Next Article in Journal
Polarizing Macrophage Functional Phenotype to Foster Cardiac Regeneration
Next Article in Special Issue
Comparison of Monoamine Oxidase-A, Aβ Plaques, Tau, and Translocator Protein Levels in Postmortem Human Alzheimer’s Disease Brain
Previous Article in Journal
Maytenus octogona Superoxide Scavenging and Anti-Inflammatory Caspase-1 Inhibition Study Using Cyclic Voltammetry and Computational Docking Techniques
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Aspects of a Diet as a New Pathway in the Prevention and Treatment of Alzheimer’s Disease

1
Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland
2
Staszica Mental Health Center, 15-071 Bialystok, Poland
3
Department of Biochemical Diagnostics, Medical University of Białystok, 15-089 Bialystok, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(13), 10751; https://doi.org/10.3390/ijms241310751
Submission received: 22 May 2023 / Revised: 19 June 2023 / Accepted: 26 June 2023 / Published: 28 June 2023
(This article belongs to the Special Issue Molecular Aspects of the Neurodegenerative Brain Diseases)

Abstract

:
Alzheimer’s disease is the most common cause of dementia in the world. Lack of an established pathology makes it difficult to develop suitable approaches and treatment for the disease. Besides known hallmarks, including amyloid β peptides cumulating in plaques and hyperphosphorylated tau forming NFTs, inflammation also plays an important role, with known connections to the diet. In AD, adhering to reasonable nutrition according to age-related principles is recommended. The diet should be high in neuroprotective foods, such as polyunsaturated fatty acids, antioxidants, and B vitamins. In addition, foods capable of rising BDNF should be considered because of the known profitable results of this molecule in AD. Adhering to beneficial diets might result in improvements in memory, cognition, and biomarkers and might even reduce the risk of developing AD. In this review, we discuss the effects of various diets, foods, and nutrients on brain health and possible connections to Alzheimer’s disease.

1. Alzheimer’s Disease

The most common cause of dementia in the world, Alzheimer’s disease (AD), puts an enormous burden on the healthcare system as a whole. Patients over the age of 65 are typically the ones who develop the illness. Every year, we see an increase in the number of impacted patients as a result of an aging society. By 2050, it is expected that there will be more than 100 million people worldwide suffering from AD dementia. It is a chronic disorder, since pathological processes begin at least 20 years before the onset of the disease. The hallmarks of AD are numerous, incompletely understood, and improperly categorized as part of the aging process. It is thought that the disease’s etiology may be influenced by both inherited and environmental factors. Despite the fact that several gene mutations are associated with AD, less than 5% of all cases are genetic. While early-onset AD (EOAD) is brought on by mutations in presenilin 1, presenilin 2, and amyloid precursor protein, sporadic, late-onset AD (LOAD) is associated with the APOE 4 gene (APP) [1]. The two main histological signs of AD are the gradual growth of neurofibrillary tangles (NFTs) formed of hyperphosphorylated tau and extracellular amyloid plaques. As a result, they cause loss of neurons and synapses [2].
As AD is currently incurable, preventative strategies are being actively discussed. The rates of clinical development of AD medications are currently limited, and the majority of medical research is focused on delaying progression rather than curing patients. It is because of the disease’s heterogeneity, underlying hallmarks, and pathogenesis, which are not yet completely understood. While there is currently no cure for Alzheimer’s disease, recent research has suggested that dietary interventions could help to prevent or delay the onset of disease.

2. Pathology of Alzheimer’s Disease

Amyloid buildup and tau protein hyperphosphorylation are the two primary pathogenic features of the illness. The aftereffects of these pathologic processes include neurodegeneration with loss of synapses and neurons, which results in macroscopical atrophy. Mixed pathology, which includes Lewy bodies and vascular disease, is a frequent occurrence, particularly in elderly adults [1]. The type 1 transmembrane protein known as amyloid precursor protein (APP) is expressed by a variety of cell types. Gamma- and beta-secretases in the central nervous system can successively cleave APP in two different ways [3]. The major components of incorrectly folded amyloid plaques, accumulating extracellularly, are Aβ40 and Aβ42, two products of APP metabolism. Aβ-42 is more prevalent than Aβ-40 inside plaques due to its higher incidence of fibrillization and insolubility. Aβ may then begin a series of processes that lead to synaptic loss and neuronal death by locating among healthy neurons and causing disruption in signaling, including neuroinflammation, and it is known that Aβ is capable of starting an immune response or creating angiopathy [4,5].
Neurons are built of a protein called tau, which assists in keeping the cytoskeleton’s microtubules intact in healthy cells [6]. It accumulates within nerve cell bodies as a result of the hyperphosphorylation that causes NFTs to tangle. Cellular proteins, which these tangles subsequently improperly engage with, are unable to carry out their typical functions [7]. A reduction in tau binding to microtubules leads to the malfunctioning of synapses. As a result of increased tau phosphorylation and intracellular tau aggregation brought on by an imbalance in tau kinase and phosphatase activity, NFTs are generated in AD patients. Last but not least, the growth of NFTs disrupts synaptic plasticity [7,8], which harms neuronal cells. According to research, Aβ accumulation may act as a trigger for the subsequent hyperphosphorylation process [9]. Moreover, there is evidence that toxic tau can boost Aβ synthesis via a feedback loop mechanism [10].
Neuroinflammation is a newly identified characteristic that is receiving a lot of attention. Even after more than 20 years of research, the processes of AD neuroinflammation are still not fully known. The two most important components of the inflammatory response in the brain are microglia and astroglia. Microglia can be of two kinds, which can be activated: M1 and M2. The M1 phenotype is regarded as “pro-inflammatory” and conventional, whereas the M2 phenotype is “anti-inflammatory” and unconventional [11]. Classical activation is brought on by the release of pro-inflammatory molecules such as IL-1, TNF, IL-6, and reactive oxygen species and is associated with pathogen defense mechanisms [12]. The M2 phenotype, which is induced by IL-4 and IL-13, on the other hand, releases neuroprotective agents such as TGF-β, IL-10, and IGF-1 [12]. By controlling inflammation, M2 microglia can help improve tissue remodeling and repair. It is noteworthy how rapidly the M1-to-M2 conversion can occur [13,14]. Moreover, senile plaque growth is promoted by the pro-inflammatory environment that active microglia surrounding the plaques generate [15,16]. These mechanisms are depicted in the Figure 1.
On the other hand, the neurotrophic factors are the growth factors promoting neuronal growth, differentiation, survival, and proper functioning in mature and developing nervous systems [17]. They interact with one of two receptors: p75 neurotrophin receptor (p75NTR), a member of the tumor necrosis factor (TNF) receptor superfamily, or tyrosine receptor kinase (Trk), one of the tropomyosin-related tyrosine kinase receptors. Neurotrophins (NGF binds to TrkA; BDNF and NT4/5 bind to TrkB; and NT3 binds to TrkC), which interact with certain Trk receptors, promote survival and growth responses. Additionally, their interaction with the P75 neurotrophic receptor (p75NTR) affects both apoptosis and brain plasticity [17]. Brain-derived neurotrophic factor (BDNF) is the most abundant neurotrophin and is responsible for promoting neuroprotection and neuroregeneration. This well-researched growth factor in the mammalian brain plays an important role in maintaining synaptic plasticity in learning and memory [18,19]. The majority of BDNF produced in the brain is delivered to presynaptic terminals and postsynaptic dendrites from the cell bodies of neurons and glial cells. The release of neurotransmitters, ion channel activity, axonal pathfinding, and neuronal excitability are all regulated by BDNF and its receptor, tropomyosin receptor kinase B (TrkB), which is localized at glutamate synapses [20]. Apoptosis, neuroinflammation, tau phosphorylation, and Aβ buildup are all linked to BDNF deficiency in the etiology of AD [21]. With the activation of TrkB and phosphatidylinositol 3-kinase (PI3K) signaling, BDNF stimulation causes tau to dephosphorylate [22]. By deregulating the glutamatergic N-methyl-d-aspartate receptor (NMDAR)/Ca2+/calpain signaling cascade, Aβ impairs BDNF function [23]. The extracellularly regulated kinase/cyclic AMP response element-binding protein (ERK/CREB) signaling pathway can increase BDNF, which can minimize the impact of Aβ on dendritic atrophy and neuronal death [24]. Increasing evidence also indicates the significance of BDNF signaling in regulating the long-term effects of Aβ buildup in AD. By controlling the neurotransmitters that are released (including glutamate and gamma-aminobutyric acid) after nuclear factor B (NF-B) activation, BDNF controls the relationship between inflammation and neuroplasticity [25]. Reduced BDNF levels were found in AD patients’ brains, blood, and cerebrospinal fluid [26,27,28] as the illness advanced. Moreover, increased cognitive performance in AD has been associated with elevated serum BDNF levels [18,29]. These results have raised interest in BDNF as a prospective biomarker for AD diagnosis or as treatment.

3. Diets and Life Situations Negatively Influencing the Disease

3.1. Obesity and Malnutrition

There is no dispute about the fact that obesity, one of the risk factors for AD, is connected to low-grade chronic inflammation. In fact, people with obesity or other metabolic illnesses have approximately a twofold-increased chance of getting AD, according to meta-analysis research [30,31]. Consuming a high-fat diet, which is abundant in high saturated fatty acids, has been linked to obesity but also to deficiencies and hippocampus-dependent learning and memory function [32,33]. Rats fed a high-fat diet depicted impaired memory, which progressed with further administration of the diet [34]. In addition, prospective epidemiological studies support the connection between high-fat diet and dementia [35]. Elevated levels of APP in adipose tissue and Aβ in plasma are two potential culprits in the development of AD in obese people. As indicated by Lee et al.’s research, increased adipocyte APP gene expression may be the cause of mid-life obesity-related increases in Aβ plasma levels. Increased transport of Aβ into the human brain may occur as a result of chronic elevation in Aβ plasma levels [31,36]. Furthermore, adipocytes and monocyte-derived macrophages in adipose tissue secrete acute-phase proteins, such as CRP, and pro-inflammatory cytokines, such as IL-6, IL-1, and TNF-alfa, which contribute to neuroinflammation [37]. Chronically consuming too many high-carbohydrate and high-saturated-fat meals while obese can have a negative influence on insulin production, which in response has a major impact on brain glucose metabolism and functioning. Loss of or diminished insulin sensitivity is one of the symptoms of insulin resistance. Reduced glycogen storage in muscle and fat cells and a decline in hepatic glucose absorption are both results of peripheral insulin resistance [31]. Peripheral insulin resistance triggers CNS insulin signaling to worsen, which then causes changes in cerebral metabolism. In fact, high insulin levels or peripheral insulin resistance lower insulin transporter levels and lower BBB permeability to insulin [38]. One of the variables underpinning the pathophysiology of AD has been identified as changes in insulin signaling in the brain [39]. According to this theory, several AD models and AD patients have decreased brain insulin receptor sensitivity, which may indicate the development of amyloid plaques in the brain.
On the other hand, in patients with AD, especially in those with moderate to severe AD, nutritional issues, particularly weight loss, are commonly observed. Moreover, it is a reliable indicator of worse outcomes in individuals suffering from the condition [40,41]. Recent research revealed that mild cognitive impairment (MCI) and early-stage AD are precursors to nutritional issues, such as appetite changes, weight loss, and sarcopenia [41,42,43]. Moreover, a prior study revealed that a low body mass index (BMI) indicates a higher risk of dementia after MCI [44]. Malnutrition issues appear to be significant though controllable aspects that may alter the prognosis of dementia, despite the fact that the precise processes behind nutritional issues in AD patients are not fully known. The biggest detrimental effects on brain function and mortality in individuals with cognitive impairment are undernutrition and weight loss, which accelerate the neurodegenerative process. In severe dementia, Hanson et al. have shown that weight loss is a predictor of mortality [45]. The most prevalent finding is that low BMI and malnutrition are associated with an increased risk of dementia and death, demonstrating the role of nutrition in the neurodegenerative process [46]. Furthermore, in patients with mild AD, malnutrition was associated with disease progression [47]. According to Yu et al., adults over 65 with a trend toward weight reduction should have their cognitive function constantly examined [48].

3.2. Diet High in Fats

Dietary fat can be divided into saturated and unsaturated dietary fats. Monounsaturated fatty acids (MUFAs) and polyunsaturated fatty acids (PUFAs), which include omega-3 fatty acids, are two categories of unsaturated fats. While MUFAs and PUFAs (“good fats”) are assumed to have positive benefits, including enhanced brain function and the avoidance of neurodegenerative diseases, saturated and trans fats (“bad fats”) are widely recognized to have deleterious impacts on human health, including correlations to AD. Unsaturated fats that have undergone partial hydrogenation, or trans fats, are used to lengthen the food shelf life span [49,50]. Evidence from studies on animals supports the harmful effects of saturated and trans fats on pathology and behavior related to AD. In a mouse model of AD (5xFAD), ingestion of a diet heavy in saturated and trans fats was enough to increase cerebrovascular Aβ deposition and hippocampus oxidative stress and promote cognitive impairment [51]. Furthermore, in an AD transgenic mouse model (APP/PS1), consumption of a diet high in fat was linked to memory impairment, a rise in Aβ monomers and plaques, and brain inflammation [50,52]. These revelations were confirmed by epidemiological studies according to which consuming saturated and trans fats increases the chance of developing Alzheimer’s disease and dementia [53,54]. A more recent meta-analysis thoroughly assessed four independent prospective cohort studies and depicted that high consumption of saturated fats is also strongly linked to an elevated risk of AD (39%) and dementia (105%) [55]. Interestingly, dietary lipids can affect how the CNS is in an inflammatory state. Microglia respond with inflammation to saturated fatty acids by secreting pro-inflammatory cytokines [56,57].

