Next Article in Journal
White Matter Magnetic Resonance Diffusion Measures in Multiple Sclerosis with Overactive Bladder
Next Article in Special Issue
The Cerebrovascular Side of Plasticity: Microvascular Architecture across Health and Neurodegenerative and Vascular Diseases
Previous Article in Journal
Reactive Astrocytosis—A Potential Contributor to Increased Suicide in Long COVID-19 Patients?
Previous Article in Special Issue
Exploring the Role of Neuroplasticity in Development, Aging, and Neurodegeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Physical Exercise and Mechanism Related to Alzheimer’s Disease: Is Gut–Brain Axis Involved?

by
Javier Sanchez-Martinez
1,
Patricio Solis-Urra
1,2,*,
Jorge Olivares-Arancibia
3 and
Julio Plaza-Diaz
4,5,6,*
1
Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, 18071 Granada, Spain
2
Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 8370134, Chile
3
AFySE Group, Research in Physical Activity and School Health, School of Physical Education, Faculty of Education, Universidad de Las Américas, Santiago 7500975, Chile
4
Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, University of Granada, 18071 Granada, Spain
5
Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
6
Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
*
Authors to whom correspondence should be addressed.
Brain Sci. 2024, 14(10), 974; https://doi.org/10.3390/brainsci14100974
Submission received: 31 August 2024 / Revised: 23 September 2024 / Accepted: 25 September 2024 / Published: 27 September 2024
(This article belongs to the Special Issue Neuroregenerative Plasticity in Health and Disease)

Abstract

:
Background: Alzheimer’s disease is a progressive neurodegenerative disease characterized by structural changes in the brain, including hippocampal atrophy, cortical thinning, amyloid plaques, and tau tangles. Due to the aging of the global population, the burden of Alzheimer’s disease is expected to increase, making the exploration of non-pharmacological interventions, such as physical exercise, an urgent priority. Results: There is emerging evidence that regular physical exercise may mitigate the structural and functional declines associated with Alzheimer’s disease. The underlying mechanisms, however, remain poorly understood. Gut–brain axis research is a promising area for further investigation. This system involves bidirectional communication between the gut microbiome and the brain. According to recent studies, the gut microbiome may influence brain health through modulating neuroinflammation, producing neuroactive compounds, and altering metabolic processes. Exercise has been shown to alter the composition of the gut microbiome, potentially impacting brain structure and function. In this review, we aim to synthesize current research on the relationship between physical exercise, structural brain changes in Alzheimer’s disease, and the gut–brain axis. Conclusions: In this study, we will investigate whether changes in the gut microbiome induced by physical exercise can mediate its neuroprotective effects, offering new insights into the prevention and treatment of Alzheimer’s disease. By integrating findings from neuroimaging studies, clinical trials, and microbiome research, this review will highlight potential mechanisms. It will also identify key gaps in the literature. This will pave the way for future research directions.

1. Structural Correlates of Alzheimer’s Disease

Alzheimer’s disease is a progressive neurodegenerative disorder that is the most common cause of dementia in older adults, affecting millions of people worldwide [1,2]. Characterized by the gradual decline in cognitive abilities, memory loss, and behavioral changes, Alzheimer’s disease profoundly impacts both patients and their families [1,2]. The disease progresses from mild cognitive impairment to severe dementia, eventually leading to the loss of basic bodily functions. Various hypotheses have been proposed to explain the onset and progression of Alzheimer’s disease. These include amyloid, cholinergic, inflammatory, tau protein, metal ions, oxidative stress, glutamate excitotoxicity, abnormal autophagy, and the microbiota–gut–brain axis [3]. Among these, the amyloid hypothesis has been particularly influential, suggesting that the accumulation of amyloid-beta (Aβ) peptides in the brain plays a central role in triggering the pathological changes associated with Alzheimer’s disease [1,4].
The brain undergoes profound structural changes driven by several mechanisms, such as neuron cell death, dendritic degeneration, metabolic slowing, and microglial activation, among others [5,6]. These mechanisms are closely related to the disorder’s progressive cognitive decline. One of the most notable changes is the accumulation of Aβ plaques, which are the extracellular deposits of misfolded Aβ peptides [7]. These plaques primarily form in the hippocampus and cortex (regions critical for memory and cognitive function) [8], disrupting neural communication and contributing to synaptic loss. Alongside amyloid plaques, neurofibrillary tangles made of hyperphosphorylated tau protein develop inside neurons [7]. These tangles disrupt the neuronal cytoskeleton, impairing intracellular transport and leading to neuronal death.
As the disease progresses, widespread neuronal loss and brain atrophy become evident, especially in the hippocampus and cortical areas [4]. This neuronal loss is accompanied by a significant shrinkage of the brain, which can be observed as a reduction in brain volume and enlargement of the ventricles on neuroimaging. Even before significant neuronal death, synaptic dysfunction occurs, characterized by reduced synaptic density and impaired neurotransmission, which are major contributors to cognitive decline [9]. Chronic neuroinflammation also plays a key role, with activated microglia and astrocytes surrounding amyloid plaques and neurofibrillary tangles [10]. While these immune cells initially attempt to clear Aβ and damaged neurons, their prolonged activation exacerbates neuronal damage and promotes further disease progression. Additionally, Alzheimer’s disease involves vascular changes, such as cerebral amyloid angiopathy, where Aβ deposits in the walls of cerebral blood vessels weaken them, increasing the risk of microbleeds and reducing the brain’s ability to clear toxins. These structural changes collectively lead to the hallmark symptoms of Alzheimer’s disease, including memory loss, confusion, language difficulties, and impaired reasoning, driving the relentless progression of the disease [4].
In Alzheimer’s disease, brain atrophy follows a characteristic pattern [1,11]. Initially, atrophy begins in the hippocampus and other structures within the medial temporal lobe (i.e., the entorhinal cortex or part of the parahippocampal gyrus). These regions are crucial for memory formation, which correlates with the early symptoms of Alzheimer’s, such as memory loss. As the disease progresses into mild to moderate stages, the atrophy spreads to the inferolateral temporal lobes, indicating that more extensive cortical regions, especially those involved in complex cognitive functions, start to deteriorate [12]. With the further advancement of the disease into moderate to severe stages, atrophy continues to affect the medial parietal lobes, areas associated with visuospatial processing and linked to the disorientation observed in more advanced Alzheimer’s patients. Finally, in the advanced stages, the frontal lobes are impacted [11]. This region governs executive functions such as decision making, personality, and behavior, leading to the severe cognitive and behavioral symptoms characteristic of late-stage Alzheimer’s disease.
Structural gray matter correlates of Alzheimer’s disease have been identified, with atrophy in key regions, particularly the medial temporal lobe, being a primary marker [11]. This atrophy includes changes in both the volume and cortical thickness, leading to the identification of a specific cortical signature of Alzheimer’s disease [13]. This signature refers to a distinct pattern of cortical thinning that can serve as a biomarker for the disease, distinguishing between cognitively unimpaired individuals and those who either have or are at risk of developing Alzheimer’s disease [13]. Numerous studies have highlighted the critical characteristics of these regions, demonstrating their significant biological implications for the development of cognitive impairment and Alzheimer’s disease [14,15]. Understanding these structural changes is crucial for advancing early diagnosis and potentially intervening before significant cognitive decline occurs.
This review aims to explore the interactions between the mechanisms implicated in the pathogenesis of Alzheimer’s disease and their associations with various microbiome compositions, with a particular focus on the role of physical exercise. We will examine how different microbiome changes are related to the mechanism of Alzheimer’s disease through these critical brain regions and hypothesize how physical exercise could modulate these interactions. Understanding how physical activity impacts the microbiome–brain axis is central to this review and could provide valuable insights into new therapeutic strategies for addressing the complex brain changes associated with Alzheimer’s disease.

