Next Article in Journal
No Difference in Liver Damage Induced by Isocaloric Fructose or Glucose in Mice with a High-Fat Diet
Previous Article in Journal
The Association Between Preoperative Sarcopenia and Sarcopenic Obesity and the Occurrence of Postoperative Complications in Patients Undergoing Pancreaticoduodenectomy for Periampullary Malignancies—A Literature Review
Previous Article in Special Issue
Association between Dietary Patterns and Cognitive Function among Qatari Adults: A Cross-Sectional Analysis of the Qatar Biobank Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases

by
Ewelina Młynarska
1,*,
Paulina Jakubowska
1,
Weronika Frąk
1,
Agata Gajewska
1,
Joanna Sornowska
1,
Sylwia Skwira
1,
Jakub Wasiak
1,
Jacek Rysz
2 and
Beata Franczyk
1
1
Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
2
Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
*
Author to whom correspondence should be addressed.
Nutrients 2024, 16(20), 3570; https://doi.org/10.3390/nu16203570
Submission received: 18 September 2024 / Revised: 9 October 2024 / Accepted: 15 October 2024 / Published: 21 October 2024
(This article belongs to the Special Issue Effects of Dietary Intake on Cognitive Function)

Abstract

:
Background/Objectives: Recent research highlights the growing interest in the impact of nutrition on cognitive health and function in disease, as dietary habits are increasingly recognized as crucial factors in relation to brain function. This focus is especially important given the rising prevalence of neurodegenerative diseases and the cognitive decline associated with poor dietary choices. Links are now being sought between brain function and the microbiota and gut–brain axis. Mechanisms are proposed that include low-grade chronic neuroinflammation, the influence of short-chain fatty acids, or the disruption of glial cells and transmitters in the brain. Methods: We reviewed the articles on pubmed. This is not a systematic review, but of the narrative type. We wanted to outline the issue and summarise the latest information. Results: The axis in question has its foundation in nutrition. It has been reported that diet, particularly the components and the timing of food intake, has an impact on cognitive processes. The Mediterranean diet is most often cited in the literature as being beneficial to health. In order to obtain a more complete view, it is worth considering other dietary patterns, even those that impair our health. Conclusions: Determining what is beneficial and what is not will allow us to develop a speronized strategy for the prevention of, and fight against, cognitive impairment. Appropriately selected supplements, the functions of which we have also discussed, may prove supportive.

1. Introduction

The relationship between dietary intake and cognitive function has garnered increasing attention in recent years, particularly considering the increasing incidence of neurodegenerative and mental health disorders, both of which are closely linked to cognitive impairment. Cognitive functions encompass a wide range of mental processes essential for navigating daily life and maintaining overall well-being. These functions include memory, attention, language, executive function, and perception. In everyday life, various factors can contribute to cognitive function, influencing individuals’ well-being. These include sleep quality, physical activity, stress levels, engagement in cognitively stimulating activities, and social interactions. Additionally, dietary habits play a crucial role, as the nutrients we ingest can significantly affect our brain function, influencing aspects such as memory, attention, and overall cognitive health. This article aims to examine the existing literature on cognitive impairment associated with various diseases and the effects of different dietary patterns and nutrients on distinct cognitive functions in these disorders. Furthermore, it identifies potential avenues for future research in this domain.

2. Cognitive Dysfunctions and Their Underlying Mechanisms in Various Diseases

Cognitive functions are essential for an individual’s capacity for managing daily life, which involves a variety of mental processes such as memory, attention, executive function, and problem-solving. However, cognitive impairment is a hallmark of several health conditions, including pathological aging, neurodegenerative diseases, psychiatric disorders, nutritional disorders and obesity. These conditions affect cognitive function by specific but often overlapping mechanisms, leading to various levels of impairment (Figure 1).

2.1. Cognitive Function and Aging

Cognitive decline is a natural part of aging, with most individuals experiencing some deterioration in memory, processing speed, and executive function, driven by several mechanisms. One of the main causes is a decrease in the hippocampus volume, which impairs memory storage and learning [1]. Additionally, oxidative stress, which naturally increases with aging, plays a crucial role in cognitive decline as the accumulation of reactive oxygen species (ROS) coupled with reduced antioxidant defenses causes oxidative damage to neurons. This process is further exacerbated by age-related mitochondrial dysfunction, leading to additional oxidative stress [2]. Furthermore, decreased neuroplasticity, accompanied by decreased synapse formation and neurogenesis in the hippocampus, limits the brain’s ability to adapt to new cognitive challenges, leading to progressive cognitive decline over time [3].

2.2. Accelerated Cognitive Decline in Pathological Aging

Pathological aging is characterized by several mechanisms that accelerate cognitive decline beyond what is typically observed in normal aging. One significant factor is enhanced neuroinflammation, where chronic activation of the microglia and astrocytes leads to persistent brain inflammation, which exacerbates neuronal damage and accelerates cognitive decline. Additionally, mitochondrial dysfunction increases reactive oxygen species (ROS) production, which heightens oxidative stress and further exacerbates neuroinflammation, creating a vicious cycle that accelerates neuronal damage and cognitive decline [4]. Additionally, cerebrovascular damage contributes to pathological aging by impairing the cerebral blood flow, which results in ischemic injury to neurons and further compromises brain function [5]. Another significant factor is the increased accumulation of pathological substances and impaired clearance mechanisms, which disrupt neuronal function and synaptic communication, thereby leading to severe cognitive impairments [6,7].

2.3. Cognitive Function in Neurodegenerative Disorders

The most common neurodegenerative diseases, including Parkinson’s disease (PD), Alzheimer’s disease (AD), frontotemporal dementia and multiple sclerosis, exhibit varied clinical profiles and underlying pathophysiology. However, they generally share a common feature: cognitive impairment, which progressively worsens as the disease advances. These impairments can manifest as difficulties with memory, concentration, spatial orientation, and the ability to perform complex tasks, as well as changes in behavior and personality. As the disease progresses, these issues usually lead to increasing challenges in everyday life.
In patients with Parkinson’s disease, neuropsychological assessments often reveal mild to moderate impairments in visuospatial skills, attention, and working memory. The evidence consistently suggests that low cerebrospinal fluid levels of amyloid-β42, a marker of comorbid Alzheimer’s disease, predict future cognitive decline and dementia in Parkinson’s disease [8].
The presence of two abnormal structures, senile plaques and neurofibrillary tangles, occurs before the neuronal death and brain degeneration seen later in Alzheimer’s disease. The plaques consist of beta-amyloid peptide aggregates that progressively form in a specific pattern as the disease develops. This pattern begins in the neocortex and spreads through the allocortex, hippocampus, basal ganglia, midbrain, and cerebellum. Neurofibrillary tangles, which are aggregates of the tau protein, also develop later and spread along a different pathway, starting in the transentorhinal region and extending through the entorhinal cortex, hippocampus, and neocortical areas. Thus, there are two distinct pathologies—amyloid and tau—each with different structural changes that appear at different stages of the disease. It has been suggested that the disorder may only be classified as a unique disease when both the amyloid and tau pathologies converge. Prior to this overlap, distinct features of cognitive dysfunction may be associated with the presence of plaques in specific brain regions. Memory loss, cognitive decline, impairment of executive functions, and loss of consciousness, among other symptoms, are observed in AD. These symptoms can emerge at various stages throughout the progression of the disease, gradually advancing along a continuum that ends in dementia and extensive brain degeneration.

2.4. Cognitive Function in Psychiatric Disorders

Cognitive dysfunction, which impacts attention, memory, executive function, and social cognition, is prevalent across various psychiatric disorders. While some mechanisms differ, there are common pathways that may underlie these cognitive impairments.
Disruptions in neurotransmitter systems, including dopamine, serotonin, and glutamate, are central to cognitive impairments in psychiatric disorders. Imbalances in dopamine transmission, such as hypoactivity in the prefrontal cortex and hyperactivity in the mesolimbic pathway, contribute to deficits in executive function and working memory, which are evident in disorders like schizophrenia [9,10]. Alterations in serotonin levels or receptor function can impair mood regulation and cognitive processes such as memory and attention, a common issue in major depressive disorder [11]. Additionally, alterations in glutamatergic and GABAergic neurotransmission impair cognitive processes in disorders like schizophrenia and mood disorders [12].
Another mechanism of cognitive impairment in psychiatric disorders may be neuroinflammation, which involves chronic activation of immune responses in the brain [13]. Elevated levels of pro-inflammatory cytokines, such as IL-6, TNF-alpha, and IL-1β, contribute to cognitive deficits by activating the microglia and astrocytes, disrupting synaptic plasticity, impairing neurogenesis, and compromising the integrity of the blood–brain barrier [14,15]. This inflammatory environment is often exacerbated by dysregulation of the hypothalamic–pituitary–adrenal (HPA) axis, which results in sustained high cortisol levels. Elevated cortisol further aggravates neuroinflammation, leading to increased neuronal damage and cognitive decline [13].
Increased oxidative stress, marked by an imbalance between the production of ROS and the body’s antioxidant defenses, plays a pivotal role in the neuronal damage and cognitive dysfunction observed in psychiatric disorders. Excessive ROS can lead to the oxidation of lipids, proteins, and DNA, particularly in critical brain regions essential for cognitive processing [16]. Furthermore, oxidative stress impairs mitochondrial function, leading to mitochondrial DNA (mtDNA) dysfunction, which further aggravates ROS production. This creates a vicious cycle where damaged mitochondria produce even more ROS, exacerbating the oxidative damage and contributing to the progressive decline in neuronal function and cognitive abilities [17]. Additionally, oxidative stress can exacerbate neuroinflammation by further activating the microglia and promoting the release of pro-inflammatory cytokines, creating a feedback loop that intensifies the neuronal damage and cognitive impairment [18].

2.5. Cognitive Function in Dietary Diseases

Dietary diseases, including type 2 diabetes, celiac disease and malnutrition, have been increasingly recognized for their significant impact on cognitive function. These conditions, often resulting from poor nutritional choices, disrupt normal metabolic processes, which can impair cognitive abilities.
In the case of type 2 diabetes, hyperglycemia, insulin resistance, and chronic inflammation are key mechanisms through which cognitive function is compromised. These metabolic disturbances lead to neuroinflammation, oxidative stress, and vascular damage, all of which contribute to cognitive decline and an increased risk of developing neurodegenerative diseases such as Alzheimer’s disease [19].
Celiac disease presents another example where the autoimmune response to gluten in individuals with celiac disease can lead to neuroinflammation and cognitive dysfunction, often termed “brain fog”, which refers to a range of symptoms, including cognitive slowing, difficulty concentrating and problems with the short- and long-term memory [20].
Moreover, malnutrition, particularly in older adults, is a significant risk factor for cognitive decline. Nutritional deficiencies, such as in B vitamins, vitamin D, and essential fatty acids, are associated with impaired cognitive function and an increased risk of dementia. Malnutrition increases the susceptibility of the aging brain to neurodegenerative processes, thereby accelerating cognitive decline [21].

