatments in the real-world setting. Additionally, administration of these antibodies is accompanied with important side effects (up to 40% incidence) called amyloid-related imaging abnormalities (ARIA), which include brain tissue oedema (ARIA-E) and microbleeds/microhaemorrhage (ARIA-H), representing the major cause of therapy discontinuation [18].
Moreover, emerging evidence highlights the complex interplay between AD and other chronic conditions, such as diabetes, depression, cardiovascular disease, and inflammatory bowel disease [19,20,21,22,23,24,25]. Several new theories have been proposed: chronic neuronal stress [26], the L-isoaspartyl methyltransferase (PIMT) hypothesis [27], the lipid invasion model [28], and the infectious theory [29].
From an etiopathogenic point of view, hereditary factors are significantly linked to the development of late-onset AD; therefore, genetics alone cannot account for the great majority of cases. The fact that many of the comorbidities associated with AD are linked to dysregulated metabolic pathways suggests that lifestyle variables have a role in disease aetiology. In this sense, lifestyle changes such as exercise and diet may interact with inherited susceptibility genes to improve cognition in AD patients [30,31,32,33]. The global AD epidemic demands a multifaceted approach that addresses both biological and lifestyle factors. Continued research into disease mechanisms, the development of innovative treatments, and the promotion of healthy lifestyle interventions is essential to improve the lives of individuals affected by this debilitating condition. Hence, this review aims to shed light on the influence of exposure factors, the “exposome”, on AD, specifically focusing on dietary factors and cyclic adenosine monophosphate (cAMP)/cyclic guanosine monophosphate (cGMP) pathways.

1.2. Exposome and AD

The concept of the exposome, which describes the totality of environmental exposures individuals encounter throughout their lifetime, offers a compelling framework for understanding the complex aetiology of AD [34,35]. Traditionally, research on AD has predominantly focused on genetic predispositions and pathological processes [36,37]. Nevertheless, an accumulating body of evidence highlights the substantial influence of environmental factors in modulating disease risk and progression [38]. This notion is particularly relevant for brain health, leading to the term “neural exposome” [39]. Understanding the neural exposome involves recognizing the interplay between various external factors [40]—ranging from air pollution and dietary habits to occupational exposures and psychosocial stressors—and their cumulative impact on brain health [41]. Many studies indicate that long-term exposure to environmental pollutants, such as particulate matter, may contribute to the development of neuroinflammation and oxidative stress, both of which are linked to AD pathology [42,43,44,45,46,47,48,49]. The first studies providing this evidence were conducted on feral dogs naturally exposed to polluted urban environments that showed enhanced oxidative damage, premature presence of diffuse amyloid plaques, and DNA damage in the olfactory bulbs, frontal cortex, and hippocampus [42,43]. The damage of these brain regions occurred in a gradient fashion (olfactory mucosa > olfactory bulb > frontal cortex), indicating the nasal pathway as a key portal entry for pollution. Interestingly, the earliest symptoms in AD include olfactory dysfunctions [44]. Subsequent investigations carried out in mice showed that a prolonged exposure to diesel exhaust particles caused an upregulation of pro-inflammatory cytokines and of the oxidative stress marker malondialdehyde, as well as neuronal morphology alterations in the hippocampus and olfactory bulb [45]. These results were confirmed by other preclinical studies demonstrating that both pre- and postnatal exposure to ambient air pollution (particulate matter diameter of 2.5 µm; PM2.5) induced short-term memory deficits in mice [46]. Furthermore, continuous exposure to PM2.5 caused neuronal loss and tau phosphorylation in the olfactory bulb, in the hippocampus, and in the cortex of 3xTg-AD mice [47]. The link between exposure to air pollution and development of neuroinflammation, oxidative stress, and AD was also shown in several human studies [48]. For instance, post-mortem analyses on human tissues showed that 40% of the exposed urbanites had tau hyperphosphorylation with pre-tangle material, and 51% had diffuse Aβ plaques, together with higher expression levels of genes involved in neuroinflammation and oxidative stress [49].
Adding to this evidence, a comprehensive study of the human exposome using a large-scale multianalyte metabolomics platform revealed that exposure to synthetic chemicals (e.g., persistent organic pollutants) is associated with adverse health outcomes, including neurodegenerative diseases [50]. This underscores the importance of addressing environmental factors in understanding and mitigating AD progression [51]. Furthermore, lifestyle factors, like nutrition and physical activity, are integral components of the neural exposome that can influence cognitive decline and vascular health, thereby indirectly affecting the onset of dementia. It is evident that both aspects are associated with pollution. Indeed, it is acknowledged that food contamination can affect the bioaccumulation of heavy metals, and that outdoor physical activity can expose individuals to particulate matter [50,51]. The complexity of the exposome lies not only in the diversity of exposures but also in the individual variability in response to these factors [52]. Genetic susceptibility can modulate how environmental exposures impact neurological health, directing attention to the importance of personalized approaches in AD research. For instance, variations in genes linked to inflammatory responses may exacerbate the effects of environmental toxins, highlighting the need for integrative models that consider both genetic and environmental influences.

1.3. Exposome and Nutrition

Nutrition is of pivotal importance within the exposome framework, as dietary habits exert a significant influence on both individual health outcomes and broader population health trends [53]. Dietary exposures are a principal determinant of health status, given the intricate and varied nature of the foods consumed by humans. Food contains essential macro- and micronutrients, as well as a wide range of non-nutrient compounds, some of which can have either beneficial or detrimental effects on human health [54]. The composition of these compounds is dependent upon several factors, including species, variety, production methods, and food processing. The assessment of the food exposome presents several unique challenges, largely due to the considerable variation in food intake patterns, the diverse chemical composition of food, and the complex interactions between different food components. However, recent technological advances have enabled the identification of dietary biomarkers in biospecimens such as blood, urine, and saliva, thereby providing a more objective approach to the evaluation of dietary exposures (Figure 1) [55].
Mass spectrometry is one of the most widely used techniques for identifying and quantifying dietary biomarkers due to its high sensitivity and specificity. MS can be coupled with other separation techniques such as liquid chromatography (LC-MS) or gas chromatography (GC-MS) to enhance resolution and detection [56,57]. Moreover, MS can be coupled to capillary electrophoresis to separate ions based on their size-to-charge ratio and is particularly useful for analyzing charged metabolites in biological fluids [58].
A universal quantitative analytical method for assessing an organic compound’s purity and spotting potential contaminants is nuclear magnetic resonance (NMR). For nutritional research, NMR can be used in conjunction with chromatography and MS techniques to identify and produce metabolic data [59]. HPLC is a versatile technique that separates compounds based on their interaction with a stationary phase. When combined with detectors such as UV/Vis, fluorescence, or electrochemical detection, HPLC can quantify specific dietary biomarkers [60]. ELISA is an immunoassay technique that uses antibodies to detect specific biomolecules. While less commonly used for dietary biomarkers compared to MS or NMR, ELISA is valuable for detecting large molecules like proteins or peptides [61,62].
The number of identified biomarkers has now surpassed 140 (http://exposome-explorer.iarc.fr, accessed on 25 February 2025), offering an important tool to investigate the influence of specific foods and dietary compounds on the development of chronic diseases such as neurodegenerative disorders [63]. The expanding use of dietary-wide association studies (DWAS) that employ biomarkers holds great promise for enhancing our understanding of the relationship between diet and long-term health outcomes [40]. Overall, nutrition plays a pivotal role in the exposome-related mechanisms from two fundamental perspectives: firstly, as a contributing factor to health, and secondly, as an influential agent in responses to environmental exposures [64,65]. Indeed, dietary patterns and nutrient intake can profoundly impact the body’s resilience against various stresses, including pollutants and toxins [66,67]. For instance, the presence of antioxidants in fruits and vegetables has been shown to mitigate oxidative stress induced by environmental contaminants [68,69,70]. Similarly, a diet rich in omega-3 fatty acids has been observed to reduce inflammatory responses. Consequently, by following a nutritionally balanced and nutrient-dense diet, individuals can enhance their ability to withstand negative health impacts associated with their exposure. Another extremely relevant aspect in the field of exposome-nutrition is the explosion of the supplements market [71]. These products are increasingly enriching the panorama of substances that are taken by the population often without expert monitoring [72]. Finally, the potential of nutrition within the exposome concept must also be analysed based on the characteristics of the subject (age, sex, concomitant diseases, therapies, etc.) [73,74,75].

1.4. Nutrition and AD

Recent scientific studies emphasize the connection between dietary habits and cognitive performance. As an example, pre-clinical studies using rodents demonstrated that cognitive decline, typical of aging and neuropsychiatric diseases, is correlated with gut microbiota impairment, which, in turn, can be affected by the quality of dietary habits [76]. Accordingly, Liu and colleagues showed that a high-fiber diet can reduce maternal obesity-induced social dysfunction and cognitive impairment in the offspring of C57BL/6J mice through the modulation of bacterial composition and production of short-chain fatty acids [77]. In line with this finding, several human studies demonstrated that maternal obesity modulates microbiota composition by decreasing the abundance of Ruminococcus, Bifidobacterium, and Blautia, which are associated with impaired cognitive functions in 36-month-old children [78,79,80]. Other studies investigated the impact of a meat-enriched diet on neurodegeneration. In fact, Quan and colleagues [81] demonstrated that a 50 g day−1 increment of processed meat is significantly associated with an increased risk of neurodegenerative cognitive disorders, whereas the consumption of fish and poultry seems less harmful.
It is worth noting that, due to the high percentage of people affected by obesity or chronic disease, the use of supplements and diet fads has become an integral part of modern health culture, growing steadily in popularity over the years [72]. These supplements are claimed to have multiple healthy effects on cognitive function, even if they are not supported by robust scientific evidence [72]. They also often lack rigorous clinical testing and may contain unregulated or harmful substances, posing risks of toxicity, adverse reactions, or interactions with prescription medications [72]. Moreover, diet fads that impose extreme caloric restriction or elimination of entire food groups can lead to nutrient deficiencies, metabolic imbalances, or disordered eating patterns. To tackle these challenges, it is essential to prioritize consumer education, healthcare provider involvement, regulatory oversight, and evidence-based research, ensuring individuals make informed health decisions while minimizing the risks linked to diet fads and supplements [72].
The intricate relationship between dietary habits and cognitive function is increasingly recognized as a critical factor in understanding and mitigating AD. A paradigm shift towards emphasizing food quality, rather than simply demonizing specific foods, is essential [82,83]. This necessitates a comprehensive evaluation of food intake, particularly in industrialized nations where overconsumption is prevalent, leading to excessive dietary loads [84,85,86]. At this regard, the notion of Metabolic Food Waste has been established as a metric to assess the implications of Western dietary practices [87]. Overnutrition, resulting from surplus caloric intake, directly contributes to overweight and obesity, highlighting the importance of addressing modifiable risk factors like malnutrition in AD prevention [88,89]. Conversely, targeted nutritional interventions hold promise for neuroprotection. Specific nutrients can modulate inflammation and oxidative stress, key pathological mechanisms in AD [90]. For instance, both ketogenic (KD) and Mediterranean diets exert anti-inflammatory effects by reducing pro-inflammatory cytokine expression, suppressing the COX-2 pathway, decreasing microglia activation, and restoring blood–brain barrier integrity [89,91,92]. Additionally, KD modulates gene expression and affects the gut–brain axis, contributing to its anti-inflammatory effects [92]. Beta-hydroxybutyrate further reduces intracellular levels of reactive oxygen species (ROS) and calcium, while increasing the activities of superoxide dismutase and catalase [93]. Similarly, the Mediterranean diet, rich in antioxidants, lowers postprandial levels of hydrogen peroxide and lipid peroxides [94]. Notably, polyphenols, abundant in this diet, combat neuroinflammation by reducing pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) and inhibiting the NF-κB pathway [95]. Their antioxidant properties neutralize ROS, boost antioxidant enzymes, strengthen the blood–brain barrier, and modulate gut microbiota, all contributing to neuroprotection. A recent study confirms the link between an anti-inflammatory diet and the risk of neurodegenerative diseases [96]. The study analysed the relationship between the Dietary Inflammatory Index (DII), which measures the inflammatory potential of a diet, and the risk of developing dementia. A diet rich in pro-inflammatory foods may increase chronic inflammation in the body, a factor associated with cognitive decline and neurodegenerative diseases [89,97,98]. In contrast, anti-inflammatory foods, such as fruit, vegetables, whole grains, and healthy fats, appear to protect the brain [96]. As a matter of fact, several studies have found a correlation between cognitive function and serum concentrations of key nutrients, such as folate, vitamin B-12, vitamin B-6, and homocysteine [84,85,86]. Among dietary patterns with established neuroprotective effects, the MIND diet, a hybrid of the Mediterranean and DASH diets, has demonstrated slower cognitive decline and reduced AD risk [99]. This diet emphasizes brain-healthy foods while limiting pro-inflammatory items. Higher adherence to the MIND diet is associated with fewer AD-related brain changes, independent of other lifestyle factors [99]. Similar benefits are observed with the Mediterranean and DASH diets, attributed to their ability to reduce oxidative stress and inflammation [100]. The DASH diet, originally designed to lower hypertension, has been associated with brain health due to its encouragement of nutrient-rich foods that help reducing oxidative stress and inflammation [101]. The integration of various diet components into the MIND diet has shown promising results in delaying neurodegenerative diseases and cognitive decline [99,101].
A diet with short- and medium-chain fatty acids (SCFAs, MCFAs, respectively) in animal models of AD (see below) has been shown to lower levels of AD biomarkers (e.g., Aβ peptide), to reduce inflammation, glia activation, oxidative stress, and neuronal loss, and to improve performances in different cognitive tasks [102]. Moreover, it was found that administration of medium-chain triglycerides, which are absorbed and metabolized via medium-chain fatty acids into ketone bodies, to AD patients significantly improved cognitive deficits [103,104].
Additionally, AD shares similarities with other inflammatory conditions linked to the Western lifestyle, where alterations in the gut microbiome and immune pathways have been associated with disease progression [105]. Supporting the link between diet and AD, a study on the AppNL−F/NL−F mouse model showed that a high-fat diet (HFD) exacerbates AD pathology by increasing Aβ plaque deposition, promoting oxidative stress, and impairing cognitive function [106]. Furthermore, the 36-month LipiDiDiet trial demonstrated that a multinutrient intervention slowed cognitive decline and brain atrophy and ameliorated memory performance in individuals with prodromal AD, thereby highlighting the role of diet in modifying AD risk [107]. A recent study from Dilmore and colleagues revealed that ketogenic and low-fat diets modulate the human metabolome and microbiome, with the KD particularly showing promise in improving gut microbial profiles and reducing cognitive decline in individuals at risk for AD [108]. This points to the possibility of delaying or treating AD through novel dietary approaches [105]. The health of gut microbiota and the integrity of brain health are interrelated. However, the impact of this relationship on AD is still difficult to establish. A preclinical study using a mouse model of AD demonstrated that Sika deer antler protein bi-directionally modulates the brain and intestinal tract via the tyrosine metabolism pathway, improving the AD symptoms through the activation of the PI3K/AKT/Nrf2 signalling pathway [109]. Probiotics may significantly contribute to mitigating the inflammatory processes that extend from the gut to the brain in the context of AD. By promoting a healthy balance of gut microbiota, these beneficial microorganisms can help reduce inflammation and oxidative stress, potentially lessening the impact of AD on cognitive function and overall brain health [110,111]. In a preclinical study, AD rats that received probiotics (Lactobacillus acidophilus, L. fermentum, Bifidobacterium lactis, and B. longum) had significantly improved spatial memory and reduced oxidative stress biomarkers compared to the AD control group [112]. Moreover, probiotics (a mixture of Lactobacillus acidophilus, Bifidobacterium bifidum, Lactobacillus casei, and Bifidobacterium lactis) improved memory deficits, synaptic and cerebral neuronal injuries, and microglial activation in a mouse model of AD [113]. The mechanism appears to involve inhibition of the caspase-11/caspase-1 pathway and reduction of ALPK1 expression.