2. Physical Exercise on Structural Correlates of Alzheimer’s Disease

Physical exercise promotes cognition and improves the symptoms of mood disorders and psychological disorders. Aerobic exercise training has been found to improve cognitive function, as well as the symptoms of depression and schizophrenia [16,17,18]. It has been demonstrated that regular aerobic exercise prevents age-related global brain atrophy and increases the volume of the left superior temporal lobe and frontal lobes, which are essential for cognition, attention, and memory [19]. Similarly, aerobic exercise has been shown to improve (in older adults aged 60–79 years) functional activation in the brain, which enhances efficiency when performing tasks, as well as regulating behavior and mood [20].
In animal models of Alzheimer’s disease, such as APP/PS1 transgenic mice, exercise interventions have been shown to impact the brain regions associated with the disease. For instance, 3 months of voluntary running led to increased volumes in the hippocampal subfields, specifically the dentate gyrus and CA1, compared to sedentary APP/PS1 mice [21]. Similarly, a 12-week treadmill exercise regimen enhanced synapse formation and increased the length and thickness of postsynaptic densities in the hippocampal CA1 region [22]. Furthermore, 10 weeks of treadmill training resulted in increased dendritic arborization in the CA1, CA3, and amygdalar basolateral neurons [23]. However, another study found no effect on medial prefrontal cortex volume after 4 months of running exercise. Despite this, the running exercise prevented the loss of dendritic spines and neurons [24]. These findings support the assumption that exercise may protect against degeneration in Alzheimer’s disease-related brain regions in this animal model.
In human studies, the effects of exercise on Alzheimer’s disease-related brain structures have been studied in older adults, but the findings remain inconclusive, and randomized controlled trials are limited. In healthy older adults, aerobic and coordination training have been associated with increased hippocampal volume [25,26], while resistance training showed no effect [27]. Aerobic exercise has been linked to increased gray and white matter volumes [28], insula cortical thickness [29], and putamen volume [30]. However, resistance training did not yield similar results for cortical gray matter and cortical white matter [27]. In older adults with mild cognitive impairment or at risk of dementia or Alzheimer’s disease, aerobic and multicomponent exercises have been found to increase hippocampal volume [31,32], while a 2-year home-based physical activity program showed no effect [33]. Additionally, high-intensity progressive resistance training protected against hippocampal atrophy one year after a 6-month intervention (three sets of eight repetitions, at 80% of the peak capacity) [34]. While evidence suggests exercise benefits in these populations, the findings are still insufficient for strong conclusions.
Few randomized controlled trials have examined the effects of exercise on brain structures in older adults with Alzheimer’s disease. Frederiksen et al. (2018) evaluated a 16-week aerobic exercise intervention (60 min, three times per week, moderate-to-high intensity) in older adults with mild to moderate Alzheimer’s disease. They found no effects on total hippocampal and subfield volumes, parahippocampal volume, basal ganglia (caudate and putamen) volumes, or regional cortical thickness [35]. However, a positive association was observed between exercise load and changes in hippocampal volume and frontal cortical thickness [35]. Similarly, Morris et al. (2017) assessed a 26-week vigorous aerobic exercise intervention (150 min, three to five times per week, 40–55% to 60–75% of heart rate reserve) in Alzheimer’s disease participants, focusing on hippocampal and gray matter volumes. While no direct effects were found, increased cardiorespiratory fitness was positively associated with bilateral hippocampal volume changes, particularly in the exercise group [36].
These findings indicate limited evidence to draw definitive conclusions about the effects of exercise on brain structures in older adults with Alzheimer’s disease. However, the positive associations between exercise load, cardiorespiratory fitness, and hippocampal volume suggest that further research is needed in this population to clarify these relationships. Although exercise has shown favorable effects on Alzheimer’s disease-related brain structures in healthy older adults and those at risk, research on its impact in patients with Alzheimer’s disease remains limited. Recent meta-analytical evidence indicates that regular exercise or physical activity improves hippocampal volume in humans, preventing volumetric declines over time [37].
Future studies should explore exercise’s effects on brain structure in Alzheimer’s disease patients, recognizing that many older adults with cognitive impairment and Alzheimer’s are capable of and interested in physical activity, which may benefit their brain health [38].

3. Gut–Brain Axis in Alzheimer’s Disease

A complex and intricate ecological network, the human microbiome comprises a variety of microorganisms (bacteria, viruses, fungi, and protozoa) that have colonized numerous anatomical locations within the human body, including the skin, oral cavity, vagina, and gastrointestinal tract [39,40]. The microbiome inhabits the human body and interacts with it. Depending on the nature of the interaction, it may be mutualistic, commensalistic, or pathogenic [41,42,43,44,45,46,47].
Selective serotonin uptake inhibitors improve gastrointestinal function in individuals with neural disorders [48]. Observations such as these have sparked curiosity among researchers, leading to an increase in research into the connection between the gastrointestinal tract and the brain [49,50,51,52,53].
The gut and the brain are intimately linked during fetal development when the central nervous system and the enteric nervous system are derived from the same tissues [49]. Gut–brain communication is bidirectional and mediated by the autonomic nervous system; efferent and afferent signals through the vagus nerve; neuroendocrine signaling through the hypothalamic–pituitary–adrenal axis; and serotonin regulation [54,55,56,57]. Specific microbiomes can affect other communication channels in the gut–brain axis, such as gamma-aminobutyric acid-glutamate and catecholamines, metabolites of microbial origin, and even hormones [58,59].
Several studies have demonstrated that Alzheimer’s disease participants have different microbiome compositions than cognitively healthy individuals [60,61,62]. There was a decrease in Bacillota, an increase in Bacteroidota, and a decrease in Bifidobacterium abundance in the microbiome of patients with Alzheimer’s disease. Positive correlations were also observed between differentially abundant genera and the biomarkers of Alzheimer’s disease in cerebrospinal fluid according to Vogt et al. [60]. According to another study, bacterial taxa in 43 patients with Alzheimer’s disease differed at taxonomic levels from those in controls, including Actinobacteriota, Bacteroides, Ruminococcus, Selenomonadales, and Lachnospiraceae [62]. In another study, as a baseline comparison, subjects with normal versus impaired cognition do not exhibit a significant difference in microbiome diversity. However, subjects with mild cognitive impairment display a number of unique microbial signatures. There was a positive correlation between Pseudomonadota and Aβ-42: Aβ-40 ratio and a negative correlation between fecal propionate and butyrate and Aβ-42 in subjects with mild cognitive impairment [61]. As compared to both healthy controls and patients without brain amyloidosis, patients with cognitive impairment had lower abundances of Eubacterium rectale and higher abundances of Escherichia/Shigella. The abundance of Escherichia/Shigella, an inflammatory bacteria taxon, was positively correlated with the presence of pro-inflammatory cytokines, such as interleukin (IL)-1β, whereas Eubacterium rectale, an anti-inflammatory bacteria taxon, was negatively correlated with the presence of those pro-inflammatory cytokines [63]. A diagram of the gut microbiome in Alzheimer’s disease can be seen in Figure 1.

4. Gut–Brain Axis and Exercise in Alzheimer’s Disease

Studies have shown that physical exercise and gut–brain axis play a significant role in the treatment and prevention of Alzheimer’s disease in both humans and animals. Below is a summary of these studies.

4.1. Animal Studies of Alzheimer’s Disease, Exercise, and the Gut–Brain Axis

In rats with a disrupted microbiome, performance on two hippocampal neurogenesis-dependent tasks was impaired. These tasks were the novelty-suppressed feeding test and the modified spontaneous location recognition task. Physical exercise mitigated the reduction in hippocampal neurogenesis caused by gut microbes in these rats [64]. A treadmill running protocol composed of 10 cycles of 6 min at high intensity, followed by two minutes at low intensity for two weeks resulted in increased antioxidant defenses and improved anti-inflammatory system performance in Alzheimer’s disease transgenic mice. As a result of these effects, pro-apoptotic proteins in intestinal lymphocytes appear to be reduced, and short-chain fatty acid (SCFA)-producing bacteria and Lactobacillus reuteri, a vitamin B12 producer, are increased, which improves cognitive function and slows the progression of biomarkers such as Aβ-42, total tau, and phosphorylated tau [65].
As a result of forced treadmill running, the gut microbiome changes in a symbiotic manner. This is due to an increase in Akkermansia muciniphila and a decrease in Bacteroides species. In addition, there is an increase in the blood–brain barrier-related protein expression and a reduction in the progression of Alzheimer’s disease-like cognitive impairments [66].
Through the production of palmitoleic acid, which is protective against inflammation and metabolic disorders, Dubosiella enrichment hinders the progression of Alzheimer’s disease in an animal model. The correlation between deoxycholate levels and cognitive scores in humans has also been supported by fecal deoxycholic acid-mediated interactions between the Alzheimer’s disease hub bacteria Erysipelatoclostridium and the disease’s occurrence [67]. In male APP/PS1 transgenic mice, exercise-induced changes in the microbiome do not significantly affect mitochondrial density and the AMPK/mTOR/S6 pathways related to protein synthesis [68]. Physical exercise has a positive effect on other organs, making Alzheimer’s disease a systemic disease [69,70].
Exercise reduced disintegrin and metalloproteinase domain-containing protein 10 and glial fibrillary acidic protein expression in the hippocampus of middle-aged APP/PS1 mice, but there were no significant alterations in circulating metabolites. Moreover, mice from the exercise group had markedly reduced abundances of the phyla Pseudomonadota and Tenericutes, genera Bacteroides and Faecalibacterium, and elevated abundances of the genus Allobaculum [71]. Exercise increased the alpha diversity index of cecal content in APP/PS1 mice, and trimethylamine N-oxide and exercise had a differential effect on gut microbiome profiles [72].