2.6. Cognitive Function in Obesity

The rate of obesity is significantly increasing, and alongside this trend, there is a corresponding rise in the occurrence of metabolic disorders such as cardiovascular disease, systemic hypertension and type 2 diabetes. Excessive adiposity leads to insulin resistance, systemic inflammation, and altered brain structure and function, especially within the fronto-mesolimbic circuitry [22]. Neuroimaging studies have consistently demonstrated that obesity is linked to reduced gray matter volume and altered functional connectivity, which are predictive of cognitive impairment. Research has shown that obesity doubles the risk of developing Alzheimer’s disease, and being obese during midlife is a strong predictor of a higher likelihood of dementia in later years [23].
A cross-sectional longitudinal study involving more than 2000 middle-aged workers found a consistent linear relationship between BMI and cognitive function, as measured by the word-list learning test, which assesses verbal learning and memory, and the digit-symbol substitution test, which evaluates attention, response speed, and visuomotor coordination [24]. The results showed that obese individuals recalled fewer words on the word-list learning test and required more time to complete the DSST compared to individuals of normal weight.
Figure 1. This figure summarizes the primary mechanisms contributing to cognitive dysfunction across various diseases, highlighting the complexity and interconnectivity of these processes.
Figure 1. This figure summarizes the primary mechanisms contributing to cognitive dysfunction across various diseases, highlighting the complexity and interconnectivity of these processes.
Nutrients 16 03570 g001

3. Microbiota and Gut–Brain Axis

One research topic that has been gaining popularity in recent years is the gut microbiota, the gut–brain axis (GBA), and their impact on whole body function, including brain activity and cognitive function. The term microbiota is used to describe the microorganisms, including commensal bacteria, that inhabit the human gastrointestinal tract. It is estimated that a person’s gut microbiome contains over 100 trillion bacteria, with their numbers increasing from the stomach to the intestines and colon [25]. While the specific composition of the microbiota varies among individuals, healthy people typically share similar proportions: 60% to 80% are Firmicutes, 20% to 40% are Bacteroidetes, and around 5% consist of Proteobacteria and Actinobacteria [26,27]. Microbiota development starts in the fetal stage and is strongly influenced by the mode of delivery, whether vaginal or cesarean. Additionally, the composition of the microbiota is affected by the infant’s diet, particularly whether they are breastfed or formula-fed [28].
Acute changes in a person’s diet account for about 20% of the variability in the human microbiota, but long-term dietary habits have a more significant impact on the microbiota composition [29]. The types of food we consume influence the growth of specific bacterial species, leading to two main enterotypes of microbiota. The first enterotype, characterized by a high presence of Bacteroides, is typical of the Western diet in highly industrialized countries, where low-fiber and high-fat diets are common. Within this enterotype, a subgroup, sometimes called Enterotype 3, is distinguished by a high abundance of Ruminococcus. The second enterotype, dominated by Prevotella, is common in less industrialized countries, where diets include less processed food and more fiber [30,31].
It is worth noting that the composition of the microbiota is individual and is affected by many factors, such as the country of residence and environmental pollution. Any diseases and medical conditions present are also of great importance. A summary of the factors affecting the microbiota composition in adults is shown in Figure 2 [32,33,34,35,36,37,38,39,40].
The human gut microbiota and host share a symbiotic relationship, which implies that the host and microorganisms benefit from one another. The host provides a habitat and sustenance for microorganisms, which in turn benefit the host’s health by increasing disease resistance and nutrient absorption [41].
The GBA plays a crucial role in maintaining human health by enabling two-way communication between the gut microbiota and the brain. Achieving and maintaining eubiosis, which involves the natural presence of beneficial microorganisms, is vital for proper functioning. The GBA involves complex interactions between various systems, including the gut-associated immune system, the enteric nervous system (ENS), the vagus nerve, and the gut microbiota itself. These interactions are facilitated by the gut microbiota, which produces important compounds such as neurotransmitters, tryptophan, and short-chain fatty acids (SCFAs) that influence both gut and brain function [42,43,44].
Both the enteric nervous system (ENS), which is composed of glial cells and enteric neurons, and the autonomic nervous system, which is controlled by the vagus nerve, are involved in regulating immunological responses, motility, and gastrointestinal secretion. The body can independently affect the gut microbiota’s makeup thanks to these processes. In turn, gut microorganisms interact with the central nervous system (CNS) via immunological, hormonal, and neurological pathways, affecting the body [45,46,47]. For instance, gut bacteria can create short-chain fatty acids (SCFAs) from fiber, such as acetate and propionate, which can control the gene expression by hyperacetylating histones. The systemic effects of SCFAs include immune system modulation, appetite regulation, improved absorption of calcium, and preservation of glucose homeostasis [48,49]. SCFAs also have neuroprotective properties. Therefore, their depletion may lead to neurodegeneration [50].
Additionally, the microbiota influences CNS function by producing neurotransmitters (like acetylcholine, catecholamines, and gamma-aminobutyric acid) and biogenic amines (like histamine), as well as altering tryptophan metabolism [51,52,53].
The proper functioning of the gut–brain axis (GBA) relies on the integrity of the intestinal barrier, which separates the contents of the gut from the rest of the body, thus controlling the exchange of fluids and solutes. When dysbiosis occurs, this barrier is compromised, leading to increased intestinal permeability, often called “leaky gut” syndrome (LGS). This condition allows microbial components like lipopolysaccharide (LPS), toxic metabolites, and inflammatory substances to enter the bloodstream [54,55]. LPS is recognized by Toll-like receptors (TLRs), which trigger the release of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), IL-6, IL-8, and IL-12, causing both local and systemic inflammation [53,56,57]. According to current theories, the LPS in the CNS promotes amyloid-β and tau accumulation as well as neuropathology, which results in neurodegenerative illnesses [58]. Moreover, the administration of LPS activates the brain microglia, increases pro-inflammatory cytokines, and can cause depressive symptoms. Prolonged microglial activation results in complement-mediated loss of dopaminergic neurons and neurodegeneration [59,60].
Chronic systemic inflammation from LGS can disrupt the hypothalamus–pituitary–adrenal (HPA) axis, which is essential for supplying energy sources like glucose, amino acids, and free fatty acids to support immune function. This disruption leads to increased release of glucocorticoids and catecholamines, resulting in hypercortisolemia and overactivity of the HPA axis, which is linked to impaired glucocorticoid receptor function [33,61]. Additionally, increased intestinal permeability can activate T-cells, potentially leading to autoimmune disorders in the gut or other organs if these activated lymphocytes spread further [62].
Systemic inflammation disrupts the blood–brain barrier (BBB), impairing its ability to regulate the movement of substances, ions, and cells into the brain. This increased permeability of the BBB has been observed in several psychiatric disorders, including those associated with cognitive decline, such as Alzheimer’s disease [63,64]. BBB impairment and inflammation result in the decreased number and reduced function of astrocytes, a type of glial cell [65]. Astrocytes possess TLR4 receptors on their surface, which, upon detecting LPS, trigger an amplified proinflammatory response [66]. Additionally, astrocytes play a crucial role in regulating gamma-aminobutyric acid (GABA) and glutamate levels by participating in the reabsorption of these neurotransmitters [67].

Influence of Gut Microbiota Metabolites and Neurotransmitters

Emerging evidence suggests that gut microbiota metabolites play a crucial role in the pathophysiology of neurodegenerative diseases such as Parkinson’s and Alzheimer’s.
LPS has been implicated in the exacerbation of neuroinflammation, suggesting its potential role in the pathogenesis of AD. The evidence supporting this hypothesis includes the elevated blood and brain LPS levels in Alzheimer’s patients, with LPS linked to increased amyloid-beta expression, aggregation, inflammation, neurotoxicity, tau phosphorylation, and microglial activation. Additionally, LPS may induce synapse and neuron loss, leading to memory deficits in AD animal models and cognitive dysfunction in humans [68].
In Parkinson’s disease, LPS has been shown to induce specific loss of midbrain dopaminergic neurons through microglial activation. Additionally, LPS induces α-synuclein expression, aggregation, and neurotoxicity, activates peripheral monocytes leading to inflammatory cytokine production, and mediates brain inflammation [69].
SCFAs, primarily produced by the gut microbiota, have garnered attention for their potential role in AD. SCFAs, particularly butyrate, acetate, and propionate, exhibit neuroprotective properties by enhancing neurotrophic factors like brain-derived neurotrophic factor (BDNF), which support neuronal health and synaptic plasticity. They also modulate neuroinflammation by inhibiting microglial activation and reducing pro-inflammatory cytokines, counteracting the inflammation associated with Alzheimer’s disease. Moreover, SCFAs help maintain blood–brain barrier (BBB) integrity, potentially preventing neurotoxic substances from entering the brain. Emerging evidence suggests that SCFAs may also influence amyloid-beta (Aβ) aggregation and tau phosphorylation, critical hallmarks of AD. Butyrate has been demonstrated to diminish the formation of neurotoxic amyloid-beta (Aβ) aggregates by inhibiting the assembly of the Aβ1–40 and Aβ1–42 peptides. In an animal model using 5xFAD mice, administering sodium butyrate (NaB) for twelve weeks led to a 40% decrease in the brain Aβ levels [70].
Some studies suggest that SCFAs may influence the aggregation of α-synuclein, a protein implicated in Parkinson’s disease. Additionally, SCFAs modulate neuroinflammation by inhibiting microglial activation and reducing pro-inflammatory cytokines, thereby protecting dopaminergic neurons from damage [71].
As mentioned, the microbiota also affects the production and metabolism of neurotransmitters. Firstly, recent research reveals that several neurotransmitters, including glutamate, GABA, serotonin, and dopamine, are among the metabolites generated by the gut bacteria [72]. GABA, an inhibitory neurotransmitter, was shown to be produced by certain bacteria species in the gut, such as Parabacteroides, Eubacterium, and Bifidobacterium, and act locally in the intestine [73,74]. The gut bacteria also create adenosine, which has local anti-inflammatory properties [75] and norepinephrine, which slows down the gut’s overall transit and reduces the number of migratory motor complexes. Additionally, it affects behavior and cognition, including learning, memory, and attention, and has an immunomodulatory impact [52,76].
Certain enzymes that can catalyze the conversion of substrates into matching neurotransmitters or precursors are encoded by genes found in certain bacteria. For example, SCFAs impact brain neurochemistry by controlling the expression levels of two enzymes: tyrosine hydroxylase, which is engaged in a rate-limiting step in the biosynthesis of dopamine, noradrenaline, and adrenaline, and tryptophan 5-hydroxylase 1, which is involved in the production of serotonin [77,78].
Moreover, the microbiota influences the metabolism of tryptophan, an essential amino acid, a precursor to serotonin (5-HT), which plays a vital role in central nervous system function. Imbalances in serotonin levels are closely linked to the development of mood disorders such as depression and anxiety. However, under inflammatory conditions, much of the tryptophan is diverted to the kynurenine pathway. LPS and pro-inflammatory cytokines activate enzymes like indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO), leading to a reduction in serotonin production. Instead, tryptophan is metabolized into kynurenine, which can be further broken down into compounds like quinolinic acid (QUIN) and kynurenic acid (KYNA). While KYNA typically has neuroprotective functions, imbalances caused by inflammation can lead to neurodegeneration and cognitive issues. QUIN, an NMDA receptor agonist, contributes to excessive synaptic stimulation, causing neuronal damage [65,79,80].

4. Diet

One of the modifiable contributors that influence cognitive health is diet [81]. The correlation between eating habits and intellectual capacity continues to emerge in recent research. There is evidence showing that a variety of mental abilities, including memory, attention or decision-making, are influenced by the type of food consumed [82]. A decline in all these functions lead to conditions such as dementia or Alzheimer’s disease [83]. Acknowledging the relationship between these components is crucial for preventing a cognitive decline in patients by incorporating dietary patterns such as the Mediterranean or Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) diet in their daily life [81].

4.1. Mediterranean Diet

The Mediterranean diet (MD) is characterized by high consumption of antioxidants from fruits and vegetables and monounsaturated fatty acids (MUFAs) from olive oil or nuts [84]. It also consists of a moderate intake of polyunsaturated fatty acids (PUFAs) from fish and poultry, and low consumption of red and processed meat [84,85]. Components such as MUFAs and PUFAs have recently been highlighted in research for their positive effects on cognitive performance and decreased risk of the decline associated with aging [84]. Some studies have also indicated that consuming fruits and vegetables, which are one the main elements of the MD, is beneficial to mental abilities [83]. There are many mechanisms linking the MD to cognitive function. One of them is the anti-oxidative effect resulting from components of the MD diet such as olive oil and nuts that are high in phenolic compounds, which may help reduce oxidative damage in the brain and potentially prevent neurodegeneration [86]. Moreover, a reduction in the cardiovascular disease risk described in 1975 by Ancel Keys is strongly linked to cognitive function via a high intake of healthy fats, which improve heart function and ensure better overall blood flow to the brain [81,84,87]. The other effect of the MD on mental abilities might be enabled by the anti-inflammatory factor in the brain [84]. As reported by Gu et al., individuals who adhered most closely to the MD had substantially lower levels of high-sensitivity CRP [88]. The influence of the MD on cognitive health has also been [84] proved by Feart et al. in their study, where participants who followed the MD had a notably reduced number of errors on the Mini-Mental State Examination (MMSE) and higher scores in higher Free and Cued Selective Reminding Test (FCSRT) [85,89]. A few years later, a group of individuals from the same study went through 3-T magnetic resonance imaging that revealed importantly preserved white matter and an increase in structural connectivity in patients on the MD [90]. All of the benefits coming from implementing the MD are undisputed, as even the World Health Organization (WHO) has put the MD in its guidelines for reducing cognitive decline and the dementia risk [91].
What we eat affects our microbiome, which influences the brain. The Mediterranean diet has a multi-directional effect on the above-mentioned axis, e.g., by modifying the microbiome, which is more diverse than those following a Western diet. Additionally, switching to a Mediterranean diet increases the amount of Prevotella and Firmicutes bacteria, which correlates with an elevation of SCFAs and decreased inflammation [26]. In one of the more interesting studies, a diet rich in whole grains (WGs) markedly reduced TNF-α and LBP, while eating fruits and vegetables (FVs) led to a decrease in IL-6 and LBP [92].