3.3. Western Diet

The phrase “Western diet” (WD) refers to a modern eating style that is typical to Western nations. It is based on ultra-processed foods that are ready for consumption; created using refined materials; and are high in salt, simple sugars, saturated fatty acids (SFAs), and cholesterol. Grain, fiber, and mono- and polyunsaturated fatty acids (MUFAs and PUFAs; particularly anti-inflammatory omega-3, -6, and -9 acids) are also insufficient in this dietary pattern. Moreover, the WD has a major negative impact on how the gut and its commensal bacteria operate, which indirectly reduces the absorption of minerals and vitamins from the diet [58,59]. There is a significant amount of proof that the WD can worsen cognition, learning, and memory in mice and humans, as well as aggravate or cause the pathogenic aspects of AD [60,61,62,63]. Two dietary elements, fat and sugar, were shown to have the most significant influence on hippocampal processes. When mice were fed a diet high in SFAs, sucrose/sugar, or both, for short or long periods, there were a variety of deficits in hippocampus-dependent learning and memory behavioral tests, which was comparable with evidence from individuals who ate following a diet high in SFAs for three days [64,65,66]. In addition, the WD is linked to a reduced total volume of the left hippocampus, which was described in research performed on 60- to 64-year-old men [59,67]. Additionally, research performed on animals confirmed the effects of the WD on the amyloidogenic accumulation of toxic Aβ peptides and tau phosphorylation. Overall, mice fed a Western diet showed a significant rise in typical biomarkers of AD, such as Aβ, Aβ plaques, tau, and ptau [68,69,70]. Moreover, biomarkers of inflammation such as TNF-alfa, IL-6, or IL-1beta were also enhanced in animals fed the WD [65,71,72]. These revelations support the negative influence of the WD on patients. Table 1 summarizes the effects of diets and life situations negatively influencing the disease.

4. Diets with Positive Influence

4.1. Mediterranean Diet

The Mediterranean diet (MeDi or MD) is a nutritional style that is popular in nations bordering the Mediterranean Sea, such as Greece or southern Italy. The traditional Mediterranean diet is characterized by high intake of fruits, vegetables, whole grains, nuts, and legumes; moderate intake of fish, poultry, and alcohol (especially red wine, with meals); and low intake of red and processed meats, with olive oil being the primary source of fat [73,74]. Considering that it contains all the necessary nutrients, such as monounsaturated fatty acids (primarily found in olive oil), polyunsaturated fatty acids (found in fatty fish), antioxidants (such as carotenoids, beta-carotene flavonoids, indoles, or lutein), vitamins (A, B types, D, and E), and minerals (calcium, iodine, magnesium, potassium, selenium, and zinc), it can be used as a nutritional model for healthy eating habits [75,76]. Many chronic illnesses have been linked to a preventive impact of the MeDi pattern.
Higher MD adherence has been linked to improved cognitive function [77,78], a slower pace of cognitive deterioration [79,80], and a decreased risk of cognitive impairment [81,82] but also AD itself [83,84]. Longitudinal research described by Anastaiou et al. depicted several connections between individual components of the MeDi and cognition [85]. For example, there was a reduction in the risk of dementia of 68.9% after every serving of fish per day [85]. The study by Ballarini et al. showed that lower Aβ42/40 ratio and pTau pathologies were observed in patients with higher adherence to a MeDi [86]. A newer randomized trial described the effects of the diet in mid-life on Alzheimer’s biomarkers. In cognitively normal people, Mediterranean-like diets improved cerebral blood flow; memory; and AD biomarkers, such as lowered Aβ40 levels and improved Aβ42/40 ratio. Compared with cognitively normal people, adults with MCI had an unusual pattern, with the Western diet having a positive impact on cerebrospinal fluid biomarkers and the MeDi elevating t-tau levels [87]. Interestingly, increasing the MeDi score, which describes adherence to the diet, in a study on Greek patients resulted in men with decreased risk of cognitive impairment but elevated risk in the women’s group [81].

4.2. DASH Diet

The DASH diet, which is similar to the Mediterranean diet in that it requires a substantial intake of plant-based foods, also restricts the consumption of SFAs, total fats, cholesterol, and salt. The DASH eating pattern encourages greater consumption of preventive nutrients such as K, Ca, Mg, fiber, and vegetable proteins while simultaneously encouraging lower intake of refined carbs and saturated fats [88]. It has been demonstrated that the DASH dietary pattern, which was created to prevent and treat hypertension, improves risk factors for cardiovascular disease (CVD), such as total cholesterol, and systolic and diastolic blood pressure [88,89].
Several studies examined the effects of the DASH diet on AD. In older adults with cognitive impairment without dementia (CIND), better verbal memory was present while adhering to the diet [90]. However, many studies describe no significant results of the DASH diet on AD [91,92,93].

4.3. MIND Diet

The Mediterranean–DASH Intervention for Neurodegenerative Delay diet, a cross between the Mediterranean and DASH diets, was created to stress nutrients linked to preventing dementia and to avoid components, such as saturated/hydrogenated fats, that have been linked to dementia [94]. The MIND diet suggests eating more than or equal to three servings of whole grains per day, which is the core of the diet [95]. The next important component of the diet is eating more than or equal to six servings of leafy green vegetables per week (in addition to one or more daily servings of other vegetables) [96]. Moreover, it also entails consuming more than or equal to two servings of berries per week [97], more than or equal to one serving of fish per week [98], more than or equal to two servings of poultry per week, more than three servings of beans per week, and more than or equal to five servings of nuts per week [99]. The MIND diet encourages the use of olive oil as the main source of fat [100] and permits one serving of wine or alcohol per day [94,101]. Moreover, this diet should not be based on red meat or processed food such as fast food and should avoid frying [94]. According to several prospective cohort studies, eating more vegetables was linked to a slower rate of cognitive decline, with green leafy vegetables showing the highest associations [96,102,103]. Due to its dietary elements, which have antioxidative, anti-inflammatory, and neuroprotective properties, the MIND diet may support brain health. Hosking et al.’s research indicated that the MIND diet was linked to a lower risk of 12-year cognitive impairment and that stronger adherence to the diet was linked to a 53% lower risk of impairment [103]. In models of adjusted logistic regression, the MIND diet was linked to a lower risk of cognitive deterioration over the course of 12 years [103]. Moreover, a decreased rate of cognitive impairment following stroke was linked to high MIND diet adherence [94].

4.4. Other Diets

4.4.1. Ketogenic Diet

Ketogenic diet (KD) refers to a high-fat, medium-protein, and low-carbohydrate diet that leads to a metabolic shift to ketosis [104]. Carbohydrates should be reduced to less than 10% of the diet, which triggers the shift from glucose metabolism to metabolizing fatty acids [105]. In neurodegenerative diseases, there is evidence that a ketogenic diet (KD) and/or exogenous ketone supplementation may be helpful in the treatment of AD patients [106]. A complicated interplay of metabolism, gut microbiome, and other mechanisms can regulate neuroinflammation in neurodegenerative diseases by activating multiple molecular and cellular pathways. Recent and accumulating studies on human and animal models have shown that the KD is beneficial in neurodegenerative diseases by modulating central and peripheral metabolism, mitochondrial function, inflammation, oxidative stress, autophagy, and the gut microbiome [107]. The effect of a ketogenic diet on the inhibition of AD development is multidirectional; the KD prevents chronic sleep deprivation (SD)-induced AD by inhibiting ferroptosis and improving the neuronal repair ability via the Sirt1/Nrf2 signaling pathway [108]. In the study by Xu et al., four months of KD improved spatial learning, spatial memory, and working memory in mice [109]. The authors observed an improvement in cognitive functions, which was associated with a restored number of neurons and synapses in both the hippocampus and the cortex. Ketogenic diet treatment also decreased amyloid plaque deposition and microglial activation, resulting in reduced neuroinflammation [109]. In clinical studies, the KD was associated with better cognition but also improved brain metabolism and AD biomarkers, with lowered tau concentrations and increased Aβ42 cerebrospinal fluid levels in patients with MCI [110]. Currently, it seems that the KD can provide therapeutic benefits to patients with neurological problems by effectively controlling the balance between pro- and antioxidant processes, and between pro-excitatory and inhibitory neurotransmitters and by modulating inflammation [111]. It is a common phenomenon that energy deprivation in neurological disorders, including Alzheimer’s disease, progresses rapidly. The ability of ketone bodies to stabilize mitochondrial energy metabolism makes them suitable intervening agents [112]. The ketogenic diet is a good candidate for adjuvant therapy, but its specific applicability depends on the type and the degree of the disease [104].

4.4.2. Fasting

Given the many health advantages of intermittent fasting (IF), it has lately grown in popularity, also considering its positive effects on AD. According to studies on the regulation of metabolic pathways, intermittent fasting (IF) has been shown to increase the life span and prevent or postpone the onset of age-related disorders. Improvements in glucose tolerance, lipid metabolism, and cognitive impairment are just a handful of the positive consequences of IF [113]. When examining the molecular processes, fasting has a significant impact on the neurochemistry and activity of the neural network, particularly in certain parts of the brain, such as brainstem, striatum, hypothalamus, and hippocampus. Several signaling pathways have been shown to play a role in the structural and functional adaptations of neuronal circuits to nutritional limitation, particularly low glucose and amino acid levels, including enhanced synaptic density and neurogenesis [75,114].
There are two types of IF: alternate day fasting (ADF) and time-restricted feeding (TRF). ADF involves 24 h of dietary restriction every other day, followed by a day of unlimited feeding. With no limitation on the quantity of food or nutrients consumed, TRF restricts daily meal consumption to 8 h or fewer. Animal experiments have also used other forms of IF, such as prolonged fasting (fasting for two or more days), the 5:2 strategy (2 fasting days interspersed with 5 days of regular feeding), and time-controlled fasting (40 h of fasting, 24 h of feeding, followed by 24 h of fasting and 80 h of feeding in each cycle) [113,115].
Although TRF is a more known type of food restriction diet and mainly used to lose weight, ADF has more positive outcomes relatively to AD. Clinical research has mainly concentrated on the improvement of ADF in obese individuals, which includes lowering weight and total cholesterol, promoting fat oxidation, and reducing insulin resistance [116,117]. In individuals who are normal weight or overweight, ADF is also helpful for cardioprotection as it decreases triacylglycerol concentrations and increases low-density lipoprotein (LDL) particle size [118]. In regards to body weight and heart disease, greater results were observed with ADF combined with exercise [119]. In the case of neurodegeneration, ADF is frequently used in mice models of neurodegeneration and aging. ADF improved synaptic plasticity, and learning and memory deficiencies in elderly mice by upregulating the expression of synaptophysin (Syn) and protein kinase CaMkinase (CaM) [120]. However, ADF affected mice models in different ways. ADF decreased the excessive amyloid buildup in the APP/PS1 animal model of AD, though it had no effect on amyloid deposition in 3xTg AD [121,122]. Furthermore, ADF enhanced neuroinflammatory responses in female 5xFAD, increased GABA signaling, and decreased neuronal function [123]. Nevertheless, after ADF intervention, the survival rate, spatial learning, and memory abilities in the AppNL-G-F mice model greatly improved, with no notable change in Aβ accumulation [124]. Moreover, in another mice model, IF has been linked to improvements in brain structure, such as a thicker CA1 pyramidal cell layer and greater expression of the dendritic protein drebrin in the hippocampus, as well as decreased oxidative stress [125].
In human studies so far, several researchers have highlighted the possible benefit of protein restriction against the aging process and aging-related chronic disorders. At this time, no research has been performed on protein or calorie restriction in human individuals diagnosed with AD [75,126]. It is difficult to imagine that severe food restrictions could be sustained for extended periods, particularly among elderly subjects with neurodegenerative diseases. However, short periods of caloric restriction were described as capable to enhance cognitive functions such as verbal memory, and 30 days of low-glycemic-index diet in patients with MCI resulted in the advancement of delayed visual memory and the CSF biomarker Aβ42 [127,128]. Positive results of the above-described diets are presented in Figure 2.

5. Beneficial Foods and Nutrients

For a long time, a wide range of human illnesses, including cancer, cardiovascular disease, respiratory diseases, infectious diseases, diabetes, obesity, metabolic syndromes, and neurological problems, have been successfully treated with natural remedies based on plants and herbs. These products have also been used to slow down the aging process. The majority of diets with numerous cell-protective properties, such as anti-inflammatory, antioxidant, and anti-arthritis properties, and capable of improving memory and cognitive abilities come from natural sources. Therefore, we aim to summarize the beneficial aspects of foods and nutrients with a known connection to AD.

5.1. Curcumin

Curcumin is a polyphenol found in turmeric and a spice with a unique yellow color that is commonly used in cooking. Due to its rich therapeutic qualities, turmeric has drawn a lot of interest among many natural medicines [129]. Curcumin may have anti-amyloid effects on AD, according to many lines of evidence. Firstly, curcumin inhibits Aβ aggregation as well as disaggregation to create fibrillar Aβ40 according to research performed in vitro [130]. Numerous in vivo investigations described that it promotes the disaggregation of existing amyloid deposits, prevents the formation of new ones, and even reduces the size of surviving deposits [131,132]. Moreover, curcumin and its derivatives suppress the synthesis of fibrillar Aβ from Aβ monomers and destabilize preformed fibrillar Aβ in vitro, showing that curcumin is protective against Aβ toxicity [133]. According to existing theories, curcumin works primarily by inhibiting NFkB, which is accomplished by preventing IkB phosphorylation and subsequent NFkB activation [134]. Curcumin can also prevent APP-cleaving enzymes such as B-secretase from functioning (BACE-1) [135,136].
Although animal studies show promising results, research performed on humans is limited and not trustworthy, which impedes the understanding of the results [137,138,139]. Only a modest number of clinical research studies have discussed the impact of curcumin on human cognitive performance, with conflicting results. While several research studies claim that curcumin has no beneficial effects on cognitive function [140,141], others show valuable results in protecting against cognitive decline [137,138]. Furthermore, neuroimaging demonstrates that curcumin could decrease Aβ brain deposits [137].