4.2. Human Studies of Alzheimer’s Disease, Exercise, and the Gut–Brain Axis

There is a link between Alzheimer’s disease and changes in the microbiome, and exercise appears to be beneficial in treating the disease. Moreover, physical activity is beneficial to the human intestinal microbiome. Therefore, this complex interaction may be advantageous for patients with Alzheimer’s disease as well as for the professional community and national budgets [73]. Next, we will present the results of a multimodal lifestyle intervention in humans that included exercise.
A 1:1 multicenter, phase 2 randomized controlled study on multimodal lifestyle intervention (including exercise, diet, and stress management, among others) was conducted on fifty-one patients with mild cognitive impairment or early dementia due to Alzheimer’s disease. The exercise component of the intervention included aerobic exercises (e.g., walking) for at least 30 min per day and strength training three times per week, either in person or via virtual consultations with an exercise physiologist. Based on the patient’s age and fitness level, an exercise prescription was developed for 20 weeks [74]. Blautia, which was found to increase in the intervention group during the intervention, has previously been associated with a lower risk of Alzheimer’s disease, possibly because it promotes gamma-aminobutyric acid production [75]. During the intervention, Eubacterium also increased in the intervention group, and prior studies have demonstrated that the Eubacterium genus (specifically Eubacterium fissicatena) protects against Alzheimer’s disease [76]. Furthermore, the relative abundance of the taxa associated with an increased risk of Alzheimer’s disease decreased in the intervention group, e.g., Prevotella and Turicibacter, the latter of which has been associated with relevant biological processes such as serotonin production [74]. Previously, Prevotella and Turicibacter rates increased with disease progression [77], but these rates decreased after the intervention. Despite these insights, research on the intricate effects of physical exercise on the gut–brain connection in humans remains relatively scarce. There is a need for human studies that evaluate only exercise as the main variable. The existing studies have primarily focused on isolated aspects rather than the comprehensive interactions between exercise, gut health, and cognitive function. The main studies discussed in this section are summarized in Table 1.
In the following section, we will present a detailed theoretical framework that can serve as a foundation for future research. This framework aims to integrate the existing knowledge and guide new studies in examining how physical exercise influences the gut–brain axis, with the goal of uncovering more nuanced and actionable insights.

5. Mechanisms Involved

Exercise’s effect on the gut microbiome is believed to contribute to the treatment or prevention of Alzheimer’s disease in several ways, including modulating neuroinflammation, amyloid metabolism, brain-derived neurotrophic factor release, and the transport of gut-derived metabolites across the blood–brain barrier.

5.1. Inflammation Reduction—Chronic Inflammation

A gut–brain axis pathway may be responsible for communication between the gut microbiome and the nervous system, which includes vagus nerve neuronal connections, the enteric nervous system, metabolism, and the immune system [78]. Exercise could modulate the gut microbiome through mechanisms that are closely related to an anti-inflammatory role, which results in a reduced level of inflammation in the gut [79]. Several mechanisms underlie exercise-induced benefits for cognition, including the gut microbiome regulation of neurogenesis and neuroinflammation [80,81]. Exercise has a neuroprotective effect, enhancing angiogenesis and neurogenesis, reducing inflammation, and reducing cerebrovascular risk factors [82,83,84]. Exercise programs that are conducted over a long period of time can reduce the risk factors for Alzheimer’s disease, improve blood flow, increase hippocampal volume, and improve neurogenesis in the brain, including swimming, walking, cycling, yoga, and bowling [85].

5.2. Amyloid Metabolism

There is evidence to support the belief that decreased Aβ clearance is an integral part of the pathomechanism of Alzheimer’s disease [86]. In the early stages of amyloid plaque accumulation, memory problems are the most common clinical symptom [85]. Additionally, Alzheimer’s disease may affect metabolism through the accumulation of pathological amyloid plaques in peripheral organs [87,88,89,90]. Based on these observations, Alzheimer’s disease is considered to be a complex systemic disease [70,91].
The gut microbiome contains several species of bacteria that can produce amyloid peptides, including Escherichia coli, Citrobacter, Mycobacteria, Klebsiella, Pseudomonas, Staphylococcus, Streptococcus, Streptomyces, Salmonella, and Bacillus [92,93]. There is a possibility that these peptides can be transmitted to the brain and accumulate there, leading to cognitive dysfunction [94]. The biomarkers of Alzheimer’s disease are also associated with a reduction in Bifidobacterium and Eubacterium rectale [95]. Despite the absence of any clear causal relationship between the gut microbiome and neurodegeneration, these results suggest that it may play an important role in the pathogenesis of dementia [96]. According to a study that compared the gut microbiome of amyloid-positive patients, amyloid-negative patients, and healthy controls, amyloid-positive patients had a low ratio of Eubacterium rectale and a high proportion of Escherichia/Shigella [97]. By regulating the immune system, differences in the gut microbiome influence amyloid accumulation in the brain [63]. While animal studies show that amyloid clearance is one of the possible mechanisms by which exercise positively impacts brain health [98,99,100], only a few human studies have been conducted and the results show no effect [101] or remain inconclusive [102].

5.3. Brain-Derived Neurotrophic Factor

Multiple studies have demonstrated that brain-derived neurotrophic factor is required for neurogenesis in the subventricular zone of the hippocampus [103,104,105]. Additionally, to modulate neurogenesis, brain-derived neurotrophic factor plays an important role in brain physiology, as well as in muscle tissue and vascular endothelial cells outside of neuronal tissue [103,104,105]. This neurotrophic factor play an important role in long-term potentiation and synaptic plasticity in the central nervous system, where it influences the morphology of mature neurons by promoting the growth of axons, the formation of dendritic arbors, and the pruning of dendrites [103,104,105,106].
Brain-derived neurotrophic factor expression was strongly negatively correlated with germ-free mice and at the phylum level of Bacillota, Bacteroidota, and Pseudomonadota. Bacillota also showed a negative correlation with neurogenesis. It is clear that the loss of healthy gut microbiome, such as in germ-free mice, has a detrimental effect on the brain, but it remains unclear why both the major phyla of the healthy microbiome (Bacillota and Bacteroidota) showed strong negative correlations [107]. Therefore, it is important to take precautions when interpreting the results at the phylum level. In addition, multiple topics concerning the role of the microbiome in neurogenesis and brain-derived neurotrophic factor expression were identified, including age, obesity, chronic stress, and antibiotic treatment. There has, however, been difficulty in making a definitive connection between a bacterial taxon and brain-derived neurotrophic factor expression or neurogenesis in adult animals, which sometimes leads to results that are inconclusive. For this purpose, studies with gnotobiotic mice that focus their attention on one taxonomic rank of bacteria would be helpful in determining the individual interactions between the microbiome, the gut, and the brain [106].
The presence of high levels of a brain-derived neurotrophic factor in the brain has been linked to improvements in memory and recollection, as well as the prevention of cognitive decline [108,109] and the brain-derived neurotrophic factor is one of the main candidates by which exercise exerts a positive effect on the brain [110,111]. In contrast, decreased levels of brain-derived neurotrophic factor are associated with poor memory function, neurodegeneration, and cognitive impairments associated with Alzheimer’s disease [112].

5.4. Transport of Gut-Derived Metabolites across the Blood–Brain Barrier

Previous studies have shown that the gut is crucial for the release of hormones, peptides, and microbial metabolites, including SCFAs, secondary bile acids, and products derived from tryptophan and polyphenols. Neuronal function and survival are significantly affected by these substances. Several of these compounds can cross the blood–brain barrier, including SCFAs, which use active membrane transporters in the endothelium to reach the central nervous system [113]. There are enteroendocrine cells within the enteric nervous system which are responsible for receiving signals from the gut microbiome directly. By secreting hormones, these cells can influence the function of brain cells across the blood–brain barrier [114].
Enteroendocrine cells are intricately connected with the vagus nerve, serving as a possible link between the gut microbiome and the brain [115]. It is possible to communicate bidirectionally between the gut and the brain via this direct connection, allowing for the exchange of signals and information that can influence a variety of physiological and neurological processes [116]. Due to the vagus nerve’s role in this communication pathway, it is important that exercise facilitates interactions between the gut microbiome and the brain, which may result in the vagus nerve mediating the gut–brain axis [117].
A number of immuno-signaling mediators, including cytokines, chemokines, and microbial-associated molecular patterns, contribute to the facilitation of communication between the gut microbiome and the brain [114]. This includes microbial constituents such as lipopolysaccharides and peptidoglycans [118]; microbial products such as enzymes, SCFAs, and neurotransmitters [119,120]; hormone release (glucocorticoids) [121]; and substrate metabolism (bile acids and tryptophan) [122,123].
Direct and indirect pathways are available for interaction between these mediators, allowing bidirectional signaling between the brain and the gut [124]. Using these signaling pathways, the gut microbiome can influence immune responses and inflammation in the brain, as well as modulate immune function in the gut through the brain [114,125].
Interestingly, the potential for physical exercise to modify blood–brain barrier permeability has been previously discussed [126], supported by a clear theoretical rationale that positions it as a significant factor in this phenomenon. This connection suggests that exercise may play a crucial role in maintaining the integrity of the blood–brain barrier, which is vital for neurological health. Figure 2 illustrates the mechanism described in the manuscript.

6. Future Directions

A multitude of mechanisms associated with Alzheimer’s disease can be modified by physical exercise. The gut–brain axis as a potential pathway to understand the effect of exercise on brain structure and the final behavioral consequences is promising [85]. The performance of cognitive functions, memories, and executive functions can be improved through a variety of activities, including swimming, walking, cycling, yoga, and bowling. Additionally, an hour-long resistance exercise program can help reduce the progression of Alzheimer’s by improving physical function long term [127,128].
Despite this, in some of the mechanisms proposed, the understanding is limited due to a variety of reasons. These include the lack of human studies, the lack of a targeted approach to the understanding of this dual mechanism, or the lack of diversity of interventions (in terms of type, duration, and population, among others). This document sheds light on the targeted approach to the gut–brain axis as a potential key mechanism in the understanding of the complexity of the disease with the final aim of improving the targeted approach to the disease and more effective disease-modifying therapies.