4.2. The Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) Diet

The MIND diet has been developed by researchers at Rush University Medical Center and merges aspects of the Mediterranean and Dietary Approaches to Stop Hypertension (DASH) diets, aiming to preserve cognitive function and protect brain health through the aging process [87]. Developed by researchers at Rush University Medical Center, the MIND diet’s main aim is to promote brain health and reduce the risk of cognitive decline. As it is a combination of diets designed for managing hypertension, the MIND diet also has a positive influence on vascular health [81]. Moreover, it has attracted considerable interest due to its potential to slow the development of neurodegenerative diseases, such as a 53% lower risk of developing Alzheimer’s disease [87]. The main components of the MIND diet are green leafy vegetables, berries, extra-virgin oil, nuts, whole grains and lean protein sources [93]. Animal products and saturated fats are limited due to the association with inflammation and cognitive decline [94]. However, it uniquely focuses on eating berries and leafy greens, while not requiring a high fruit intake or more than one weekly fish meal, as in the Mediterranean and DASH diets [94]. The unique elements such as berries in studies have shown a protective influence on cognitive loss [95]. As research continues to explore this issue, the MIND diet stands out as a powerful tool for maintaining mental acuity and protecting against cognitive decline.

4.3. Vegetarian Diet

The relationship between diet and cognitive function has garnered increasing attention in recent years, particularly as researchers explore how various dietary patterns influence brain health. One of the areas of interest is the vegetarian diet. However, the evidence regarding the cognitive and mental effects of a plant-based diet remains uncertain [96]. A well-planned vegetarian diet is typically rich in nutrient-dense foods such as fruits, vegetables, whole grains, nuts, and seeds. It is typically low in saturated fats and cholesterol, promoting a healthier blood lipid profile. Moreover, it consists of foods high in polyphenols like cocoa, berries or nuts, which provide significant benefits for memory and cognitive functions, particularly in areas like attention and processing speed [97]. On the contrary, a plant-based diet may be deficient in certain vitamins, such as B12 and micronutrients, potentially leading to nutritional deficiencies. B12, also known as cobalamin, is crucial for maintaining neurological health and cognitive function. It is essential for the synthesis of myelin and is also involved in energy metabolism. Deficiency can impair cognitive functions, leading to issues like fatigue, poor concentration, and memory problems leading to dementia. Although a vegetarian diet brings in many nutritional components that have a positive impact on cognitive function, is it important to supplement any deficient nutrients during it, as this can provide significant benefits for the memory [98].

4.4. The Ketogenic Diet

The ketogenic diet is the intake of high-fat and low-carbohydrate foods. It leads to the formation and consumption of ketones as an alternative energy source. Interestingly, in Alzheimer’s disease, there are a reduced number of receptors in the brain for insulin, and there is also the phenomenon of brain insulin resistance [99]. There are studies, using scales such as the MMSE, ADAS-Cog and NM, supporting the thesis that a ketogenic diet can improve the cognitive function of people with Alzheimer’s disease. Undoubtedly, the disadvantage is the emerging hyperlipidemia, threatening cardiovascular health, and vitamin deficiencies [100].
One literature review assessed that for patients with Alzheimer’s, Parkinson’s and mild cognitive impairment, the ketogenic diet minimally improves cognitive function compared to the Mediterranean diet, modified Atkins diet and low-carbohydrate diet, or ketogenic medium-chain triglyceride (kMCT) supplementation [101]. It is currently considered preferable to use MCT oil and BOHB-containing supplements in addition to the standard diet than to use a ketogenic diet in patients with Alzheimer’s disease and Parkinson’s disease [102].

4.5. Time Restriction in Eating (TRE) and Intermittent Fasting (IFA)

TRE and IFA are dietary approaches aimed at reducing food intake throughout the day. Both lead to increased glucose uptake in the brain, which works to prevent neurodegenerative diseases, and also modulate the microbiome. IFA has been proven to have beneficial effects on synaptic plasticity, neurogenesis and BDNF-mediated neuroprotection [103].
In Alzheimer’s disease, this diet causes a decrease in β-amyloid, better synaptic adaptation in the hippocampus, and improved cognitive function, and it keeps the vascular system healthy [104].
Interestingly, intermittent fasting increases Lachnospiraceae and thus butyric acid, which has health-promoting effects [105].

4.6. Ultra-Processed Foods (UPFs)

Ultra-processed foods (UPFs) are favored by the public because of their low cost, time-saving nature and taste. The known and visible effects are obesity and cardiovascular disease [106]. However, what we take into our bodies also has consequences at a mental level. UPF includes, for example, salt, sugar, fats, hydrolyzed proteins, modified starches and hydrogenated or interesterified oil. When consumed in higher amounts, they induce low-grade systemic inflammatory and oxidative changes [107,108]. These changes are seen in the form of dysbiosis, which in turn affects the gut–brain axis [108]. Links have been established between modified foods and brain inflammation, anxiety–depressive behavior and cognitive decline [109]. In a cross-sectional study of a sample of 3632 participants over 60 years of age, it was found that in the animal fluency cognitive test to assess language and executive function in older people, the consumption of highly processed foods correlates with worse outcomes [110].
A recent meta-analysis of 28 studies found that long-term and excessive consumption of UPF has a negative impact on cognitive function, but it is not associated with an increased risk of Alzheimer’s disease or dementia [111]. A different conclusion was reached by the authors of a systematic review in which, after analyzing cohort studies, they found that UPF correlates with this neurodegenerative disease [112].
It has been shown in an animal model (mice) that long-term, 12-month exposure to advanced lipid peroxidation products (ALEs) contained in this food pattern significantly impairs cognitive functions related to the working memory, long-term memory, spatial memory and learning. The study also confirmed that the gut–brain microbiota remains an important link through interactions with ALEs [109].
Mitochondrial dysfunction is implicated in cognitive decline. The exact mechanisms are not fully understood, but it was shown that salt significantly inhibited mitochondrial division and blocked mitophagy, consequently disrupting mitochondrial function and negatively affecting synaptic plasticity. The protective factor was found to be SIRT3, which promotes mitochondrial division and promotes the PINK1/Parkin-dependent pathway associated with proper mitophagy [113].
There are reports that excess AGEs in the circulatory system and skin are linked to cognitive disabilities, dementia, as well as depression and schizophrenia. Underlying the pathophysiology is oxidative stress and neuroinflammation [114].
A study by Varun M. Bhave et al. showed that UPF negatively affects memory and fluency assessments in older adults, regardless of risk factors and adherence to recommended dietary patterns such as the MIND, MD, and DASH [115].
It is also essential to be aware that UPFs have different potential for harm among their categories, as shown by the NOVA classification. Based on the example of the Galit Weinstein study, highly processed meat and oils were shown to correlate with accelerated cognitive decline, such as executive functions and global cognition, in older people with type 2 diabetes, especially among women and obese people, while food calories did not find a link. In the same study, it was shown that UPF, originating from dairy products, bread, biscuits and starch, was not related to changes in cognitive function.
Intestinal dysbiosis may have its origins in the high-fructose diet that is part of the Western diet. Modification of the microbiota composition has been shown to be a critical factor in mouse hippocampal neuroinflammation. The conclusions from this study include a protective role for SCFAs against the defective NLRP6 inflammasome of the colon, which is responsible for impairment of the intestinal epithelial barrier [116].
It is considered that the use of this diet leads to damage to the intestinal barrier and thus the previously discussed increased intestinal permeability [117].
The negative effects include cognitive processes. One study evaluating the effects of a high-fat and high-sugar diet noted similarities such as increased Clostridiales compared to a normal diet. Increased Clostridiales and decreased Bacteroidales in high-fat diets were correlated with impaired cognitive flexibility. Interestingly, in the high-sugar diet group, deterioration of the long-term and short-term memory could be observed compared to mice on a normal diet [118].

5. Role of Supplementation on Cognitive Functions

It is widely recognized that cognitive health may be supported by maintaining a healthy lifestyle, comprising a balanced diet, regular physical activity, and mental engagement, thus prolonging life expectancy and delaying the onset of dementia. Additionally, the role of dietary supplementation has become increasingly significant. In recent years, studies have emphasized the potential role of nutritional supplements in supporting and enhancing cognitive function [119,120].
Research has shown the beneficial effects of folic acid, vitamin B12, and vitamin B6 in maintaining cognitive function [121]. Ma et al. found that 24-month folic acid (FA) supplementation improves cognitive function in mild cognitive impairment (MCI) [122]. The role of FA and B12 supplementation in elderly healthy people or people with any dementia or cognitive impairment was studied. FA and vitamin B12 supplementation showed a positive therapeutic effect in improving cognitive functions. When considering the combination of FA and vitamin B12, it was noted that it was significantly superior to either folic acid or vitamin B12 alone [123,124,125]. Elevated serum homocysteine levels have been associated with the development of Alzheimer’s dementia. The study by Sun et al. determined that supplementation with vitamins B6, B12, and FA decreased the homocysteine concentrations in patients with mild to moderate Alzheimer’s disease [126].
Further, these studies confirmed that supplementation with FA and docosahexaenoic acid (DHA), either alone or combined, significantly improves cognitive function and reduces plasma inflammatory cytokines in individuals with MCI. However, the combined use of FA and DHA is more effective than each nutrient taken separately [127]. Zhang et al. conducted a study that suggests that DHA supplementation (2 g/day) for 12 months in individuals with MCI can significantly enhance cognitive function and slow the progression of hippocampal atrophy [128,129].
Additionally, it has been shown that in healthy people, omega-3 fatty acids support cognitive performance and brain health [130,131,132]. Fatty acids, in particular, eicosapentaenoic acid (EPA) and DHA, are essential components of brain cell membranes and play a role in several neurophysiological functions [133]. DHA, the most prevalent omega-3 fatty acid in the brain, is well recognized for its contribution to the enhancement of neuronal signaling and synaptic plasticity, two essential components of cognitive functions like learning and memory [134]. Moreover, EPA supports cognitive health through its anti-inflammatory abilities, which helps to lessen neuroinflammation, which is a factor in both cognitive decline and the onset of neurodegenerative diseases [135]. Furthermore, increasing dietary consumption of omega-3 fatty acids and carotenoids may help lower the risk of dementia and cognitive deterioration in later life [136,137]. Nonetheless, these studies have shown no statistically significant effect on cognitive function [138,139,140,141]. Further research is needed to establish the complex interplay between omega-3 fatty acids supplementation and MCI pathophysiology, and therefore, to develop novel effective therapeutic interventions.
Adequate levels of vitamin D may support cognitive health, potentially improving memory and overall cognitive performance, particularly in older adults [142,143]. Supplementation is especially important for the health of the cerebrovascular system, as vitamin D lowers the risk of cerebrovascular diseases, which can affect cognitive function and maintain blood flow to the brain [144]. It has promise as a critical nutrient in preserving brain health through its roles in neuroprotection, neurotransmission, and brain plasticity [145,146]. Yang et al. noted that vitamin D in adults with MCI improves cognitive function through reducing oxidative stress regulated by increased telomere length [147]. Yet while these findings are encouraging, more research is required to definitively prove the link between vitamin D supplementations.
Maintaining cognitive health requires minerals like copper, zinc, iron, magnesium, selenium, and zinc, especially in the elderly population. Supplementing with them can help with a range of brain functions, including cognitive function, and lower the risk of neurodegenerative disorders [148].
On the contrary, the meta-analysis by McCleery et al. included 28 studies with over 83,000 participants and found limited evidence that vitamin or mineral supplements significantly improve cognitive function or reduce the dementia risk. B vitamins, antioxidant vitamins, vitamin D, calcium, zinc, and copper showed little to no effect on cognitive function in healthy older adults. The studies generally had low- to moderate-certainty evidence, and adverse events were rarely reported. While there was some evidence of benefit from long-term antioxidant use, overall, the supplements did not have a significant impact on cognition or dementia prevention [149].
Nutritional supplements can play a supportive role in enhancing cognitive function, particularly in cases of dietary insufficiencies or increased demands on brain health. B vitamins, omega-3 fatty acids, B vitamins, and D vitamins all show promise in improving aspects of cognition such as memory, attention, and mood. Further investigation is warranted to advance our knowledge and establish novel treatments and preventive strategies. Thus, the benefits of supplements should be considered as part of a broader approach to cognitive health, including a balanced diet, regular physical activity, and mental exercises.