5.2. Coffee and Tea

Coffee is a beverage that contains a great amount of polyphenols. Moreover, it is known that caffeine increases human information processing, attentiveness, and reaction time [142]. The positive outcomes of caffeine are connected to antagonizing A1 adenosine receptors in the hippocampus and cortex [143]. Caffeine has well-known anti-inflammatory characteristics that include reducing the invasion of immune cells from the periphery, attenuating pro-inflammatory mediators, and inhibiting microglia activation. Greater plasma caffeine concentrations were associated with lower levels of inflammatory cytokines in the hippocampal region [56,144]. Animal studies imply that coffee with caffeine has a crucial function, since it lowers plasma Aβ levels, as opposed to caffeine-free coffee [145]. A meta-analysis by Wu et al. has shown that a diminished risk of development of dementias in humans (including AD) was observed with daily consumption of one–two cups of coffee [146]. Interestingly, research part of the Italian Longitudinal Study on Aging (ILSA) suggested that older people with normal cognitive function who increased their coffee consumption had a higher rate of developing MCI, while constant-over-time, moderate coffee consumption was linked to a lower rate of MCI incidence [147,148]. However, research does not support the theory that caffeine could prevent AD [149], although as reported in epidemiological research, drinking coffee and caffeine can possibly help treat AD [147].
One of the most popular beverages worldwide is tea, as it is the most consumed, second only to water. It is a byproduct of the plant Camellia sinensis (L.) O. Kuntze. Regardless of how much fermentation has occurred, green tea, black tea, and oolong tea are all produced from the same plant [150,151]. The health advantages of green tea have been the subject of research the most, including its effects on diabetes, cancer, cardiovascular diseases, and neurological disorders [152,153]. It mainly contains polyphenols. Despite the fact that the mechanisms behind green tea’s preventative impact are not fully understood, there are some hypotheses. It has been shown that oxidative stress has a role in the etiology of both AD and vascular dementia (VaD) [154,155]. Firstly, green tea catechins working in the brain have antioxidant properties with known strong free radical scavenger abilities [156]. Then, green tea polyphenols have an anti-inflammatory effect via the inhibition of nuclear factor kappa-beta activation [157]. Interestingly, epigallocatechin gallate (EGCG), the main component of green tea catechins, has shown inhibitory properties against Aβ accumulation [154,158]. Mice models of AD fed EGCG for weeks depicted an indicative decrease in Aβ aggregation and lowered neurotoxicity associated with amyloid [159]. Moreover, based on research by Tomata et al., drinking green tea frequently was substantially linked with having a low risk of dementia [160]. In a rat model of Alzheimer’s disease, green tea was found to improve memory and prevent oxidative stress and damage to the hippocampus [161].

5.3. Cocoa

Theobroma cocoa beans, which are often consumed by humans, are used to make cocoa (powder) and chocolate. Such products are abundant in flavonoids, the greatest category of dietary polyphenols and antioxidant compounds [162,163]. Cocoa beans are reported to have more antioxidant activity than green tea, red wine, or blueberries [164]. The polyphenol content in cocoa and chocolate is not the only difference between them; cocoa is a ground-up form of cocoa beans, whereas chocolate is a mixture of cocoa, cocoa butter, sugar, and other components that are combined to create a solid food product [165]. Chocolate has varying amounts of cocoa, which vary from 30–70% in dark chocolate to 7–15% in milk chocolate [166]. Additionally, the bioavailability of cocoa flavanols tends to be regulated by the complexity of the food matrix; therefore, consuming dark chocolate may result in less effective absorption of these chemicals than ingesting cocoa powder [163]. Human studies have demonstrated that consuming a particular combination of flavanols in cocoa can lower blood pressure, enhance insulin sensitivity, and improve cerebral perfusion and blood flow [167,168,169]. Research on the effects of cocoa in the rat pheochromocytoma PC12 cell line treated with Aβ(25–35) has shown that cocoa flavonoids have neuroprotective effects, preventing Aβ-induced cell death [170,171]. Interestingly, the findings of mouse models show that cocoa extracts may successfully stop the oligomerization of Aβ [172]. These revelations prompted researchers to conduct the study called CoCoA Study (Cocoa, Cognition, and Aging). They investigated the possibility that dietary flavanols might enhance cognitive performance in participants with MCI [173]. Desideri et al. showed that consuming a beverage fortified with cocoa flavanols (CFs) improved cognitive functions in adults diagnosed with MCI and lowered blood pressure and insulin resistance [173]. The results were confirmed by a randomized controlled trial showing comparable outcomes [168].

5.4. Vitamins

It was observed that patients’ cognitive abilities can be enhanced by having them consume a diet high in vitamins, especially those with antioxidant properties. Oxidative stress causes the reduction of O2 to H2O in mitochondria, which promotes the production of reactive oxygen species (ROS). As a consequence of their high reactivity, they interact with many molecules and structures. All macromolecules are observed to be oxidized quite early in AD patients’ brains. In AD, oxidation occurs in lipids, proteins, nucleic acids, and polysaccharides. Therefore, antioxidants such as vitamins are widely discussed as beneficial in AD [174]. Vitamins from the B family vary in terms of functions and structure. Folates play a significant role in the etiology of neurological illnesses by providing the methyl groups required for DNA methylation [175,176]. Homocysteine by itself is related to increases in cardiovascular risk and cognitive impairment. The metabolism of homocysteine is linked to the positive effect of vitamin B on cognitive performance. There is a reduction in DNA methylation in AD patients, and insufficiency of these vitamins leads to a buildup of homocysteine in the body that affects the genes connected to AD with their overexpression [176]. Therefore, researchers studied the supplementation of vitamin B, and the results showed decreased concentrations of homocysteine with simultaneous better results in MMSE, and semantic and episodic memory [177]. The results were tested in AD patients with similar results while supplementing folic acid [178]. However, the results are not consistent, with several publications not documenting a significant impact on cognitive decline or even documenting no connection [179,180]. In conclusion, the examination of individual articles demonstrates that the impact of nutritional intervention in the form of dietary supplementation with vitamins B appears to be ambiguous. High blood levels of vitamin B12 may be a protective factor, even if it has been noted that testing for the presence of folate and vitamin B12 is unreliable [175].
High frequency of vitamin D hypovitaminosis, particularly in the elderly, is caused by dietary habits and inadequate sun exposure. According to recent studies, more than 50% of central Europeans over 60 may not be getting sufficient amounts of vitamin D. Vitamin D supplementation seems to be especially beneficial for this sensitive age group, since vitamin D hypovitaminosis is linked to numerous disorders, including AD [181,182,183]. It is described that it can double the risk of dementia and increase the risk of developing AD by 21% [184,185]. Ghahremani et al. showed that when compared with no exposure, dementia incidence was 40% lower when vitamin D was provided [186]. Moreover, the benefits of vitamin D were much stronger in women than in men and in those with normal cognition than in people with mild cognitive impairment, and apolipoprotein E4 non-carriers had much stronger vitamin D effects than carriers [186]. Several research studies found changed plasma Aβ1-40 levels, slower development of AD symptomatology, and lowered Aβ-related biomarkers following vitamin D treatment, which would indicate a lower risk of AD [187,188].
Vitamin E is a fat-soluble vitamin that shows neuroprotective and anti-inflammatory properties [189,190]. Therefore, researchers have considered it for potential therapeutic usage in AD. A meta-analysis by Lopes da Silva et al. showed lower plasma levels of vitamin E in AD patients in comparison to cognitively healthy controls; moreover, they came to the conclusion that the patients’ malnutrition was not the cause of this result [191]. This was also confirmed by a newer and bigger study by Mullan et al. [192]. Additionally, a number of prospective cohort studies examined plasma vitamin E levels and the probability of developing AD, with results showing significant correlation with higher levels of vitamin E and reduced risk of cognitive impairment or even reduced risk of developing AD [193,194]. Thus, supplementing this vitamin was also studied, with interesting evidence. Greater dietary vitamin E intake was linked to a reduced risk of AD, although the effect was only shown in people who did not have the ApoE4 risk allele [195,196]. Interestingly, the same results were not obtained when supplementation of this vitamin was non-dietary [195]. In randomized trials, supplementing α-tocopherol alone led to a slower functional decline compared with the placebo group. Furthermore, this study also showed that the combination of memantine and α-tocopherol was not as beneficial as α-tocopherol alone [197]. However, many studies show no changes in patients’ behavior, biomarkers, or cognitive decline; therefore, the connection between vitamin E and treating AD is still being discussed [198,199].

5.5. Foods Rising BDNF

As described above, BDNF is an interesting factor playing a role in the brain also in AD. Specifically altering patients’ diet may be a useful strategy for influencing BDNF and maintaining or even enhancing cognitive and metabolic health. Thus, numerous intervention trials with the goal of increasing BDNF levels have been conducted as a result of the fact that higher levels are linked to better cognitive function [200]. Although various research studies have shown no significant increase in BDNF in connection with dietary interventions, there are some with interesting outcomes [117,201]. Suzuki et al. reported that in female MCI patients, consuming mold-fermented cheese significantly increased serum BDNF levels as compared with non-mold-fermented cheese administration [202]. In healthy subjects, intake of kernel-based whole grain (WG) rye products was described as capable to increase BDNF concentrations [203]. In addition, zinc supplementation had beneficial effects on women with premenstrual syndrome and obese ones [204,205]. Moreover, curcumin, further to the above-described beneficial effects on AD itself, was also described as influencing BDNF levels in the plasma of depressed patients [206]. However, this area needs more research, especially in connection to AD.

6. Summary

Overall, there is growing evidence that the molecular aspects of a diet can play an important role in the prevention and treatment of Alzheimer’s disease. By consuming a diet that is rich in omega-3 fatty acids, antioxidants, and plant-based foods, individuals may be able to reduce their risk of developing Alzheimer’s disease and slow the progression of the disease if it does occur. In our article, we discuss the potential of a specific diet and diet interventions to prevent and treat Alzheimer’s disease. We reviewed the literature on the role of diet in Alzheimer’s disease and propose new aspects for preventing and treating the disease based on the molecular aspects of a specific diet. Research performed on diets rich in fruits, vegetables, whole grains, legumes, nuts, fish, and olive oil suggests that diet can modify the molecular pathways involved in Alzheimer’s disease by reducing inflammation, oxidative stress, and insulin resistance. We also discuss diets and life situations negatively influencing the disease. Research shows that both obesity and malnutrition have a significantly bad influence on the course of AD or result in a higher chance of developing the disease. Moreover, diets high in fat and sugar intake contribute to higher levels of AD and inflammatory biomarkers. This review suggests an important role of food intake in the course of the disease and suggests potential dietary intervention in the treatment of AD.