Author Contributions

Conceptualization, P.S.-U. and J.P.-D.; methodology, J.S.-M., P.S.-U., J.O.-A. and J.P.-D.; writing—original draft preparation, J.S.-M., P.S.-U., J.O.-A. and J.P.-D.; writing—review and editing, J.S.-M., P.S.-U., J.O.-A. and J.P.-D.; visualization, J.S.-M., P.S.-U. and J.P.-D.; supervision, P.S.-U. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Julio Plaza-Diaz is part of the “UGR Plan Propio de Investigación 2016” and the “Excellence actions: Unit of Excellence on Exercise and Health (UCEES), University of Granada”. Javier Sanchez-Martinez is supported by the National Agency for Research and Development (ANID)/Scholarship Program/DOCTORADO BECAS CHILE/2022–(Grant Nº72220164). Patricio Solis-Urra was supported by MICIU/AEI/10.13039/501100011033 and FEDER, UE (PID2022-137399OB-I00).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef] [PubMed]
  2. Livingston, G.; Huntley, J.; Liu, K.Y.; Costafreda, S.G.; Selbaek, G.; Alladi, S.; Ames, D.; Banerjee, S.; Burns, A.; Brayne, C.; et al. Dementia prevention, intervention, and care: 2024 report of the Lancet standing Commission. Lancet 2024, 404, 572–628. [Google Scholar] [CrossRef]
  3. Zhang, J.; Zhang, Y.; Wang, J.; Xia, Y.; Zhang, J.; Chen, L. Recent advances in Alzheimer’s disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduct. Target. Ther. 2024, 9, 211. [Google Scholar] [CrossRef] [PubMed]
  4. Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chetelat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef]
  5. Blinkouskaya, Y.; Cacoilo, A.; Gollamudi, T.; Jalalian, S.; Weickenmeier, J. Brain aging mechanisms with mechanical manifestations. Mech. Ageing Dev. 2021, 200, 111575. [Google Scholar] [CrossRef]
  6. Gao, C.; Jiang, J.; Tan, Y.; Chen, S. Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 359. [Google Scholar] [CrossRef] [PubMed]
  7. Karran, E.; De Strooper, B. The amyloid hypothesis in Alzheimer disease: New insights from new therapeutics. Nat. Rev. Drug Discov. 2022, 21, 306–318. [Google Scholar] [CrossRef] [PubMed]
  8. Rao, Y.L.; Ganaraja, B.; Murlimanju, B.V.; Joy, T.; Krishnamurthy, A.; Agrawal, A. Hippocampus and its involvement in Alzheimer’s disease: A review. 3 Biotech. 2022, 12, 55. [Google Scholar] [CrossRef]
  9. DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef]
  10. Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2021, 17, 157–172. [Google Scholar] [CrossRef]
  11. Smith, A.D. Imaging the progression of Alzheimer pathology through the brain. Proc. Natl. Acad. Sci. USA 2002, 99, 4135–4137. [Google Scholar] [CrossRef] [PubMed]
  12. van de Mortel, L.A.; Thomas, R.M.; van Wingen, G.A.; Alzheimer’s Disease Neuroimaging, I. Grey Matter Loss at Different Stages of Cognitive Decline: A Role for the Thalamus in Developing Alzheimer’s Disease. J. Alzheimers Dis. 2021, 83, 705–720. [Google Scholar] [CrossRef] [PubMed]
  13. Dickerson, B.C.; Bakkour, A.; Salat, D.H.; Feczko, E.; Pacheco, J.; Greve, D.N.; Grodstein, F.; Wright, C.I.; Blacker, D.; Rosas, H.D. The cortical signature of Alzheimer’s disease: Regionally specific cortical thinning relates to symptom severity in very mild to mild AD dementia and is detectable in asymptomatic amyloid-positive individuals. Cereb. Cortex 2009, 19, 497–510. [Google Scholar] [CrossRef] [PubMed]
  14. Pelkmans, W.; Vromen, E.M.; Dicks, E.; Scheltens, P.; Teunissen, C.E.; Barkhof, F.; van der Flier, W.M.; Tijms, B.M.; Alzheimer’s Disease Neuroimaging, I. Grey matter network markers identify individuals with prodromal Alzheimer’s disease who will show rapid clinical decline. Brain Commun. 2022, 4, fcac026. [Google Scholar] [CrossRef]
  15. Vogel, J.W.; Young, A.L.; Oxtoby, N.P.; Smith, R.; Ossenkoppele, R.; Strandberg, O.T.; La Joie, R.; Aksman, L.M.; Grothe, M.J.; Iturria-Medina, Y.; et al. Four distinct trajectories of tau deposition identified in Alzheimer’s disease. Nat. Med. 2021, 27, 871–881. [Google Scholar] [CrossRef]
  16. Wu, M.H.; Lee, C.P.; Hsu, S.C.; Chang, C.M.; Chen, C.Y. Effectiveness of high-intensity interval training on the mental and physical health of people with chronic schizophrenia. Neuropsychiatr. Dis. Treat. 2015, 11, 1255–1263. [Google Scholar] [CrossRef]
  17. McNeil, J.K.; LeBlanc, E.M.; Joyner, M. The effect of exercise on depressive symptoms in the moderately depressed elderly. Psychol. Aging 1991, 6, 487–488. [Google Scholar] [CrossRef]
  18. Quaney, B.M.; Boyd, L.A.; McDowd, J.M.; Zahner, L.H.; He, J.; Mayo, M.S.; Macko, R.F. Aerobic exercise improves cognition and motor function poststroke. Neurorehabil. Neural Repair. 2009, 23, 879–885. [Google Scholar] [CrossRef]
  19. Colcombe, S.J.; Erickson, K.I.; Scalf, P.E.; Kim, J.S.; Prakash, R.; McAuley, E.; Elavsky, S.; Marquez, D.X.; Hu, L.; Kramer, A.F. Aerobic exercise training increases brain volume in aging humans. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2006, 61, 1166–1170. [Google Scholar] [CrossRef]
  20. Gomez-Pinilla, F.; Hillman, C. The influence of exercise on cognitive abilities. Compr. Physiol. 2013, 3, 403–428. [Google Scholar] [CrossRef]
  21. Zhang, S.S.; Zhu, L.; Peng, Y.; Zhang, L.; Chao, F.L.; Jiang, L.; Xiao, Q.; Liang, X.; Tang, J.; Yang, H.; et al. Long-term running exercise improves cognitive function and promotes microglial glucose metabolism and morphological plasticity in the hippocampus of APP/PS1 mice. J. Neuroinflamm. 2022, 19, 34. [Google Scholar] [CrossRef] [PubMed]
  22. Li, B.; Mao, Q.; Zhao, N.; Xia, J.; Zhao, Y.; Xu, B. Treadmill exercise overcomes memory deficits related to synaptic plasticity through modulating ionic glutamate receptors. Behav. Brain Res. 2021, 414, 113502. [Google Scholar] [CrossRef] [PubMed]
  23. Lin, T.W.; Shih, Y.H.; Chen, S.J.; Lien, C.H.; Chang, C.Y.; Huang, T.Y.; Chen, S.H.; Jen, C.J.; Kuo, Y.M. Running exercise delays neurodegeneration in amygdala and hippocampus of Alzheimer’s disease (APP/PS1) transgenic mice. Neurobiol. Learn. Mem. 2015, 118, 189–197. [Google Scholar] [CrossRef] [PubMed]
  24. Zhu, L.; Fan, J.H.; Chao, F.L.; Zhou, C.N.; Jiang, L.; Zhang, Y.; Luo, Y.M.; Zhang, L.; Xiao, Q.; Yang, H.; et al. Running exercise protects spinophilin-immunoreactive puncta and neurons in the medial prefrontal cortex of APP/PS1 transgenic mice. J. Comp. Neurol. 2022, 530, 858–870. [Google Scholar] [CrossRef] [PubMed]
  25. Erickson, K.I.; Voss, M.W.; Prakash, R.S.; Basak, C.; Szabo, A.; Chaddock, L.; Kim, J.S.; Heo, S.; Alves, H.; White, S.M.; et al. Exercise training increases size of hippocampus and improves memory. Proc. Natl. Acad. Sci. USA 2011, 108, 3017–3022. [Google Scholar] [CrossRef]
  26. Niemann, C.; Godde, B.; Voelcker-Rehage, C. Not only cardiovascular, but also coordinative exercise increases hippocampal volume in older adults. Front. Aging Neurosci. 2014, 6, 170. [Google Scholar] [CrossRef]
  27. Best, J.R.; Chiu, B.K.; Liang Hsu, C.; Nagamatsu, L.S.; Liu-Ambrose, T. Long-Term Effects of Resistance Exercise Training on Cognition and Brain Volume in Older Women: Results from a Randomized Controlled Trial. J. Int. Neuropsychol. Soc. 2015, 21, 745–756. [Google Scholar] [CrossRef]