6. Potential Microbiota-Targeted Therapies for Cognitive Impairment Disorders

Research has demonstrated that individuals with PD and AD often exhibit altered gut microbiota profiles, characterized by a decrease in beneficial bacteria and an increase in pro-inflammatory species. This state may contribute to the neuroinflammatory processes and neurodegeneration observed in these conditions [150,151]. Targeting the gut microbiota to restore the balance may thus have significant therapeutic implications.
Prebiotics are food components that are selectively utilized by host bacteria, providing health benefits. According to Liu et al.’s findings, mannan oligosaccharide (MOS) improves behavioral and cognitive impairments, possibly via altering the gut microbiome and increasing SCFA production, proposing MOS as a potential microbiota-targeted therapy for neurodegenerative illnesses such as Alzheimer’s. MOS significantly reduces Aβ accumulation in the cortex, hippocampus, and amygdala, while also balancing brain redox status and suppressing neuroinflammatory responses. Additionally, MOS alleviates HPA axis dysfunction by lowering the corticosterone (CORT) and corticotropin-releasing hormone (CRH) levels while increasing norepinephrine (NE) expression [152].
Probiotics are live microorganisms, which, when consumed in adequate amounts, provide health advantages to the host. Interestingly, probiotics have been shown in both PD and AD models to reduce neuroinflammation, improve gut barrier integrity, and increase synthesis of neuroprotective metabolites such SCFAs [150,151]. According to Castelli et al., supplementing with a probiotic cocktail that included Lactobacillus acidophilus, Bifidobacterium animalis lactis, and L. rhamnosus GG increased butyrate synthesis, which in turn decreased the loss of nigral dopaminergic neurons [153]. Nimgampalle et al. examined the effects of Lactobacillus plantarum MTCC 1325 on D-galactose-induced AD in mice. The findings revealed a decrease in neurofibrillary tangles (NFTs) and amyloid plaques, along with increased acetylcholine (ACh) levels in the hippocampus and cerebral cortex. Additionally, the memory and spatial learning were improved in the treated mice. These effects are likely due to the production of ACh by Lactobacillus plantarum MTCC 1325, a neurotransmitter with potent antioxidant properties that may stabilize the amyloid-beta structure and reduce Aβ aggregation [154].
FMT has emerged as a promising therapeutic intervention for a range of gastrointestinal disorders, primarily focusing on conditions like Clostridioides difficile infection [155]; however, its potential associations with cognitive impairment are drawing increasing attention in the field of neurogastroenterology. Recent studies suggest that the gut–brain axis, a bidirectional communication system between the gastrointestinal tract and the central nervous system, plays a crucial role in cognitive functions and mental health [156,157]. Alterations in the gut microbiota composition have been linked to various neurological conditions, including Alzheimer’s disease [158]. Emerging research indicates that restoring microbial diversity through FMT may influence neuroprotective pathways, modulate inflammatory responses, and impact neurotransmitter levels, including those of serotonin and dopamine, which are vital for cognitive health [159,160]. Animal studies have shown that FMT can improve cognitive function in models of neurodegeneration, suggesting that the gut microbiota may directly influence neurodevelopment and neuroplasticity. While human studies remain limited, preliminary findings indicate potential improvements in cognitive performance and mood in individuals undergoing FMT for conditions associated with dysbiosis [161,162]. As the understanding of the complex interplay between the gut microbiota and brain health evolves, these findings may provide new therapeutic strategies for managing cognitive impairment, although further research is crucial.

7. Conclusions

In conclusion, the underlying processes through which dietary intake influences cognitive function are complex and multifaceted, involving the modulation of neuroinflammation, oxidative stress, synaptic plasticity, and, increasingly recognized, the gut–brain axis [81]. Nutrient-rich diets, particularly those high in omega-3 fatty acids, antioxidants, and vitamins, are linked to improved cognitive function, delayed decline, and reduced risk of neurodegenerative diseases [163,164]. In contrast, diets rich in saturated fats, sugars, and processed foods contribute to cognitive impairment and dementia [165,166]. The gut–brain axis plays a critical role, as a healthy gut microbiome supports brain health, while poor nutrition disrupts this balance. Holistic dietary patterns like the Mediterranean and DASH diets, rich in fruits, vegetables, whole grains, and healthy fats, benefit both brain and gut health, enhancing overall cognitive function [167].
However, while the correlation between diet and cognitive function is compelling, establishing causality remains challenging due to the inherent complexity of diet-related research, the influence of confounding variables, and the long-term nature of cognitive outcomes. Future research should aim to further elucidate these mechanisms, particularly the role of the gut–brain axis, employ more rigorous methodologies, and explore the potential of personalized nutrition interventions to optimize cognitive function.