Author Contributions

All authors produced the idea of the study. J.D. and B.M. coordinated project funding. J.D., J.M. and P.R. drafted the manuscript, and B.M. substantively revised it; visualization, J.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Medical University of Bialystok, Poland; grant number B.SUB.23.165 and B.SUB.23.500.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Mendez, M.F. Early-Onset Alzheimer’s Disease. Neurol. Clin. 2017, 35, 263. [Google Scholar] [CrossRef] [Green Version]
  2. Khan, S.; Barve, K.H.; Kumar, M.S. Recent Advancements in Pathogenesis, Diagnostics and Treatment of Alzheimer’s Disease. Curr. Neuropharmacol. 2020, 18, 1106. [Google Scholar] [CrossRef]
  3. O’Brien, R.J.; Wong, P.C. Amyloid Precursor Protein Processing and Alzheimer’s Disease. Annu. Rev. Neurosci. 2011, 34, 185–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Reiss, A.B.; Arain, H.A.; Stecker, M.M.; Siegart, N.M.; Kasselman, L.J. Amyloid Toxicity in Alzheimer’s Disease. Rev. Neurosci. 2018, 29, 613–627. [Google Scholar] [CrossRef] [PubMed]
  5. Ashrafian, H.; Zadeh, E.H.; Khan, R.H. Review on Alzheimer’s Disease: Inhibition of Amyloid Beta and Tau Tangle Formation. Int. J. Biol. Macromol. 2021, 167, 382–394. [Google Scholar] [CrossRef]
  6. Weingarten, M.D.; Lockwood, A.H.; Hwo, S.Y.; Kirschner, M.W. A Protein Factor Essential for Microtubule Assembly. Proc. Natl. Acad. Sci. USA 1975, 72, 1858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Naseri, N.N.; Wang, H.; Guo, J.; Sharma, M.; Luo, W. The Complexity of Tau in Alzheimer’s Disease. Neurosci. Lett. 2019, 705, 183. [Google Scholar] [CrossRef] [PubMed]
  8. Martin, L.; Latypova, X.; Wilson, C.M.; Magnaudeix, A.; Perrin, M.-L.; Yardin, C.; Terro, F. Tau Protein Kinases: Involvement in Alzheimer’s Disease. Ageing Res. Rev. 2013, 12, 289–309. [Google Scholar] [CrossRef]
  9. Bloom, G.S. Amyloid-β and Tau: The Trigger and Bullet in Alzheimer Disease Pathogenesis. JAMA Neurol. 2014, 71, 505–508. [Google Scholar] [CrossRef] [Green Version]
  10. Huang, H.C.; Jiang, Z.F. Accumulated Amyloid-Beta Peptide and Hyperphosphorylated Tau Protein: Relationship and Links in Alzheimer’s Disease. J. Alzheimers Dis. 2009, 16, 15–27. [Google Scholar] [CrossRef]
  11. Wang, C.; Wang, Q.; Lou, Y.; Xu, J.; Feng, Z.; Chen, Y.; Tang, Q.; Zheng, G.; Zhang, Z.; Wu, Y.; et al. Salidroside Attenuates Neuroinflammation and Improves Functional Recovery after Spinal Cord Injury through Microglia Polarization Regulation. J. Cell Mol. Med. 2018, 22, 1148–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Tang, Y.; Le, W. Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Mol. Neurobiol. 2015, 53, 1181–1194. [Google Scholar] [CrossRef] [PubMed]
  13. Jin, X.; Liu, M.-Y.Y.; Zhang, D.-F.F.; Zhong, X.; Du, K.; Qian, P.; Gao, H.; Wei, M.-J.J. Natural Products as a Potential Modulator of Microglial Polarization in Neurodegenerative Diseases. Pharmacol. Res. 2019, 145, 104253. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, Z.Z.; Zhang, Z.Z.; Lu, H.; Yang, Q.; Wu, H.; Wang, J. Microglial Polarization and Inflammatory Mediators After Intracerebral Hemorrhage. Mol. Neurobiol. 2017, 54, 1874–1886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Frank, S.; Burbach, G.J.; Bonin, M.; Walter, M.; Streit, W.; Bechmann, I.; Deller, T. TREM2 Is Upregulated in Amyloid Plaque-Associated Microglia in Aged APP23 Transgenic Mice. Glia 2008, 56, 1438–1447. [Google Scholar] [CrossRef]
  16. Hickman, S.E.; El Khoury, J. TREM2 and the Neuroimmunology of Alzheimer’s Disease. Biochem. Pharmacol. 2014, 88, 495–498. [Google Scholar] [CrossRef] [Green Version]
  17. Nasrolahi, A.; Javaherforooshzadeh, F.; Jafarzadeh-Gharehziaaddin, M.; Mahmoudi, J.; Asl, K.D.; Shabani, Z. Therapeutic Potential of Neurotrophic Factors in Alzheimer’s Disease. Mol. Biol. Rep. 2022, 49, 2345–2357. [Google Scholar] [CrossRef]
  18. Gao, L.; Zhang, Y.; Sterling, K.; Song, W. Brain-Derived Neurotrophic Factor in Alzheimer’s Disease and Its Pharmaceutical Potential. Transl. Neurodegener. 2022, 11, 4. [Google Scholar] [CrossRef]
  19. Edelmann, E.; Cepeda-Prado, E.; Franck, M.; Lichtenecker, P.; Brigadski, T.; Leßmann, V. Theta Burst Firing Recruits BDNF Release and Signaling in Postsynaptic CA1 Neurons in Spike-Timing-Dependent LTP. Neuron 2015, 86, 1041–1054. [Google Scholar] [CrossRef] [Green Version]
  20. Schinder, A.F.; Poo, M. ming The Neurotrophin Hypothesis for Synaptic Plasticity. Trends Neurosci. 2000, 23, 639–645. [Google Scholar] [CrossRef]
  21. Wang, Z.H.; Xiang, J.; Liu, X.; Yu, S.P.; Manfredsson, F.P.; Sandoval, I.M.; Wu, S.; Wang, J.Z.; Ye, K. Deficiency in BDNF/TrkB Neurotrophic Activity Stimulates δ-Secretase by Upregulating C/EBPβ in Alzheimer’s Disease. Cell Rep. 2019, 28, 655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Elliott, E.; Atlas, R.; Lange, A.; Ginzburg, I. Brain-Derived Neurotrophic Factor Induces a Rapid Dephosphorylation of Tau Protein through a PI-3 Kinase Signalling Mechanism. Eur. J. Neurosci. 2005, 22, 1081–1089. [Google Scholar] [CrossRef] [PubMed]
  23. Tanqueiro, S.R.; Ramalho, R.M.; Rodrigues, T.M.; Lopes, L.V.; Sebastião, A.M.; Diógenes, M.J. Inhibition of NMDA Receptors Prevents the Loss of BDNF Function Induced by Amyloid β. Front. Pharmacol. 2018, 9, 237. [Google Scholar] [CrossRef] [PubMed]
  24. Meng, C.; He, Z.; Xing, D. Low-Level Laser Therapy Rescues Dendrite Atrophy via Upregulating BDNF Expression: Implications for Alzheimer’s Disease. J. Neurosci. 2013, 33, 13505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Galic, M.A.; Riazi, K.; Pittman, Q.J. Cytokines and Brain Excitability. Front. Neuroendocrinol. 2012, 33, 116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Peng, S.; Wuu, J.; Mufson, E.J.; Fahnestock, M. Precursor Form of Brain-Derived Neurotrophic Factor and Mature Brain-Derived Neurotrophic Factor Are Decreased in the Pre-Clinical Stages of Alzheimer’s Disease. J Neurochem 2005, 93, 1412–1421. [Google Scholar] [CrossRef]
  27. Ng, T.K.S.; Ho, C.S.H.; Tam, W.W.S.; Kua, E.H.; Ho, R.C.M. Decreased Serum Brain-Derived Neurotrophic Factor (BDNF) Levels in Patients with Alzheimer’s Disease (AD): A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2019, 20, 257. [Google Scholar] [CrossRef] [Green Version]
  28. Forlenza, O.V.; Diniz, B.S.; Teixeira, A.L.; Radanovic, M.; Talib, L.L.; Rocha, N.P.; Gattaz, W.F. Lower Cerebrospinal Fluid Concentration of Brain-Derived Neurotrophic Factor Predicts Progression from Mild Cognitive Impairment to Alzheimer’s Disease. Neuromolecular. Med. 2015, 17, 326–332. [Google Scholar] [CrossRef]
  29. Weinstein, G.; Beiser, A.S.; Choi, S.H.; Preis, S.R.; Chen, T.C.; Vorgas, D.; Au, R.; Pikula, A.; Wolf, P.A.; DeStefano, A.L.; et al. Serum Brain-Derived Neurotrophic Factor and the Risk for Dementia: The Framingham Heart Study. JAMA Neurol. 2014, 71, 55. [Google Scholar] [CrossRef]
  30. Gudala, K.; Bansal, D.; Schifano, F.; Bhansali, A. Diabetes Mellitus and Risk of Dementia: A Meta-Analysis of Prospective Observational Studies. J. Diabetes Investig. 2013, 4, 640–650. [Google Scholar] [CrossRef] [Green Version]
  31. Picone, P.; di Carlo, M.; Nuzzo, D. Obesity and Alzheimer’s Disease: Molecular Bases. Eur. J. Neurosci. 2020, 52, 3944–3950. [Google Scholar] [CrossRef] [PubMed]
  32. Pistell, P.J.; Morrison, C.D.; Gupta, S.; Knight, A.G.; Keller, J.N.; Ingram, D.K.; Bruce-Keller, A.J. Cognitive Impairment Following High Fat Diet Consumption Is Associated with Brain Inflammation. J. Neuroimmunol. 2010, 219, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Winocur, G.; Greenwood, C.E. Studies of the Effects of High Fat Diets on Cognitive Function in a Rat Model. Neurobiol. Aging 2005, 26 (Suppl. 1), 46–49. [Google Scholar] [CrossRef] [PubMed]
  34. Sobesky, J.L.; Barrientos, R.M.; De May, H.S.; Thompson, B.M.; Weber, M.D.; Watkins, L.R.; Maier, S.F. High-Fat Diet Consumption Disrupts Memory and Primes Elevations in Hippocampal IL-1β, an Effect That Can Be Prevented with Dietary Reversal or IL-1 Receptor Antagonism. Brain Behav. Immun. 2014, 42, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Morris, M.C.; Evans, D.A.; Bienias, J.L.; Tangney, C.C.; Bennett, D.A.; Aggarwal, N.; Schneider, J.; Wilson, R.S. Dietary Fats and the Risk of Incident Alzheimer Disease. Arch. Neurol. 2003, 60, 194–200. [Google Scholar] [CrossRef] [PubMed]
  36. Lee, Y.H.; Martin, J.M.; Maple, R.L.; Tharp, W.G.; Pratley, R.E. Plasma Amyloid-Beta Peptide Levels Correlate with Adipocyte Amyloid Precursor Protein Gene Expression in Obese Individuals. Neuroendocrinology 2009, 90, 383–390. [Google Scholar] [CrossRef]
  37. Grant, R.W.; Dixit, V.D. Adipose Tissue as an Immunological Organ. Obesity 2015, 23, 512–518. [Google Scholar] [CrossRef] [Green Version]
  38. Arnold, S.E.; Lucki, I.; Brookshire, B.R.; Carlson, G.C.; Browne, C.A.; Kazi, H.; Bang, S.; Choi, B.R.; Chen, Y.; McMullen, M.F.; et al. High Fat Diet Produces Brain Insulin Resistance, Synaptodendritic Abnormalities and Altered Behavior in Mice. Neurobiol. Dis. 2014, 67, 79–87. [Google Scholar] [CrossRef] [Green Version]
  39. de la Monte, S.M. Brain Insulin Resistance and Deficiency as Therapeutic Targets in Alzheimer’s Disease. Curr. Alzheimer Res. 2012, 9, 35. [Google Scholar] [CrossRef]
  40. Soto, M.E.; Secher, M.; Gillette-Guyonnet, S.; Van Kan, G.A.; Andrieu, S.; Nourhashemi, F.; Rolland, Y.; Vellas, B. Weight Loss and Rapid Cognitive Decline in Community-Dwelling Patients with Alzheimer’s Disease. J. Alzheimers Dis. 2012, 28, 647–654. [Google Scholar] [CrossRef]
  41. Kimura, A.; Sugimoto, T.; Kitamori, K.; Saji, N.; Niida, S.; Toba, K.; Sakurai, T. Malnutrition Is Associated with Behavioral and Psychiatric Symptoms of Dementia in Older Women with Mild Cognitive Impairment and Early-Stage Alzheimer’s Disease. Nutrients 2019, 11, 1951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Suma, S.; Watanabe, Y.; Hirano, H.; Kimura, A.; Edahiro, A.; Awata, S.; Yamashita, Y.; Matsushita, K.; Arai, H.; Sakurai, T. Factors Affecting the Appetites of Persons with Alzheimer’s Disease and Mild Cognitive Impairment. Geriatr. Gerontol. Int. 2018, 18, 1236–1243. [Google Scholar] [CrossRef] [PubMed]
  43. Sugimoto, T.; Ono, R.; Murata, S.; Saji, N.; Matsui, Y.; Niida, S.; Toba, K.; Sakurai, T. Prevalence and Associated Factors of Sarcopenia in Elderly Subjects with Amnestic Mild Cognitive Impairment or Alzheimer Disease. Curr. Alzheimer. Res. 2016, 13, 718–726. [Google Scholar] [CrossRef] [PubMed]
  44. Ye, B.S.; Jang, E.Y.; Kim, S.Y.; Kim, E.J.; Park, S.A.; Lee, Y.; Hong, C.H.; Choi, S.H.; Yoon, B.; Yoon, S.J.; et al. Unstable Body Mass Index and Progression to Probable Alzheimer’s Disease Dementia in Patients with Amnestic Mild Cognitive Impairment. J. Alzheimers Dis. 2016, 49, 483–491. [Google Scholar] [CrossRef]
  45. Hanson, L.C.; Ersek, M.; Lin, F.C.; Carey, T.S. Outcomes of Feeding Problems in Advanced Dementia in a Nursing Home Population. J. Am. Geriatr. Soc. 2013, 61, 1692–1697. [Google Scholar] [CrossRef] [Green Version]
  46. Shimizu, T.; Nagaoka, U.; Nakayama, Y.; Kawata, A.; Kugimoto, C.; Kuroiwa, Y.; Kawai, M.; Shimohata, T.; Nishizawa, M.; Mihara, B.; et al. Reduction Rate of Body Mass Index Predicts Prognosis for Survival in Amyotrophic Lateral Sclerosis: A Multicenter Study in Japan. Amyotroph. Lateral Scler. 2012, 13, 363–366. [Google Scholar] [CrossRef]
  47. Ousset, P.J.; Nourhashemi, F.; Reynish, E.; Vellas, B. Nutritional Status Is Associated with Disease Progression in Very Mild Alzheimer Disease. Alzheimer Dis. Assoc. Disord. 2008, 22, 66–71. [Google Scholar] [CrossRef]