  28. Gow, A.J.; Bastin, M.E.; Munoz Maniega, S.; Valdes Hernandez, M.C.; Morris, Z.; Murray, C.; Royle, N.A.; Starr, J.M.; Deary, I.J.; Wardlaw, J.M. Neuroprotective lifestyles and the aging brain: Activity, atrophy, and white matter integrity. Neurology 2012, 79, 1802–1808. [Google Scholar] [CrossRef]
  29. Martin-Willett, R.; Ellingson, J.E.; Fries, J.; Helmuth, T.; Karoly, H.; Giordano, G.; Calhoun, V.D.; Bryan, A.D. Few Structural Brain Changes Associated with Moderate-Intensity Interval Training and Low-Intensity Continuous Training in a Randomized Trial of Fitness and Older Adults. J. Aging Phys. Act. 2021, 29, 505–515. [Google Scholar] [CrossRef]
  30. Nagamatsu, L.S.; Weinstein, A.M.; Erickson, K.I.; Fanning, J.; Awick, E.A.; Kramer, A.F.; McAuley, E. Exercise Mode Moderates the Relationship Between Mobility and Basal Ganglia Volume in Healthy Older Adults. J. Am. Geriatr. Soc. 2016, 64, 102–108. [Google Scholar] [CrossRef]
  31. ten Brinke, L.F.; Bolandzadeh, N.; Nagamatsu, L.S.; Hsu, C.L.; Davis, J.C.; Miran-Khan, K.; Liu-Ambrose, T. Aerobic exercise increases hippocampal volume in older women with probable mild cognitive impairment: A 6-month randomised controlled trial. Br. J. Sports Med. 2015, 49, 248–254. [Google Scholar] [CrossRef] [PubMed]
  32. Teixeira, C.V.L.; Ribeiro de Rezende, T.J.; Weiler, M.; Magalhaes, T.N.C.; Carletti-Cassani, A.; Silva, T.; Joaquim, H.P.G.; Talib, L.L.; Forlenza, O.V.; Franco, M.P.; et al. Cognitive and structural cerebral changes in amnestic mild cognitive impairment due to Alzheimer’s disease after multicomponent training. Alzheimers Dement. 2018, 4, 473–480. [Google Scholar] [CrossRef] [PubMed]
  33. Venkatraman, V.K.; Sanderson, A.; Cox, K.L.; Ellis, K.A.; Steward, C.; Phal, P.M.; Gorelik, A.; Sharman, M.J.; Villemagne, V.L.; Lai, M.; et al. Effect of a 24-month physical activity program on brain changes in older adults at risk of Alzheimer’s disease: The AIBL active trial. Neurobiol. Aging 2020, 89, 132–141. [Google Scholar] [CrossRef] [PubMed]
  34. Broadhouse, K.M.; Singh, M.F.; Suo, C.; Gates, N.; Wen, W.; Brodaty, H.; Jain, N.; Wilson, G.C.; Meiklejohn, J.; Singh, N.; et al. Hippocampal plasticity underpins long-term cognitive gains from resistance exercise in MCI. Neuroimage Clin. 2020, 25, 102182. [Google Scholar] [CrossRef]
  35. Frederiksen, K.S.; Larsen, C.T.; Hasselbalch, S.G.; Christensen, A.N.; Hogh, P.; Wermuth, L.; Andersen, B.B.; Siebner, H.R.; Garde, E. A 16-Week Aerobic Exercise Intervention Does Not Affect Hippocampal Volume and Cortical Thickness in Mild to Moderate Alzheimer’s Disease. Front. Aging Neurosci. 2018, 10, 293. [Google Scholar] [CrossRef]
  36. Morris, J.K.; Vidoni, E.D.; Johnson, D.K.; Van Sciver, A.; Mahnken, J.D.; Honea, R.A.; Wilkins, H.M.; Brooks, W.M.; Billinger, S.A.; Swerdlow, R.H.; et al. Aerobic exercise for Alzheimer’s disease: A randomized controlled pilot trial. PLoS ONE 2017, 12, e0170547. [Google Scholar] [CrossRef]
  37. Firth, J.; Stubbs, B.; Vancampfort, D.; Schuch, F.; Lagopoulos, J.; Rosenbaum, S.; Ward, P.B. Effect of aerobic exercise on hippocampal volume in humans: A systematic review and meta-analysis. Neuroimage 2018, 166, 230–238. [Google Scholar] [CrossRef]
  38. Bechard, L.E.; Beaton, D.; McGilton, K.S.; Tartaglia, M.C.; Black, S.E. Physical activity perceptions, experiences, and beliefs of older adults with mild cognitive impairment or Alzheimer’s disease and their care partners. Appl. Physiol. Nutr. Metab. 2020, 45, 1216–1224. [Google Scholar] [CrossRef] [PubMed]
  39. Feng, K.; Ren, F.; Xing, Z.; Zhao, Y.; Yang, C.; Liu, J.; Shang, Q.; Wang, X.; Wang, X. Microbiome and its implications in oncogenesis: A Mendelian randomization perspective. Am. J. Cancer Res. 2023, 13, 5785. [Google Scholar]
  40. Pastor-Villaescusa, B.; Plaza-Diaz, J.; Egea-Zorrilla, A.; Leis, R.; Bueno, G.; Hoyos, R.; Vazquez-Cobela, R.; Latorre, M.; Canete, M.D.; Caballero-Villarraso, J.; et al. Evaluation of the gut microbiota after metformin intervention in children with obesity: A metagenomic study of a randomized controlled trial. Biomed. Pharmacother. 2021, 134, 111117. [Google Scholar] [CrossRef]
  41. Dalton, A.; Mermier, C.; Zuhl, M. Exercise influence on the microbiome-gut-brain axis. Gut Microbes 2019, 10, 555–568. [Google Scholar] [CrossRef] [PubMed]
  42. Ogunrinola, G.A.; Oyewale, J.O.; Oshamika, O.O.; Olasehinde, G.I. The human microbiome and its impacts on health. Int. J. Microbiol. 2020, 2020, 8045646. [Google Scholar] [CrossRef]
  43. Plaza-Diaz, J.; Álvarez-Mercado, A.I. The interplay between microbiota and chemotherapy-derived metabolites in breast cancer. Metabolites 2023, 13, 703. [Google Scholar] [CrossRef]
  44. Plaza-Diaz, J. Nutrition, microbiota and noncommunicable diseases. Nutrients 2020, 12, 1971. [Google Scholar] [CrossRef] [PubMed]
  45. Plaza-Diaz, J.; Bernal, M.J.; Schutte, S.; Chenoll, E.; Genoves, S.; Codoner, F.M.; Gil, A.; Sanchez-Siles, L.M. Effects of Whole-Grain and Sugar Content in Infant Cereals on Gut Microbiota at Weaning: A Randomized Trial. Nutrients 2021, 13, 1496. [Google Scholar] [CrossRef]
  46. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Morales, J.; Martin-Masot, R.; Climent, E.; Silva, A.; Martinez-Blanch, J.F.; Enrique, M.; Tortajada, M.; Ramon, D.; et al. Innova 2020: A Follow-up Study of the Fecal Microbiota of Infants Using a Novel Infant Formula between 6 Months and 12 Months of Age. Int. J. Mol. Sci. 2023, 24, 7392. [Google Scholar] [CrossRef] [PubMed]
  47. Plaza-Diaz, J.; Gomez-Fernandez, A.; Chueca, N.; Torre-Aguilar, M.J.; Gil, A.; Perez-Navero, J.L.; Flores-Rojas, K.; Martin-Borreguero, P.; Solis-Urra, P.; Ruiz-Ojeda, F.J.; et al. Autism Spectrum Disorder (ASD) with and without Mental Regression is Associated with Changes in the Fecal Microbiota. Nutrients 2019, 11, 337. [Google Scholar] [CrossRef] [PubMed]
  48. Thorkelson, G.; Bielefeldt, K.; Szigethy, E. Empirically Supported Use of Psychiatric Medications in Adolescents and Adults with IBD. Inflamm. Bowel Dis. 2016, 22, 1509–1522. [Google Scholar] [CrossRef]
  49. Sharon, G.; Sampson, T.R.; Geschwind, D.H.; Mazmanian, S.K. The Central Nervous System and the Gut Microbiome. Cell 2016, 167, 915–932. [Google Scholar] [CrossRef]
  50. Foster, J.A.; Rinaman, L.; Cryan, J.F. Stress & the gut-brain axis: Regulation by the microbiome. Neurobiol. Stress 2017, 7, 124–136. [Google Scholar] [CrossRef]
  51. Burokas, A.; Arboleya, S.; Moloney, R.D.; Peterson, V.L.; Murphy, K.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice. Biol. Psychiatry 2017, 82, 472–487. [Google Scholar] [CrossRef]
  52. Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G. Irritable bowel syndrome: A microbiome-gut-brain axis disorder? World J. Gastroenterol. 2014, 20, 14105–14125. [Google Scholar] [CrossRef] [PubMed]
  53. Cryan, J.F.; O’Mahony, S.M. The microbiome-gut-brain axis: From bowel to behavior. Neurogastroenterol. Motil. 2011, 23, 187–192. [Google Scholar] [CrossRef] [PubMed]
  54. Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbe communication in health and disease. Front. Physiol. 2011, 2, 94. [Google Scholar] [CrossRef]
  55. Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef] [PubMed]