1.5. cAMP/cGMP Pathways and AD

Cyclic adenosine and guanosine monophosphate (cAMP and cGMP, respectively) are two classical second messengers that are synthesised in cells by different isoforms of adenylyl and guanylyl cyclases (AC; GC) starting from the respective triphosphate substrates [114,115]. While the AC activity is mainly modulated by G-protein-coupled receptors through Gi/Gs protein, the two GC families have different structures, cellular localizations, and mechanisms of activation. In particular, the soluble form of this enzyme represents the heme-based gasoreceptor of the intra/intercellular transmitter nitric oxide (NO), which, in turn, is produced by different NO synthases (namely neuronal, endothelial, and macrophagic NOS), transforming L-arginine into L-citrulline [115]. Moreover, NO can also derive from dietary nitrate, present in several green-leafy vegetables and root vegetables, via the nitrate–nitrite–NO pathway [116]. Interestingly, it has been proposed that flavonoids, a group of natural compounds present in various foods (see below), can enhance NO production through increased expression and activation of endothelial NOS [117]. Similarly, omega-3 polyunsaturated fatty acids (ω3 PUFAs), such as docosahexaenoic acid (DHA) and eicosapentanoic acid (EPA), are able to increase NO, also in this case via endothelial NOS upregulation [118].
It is well established that the two cyclic nucleotides play a crucial role in learning and memory formation/consolidation, basically acting in concert to trigger and maintain the synaptic plasticity process known as long term potentiation (LTP), and, indeed, their modulation has been explored as a potential therapeutic strategy for AD [114,115,119].
Despite being among the most extensively researched therapeutic strategies, modulators of these pathways have exhibited partial outcomes in clinical trials on AD patients, only in part corroborating the preclinical evidence [115,120,121]. It is reasonable that nutrition, as a fundamental element of the exposome, has the capacity to contribute to the homeostasis of cyclic nucleotides. A notable finding in the extant literature is the observation of reduced arginine and citrulline concentrations in the cerebrospinal fluid of individuals diagnosed with AD [122,123,124], which may negatively impact the function of the NO/cGMP pathway. Of note, research has demonstrated that elderly patients afflicted with cerebrovascular disease exhibited enhanced cognitive functions and a reduction in lipid peroxidation levels in response to oral administration of L-Arg [125]. Interestingly, it has been shown that intracerebroventricular administration of arginine to 3xTg-AD mice improved spatial memory acquisition [126]. A recent preclinical study also demonstrated that L-Arg and limonoid supplementation in AD mice reestablished the intracellular GSH levels, reduced oxidative stress, suppressed the secretion of proinflammatory cytokines, and balanced intestinal microflora. These effects were due to the enhancement of communication between gut, pancreas, liver, and brain to produce metabolites that promote host homeostasis [127]. Finally, an oral supplementation with citrulline, which is also a precursor of arginine, was able to improve spatial memory in an AD mouse model, an effect apparently associated with the increase of arginine in the CSF and of NO in the hippocampus [128].
As recently reviewed [129], dietary nitrate can also influence cerebral blood flow and cognitive functions, although there is still no clear evidence, with several clinical trials showing significant beneficial effects while others did not. However, a prospective study on participants of the Rush Memory and Aging Project followed for 4.7 years reported that daily consumption of three green-leafy vegetables (spinach, cabbages, lettuce), containing several bioactive compounds including nitrate, was associated with slower cognitive decline in the higher intake group (median 1.3 servings/day), equivalent to being 11 years younger. Of note, a higher intake of each of the identified nutrients and bioactive compounds (phylloquinone, lutein, folate, α-tocopherol, kaempferol, and nitrate) was associated with a slower cognitive decline [130]. In addition, in a recent prospective study, Rajendra et al. [131] examined the association between the habitual intake of dietary nitrate on older adults of the Australian Imaging, Biomarkers and Lifestyle Study of Aging, cognitively normal at baseline and stratified by APOEε4 status, a polymorphism of the APOE gene known to increase the risk of developing AD. A higher dietary nitrate intake (60 mg/day) was associated with a better language performance in APOEε4 non-carriers, whereas APOEε4 carriers showed better episodic recall memory and recognition memory over 126 months. However, no relationship of dietary nitrate intake with cognitive decline was observed. The same research group also investigated the association between dietary nitrate intake and dementia-related mortality among participants of the Australian Diabetes, Obesity, and Lifestyle study followed over a period of 17 years [132]. They found that participants with the highest intakes of dietary nitrate derived from plants (vegetables, fruits, cereals, herbs, spices, pulses, and nuts; median intake of 98 mg/day) or from just vegetables (median intake of 78 mg/day) had a 57% and 66% lower risk of dementia-related mortality, respectively. As for flavonoids, it remains to be established whether their documented beneficial effects on cognitive functions and in reducing the risk of AD [133] can be attributed, at least in part, to the activation of the NOS/sGC pathway in the cardiovascular and central nervous systems. Regarding ω3 PUFAS, a recent meta-analysis of 21 trials revealed that a one serving/week increment of dietary fish or 0.1 g/day of DHA was associated with a lower risk for dementia and AD, whereas the risk for MCI was lower with an increment of 8 g/day of total PUFAs [134]. However, other studies did not confirm such an association. Therefore, although the existing data favour the idea that an increased consumption of ω3 PUFAS can reduce the risk of cognitive decline and AD, more studies are needed to definitively answer this question and to establish whether this effect is mediated by NO/cGMP.
The importance of cyclic nucleotides in various physiological processes suggests that dietary factors could potentially influence their regulation and signalling [135]. This aspect could be pertinent in determining the actual efficacy of drug treatments in clinical trials and in evaluating their significance in the set endpoints.

1.6. Phosphodiesterase Enzymes (PDEs)

The identification of cyclic AMP phosphodiesterase activity in 1962 spurred extensive research into its biochemical characterization, tissue distribution, and physiological functions. Phosphodiesterases (PDEs) are a family of enzymes that catalyse the hydrolysis of phosphodiester bonds in cyclic nucleotides, namely cAMP cGMP, thus terminating their second messenger functions [136]. By regulating the levels of cAMP and cGMP, PDEs play a pivotal role in maintaining cellular homeostasis and influencing metabolic pathways [137]. Subsequent research led to the identification of numerous PDE isozymes, characterized by distinct substrate specificities, regulatory mechanisms, and inhibitor sensitivities. This has culminated in the establishment of a standardized PDE classification system, recognizing eleven distinct PDE families, encoded by 21 genes with multiple variants [138]. Despite sharing a conserved C-terminal catalytic domain, PDEs exhibit variation in their N-terminal regions. Classification of these enzymes is based on C-terminal homology [139]. The substrate specificities exhibited by different PDE families are distinct: some target both cyclic nucleotides (PDE1, 2, 3, 10, 11), while others are selective for cAMP (PDE4, 7, 8) or cGMP (PDE5, 6, 9). They are distinguished by isoform diversity, tissue-specific expression, and subcellular localization, rendering them appealing drug targets. This highlights the potential for targeted interventions also through dietary modifications that can influence specific PDE pathways, ultimately shaping health outcomes. Therapeutic development efforts have yielded numerous selective PDE inhibitors with diverse applications, including the treatment of central nervous system (CNS) diseases such as AD [140].

1.7. PDEs and AD

PDE inhibitors have been identified as promising therapeutic agents for the treatment of AD [114,115,119] since, as reported above, cAMP and cGMP signalling are crucial for memory formation and consolidation. These pathways exert significant influence on corticostriatal and hippocampal circuits, impacting both pre- and postsynaptic neurons [141]. Dysregulation of these pathways has been strongly implicated in the pathogenesis of neurodegenerative disorders, including AD [142,143,144,145].
Reduced adenylyl cyclase (AC) activity in the hippocampus and temporal cortex, coupled with increased expression of specific PDE isoforms (e.g., PDE3, PDE4D, PDE7, and PDE8B), has been observed in AD patients [108]. This leads to decreased cAMP-dependent protein kinase A (PKA) activity, as evidenced by reduced cAMP binding to PKA and decreased PKA expression and activity, particularly in severe cases with high amyloid-β (Aβ) burden [146]. Furthermore, cAMP-mediated phosphorylation of Tau at specific sites may contribute to the reduction of neurofibrillary tangles, another pathological hallmark of AD [147,148,149]. Indeed, strategies aimed at elevating cAMP levels, such as PDE4 inhibition, have shown promise in reversing memory deficits in AD [150,151,152,153,154]. These and other observations align with the hypothesis that the cAMP/PKA/cAMP response element-binding protein (CREB) pathway plays a crucial role in mitigating or preventing AD-associated deficits.
As for the cGMP signalling pathway, reduced soluble guanylyl cyclase (sGC) activity has been observed in the superior temporal cortex of AD patient and in reactive astrocytes surrounding Aβ plaques [155,156]. This is accompanied by decreased cGMP levels in the cerebrospinal fluid and platelets of AD patients, which correlates with the severity of memory impairment [143,157]. While some studies have reported increased activity of the NOS/sGC system [158,159], the overall evidence suggests a decline in cGMP signalling in AD. Accordingly, a marked increase in PDE5A mRNA and protein expression has been observed in the temporal and entorhinal cortex of individuals diagnosed with AD [160,161]. In this view, it is worth noting that the PDE5 selective inhibitor vardenafil was able to rescue impaired LTP in functional synapses isolated from parietal cortex samples of AD patients [162]. Recently, a systematic review and meta-analysis of six studies comparing the occurrence of AD between PDE5 inhibitor users and non-users reported a significantly lower risk of developing the disease in the former group [163].
Although inhibitors of cGMP-specific PDEs, namely PDE5 and PDE9, have been shown to exert neuroprotective and memory-enhancing effects in a variety of AD animal models, their translation in clinical trials has not yet yielded consistent results [164,165].

1.8. PDEs and Nutrition

A number of naturally occurring compounds present in foodstuffs, including bioactive constituents in a variety of plant extracts, have been shown to exert inhibitory effects on PDE enzymes [166,167]. Furthermore, these compounds have been demonstrated to possess neuroprotective properties that can enhance cognitive function in individuals with dementia and other neurodegenerative disorders [168]. The inhibition of PDE enzymes by natural compounds found in food was initially observed with natural alkaloids such as caffeine [169]. Caffeine, initially known for its bronchodilation properties, was subsequently identified as a non-selective PDE inhibitor, exerting influence over multiple PDE isoforms [169]. This non-selective inhibition contributes to its effects on the CNS and has inspired research into its potential neuroprotective actions, including improvements in cognitive function and support in neurodegenerative disorders. In addition to caffeine, theophylline, another alkaloid present in trace amounts in tea, also functions as a non-selective PDE inhibitor, exhibiting comparable effects on smooth muscle relaxation and bronchodilation [170]. Flavonoids, a group of naturally occurring compounds found in fruits, vegetables, and beverages like tea, have demonstrated PDE inhibitory effects [171]. Key flavonoids, such as quercetin, kaempferol, and catechins act not only as antioxidants but also as modulators of cAMP and cGMP signalling pathways. By selectively inhibiting specific PDE isoforms, they enhance endothelial function, reduce inflammation, and contribute to improved cardiovascular health. Additionally, naringin, a flavonoid present in citrus fruits such as grapefruit, has been investigated as a natural PDE inhibitor. It is particularly noteworthy for its capacity to inhibit cGMP hydrolysis by PDE enzymes, thereby exerting vasorelaxant effects that may ameliorate cardiovascular health and enhance blood flow in the brain, potentially offering protective effects against neurodegenerative conditions [172]. Epigallocatechin-3-gallate (EGCG), a compound present in abundance in green tea, can inhibit all PDE isoforms with some more efficacy on PDE1 and 2. Such a broad inhibitory activity demonstrates significant potential in inducing vasorelaxation, thereby supporting cognitive function and offering neuroprotection against AD [173]. Also, ω3 PUFAs, particularly those derived from fish oil, exhibit inhibitory effects on specific PDE isoforms. This modulation of PDE activity contributes to anti-inflammatory responses and enhances insulin sensitivity, highlighting the critical role of dietary fat quality in metabolic health [174]. Furthermore, the impact of dietary nitrates, abundant in leafy green vegetables and beets, exemplifies the intricate interplay with cGMP signalling, since they are converted to nitric oxide within the body, thereby increasing cGMP levels (see above). While this is a direct effect of a nutrient, it raises questions about the subsequent regulation of PDEs, which might adapt to altered cGMP levels [116]. The implications of PDE activity in relation to nutrition extend to various disease states, including obesity, type 2 diabetes, and cardiovascular diseases, which are also risk factors for AD. Elevated PDE activity has been associated with insulin resistance, indicating that dietary interventions aimed at PDE inhibition may enhance glucose metabolism. For instance, the integration of flavonoid-rich foods into the diet has been shown to bolster cAMP signalling pathways, which may in turn enhance insulin sensitivity and glucose uptake in target tissues [175].
In the domain of cardiovascular health, the interplay between PDEs and nutrition holds particular pertinence. It is widely recognized that chronic inflammation and endothelial dysfunction are common precursors to cardiovascular diseases that, in turn, are regarded as possible risk factors for AD. Intriguingly, olive oil phenols were shown to inhibit PDE3, thus possessing anti-platelet aggregation properties [176]. Consequently, dietary interventions aimed at increasing the intake of PDE-inhibiting foods could serve as non-pharmacological strategies to enhance cardiovascular health. One such example is the Mediterranean diet, which is abundant in fruits, vegetables, whole grains, and healthy fats. This diet has been associated with improved endothelial function and a reduced risk of cardiovascular disease and AD [177,178]. It can be hypothesized that part of these beneficial effects may be attributable to the impact of the Mediterranean diet on PDE activity. The insights gained from understanding the relationship between PDEs and nutrition open doors for novel therapeutic strategies. Nutritional interventions that promote PDE inhibition could play a pivotal role in the prevention and management of chronic diseases. Nevertheless, it is imperative to approach this field with a degree of caution. While dietary supplements that act as PDE inhibitors are emerging, their long-term safety and efficacy require rigorous research.
Furthermore, personalized nutrition represents a promising avenue for maximizing the benefits of PDE inhibition. Genetic variations can influence individual responses to dietary components, suggesting that personalization could enhance the impact of specific diets on PDE activity. Future studies that incorporate genomics and metabolomics into nutritional research could elucidate tailored approaches to harness the power of PDE modulation through diet.