  48. Yu, J.T.; Xu, W.; Tan, C.C.; Andrieu, S.; Suckling, J.; Evangelou, E.; Pan, A.; Zhang, C.; Jia, J.; Feng, L.; et al. Review: Evidence-Based Prevention of Alzheimer’s Disease: Systematic Review and Meta-Analysis of 243 Observational Prospective Studies and 153 Randomised Controlled Trials. J. Neurol. Neurosurg. Psychiatry 2020, 91, 1201. [Google Scholar] [CrossRef]
  49. Ludwig, D.S.; Willett, W.C.; Volek, J.S.; Neuhouser, M.L. Dietary Fat: From Foe to Friend? Science 2018, 362, 764–770. [Google Scholar] [CrossRef] [Green Version]
  50. Frausto, D.M.; Forsyth, C.B.; Keshavarzian, A.; Voigt, R.M. Dietary Regulation of Gut-Brain Axis in Alzheimer’s Disease: Importance of Microbiota Metabolites. Front. Neurosci. 2021, 15, 736814. [Google Scholar] [CrossRef]
  51. Lin, B.; Hasegawa, Y.; Takane, K.; Koibuchi, N.; Cao, C.; Kim-Mitsuyama, S. High-Fat-Diet Intake Enhances Cerebral Amyloid Angiopathy and Cognitive Impairment in a Mouse Model of Alzheimer’s Disease, Independently of Metabolic Disorders. J. Am. Heart Assoc. Cardiovasc. Cerebrovasc. Dis. 2016, 5, e003154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Bracko, O.; Vinarcsik, L.K.; Cruz Hernández, J.C.; Ruiz-Uribe, N.E.; Haft-Javaherian, M.; Falkenhain, K.; Ramanauskaite, E.M.; Ali, M.; Mohapatra, A.; Swallow, M.A.; et al. High Fat Diet Worsens Alzheimer’s Disease-Related Behavioral Abnormalities and Neuropathology in APP/PS1 Mice, but Not by Synergistically Decreasing Cerebral Blood Flow. Sci. Rep. 2020, 10, 9884. [Google Scholar] [CrossRef] [PubMed]
  53. Morris, M.C.; Tangney, C.C.; Bienias, J.L.; Evans, D.A.; Wilson, R.S. Validity and Reproducibility of a Food Frequency Questionnaire by Cognition in an Older Biracial Sample. Am. J. Epidemiol. 2003, 158, 1213–1217. [Google Scholar] [CrossRef] [Green Version]
  54. Laitinen, M.H.; Ngandu, T.; Rovio, S.; Helkala, E.L.; Uusitalo, U.; Viitanen, M.; Nissinen, A.; Tuomilehto, J.; Soininen, H.; Kivipelto, M. Fat Intake at Midlife and Risk of Dementia and Alzheimer’s Disease: A Population-Based Study. Dement. Geriatr. Cogn. Disord. 2006, 22, 99–107. [Google Scholar] [CrossRef] [PubMed]
  55. Ruan, Y.; Tang, J.; Guo, X.; Li, K.; Li, D. Dietary Fat Intake and Risk of Alzheimer’s Disease and Dementia: A Meta-Analysis of Cohort Studies. Curr. Alzheimer Res. 2018, 15, 869–876. [Google Scholar] [CrossRef] [PubMed]
  56. Szczechowiak, K.; Diniz, B.S.; Leszek, J. Diet and Alzheimer’s Dementia—Nutritional Approach to Modulate Inflammation. Pharmacol. Biochem. Behav. 2019, 184, 172743. [Google Scholar] [CrossRef] [PubMed]
  57. Velloso, L.A.; Folli, F.; Saad, M.J. TLR4 at the Crossroads of Nutrients, Gut Microbiota, and Metabolic Inflammation. Endocr. Rev. 2015, 36, 245–271. [Google Scholar] [CrossRef] [Green Version]
  58. Lustig, R.H. Ultraprocessed Food: Addictive, Toxic, and Ready for Regulation. Nutrients 2020, 12, 3401. [Google Scholar] [CrossRef]
  59. Więckowska-Gacek, A.; Mietelska-Porowska, A.; Wydrych, M.; Wojda, U. Western Diet as a Trigger of Alzheimer’s Disease: From Metabolic Syndrome and Systemic Inflammation to Neuroinflammation and Neurodegeneration. Ageing Res. Rev. 2021, 70, 101397. [Google Scholar] [CrossRef]
  60. Ke, X.; Fu, Q.; Sterrett, J.; Hillard, C.J.; Lane, R.H.; Majnik, A. Adverse Maternal Environment and Western Diet Impairs Cognitive Function and Alters Hippocampal Glucocorticoid Receptor Promoter Methylation in Male Mice. Physiol. Rep. 2020, 8, e14407. [Google Scholar] [CrossRef]
  61. Leigh, S.J.; Morris, M.J. Diet, Inflammation and the Gut Microbiome: Mechanisms for Obesity-Associated Cognitive Impairment. Biochim. Et Biophys. Acta (BBA)-Mol. Basis Dis. 2020, 1866, 165767. [Google Scholar] [CrossRef] [PubMed]
  62. Leigh Gibson, E.; Barr, S.; Jeanes, Y.M. Habitual Fat Intake Predicts Memory Function in Younger Women. Front. Hum. Neurosci. 2013, 7, 838. [Google Scholar] [CrossRef] [Green Version]
  63. Stevenson, R.J.; Francis, H.M.; Attuquayefio, T.; Gupta, D.; Yeomans, M.R.; Oaten, M.J.; Davidson, T. Hippocampal-Dependent Appetitive Control Is Impaired by Experimental Exposure to a Western-Style Diet. R. Soc. Open Sci. 2020, 7, 191338. [Google Scholar] [CrossRef] [Green Version]
  64. Rebai, R.; Jasmin, L.; Boudah, A. Agomelatine Effects on Fat-Enriched Diet Induced Neuroinflammation and Depression-like Behavior in Rats. Biomed. Pharmacother. 2021, 135, 111246. [Google Scholar] [CrossRef] [PubMed]
  65. Alghamdi, B.S. The Effect of Short-Term Feeding of a High-Coconut Oil or High-Fat Diet on Neuroinflammation and the Performance of an Object–Place Task in Rats. Neurochem. Res. 2021, 46, 287–298. [Google Scholar] [CrossRef]
  66. Attuquayefio, T.; Stevenson, R.J.; Oaten, M.J.; Francis, H.M. A Four-Day Western-Style Dietary Intervention Causes Reductions in Hippocampal-Dependent Learning and Memory and Interoceptive Sensitivity. PLoS ONE 2017, 12, e0172645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Jacka, F.N.; Cherbuin, N.; Anstey, K.J.; Sachdev, P.; Butterworth, P. Western Diet Is Associated with a Smaller Hippocampus: A Longitudinal Investigation. BMC Med. 2015, 13, 215. [Google Scholar] [CrossRef] [Green Version]
  68. Ullrich, C.; Pirchl, M.; Humpel, C. Hypercholesterolemia in Rats Impairs the Cholinergic System and Leads to Memory Deficits. Mol. Cell. Neurosci. 2010, 45, 408–417. [Google Scholar] [CrossRef]
  69. Selvi, Y.; Gergerlioglu, H.S.; Akbaba, N.; Oz, M.; Kandeger, A.; Demir, E.A.; Yerlikaya, F.H.; Nurullahoglu-Atalik, K.E. Impact of Enriched Environment on Production of Tau, Amyloid Precursor Protein and, Amyloid-β Peptide in High-Fat and High-Sucrose-Fed Rats. Acta Neuropsychiatr. 2017, 29, 291–298. [Google Scholar] [CrossRef]
  70. Hohsfield, L.A.; Daschil, N.; Orädd, G.; Strömberg, I.; Humpel, C. Vascular Pathology of 20-Month-Old Hypercholesterolemia Mice in Comparison to Triple-Transgenic and APPSwDI Alzheimer’s Disease Mouse Models. Mol. Cell. Neurosci. 2014, 63, 83–95. [Google Scholar] [CrossRef] [Green Version]
  71. Kang, J.; Wang, Z.; Oteiza, P.I. (−)-Epicatechin Mitigates High Fat Diet-Induced Neuroinflammation and Altered Behavior in Mice. Food. Funct. 2020, 11, 5065–5076. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, Y.; Fu, X.; Lan, N.; Li, S.; Zhang, J.; Wang, S.; Li, C.; Shang, Y.; Huang, T.; Zhang, L. Luteolin Protects against High Fat Diet-Induced Cognitive Deficits in Obesity Mice. Behav. Brain Res. 2014, 267, 178–188. [Google Scholar] [CrossRef]
  73. Trichopoulou, A.; Martínez-González, M.A.; Tong, T.Y.N.; Forouhi, N.G.; Khandelwal, S.; Prabhakaran, D.; Mozaffarian, D.; de Lorgeril, M. Definitions and Potential Health Benefits of the Mediterranean Diet: Views from Experts around the World. BMC Med. 2014, 12, 112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Davis, C.; Bryan, J.; Hodgson, J.; Murphy, K. Definition of the Mediterranean Diet: A Literature Review. Nutrients 2015, 7, 9139–9153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Cremonini, A.L.; Caffa, I.; Cea, M.; Nencioni, A.; Odetti, P.; Monacelli, F. Nutrients in the Prevention of Alzheimer’s Disease. Oxid. Med. Cell Longev. 2019, 2019, 9874159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Knight, A.; Bryan, J.; Murphy, K. Is the Mediterranean Diet a Feasible Approach to Preserving Cognitive Function and Reducing Risk of Dementia for Older Adults in Western Countries? New Insights and Future Directions. Ageing Res. Rev. 2016, 25, 85–101. [Google Scholar] [CrossRef]
  77. McEvoy, C.T.; Guyer, H.; Langa, K.M.; Yaffe, K. Neuroprotective Diets Are Associated with Better Cognitive Function: The Health and Retirement Study. J. Am. Geriatr. Soc. 2017, 65, 1857. [Google Scholar] [CrossRef] [Green Version]
  78. Zbeida, M.; Goldsmith, R.; Shimony, T.; Yardi, H.; Naggan, L.; Shahar, D.R. Mediterranean Diet and Functional Indicators among Older Adults in Non-Mediterranean and Mediterranean Countries. J. Nutr. Health Aging 2014, 18, 411–418. [Google Scholar] [CrossRef]
  79. Anastasiou, C.A.; Yannakoulia, M.; Kontogianni, M.D.; Kosmidis, M.H.; Mamalaki, E.; Dardiotis, E.; Hadjigeorgiou, G.; Sakka, P.; Tsapanou, A.; Lykou, A.; et al. Mediterranean Lifestyle in Relation to Cognitive Health: Results from the HELIAD Study. Nutrients 2018, 10, 1557. [Google Scholar] [CrossRef] [Green Version]
  80. Galbete, C.; Toledo, E.; Toledo, J.B.; Bes-Rastrollo, M.; Buil-Cosiales, P.; Marti, A.; Guillén-Grima, F.; Martínez-González, M.A. Mediterranean Diet and Cognitive Function: The SUN Project. J. Nutr. Health Aging 2015, 19, 305–312. [Google Scholar] [CrossRef]
  81. Katsiardanis, K.; Diamantaras, A.A.; Dessypris, N.; Michelakos, T.; Anastasiou, A.; Katsiardani, K.P.; Kanavidis, P.; Papadopoulos, F.C.; Stefanadis, C.; Panagiotakos, D.B.; et al. Cognitive Impairment and Dietary Habits among Elders: The Velestino Study. J. Med. Food 2013, 16, 343–350. [Google Scholar] [CrossRef] [PubMed]
  82. Tsivgoulis, G.; Judd, S.; Letter, A.J.; Alexandrov, A.V.; Howard, G.; Nahab, F.; Unverzagt, F.W.; Moy, C.; Howard, V.J.; Kissela, B.; et al. Adherence to a Mediterranean Diet and Risk of Incident Cognitive Impairment. Neurology 2013, 80, 1684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Scarmeas, N.; Stern, Y.; Tang, M.X.; Mayeux, R.; Luchsinger, J.A. Mediterranean Diet and Risk for Alzheimer’s Disease. Ann. Neurol. 2006, 59, 912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. 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. [Google Scholar] [CrossRef] [Green Version]
  85. Anastasiou, C.A.; Yannakoulia, M.; Kosmidis, M.H.; Dardiotis, E.; Hadjigeorgiou, G.M.; Sakka, P.; Arampatzi, X.; Bougea, A.; Labropoulos, I.; Scarmeas, N. Mediterranean Diet and Cognitive Health: Initial Results from the Hellenic Longitudinal Investigation of Ageing and Diet. PLoS ONE 2017, 12, e018204. [Google Scholar] [CrossRef] [Green Version]
  86. Ballarini, T.; van Lent, D.M.; Brunner, J.; Schröder, A.; Wolfsgruber, S.; Altenstein, S.; Brosseron, F.; Buerger, K.; Dechent, P.; Dobisch, L.; et al. Mediterranean Diet, Alzheimer Disease Biomarkers, and Brain Atrophy in Old Age. Neurology 2021, 96, e2920. [Google Scholar] [CrossRef]
  87. Hoscheidt, S.; Sanderlin, A.H.; Baker, L.D.; Jung, Y.; Lockhart, S.; Kellar, D.; Whitlow, C.T.; Hanson, A.J.; Friedman, S.; Register, T.; et al. Mediterranean and Western Diet Effects on Alzheimer’s Disease Biomarkers, Cerebral Perfusion, and Cognition in Mid-life: A Randomized Trial. Alzheimer’s Dement. 2022, 18, 457. [Google Scholar] [CrossRef]
  88. Siervo, M.; Lara, J.; Chowdhury, S.; Ashor, A.; Oggioni, C.; Mathers, J.C. Effects of the Dietary Approach to Stop Hypertension (DASH) Diet on Cardiovascular Risk Factors: A Systematic Review and Meta-Analysis. Br. J. Nutr. 2015, 113, 1–15. [Google Scholar] [CrossRef] [Green Version]
  89. Van Den Brink, A.C.; Brouwer-Brolsma, E.M.; Berendsen, A.A.M.; Van De Rest, O. The Mediterranean, Dietary Approaches to Stop Hypertension (DASH), and Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) Diets Are Associated with Less Cognitive Decline and a Lower Risk of Alzheimer’s Disease—A Review. Adv. Nutr. 2019, 10, 1040. [Google Scholar] [CrossRef] [Green Version]
  90. Blumenthal, J.A.; Smith, P.J.; Mabe, S.; Hinderliter, A.; Welsh-Bohmer, K.; Browndyke, J.N.; Lin, P.H.; Kraus, W.; Doraiswamy, P.M.; Burke, J.; et al. Lifestyle and Neurocognition in Older Adults with Cardiovascular Risk Factors and Cognitive Impairment. Psychosom. Med. 2017, 79, 719. [Google Scholar] [CrossRef]