Author Contributions

Conceptualization, B.F., E.M. and J.R.; methodology, E.M., P.J., W.F., A.G., J.S., S.S. and J.W.; software, E.M.; validation, B.F., E.M. and J.R.; formal analysis, E.M., P.J., W.F., A.G., J.S., S.S. and J.W.; investigation, E.M., P.J., W.F., A.G., J.S., S.S. and J.W.; resources, B.F., E.M. and J.R; data curation, E.M.; writing—original draft preparation, E.M., P.J., W.F., A.G., J.S., S.S. and J.W.; writing—review and editing, E.M.; visualization, E.M., P.J., W.F., A.G., J.S., S.S. and J.W.; supervision, B.F., E.M. and J.R; project administration, B.F., E.M. and J.R.; funding acquisition, B.F. and J.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bettio, L.E.B.; Rajendran, L.; Gil-Mohapel, J. The Effects of Aging in the Hippocampus and Cognitive Decline. Neurosci. Biobehav. Rev. 2017, 79, 66–86. [Google Scholar] [CrossRef] [PubMed]
  2. Kandlur, A.; Satyamoorthy, K.; Gangadharan, G. Oxidative Stress in Cognitive and Epigenetic Aging: A Retrospective Glance. Front. Mol. Neurosci. 2020, 13, 41. [Google Scholar] [CrossRef] [PubMed]
  3. Marzola, P.; Melzer, T.; Pavesi, E.; Gil-Mohapel, J.; Brocardo, P.S. Exploring the Role of Neuroplasticity in Development, Aging, and Neurodegeneration. Brain Sci. 2023, 13, 1610. [Google Scholar] [CrossRef] [PubMed]
  4. Jin, M.; Cai, S.Q. Mechanisms Underlying Brain Aging Under Normal and Pathological Conditions. Neurosci. Bull. 2023, 39, 303–314. [Google Scholar] [CrossRef] [PubMed]
  5. Zimmerman, B.; Rypma, B.; Gratton, G.; Fabiani, M. Age-Related Changes in Cerebrovascular Health and Their Effects on Neural Function and Cognition: A Comprehensive Review. Psychophysiology 2021, 58, e13796. [Google Scholar] [CrossRef]
  6. Mattson, M.P.; Arumugam, T.V. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metab. 2018, 27, 1176–1199. [Google Scholar] [CrossRef]
  7. Mormino, E.C.; Papp, K.V. Amyloid Accumulation and Cognitive Decline in Clinically Normal Older Individuals: Implications for Aging and Early Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 64, S633–S646. [Google Scholar] [CrossRef]
  8. Aarsland, D.; Creese, B.; Politis, M.; Chaudhuri, K.R.; Ffytche, D.H.; Weintraub, D.; Ballard, C. Cognitive Decline in Parkinson Disease. Nat. Rev. Neurol. 2017, 13, 217–231. [Google Scholar] [CrossRef]
  9. Brisch, R.; Saniotis, A.; Wolf, R.; Bielau, H.; Bernstein, H.G.; Steiner, J.; Bogerts, B.; Braun, K.; Kumaratilake, J.; Henneberg, M.; et al. The Role of Dopamine in Schizophrenia from a Neurobiological and Evolutionary Perspective: Old Fashioned, but Still in Vogue. Front. Psychiatry 2014, 5, 47. [Google Scholar]
  10. Li, H.; Hirano, S.; Furukawa, S.; Nakano, Y.; Kojima, K.; Ishikawa, A.; Tai, H.; Horikoshi, T.; Iimori, T.; Uno, T.; et al. The Relationship Between the Striatal Dopaminergic Neuronal and Cognitive Function with Aging. Front. Aging Neurosci. 2020, 12, 41. [Google Scholar] [CrossRef]
  11. Štrac, D.Š.; Pivac, N.; Mück-Šeler, D. The Serotonergic System and Cognitive Function. Transl. Neurosci. 2016, 7, 35–49. [Google Scholar] [CrossRef] [PubMed]
  12. Reddy-Thootkur, M.; Kraguljac, N.V.; Lahti, A.C. The Role of Glutamate and GABA in Cognitive Dysfunction in Schizophrenia and Mood Disorders—A Systematic Review of Magnetic Resonance Spectroscopy Studies. Schizophr. Res. 2022, 249, 74–84. [Google Scholar] [CrossRef]
  13. Fourrier, C.; Singhal, G.; Baune, B.T. Neuroinflammation and Cognition across Psychiatric Conditions. CNS Spectrums 2019, 24, 4–15. [Google Scholar] [CrossRef]
  14. Yang, J.; Ran, M.; Li, H.; Lin, Y.; Ma, K.; Yang, Y.; Fu, X.; Yang, S. New Insight into Neurological Degeneration: Inflammatory Cytokines and Blood–Brain Barrier. Front. Mol. Neurosci. 2022, 15, 1013933. [Google Scholar] [CrossRef]
  15. Gąssowska-Dobrowolska, M.; Chlubek, M.; Kolasa, A.; Tomasiak, P.; Korbecki, J.; Skowrońska, K.; Tarnowski, M.; Masztalewicz, M.; Baranowska-Bosiacka, I. Microglia and Astroglia—The Potential Role in Neuroinflammation Induced by Pre- and Neonatal Exposure to Lead (Pb). Int. J. Mol. Sci. 2023, 24, 9903. [Google Scholar] [CrossRef] [PubMed]
  16. Franzoni, F.; Scarfò, G.; Guidotti, S.; Fusi, J.; Asomov, M.; Pruneti, C. Oxidative Stress and Cognitive Decline: The Neuroprotective Role of Natural Antioxidants. Front. Neurosci. 2021, 15, 729757. [Google Scholar] [CrossRef]
  17. Daniels, T.E.; Olsen, E.M.; Tyrka, A.R. Stress and Psychiatric Disorders: The Role of Mitochondria. Annu. Rev. Clin. Psychol. 2020, 16, 165–186. [Google Scholar] [CrossRef] [PubMed]
  18. Teleanu, D.M.; Niculescu, A.G.; Lungu, I.I.; Radu, C.I.; Vladâcenco, O.; Roza, E.; Costăchescu, B.; Grumezescu, A.M.; Teleanu, R.I. An Overview of Oxidative Stress, Neuroinflammation and Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 5938. [Google Scholar] [CrossRef]
  19. Biessels, G.; Despa, F. Cognitive Decline and Dementia in Diabetes Mellitus: Mechanisms and Clinical Implications. Nat. Rev. Endocrinol. 2018, 14, 591–604. [Google Scholar] [CrossRef]
  20. Croall, I.D.; Tooth, C.; Venneri, A.; Poyser, C.; Sanders, D.S.; Hoggard, N.; Hadjivassiliou, M. Cognitive Impairment in Coeliac Disease with Respect to Disease Duration and Gluten-Free Diet Adherence: A Pilot Study. Nutrients 2020, 12, 2028. [Google Scholar] [CrossRef]
  21. Saghafi-Asl, M.; Aliasgharzadeh, S.; Asghari-Jafarabadi, M. Factors Influencing Weight Management Behavior among College Students: An Application of the Health Belief Model. PLoS ONE 2020, 15, e0228058. [Google Scholar] [CrossRef] [PubMed]
  22. Li, G.; Hu, Y.; Zhang, W.; Wang, J.; Ji, W.; Manza, P.; Volkow, N.D.; Zhang, Y.; Wang, G.J. Brain Functional and Structural Magnetic Resonance Imaging of Obesity and Weight Loss Interventions. Mol. Psychiatry 2023, 28, 1466–1479. [Google Scholar] [CrossRef]
  23. Anstey, K.J.; Cherbuin, N.; Budge, M.; Young, J. Body Mass Index in Midlife and Late-Life as a Risk Factor for Dementia: A Meta-Analysis of Prospective Studies. Obes. Rev. 2011, 12, e426–e437. [Google Scholar] [CrossRef] [PubMed]
  24. Nguyen, J.C.D.; Killcross, A.S.; Jenkins, T.A. Obesity and Cognitive Decline: Role of Inflammation and Vascular Changes. Front. Neurosci. 2014, 8, 375. [Google Scholar] [CrossRef]
  25. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.; Gasbarrini, A.; Mele, M. What Is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed]
  26. Beam, A.; Clinger, E.; Hao, L. Effect of Diet and Dietary Components on the Composition of the Gut Microbiota. Nutrients 2021, 13, 2795. [Google Scholar] [CrossRef]
  27. McCallum, G.; Tropini, C. The Gut Microbiota and Its Biogeography. Nat. Rev. Microbiol. 2024, 22, 105–118. [Google Scholar] [CrossRef] [PubMed]
  28. Pantazi, A.C.; Balasa, A.L.; Mihai, C.M.; Chisnoiu, T.; Lupu, V.V.; Kassim, M.A.K.; Mihai, L.; Frecus, C.E.; Chirila, S.I.; Lupu, A.; et al. Development of Gut Microbiota in the First 1000 Days after Birth and Potential Interventions. Nutrients 2023, 15, 3647. [Google Scholar] [CrossRef] [PubMed]
  29. Leeming, E.R.; Johnson, A.J.; Spector, T.D.; Le Roy, C.I. Effect of Diet on the Gut Microbiota: Rethinking Intervention Duration. Nutrients 2019, 11, 2862. [Google Scholar] [CrossRef]
  30. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.-Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef]
  31. Campaniello, D.; Corbo, M.R.; Sinigaglia, M.; Speranza, B.; Racioppo, A.; Altieri, C.; Bevilacqua, A. How Diet and Physical Activity Modulate Gut Microbiota: Evidence, and Perspectives. Nutrients 2022, 14, 2456. [Google Scholar] [CrossRef] [PubMed]
  32. Młynarska, E.; Wasiak, J.; Gajewska, A.; Steć, G.; Jasińska, J.; Rysz, J.; Franczyk, B. Exploring the Significance of Gut Microbiota in Diabetes Pathogenesis and Management-A Narrative Review. Nutrients 2024, 16, 1938. [Google Scholar] [CrossRef]
  33. Wu, Q.; Xu, Z.; Song, S.; Zhang, H.; Zhang, W.; Liu, L.; Chen, Y.; Sun, J. Gut Microbiota Modulates Stress-Induced Hypertension through the HPA Axis. Brain Res. Bull. 2020, 162, 49–58. [Google Scholar] [CrossRef]
  34. Geng, J.; Ni, Q.; Sun, W.; Li, L.; Feng, X. The Links between Gut Microbiota and Obesity and Obesity Related Diseases. Biomed. Pharmacother. 2022, 147, 112678. [Google Scholar] [CrossRef] [PubMed]
  35. Ohkusa, T.; Koido, S.; Nishikawa, Y.; Sato, N. Gut Microbiota and Chronic Constipation: A Review and Update. Front. Med. 2019, 6, 19. [Google Scholar] [CrossRef] [PubMed]
  36. Napolitano, M.; Fasulo, E.; Ungaro, F.; Massimino, L.; Sinagra, E.; Danese, S.; Mandarino, F.V. Gut Dysbiosis in Irritable Bowel Syndrome: A Narrative Review on Correlation with Disease Subtypes and Novel Therapeutic Implications. Microorganisms 2023, 11, 2369. [Google Scholar] [CrossRef]
  37. Mandarino, F.V.; Sinagra, E.; Barchi, A.; Verga, M.C.; Brinch, D.; Raimondo, D.; Danese, S. Gastroparesis: The Complex Interplay with Microbiota and the Role of Exogenous Infections in the Pathogenesis of the Disease. Microorganisms 2023, 11, 1122. [Google Scholar] [CrossRef] [PubMed]
  38. Rahman, M.M.; Islam, F.; Or-Rashid, M.H.; Al Mamun, A.; Rahaman, M.S.; Islam, M.M.; Meem, A.F.K.; Sutradhar, P.R.; Mitra, S.; Mimi, A.A.; et al. The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation. Front. Cell. Infect. Microbiol. 2022, 12, 903570. [Google Scholar] [CrossRef]
  39. Yang, T.; Richards, E.M.; Pepine, C.J.; Raizada, M.K. The Gut Microbiota and the Brain–Gut–Kidney Axis in Hypertension and Chronic Kidney Disease. Nat. Rev. Nephrol. 2018, 14, 442–456. [Google Scholar] [CrossRef]
  40. Zmora, N.; Suez, J.; Elinav, E. You Are What You Eat: Diet, Health and the Gut Microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 35–56. [Google Scholar] [CrossRef]
  41. Colella, M.; Charitos, I.A.; Ballini, A.; Cafiero, C.; Topi, S.; Palmirotta, R.; Santacroce, L. Microbiota Revolution: How Gut Microbes Regulate Our Lives. World J. Gastroenterol. 2023, 29, 4368–4383. [Google Scholar] [CrossRef] [PubMed]
  42. Grosso, G. Nutritional Psychiatry: How Diet Affects Brain through Gut Microbiota. Nutrients 2021, 13, 1282. [Google Scholar] [CrossRef] [PubMed]
  43. Margolis, K.G.; Cryan, J.F.; Mayer, E.A. The Microbiota-Gut-Brain Axis: From Motility to Mood. Gastroenterology 2021, 160, 1486–1501. [Google Scholar] [CrossRef] [PubMed]
  44. Mayer, E.A.; Nance, K.; Chen, S. The Gut–Brain Axis. Annu. Rev. Med. 2022, 73, 439–453. [Google Scholar] [CrossRef] [PubMed]