2. Transgenic AD Mouse Models and Nutrition

As previously delineated, the dynamic interplay between AD and nutrition could prove pivotal in the development of effective therapeutic interventions for dementia. Consequently, the utilization of animal models of AD is crucial. Indeed, AD mouse models have been widely employed to examine how nutrition affects disease onset and progression [179]. These models exhibit various features of AD, such as amyloid pathology, cholinergic deficits, neurodegeneration, and cognitive impairment [180]. In this section, we will provide an overview of the most frequently used mouse models of AD within the domain of nutrition (Table 1).

2.1. Tg2576 Mouse Model

The Tg2576 mouse is a single transgenic model overexpressing the human APP Swedish mutation, in which elevated levels of extracellular Aβ can be detected at six months, whereas insoluble plaques start emerging one month later, and behavioural alterations in cognitive domain (e.g., spatial and working memory) manifest at eight to ten months [181,182].
A combination of epigallocatechin-3-gallate, docosahexaenoic acid, and α-lipoic acid effectively reduced Aβ deposition, microglia activation, and memory deficits in the Tg2576 model [183]. Long-term pomegranate supplementation inhibited neuroinflammation and ameliorated the loss of synaptic structure proteins in AD mice [184,185]. Furthermore, dietary supplementation with palm fruits significantly improved learning and memory deficits, enhanced motor coordination, and reduced anxiety, while concurrently decreasing plasma Aβ40 and Aβ42 levels [186]. Similarly, figs are active on these transgenic mice, reducing oxidative damage and improving cognitive skills [187,188]. Specifically, dietary supplementation with Omani figs for 15 months in Tg2576 mice significantly attenuated oxidative damage, restored antioxidant status, and improved spatial learning and memory, compared to control-fed Tg2576 mice [185,187]. Moreover, it has been reported that adequate intake of docosahexaenoic acid, a n-3 PUFA found predominantly in marine fish that can also be biosynthesized in mammals from linolenic acid, significantly reduced Aβ levels and plaque burden, and modulated APP processing pathways in aged Tg2576 mice [189].
Conversely, a diet deficient in folate, vitamin B6, and B12 accelerated brain amyloidosis in Tg2576 mice [190], and diet-induced hyperhomocysteinemia increased Aβ formation and deposition in the same mouse model [191,192].

2.2. APP/PS1 Mouse Model

APP/PS1 mice were generated by combining the human APP Swedish mutation with the human PSEN1 carrying the delta E9 mutation. In this AD mouse model, amyloid plaques start developing at 4–6 months, and deficits across cognitive domains manifest as early as 6 months [181,182,193]. Using this model, it has been shown that a 3-month treatment with pomegranate extract attenuated Aβ plaque load and microgliosis and was able to improve spatial as well as working memory [194]. Pomegranate was also found to inhibit PC12 neuronal death caused by Aβ-induced oxidative stress and to improve learning and memory deficits in mice injected with Aβ42 [195]. Similarly, another study in APP/PS1 mice reported that urolithin A, a major metabolite of ellagic acid (derived from the hydrolysis of ellagitannins present in pomegranate and berries), provided neuroprotection by rescuing neurons from apoptosis and by stimulating neurogenesis through the activation of anti-inflammatory signalling pathways, reduced amyloid deposition and microgliosis, and improved spatial learning and memory [196]. Interestingly, metabolomic analysis of serum samples from APP/PS1 mice revealed significant alterations in various metabolic pathways, including energy metabolism, amino acid homeostasis, and membrane lipid metabolism, compared to wild-type mice [197]. In addition, a deficiency in folic acid, which is observed in AD patients [198], has been shown to exacerbate Aβ plaque accumulation in the brain of APP/PS1 mice and to worsen spatial memory but not learning ability [199]. This deficiency also resulted in the diminished expression of specific miRNAs [199] associated with amyloid formation, which may play a crucial role in the progression of neurodegenerative diseases. Moreover, by combining APP/PS1 characteristics with a mutation in cystathionine-beta-synthase, a mouse model with significantly elevated serum homocysteine levels was generated and used to investigate hyperhomocysteinemia that, beside genetic alterations, can result from dietary deficits (e.g., folate, vitamin B6 and B12) and is considered an AD risk factor [200]. Intriguingly, it was found that Aβ40 and Aβ42 levels were markedly increased in the brains of female, but not male, mice, thus emphasising a gender effect that deserves further investigations [198]. Finally, in APP/PS1 mice, a restriction in the intake of methionine, an amino acid especially abundant in red meat, has been reported to improve the gut microbiota and faecal metabolites, including SCFAs, able to reduce oxidative stress, enhance energy metabolism, and alleviate Aβ−induced cytotoxicity [201]. Of note, these effects were accompanied with a clear improvement of spatial and non-spatial memory. Remarkably, a faecal microbiota transplantation from transgenic mice with a methionine diet restriction to APP/PS1 mice also reversed the reduction in SCFAs, improving memory and learning scores [201].

2.3. 5xFAD Mouse Model

The 5XFAD mouse bears the human APP Swedish (KM670/671NL), London (V717I), and Florida (I716V) mutations together with PSEN1 mutations (M146L/L286V). On average, these mice show Aβ42 extracellular deposition by 2 months of age, amyloid plaques at around 6 months, and alterations in different cognitive domains starting at 4–6 months [181,182]. This early-onset transgenic model of AD has been used to study the effects of nutrition on the progression of AD pathology [202].
High-fat diet has been shown to impair energy metabolism in male 5XFAD mice, leading to increased locomotor activity, energy expenditure, and food intake [203]. The microbiome changes caused by a high-fat diet elevated the expression of apoptotic, microglial, and amyloidogenic genes in the hippocampuses of 5XFAD mice. A synbiotic formulation composed of Clostridium sporogenes and xylan enhanced the cognitive performance and spatial memory faculties in this animal model [204]. Interestingly, a Corinthian currant-supplemented diet during the early stages of AD in 5XFAD mice reduced brain oxidative stress through a mechanism involving the Paraoxonase-1 enzyme [205]. In this mouse model, however, the PUFAs ratio did not modify the pathological processes relevant to AD [206]. Oleocanthal, a naturally occurring phenolic secoiridoid isolated from extra-virgin olive oil, has been demonstrated to improve anxiety-like behaviour symptoms and increase sleeping hours in these transgenic mice [207].

2.4. 3xTg-AD Mouse Model

This mouse model was developed to replicate tau hyperphosphorylation and neurofibrillary tangle (NTF) formation that, in addition to the amyloid pathology, characterize human AD. Indeed, 3xTg-AD mice contain the APP Swedish, the human PSEN1 (M146V), and the MAPT (P301L) mutations and show increased Aβ42 levels at 4 months, Aβ plaques around 9 months, NTF at approximately 12 months, and initial cognitive deficits at 4 months [181,182].
In 2024, it was reported that curcumin effectively mitigated cognitive impairments in 3xTg-AD mice subjected to a high-fat, high-sugar diet. This effect was associated with a modulation of the gut microbiota, leading to a reduction in fatty acid synthesis, altered cholesterol metabolism, and suppressed adipogenesis-related pathways in the liver [208]. Resveratrol, another polyphenol, has also demonstrated neuroprotective properties in this mouse model. It has been shown to enhance the effects of exercise training by modulating the toxicity of Aβ oligomers [209]. In this triple transgenic AD mouse model, chronic quercetin administration exhibited neuroprotective effects, and a blueberry polyphenolic extract significantly improved memory performance [210,211]. Conversely, a diet high in saturated fats and deficient in n-3 and n-6 PUFAs has been demonstrated to elevate Aβ concentrations and insoluble tau levels in these AD mice. This diet also decreased cortical levels of drebrin, a postsynaptic marker, suggesting that high-fat consumption combined with low n-3 PUFA intake can promote AD-like neuropathology [212]. These results have been recently confirmed and extended, as it has been shown that the high-fat-diet-induced enhancement of Aβ deposition and tau phosphorylation was associated with microglia damage, neuroinflammation, and marked memory impairment [213]. Consistent with these findings, mice fed a high-fat diet supplemented with branched-chain amino acids exhibited increased tau neuropathology [214]. Moreover, a diet enriched in palmitate and deficient in linoleate intensified oxidative stress and Aβ accumulation within the hippocampus, a process mediated by the activation of NF-κB transcription [215]. In contrast, a diet with high tryptophan levels has been demonstrated to decrease intraneuronal Aβ density in the triple mutant mice. This suggests that augmenting dietary tryptophan intake, and consequently enhancing serotonin neurotransmission, may represent effective strategies for mitigating plaque pathology in AD [216].
Table 1. Nutritional and metabolic insights in transgenic AD mouse models.
Table 1. Nutritional and metabolic insights in transgenic AD mouse models.
Transgenic AD Mouse ModelMechanismsReferences
Tg2576
  • Aβ deposition: reduced by epigallocatechin-3-gallate, DHA, α-lipoic acid and dietary supplementation with palm fruit; exacerbated by vitamin deficiencies.
[183,186,191,192]
  • Neuroinflammation: inhibited by pomegranate supplementation.
[184,185]
  • Oxidative stress: fig extract supplementation reduces oxidative damage.
[187,188]
  • Cognitive performance: improved by palm fruit and fig supplementation.
[186,187,188]
APP/PS1
  • Aβ deposition: attenuated by pomegranate extract and exacerbated by folic acid deficiency.
[194,199]
  • Neuroinflammation: urolithin A reduces inflammation and stimulates neurogenesis.
[196]
  • Oxidative stress: reduced by pomegranate extract.
[195]
  • Cognitive performance: improved by urolithin A and microbiota transplantation.
[196,201]
5xFAD
  • Aβ deposition: high-fat diet elevates amyloidogenic gene expression in the hippocampus.
[203]
  • Oxidative stress: Corinthian currant-supplemented diet reduces brain oxidative stress via Paraoxonase-1 enzyme.
[205]
  • Cognitive performance: improved by a symbiotic formulation composed of Clostridium sporogenes and xylan.
[204]
3xTg-AD
  • Aβ deposition: elevated by high-fat and low PUFAs diets; reduced by high tryptophan levels diets.
[212,215,216]
  • Neuroinflammation: resveratrol and quercetin show neuroprotective effects.
[209,210]
  • Oxidative stress: a diet enriched in palmitate and deficient in linoleate intensified oxidative stress within the hippocampus.
[215]
  • Cognitive performance: improved by blueberry polyphenols.
[211]

3. Concluding Remarks

The exposome, particularly the role of nutrition, is crucial in understanding both the causes (aetiopathogenesis) and potential treatments (therapeutic aspects) of AD (Figure 2). This critical understanding is imperative to overcome the current limitations in AD treatment, often referred to as the “therapeutic impasse”.
Research has demonstrated that numerous dietary constituents possess significant modulatory effects that can enhance the efficacy of existing drug-based therapies. Considering the main mechanisms identified, various dietary changes can be tested to decrease Aβ plaque accumulation, regulate the brain–gut axis, lower oxidative stress and neuroinflammation, and alleviate comorbidities such as anxiety and depression. Furthermore, identifying and addressing dietary deficiencies is essential. Finally, recognizing the importance of translating this evidence into clinical practice through well-designed randomized controlled trials, we emphasize the importance of animal models in AD research. Given the promising results observed in AD transgenic mice summarised in this review, which, however, model the very limited percentage of familial AD, naturally occurring models such as SAMP8 mice appear to be increasingly valuable for investigating the impact of nutrition on the pathogenesis of the vast majority of sporadic AD and for developing novel therapeutic/nutraceutical strategies. These animals display a phenotype of accelerated aging and, in our opinion, may be the right model for analysing the impact of nutrition on the development and control of dementia. Indeed, they help to overcome potential bias due to the possibility that genetic variants may alter the microbiome directly, which can result in a specific disease phenotype [217,218].

Author Contributions

Conceptualization of the review article, E.F. and M.G; validation, E.F. and M.G.; writing—original draft preparation, M.M., C.T., E.F. and M.G.; writing—review and editing, M.M., C.T., E.F. and M.G. All authors have read and agreed to the published version of the manuscript.