  56. Forsythe, P.; Kunze, W.; Bienenstock, J. Moody microbes or fecal phrenology: What do we know about the microbiota-gut-brain axis? BMC Med. 2016, 14, 58. [Google Scholar] [CrossRef]
  57. Brookes, S.J.; Spencer, N.J.; Costa, M.; Zagorodnyuk, V.P. Extrinsic primary afferent signalling in the gut. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 286–296. [Google Scholar] [CrossRef]
  58. Yunes, R.A.; Poluektova, E.U.; Dyachkova, M.S.; Klimina, K.M.; Kovtun, A.S.; Averina, O.V.; Orlova, V.S.; Danilenko, V.N. GABA production and structure of gadB/gadC genes in Lactobacillus and Bifidobacterium strains from human microbiota. Anaerobe 2016, 42, 197–204. [Google Scholar] [CrossRef]
  59. Asano, Y.; Hiramoto, T.; Nishino, R.; Aiba, Y.; Kimura, T.; Yoshihara, K.; Koga, Y.; Sudo, N. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G1288–G1295. [Google Scholar] [CrossRef]
  60. Vogt, N.M.; Kerby, R.L.; Dill-McFarland, K.A.; Harding, S.J.; Merluzzi, A.P.; Johnson, S.C.; Carlsson, C.M.; Asthana, S.; Zetterberg, H.; Blennow, K.; et al. Gut microbiome alterations in Alzheimer’s disease. Sci. Rep. 2017, 7, 13537. [Google Scholar] [CrossRef]
  61. Nagpal, R.; Neth, B.J.; Wang, S.; Craft, S.; Yadav, H. Modified Mediterranean-ketogenic diet modulates gut microbiome and short-chain fatty acids in association with Alzheimer’s disease markers in subjects with mild cognitive impairment. eBioMedicine 2019, 47, 529–542. [Google Scholar] [CrossRef] [PubMed]
  62. Zhuang, Z.Q.; Shen, L.L.; Li, W.W.; Fu, X.; Zeng, F.; Gui, L.; Lu, Y.; Cai, M.; Zhu, C.; Tan, Y.L.; et al. Gut Microbiota is Altered in Patients with Alzheimer’s Disease. J. Alzheimers Dis. 2018, 63, 1337–1346. [Google Scholar] [CrossRef]
  63. Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef]
  64. Nicolas, S.; Dohm-Hansen, S.; Lavelle, A.; Bastiaanssen, T.F.S.; English, J.A.; Cryan, J.F.; Nolan, Y.M. Exercise mitigates a gut microbiota-mediated reduction in adult hippocampal neurogenesis and associated behaviours in rats. Transl. Psychiatry 2024, 14, 195. [Google Scholar] [CrossRef] [PubMed]
  65. Abraham, D.; Feher, J.; Scuderi, G.L.; Szabo, D.; Dobolyi, A.; Cservenak, M.; Juhasz, J.; Ligeti, B.; Pongor, S.; Gomez-Cabrera, M.C. Exercise and probiotics attenuate the development of Alzheimer’s disease in transgenic mice: Role of microbiome. Exp. Gerontol. 2019, 115, 122–131. [Google Scholar] [CrossRef] [PubMed]
  66. Jin, Y.; Kim, T.; Kang, H. Forced treadmill running modifies gut microbiota with alleviations of cognitive impairment and Alzheimer’s disease pathology in 3xTg-AD mice. Physiol. Behav. 2023, 264, 114145. [Google Scholar] [CrossRef]
  67. Chen, Y.; Li, Y.; Fan, Y.; Chen, S.; Chen, L.; Chen, Y.; Chen, Y. Gut microbiota-driven metabolic alterations reveal gut-brain communication in Alzheimer’s disease model mice. Gut Microbes 2024, 16, 2302310. [Google Scholar] [CrossRef]
  68. Teglas, T.; Abraham, D.; Jokai, M.; Kondo, S.; Mohammadi, R.; Feher, J.; Szabo, D.; Wilhelm, M.; Radak, Z. Exercise combined with a probiotics treatment alters the microbiome, but moderately affects signalling pathways in the liver of male APP/PS1 transgenic mice. Biogerontology 2020, 21, 807–815. [Google Scholar] [CrossRef]
  69. Gonzalez-Dominguez, R.; Garcia-Barrera, T.; Vitorica, J.; Gomez-Ariza, J.L. Metabolomic investigation of systemic manifestations associated with Alzheimer’s disease in the APP/PS1 transgenic mouse model. Mol. Biosyst. 2015, 11, 2429–2440. [Google Scholar] [CrossRef]
  70. Wang, J.; Gu, B.J.; Masters, C.L.; Wang, Y.J. A systemic view of Alzheimer disease—insights from amyloid-beta metabolism beyond the brain. Nat. Rev. Neurol. 2017, 13, 612–623. [Google Scholar] [CrossRef]
  71. Wang, G.; Zhou, H.H.; Luo, L.; Qin, L.Q.; Yin, J.; Yu, Z.; Zhang, L.; Wan, Z. Voluntary wheel running is capable of improving cognitive function only in the young but not the middle-aged male APPSwe/PS1De9 mice. Neurochem. Int. 2021, 145, 105010. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, Y.; Wang, G.; Li, R.; Liu, R.; Yu, Z.; Zhang, Z.; Wan, Z. Trimethylamine N-oxide aggravated cognitive impairment from APP/PS1 mice and protective roles of voluntary exercise. Neurochem. Int. 2023, 162, 105459. [Google Scholar] [CrossRef] [PubMed]
  73. Schlegel, P.; Novotny, M.; Klimova, B.; Valis, M. “Muscle-Gut-Brain Axis”: Can Physical Activity Help Patients with Alzheimer’s Disease Due to Microbiome Modulation? J. Alzheimers Dis. 2019, 71, 861–878. [Google Scholar] [CrossRef]
  74. Ornish, D.; Madison, C.; Kivipelto, M.; Kemp, C.; McCulloch, C.E.; Galasko, D.; Artz, J.; Rentz, D.; Lin, J.; Norman, K.; et al. Effects of intensive lifestyle changes on the progression of mild cognitive impairment or early dementia due to Alzheimer’s disease: A randomized, controlled clinical trial. Alzheimers Res. Ther. 2024, 16, 122. [Google Scholar] [CrossRef]
  75. Zhuang, Z.; Yang, R.; Wang, W.; Qi, L.; Huang, T. Associations between gut microbiota and Alzheimer’s disease, major depressive disorder, and schizophrenia. J. Neuroinflamm. 2020, 17, 288. [Google Scholar] [CrossRef] [PubMed]
  76. Cammann, D.; Lu, Y.; Cummings, M.J.; Zhang, M.L.; Cue, J.M.; Do, J.; Ebersole, J.; Chen, X.; Oh, E.C.; Cummings, J.L.; et al. Genetic correlations between Alzheimer’s disease and gut microbiome genera. Sci. Rep. 2023, 13, 5258. [Google Scholar] [CrossRef]
  77. Borsom, E.M.; Conn, K.; Keefe, C.R.; Herman, C.; Orsini, G.M.; Hirsch, A.H.; Palma Avila, M.; Testo, G.; Jaramillo, S.A.; Bolyen, E.; et al. Predicting Neurodegenerative Disease Using Prepathology Gut Microbiota Composition: A Longitudinal Study in Mice Modeling Alzheimer’s Disease Pathologies. Microbiol. Spectr. 2023, 11, e0345822. [Google Scholar] [CrossRef]
  78. Westfall, S.; Lomis, N.; Kahouli, I.; Dia, S.Y.; Singh, S.P.; Prakash, S. Microbiome, probiotics and neurodegenerative diseases: Deciphering the gut brain axis. Cell. Mol. Life Sci. 2017, 74, 3769–3787. [Google Scholar] [CrossRef]
  79. Gubert, C.; Kong, G.; Renoir, T.; Hannan, A.J. Exercise, diet and stress as modulators of gut microbiota: Implications for neurodegenerative diseases. Neurobiol. Dis. 2020, 134, 104621. [Google Scholar] [CrossRef]
  80. Erny, D.; Hrabe de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef]
  81. Ogbonnaya, E.S.; Clarke, G.; Shanahan, F.; Dinan, T.G.; Cryan, J.F.; O’Leary, O.F. Adult Hippocampal Neurogenesis Is Regulated by the Microbiome. Biol. Psychiatry 2015, 78, e7–e9. [Google Scholar] [CrossRef] [PubMed]
  82. Boyle, C.P.; Raji, C.A.; Erickson, K.I.; Lopez, O.L.; Becker, J.T.; Gach, H.M.; Longstreth, W.T., Jr.; Teverovskiy, L.; Kuller, L.H.; Carmichael, O.T.; et al. Physical activity, body mass index, and brain atrophy in Alzheimer’s disease. Neurobiol. Aging 2015, 36 (Suppl. S1), S194–S202. [Google Scholar] [CrossRef] [PubMed]
  83. Braskie, M.N.; Boyle, C.P.; Rajagopalan, P.; Gutman, B.A.; Toga, A.W.; Raji, C.A.; Tracy, R.P.; Kuller, L.H.; Becker, J.T.; Lopez, O.L.; et al. Physical activity, inflammation, and volume of the aging brain. Neuroscience 2014, 273, 199–209. [Google Scholar] [CrossRef] [PubMed]