  45. Nguyen, T.T.; Baumann, P.; Tüscher, O.; Schick, S.; Endres, K. The Aging Enteric Nervous System. Int. J. Mol. Sci. 2023, 24, 9471. [Google Scholar] [CrossRef]
  46. Cavin, J.-B.; Cuddihey, H.; MacNaughton, W.K.; Sharkey, K.A. Acute Regulation of Intestinal Ion Transport and Permeability in Response to Luminal Nutrients: The Role of the Enteric Nervous System. Am. J. Physiol.-Gastrointest. Liver Physiol. 2020, 318, G254–G264. [Google Scholar] [CrossRef]
  47. Furness, J.B. The Enteric Nervous System and Neurogastroenterology. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 286–294. [Google Scholar] [CrossRef]
  48. Martin-Gallausiaux, C.; Marinelli, L.; Blottière, H.M.; Larraufie, P.; Lapaque, N. SCFA: Mechanisms and Functional Importance in the Gut. Proc. Nutr. Soc. 2021, 80, 37–49. [Google Scholar] [CrossRef]
  49. Komorniak, N.; Martynova-Van Kley, A.; Nalian, A.; Wroński, M.; Kaseja, K.; Kowalewski, B.; Kaźmierczak-Siedlecka, K.; Łoniewski, I.; Kaczmarczyk, M.; Podsiadło, K.; et al. Association between Fecal Microbiota, SCFA, Gut Integrity Markers and Depressive Symptoms in Patients Treated in the Past with Bariatric Surgery—The Cross-Sectional Study. Nutrients 2022, 14, 5372. [Google Scholar] [CrossRef]
  50. Cheung, S.G.; Goldenthal, A.R.; Uhlemann, A.-C.; Mann, J.J.; Miller, J.M.; Sublette, M.E. Systematic Review of Gut Microbiota and Major Depression. Front. Psychiatry 2019, 10, 34. [Google Scholar] [CrossRef]
  51. Obrenovich, M.E.M. Leaky Gut, Leaky Brain? Microorganisms 2018, 6, 107. [Google Scholar] [CrossRef] [PubMed]
  52. Dicks, L.M.T. Gut Bacteria and Neurotransmitters. Microorganisms 2022, 10, 1838. [Google Scholar] [CrossRef] [PubMed]
  53. Wasiak, J.; Gawlik-Kotelnicka, O. Intestinal Permeability and Its Significance in Psychiatric Disorders—A Narrative Review and Future Perspectives. Behav. Brain Res. 2023, 448, 114459. [Google Scholar] [CrossRef]
  54. Odenwald, M.A.; Turner, J.R. Intestinal Permeability Defects: Is It Time to Treat? Clin. Gastroenterol. Hepatol. 2013, 11, 1075–1083. [Google Scholar] [CrossRef]
  55. Quigley, E.M.M. Leaky Gut-Concept or Clinical Entity? Curr. Opin. Gastroenterol. 2016, 32, 74–79. [Google Scholar] [CrossRef] [PubMed]
  56. Mu, Q.; Kirby, J.; Reilly, C.M.; Luo, X.M. Leaky Gut as a Danger Signal for Autoimmune Diseases. Front. Immunol. 2017, 8, 598. [Google Scholar] [CrossRef]
  57. Vaure, C.; Liu, Y. A Comparative Review of Toll-Like Receptor 4 Expression and Functionality in Different Animal Species. Front. Immunol. 2014, 5, 316. [Google Scholar] [CrossRef]
  58. Brown, G.C. The Endotoxin Hypothesis of Neurodegeneration. J. Neuroinflamm. 2019, 16, 180. [Google Scholar] [CrossRef]
  59. Sandiego, C.M.; Gallezot, J.-D.; Pittman, B.; Nabulsi, N.; Lim, K.; Lin, S.-F.; Matuskey, D.; Lee, J.-Y.; O’Connor, K.C.; Huang, Y.; et al. Imaging Robust Microglial Activation after Lipopolysaccharide Administration in Humans with PET. Proc. Natl. Acad. Sci. USA 2015, 112, 12468–12473. [Google Scholar] [CrossRef]
  60. Bodea, L.-G.; Wang, Y.; Linnartz-Gerlach, B.; Kopatz, J.; Sinkkonen, L.; Musgrove, R.; Kaoma, T.; Muller, A.; Vallar, L.; Di Monte, D.A.; et al. Neurodegeneration by Activation of the Microglial Complement-Phagosome Pathway. J. Neurosci. 2014, 34, 8546–8556. [Google Scholar] [CrossRef]
  61. Misiak, B.; Łoniewski, I.; Marlicz, W.; Frydecka, D.; Szulc, A.; Rudzki, L.; Samochowiec, J. The HPA Axis Dysregulation in Severe Mental Illness: Can We Shift the Blame to Gut Microbiota? Prog. Neuropsychopharmacol. Biol. Psychiatry 2020, 102, 109951. [Google Scholar] [CrossRef] [PubMed]
  62. Kinashi, Y.; Hase, K. Partners in Leaky Gut Syndrome: Intestinal Dysbiosis and Autoimmunity. Front. Immunol. 2021, 12, 673708. [Google Scholar] [CrossRef]
  63. Kealy, J.; Greene, C.; Campbell, M. Blood-Brain Barrier Regulation in Psychiatric Disorders. Neurosci. Lett. 2020, 726, 133664. [Google Scholar] [CrossRef]
  64. Daneman, R.; Prat, A. The Blood–Brain Barrier. Cold Spring Harb. Perspect. Biol. 2015, 7, a020412. [Google Scholar] [CrossRef]
  65. Rudzki, L.; Maes, M. The Microbiota-Gut-Immune-Glia (MGIG) Axis in Major Depression. Mol. Neurobiol. 2020, 57, 4269–4295. [Google Scholar] [CrossRef]
  66. Sofroniew, M.V. Astrocyte Barriers to Neurotoxic Inflammation. Nat. Rev. Neurosci. 2015, 16, 249–263. [Google Scholar] [CrossRef] [PubMed]
  67. Dienel, S.J.; Enwright, J.F.; Hoftman, G.D.; Lewis, D.A. Markers of Glutamate and GABA Neurotransmission in the Prefrontal Cortex of Schizophrenia Subjects: Disease Effects Differ across Anatomical Levels of Resolution. Schizophr. Res. 2020, 217, 86–94. [Google Scholar] [CrossRef]
  68. Brown, G.; Heneka, M. The Endotoxin Hypothesis of Alzheimer’s Disease. Mol. Neurodegener. 2024, 19, 30. [Google Scholar] [CrossRef] [PubMed]
  69. Brown, G.C.; Camacho, M.; Williams-Gray, C.H. The Endotoxin Hypothesis of Parkinson’s Disease. Mov. Disord. 2023, 38, 1143–1155. [Google Scholar] [CrossRef]
  70. Huang, Y.; Wang, Y.F.; Miao, J.; Zheng, R.F.; Li, J.Y. Short-Chain Fatty Acids: Important Components of the Gut-Brain Axis against AD. Biomed. Pharmacother. 2024, 175, 116601. [Google Scholar] [CrossRef]
  71. Kalyanaraman, B.; Cheng, G.; Hardy, M. Gut Microbiome, Short-Chain Fatty Acids, Alpha-Synuclein, Neuroinflammation, and ROS/RNS: Relevance to Parkinson’s Disease and Therapeutic Implications. Redox Biol. 2024, 71, 103092. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef] [PubMed]
  73. Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The Short-Chain Fatty Acid Acetate Reduces Appetite via a Central Homeostatic Mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef]
  74. Strandwitz, P.; Kim, K.H.; Terekhova, D.; Liu, J.K.; Sharma, A.; Levering, J.; McDonald, D.; Dietrich, D.; Ramadhar, T.R.; Lekbua, A.; et al. GABA-Modulating Bacteria of the Human Gut Microbiota. Nat. Microbiol. 2018, 4, 396–403. [Google Scholar] [CrossRef]
  75. Estrela, A.B.; Abraham, W.-R. Adenosine in the Inflamed Gut: A Janus Faced Compound. Curr. Med. Chem. 2011, 18, 2791–2815. [Google Scholar] [CrossRef]
  76. Mittal, R.; Debs, L.H.; Patel, A.P.; Nguyen, D.; Patel, K.; O’Connor, G.; Grati, M.; Mittal, J.; Yan, D.; Eshraghi, A.A.; et al. Neurotransmitters: The Critical Modulators Regulating Gut–Brain Axis. J. Cell. Physiol. 2017, 232, 2359–2372. [Google Scholar] [CrossRef]
  77. Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The Role of Short-Chain Fatty Acids in Microbiota–Gut–Brain Communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef]
  78. González-Arancibia, C.; Urrutia-Piñones, J.; Illanes-González, J.; Martinez-Pinto, J.; Sotomayor-Zárate, R.; Julio-Pieper, M.; Bravo, J.A. Do Your Gut Microbes Affect Your Brain Dopamine? Psychopharmacology 2019, 236, 1611–1622. [Google Scholar] [CrossRef] [PubMed]
  79. Rudzki, L.; Maes, M. From “Leaky Gut” to Impaired Glia-Neuron Communication in Depression. In Major Depressive Disorder: Rethinking and Understanding Recent Discoveries; Springer: Singapore, 2021; pp. 129–155. [Google Scholar]
  80. Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G. Kynurenine Pathway Metabolism and the Microbiota-Gut-Brain Axis. Neuropharmacology 2017, 112, 399–412. [Google Scholar] [CrossRef]
  81. Puri, S.; Shaheen, M.; Grover, B. Nutrition and Cognitive Health: A Life Course Approach. Front. Public Health 2023, 11, 1023907. [Google Scholar] [CrossRef]
  82. Attuquayefio, T.; Stevenson, R.J. A Systematic Review of Longer-Term Dietary Interventions on Human Cognitive Function: Emerging Patterns and Future Directions. Appetite 2015, 95, 554–570. [Google Scholar] [CrossRef] [PubMed]
  83. Fortune, N.C.; Harville, E.W.; Guralnik, J.M.; Gustat, J.; Chen, W.; Qi, L.; Bazzano, L.A. Dietary Intake and Cognitive Function: Evidence from the Bogalusa Heart Study. Am. J. Clin. Nutr. 2019, 109, 1656–1663. [Google Scholar] [CrossRef] [PubMed]
  84. Petersson, S.D.; Philippou, E. Mediterranean Diet, Cognitive Function, and Dementia: A Systematic Review of the Evidence. Adv. Nutr. 2016, 7, 889–904. [Google Scholar] [CrossRef] [PubMed]
  85. Siervo, M.; Shannon, O.M.; Llewellyn, D.J.; Stephan, B.C.; Fontana, L. Mediterranean Diet and Cognitive Function: From Methodology to Mechanisms of Action. Free Radic. Biol. Med. 2021, 176, 105–117. [Google Scholar] [CrossRef]
  86. Valls-Pedret, C.; Sala-Vila, A.; Serra-Mir, M.; Corella, D.; de la Torre, R.; Martínez-González, M.Á.; Martínez-Lapiscina, E.H.; Fitó, M.; Pérez-Heras, A.; Salas-Salvadó, J.; et al. Mediterranean Diet and Age-Related Cognitive Decline. JAMA Intern Med. 2015, 175, 1094. [Google Scholar] [CrossRef]
  87. Cena, H.; Calder, P.C. Defining a Healthy Diet: Evidence for the Role of Contemporary Dietary Patterns in Health and Disease. Nutrients 2020, 12, 334. [Google Scholar] [CrossRef]
  88. Gu, Y.; Luchsinger, J.A.; Stern, Y.; Scarmeas, N. Mediterranean Diet, Inflammatory and Metabolic Biomarkers, and Risk of Alzheimer’s Disease. J. Alzheimer’s Dis. 2010, 22, 483–492. [Google Scholar] [CrossRef]
  89. Féart, C. Adherence to a Mediterranean Diet, Cognitive Decline, and Risk of Dementia. JAMA 2009, 302, 638. [Google Scholar] [CrossRef]
  90. Pelletier, A.; Barul, C.; Féart, C.; Helmer, C.; Bernard, C.; Periot, O.; Dilharreguy, B.; Dartigues, J.; Allard, M.; Barberger-Gateau, P.; et al. Mediterranean Diet and Preserved Brain Structural Connectivity in Older Subjects. Alzheimer’s Dement. 2015, 11, 1023–1031. [Google Scholar] [CrossRef]
  91. World Health Organization. Risk Reduction of Cognitive Decline and Dementia: WHO Guidelines; World Health Organization: Geneva, Switzerland, 2019. [Google Scholar]
  92. Kopf, J.C.; Suhr, M.J.; Clarke, J.; Eyun, S.; Riethoven, J.-J.M.; Ramer-Tait, A.E.; Rose, D.J. Role of Whole Grains versus Fruits and Vegetables in Reducing Subclinical Inflammation and Promoting Gastrointestinal Health in Individuals Affected by Overweight and Obesity: A Randomized Controlled Trial. Nutr. J. 2018, 17, 72. [Google Scholar] [CrossRef]
  93. Liu, X.; Morris, M.C.; Dhana, K.; Ventrelle, J.; Johnson, K.; Bishop, L.; Hollings, C.S.; Boulin, A.; Laranjo, N.; Stubbs, B.J.; et al. Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) Study: Rationale, Design and Baseline Characteristics of a Randomized Control Trial of the MIND Diet on Cognitive Decline. Contemp. Clin. Trials 2021, 102, 106270. [Google Scholar] [CrossRef] [PubMed]