Funding

We acknowledge the funding by Development and Cohesion Plan of the Italian Ministry of Health, Trajectory 4—Biotechnologies, bioinformatics, and pharmaceutical development project title: “Hybrid Hub (H2UB): Cellular and computational models, micro- and nanotechnologies for the personalisation of innovative therapies”, project code T4-AN-10.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Nasb, M.; Tao, W.; Chen, N. Alzheimer’s Disease Puzzle: Delving into Pathogenesis Hypotheses. Aging Dis. 2024, 15, 43–73. [Google Scholar] [CrossRef] [PubMed]
  2. Aguzzoli, C.S.; Anstey, K.J.; Atri, A.; Barbarino, P.; Benoist, C.; Brijnath, B.; Bruno, M.A.; Cose, L.; Darge, D.; Dean, W.; et al. World Alzheimer Report 2024 Global Changes in Attitudes to Dementia Contributors: Survey Translators. Alzheimer’s Disease International: London, UK, 2024. [Google Scholar]
  3. Zhong, S.; Lou, J.; Ma, K.; Shu, Z.; Chen, L.; Li, C.; Ye, Q.; Zhou, L.; Shen, Y.; Ye, X.; et al. Disentangling In-Vivo Microstructural Changes of White and Gray Matter in Mild Cognitive Impairment and Alzheimer’s Disease: A Systematic Review and Meta-Analysis. Brain Imaging Behav. 2023, 17, 764–777. [Google Scholar] [CrossRef] [PubMed]
  4. Żukowska, J.; Moss, S.J.; Subramanian, V.; Acharya, K.R. Molecular Basis of Selective Amyloid-β Degrading Enzymes in Alzheimer’s Disease. FEBS J. 2024, 291, 2999–3029. [Google Scholar] [PubMed]
  5. Zott, B.; Nästle, L.; Grienberger, C.; Unger, F.; Knauer, M.M.; Wolf, C.; Keskin-Dargin, A.; Feuerbach, A.; Busche, M.A.; Skerra, A.; et al. β-Amyloid Monomer Scavenging by an Anticalin Protein Prevents Neuronal Hyperactivity in Mouse Models of Alzheimer’s Disease. Nat. Commun. 2024, 15, 5819. [Google Scholar] [CrossRef]
  6. Hong, S.; Beja-Glasser, V.F.; Nfonoyim, B.M.; Frouin, A.; Li, S.; Ramakrishnan, S.; Merry, K.M.; Shi, Q.; Rosenthal, A.; Barres, B.A.; et al. Complement and Microglia Mediate Early Synapse Loss in Alzheimer Mouse Models. Science 2016, 352, 712–716. [Google Scholar] [CrossRef]
  7. Jin, M.; Shepardson, N.; Yang, T.; Chen, G.; Walsh, D.; Selkoe, D.J. Soluble Amyloid β-Protein Dimers Isolated from Alzheimer Cortex Directly Induce Tau Hyperphosphorylation and Neuritic Degeneration. Proc. Natl. Acad. Sci. USA 2011, 108, 5819–5824. [Google Scholar] [CrossRef]
  8. Kadowaki, H.; Nishitoh, H.; Urano, F.; Sadamitsu, C.; Matsuzawa, A.; Takeda, K.; Masutani, H.; Yodoi, J.; Urano, Y.; Nagano, T.; et al. Amyloid β Induces Neuronal Cell Death through ROS-Mediated ASK1 Activation. Cell Death Differ. 2005, 12, 19–24. [Google Scholar] [CrossRef]
  9. Zubčić, K.; Franić, D.; Pravica, M.; Hof, P.R.; Šimić, G.; Boban, M. Effects of Heterologous Human Tau Protein Expression in Yeast Models of Proteotoxic Stress Response. CNS Neurosci. Ther. 2024, 30, e14304. [Google Scholar] [CrossRef]
  10. Šimić, G.; Babić Leko, M.; Wray, S.; Harrington, C.; Delalle, I.; Jovanov-Milošević, N.; Bažadona, D.; Buée, L.; De Silva, R.; Di Giovanni, G.; et al. Tau Protein Hyperphosphorylation and Aggregation in Alzheimer’s Disease and Other Tauopathies, and Possible Neuroprotective Strategies. Biomolecules 2016, 6, 6. [Google Scholar] [CrossRef]
  11. Wegmann, S.; Maury, E.A.; Kirk, M.J.; Saqran, L.; Roe, A.; DeVos, S.L.; Nicholls, S.; Fan, Z.; Takeda, S.; Cagsal-Getkin, O.; et al. Removing Endogenous Tau Does Not Prevent Tau Propagation yet Reduces Its Neurotoxicity. EMBO J. 2015, 34, 3028–3041. [Google Scholar] [CrossRef]
  12. Congdon, E.E.; Duff, K.E. Is Tau Aggregation Toxic or Protective? J. Alzheimer’s Dis. 2008, 14, 453–457. [Google Scholar] [CrossRef]
  13. Zuppe, H.; Reed, E. Common Cytokine Receptor Gamma Chain Family Cytokines Activate MAPK, PI3K, and JAK/STAT Pathways in Microglia to Influence Alzheimer’s Disease. Front Mol. Neurosci. 2024, 17, 1441691. [Google Scholar] [CrossRef] [PubMed]
  14. Swanson, A.; Wolf, T.; Sitzmann, A.; Willette, A.A. Neuroinflammation in Alzheimer’s Disease: Pleiotropic Roles for Cytokines and Neuronal Pentraxins. Behav. Brain Res. 2018, 347, 49–56. [Google Scholar] [CrossRef] [PubMed]
  15. Pugazhenthi, S.; Qin, L.; Reddy, P.H. Common Neurodegenerative Pathways in Obesity, Diabetes, and Alzheimer’s Disease. Biochim. Et Biophys. Acta (BBA) Mol. Basis Dis. 2017, 1863, 1037–1045. [Google Scholar] [CrossRef]
  16. Newcombe, E.A.; Camats-Perna, J.; Silva, M.L.; Valmas, N.; Huat, T.J.; Medeiros, R. Inflammation: The Link between Comorbidities, Genetics, and Alzheimer’s Disease. J. Neuroinflammation 2018, 15, 276. [Google Scholar] [CrossRef]
  17. Terao, I.; Kodama, W. Comparative Efficacy, Tolerability and Acceptability of Donanemab, Lecanemab, Aducanumab and Lithium on Cognitive Function in Mild Cognitive Impairment and Alzheimer’s Disease: A Systematic Review and Network Meta-Analysis. Ageing Res. Rev. 2024, 94, 102203. [Google Scholar] [CrossRef]
  18. Fedele, E. Anti-Amyloid Therapies for Alzheimer’s Disease and the Amyloid Cascade Hypothesis. Int. J. Mol. Sci. 2023, 24, 14499. [Google Scholar] [CrossRef]
  19. Santiago, J.A.; Potashkin, J.A. The Impact of Disease Comorbidities in Alzheimer’s Disease. Front. Aging Neurosci. 2021, 13, 631770. [Google Scholar] [CrossRef]
  20. Herrera, P.A.; Campos-Romero, S.; Szabo, W.; Martínez, P.; Guajardo, V.; Rojas, G. Understanding the Relationship between Depression and Chronic Diseases Such as Diabetes and Hypertension: A Grounded Theory Study. Int. J. Environ. Res. Public Health 2021, 18, 12130. [Google Scholar] [CrossRef]
  21. Doroszkiewicz, J.; Mroczko, J.; Winkel, I.; Mroczko, B. Metabolic and Immune System Dysregulation: Unraveling the Connections between Alzheimer’s Disease, Diabetes, Inflammatory Bowel Diseases, and Rheumatoid Arthritis. J. Clin. Med. 2024, 13, 5057. [Google Scholar] [CrossRef]
  22. Casserly, I.P.; Topol, E.J. Convergence of Atherosclerosis and Alzheimer’s Disease: Cholesterol, Inflammation, and Misfolded Proteins. Discov. Med. 2004, 4, 149–156. [Google Scholar] [PubMed]
  23. Chatterjee, S.; Mudher, A. Alzheimer’s Disease and Type 2 Diabetes: A Critical Assessment of the Shared Pathological Traits. Front. Neurosci. 2018, 12, 383. [Google Scholar] [CrossRef]
  24. Ownby, R.L.; Crocco, E.; Acevedo, A.; John, V.; Loewenstein, D. Depression and Risk for Alzheimer Disease. Arch. Gen. Psychiatry 2006, 63, 530. [Google Scholar] [CrossRef] [PubMed]
  25. Zhou, X.; Hetrick, S.E.; Cuijpers, P.; Qin, B.; Barth, J.; Whittington, C.J.; Cohen, D.; Del Giovane, C.; Liu, Y.; Michael, K.D.; et al. Comparative Efficacy and Acceptability of Psychotherapies for Depression in Children and Adolescents: A Systematic Review and Network Meta-Analysis. World Psychiatry 2015, 14, 207–222. [Google Scholar] [CrossRef] [PubMed]
  26. Decourt, B.; D’Souza, G.X.; Shi, J.; Ritter, A.; Suazo, J.; Sabbagh, M.N. The Cause of Alzheimer’s Disease: The Theory of Multipathology Convergence to Chronic Neuronal Stress. Aging Dis. 2022, 13, 37. [Google Scholar] [CrossRef]
  27. D’Alessandro, A.; Lukens, J.R.; Zimring, J.C. The Role of PIMT in Alzheimer’s Disease Pathogenesis: A Novel Hypothesis. Alzheimer’s Dement. 2023, 19, 5296–5302. [Google Scholar] [CrossRef]
  28. Rudge, J.D. A New Hypothesis for Alzheimer’s Disease: The Lipid Invasion Model. J. Alzheimers Dis. Rep. 2022, 6, 129–161. [Google Scholar] [CrossRef]
  29. Ganz, T.; Fainstein, N.; Ben-Hur, T. When the Infectious Environment Meets the AD Brain. Mol. Neurodegener. 2022, 17, 53. [Google Scholar] [CrossRef]
  30. Abraham, M.J.; El Sherbini, A.; El-Diasty, M.; Askari, S.; Szewczuk, M.R. Restoring Epigenetic Reprogramming with Diet and Exercise to Improve Health-Related Metabolic Diseases. Biomolecules 2023, 13, 318. [Google Scholar] [CrossRef]
  31. Galkin, F.; Kovalchuk, O.; Koldasbayeva, D.; Zhavoronkov, A.; Bischof, E. Stress, Diet, Exercise: Common Environmental Factors and Their Impact on Epigenetic Age. Ageing Res. Rev. 2023, 88, 101956. [Google Scholar] [CrossRef]
  32. Yang, L.; Yuan, Z.; Peng, C. Effects of Aerobic Exercise on Cognitive Function and Quality of Life in Patients with Alzheimer’s Disease: A Systematic Review and Meta-Analysis. BMJ Open 2025, 15, e090623. [Google Scholar] [CrossRef] [PubMed]
  33. de Sá Leitão, C.V.F.; de Faria Moraes, B.; Leite, G.A.P.D.; Duarte, A.G.; da Silva, M.V.G.; de Oliveira, G.M.; Andrade, F.A.B.; da Silva, J.A.B.; dos Santos, R.C.C.; Figueiredo, G.S.; et al. Twelve Weeks of Exercise Training Improves Cognitive Status, Physical Performance and Quality of Life in Alzheimer’s Disease: A Systematic Review and Meta-Analysis. Ageing Res. Rev. 2025, 104, 102655. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, Y.-L.; Hao, Y.-N.; Ge, Y.-J.; Zhang, Y.; Huang, L.-Y.; Fu, Y.; Zhang, D.-D.; Ou, Y.-N.; Cao, X.-P.; Feng, J.-F.; et al. Variables Associated with Cognitive Function: An Exposome-Wide and Mendelian Randomization Analysis. Alzheimers Res. Ther. 2025, 17, 13. [Google Scholar] [CrossRef] [PubMed]
  35. Wild, C.P. Complementing the Genome with an “Exposome”: The Outstanding Challenge of Environmental Exposure Measurement in Molecular Epidemiology. Cancer Epidemiol. Biomark. Prev. 2005, 14, 1847–1850. [Google Scholar] [CrossRef]
  36. Chen, H.; Kwong, J.C.; Copes, R.; Tu, K.; Villeneuve, P.J.; van Donkelaar, A.; Hystad, P.; Martin, R.V.; Murray, B.J.; Jessiman, B.; et al. Living near Major Roads and the Incidence of Dementia, Parkinson’s Disease, and Multiple Sclerosis: A Population-Based Cohort Study. Lancet 2017, 389, 718–726. [Google Scholar] [CrossRef]
  37. Hussain, R.; Graham, U.; Elder, A.; Nedergaard, M. Air Pollution, Glymphatic Impairment, and Alzheimer’s Disease. Trends Neurosci. 2023, 46, 901–911. [Google Scholar] [CrossRef]
  38. Finch, C.E.; Kulminski, A.M. The Alzheimer’s Disease Exposome. Alzheimer’s Dement. 2019, 15, 1123–1132. [Google Scholar] [CrossRef]
  39. Granov, R.; Vedad, S.; Wang, S.-H.; Durham, A.; Shah, D.; Pasinetti, G.M. The Role of the Neural Exposome as a Novel Strategy to Identify and Mitigate Health Inequities in Alzheimer’s Disease and Related Dementias. Mol. Neurobiol. 2025, 62, 1205–1224. [Google Scholar] [CrossRef]
  40. Dagnino, S.; Macherone, A. Unraveling The Exposome: A Practical View; Springer International Publishing AG, part of Springer Nature 2019: Cham, Switzerland, 2019; ISBN 9783319893211. [Google Scholar]
  41. Vermeulen, R.; Schymanski, E.L.; Barabási, A.-L.; Miller, G.W. The Exposome and Health: Where Chemistry Meets Biology. Science 2020, 367, 392–396. [Google Scholar] [CrossRef]
  42. Calderón-Garcidueñas, L.; Azzarelli, B.; Acuna, H.; Garcia, R.; Gambling, T.M.; Osnaya, N.; Monroy, S.; Del Rosario Tizapantzi, M.; Carson, J.L.; Villarreal-Calderon, A.; et al. Air Pollution and Brain Damage. Toxicol. Pathol. 2002, 30, 373–389. [Google Scholar] [CrossRef]
  43. Calderon-Garciduenas, L.; Maronpot, R.R.; Torres-Jardon, R.; Henriquez-Roldan, C.; Schoonhoven, R.; Acuna-Ayala, H.; Villarreal-Calderon, A.; Nakamura, J.; Fernando, R.; Reed, W.; et al. DNA Damage in Nasal and Brain Tissues of Canines Exposed to Air Pollutants Is Associated with Evidence of Chronic Brain Inflammation and Neurodegeneration. Toxicol. Pathol. 2003, 31, 524–538. [Google Scholar] [CrossRef] [PubMed]
  44. Doty, R.L. The Olfactory Vector Hypothesis of Neurodegenerative Disease: Is It Viable? Ann. Neurol. 2008, 63, 7–15. [Google Scholar] [CrossRef]
  45. Ehsanifar, M.; Montazeri, Z.; Taheri, M.A.; Rafati, M.; Behjati, M.; Karimian, M. Hippocampal Inflammation and Oxidative Stress Following Exposure to Diesel Exhaust Nanoparticles in Male and Female Mice. Neurochem. Int. 2021, 145, 104989. [Google Scholar] [CrossRef] [PubMed]
  46. Zanchi, A.C.T.; Fagundes, L.S.; Barbosa, F.; Bernardi, R.; Rhoden, C.R.; Saldiva, P.H.N.; do Valle, A.C. Pre and Post-Natal Exposure to Ambient Level of Air Pollution Impairs Memory of Rats: The Role of Oxidative Stress. Inhal. Toxicol. 2010, 22, 910–918. [Google Scholar] [CrossRef] [PubMed]