  84. De la Rosa, A.; Olaso-Gonzalez, G.; Arc-Chagnaud, C.; Millan, F.; Salvador-Pascual, A.; Garcia-Lucerga, C.; Blasco-Lafarga, C.; Garcia-Dominguez, E.; Carretero, A.; Correas, A.G.; et al. Physical exercise in the prevention and treatment of Alzheimer’s disease. J. Sport. Health Sci. 2020, 9, 394–404. [Google Scholar] [CrossRef]
  85. Meng, Q.; Lin, M.S.; Tzeng, I.S. Relationship Between Exercise and Alzheimer’s Disease: A Narrative Literature Review. Front. Neurosci. 2020, 14, 131. [Google Scholar] [CrossRef]
  86. Tavares, R.S.; Martins, S.; Almeida-Santos, T.; Sousa, A.P.; Ramalho-Santos, J.; da Cruz, E.S.O.A. Alzheimer’s disease-related amyloid-beta(1–42) peptide induces the loss of human sperm function. Cell Tissue Res. 2017, 369, 647–651. [Google Scholar] [CrossRef]
  87. Mattson, M.P. Pathways towards and away from Alzheimer’s disease. Nature 2004, 430, 631–639. [Google Scholar] [CrossRef] [PubMed]
  88. Kawarabayashi, T.; Younkin, L.H.; Saido, T.C.; Shoji, M.; Ashe, K.H.; Younkin, S.G. Age-dependent changes in brain, CSF, and plasma amyloid (beta) protein in the Tg2576 transgenic mouse model of Alzheimer’s disease. J. Neurosci. 2001, 21, 372–381. [Google Scholar] [CrossRef]
  89. Kheirbakhsh, R.; Haddadi, M.; Muhammadnejad, A.; Abdollahi, A.; Shahi, F.; Amanpour-Gharaei, B.; Abrahim-Habibi, A.; Barati, T.; Amanpour, S. Long-term behavioral, histological, biochemical and hematological evaluations of amyloid beta-induced Alzheimer’s disease in rat. Acta Neurobiol. Exp. 2018, 78, 51–59. [Google Scholar] [CrossRef]
  90. Arbor, S. Targeting amyloid precursor protein shuttling and processing—long before amyloid beta formation. Neural Regen. Res. 2017, 12, 207–209. [Google Scholar] [CrossRef]
  91. Morris, J.K.; Honea, R.A.; Vidoni, E.D.; Swerdlow, R.H.; Burns, J.M. Is Alzheimer’s disease a systemic disease? Biochim. Biophys. Acta 2014, 1842, 1340–1349. [Google Scholar] [CrossRef] [PubMed]
  92. Alkasir, R.; Li, J.; Li, X.; Jin, M.; Zhu, B. Human gut microbiota: The links with dementia development. Protein Cell 2017, 8, 90–102. [Google Scholar] [CrossRef] [PubMed]
  93. Degn, S.E.; Jensen, L.; Olszowski, T.; Jensenius, J.C.; Thiel, S. Co-complexes of MASP-1 and MASP-2 associated with the soluble pattern-recognition molecules drive lectin pathway activation in a manner inhibitable by MAp44. J. Immunol. 2013, 191, 1334–1345. [Google Scholar] [CrossRef] [PubMed]
  94. Friedland, R.P.; Chapman, M.R. The role of microbial amyloid in neurodegeneration. PLoS Pathog. 2017, 13, e1006654. [Google Scholar] [CrossRef]
  95. Welcome, M.O. Current Perspectives and Mechanisms of Relationship between Intestinal Microbiota Dysfunction and Dementia: A Review. Dement. Geriatr. Cogn. Dis. Extra 2018, 8, 360–381. [Google Scholar] [CrossRef]
  96. Chen, W.W.; Zhang, X.; Huang, W.J. Role of neuroinflammation in neurodegenerative diseases (Review). Mol. Med. Rep. 2016, 13, 3391–3396. [Google Scholar] [CrossRef] [PubMed]
  97. Kelaiditi, E.; Cesari, M.; Canevelli, M.; van Kan, G.A.; Ousset, P.J.; Gillette-Guyonnet, S.; Ritz, P.; Duveau, F.; Soto, M.E.; Provencher, V.; et al. Cognitive frailty: Rational and definition from an (I.A.N.A./I.A.G.G.) international consensus group. J. Nutr. Health Aging 2013, 17, 726–734. [Google Scholar] [CrossRef]
  98. Adlard, P.A.; Perreau, V.M.; Pop, V.; Cotman, C.W. Voluntary exercise decreases amyloid load in a transgenic model of Alzheimer’s disease. J. Neurosci. 2005, 25, 4217–4221. [Google Scholar] [CrossRef]
  99. Radak, Z.; Ihasz, F.; Koltai, E.; Goto, S.; Taylor, A.W.; Boldogh, I. The redox-associated adaptive response of brain to physical exercise. Free Radic. Res. 2014, 48, 84–92. [Google Scholar] [CrossRef]
  100. Khodadadi, D.; Gharakhanlou, R.; Naghdi, N.; Salimi, M.; Azimi, M.; Shahed, A.; Heysieattalab, S. Treadmill Exercise Ameliorates Spatial Learning and Memory Deficits Through Improving the Clearance of Peripheral and Central Amyloid-Beta Levels. Neurochem. Res. 2018, 43, 1561–1574. [Google Scholar] [CrossRef]
  101. Vidoni, E.D.; Morris, J.K.; Watts, A.; Perry, M.; Clutton, J.; Van Sciver, A.; Kamat, A.S.; Mahnken, J.; Hunt, S.L.; Townley, R.; et al. Effect of aerobic exercise on amyloid accumulation in preclinical Alzheimer’s: A 1-year randomized controlled trial. PLoS ONE 2021, 16, e0244893. [Google Scholar] [CrossRef] [PubMed]
  102. Rodriguez-Ayllon, M.; Solis-Urra, P.; Arroyo-Avila, C.; Alvarez-Ortega, M.; Molina-Garcia, P.; Molina-Hidalgo, C.; Gomez-Rio, M.; Brown, B.; Erickson, K.I.; Esteban-Cornejo, I. Physical activity and amyloid beta in middle-aged and older adults: A systematic review and meta-analysis. J. Sport Health Sci. 2024, 13, 133–144. [Google Scholar] [CrossRef] [PubMed]
  103. Bath, K.G.; Akins, M.R.; Lee, F.S. BDNF control of adult SVZ neurogenesis. Dev. Psychobiol. 2012, 54, 578–589. [Google Scholar] [CrossRef] [PubMed]
  104. Rossi, C.; Angelucci, A.; Costantin, L.; Braschi, C.; Mazzantini, M.; Babbini, F.; Fabbri, M.E.; Tessarollo, L.; Maffei, L.; Berardi, N.; et al. Brain-derived neurotrophic factor (BDNF) is required for the enhancement of hippocampal neurogenesis following environmental enrichment. Eur. J. Neurosci. 2006, 24, 1850–1856. [Google Scholar] [CrossRef]
  105. Scharfman, H.; Goodman, J.; Macleod, A.; Phani, S.; Antonelli, C.; Croll, S. Increased neurogenesis and the ectopic granule cells after intrahippocampal BDNF infusion in adult rats. Exp. Neurol. 2005, 192, 348–356. [Google Scholar] [CrossRef]
  106. Agnihotri, N.; Mohajeri, M.H. Involvement of Intestinal Microbiota in Adult Neurogenesis and the Expression of Brain-Derived Neurotrophic Factor. Int. J. Mol. Sci. 2022, 23, 15934. [Google Scholar] [CrossRef] [PubMed]
  107. Montagnani, M.; Bottalico, L.; Potenza, M.A.; Charitos, I.A.; Topi, S.; Colella, M.; Santacroce, L. The Crosstalk between Gut Microbiota and Nervous System: A Bidirectional Interaction between Microorganisms and Metabolome. Int. J. Mol. Sci. 2023, 24, 10322. [Google Scholar] [CrossRef]
  108. Lu, B.; Nagappan, G.; Guan, X.; Nathan, P.J.; Wren, P. BDNF-based synaptic repair as a disease-modifying strategy for neurodegenerative diseases. Nat. Rev. Neurosci. 2013, 14, 401–416. [Google Scholar] [CrossRef]
  109. Lu, B.; Nagappan, G.; Lu, Y. BDNF and synaptic plasticity, cognitive function, and dysfunction. Handb. Exp. Pharmacol. 2014, 220, 223–250. [Google Scholar] [CrossRef]
  110. Zhang, S.; Gu, B.; Zhen, K.; Du, L.; Lv, Y.; Yu, L. Effects of exercise on brain-derived neurotrophic factor in Alzheimer’s disease models: A systematic review and meta-analysis. Arch. Gerontol. Geriatr. 2024, 126, 105538. [Google Scholar] [CrossRef]
  111. Ruiz-Gonzalez, D.; Hernandez-Martinez, A.; Valenzuela, P.L.; Morales, J.S.; Soriano-Maldonado, A. Effects of physical exercise on plasma brain-derived neurotrophic factor in neurodegenerative disorders: A systematic review and meta-analysis of randomized controlled trials. Neurosci. Biobehav. Rev. 2021, 128, 394–405. [Google Scholar] [CrossRef] [PubMed]
  112. Zuccato, C.; Cattaneo, E. Brain-derived neurotrophic factor in neurodegenerative diseases. Nat. Rev. Neurol. 2009, 5, 311–322. [Google Scholar] [CrossRef] [PubMed]