  94. Morris, M.C.; Tangney, C.C.; Wang, Y.; Sacks, F.M.; Barnes, L.L.; Bennett, D.A.; Aggarwal, N.T. MIND Diet Slows Cognitive Decline with Aging. Alzheimer’s Dement. 2015, 11, 1015–1022. [Google Scholar] [CrossRef] [PubMed]
  95. Barnes, L.L.; Dhana, K.; Liu, X.; Carey, V.J.; Ventrelle, J.; Johnson, K.; Hollings, C.S.; Bishop, L.; Laranjo, N.; Stubbs, B.J.; et al. Trial of the MIND Diet for Prevention of Cognitive Decline in Older Persons. N. Engl. J. Med. 2023, 389, 602–611. [Google Scholar] [CrossRef]
  96. Medawar, E.; Huhn, S.; Villringer, A.; Veronica Witte, A. The Effects of Plant-Based Diets on the Body and the Brain: A Systematic Review. Transl. Psychiatry 2019, 9, 226. [Google Scholar] [CrossRef]
  97. Rajaram, S.; Jones, J.; Lee, G.J. Plant-Based Dietary Patterns, Plant Foods, and Age-Related Cognitive Decline. Adv. Nutr. 2019, 10, S422–S436. [Google Scholar] [CrossRef]
  98. Katonova, A.; Sheardova, K.; Amlerova, J.; Angelucci, F.; Hort, J. Effect of a Vegan Diet on Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 14924. [Google Scholar] [CrossRef] [PubMed]
  99. Devranis, P.; Vassilopoulou, Ε.; Tsironis, V.; Sotiriadis, P.M.; Chourdakis, M.; Aivaliotis, M.; Tsolaki, M. Mediterranean Diet, Ketogenic Diet or MIND Diet for Aging Populations with Cognitive Decline: A Systematic Review. Life 2023, 13, 173. [Google Scholar] [CrossRef]
  100. Rong, L.; Peng, Y.; Shen, Q.; Chen, K.; Fang, B.; Li, W. Effects of Ketogenic Diet on Cognitive Function of Patients with Alzheimer’s Disease: A Systematic Review and Meta-Analysis. J. Nutr. Health Aging 2024, 28, 100306. [Google Scholar] [CrossRef]
  101. Price, S.; Ruppar, T.M. Ketogenic Therapies in Parkinson’s Disease, Alzheimer’s Disease, and Mild Cognitive Impairment: An Integrative Review. Appl. Nurs. Res. 2023, 74, 151745. [Google Scholar] [CrossRef]
  102. Grochowska, K.; Przeliorz, A. The Effect of the Ketogenic Diet on the Therapy of Neurodegenerative Diseases and Its Impact on Improving Cognitive Functions. Dement Geriatr. Cogn. Dis. Extra 2022, 12, 100–106. [Google Scholar] [CrossRef]
  103. Sharifi, S.; Rostami, F.; Babaei Khorzoughi, K.; Rahmati, M. Effect of Time-Restricted Eating and Intermittent Fasting on Cognitive Function and Mental Health in Older Adults: A Systematic Review. Prev. Med. Rep. 2024, 42, 102757. [Google Scholar] [CrossRef] [PubMed]
  104. Elias, A.; Padinjakara, N.; Lautenschlager, N.T. Effects of Intermittent Fasting on Cognitive Health and Alzheimer’s Disease. Nutr. Rev. 2023, 81, 1225–1233. [Google Scholar] [CrossRef] [PubMed]
  105. Su, J.; Wang, Y.; Zhang, X.; Ma, M.; Xie, Z.; Pan, Q.; Ma, Z.; Peppelenbosch, M.P. Remodeling of the Gut Microbiome during Ramadan-Associated Intermittent Fasting. Am. J. Clin. Nutr. 2021, 113, 1332–1342. [Google Scholar] [CrossRef] [PubMed]
  106. Elizabeth, L.; Machado, P.; Zinöcker, M.; Baker, P.; Lawrence, M. Ultra-Processed Foods and Health Outcomes: A Narrative Review. Nutrients 2020, 12, 1955. [Google Scholar] [CrossRef]
  107. Martínez Leo, E.E.; Segura Campos, M.R. Effect of ultra-processed diet on gut microbiota and thus its role in neurodegenerative diseases. Nutrition 2020, 71, 110609. [Google Scholar] [CrossRef] [PubMed]
  108. Song, Z.; Song, R.; Liu, Y.; Wu, Z.; Zhang, X. Effects of Ultra-Processed Foods on the Microbiota-Gut-Brain Axis: The Bread-and-Butter Issue. Food Res. Int. 2023, 167, 112730. [Google Scholar] [CrossRef]
  109. Wang, Y.; Fan, D.; Zhang, Y.; Wang, J.; Dong, L.; Hu, Y.; Wang, S. Long-Term Exposure to Advanced Lipid Peroxidation End Products Impairs Cognitive Function through Microbiota-Gut-Brain Axis. Food Chem. 2024, 461, 140864. [Google Scholar] [CrossRef]
  110. R Cardoso, B.; Machado, P.; Steele, E.M. Association between Ultra-Processed Food Consumption and Cognitive Performance in US Older Adults: A Cross-Sectional Analysis of the NHANES 2011–2014. Eur. J. Nutr. 2022, 61, 3975–3985. [Google Scholar] [CrossRef]
  111. Pourmotabbed, A.; Talebi, S.; Mehrabani, S.; Babaei, A.; Khosroshahi, R.A.; Bagheri, R.; Wong, A.; Ghoreishy, S.M.; Amirian, P.; Zarpoosh, M.; et al. The Association of Ultra-Processed Food Intake with Neurodegenerative Disorders: A Systematic Review and Dose-Response Meta-Analysis of Large-Scale Cohorts. Nutr. Neurosci. 2024, 1–14. [Google Scholar] [CrossRef]
  112. Claudino, P.A.; Bueno, N.B.; Piloneto, S.; Halaiko, D.; Azevedo de Sousa, L.P.; Barroso Jara Maia, C.H.; Netto, B.D.M. Consumption of Ultra-Processed Foods and Risk for Alzheimer’s Disease: A Systematic Review. Front. Nutr. 2023, 10, 1288749. [Google Scholar] [CrossRef]
  113. Fan, H.; Yuan, M.; Wang, S.; Yang, X.; Shu, L.; Pu, Y.; Zou, Q.; Zhang, X.; Wang, C.; Cai, Z. Dietary Salt Promotes Cognitive Impairment through Repression of SIRT3/PINK1-Mediated Mitophagy and Fission. Mol. Cell. Biochem. 2024. [Google Scholar] [CrossRef] [PubMed]
  114. D’Cunha, N.M.; Sergi, D.; Lane, M.M.; Naumovski, N.; Gamage, E.; Rajendran, A.; Kouvari, M.; Gauci, S.; Dissanayka, T.; Marx, W.; et al. The Effects of Dietary Advanced Glycation End-Products on Neurocognitive and Mental Disorders. Nutrients 2022, 14, 2421. [Google Scholar] [CrossRef] [PubMed]
  115. Bhave, V.M.; Oladele, C.R.; Ament, Z.; Kijpaisalratana, N.; Jones, A.C.; Couch, C.A.; Patki, A.; Garcia Guarniz, A.-L.; Bennett, A.; Crowe, M.; et al. Associations Between Ultra-Processed Food Consumption and Adverse Brain Health Outcomes. Neurology 2024, 102, e209432. [Google Scholar] [CrossRef]
  116. Li, J.-M.; Yu, R.; Zhang, L.-P.; Wen, S.-Y.; Wang, S.-J.; Zhang, X.-Y.; Xu, Q.; Kong, L.-D. Dietary Fructose-Induced Gut Dysbiosis Promotes Mouse Hippocampal Neuroinflammation: A Benefit of Short-Chain Fatty Acids. Microbiome 2019, 7, 98. [Google Scholar] [CrossRef]
  117. Ayten, Ş.; Bilici, S. Modulation of Gut Microbiota Through Dietary Intervention in Neuroinflammation and Alzheimer’s and Parkinson’s Diseases. Curr. Nutr. Rep. 2024, 13, 82–96. [Google Scholar] [CrossRef]
  118. Magnusson, K.R.; Hauck, L.; Jeffrey, B.M.; Elias, V.; Humphrey, A.; Nath, R.; Perrone, A.; Bermudez, L.E. Relationships between Diet-Related Changes in the Gut Microbiome and Cognitive Flexibility. Neuroscience 2015, 300, 128–140. [Google Scholar] [CrossRef]
  119. Dhana, K.; Franco, O.H.; Ritz, E.M.; Ford, C.N.; Desai, P.; Krueger, K.R.; Holland, T.M.; Dhana, A.; Liu, X.; Aggarwal, N.T.; et al. Healthy Lifestyle and Life Expectancy with and without Alzheimer’s Dementia: Population Based Cohort Study. BMJ 2022, 377, e068390. [Google Scholar] [CrossRef]
  120. Dhana, K.; Evans, D.A.; Rajan, K.B.; Bennett, D.A.; Morris, M.C. Healthy Lifestyle and the Risk of Alzheimer Dementia: Findings from 2 Longitudinal Studies. Neurology 2020, 95, e374–e383. [Google Scholar] [CrossRef] [PubMed]
  121. Ma, F.; Wu, T.; Zhao, J.; Han, F.; Marseglia, A.; Liu, H.; Huang, G. Effects of 6-Month Folic Acid Supplementation on Cognitive Function and Blood Biomarkers in Mild Cognitive Impairment: A Randomized Controlled Trial in China. J. Gerontol.—Ser. A Biol. Sci. Med. Sci. 2016, 71, 1376–1383. [Google Scholar] [CrossRef]
  122. Ma, F.; Li, Q.; Zhou, X.; Zhao, J.; Song, A.; Li, W.; Liu, H.; Xu, W.; Huang, G. Effects of Folic Acid Supplementation on Cognitive Function and Aβ-Related Biomarkers in Mild Cognitive Impairment: A Randomized Controlled Trial. Eur. J. Nutr. 2019, 58, 345–356. [Google Scholar] [CrossRef]
  123. Malouf, R.; Evans, J.G. Folic Acid with or without Vitamin B12 for the Prevention and Treatment of Healthy Elderly and Demented People. Cochrane Database Syst. Rev. 2008, 2008, CD004514. [Google Scholar] [CrossRef] [PubMed]
  124. Ma, F.; Zhou, X.; Li, Q.; Zhao, J.; Song, A.; An, P.; Du, Y.; Xu, W.; Huang, G. Effects of Folic Acid and Vitamin B12, Alone and in Combination on Cognitive Function and Inflammatory Factors in the Elderly with Mild Cognitive Impairment: A Single-Blind Experimental Design. Curr. Alzheimer Res. 2019, 16, 622–632. [Google Scholar] [CrossRef] [PubMed]
  125. Chen, H.; Liu, S.; Ge, B.; Zhou, D.; Li, M.; Li, W.; Ma, F.; Liu, Z.; Ji, Y.; Huang, G. Effects of Folic Acid and Vitamin B12 Supplementation on Cognitive Impairment and Inflammation in Patients with Alzheimer’s Disease: A Randomized, Single-Blinded, Placebo-Controlled Trial. J. Prev. Alzheimer’s Dis. 2021, 8, 249–256. [Google Scholar] [CrossRef] [PubMed]
  126. Sun, Y.; Lu, C.J.; Chien, K.L.; Chen, S.T.; Chen, R.C. Efficacy of Multivitamin Supplementation Containing Vitamins B6 and B12 and Folic Acid as Adjunctive Treatment with a Cholinesterase Inhibitor in Alzheimer’s Disease: A 26-Week, Randomized, Double-Blind, Placebo-Controlled Study in Taiwanese Patients. Clin. Ther. 2007, 29, 2204–2214. [Google Scholar] [CrossRef]
  127. Li, M.; Li, W.; Gao, Y.; Chen, Y.; Bai, D.; Weng, J.; Du, Y.; Ma, F.; Wang, X.; Liu, H.; et al. Effect of Folic Acid Combined with Docosahexaenoic Acid Intervention on Mild Cognitive Impairment in Elderly: A Randomized Double-Blind, Placebo-Controlled Trial. Eur. J. Nutr. 2021, 60, 1795–1808. [Google Scholar] [CrossRef]
  128. Zhang, Y.P.; Miao, R.; Li, Q.; Wu, T.; Ma, F. Effects of DHA Supplementation on Hippocampal Volume and Cognitive Function in Older Adults with Mild Cognitive Impairment: A 12-Month Randomized, Double-Blind, Placebo-Controlled Trial. J. Alzheimer’s Dis. 2017, 55, 497–507. [Google Scholar] [CrossRef]
  129. Zhang, Y.P.; Lou, Y.; Hu, J.; Miao, R.; Ma, F. DHA Supplementation Improves Cognitive Function via Enhancing Aβ-Mediated Autophagy in Chinese Elderly with Mild Cognitive Impairment: A Randomised Placebo-Controlled Trial. J. Neurol. Neurosurg. Psychiatry 2018, 89, 382–388. [Google Scholar] [CrossRef]
  130. Atmadja, T.; Kusharto, C.; Sinaga, T. Supplementation of Catfish (Clarias Gariepinus) Oil Enriched with Omega-3 Soft Capsule Improves Oxidative Stress and Cognitive Function in Elderly. J. Nutr. Sci. Vitaminol. 2020, 66, S47–S50. [Google Scholar] [CrossRef]
  131. Nolan, J.M.; Power, R.; Howard, A.N.; Bergin, P.; Roche, W.; Prado-Cabrero, A.; Pope, G.; Cooke, J.; Power, T.; Mulcahy, R. Supplementation with Carotenoids, Omega-3 Fatty Acids, and Vitamin E Has a Positive Effect on the Symptoms and Progression of Alzheimer’s Disease. J. Alzheimer’s Dis. 2022, 90, 233–249. [Google Scholar] [CrossRef]