  91. Haring, B.; Wu, C.; Mossavar-Rahmani, Y.; Snetselaar, L.; Brunner, R.; Wallace, R.B.; Neuhouser, M.L.; Wassertheil-Smoller, S. No Association between Dietary Patterns and Risk for Cognitive Decline in Older Women with Nine-Year Follow-up: Data from the Women’s Health Initiative Memory Study. J. Acad. Nutr. Diet. 2016, 116, 921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Smith, P.J.; Blumenthal, J.A.; Babyak, M.A.; Craighead, L.; Welsh-Bohmer, K.A.; Browndyke, J.N.; Strauman, T.A.; Sherwood, A. Effects of the Dietary Approaches to Stop Hypertension Diet, Exercise, and Caloric Restriction on Neurocognition in Overweight Adults with High Blood Pressure. Hypertension 2010, 55, 1331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Shakersain, B.; Rizzuto, D.; Larsson, S.C.; Faxén-Irving, G.; Fratiglioni, L.; Xu, W.L. The Nordic Prudent Diet Reduces Risk of Cognitive Decline in the Swedish Older Adults: A Population-Based Cohort Study. Nutrients 2018, 10, 229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Cherian, L.; Wang, Y.; Fakuda, K.; Leurgans, S.; Aggarwal, N.; Morris, M. Mediterranean-Dash Intervention for Neurodegenerative Delay (MIND) Diet Slows Cognitive Decline after Stroke. J. Prev. Alzheimers Dis. 2019, 6, 267. [Google Scholar] [CrossRef] [PubMed]
  95. Wengreen, H.; Munger, R.G.; Cutler, A.; Quach, A.; Bowles, A.; Corcoran, C.; Tschanz, J.A.T.; Norton, M.C.; Welsh-Bohmer, K.A. Prospective Study of Dietary Approaches to Stop Hypertension– and Mediterranean-Style Dietary Patterns and Age-Related Cognitive Change: The Cache County Study on Memory, Health and Aging. Am. J. Clin. Nutr. 2013, 98, 1263. [Google Scholar] [CrossRef] [Green Version]
  96. Morris, M.C.; Evans, D.A.; Tangney, C.C.; Bienias, J.L.; Wilson, R.S. Associations of Vegetable and Fruit Consumption with Age-Related Cognitive Change. Neurology 2006, 67, 1370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Devore, E.E.; Kang, J.H.; Breteler, M.M.B.; Grodstein, F. Dietary Intake of Berries and Flavonoids in Relation to Cognitive Decline. Ann. Neurol. 2012, 72, 135. [Google Scholar] [CrossRef]
  98. Morris, M.C.; Evans, D.A.; Tangney, C.C.; Bienias, J.L.; Wilson, R.S. Fish Consumption and Cognitive Decline with Age in a Large Community Study. Arch. Neurol. 2005, 62, 1849–1853. [Google Scholar] [CrossRef] [Green Version]
  99. Urpi-Sarda, M.; Casas, R.; Chiva-Blanch, G.; Romero-Mamani, E.S.; Valderas-Martínez, P.; Arranz, S.; Andres-Lacueva, C.; Llorach, R.; Medina-Remón, A.; Lamuela-Raventos, R.M.; et al. Virgin Olive Oil and Nuts as Key Foods of the Mediterranean Diet Effects on Inflammatory Biomakers Related to Atherosclerosis. Pharmacol. Res. 2012, 65, 577–583. [Google Scholar] [CrossRef]
  100. Martínez-Lapiscina, E.H.; Clavero, P.; Toledo, E.; Estruch, R.; Salas-Salvadó, J.; San Julián, B.; Sanchez-Tainta, A.; Ros, E.; Valls-Pedret, C.; Martinez-Gonzalez, M.Á. Mediterranean Diet Improves Cognition: The PREDIMED-NAVARRA Randomised Trial. J. Neurol. Neurosurg. Psychiatry 2013, 84, 1318–1325. [Google Scholar] [CrossRef] [Green Version]
  101. Nutritional Factors and Risk of Incident Dementia in the PAQUID Longitudinal Cohort. Available online: https://psycnet.apa.org/record/2004-14797-001 (accessed on 21 March 2023).
  102. Chen, X.; Huang, Y.; Cheng, H.G. Lower Intake of Vegetables and Legumes Associated with Cognitive Decline among Illiterate Elderly Chinese: A 3-Year Cohort Study. J. Nutr. Health Aging 2012, 16, 549–552. [Google Scholar] [CrossRef] [PubMed]
  103. Hosking, D.E.; Eramudugolla, R.; Cherbuin, N.; Anstey, K.J. MIND Not Mediterranean Diet Related to 12-Year Incidence of Cognitive Impairment in an Australian Longitudinal Cohort Study. Alzheimers Dement. 2019, 15, 581–589. [Google Scholar] [CrossRef] [PubMed]
  104. Tao, Y.; Leng, S.X.; Zhang, H. Ketogenic Diet: An Effective Treatment Approach for Neurodegenerative Diseases. Curr. Neuropharmacol. 2022, 20, 2303–2319. [Google Scholar] [CrossRef] [PubMed]
  105. Rusek, M.; Pluta, R.; Ułamek-Kozioł, M.; Czuczwar, S.J. Ketogenic Diet in Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 3892. [Google Scholar] [CrossRef] [Green Version]
  106. Hersant, H.; Grossberg, G. The Ketogenic Diet and Alzheimer’s Disease. J. Nutr. Health Aging 2022, 26, 606–614. [Google Scholar] [CrossRef]
  107. Jiang, Z.; Yin, X.; Wang, M.; Chen, T.; Wang, Y.; Gao, Z.; Wang, Z. Effects of Ketogenic Diet on Neuroinflammation in Neurodegenerative Diseases. Aging Dis. 2022, 13, 1146–1165. [Google Scholar] [CrossRef]
  108. Yang, Y.; Wang, X.; Xiao, A.; Han, J.; Wang, Z.; Wen, M. Ketogenic Diet Prevents Chronic Sleep Deprivation-Induced Alzheimer’s Disease by Inhibiting Iron Dyshomeostasis and Promoting Repair via Sirt1/Nrf2 Pathway. Front. Aging Neurosci. 2022, 14, 1013. [Google Scholar] [CrossRef]
  109. Xu, Y.; Jiang, C.; Wu, J.; Liu, P.; Deng, X.; Zhang, Y.; Peng, B.; Zhu, Y. Ketogenic Diet Ameliorates Cognitive Impairment and Neuroinflammation in a Mouse Model of Alzheimer’s Disease. CNS Neurosci. Ther. 2022, 28, 580–592. [Google Scholar] [CrossRef]
  110. Neth, B.J.; Mintz, A.; Whitlow, C.; Jung, Y.; Solingapuram Sai, K.; Register, T.C.; Kellar, D.; Lockhart, S.N.; Hoscheidt, S.; Maldjian, J.; et al. Modified Ketogenic Diet Is Associated with Improved Cerebrospinal Fluid Biomarker Profile, Cerebral Perfusion, and Cerebral Ketone Body Uptake in Older Adults at Risk for Alzheimer’s Disease: A Pilot Study. Neurobiol. Aging 2020, 86, 54–63. [Google Scholar] [CrossRef]
  111. Pietrzak, D.; Kasperek, K.; Rękawek, P.; Piątkowska-Chmiel, I. The Therapeutic Role of Ketogenic Diet in Neurological Disorders. Nutrients 2022, 14, 1952. [Google Scholar] [CrossRef]
  112. Sridharan, B.; Lee, M.-J. Ketogenic Diet: A Promising Neuroprotective Composition for Managing Alzheimer’s Diseases and Its Pathological Mechanisms. Curr. Mol. Med. 2021, 22, 640–656. [Google Scholar] [CrossRef]
  113. Zhao, Y.; Jia, M.; Chen, W.; Liu, Z. The Neuroprotective Effects of Intermittent Fasting on Brain Aging and Neurodegenerative Diseases via Regulating Mitochondrial Function. Free Radic. Biol. Med. 2022, 182, 206–218. [Google Scholar] [CrossRef]
  114. Mattson, M.P.; Moehl, K.; Ghena, N.; Schmaedick, M.; Cheng, A. Intermittent Metabolic Switching, Neuroplasticity and Brain Health. Nat. Rev. Neurosci. 2018, 19, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Mattson, M.P.; Longo, V.D.; Harvie, M. Impact of Intermittent Fasting on Health and Disease Processes. Ageing Res. Rev. 2017, 39, 46–58. [Google Scholar] [CrossRef] [PubMed]
  116. Tinsley, G.M.; La Bounty, P.M. Effects of Intermittent Fasting on Body Composition and Clinical Health Markers in Humans. Nutr. Rev. 2015, 73, 661–674. [Google Scholar] [CrossRef] [PubMed]
  117. Catenacci, V.A.; Pan, Z.; Ostendorf, D.; Brannon, S.; Gozansky, W.S.; Mattson, M.P.; Martin, B.; MacLean, P.S.; Melanson, E.L.; Troy Donahoo, W. A Randomized Pilot Study Comparing Zero-Calorie Alternate-Day Fasting to Daily Caloric Restriction in Adults with Obesity. Obesity 2016, 24, 1874–1883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Varady, K.A.; Bhutani, S.; Klempel, M.C.; Kroeger, C.M.; Trepanowski, J.F.; Haus, J.M.; Hoddy, K.K.; Calvo, Y. Alternate Day Fasting for Weight Loss in Normal Weight and Overweight Subjects: A Randomized Controlled Trial. Nutr. J. 2013, 12, 146. [Google Scholar] [CrossRef] [Green Version]
  119. Bhutani, S.; Klempel, M.C.; Kroeger, C.M.; Trepanowski, J.F.; Varady, K.A. Alternate Day Fasting and Endurance Exercise Combine to Reduce Body Weight and Favorably Alter Plasma Lipids in Obese Humans. Obesity 2013, 21, 1370–1379. [Google Scholar] [CrossRef] [PubMed]
  120. Singh, R.; Lakhanpal, D.; Kumar, S.; Sharma, S.; Kataria, H.; Kaur, M.; Kaur, G. Late-Onset Intermittent Fasting Dietary Restriction as a Potential Intervention to Retard Age-Associated Brain Function Impairments in Male Rats. Age 2012, 34, 917–933. [Google Scholar] [CrossRef] [Green Version]
  121. Zhang, J.; Zhan, Z.; Li, X.; Xing, A.; Jiang, C.; Chen, Y.; Shi, W.; An, L. Intermittent Fasting Protects against Alzheimer’s Disease Possible through Restoring Aquaporin-4 Polarity. Front. Mol. Neurosci. 2017, 10, 395. [Google Scholar] [CrossRef] [Green Version]
  122. Halagappa, V.K.M.; Guo, Z.; Pearson, M.; Matsuoka, Y.; Cutler, R.G.; LaFerla, F.M.; Mattson, M.P. Intermittent Fasting and Caloric Restriction Ameliorate Age-Related Behavioral Deficits in the Triple-Transgenic Mouse Model of Alzheimer’s Disease. Neurobiol. Dis. 2007, 26, 212–220. [Google Scholar] [CrossRef] [PubMed]
  123. Lazic, D.; Tesic, V.; Jovanovic, M.; Brkic, M.; Milanovic, D.; Zlokovic, B.V.; Kanazir, S.; Perovic, M. Every-Other-Day Feeding Exacerbates Inflammation and Neuronal Deficits in 5XFAD Mouse Model of Alzheimer’s Disease. Neurobiol. Dis. 2020, 136, 104745. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, Y.; Cheng, A.; Li, Y.J.; Yang, Y.; Kishimoto, Y.; Zhang, S.; Wang, Y.; Wan, R.; Raefsky, S.M.; Lu, D.; et al. SIRT3 Mediates Hippocampal Synaptic Adaptations to Intermittent Fasting and Ameliorates Deficits in APP Mutant Mice. Nat. Commun. 2019, 10, 1886. [Google Scholar] [CrossRef] [Green Version]
  125. Li, L.; Wang, Z.; Zuo, Z. Chronic Intermittent Fasting Improves Cognitive Functions and Brain Structures in Mice. PLoS ONE 2013, 8, 66069. [Google Scholar] [CrossRef] [Green Version]
  126. Mirzaei, H.; Suarez, J.A.; Longo, V.D. Protein and Amino Acid Restriction, Aging and Disease: From Yeast to Humans. Trends Endocrinol. Metab. 2014, 25, 558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Witte, A.V.; Fobker, M.; Gellner, R.; Knecht, S.; Flöel, A. From the Cover: Caloric Restriction Improves Memory in Elderly Humans. Proc. Natl. Acad. Sci. USA 2009, 106, 1255. [Google Scholar] [CrossRef] [Green Version]
  128. Bayer-Carter, J.L.; Green, P.S.; Montine, T.J.; Van-Fossen, B.; Baker, L.D.; Watson, G.S.; Bonner, L.M.; Callaghan, M.; Leverenz, J.B.; Walter, B.K.; et al. Diet Intervention and Cerebrospinal Fluid Biomarkers in Amnestic Mild Cognitive Impairment. Arch. Neurol. 2011, 68, 743. [Google Scholar] [CrossRef] [Green Version]
  129. Kunnumakkara, A.B.; Bordoloi, D.; Padmavathi, G.; Monisha, J.; Roy, N.K.; Prasad, S.; Aggarwal, B.B. Curcumin, the Golden Nutraceutical: Multitargeting for Multiple Chronic Diseases. Br. J. Pharmacol. 2017, 174, 1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; et al. Curcumin Inhibits Formation of Amyloid Beta Oligomers and Fibrils, Binds Plaques, and Reduces Amyloid in Vivo. J. Biol. Chem. 2005, 280, 5892–5901. [Google Scholar] [CrossRef] [Green Version]
  131. Garcia-Alloza, M.; Borrelli, L.A.; Rozkalne, A.; Hyman, B.T.; Bacskai, B.J. Curcumin Labels Amyloid Pathology in Vivo, Disrupts Existing Plaques, and Partially Restores Distorted Neurites in an Alzheimer Mouse Model. J. Neurochem. 2007, 102, 1095–1104. [Google Scholar] [CrossRef]
  132. Reddy, P.H.; Manczak, M.; Yin, X.; Grady, M.C.; Mitchell, A.; Tonk, S.; Kuruva, C.S.; Bhatti, J.S.; Kandimalla, R.; Vijayan, M.; et al. Protective Effects of Indian Spice Curcumin Against Amyloid Beta in Alzheimer’s Disease. J. Alzheimers Dis. 2018, 61, 843. [Google Scholar] [CrossRef] [PubMed]
  133. Park, S.Y.; Kim, D.S.H.L. Discovery of Natural Products from Curcuma Longa That Protect Cells from Beta-Amyloid Insult: A Drug Discovery Effort against Alzheimer’s Disease. J. Nat. Prod. 2002, 65, 1227–1231. [Google Scholar] [CrossRef] [PubMed]
  134. Ray, B.; Lahiri, D.K. Neuroinflammation in Alzheimer’s Disease: Different Molecular Targets and Potential Therapeutic Agents Including Curcumin. Curr. Opin. Pharmacol. 2009, 9, 434–444. [Google Scholar] [CrossRef] [PubMed]