  47. Lee, S.-H.; Chen, Y.-H.; Chien, C.-C.; Yan, Y.-H.; Chen, H.-C.; Chuang, H.-C.; Hsieh, H.-I.; Cho, K.-H.; Kuo, L.-W.; Chou, C.C.-K.; et al. Three Month Inhalation Exposure to Low-Level PM2.5 Induced Brain Toxicity in an Alzheimer’s Disease Mouse Model. PLoS ONE 2021, 16, e0254587. [Google Scholar] [CrossRef]
  48. You, R.; Ho, Y.-S.; Chang, R.C.-C. The Pathogenic Effects of Particulate Matter on Neurodegeneration: A Review. J. Biomed. Sci. 2022, 29, 15. [Google Scholar] [CrossRef]
  49. Calderón-Garcidueñas, L.; Kavanaugh, M.; Block, M.; D’Angiulli, A.; Delgado-Chávez, R.; Torres-Jardón, R.; González-Maciel, A.; Reynoso-Robles, R.; Osnaya, N.; Villarreal-Calderon, R.; et al. Neuroinflammation, Hyperphosphorylated Tau, Diffuse Amyloid Plaques, and Down-Regulation of the Cellular Prion Protein in Air Pollution Exposed Children and Young Adults. J. Alzheimer’s Dis. 2012, 28, 93–107. [Google Scholar] [CrossRef]
  50. González-Domínguez, R.; Jáuregui, O.; Queipo-Ortuño, M.I.; Andrés-Lacueva, C. Characterization of the Human Exposome by a Comprehensive and Quantitative Large-Scale Multianalyte Metabolomics Platform. Anal. Chem. 2020, 92, 13767–13775. [Google Scholar] [CrossRef]
  51. Zhang, P.; Carlsten, C.; Chaleckis, R.; Hanhineva, K.; Huang, M.; Isobe, T.; Koistinen, V.M.; Meister, I.; Papazian, S.; Sdougkou, K.; et al. Defining the Scope of Exposome Studies and Research Needs from a Multidisciplinary Perspective. Environ. Sci. Technol. Lett. 2021, 8, 839–852. [Google Scholar]
  52. Sillé, F.C.M.; Karakitsios, S.; Kleensang, A.; Koehler, K.; Maertens, A.; Prasse, C.; Quiros-Alcala, L.; Ramachandran, G.; Rappaport, S.M.; Rule, A.M.; et al. The Exposome—A New Approach for Risk Assessment. ALTEX 2020, 37, 3–23. [Google Scholar] [CrossRef]
  53. Rushing, B.R.; Thessen, A.E.; Soliman, G.A.; Ramesh, A.; Sumner, S.C.J. The Exposome and Nutritional Pharmacology and Toxicology: A New Application for Metabolomics. Exposome 2023, 3, osad008. [Google Scholar] [CrossRef]
  54. Rattan, S.I.S.; Kaur, G. Nutrition, Food and Diet in Health and Longevity: We Eat What We Are. Nutrients 2022, 14, 5376. [Google Scholar] [CrossRef]
  55. Dettmer, K.; Aronov, P.A.; Hammock, B.D. Mass Spectrometry-based Metabolomics. Mass. Spectrom. Rev. 2007, 26, 51–78. [Google Scholar] [CrossRef]
  56. Grand-Guillaume Perrenoud, A.; Guillarme, D.; Boccard, J.; Veuthey, J.-L.; Barron, D.; Moco, S. Ultra-High Performance Supercritical Fluid Chromatography Coupled with Quadrupole-Time-of-Flight Mass Spectrometry as a Performing Tool for Bioactive Analysis. J. Chromatogr. A 2016, 1450, 101–111. [Google Scholar] [CrossRef] [PubMed]
  57. Liebich, H.M.; Gesele, E. Profiling of Organic Acids by Capillary Gas Chromatography–Mass Spectrometry after Direct Methylation in Urine Using Trimethyloxonium Tetrafluoroborate. J. Chromatogr. A 1999, 843, 237–245. [Google Scholar] [CrossRef]
  58. Sasaki, K.; Sagawa, H.; Suzuki, M.; Yamamoto, H.; Tomita, M.; Soga, T.; Ohashi, Y. Metabolomics Platform with Capillary Electrophoresis Coupled with High-Resolution Mass Spectrometry for Plasma Analysis. Anal. Chem. 2019, 91, 1295–1301. [Google Scholar] [CrossRef] [PubMed]
  59. Chowdhury, C.R.; Kavitake, D.; Jaiswal, K.K.; Jaiswal, K.S.; Reddy, G.B.; Agarwal, V.; Shetty, P.H. NMR-Based Metabolomics as a Significant Tool for Human Nutritional Research and Health Applications. Food Biosci. 2023, 53, 102538. [Google Scholar] [CrossRef]
  60. Guyot, S.; Marnet, N.; Laraba, D.; Sanoner, P.; Drilleau, J.-F. Reversed-Phase HPLC Following Thiolysis for Quantitative Estimation and Characterization of the Four Main Classes of Phenolic Compounds in Different Tissue Zones of a French Cider Apple Variety (Malus Domestica Var. Kermerrien). J. Agric. Food Chem. 1998, 46, 1698–1705. [Google Scholar] [CrossRef]
  61. Mazanova, A.O. Development and Validation Of Immunoenzyme Test-System For Determination Of 25-Hydroxyvitamin D In Blood Serum. Biotechnol. Acta 2016, 9, 28–36. [Google Scholar] [CrossRef]
  62. Tabung, F.K.; Smith-Warner, S.A.; Chavarro, J.E.; Wu, K.; Fuchs, C.S.; Hu, F.B.; Chan, A.T.; Willett, W.C.; Giovannucci, E.L. Development and Validation of an Empirical Dietary Inflammatory Index. J. Nutr. 2016, 146, 1560–1570. [Google Scholar] [CrossRef]
  63. Neveu, V.; Moussy, A.; Rouaix, H.; Wedekind, R.; Pon, A.; Knox, C.; Wishart, D.S.; Scalbert, A. Exposome-Explorer: A Manually-Curated Database on Biomarkers of Exposure to Dietary and Environmental Factors. Nucleic Acids Res. 2017, 45, D979–D984. [Google Scholar] [CrossRef]
  64. Vieujean, S.; Caron, B.; Haghnejad, V.; Jouzeau, J.-Y.; Netter, P.; Heba, A.-C.; Ndiaye, N.C.; Moulin, D.; Barreto, G.; Danese, S.; et al. Impact of the Exposome on the Epigenome in Inflammatory Bowel Disease Patients and Animal Models. Int. J. Mol. Sci. 2022, 23, 7611. [Google Scholar] [CrossRef] [PubMed]
  65. Vojdani, A.; Vojdani, E. The Role of Exposomes in the Pathophysiology of Autoimmune Diseases I: Toxic Chemicals and Food. Pathophysiology 2021, 28, 513–543. [Google Scholar] [CrossRef] [PubMed]
  66. Dietary Patterns to Prevent and Manage Diet-Related Disease Across the Lifespan; Snair, M., Ed.; National Academies Press: Washington, DC, USA, 2023; ISBN 978-0-309-71329-0. [Google Scholar]
  67. Santos, L. The Impact of Nutrition and Lifestyle Modification on Health. Eur. J. Intern. Med. 2022, 97, 18–25. [Google Scholar] [CrossRef] [PubMed]
  68. Prior, R.L. Fruits and Vegetables in the Prevention of Cellular Oxidative Damage. Am. J. Clin. Nutr. 2003, 78, 570S–578S. [Google Scholar] [CrossRef]
  69. Rahaman, M.M.; Hossain, R.; Herrera-Bravo, J.; Islam, M.T.; Atolani, O.; Adeyemi, O.S.; Owolodun, O.A.; Kambizi, L.; Daştan, S.D.; Calina, D.; et al. Natural Antioxidants from Some Fruits, Seeds, Foods, Natural Products, and Associated Health Benefits: An Update. Food Sci. Nutr. 2023, 11, 1657–1670. [Google Scholar] [CrossRef]
  70. Landete, J.M. Dietary Intake of Natural Antioxidants: Vitamins and Polyphenols. Crit. Rev. Food Sci. Nutr. 2013, 53, 706–721. [Google Scholar] [CrossRef]
  71. Djaoudene, O.; Romano, A.; Bradai, Y.D.; Zebiri, F.; Ouchene, A.; Yousfi, Y.; Amrane-Abider, M.; Sahraoui-Remini, Y.; Madani, K. A Global Overview of Dietary Supplements: Regulation, Market Trends, Usage during the COVID-19 Pandemic, and Health Effects. Nutrients 2023, 15, 3320. [Google Scholar] [CrossRef]
  72. Khubchandani, J.; Batra, K. Diet Fads and Supplements: Navigating the Allure, Risks, and Reality. J. Med. Surg. Public Health 2024, 4, 100168. [Google Scholar] [CrossRef]
  73. Nohesara, S.; Abdolmaleky, H.M.; Dickerson, F.; Pinto-Tomás, A.A.; Jeste, D.V.; Thiagalingam, S. Maternal Gut Microbiome-Mediated Epigenetic Modifications in Cognitive Development and Impairments: A New Frontier for Therapeutic Innovation. Nutrients 2024, 16, 4355. [Google Scholar] [CrossRef]
  74. Gold, E.; Perez de Bronner, S.; Goday, P.S. Nutrition Considerations in the Transgender and Gender-diverse Patient. Nutr. Clin. Pract. 2024, 39, 366–372. [Google Scholar] [CrossRef] [PubMed]
  75. Shiels, P.G.; Painer, J.; Natterson-Horowitz, B.; Johnson, R.J.; Miranda, J.J.; Stenvinkel, P. Manipulating the Exposome to Enable Better Ageing. Biochem. J. 2021, 478, 2889–2898. [Google Scholar] [CrossRef] [PubMed]
  76. Młynarska, E.; Jakubowska, P.; Frąk, W.; Gajewska, A.; Sornowska, J.; Skwira, S.; Wasiak, J.; Rysz, J.; Franczyk, B. Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases. Nutrients 2024, 16, 3570. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, X.; Li, X.; Xia, B.; Jin, X.; Zou, Q.; Zeng, Z.; Zhao, W.; Yan, S.; Li, L.; Yuan, S.; et al. High-Fiber Diet Mitigates Maternal Obesity-Induced Cognitive and Social Dysfunction in the Offspring via Gut-Brain Axis. Cell Metab. 2021, 33, 923–938.e6. [Google Scholar] [CrossRef]
  78. Guzzardi, M.A.; Ederveen, T.H.A.; Rizzo, F.; Weisz, A.; Collado, M.C.; Muratori, F.; Gross, G.; Alkema, W.; Iozzo, P. Maternal Pre-Pregnancy Overweight and Neonatal Gut Bacterial Colonization Are Associated with Cognitive Development and Gut Microbiota Composition in Pre-School-Age Offspring. Brain Behav. Immun. 2022, 100, 311–320. [Google Scholar] [CrossRef]
  79. Contu, L.; Hawkes, C. A Review of the Impact of Maternal Obesity on the Cognitive Function and Mental Health of the Offspring. Int. J. Mol. Sci. 2017, 18, 1093. [Google Scholar] [CrossRef]
  80. Monthé-Drèze, C.; Rifas-Shiman, S.L.; Gold, D.R.; Oken, E.; Sen, S. Maternal Obesity and Offspring Cognition: The Role of Inflammation. Pediatr. Res. 2019, 85, 799–806. [Google Scholar] [CrossRef]
  81. Quan, W.; Xu, Y.; Luo, J.; Zeng, M.; He, Z.; Shen, Q.; Chen, J. Association of Dietary Meat Consumption Habits with Neurodegenerative Cognitive Impairment: An Updated Systematic Review and Dose–Response Meta-Analysis of 24 Prospective Cohort Studies. Food Funct. 2022, 13, 12590–12601. [Google Scholar] [CrossRef]
  82. Lockyer, S.; Spiro, A.; Berry, S.; He, J.; Loth, S.; Martinez-Inchausti, A.; Mellor, D.; Raats, M.; Sokolović, M.; Vijaykumar, S.; et al. How Do We Differentiate Not Demonise—Is There a Role for Healthier Processed Foods in an Age of Food Insecurity? Proceedings of a Roundtable Event. Nutr. Bull. 2023, 48, 278–295. [Google Scholar] [CrossRef]
  83. Bhardwaj, R.L.; Parashar, A.; Parewa, H.P.; Vyas, L. An Alarming Decline in the Nutritional Quality of Foods: The Biggest Challenge for Future Generations’ Health. Foods 2024, 13, 877. [Google Scholar] [CrossRef]
  84. McNamara, A.E.; Brennan, L. Potential of Food Intake Biomarkers in Nutrition Research. Proc. Nutr. Soc. 2020, 79, 487–497. [Google Scholar] [CrossRef]
  85. Vinci, G.; Prencipe, S.A.; Ruggieri, R.; Ruggeri, M. How Much Does Overnutrition Weigh? The Environmental and Social Impacts of Metabolic Food Waste in Italy. Sci. Total Environ. 2024, 947, 174420. [Google Scholar] [CrossRef] [PubMed]
  86. Franco, S.; Barbanera, M.; Moscetti, R.; Cicatiello, C.; Secondi, L.; Massantini, R. Overnutrition Is a Significant Component of Food Waste and Has a Large Environmental Impact. Sci. Rep. 2022, 12, 8166. [Google Scholar] [CrossRef]
  87. Serafini, M.; Toti, E. Unsustainability of Obesity: Metabolic Food Waste. Front. Nutr. 2016, 3, 40. [Google Scholar] [CrossRef]
  88. Unsal, P.; Guner, M.; Ozsurekci, C.; Balli, N.; Bas, A.O.; Ozturk, Y.; Dikmeer, A.; Burkuk, S.; Koca, M.; Balci, C.; et al. Prevalence of Nutrition Disorders and Nutrition-related Conditions in Older Patients with Alzheimer’s Disease. Nutr. Clin. Pract. 2023, 38, 1142–1153. [Google Scholar] [CrossRef]
  89. McGrattan, A.M.; McGuinness, B.; McKinley, M.C.; Kee, F.; Passmore, P.; Woodside, J.V.; McEvoy, C.T. Diet and Inflammation in Cognitive Ageing and Alzheimer’s Disease. Curr. Nutr. Rep. 2019, 8, 53–65. [Google Scholar] [CrossRef]
  90. Wang, J.; Song, Y.; Gao, M.; Bai, X.; Chen, Z. Neuroprotective Effect of Several Phytochemicals and Its Potential Application in the Prevention of Neurodegenerative Diseases. Geriatrics 2016, 1, 29. [Google Scholar] [CrossRef]
  91. Devassy, J.G.; Leng, S.; Gabbs, M.; Monirujjaman, M.; Aukema, H.M. Omega-3 Polyunsaturated Fatty Acids and Oxylipins in Neuroinflammation and Management of Alzheimer Disease. Adv. Nutr. 2016, 7, 905–916. [Google Scholar] [CrossRef]
  92. Dyńka, D.; Kowalcze, K.; Paziewska, A. The Role of Ketogenic Diet in the Treatment of Neurological Diseases. Nutrients 2022, 14, 5003. [Google Scholar] [CrossRef]
  93. Xie, G.; Tian, W.; Wei, T.; Liu, F. The Neuroprotective Effects of β-Hydroxybutyrate on Aβ-Injected Rat Hippocampus in Vivo and in Aβ-Treated PC-12 Cells in Vitro. Free Radic. Res. 2015, 49, 139–150. [Google Scholar] [CrossRef]
  94. Yubero-Serrano, E.M.; Garcia-Rios, A.; Delgado-Lista, J.; Delgado-Casado, N.; Perez-Martinez, P.; Rodriguez-Cantalejo, F.; Fuentes, F.; Cruz-Teno, C.; Tunez, I.; Tasset-Cuevas, I.; et al. Postprandial Effects of the Mediterranean Diet on Oxidant and Antioxidant Status in Elderly Men and Women. J. Am. Geriatr. Soc. 2011, 59, 938–940. [Google Scholar] [CrossRef] [PubMed]