  132. Patan, M.J.; Kennedy, D.O.; Husberg, C.; Hustvedt, S.O.; Calder, P.C.; Khan, J.; Forster, J.; Jackson, P.A. Supplementation with Oil Rich in Eicosapentaenoic Acid, but Not in Docosahexaenoic Acid, Improves Global Cognitive Function in Healthy, Young Adults: Results from Randomized Controlled Trials. Am. J. Clin. Nutr. 2021, 114, 914–924. [Google Scholar] [CrossRef]
  133. Djuricic, I.; Calder, P.C. Beneficial Outcomes of Omega-6 and Omega-3 Polyunsaturated Fatty Acids on Human Health: An Update for 2021. Nutrients 2021, 13, 2421. [Google Scholar] [CrossRef] [PubMed]
  134. Glück, T.; Alter, P. Marine Omega-3 Highly Unsaturated Fatty Acids: From Mechanisms to Clinical Implications in Heart Failure and Arrhythmias. Vasc. Pharmacol. 2016, 82, 11–19. [Google Scholar] [CrossRef] [PubMed]
  135. Poddar, J.; Pradhan, M.; Ganguly, G.; Chakrabarti, S. Biochemical Deficits and Cognitive Decline in Brain Aging: Intervention by Dietary Supplements. J. Chem. Neuroanat. 2019, 95, 70–80. [Google Scholar] [CrossRef] [PubMed]
  136. Power, R.; Nolan, J.M.; Prado-Cabrero, A.; Roche, W.; Coen, R.; Power, T.; Mulcahy, R. Omega-3 Fatty Acid, Carotenoid and Vitamin E Supplementation Improves Working Memory in Older Adults: A Randomised Clinical Trial. Clin. Nutr. 2022, 41, 405–414. [Google Scholar] [CrossRef]
  137. Danthiir, V.; Burns, N.R.; Nettelbeck, T.; Wilson, C.; Wittert, G. The Older People, Omega-3, and Cognitive Health (EPOCH) Trial Design and Methodology: A Randomised, Double-Blind, Controlled Trial Investigating the Effect of Long-Chain Omega-3 Fatty Acids on Cognitive Ageing and Wellbeing in Cognitively Healthy Older Adults. Nutr. J. 2011, 10, 117. [Google Scholar] [CrossRef]
  138. Chew, E.Y.; Clemons, T.E.; Agrón, E.; Launer, L.J.; Grodstein, F.; Bernstein, P.S. Effect of Omega-3 Fatty Acids, Lutein/Zeaxanthin, or Other Nutrient Supplementation on Cognitive Function: The AREDS2 Randomized Clinical Trial. JAMA 2015, 314, 791–801. [Google Scholar] [CrossRef]
  139. Giudici, K.V.; de Souto Barreto, P.; Beard, J.; Cantet, C.; Araujo de Carvalho, I.; Rolland, Y.; Vellas, B. Effect of Long-Term Omega-3 Supplementation and a Lifestyle Multidomain Intervention on Intrinsic Capacity among Community-Dwelling Older Adults: Secondary Analysis of a Randomized, Placebo-Controlled Trial (MAPT Study). Maturitas 2020, 141, 39–45. [Google Scholar] [CrossRef]
  140. Lin, P.Y.; Cheng, C.; Satyanarayanan, S.K.; Chiu, L.T.; Chien, Y.C.; Chuu, C.P.; Lan, T.H.; Su, K.P. Omega-3 Fatty Acids and Blood-Based Biomarkers in Alzheimer’s Disease and Mild Cognitive Impairment: A Randomized Placebo-Controlled Trial. Brain Behav. Immun. 2022, 99, 289–298. [Google Scholar] [CrossRef]
  141. Mengelberg, A.; Leathem, J.; Podd, J.; Hill, S.; Conlon, C. The Effects of Docosahexaenoic Acid Supplementation on Cognition and Well-Being in Mild Cognitive Impairment: A 12-Month Randomised Controlled Trial. Int. J. Geriatr. Psychiatry 2022, 37, 1–12. [Google Scholar] [CrossRef]
  142. 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]
  143. Ghaderi, A.; Rasouli-Azad, M.; Farhadi, M.H.; Mirhosseini, N.; Motmaen, M.; Pishyareh, E.; Omidi, A.; Asemi, Z. Exploring the Effects of Vitamin D Supplementation on Cognitive Functions and Mental Health Status in Subjects under Methadone Maintenance Treatment. J. Addict. Med. 2020, 14, 18–25. [Google Scholar] [CrossRef] [PubMed]
  144. Wang, W.; Li, Y.; Meng, X. Vitamin D and Neurodegenerative Diseases. Heliyon 2023, 9, e12877. [Google Scholar] [CrossRef] [PubMed]
  145. Maharjan, R.; Diaz Bustamante, L.; Ghattas, K.N.; Ilyas, S.; Al-Refai, R.; Khan, S. Role of Lifestyle in Neuroplasticity and Neurogenesis in an Aging Brain. Cureus 2020, 12, e10639. [Google Scholar] [CrossRef]
  146. Deluca, G.C.; Kimball, S.M.; Kolasinski, J.; Ramagopalan, S.V.; Ebers, G.C. Review: The Role of Vitamin D in Nervous System Health and Disease. Neuropathol. Appl. Neurobiol. 2013, 39, 458–484. [Google Scholar] [CrossRef] [PubMed]
  147. Yang, T.; Wang, H.; Xiong, Y.; Chen, C.; Duan, K.; Jia, J.; Ma, F. Vitamin D Supplementation Improves Cognitive Function through Reducing Oxidative Stress Regulated by Telomere Length in Older Adults with Mild Cognitive Impairment: A 12-Month Randomized Controlled Trial. J. Alzheimer’s Dis. 2020, 78, 1509–1518. [Google Scholar] [CrossRef]
  148. Klimova, B.; Dziuba, S.; Cierniak-Emerych, A. The Effect of Healthy Diet on Cognitive Performance Among Healthy Seniors—A Mini Review. Front. Hum. Neurosci. 2020, 14, 325. [Google Scholar] [CrossRef]
  149. Mccleery, J.; Abraham, R.P.; Denton, D.A.; Rutjes, A.W.S.; Chong, L.Y.; Al-Assaf, A.S.; Griffith, D.J.; Rafeeq, S.; Yaman, H.; Malik, M.A.; et al. Vitamin and Mineral Supplementation for Preventing Dementia or Delaying Cognitive Decline in People with Mild Cognitive Impairment. Cochrane Database Syst. Rev. 2018, 2018, CD011905. [Google Scholar] [CrossRef]
  150. Naomi, R.; Embong, H.; Othman, F.; Ghazi, H.F.; Maruthey, N.; Bahari, H. Probiotics for Alzheimer’s Disease: A Systematic Review. Nutrients 2022, 14, 20. [Google Scholar] [CrossRef]
  151. Tan, A.H.; Hor, J.W.; Chong, C.W.; Lim, S.Y. Probiotics for Parkinson’s Disease: Current Evidence and Future Directions. JGH Open 2021, 5, 414–419. [Google Scholar] [CrossRef]
  152. Liu, Q.; Xi, Y.; Wang, Q.; Liu, J.; Li, P.; Meng, X.; Liu, K.; Chen, W.; Liu, X.; Liu, Z. Mannan Oligosaccharide Attenuates Cognitive and Behavioral Disorders in the 5xFAD Alzheimer’s Disease Mouse Model via Regulating the Gut Microbiota-Brain Axis. Brain Behav. Immun. 2021, 95, 330–343. [Google Scholar] [CrossRef]
  153. Castelli, V.; d’Angelo, M.; Lombardi, F.; Alfonsetti, M.; Antonosante, A.; Catanesi, M.; Benedetti, E.; Palumbo, P.; Cifone, M.G.; Giordano, A.; et al. Effects of the Probiotic Formulation SLAB51 in in Vitro and in Vivo Parkinson’s Disease Models. Aging 2020, 12, 4641–4659. [Google Scholar] [CrossRef] [PubMed]
  154. Nimgampalle, M. Anti-Alzheimer Properties of Probiotic, Lactobacillus Plantarum MTCC 1325 in Alzheimer’s Disease Induced Albino Rats. J. Clin. Diagn. Res. 2017, 11, KC01–KC05. [Google Scholar] [CrossRef] [PubMed]
  155. Imdad, A.; Minkoff, N.Z.; Tanner-Smith, E.E.; Zackular, J.P.; Acra, S.; Nicholson, M.R. Fecal Microbiota Transplantation for the Treatment of Recurrent Clostridioides difficile (Clostridium difficile). Cochrane Database Syst. Rev. 2021, 2021, CD013871. [Google Scholar] [CrossRef]
  156. Socała, K.; Doboszewska, U.; Szopa, A.; Serefko, A.; Włodarczyk, M.; Zielińska, A.; Poleszak, E.; Fichna, J.; Wlaź, P. The Role of Microbiota-Gut-Brain Axis in Neuropsychiatric and Neurological Disorders. Pharmacol. Res. 2021, 172, 105840. [Google Scholar] [CrossRef]
  157. Warner, B.B. The Contribution of the Gut Microbiome to Neurodevelopment and Neuropsychiatric Disorders. Pediatr. Res. 2019, 85, 216–224. [Google Scholar] [CrossRef]
  158. Kesika, P.; Suganthy, N.; Sivamaruthi, B.S.; Chaiyasut, C. Role of Gut-Brain Axis, Gut Microbial Composition, and Probiotic Intervention in Alzheimer’s Disease. Life Sci. 2021, 264, 118627. [Google Scholar] [CrossRef]
  159. Bhatt, S.; Kanoujia, J.; Mohana Lakshmi, S.; Patil, C.R.; Gupta, G.; Chellappan, D.K.; Dua, K. Role of Brain-Gut-Microbiota Axis in Depression: Emerging Therapeutic Avenues. CNS Neurol. Disord. Drug Targets 2023, 22, 276–288. [Google Scholar] [CrossRef] [PubMed]
  160. Chen, C.-Y.; Wang, Y.-F.; Lei, L.; Zhang, Y. Impacts of Microbiota and Its Metabolites through Gut-Brain Axis on Pathophysiology of Major Depressive Disorder. Life Sci. 2024, 351, 122815. [Google Scholar] [CrossRef] [PubMed]
  161. Mossad, O.; Nent, E.; Woltemate, S.; Folschweiller, S.; Buescher, J.M.; Schnepf, D.; Erny, D.; Staeheli, P.; Bartos, M.; Szalay, A.; et al. Microbiota-Dependent Increase in δ-Valerobetaine Alters Neuronal Function and Is Responsible for Age-Related Cognitive Decline. Nat. Aging 2021, 1, 1127–1136. [Google Scholar] [CrossRef]
  162. Xiong, Y.; Pu, Y.; Li, L.; Su, Y.; Niu, J.; Xiao, Z. Gut Microbiota-derived Metabolite Trimethylamine N -oxide Aggravates Cognitive Dysfunction Induced by Femoral Fracture Operation in Mice. Kaohsiung J. Med. Sci. 2024, 40, 732–743. [Google Scholar] [CrossRef]
  163. Parletta, N.; Milte, C.M.; Meyer, B.J. Nutritional Modulation of Cognitive Function and Mental Health. J. Nutr. Biochem. 2013, 24, 725–743. [Google Scholar] [CrossRef] [PubMed]
  164. Bianchi, V.E.; Herrera, P.F.; Laura, R. Effect of Nutrition on Neurodegenerative Diseases. A Systematic Review. Nutr. Neurosci. 2021, 24, 810–834. [Google Scholar] [CrossRef] [PubMed]
  165. Freeman, L.R.; Haley-Zitlin, V.; Rosenberger, D.S.; Granholm, A.C. Damaging Effects of a High-Fat Diet to the Brain and Cognition: A Review of Proposed Mechanisms. Nutr. Neurosci. 2014, 17, 241–251. [Google Scholar] [CrossRef] [PubMed]
  166. Beilharz, J.E.; Maniam, J.; Morris, M.J. Diet-Induced Cognitive Deficits: The Role of Fat and Sugar, Potential Mechanisms and Nutritional Interventions. Nutrients 2015, 7, 6719–6738. [Google Scholar] [CrossRef]
  167. 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, 1040–1065. [Google Scholar] [CrossRef]
Figure 2. Factors affecting the microbiota composition.
Figure 2. Factors affecting the microbiota composition.
Nutrients 16 03570 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Młynarska, E.; Jakubowska, P.; Frąk, W.; Gajewska, A.; Sornowska, J.; Skwira, S.; Wasiak, J.; Rysz, J.; Franczyk, B. Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases. Nutrients 2024, 16, 3570. https://doi.org/10.3390/nu16203570

AMA Style

Młynarska E, Jakubowska P, Frąk W, Gajewska A, Sornowska J, Skwira S, Wasiak J, Rysz J, Franczyk B. Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases. Nutrients. 2024; 16(20):3570. https://doi.org/10.3390/nu16203570

Chicago/Turabian Style

Młynarska, Ewelina, Paulina Jakubowska, Weronika Frąk, Agata Gajewska, Joanna Sornowska, Sylwia Skwira, Jakub Wasiak, Jacek Rysz, and Beata Franczyk. 2024. "Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases" Nutrients 16, no. 20: 3570. https://doi.org/10.3390/nu16203570

APA Style

Młynarska, E., Jakubowska, P., Frąk, W., Gajewska, A., Sornowska, J., Skwira, S., Wasiak, J., Rysz, J., & Franczyk, B. (2024). Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases. Nutrients, 16(20), 3570. https://doi.org/10.3390/nu16203570

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