  113. Vijay, N.; Morris, M.E. Role of monocarboxylate transporters in drug delivery to the brain. Curr. Pharm. Des. 2014, 20, 1487–1498. [Google Scholar] [CrossRef]
  114. Rojas-Valverde, D.; Bonilla, D.A.; Gomez-Miranda, L.M.; Calleja-Nunez, J.J.; Arias, N.; Martinez-Guardado, I. Examining the Interaction between Exercise, Gut Microbiota, and Neurodegeneration: Future Research Directions. Biomedicines 2023, 11, 2267. [Google Scholar] [CrossRef]
  115. Liu, L.; Huh, J.R.; Shah, K. Microbiota and the gut-brain-axis: Implications for new therapeutic design in the CNS. eBioMedicine 2022, 77, 103908. [Google Scholar] [CrossRef] [PubMed]
  116. Lu, S.; Zhao, Q.; Guan, Y.; Sun, Z.; Li, W.; Guo, S.; Zhang, A. The communication mechanism of the gut-brain axis and its effect on central nervous system diseases: A systematic review. Biomed. Pharmacother. 2024, 178, 117207. [Google Scholar] [CrossRef]
  117. Matteoli, G.; Boeckxstaens, G.E. The vagal innervation of the gut and immune homeostasis. Gut 2013, 62, 1214–1222. [Google Scholar] [CrossRef]
  118. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  119. Mirzaei, R.; Bouzari, B.; Hosseini-Fard, S.R.; Mazaheri, M.; Ahmadyousefi, Y.; Abdi, M.; Jalalifar, S.; Karimitabar, Z.; Teimoori, A.; Keyvani, H.; et al. Role of microbiota-derived short-chain fatty acids in nervous system disorders. Biomed. Pharmacother. 2021, 139, 111661. [Google Scholar] [CrossRef]
  120. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
  121. Tetel, M.J.; de Vries, G.J.; Melcangi, R.C.; Panzica, G.; O’Mahony, S.M. Steroids, stress and the gut microbiome-brain axis. J. Neuroendocrinol. 2018, 30, e12548. [Google Scholar] [CrossRef] [PubMed]
  122. Staley, C.; Weingarden, A.R.; Khoruts, A.; Sadowsky, M.J. Interaction of gut microbiota with bile acid metabolism and its influence on disease states. Appl. Microbiol. Biotechnol. 2017, 101, 47–64. [Google Scholar] [CrossRef]
  123. O’Mahony, S.M.; Clarke, G.; Borre, Y.E.; Dinan, T.G.; Cryan, J.F. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 2015, 277, 32–48. [Google Scholar] [CrossRef]
  124. Kasarello, K.; Cudnoch-Jedrzejewska, A.; Czarzasta, K. Communication of gut microbiota and brain via immune and neuroendocrine signaling. Front. Microbiol. 2023, 14, 1118529. [Google Scholar] [CrossRef] [PubMed]
  125. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  126. Malkiewicz, M.A.; Szarmach, A.; Sabisz, A.; Cubala, W.J.; Szurowska, E.; Winklewski, P.J. Blood-brain barrier permeability and physical exercise. J. Neuroinflamm. 2019, 16, 15. [Google Scholar] [CrossRef] [PubMed]
  127. Cass, S.P. Alzheimer’s disease and exercise: A literature review. Curr. Sports Med. Rep. 2017, 16, 19–22. [Google Scholar] [CrossRef]
  128. Hoffmann, K.; Sobol, N.A.; Frederiksen, K.S.; Beyer, N.; Vogel, A.; Vestergaard, K.; Brændgaard, H.; Gottrup, H.; Lolk, A.; Wermuth, L. Moderate-to-high intensity physical exercise in patients with Alzheimer’s disease: A randomized controlled trial. J. Alzheimer’s Dis. 2016, 50, 443–453. [Google Scholar] [CrossRef]
Figure 1. Alzheimer’s disease and the gut microbiome.
Figure 1. Alzheimer’s disease and the gut microbiome.
Brainsci 14 00974 g001
Figure 2. Involvement of physical activity in Alzheimer’s disease mechanisms. A black arrow indicates the mechanism implicated in Alzheimer’s disease, and a blue arrow indicates the impact of physical activity on the disease.
Figure 2. Involvement of physical activity in Alzheimer’s disease mechanisms. A black arrow indicates the mechanism implicated in Alzheimer’s disease, and a blue arrow indicates the impact of physical activity on the disease.
Brainsci 14 00974 g002
Table 1. Research on the gut–brain axis and exercise in Alzheimer’s disease.
Table 1. Research on the gut–brain axis and exercise in Alzheimer’s disease.
ReferenceProtocol for ExerciseAlzheimer’s Disease and the Microbiome
Nicolas, S. et al. [64]For ten weeks, rats were housed in pairs in either standard housing cages or cages that provided continuous and free access to running wheels.These rats were able to mitigate the reduction in hippocampal neurogenesis caused by gut microbes through physical exercise.
Abraham, D. et al. [65]Ten cycles of 6 min at high intensity, followed by two minutes at low intensity for two weeks in mice.There was an increase in Lactobacillus reuteri and bacteria that produce short-chain fatty acids, which resulted in improved cognitive function and slowed the progression of Alzheimer’s disease biomarkers.
Jin, K. et al. [66]A motor-driven treadmill for rodents was used to train mice five days a week for 18 weeks. In each session, a warm-up period of five minutes at a speed of five meters per minute was followed by five minutes at a speed of eight meters per minute and a maximum speed of eleven meters per minute for twenty minutes. Once the maximum speed reached 15 m/min, it was increased by 1 m/min every four weeks.There has been an increase in Akkermansia muciniphila and a decrease in Bacteroides species. A reduction in the progression of Alzheimer’s disease-like cognitive impairment is also observed, as well as an increase in the blood–brain barrier-related protein expression.
Teglas, T. et al. [68]The training was conducted four times a week for 60 min in mice. There were ten cycles of training, each consisting of four minutes at a high intensity (20 m/min) and two minutes at a low intensity (10 m/min).Changes in the microbiome caused by exercise do not significantly affect the mitochondrial density and the AMPK/mTOR/S6 pathways related to protein synthesis.
Wang, G. et al. [71]The running wheel was accessed freely and unlimitedly by micePseudomonadota and Tenericutes, Bacteroides, and Faecalibacterium were markedly reduced in the exercise group, while Allobaculum was increased
Zhang, Y. et al. [72]The running wheel was accessed freely and unlimitedly by miceIn APP/PS1 mice, exercise increased the alpha diversity index of cecal content
Ornish, D. et al. [74]A 20-week program of aerobic exercise (e.g., walking) and strength training exercises in humansThere was an increase in Blautia and Eubacterium in the intervention group during the intervention. Additionally, in the intervention group, the relative abundance of the taxa associated with an increased risk of Alzheimer’s disease decreased, e.g., Prevotella and Turicibacter
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

Sanchez-Martinez, J.; Solis-Urra, P.; Olivares-Arancibia, J.; Plaza-Diaz, J. Physical Exercise and Mechanism Related to Alzheimer’s Disease: Is Gut–Brain Axis Involved? Brain Sci. 2024, 14, 974. https://doi.org/10.3390/brainsci14100974

AMA Style

Sanchez-Martinez J, Solis-Urra P, Olivares-Arancibia J, Plaza-Diaz J. Physical Exercise and Mechanism Related to Alzheimer’s Disease: Is Gut–Brain Axis Involved? Brain Sciences. 2024; 14(10):974. https://doi.org/10.3390/brainsci14100974

Chicago/Turabian Style

Sanchez-Martinez, Javier, Patricio Solis-Urra, Jorge Olivares-Arancibia, and Julio Plaza-Diaz. 2024. "Physical Exercise and Mechanism Related to Alzheimer’s Disease: Is Gut–Brain Axis Involved?" Brain Sciences 14, no. 10: 974. https://doi.org/10.3390/brainsci14100974

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