  95. Grabska-Kobyłecka, I.; Szpakowski, P.; Król, A.; Książek-Winiarek, D.; Kobyłecki, A.; Głąbiński, A.; Nowak, D. Polyphenols and Their Impact on the Prevention of Neurodegenerative Diseases and Development. Nutrients 2023, 15, 3454. [Google Scholar] [CrossRef]
  96. van Lent, D.M.; Mesa, H.G.; Short, M.I.; Gonzales, M.M.; Aparicio, H.J.; Salinas, J.; Yuan, C.; Jacques, P.F.; Beiser, A.; Seshadri, S.; et al. Association between Dietary Inflammatory Index Score and Incident Dementia. Alzheimer’s Dement. 2024. [Google Scholar] [CrossRef] [PubMed]
  97. Gu, Y.; Manly, J.J.; Mayeux, R.P.; Brickman, A.M. An Inflammation-Related Nutrient Pattern Is Associated with Both Brain and Cognitive Measures in a Multiethnic Elderly Population. Curr. Alzheimer Res. 2018, 15, 493–501. [Google Scholar] [CrossRef]
  98. Duggan, M.R.; Butler, L.; Peng, Z.; Daya, G.N.; Moghekar, A.; An, Y.; Rapp, S.R.; Hayden, K.M.; Shadyab, A.H.; Natale, G.; et al. Plasma Proteins Related to Inflammatory Diet Predict Future Cognitive Impairment. Mol. Psychiatry 2023, 28, 1599–1609. [Google Scholar] [CrossRef] [PubMed]
  99. Agarwal, P.; Leurgans, S.E.; Agrawal, S.; Aggarwal, N.T.; Cherian, L.J.; James, B.D.; Dhana, K.; Barnes, L.L.; Bennett, D.A.; Schneider, J.A. Association of Mediterranean-DASH Intervention for Neurodegenerative Delay and Mediterranean Diets with Alzheimer Disease Pathology. Neurology 2023, 100, e2259–e2268. [Google Scholar] [CrossRef]
  100. Fernández-Sanz, P.; Ruiz-Gabarre, D.; García-Escudero, V. Modulating Effect of Diet on Alzheimer’s Disease. Diseases 2019, 7, 12. [Google Scholar] [CrossRef]
  101. Couch, C.A.; Ament, Z.; Patki, A.; Kijpaisalratana, N.; Bhave, V.; Jones, A.C.; Armstrong, N.D.; Cheung, K.L.; Kimberly, W.T.; Tiwari, H.K.; et al. The Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) Diet and Metabolites in Chronic Kidney Disease. Nutrients 2024, 16, 2458. [Google Scholar] [CrossRef]
  102. Ameen, A.O.; Freude, K.; Aldana, B.I. Fats, Friends or Foes: Investigating the Role of Short- and Medium-Chain Fatty Acids in Alzheimer’s Disease. Biomedicines 2022, 10, 2778. [Google Scholar] [CrossRef]
  103. Henderson, S.T.; Vogel, J.L.; Barr, L.J.; Garvin, F.; Jones, J.J.; Costantini, L.C. Study of the Ketogenic Agent AC-1202 in Mild to Moderate Alzheimer’s Disease: A Randomized, Double-Blind, Placebo-Controlled, Multicenter Trial. Nutr. Metab. 2009, 6, 31. [Google Scholar] [CrossRef]
  104. Ota, M.; Matsuo, J.; Ishida, I.; Takano, H.; Yokoi, Y.; Hori, H.; Yoshida, S.; Ashida, K.; Nakamura, K.; Takahashi, T.; 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, 232–236. [Google Scholar] [CrossRef]
  105. Zhou, Y.; Xie, L.; Schröder, J.; Schuster, I.S.; Nakai, M.; Sun, G.; Sun, Y.B.Y.; Mariño, E.; Degli-Esposti, M.A.; Marques, F.Z.; et al. Dietary Fiber and Microbiota Metabolite Receptors Enhance Cognition and Alleviate Disease in the 5xFAD Mouse Model of Alzheimer’s Disease. J. Neurosci. 2023, 43, 6460–6475. [Google Scholar] [CrossRef]
  106. Mazzei, G.; Ikegami, R.; Abolhassani, N.; Haruyama, N.; Sakumi, K.; Saito, T.; Saido, T.C.; Nakabeppu, Y. A High-Fat Diet Exacerbates the Alzheimer’s Disease Pathology in the Hippocampus of the AppNL−F/NL−F Knock-in Mouse Model. Aging Cell 2021, 20, e13429. [Google Scholar] [CrossRef] [PubMed]
  107. Soininen, H.; Solomon, A.; Visser, P.J.; Hendrix, S.B.; Blennow, K.; Kivipelto, M.; Hartmann, T. 36-Month LipiDiDiet Multinutrient Clinical Trial in Prodromal Alzheimer’s Disease. Alzheimer’s Dement. 2021, 17, 29–40. [Google Scholar] [CrossRef]
  108. Dilmore, A.H.; Martino, C.; Neth, B.J.; West, K.A.; Zemlin, J.; Rahman, G.; Panitchpakdi, M.; Meehan, M.J.; Weldon, K.C.; Blach, C.; et al. Effects of a Ketogenic and Low-Fat Diet on the Human Metabolome, Microbiome, and Foodome in Adults at Risk for Alzheimer’s Disease. Alzheimer’s Dement. 2023, 19, 4805–4816. [Google Scholar] [CrossRef]
  109. Li, L.; Wang, L.; Ding, W.; Wu, J.; Liu, F.; Liu, J.; Zhang, J.; Wang, J. The Improvement Effects of Sika Deer Antler Protein in an Alzheimer’s Disease Mouse Model via the Microbe–Gut–Brain Axis. Food Sci. Nutr. 2025, 13, e4656. [Google Scholar] [CrossRef] [PubMed]
  110. Prajapati, S.K.; Wang, S.; Mishra, S.P.; Jain, S.; Yadav, H. Protection of Alzheimer’s Disease Progression by a Human-Origin Probiotics Cocktail. Sci. Rep. 2025, 15, 1589. [Google Scholar] [CrossRef] [PubMed]
  111. Rezaei Asl, Z.; Sepehri, G.; Salami, M. Probiotic Treatment Improves the Impaired Spatial Cognitive Performance and Restores Synaptic Plasticity in an Animal Model of Alzheimer’s Disease. Behav. Brain Res. 2019, 376, 112183. [Google Scholar] [CrossRef]
  112. Athari Nik Azm, S.; Djazayeri, A.; Safa, M.; Azami, K.; Ahmadvand, B.; Sabbaghziarani, F.; Sharifzadeh, M.; Vafa, M. Lactobacilli and Bifidobacteria Ameliorate Memory and Learning Deficits and Oxidative Stress in β-Amyloid (1–42) Injected Rats. Appl. Physiol. Nutr. Metab. 2018, 43, 718–726. [Google Scholar] [CrossRef]
  113. Li, S.-C.; Hsu, W.-F.; Chang, J.-S.; Shih, C.-K. Combination of Lactobacillus Acidophilus and Bifidobacterium Animalis Subsp. Lactis Shows a Stronger Anti-Inflammatory Effect than Individual Strains in HT-29 Cells. Nutrients 2019, 11, 969. [Google Scholar] [CrossRef]
  114. Ricciarelli, R.; Fedele, E. Phosphodiesterase 4D: An Enzyme to Remember. Br. J. Pharmacol. 2015, 172, 4785–4789. [Google Scholar] [CrossRef] [PubMed]
  115. Fedele, E.; Ricciarelli, R. Memory Enhancers for Alzheimer’s Dementia: Focus on CGMP. Pharmaceuticals 2021, 14, 61. [Google Scholar] [CrossRef] [PubMed]
  116. Lundberg, J.O.; Weitzberg, E.; Gladwin, M.T. The Nitrate–Nitrite–Nitric Oxide Pathway in Physiology and Therapeutics. Nat. Rev. Drug. Discov. 2008, 7, 156–167. [Google Scholar] [CrossRef] [PubMed]
  117. Bondonno, C.P.; Croft, K.D.; Ward, N.; Considine, M.J.; Hodgson, J.M. Dietary Flavonoids and Nitrate: Effects on Nitric Oxide and Vascular Function. Nutr. Rev. 2015, 73, 216–235. [Google Scholar] [CrossRef]
  118. López, D.; Orta, X.; Casós, K.; Sáiz, M.P.; Puig-Parellada, P.; Farriol, M.; Mitjavila, M.T. Upregulation of Endothelial Nitric Oxide Synthase in Rat Aorta after Ingestion of Fish Oil-Rich Diet. Am. J. Physiol. -Heart Circ. Physiol. 2004, 287, H567–H572. [Google Scholar] [CrossRef]
  119. Ricciarelli, R.; Fedele, E. CAMP, CGMP and Amyloid β: Three Ideal Partners for Memory Formation. Trends Neurosci. 2018, 41, 255–266. [Google Scholar] [CrossRef]
  120. Sanders, O.; Rajagopal, L. Phosphodiesterase Inhibitors for Alzheimer’s Disease: A Systematic Review of Clinical Trials and Epidemiology with a Mechanistic Rationale. J. Alzheimers Dis. Rep. 2020, 4, 185–215. [Google Scholar] [CrossRef]
  121. Thal, L.J.; Salmon, D.P.; Lasker, B.; Bower, D.; Klauber, M.R. The Safety and Lack of Efficacy of Vinpocetine in Alzheimer’s Disease. J. Am. Geriatr. Soc. 1989, 37, 515–520. [Google Scholar] [CrossRef]
  122. Arlt, S.; Schwedhelm, E.; Kölsch, H.; Jahn, H.; Linnebank, M.; Smulders, Y.; Jessen, F.; Böger, R.H.; Popp, J. Dimethylarginines, Homocysteine Metabolism, and Cerebrospinal Fluid Markers for Alzheimer’s Disease. J. Alzheimer’s Dis. 2012, 31, 751–758. [Google Scholar] [CrossRef]
  123. Fonteh, A.N.; Harrington, R.J.; Tsai, A.; Liao, P.; Harrington, M.G. Free Amino Acid and Dipeptide Changes in the Body Fluids from Alzheimer’s Disease Subjects. Amino Acids 2007, 32, 213–224. [Google Scholar] [CrossRef]
  124. Ibáñez, C.; Simó, C.; Martín-Álvarez, P.J.; Kivipelto, M.; Winblad, B.; Cedazo-Mínguez, A.; Cifuentes, A. Toward a Predictive Model of Alzheimer’s Disease Progression Using Capillary Electrophoresis–Mass Spectrometry Metabolomics. Anal. Chem. 2012, 84, 8532–8540. [Google Scholar] [CrossRef] [PubMed]
  125. Ohtsuka, Y.; Nakaya, J. Effect of Oral Administration of L-Arginine on Senile Dementia. Am. J. Med. 2000, 108, 439. [Google Scholar] [CrossRef]
  126. Fonar, G.; Polis, B.; Meirson, T.; Maltsev, A.; Elliott, E.; Samson, A.O. Intracerebroventricular Administration of L-Arginine Improves Spatial Memory Acquisition in Triple Transgenic Mice via Reduction of Oxidative Stress and Apoptosis. Transl. Neurosci. 2018, 9, 43–53. [Google Scholar] [CrossRef] [PubMed]
  127. Minamisawa, M.; Sato, Y.; Ishiguro, E.; Taniai, T.; Sakamoto, T.; Kawai, G.; Saito, T.; Saido, T.C. Amelioration of Alzheimer’s Disease by Gut-Pancreas-Liver-Brain Interaction in an App Knock-In Mouse Model. Life 2021, 12, 34. [Google Scholar] [CrossRef] [PubMed]
  128. Martínez-González, K.; Serrano-Cuevas, L.; Almeida-Gutiérrez, E.; Flores-Chavez, S.; Mejía-Aranguré, J.M.; Garcia-delaTorre, P. Citrulline Supplementation Improves Spatial Memory in a Murine Model for Alzheimer’s Disease. Nutrition 2021, 90, 111248. [Google Scholar] [CrossRef]
  129. Rajendra, A.; Bondonno, N.P.; Rainey-Smith, S.R.; Gardener, S.L.; Hodgson, J.M.; Bondonno, C.P. Potential Role of Dietary Nitrate in Relation to Cardiovascular and Cerebrovascular Health, Cognition, Cognitive Decline and Dementia: A Review. Food Funct. 2022, 13, 12572–12589. [Google Scholar] [CrossRef]
  130. Morris, M.C.; Wang, Y.; Barnes, L.L.; Bennett, D.A.; Dawson-Hughes, B.; Booth, S.L. Nutrients and Bioactives in Green Leafy Vegetables and Cognitive Decline. Neurology 2018, 90, e214–e222. [Google Scholar] [CrossRef]
  131. Rajendra, A.; Bondonno, N.P.; Murray, K.; Zhong, L.; Rainey-Smith, S.R.; Gardener, S.L.; Blekkenhorst, L.C.; Ames, D.; Maruff, P.; Martins, R.N.; et al. Habitual Dietary Nitrate Intake and Cognition in the Australian Imaging, Biomarkers and Lifestyle Study of Ageing: A Prospective Cohort Study. Clin. Nutr. 2023, 42, 1251–1259. [Google Scholar] [CrossRef]
  132. Rajendra, A.; Bondonno, N.P.; Zhong, L.; Radavelli-Bagatini, S.; Murray, K.; Rainey-Smith, S.R.; Gardener, S.L.; Blekkenhorst, L.C.; Magliano, D.J.; Shaw, J.E.; et al. Plant but Not Animal Sourced Nitrate Intake Is Associated with Lower Dementia-Related Mortality in the Australian Diabetes, Obesity, and Lifestyle Study. Front. Nutr. 2024, 11, 1327042. [Google Scholar] [CrossRef]
  133. Hunt, T.; Pontifex, M.G.; Vauzour, D. (Poly)Phenols and Brain Health—beyond Their Antioxidant Capacity. FEBS Lett. 2024, 598, 2949–2962. [Google Scholar] [CrossRef]
  134. Zhang, Y.; Chen, J.; Qiu, J.; Li, Y.; Wang, J.; Jiao, J. Intakes of Fish and Polyunsaturated Fatty Acids and Mild-to-Severe Cognitive Impairment Risks: A Dose-Response Meta-Analysis of 21 Cohort Studies. Am. J. Clin. Nutr. 2016, 103, 330–340. [Google Scholar] [CrossRef] [PubMed]
  135. Arshad, N.; Visweswariah, S.S. Cyclic Nucleotide Signaling in Intestinal Epithelia: Getting to the Gut of the Matter. WIREs Syst. Biol. Med. 2013, 5, 409–424. [Google Scholar] [CrossRef]
  136. Azevedo, M.F.; Faucz, F.R.; Bimpaki, E.; Horvath, A.; Levy, I.; de Alexandre, R.B.; Ahmad, F.; Manganiello, V.; Stratakis, C.A. Clinical and Molecular Genetics of the Phosphodiesterases (PDEs). Endocr. Rev. 2014, 35, 195–233. [Google Scholar] [CrossRef]
  137. Ahmad, F.; Murata, T.; Shimizu, K.; Degerman, E.; Maurice, D.; Manganiello, V. Cyclic Nucleotide Phosphodiesterases: Important Signaling Modulators and Therapeutic Targets. Oral Dis. 2015, 21, e25–e50. [Google Scholar] [CrossRef]
  138. Lugnier, C. Cyclic Nucleotide Phosphodiesterase (PDE) Superfamily: A New Target for the Development of Specific Therapeutic Agents. Pharmacol. Ther. 2006, 109, 366–398. [Google Scholar] [CrossRef] [PubMed]
  139. Newton, A.C.; Bootman, M.D.; Scott, J.D. Second Messengers. Cold Spring Harb. Perspect. Biol. 2016, 8, a005926. [Google Scholar] [CrossRef] [PubMed]
  140. Bondarev, A.D.; Attwood, M.M.; Jonsson, J.; Chubarev, V.N.; Tarasov, V.V.; Liu, W.; Schiöth, H.B. Recent Developments of Phosphodiesterase Inhibitors: Clinical Trials, Emerging Indications and Novel Molecules. Front. Pharmacol. 2022, 13, 1057083. [Google Scholar] [CrossRef]