  135. Konno, H.; Endo, H.; Ise, S.; Miyazaki, K.; Aoki, H.; Sanjoh, A.; Kobayashi, K.; Hattori, Y.; Akaji, K. Synthesis and Evaluation of Curcumin Derivatives toward an Inhibitor of Beta-Site Amyloid Precursor Protein Cleaving Enzyme 1. Bioorg. Med. Chem. Lett. 2014, 24, 685–690. [Google Scholar] [CrossRef] [PubMed]
  136. Doroszkiewicz, J.; Mroczko, P.; Kulczyńska-Przybik, A. Inflammation in the CNS: Understanding Various Aspects of the Pathogenesis of Alzheimer’s Disease. Curr. Alzheimer Res. 2021, 19, 16–31. [Google Scholar] [CrossRef]
  137. Small, G.W.; Siddarth, P.; Li, Z.; Miller, K.J.; Ercoli, L.; Emerson, N.D.; Martinez, J.; Wong, K.P.; Liu, J.; Merrill, D.A.; et al. Memory and Brain Amyloid and Tau Effects of a Bioavailable Form of Curcumin in Non-Demented Adults: A Double-Blind, Placebo-Controlled 18-Month Trial. Am. J. Geriatr. Psychiatry 2018, 26, 266–277. [Google Scholar] [CrossRef]
  138. Rainey-Smith, S.R.; Brown, B.M.; Sohrabi, H.R.; Shah, T.; Goozee, K.G.; Gupta, V.B.; Martins, R.N. Curcumin and Cognition: A Randomised, Placebo-Controlled, Double-Blind Study of Community-Dwelling Older Adults. Br. J. Nutr. 2016, 115, 2106–2113. [Google Scholar] [CrossRef] [Green Version]
  139. Hoppe, J.B.; Coradini, K.; Frozza, R.L.; Oliveira, C.M.; Meneghetti, A.B.; Bernardi, A.; Pires, E.S.; Beck, R.C.R.; Salbego, C.G. Free and Nanoencapsulated Curcumin Suppress β-Amyloid-Induced Cognitive Impairments in Rats: Involvement of BDNF and Akt/GSK-3β Signaling Pathway. Neurobiol. Learn Mem. 2013, 106, 134–144. [Google Scholar] [CrossRef]
  140. Ringman, J.M.; Frautschy, S.A.; Teng, E.; Begum, A.N.; Bardens, J.; Beigi, M.; Gylys, K.H.; Badmaev, V.; Heath, D.D.; Apostolova, L.G.; et al. Oral Curcumin for Alzheimer’s Disease: Tolerability and Efficacy in a 24-Week Randomized, Double Blind, Placebo-Controlled Study. Alzheimers Res. Ther. 2012, 4, 43. [Google Scholar] [CrossRef] [Green Version]
  141. Baum, L.; Lam, C.W.K.; Cheung, S.K.K.; Kwok, T.; Lui, V.; Tsoh, J.; Lam, L.; Leung, V.; Hui, E.; Ng, C.; et al. Six-Month Randomized, Placebo-Controlled, Double-Blind, Pilot Clinical Trial of Curcumin in Patients with Alzheimer Disease. J. Clin. Psychopharmacol. 2008, 28, 110–113. [Google Scholar] [CrossRef] [Green Version]
  142. Santos, C.; Lunet, N.; Azevedo, A.; De Mendonça, A.; Ritchie, K.; Barros, H. Caffeine Intake Is Associated with a Lower Risk of Cognitive Decline: A Cohort Study from Portugal. J. Alzheimers Dis. 2010, 20 (Suppl. 1), S175–S185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Arab, L.; Khan, F.; Lam, H. Epidemiologic Evidence of a Relationship between Tea, Coffee, or Caffeine Consumption and Cognitive Decline. Adv. Nutr. 2013, 4, 115–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Madeira, M.H.; Boia, R.; Ambrósio, A.F.; Santiago, A.R. Having a Coffee Break: The Impact of Caffeine Consumption on Microglia-Mediated Inflammation in Neurodegenerative Diseases. Mediators Inflamm. 2017, 2017, 4761081. [Google Scholar] [CrossRef] [Green Version]
  145. Arendash, G.W.; Cao, C. Caffeine and Coffee as Therapeutics Against Alzheimer’s Disease. J. Alzheimer’s Dis. 2010, 20, S117–S126. [Google Scholar] [CrossRef] [Green Version]
  146. Wu, L.; Sun, D.; He, Y. Coffee Intake and the Incident Risk of Cognitive Disorders: A Dose–Response Meta-Analysis of Nine Prospective Cohort Studies. Clin. Nutr. 2017, 36, 730–736. [Google Scholar] [CrossRef] [PubMed]
  147. Panza, F.; Solfrizzi, V.; Barulli, M.R.; Bonfiglio, C.; Guerra, V.; Osella, A.; Seripa, D.; Sabbà, C.; Pilotto, A.; Logroscino, G. Coffee, Tea, and Caffeine Consumption and Prevention of Late-Life Cognitive Decline and Dementia: A Systematic Review. J. Nutr. Health Aging 2015, 19, 313–328. [Google Scholar] [CrossRef]
  148. Solfrizzi, V.; Custodero, C.; Lozupone, M.; Imbimbo, B.P.; Valiani, V.; Agosti, P.; Schilardi, A.; D’Introno, A.; La Montagna, M.; Calvani, M.; et al. Relationships of Dietary Patterns, Foods, and Micro- and Macronutrients with Alzheimer’s Disease and Late-Life Cognitive Disorders: A Systematic Review. J. Alzheimers Dis. 2017, 59, 815–849. [Google Scholar] [CrossRef] [Green Version]
  149. Zhou, A.; Taylor, A.E.; Karhunen, V.; Zhan, Y.; Rovio, S.P.; Lahti, J.; Sjögren, P.; Byberg, L.; Lyall, D.M.; Auvinen, J.; et al. Habitual Coffee Consumption and Cognitive Function: A Mendelian Randomization Meta-Analysis in up to 415,530 Participants. Sci. Rep. 2018, 8, 7526. [Google Scholar] [CrossRef] [Green Version]
  150. Pervin, M.; Unno, K.; Ohishi, T.; Tanabe, H.; Miyoshi, N.; Nakamura, Y. Beneficial Effects of Green Tea Catechins on Neurodegenerative Diseases. Mol. A J. Synth. Chem. Nat. Prod. Chem. 2018, 23, 1297. [Google Scholar] [CrossRef] [Green Version]
  151. Suzuki, T.; Pervin, M.; Goto, S.; Isemura, M.; Nakamura, Y. Beneficial Effects of Tea and the Green Tea Catechin Epigallocatechin-3-Gallate on Obesity. Molecules 2016, 21, 1305. [Google Scholar] [CrossRef] [Green Version]
  152. Khan, N.; Mukhtar, H. Tea Polyphenols in Promotion of Human Health. Nutrients 2019, 11, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Chieng, D.; Kistler, P.M. Coffee and Tea on Cardiovascular Disease (CVD) Prevention. Trends Cardiovasc. Med. 2022, 32, 399–405. [Google Scholar] [CrossRef] [PubMed]
  154. Kakutani, S.; Watanabe, H.; Murayama, N. Green Tea Intake and Risks for Dementia, Alzheimer’s Disease, Mild Cognitive Impairment, and Cognitive Impairment: A Systematic Review. Nutrients 2019, 11, 1165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Luca, M.; Luca, A.; Calandra, C. The Role of Oxidative Damage in the Pathogenesis and Progression of Alzheimer’s Disease and Vascular Dementia. Oxid. Med. Cell Longev. 2015, 2015, 504678. [Google Scholar] [CrossRef] [Green Version]
  156. Molino, S.; Dossena, M.; Buonocore, D.; Ferrari, F.; Venturini, L.; Ricevuti, G.; Verri, M. Polyphenols in Dementia: From Molecular Basis to Clinical Trials. Life Sci. 2016, 161, 69–77. [Google Scholar] [CrossRef]
  157. Spagnuolo, C.; Moccia, S.; Russo, G.L. Anti-Inflammatory Effects of Flavonoids in Neurodegenerative Disorders. Eur. J. Med. Chem. 2018, 153, 105–115. [Google Scholar] [CrossRef]
  158. Afzal, M.; Safer, A.M.; Menon, M. Green Tea Polyphenols and Their Potential Role in Health and Disease. Inflammopharmacology 2015, 23, 151–161. [Google Scholar] [CrossRef]
  159. Prasanth, M.I.; Sivamaruthi, B.S.; Chaiyasut, C.; Tencomnao, T. A Review of the Role of Green Tea (Camellia Sinensis) in Antiphotoaging, Stress Resistance, Neuroprotection, and Autophagy. Nutrients 2019, 11, 474. [Google Scholar] [CrossRef] [Green Version]
  160. Tomata, Y.; Sugiyama, K.; Kaiho, Y.; Honkura, K.; Watanabe, T.; Zhang, S.; Sugawara, Y.; Tsuji, I. Green Tea Consumption and the Risk of Incident Dementia in Elderly Japanese: The Ohsaki Cohort 2006 Study. Am. J. Geriatr. Psychiatry 2016, 24, 881–889. [Google Scholar] [CrossRef] [Green Version]
  161. Schimidt, H.L.; Garcia, A.; Martins, A.; Mello-Carpes, P.B.; Carpes, F.P. Green Tea Supplementation Produces Better Neuroprotective Effects than Red and Black Tea in Alzheimer-like Rat Model. Food Res. Int. 2017, 100, 442–448. [Google Scholar] [CrossRef]
  162. Gu, L.; Kelm, M.A.; Hammerstone, J.F.; Beecher, G.; Holden, J.; Haytowitz, D.; Gebhardt, S.; Prior, R.L. Concentrations of Proanthocyanidins in Common Foods and Estimations of Normal Consumption. J. Nutr. 2004, 134, 613–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Zeli, C.; Lombardo, M.; Storz, M.A.; Ottaviani, M.; Rizzo, G. Chocolate and Cocoa-Derived Biomolecules for Brain Cognition during Ageing. Antioxidants 2022, 11, 1353. [Google Scholar] [CrossRef]
  164. Septianti, E.; Salengke; Langkong, J. Profile of Bioactive Compounds, Antioxidant and Aromatic Component from Several Clones of Cocoa Beans during Fermentation. IOP Conf. Ser. Earth Environ. Sci. 2020, 575, 012009. [Google Scholar] [CrossRef]
  165. Cooper, K.A.; Donovan, J.L.; Waterhouse, A.I.; Williamson, G. Cocoa and Health: A Decade of Research. Br. J. Nutr. 2008, 99, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Lazarus, S.A.; Hammerstone, J.F.; Schmitz, H.H. Chocolate Contains Additional Flavonoids Not Found in Tea. Lancet 1999, 354, 1825. [Google Scholar] [CrossRef] [PubMed]
  167. Grassi, D.; Desideri, G.; Necozione, S.; Lippi, C.; Casale, R.; Properzi, G.; Blumberg, J.B.; Ferri, C. Blood Pressure Is Reduced and Insulin Sensitivity Increased in Glucose-Intolerant, Hypertensive Subjects after 15 Days of Consuming High-Polyphenol Dark Chocolate. J. Nutr. 2008, 138, 1671–1676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Mastroiacovo, D.; Kwik-Uribe, C.; Grassi, D.; Necozione, S.; Raffaele, A.; Pistacchio, L.; Righetti, R.; Bocale, R.; Lechiara, M.C.; Marini, C.; et al. Cocoa Flavanol Consumption Improves Cognitive Function, Blood Pressure Control, and Metabolic Profile in Elderly Subjects: The Cocoa, Cognition, and Aging (CoCoA) Study—A Randomized Controlled Trial. Am. J. Clin. Nutr. 2015, 101, 538. [Google Scholar] [CrossRef] [Green Version]
  169. Sorond, F.A.; Lipsitz, L.A.; Hollenberg, N.K.; Fisher, N.D.L. Cerebral Blood Flow Response to Flavanol-Rich Cocoa in Healthy Elderly Humans. Neuropsychiatr. Dis. Treat. 2008, 4, 433. [Google Scholar] [CrossRef] [Green Version]
  170. Jin Heo, H.; Lee, C.Y. Epicatechin and Catechin in Cocoa Inhibit Amyloid Beta Protein Induced Apoptosis. J. Agric. Food Chem. 2005, 53, 1445–1448. [Google Scholar] [CrossRef]
  171. Cimini, A.; Gentile, R.; D’Angelo, B.; Benedetti, E.; Cristiano, L.; Avantaggiati, M.L.; Giordano, A.; Ferri, C.; Desideri, G. Cocoa Powder Triggers Neuroprotective and Preventive Effects in a Human Alzheimer’s Disease Model by Modulating BDNF Signaling Pathway. J. Cell Biochem. 2013, 114, 2209. [Google Scholar] [CrossRef] [Green Version]
  172. Wang, J.; Varghese, M.; Ono, K.; Yamada, M.; Levine, S.; Tzavaras, N.; Gong, B.; Hurst, W.J.; Blitzer, R.D.; Pasinetti, G.M. Cocoa Extracts Reduce Oligomerization of Amyloid-β: Implications for Cognitive Improvement in Alzheimer’s Disease. J. Alzheimers Dis. 2014, 41, 643–650. [Google Scholar] [CrossRef] [PubMed]
  173. Desideri, G.; Kwik-Uribe, C.; Grassi, D.; Necozione, S.; Ghiadoni, L.; Mastroiacovo, D.; Raffaele, A.; Ferri, L.; Bocale, R.; Lechiara, M.C.; et al. Benefits in Cognitive Function, Blood Pressure, and Insulin Resistance through Cocoa Flavanol Consumption in Elderly Subjects with Mild Cognitive Impairment: The Cocoa, Cognition, and Aging (CoCoA) Study. Hypertension 2012, 60, 794–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Mosconi, L.; Pupi, A.; De Leon, M.J. Brain Glucose Hypometabolism and Oxidative Stress in Preclinical Alzheimer’s Disease. Ann. N. Y. Acad. Sci. 2008, 1147, 180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Mielech, A.; Puścion-Jakubik, A.; Markiewicz-żukowska, R.; Socha, K. Vitamins in Alzheimer’s Disease—Review of the Latest Reports. Nutrients 2020, 12, 3458. [Google Scholar] [CrossRef]
  176. Smith, P.J.; Blumenthal, J.A. Dietary Factors and Cognitive Decline. J. Prev. Alzheimers Dis. 2016, 3, 53. [Google Scholar] [CrossRef]
  177. De Jager, C.A.; Oulhaj, A.; Jacoby, R.; Refsum, H.; Smith, A.D. Cognitive and Clinical Outcomes of Homocysteine-Lowering B-Vitamin Treatment in Mild Cognitive Impairment: A Randomized Controlled Trial. Int. J. Geriatr. Psychiatry 2012, 27, 592–600. [Google Scholar] [CrossRef]