  141. Heckman, P.R.A.; Blokland, A.; Bollen, E.P.P.; Prickaerts, J. Phosphodiesterase Inhibition and Modulation of Corticostriatal and Hippocampal Circuits: Clinical Overview and Translational Considerations. Neurosci. Biobehav. Rev. 2018, 87, 233–254. [Google Scholar] [CrossRef]
  142. Gulisano, W.; Tropea, M.R.; Arancio, O.; Palmeri, A.; Puzzo, D. Sub-Efficacious Doses of Phosphodiesterase 4 and 5 Inhibitors Improve Memory in a Mouse Model of Alzheimer’s Disease. Neuropharmacology 2018, 138, 151–159. [Google Scholar] [CrossRef]
  143. Hesse, R.; Lausser, L.; Gummert, P.; Schmid, F.; Wahler, A.; Schnack, C.; Kroker, K.S.; Otto, M.; Tumani, H.; Kestler, H.A.; et al. Reduced CGMP Levels in CSF of AD Patients Correlate with Severity of Dementia and Current Depression. Alzheimers Res. Ther. 2017, 9, 17. [Google Scholar] [CrossRef]
  144. Kelly, M.P. Cyclic Nucleotide Signaling Changes Associated with Normal Aging and Age-Related Diseases of the Brain. Cell Signal 2018, 42, 281–291. [Google Scholar] [CrossRef] [PubMed]
  145. Yamamoto, M.; Götz, M.E.; Ozawa, H.; Luckhaus, C.; Saito, T.; Rösler, M.; Riederer, P. Hippocampal Level of Neural Specific Adenylyl Cyclase Type I Is Decreased in Alzheimer’s Disease. Biochim. Et Biophys. Acta (BBA) Mol. Basis Dis. 2000, 1535, 60–68. [Google Scholar] [CrossRef]
  146. Xiang, Y.; Naik, S.; Zhao, L.; Shi, J.; Ke, H. Emerging Phosphodiesterase Inhibitors for Treatment of Neurodegenerative Diseases. Med. Res. Rev. 2024, 44, 1404–1445. [Google Scholar] [CrossRef] [PubMed]
  147. Brandt, R.; Lee, G.; Teplow, D.B.; Shalloway, D.; Abdel-Ghany, M. Differential Effect of Phosphorylation and Substrate Modulation on Tau’s Ability to Promote Microtubule Growth and Nucleation. J. Biol. Chem. 1994, 269, 11776–11782. [Google Scholar] [CrossRef]
  148. Schneider, A.; Biernat, J.; von Bergen, M.; Mandelkow, E.; Mandelkow, E.-M. Phosphorylation That Detaches Tau Protein from Microtubules (Ser262, Ser214) Also Protects It against Aggregation into Alzheimer Paired Helical Filaments. Biochemistry 1999, 38, 3549–3558. [Google Scholar] [CrossRef]
  149. Villa, V.; Montalto, G.; Caudano, F.; Fedele, E.; Ricciarelli, R. Selective Inhibition of Phosphodiesterase 4D Increases Tau Phosphorylation at Ser214 Residue. BioFactors 2022, 48, 1111–1117. [Google Scholar] [CrossRef]
  150. Zhang, C.; Cheng, Y.; Wang, H.; Wang, C.; Wilson, S.P.; Xu, J.; Zhang, H.-T. RNA Interference-Mediated Knockdown of Long-Form Phosphodiesterase-4D (PDE4D) Enzyme Reverses Amyloid-Β42-Induced Memory Deficits in Mice. J. Alzheimer’s Dis. 2013, 38, 269–280. [Google Scholar] [CrossRef]
  151. Wang, C.; Yang, X.-M.; Zhuo, Y.-Y.; Zhou, H.; Lin, H.-B.; Cheng, Y.-F.; Xu, J.-P.; Zhang, H.-T. The Phosphodiesterase-4 Inhibitor Rolipram Reverses Aβ-Induced Cognitive Impairment and Neuroinflammatory and Apoptotic Responses in Rats. Int. J. Neuropsychopharmacol. 2012, 15, 749–766. [Google Scholar] [CrossRef]
  152. Kumari, S.; Bagri, K.; Deshmukh, R. Connecting Dots: Preclinical Foundations to Clinical Realities of PDE4 Inhibitors in Alzheimer’s Disease. Inflammopharmacology 2025, 33, 593–603. [Google Scholar] [CrossRef]
  153. Prickaerts, J.; Kerckhoffs, J.; Possemis, N.; van Overveld, W.; Verbeek, F.; Grooters, T.; Sambeth, A.; Blokland, A. Roflumilast and Cognition Enhancement: A Translational Perspective. Biomed. Pharmacother. 2024, 181, 117707. [Google Scholar] [CrossRef]
  154. Donders, Z.; Skorupska, I.J.; Willems, E.; Mussen, F.; Broeckhoven, J.V.; Carlier, A.; Schepers, M.; Vanmierlo, T. Beyond PDE4 Inhibition: A Comprehensive Review on Downstream CAMP Signaling in the Central Nervous System. Biomed. Pharmacother. 2024, 177, 117009. [Google Scholar] [CrossRef]
  155. Bonkale, W.L.; Winblad, B.; Ravid, R.; Cowburn, R.F. Reduced Nitric Oxide Responsive Soluble Guanylyl Cyclase Activity in the Superior Temporal Cortex of Patients with Alzheimer’s Disease. Neurosci. Lett. 1995, 187, 5–8. [Google Scholar] [CrossRef] [PubMed]
  156. BALTRONS, M.; PIFARRE, P.; FERRER, I.; CAROT, J.; GARCIA, A. Reduced Expression of NO-Sensitive Guanylyl Cyclase in Reactive Astrocytes of Alzheimer Disease, Creutzfeldt–Jakob Disease, and Multiple Sclerosis Brains. Neurobiol. Dis. 2004, 17, 462–472. [Google Scholar] [CrossRef] [PubMed]
  157. Ugarte, A.; Gil-Bea, F.; García-Barroso, C.; Cedazo-Minguez, Á.; Ramírez, M.J.; Franco, R.; García-Osta, A.; Oyarzabal, J.; Cuadrado-Tejedor, M. Decreased Levels of Guanosine 3′, 5′-monophosphate (c GMP) in Cerebrospinal Fluid (CSF) Are Associated with Cognitive Decline and Amyloid Pathology in A Lzheimer’s Disease. Neuropathol. Appl. Neurobiol. 2015, 41, 471–482. [Google Scholar] [CrossRef]
  158. Yew, D.T.; Wong, H.W.; Li, W.P.; Lai, H.W.L.; Yu, W.A. Nitric Oxide Synthase Neurons in Different Areas of Normal Aged and Alzheimer’s Brains. Neuroscience 1999, 89, 675–686. [Google Scholar] [CrossRef]
  159. Gunn, A.P.; Wong, B.X.; McLean, C.; Fowler, C.; Barnard, P.J.; Duce, J.A.; Roberts, B.R. Increased Glutaminyl Cyclase Activity in Brains of Alzheimer’s Disease Individuals. J. Neurochem. 2021, 156, 979–987. [Google Scholar] [CrossRef]
  160. Xu, M.; Zhang, D.; Luo, R.; Wu, Y.; Zhou, H.; Kong, L.; Bi, R.; Yao, Y. A Systematic Integrated Analysis of Brain Expression Profiles Reveals YAP1 and Other Prioritized Hub Genes as Important Upstream Regulators in Alzheimer’s Disease. Alzheimer’s Dement. 2018, 14, 215–229. [Google Scholar] [CrossRef]
  161. Sanders, O. Sildenafil for the Treatment of Alzheimer’s Disease: A Systematic Review. J. Alzheimers Dis. Rep. 2020, 4, 91–106. [Google Scholar] [CrossRef]
  162. Prieto, G.A.; Trieu, B.H.; Dang, C.T.; Bilousova, T.; Gylys, K.H.; Berchtold, N.C.; Lynch, G.; Cotman, C.W. Pharmacological Rescue of Long-Term Potentiation in Alzheimer Diseased Synapses. J. Neurosci. 2017, 37, 1197–1212. [Google Scholar] [CrossRef] [PubMed]
  163. Abouelmagd, M.E.; Abdelmeseh, M.; Elrosasy, A.; Saad, Y.H.; Alnajjar, A.Z.; Eid, M.; Hassan, A.; Abbas, A. Phosphodiesterase-5 Inhibitors Use and the Risk of Alzheimer’s Disease: A Systematic Review and Meta-Analysis. Neurol. Sci. 2024, 45, 5261–5270. [Google Scholar] [CrossRef]
  164. Crescioli, C.; Paronetto, M.P. The Emerging Role of Phosphodiesterase 5 Inhibition in Neurological Disorders: The State of the Art. Cells 2024, 13, 1720. [Google Scholar] [CrossRef]
  165. Justo, A.F.O.; Toscano, E.C. de B.; Farias-Itao, D.S.; Suemoto, C.K. The Action of Phosphodiesterase-5 Inhibitors on β-Amyloid Pathology and Cognition in Experimental Alzheimer’s Disease: A Systematic Review. Life Sci. 2023, 320, 121570. [Google Scholar] [CrossRef] [PubMed]
  166. Röhrig, T.; Pacjuk, O.; Hernández-Huguet, S.; Körner, J.; Scherer, K.; Richling, E. Inhibition of Cyclic Adenosine Monophosphate-Specific Phosphodiesterase by Various Food Plant-Derived Phytotherapeutic Agents. Medicines 2017, 4, 80. [Google Scholar] [CrossRef]
  167. Göttel, C.; Niesen, S.; Daub, V.; Werle, T.; Bakuradze, T.; Winterhalter, P.; Richling, E. In Vitro Inhibition of Phosphodiesterase 3B (PDE 3B) by Anthocyanin-Rich Fruit Juice Extracts and Selected Anthocyanins. Int. J. Mol. Sci. 2020, 21, 6934. [Google Scholar] [CrossRef]
  168. Zanforlin, E.; Zagotto, G.; Ribaudo, G. An Overview of New Possible Treatments of Alzheimer’s Disease, Based on Natural Products and Semi-Synthetic Compounds. Curr. Med. Chem. 2017, 24, 3749–3773. [Google Scholar] [CrossRef] [PubMed]
  169. HAYASHI, S.; OZAWA, H. Studies on 3, 7-Dimethyl-1-(5-Oxo-Hexyl)-Xanthine (BL 191). I. Cyclic 3’, 5’-Nucleotide Phosphodiesterase (PDE) and the Inhibitory Effect of BL 191 on PDE in Rat Brain and Heart. Chem. Pharm. Bull. 1974, 22, 587–593. [Google Scholar] [CrossRef] [PubMed]
  170. Rabe, K.; Magnussen, H.; Dent, G. Theophylline and Selective PDE Inhibitors as Bronchodilators and Smooth Muscle Relaxants. Eur. Respir. J. 1995, 8, 637–642. [Google Scholar] [CrossRef]
  171. Orhan, I.E.; Rauf, A.; Saleem, M.; Khalil, A.A. Natural Molecules as Talented Inhibitors of Nucleotide Pyrophosphatases/ Phosphodiesterases (PDEs). Curr. Top Med. Chem. 2022, 22, 209–228. [Google Scholar] [CrossRef]
  172. Singh, A.; Kumar Singh, N. Pre-Clinical Evidence-Based Neuroprotective Potential of Naringin against Alzheimer’s Disease-like Pathology: A Comprehensive Review. Curr. Pharm. Biotechnol. 2024, 25, 1112–1123. [Google Scholar] [CrossRef]
  173. Ribaudo, G.; Memo, M.; Gianoncelli, A. A Perspective on Natural and Nature-Inspired Small Molecules Targeting Phosphodiesterase 9 (PDE9): Chances and Challenges against Neurodegeneration. Pharmaceuticals 2021, 14, 58. [Google Scholar] [CrossRef]
  174. Giacobbe, J.; Benoiton, B.; Zunszain, P.; Pariante, C.M.; Borsini, A. The Anti-Inflammatory Role of Omega-3 Polyunsaturated Fatty Acids Metabolites in Pre-Clinical Models of Psychiatric, Neurodegenerative, and Neurological Disorders. Front. Psychiatry 2020, 11, 122. [Google Scholar] [CrossRef]
  175. Thompson, A.S.; Jennings, A.; Bondonno, N.P.; Tresserra-Rimbau, A.; Parmenter, B.H.; Hill, C.; Perez-Cornago, A.; Kühn, T.; Cassidy, A. Higher Habitual Intakes of Flavonoids and Flavonoid-Rich Foods Are Associated with a Lower Incidence of Type 2 Diabetes in the UK Biobank Cohort. Nutr. Diabetes 2024, 14, 32. [Google Scholar] [CrossRef]
  176. Dell’Agli, M.; Maschi, O.; Galli, G.V.; Fagnani, R.; Dal Cero, E.; Caruso, D.; Bosisio, E. Inhibition of Platelet Aggregation by Olive Oil Phenols via CAMP-Phosphodiesterase. Br. J. Nutr. 2008, 99, 945–951. [Google Scholar] [CrossRef]
  177. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.-I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. New Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef]
  178. Fekete, M.; Varga, P.; Ungvari, Z.; Fekete, J.T.; Buda, A.; Szappanos, Á.; Lehoczki, A.; Mózes, N.; Grosso, G.; Godos, J.; et al. The Role of the Mediterranean Diet in Reducing the Risk of Cognitive Impairement, Dementia, and Alzheimer’s Disease: A Meta-Analysis. Geroscience 2025, online ahead of print. [Google Scholar] [CrossRef]
  179. Duff, K. Transgenic Mouse Models of Alzheimer’s Disease: Phenotype and Mechanisms of Pathogenesis. Biochem. Soc. Symp. 2001, 67, 195–202. [Google Scholar] [CrossRef]
  180. Kosel, F.; Pelley, J.M.S.; Franklin, T.B. Behavioural and Psychological Symptoms of Dementia in Mouse Models of Alzheimer’s Disease-Related Pathology. Neurosci. Biobehav. Rev. 2020, 112, 634–647. [Google Scholar] [CrossRef] [PubMed]
  181. Li, X.; Quan, M.; Wei, Y.; Wang, W.; Xu, L.; Wang, Q.; Jia, J. Critical Thinking of Alzheimer’s Transgenic Mouse Model: Current Research and Future Perspective. Sci. China Life Sci. 2023, 66, 2711–2754. [Google Scholar] [CrossRef]
  182. De Plano, L.M.; Saitta, A.; Oddo, S.; Caccamo, A. Navigating Alzheimer’s Disease Mouse Models: Age-Related Pathology and Cognitive Deficits. Biomolecules 2024, 14, 1405. [Google Scholar] [CrossRef]
  183. Sharman, M.J.; Gyengesi, E.; Liang, H.; Chatterjee, P.; Karl, T.; Li, Q.-X.; Wenk, M.R.; Halliwell, B.; Martins, R.N.; Münch, G. Assessment of Diets Containing Curcumin, Epigallocatechin-3-Gallate, Docosahexaenoic Acid and α-Lipoic Acid on Amyloid Load and Inflammation in a Male Transgenic Mouse Model of Alzheimer’s Disease: Are Combinations More Effective? Neurobiol. Dis. 2019, 124, 505–519. [Google Scholar] [CrossRef]
  184. Braidy, N.; Essa, M.M.; Poljak, A.; Selvaraju, S.; Al-Adawi, S.; Manivasagm, T.; Thenmozhi, A.J.; Ooi, L.; Sachdev, P.; Guillemin, G.J. Consumption of Pomegranates Improves Synaptic Function in a Transgenic Mice Model of Alzheimer’s Disease. Oncotarget 2016, 7, 64589–64604. [Google Scholar] [CrossRef] [PubMed]
  185. Essa, M.M.; Subash, S.; Akbar, M.; Al-Adawi, S.; Guillemin, G.J. Long-Term Dietary Supplementation of Pomegranates, Figs and Dates Alleviate Neuroinflammation in a Transgenic Mouse Model of Alzheimer’s Disease. PLoS ONE 2015, 10, e0120964. [Google Scholar] [CrossRef]
  186. Essa, M.; Braidy, N.; Awlad-Thani, K.; Vaishnav, R.; Al-Asmi, A.; Guillemin, G.; Al-Adawi, S.; Subash, S. Diet Rich in Date Palm Fruits Improves Memory, Learning and Reduces Beta Amyloid in Transgenic Mouse Model of Alzheimer′s Disease. J. Ayurveda Integr. Med. 2015, 6, 111. [Google Scholar] [CrossRef]
  187. Subash, S.; Essa, M.M.; Al-Asmi, A.; Al-Adawi, S.; Vaishnav, R. Chronic Dietary Supplementation of 4% Figs on the Modification of Oxidative Stress in Alzheimer’s Disease Transgenic Mouse Model. Biomed. Res. Int. 2014, 2014, 1–8. [Google Scholar] [CrossRef]
  188. Subash, S.; Essa, M.M.; Braidy, N.; Al-Jabri, A.; Vaishnav, R.; Al-Adawi, S.; Al-Asmi, A.; Guillemin, G.J. Consumption of Fig Fruits Grown in Oman Can Improve Memory, Anxiety, and Learning Skills in a Transgenic Mice Model of Alzheimer’s Disease. Nutr. Neurosci. 2016, 19, 475–483. [Google Scholar] [CrossRef]
  189. Lim, G.P.; Calon, F.; Morihara, T.; Yang, F.; Teter, B.; Ubeda, O.; Salem, N.; Frautschy, S.A.; Cole, G.M. A Diet Enriched with the Omega-3 Fatty Acid Docosahexaenoic Acid Reduces Amyloid Burden in an Aged Alzheimer Mouse Model. J. Neurosci. 2005, 25, 3032–3040. [Google Scholar] [CrossRef]
  190. Zhuo, J.-M.; Praticò, D. Acceleration of Brain Amyloidosis in an Alzheimer’s Disease Mouse Model by a Folate, Vitamin B6 and B12-Deficient Diet. Exp. Gerontol. 2010, 45, 195–201. [Google Scholar] [CrossRef]
  191. Bernardo, A.; McCord, M.; Troen, A.M.; Allison, J.D.; McDonald, M.P. Impaired Spatial Memory in APP-Overexpressing Mice on a Homocysteinemia-Inducing Diet. Neurobiol. Aging 2007, 28, 1195–1205. [Google Scholar] [CrossRef]
  192. Zhuo, J.-M.; Portugal, G.; Kruger, W.; Wang, H.; Gould, T.; Pratico, D. Diet-Induced Hyperhomocysteinemia Increases Amyloid-beta Formation and Deposition in a Mouse Model of Alzheimer’s Disease. Curr. Alzheimer Res. 2010, 7, 140–149. [Google Scholar] [CrossRef]
  193. Huang, H.; Nie, S.; Cao, M.; Marshall, C.; Gao, J.; Xiao, N.; Hu, G.; Xiao, M. Characterization of AD-like Phenotype in Aged APPSwe/PS1dE9 Mice. Age 2016, 38, 303–322. [Google Scholar] [CrossRef]
  194. Rojanathammanee, L.; Puig, K.L.; Combs, C.K. Pomegranate Polyphenols and Extract Inhibit Nuclear Factor of Activated T-Cell Activity and Microglial Activation In Vitro and in a Transgenic Mouse Model of Alzheimer Disease. J. Nutr. 2013, 143, 597–605. [Google Scholar] [CrossRef] [PubMed]
  195. Choi, S.J.; Lee, J.-H.; Heo, H.J.; Cho, H.Y.; Kim, H.K.; Kim, C.-J.; Kim, M.O.; Suh, S.H.; Shin, D.-H. Punica Granatum Protects Against Oxidative Stress in PC12 Cells and Oxidative Stress-Induced Alzheimer’s Symptoms in Mice. J. Med. Food 2011, 14, 695–701. [Google Scholar] [CrossRef] [PubMed]
  196. Gong, Z.; Huang, J.; Xu, B.; Ou, Z.; Zhang, L.; Lin, X.; Ye, X.; Kong, X.; Long, D.; Sun, X.; et al. Urolithin A Attenuates Memory Impairment and Neuroinflammation in APP/PS1 Mice. J. Neuroinflammation. 2019, 16, 62. [Google Scholar] [CrossRef]
  197. González-Domínguez, R.; García-Barrera, T.; Vitorica, J.; Gómez-Ariza, J.L. Deciphering Metabolic Abnormalities Associated with Alzheimer’s Disease in the APP/PS1 Mouse Model Using Integrated Metabolomic Approaches. Biochimie 2015, 110, 119–128. [Google Scholar] [CrossRef]
  198. Smach, M.A.; Jacob, N.; Golmard, J.-L.; Charfeddine, B.; Lammouchi, T.; Ben Othman, L.; Dridi, H.; Bennamou, S.; Limem, K. Folate and Homocysteine in the Cerebrospinal Fluid of Patients with Alzheimer’s Disease or Dementia: A Case Control Study. Eur. Neurol. 2011, 65, 270–278. [Google Scholar] [CrossRef]
  199. Liu, H.; Tian, T.; Qin, S.; Li, W.; Zhang, X.; Wang, X.; Gao, Y.; Huang, G. Folic Acid Deficiency Enhances Abeta Accumulation in APP/PS1 Mice Brain and Decreases Amyloid-Associated MiRNAs Expression. J. Nutr. Biochem. 2015, 26, 1502–1508. [Google Scholar] [CrossRef] [PubMed]
  200. Pacheco-Quinto, J.; Rodriguez de Turco, E.B.; DeRosa, S.; Howard, A.; Cruz-Sanchez, F.; Sambamurti, K.; Refolo, L.; Petanceska, S.; Pappolla, M.A. Hyperhomocysteinemic Alzheimer’s Mouse Model of Amyloidosis Shows Increased Brain Amyloid β Peptide Levels. Neurobiol. Dis. 2006, 22, 651–656. [Google Scholar] [CrossRef]
  201. Yu, R.; Zhang, H.; Chen, R.; Lin, Y.; Xu, J.; Fang, Z.; Ru, Y.; Fan, C.; Wu, G. Fecal Microbiota Transplantation from Methionine-Restricted Diet Mouse Donors Improves Alzheimer’s Learning and Memory Abilities Through Short-Chain Fatty Acids. Foods 2025, 14, 101. [Google Scholar] [CrossRef]
  202. Griñán-Ferré, C.; Sarroca, S.; Ivanova, A.; Puigoriol-Illamola, D.; Aguado, F.; Camins, A.; Sanfeliu, C.; Pallàs, M. Epigenetic Mechanisms Underlying Cognitive Impairment and Alzheimer Disease Hallmarks in 5XFAD Mice. Aging 2016, 8, 664–684. [Google Scholar] [CrossRef]
  203. Reilly, A.M.; Tsai, A.P.; Lin, P.B.; Ericsson, A.C.; Oblak, A.L.; Ren, H. Metabolic Defects Caused by High-Fat Diet Modify Disease Risk through Inflammatory and Amyloidogenic Pathways in a Mouse Model of Alzheimer’s Disease. Nutrients 2020, 12, 2977. [Google Scholar] [CrossRef]
  204. Li, L.; Yang, C.; Jia, M.; Wang, Y.; Zhao, Y.; Li, Q.; Gong, J.; He, Y.; Xu, K.; Liu, X.; et al. Synbiotic Therapy with Clostridium Sporogenes and Xylan Promotes Gut-Derived Indole-3-Propionic Acid and Improves Cognitive Impairments in an Alzheimer’s Disease Mouse Model. Food Funct. 2024, 15, 7865–7882. [Google Scholar] [CrossRef] [PubMed]
  205. Lymperopoulos, D.; Dedemadi, A.-G.; Voulgari, M.-L.; Georgiou, E.; Dafnis, I.; Mountaki, C.; Panagopoulou, E.A.; Karvelas, M.; Chiou, A.; Karathanos, V.T.; et al. Corinthian Currants Promote the Expression of Paraoxonase-1 and Enhance the Antioxidant Status in Serum and Brain of 5xFAD Mouse Model of Alzheimer’s Disease. Biomolecules 2024, 14, 426. [Google Scholar] [CrossRef]
  206. Galeano, P.; de Ceglia, M.; Mastrogiovanni, M.; Campanelli, L.; Medina-Vera, D.; Campolo, N.; Novack, G.V.; Rosell-Valle, C.; Suárez, J.; Aicardo, A.; et al. The Effect of Fat Intake with Increased Omega-6-to-Omega-3 Polyunsaturated Fatty Acid Ratio in Animal Models of Early and Late Alzheimer’s Disease-like Pathogenesis. Int. J. Mol. Sci. 2023, 24, 17009. [Google Scholar] [CrossRef] [PubMed]
  207. Yang, E.; Wang, J.; Woodie, L.N.; Greene, M.W.; Kaddoumi, A. Oleocanthal Ameliorates Metabolic and Behavioral Phenotypes in a Mouse Model of Alzheimer’s Disease. Molecules 2023, 28, 5592. [Google Scholar] [CrossRef] [PubMed]
  208. Lamichhane, G.; Liu, J.; Lee, S.-J.; Lee, D.-Y.; Zhang, G.; Kim, Y. Curcumin Mitigates the High-Fat High-Sugar Diet-Induced Impairment of Spatial Memory, Hepatic Metabolism, and the Alteration of the Gut Microbiome in Alzheimer’s Disease-Induced (3xTg-AD) Mice. Nutrients 2024, 16, 240. [Google Scholar] [CrossRef]
  209. Broderick, T.L.; Rasool, S.; Li, R.; Zhang, Y.; Anderson, M.; Al-Nakkash, L.; Plochocki, J.H.; Geetha, T.; Babu, J.R. Neuroprotective Effects of Chronic Resveratrol Treatment and Exercise Training in the 3xTg-AD Mouse Model of Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 7337. [Google Scholar] [CrossRef]
  210. Paula, P.-C.; Angelica Maria, S.-G.; Luis, C.-H.; Gloria Patricia, C.-G. Preventive Effect of Quercetin in a Triple Transgenic Alzheimer’s Disease Mice Model. Molecules 2019, 24, 2287. [Google Scholar] [CrossRef]
  211. Dal-Pan, A.; Dudonné, S.; Bourassa, P.; Bourdoulous, M.; Tremblay, C.; Desjardins, Y.; Calon, F. Cognitive-Enhancing Effects of a Polyphenols-Rich Extract from Fruits without Changes in Neuropathology in an Animal Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2016, 55, 115–135. [Google Scholar] [CrossRef]
  212. Julien, C.; Tremblay, C.; Phivilay, A.; Berthiaume, L.; Émond, V.; Julien, P.; Calon, F. High-Fat Diet Aggravates Amyloid-Beta and Tau Pathologies in the 3xTg-AD Mouse Model. Neurobiol. Aging 2010, 31, 1516–1531. [Google Scholar] [CrossRef]
  213. Liang, Z.; Gong, X.; Zhao, Y.; Zhao, Y.; Yu, J.; Huang, T.; Yang, C.; Wu, L.; Huang, M.; Wang, X.; et al. Long-Term High-Fat Diet Consumption Aggravates Β-Amyloid Deposition and Tau Pathology Accompanied by Microglial Activation in an Alzheimer’s Disease Model. Mol. Nutr. Food. Res. 2024, 68, e2300669. [Google Scholar] [CrossRef]
  214. Tournissac, M.; Vandal, M.; Tremblay, C.; Bourassa, P.; Vancassel, S.; Emond, V.; Gangloff, A.; Calon, F. Dietary Intake of Branched-chain Amino Acids in a Mouse Model of Alzheimer’s Disease: Effects on Survival, Behavior, and Neuropathology. Alzheimer’s Dement.: Transl. Res. Clin. Interv. 2018, 4, 677–687. [Google Scholar] [CrossRef]
  215. Marwarha, G.; Claycombe-Larson, K.; Lund, J.; Schommer, J.; Ghribi, O. A Diet Enriched in Palmitate and Deficient in Linoleate Exacerbates Oxidative Stress and Amyloid-β Burden in the Hippocampus of 3xTg-AD Mouse Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2019, 68, 219–237. [Google Scholar] [CrossRef]
  216. Noristani, H.N.; Verkhratsky, A.; Rodríguez, J.J. High Tryptophan Diet Reduces CA1 Intraneuronal Β-amyloid in the Triple Transgenic Mouse Model of Alzheimer’s Disease. Aging Cell 2012, 11, 810–822. [Google Scholar] [CrossRef] [PubMed]
  217. Bubier, J.A.; Chesler, E.J.; Weinstock, G.M. Host Genetic Control of Gut Microbiome Composition. Mamm. Genome 2021, 32, 263–281. [Google Scholar] [CrossRef]
  218. Onisiforou, A.; Charalambous, E.G.; Zanos, P. Shattering the Amyloid Illusion: The Microbial Enigma of Alzheimer’s Disease Pathogenesis—From Gut Microbiota and Viruses to Brain Biofilms. Microorganisms 2025, 13, 90. [Google Scholar] [CrossRef]
Figure 1. The figure illustrates the common workflow to characterize dietary biomarkers.
Figure 1. The figure illustrates the common workflow to characterize dietary biomarkers.
Ijms 26 03015 g001
Figure 2. The figure illustrates the comparison between a healthy brain and an atrophic brain associated with AD. It highlights environmental and lifestyle factors influencing brain health, including a healthy diet and supplements as protective measures, versus air pollution and high-fat diets as risk factors for developing AD. Key pathological mechanisms linked to the exposome are shown, including neuroinflammation, gut–brain axis disruption, Aβ deposition, oxidative stress, and altered cAMP/cGMP pathways. The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license (https://smart.servier.com/, accessed on 21 January 2025).
Figure 2. The figure illustrates the comparison between a healthy brain and an atrophic brain associated with AD. It highlights environmental and lifestyle factors influencing brain health, including a healthy diet and supplements as protective measures, versus air pollution and high-fat diets as risk factors for developing AD. Key pathological mechanisms linked to the exposome are shown, including neuroinflammation, gut–brain axis disruption, Aβ deposition, oxidative stress, and altered cAMP/cGMP pathways. The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license (https://smart.servier.com/, accessed on 21 January 2025).
Ijms 26 03015 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Monaco, M.; Torazza, C.; Fedele, E.; Grilli, M. The Impact of the Exposome on Alzheimer’s Disease: The Influence of Nutrition. Int. J. Mol. Sci. 2025, 26, 3015. https://doi.org/10.3390/ijms26073015

AMA Style

Monaco M, Torazza C, Fedele E, Grilli M. The Impact of the Exposome on Alzheimer’s Disease: The Influence of Nutrition. International Journal of Molecular Sciences. 2025; 26(7):3015. https://doi.org/10.3390/ijms26073015

Chicago/Turabian Style

Monaco, Martina, Carola Torazza, Ernesto Fedele, and Massimo Grilli. 2025. "The Impact of the Exposome on Alzheimer’s Disease: The Influence of Nutrition" International Journal of Molecular Sciences 26, no. 7: 3015. https://doi.org/10.3390/ijms26073015

APA Style

Monaco, M., Torazza, C., Fedele, E., & Grilli, M. (2025). The Impact of the Exposome on Alzheimer’s Disease: The Influence of Nutrition. International Journal of Molecular Sciences, 26(7), 3015. https://doi.org/10.3390/ijms26073015

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

Article Metrics

Back to TopTop