  178. Chen, H.; Liu, S.; Ji, L.; Wu, T.; Ji, Y.; Zhou, Y.; Zheng, M.; Zhang, M.; Xu, W.; Huang, G. Folic Acid Supplementation Mitigates Alzheimer’s Disease by Reducing Inflammation: A Randomized Controlled Trial. Mediators Inflamm. 2016, 2016, 5912146. [Google Scholar] [CrossRef] [Green Version]
  179. Kwok, T.; Wu, Y.; Lee, J.; Lee, R.; Yung, C.Y.; Choi, G.; Lee, V.; Harrison, J.; Lam, L.; Mok, V. A Randomized Placebo-Controlled Trial of Using B Vitamins to Prevent Cognitive Decline in Older Mild Cognitive Impairment Patients. Clin. Nutr. 2020, 39, 2399–2405. [Google Scholar] [CrossRef] [Green Version]
  180. Van Der Zwaluw, N.L.; Dhonukshe-Rutten, R.A.M.; Van Wijngaarden, J.P.V.; Brouwer-Brolsma, E.M.; Van De Rest, O.V.D.; In’t Veld, P.H.; Enneman, A.W.; Van Dijk, S.C.; Ham, A.C.; Swart, K.M.A.; et al. Results of 2-Year Vitamin B Treatment on Cognitive Performance: Secondary Data from an RCT. Neurology 2014, 83, 2158–2166. [Google Scholar] [CrossRef]
  181. Thiel, A.; Hermanns, C.; Lauer, A.A.; Reichrath, J.; Erhardt, T.; Hartmann, T.; Grimm, M.O.W.; Grimm, H.S. Vitamin D and Its Analogues: From Differences in Molecular Mechanisms to Potential Benefits of Adapted Use in the Treatment of Alzheimer’s Disease. Nutrients 2023, 15, 1684. [Google Scholar] [CrossRef]
  182. Amrein, K.; Scherkl, M.; Hoffmann, M.; Neuwersch-Sommeregger, S.; Köstenberger, M.; Tmava Berisha, A.; Martucci, G.; Pilz, S.; Malle, O. Vitamin D Deficiency 2.0: An Update on the Current Status Worldwide. Eur. J. Clin. Nutr. 2020, 74, 1498. [Google Scholar] [CrossRef]
  183. Kiderman, D.; Ben-Shabat, N.; Tsur, A.M.; Anis, S.; Watad, A.; Cohen, A.D.; Paz, Z.; Amital, H. Vitamin D Insufficiency Is Associated with Higher Incidence of Dementia, a Large Community-Based Retrospective Cohort Study. J. Geriatr. Psychiatry Neurol. online ahead of print. 2023. [Google Scholar] [CrossRef]
  184. Chai, B.; Gao, F.; Wu, R.; Dong, T.; Gu, C.; Lin, Q.; Zhang, Y. Vitamin D Deficiency as a Risk Factor for Dementia and Alzheimer’s Disease: An Updated Meta-Analysis. BMC Neurol. 2019, 19, 284. [Google Scholar] [CrossRef] [PubMed]
  185. Shen, L.; Ji, H.F. Vitamin D Deficiency Is Associated with Increased Risk of Alzheimer’s Disease and Dementia: Evidence from Meta-Analysis. Nutr. J. 2015, 14, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Ghahremani, M.; Smith, E.E.; Chen, H.; Creese, B.; Goodarzi, Z.; Ismail, Z. Vitamin D Supplementation and Incident Dementia: Effects of Sex, APOE, and Baseline Cognitive Status. Alzheimers Dement. 2023, 15, 12404. [Google Scholar] [CrossRef] [PubMed]
  187. Miller, B.J.; Whisner, C.M.; Johnston, C.S. Vitamin D Supplementation Appears to Increase Plasma Aβ40 in Vitamin D Insufficient Older Adults: A Pilot Randomized Controlled Trial. J. Alzheimers Dis. 2016, 52, 843–847. [Google Scholar] [CrossRef]
  188. Jia, J.; Hu, J.; Huo, X.; Miao, R.; Zhang, Y.; Ma, F. Effects of Vitamin D Supplementation on Cognitive Function and Blood Aβ-Related Biomarkers in Older Adults with Alzheimer’s Disease: A Randomised, Double-Blind, Placebo-Controlled Trial. J. Neurol. Neurosurg. Psychiatry 2019, 90, 1347–1352. [Google Scholar] [CrossRef]
  189. Reiter, E.; Jiang, Q.; Christen, S. Anti-Inflammatory Properties of α- and γ-Tocopherol. Mol. Aspects Med. 2007, 28, 668. [Google Scholar] [CrossRef] [Green Version]
  190. Jiang, Q. Natural Forms of Vitamin E: Metabolism, Antioxidant and Anti-Inflammatory Activities and the Role in Disease Prevention and Therapy. Free Radic. Biol. Med. 2014, 72, 76. [Google Scholar] [CrossRef] [Green Version]
  191. Lopes Da Silva, S.; Vellas, B.; Elemans, S.; Luchsinger, J.; Kamphuis, P.; Yaffe, K.; Sijben, J.; Groenendijk, M.; Stijnen, T. Plasma Nutrient Status of Patients with Alzheimer’s Disease: Systematic Review and Meta-Analysis. Alzheimers Dement. 2014, 10, 485–502. [Google Scholar] [CrossRef] [Green Version]
  192. Mullan, K.; Cardwell, C.R.; McGuinness, B.; Woodside, J.V.; McKay, G.J. Plasma Antioxidant Status in Patients with Alzheimer’s Disease and Cognitively Intact Elderly: A Meta-Analysis of Case-Control Studies. J. Alzheimers Dis. 2018, 62, 305–317. [Google Scholar] [CrossRef] [Green Version]
  193. Mangialasche, F.; Kivipelto, M.; Mecocci, P.; Rizzuto, D.; Palmer, K.; Winblad, B.; Fratiglioni, L. High Plasma Levels of Vitamin E Forms and Reduced Alzheimer’s Disease Risk in Advanced Age. J. Alzheimers Dis. 2010, 20, 1029–1037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Mangialasche, F.; Solomon, A.; Kåreholt, I.; Hooshmand, B.; Cecchetti, R.; Fratiglioni, L.; Soininen, H.; Laatikainen, T.; Mecocci, P.; Kivipelto, M. Serum Levels of Vitamin E Forms and Risk of Cognitive Impairment in a Finnish Cohort of Older Adults. Exp. Gerontol. 2013, 48, 1428–1435. [Google Scholar] [CrossRef] [PubMed]
  195. Morris, M.C.; Evans, D.A.; Bienias, J.L.; Tangney, C.C.; Bennett, D.A.; Aggarwal, N.; Wilson, R.S.; Scherr, P.A. Dietary Intake of Antioxidant Nutrients and the Risk of Incident Alzheimer Disease in a Biracial Community Study. JAMA 2002, 287, 3230–3237. [Google Scholar] [CrossRef]
  196. Browne, D.; McGuinness, B.; Woodside, J.V.; McKay, G.J. Vitamin E and Alzheimer’s Disease: What Do We Know so Far? Clin. Interv. Aging 2019, 14, 1303. [Google Scholar] [CrossRef] [Green Version]
  197. Dysken, M.W.; Sano, M.; Asthana, S.; Vertrees, J.E.; Pallaki, M.; Llorente, M.; Love, S.; Schellenberg, G.D.; McCarten, J.R.; Malphurs, J.; et al. Effect of Vitamin E and Memantine on Functional Decline in Alzheimer Disease: The TEAM-AD VA Cooperative Randomized Trial. J. Am. Med. Assoc. 2014, 311, 33. [Google Scholar] [CrossRef] [PubMed]
  198. Kryscio, R.J.; Abner, E.L.; Caban-Holt, A.; Lovell, M.; Goodman, P.; Darke, A.K.; Yee, M.; Crowley, J.; Schmitt, F.A. Association of Antioxidant Supplement Use and Dementia in the Prevention of Alzheimer’s Disease by Vitamin E and Selenium Trial (PREADViSE). JAMA Neurol. 2017, 74, 567. [Google Scholar] [CrossRef] [PubMed]
  199. Von Arnim, C.A.F.; Herbolsheimer, F.; Nikolaus, T.; Peter, R.; Biesalski, H.K.; Ludolph, A.C.; Riepe, M.; Nagel, G. Dietary Antioxidants and Dementia in a Population-Based Case-Control Study among Older People in South Germany. J. Alzheimers Dis. 2012, 31, 717–724. [Google Scholar] [CrossRef] [Green Version]
  200. Gravesteijn, E.; Mensink, R.P.; Plat, J. Effects of Nutritional Interventions on BDNF Concentrations in Humans: A Systematic Review. Nutr. Neurosci. 2022, 25, 1425–1436. [Google Scholar] [CrossRef]
  201. Sánchez-Villegas, A.; Galbete, C.; Martinez-González, M.A.; Martinez, J.A.; Razquin, C.; Salas-Salvadó, J.; Estruch, R.; Buil-Cosiales, P.; Martí, A. The Effect of the Mediterranean Diet on Plasma Brain-Derived Neurotrophic Factor (BDNF) Levels: The PREDIMED-NAVARRA Randomized Trial. Nutr. Neurosci. 2013, 14, 195–201. [Google Scholar] [CrossRef]
  202. Suzuki, T.; Kojima, N.; Osuka, Y.; Tokui, Y.; Takasugi, S.; Kawashima, A.; Yamaji, T.; Hosoi, E.; Won, C.W.; Kim, H. The Effects of Mold-Fermented Cheese on Brain-Derived Neurotrophic Factor in Community-Dwelling Older Japanese Women with Mild Cognitive Impairment: A Randomized, Controlled, Crossover Trial. J. Am. Med. Dir. Assoc. 2019, 20, 1509–1514. [Google Scholar] [CrossRef]
  203. Sandberg, J.C.; Björck, I.M.E.; Nilsson, A.C. Increased Plasma Brain-Derived Neurotrophic Factor 10.5 h after Intake of Whole Grain Rye-Based Products in Healthy Subjects. Nutrients 2018, 10, 1097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Jafari, F.; Amani, R.; Tarrahi, M.J. Effect of Zinc Supplementation on Physical and Psychological Symptoms, Biomarkers of Inflammation, Oxidative Stress, and Brain-Derived Neurotrophic Factor in Young Women with Premenstrual Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Biol. Trace Elem. Res. 2020, 194, 89–95. [Google Scholar] [CrossRef] [PubMed]
  205. Solati, Z.; Jazayeri, S.; Tehrani-Doost, M.; Mahmoodianfard, S.; Gohari, M.R. Zinc Monotherapy Increases Serum Brain-Derived Neurotrophic Factor (BDNF) Levels and Decreases Depressive Symptoms in Overweight or Obese Subjects: A Double-Blind, Randomized, Placebo-Controlled Trial. Nutr. Neurosci. 2014, 18, 162–168. [Google Scholar] [CrossRef] [PubMed]
  206. Yu, J.J.; Pei, L.B.; Zhang, Y.; Wen, Z.Y.; Yang, J.L. Chronic Supplementation of Curcumin Enhances the Efficacy of Antidepressants in Major Depressive Disorder: A Randomized, Double-Blind, Placebo-Controlled Pilot Study. J. Clin. Psychopharmacol. 2015, 35, 406–410. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms underlying Alzheimer’s disease pathology.
Figure 1. Mechanisms underlying Alzheimer’s disease pathology.
Ijms 24 10751 g001
Figure 2. Illustration of the positive effects of diets.
Figure 2. Illustration of the positive effects of diets.
Ijms 24 10751 g002
Table 1. The effects of diets and life situations negatively influencing the Alzheimer’s disease.
Table 1. The effects of diets and life situations negatively influencing the Alzheimer’s disease.
AuthorsType of Diet/Life SituationStudy DesignStudy PopulationResultsReference
Lee et al.ObesityPilot study10 obese patients before and after weight loss interventionChanges in adipocyte APP expression correlated with changes in plasma Aβ40 levels (R = 0.74, p = 0.01).[36]
Ye et al.MalnutritionLongitudinal study747 patients with aMCIThe underweight group had a higher risk of probable Alzheimer’s disease dementia (pADD), and the decreased-BMI (HR: 2.29, 95% CI: 1.41–3.72) groups were at increased risk of progression to pADD.[44]
Hanson et al.MalnutritionProspective cohort256 nursing home residents with advanced dementia and feeding problemsSignificant mortality risk in patients with feeding problems[45]
Laitinen et al.High-fat dietPopulation-based study1449 patients with 117 who had dementiaModerate intake of polyunsaturated fats in mid-life decreased the risk of dementia even after adjustment for demographic variables, especially among the ApoE epsilon4 carriers.[54]
Ruan et al.High-fat dietMeta-analysis of cohort studies8630 participants and 633 casesA higher dietary saturated fat intake was significantly associated with increased risks of 39% and 105% for AD and dementia.[55]
Jacka et al.Western dietLongitudinal study255 persons from the Personality and Total Health Through Life StudyHigher consumption of an unhealthy “Western” dietary pattern was associated with smaller left hippocampal volume.[67]
Gibson et al.Western dietObservational study23 women with polycystic ovary syndrome from 25 to 45 years oldGreater intakes of saturated and trans fats, and higher saturated-to-unsaturated fat ratio (Sat:UFA) were associated with more errors in the visuospatial task and poorer word recall and recognition.[62]
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

Doroszkiewicz, J.; Mroczko, J.; Rutkowski, P.; Mroczko, B. Molecular Aspects of a Diet as a New Pathway in the Prevention and Treatment of Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 10751. https://doi.org/10.3390/ijms241310751

AMA Style

Doroszkiewicz J, Mroczko J, Rutkowski P, Mroczko B. Molecular Aspects of a Diet as a New Pathway in the Prevention and Treatment of Alzheimer’s Disease. International Journal of Molecular Sciences. 2023; 24(13):10751. https://doi.org/10.3390/ijms241310751

Chicago/Turabian Style

Doroszkiewicz, Julia, Jan Mroczko, Piotr Rutkowski, and Barbara Mroczko. 2023. "Molecular Aspects of a Diet as a New Pathway in the Prevention and Treatment of Alzheimer’s Disease" International Journal of Molecular Sciences 24, no. 13: 10751. https://doi.org/10.3390/ijms241310751

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