Next Article in Journal
The Role of NMDA Receptor Partial Antagonist, Carbamathione, as a Therapeutic Agent for Transient Global Ischemia
Next Article in Special Issue
The Optic Nerve at Stake: Update on Environmental Factors Modulating Expression of Leber’s Hereditary Optic Neuropathy
Previous Article in Journal
Molecular Research on Alzheimer’s Disease
Previous Article in Special Issue
Air Pollution: A Silent Key Driver of Dementia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Life Experience Matters: Enrichment and Stress Can Influence the Likelihood of Developing Alzheimer’s Disease via Gut Microbiome

Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 22 May 2023 / Revised: 21 June 2023 / Accepted: 30 June 2023 / Published: 3 July 2023

Abstract

:
Alzheimer’s disease (AD) is a chronic neurodegenerative disease, characterized by the presence of β-amyloid (Aβ) plaques and neurofibrillary tangles (NFTs) formed from abnormally phosphorylated tau proteins (ptau). To date, there is no cure for AD. Earlier therapeutic efforts have focused on the clinical stages of AD. Despite paramount efforts and costs, pharmaceutical interventions including antibody therapies targeting Aβ have largely failed. This highlights the need to alternate treatment strategies and a shift of focus to early pre-clinical stages. Approximately 25–40% of AD cases can be attributed to environmental factors including chronic stress. Gut dysbiosis has been associated with stress and the pathogenesis of AD and can increase both Aβ and NFTs in animal models of the disease. Both stress and enrichment have been shown to alter AD progression and gut health. Targeting stress-induced gut dysbiosis through probiotic supplementation could provide a promising intervention to delay disease progression. In this review, we discuss the effects of stress, enrichment, and gut dysbiosis in AD models and the promising evidence from probiotic intervention studies.

Graphical Abstract

1. Alzheimer’s Disease: Still No Cure

Dementia is a set of symptoms, including but not limited to memory loss, problem-solving issues, and mood changes, spanning decades and not considered a “normal” part of aging [1,2]. Alzheimer’s disease (AD) is the leading cause of dementia, being responsible for 60–80% of dementia diagnoses and typically, though not exclusively, affecting people aged 65 years and older. AD is a chronic neurodegenerative disease, characterized by the presence of β-amyloid (Aβ) plaques and neurofibrillary tangles (NFTs) formed from abnormally phosphorylated tau proteins (ptau) [3]. Aβ plaques accumulate in the extracellular space between neurons, while NFTs accumulate within the neurons themselves, each preventing normal cellular functioning and communication, eventually leading to their deterioration [4,5]. Aβ and NFT can be examined post-mortem, as well as detected using fluid and imaging biomarkers in live patients [3,6]. Symptoms of AD appear to gradually and progressively worsen from mild cognitive impairment (MCI) to severe impairment, changes in personality, and physical disabilities requiring constant care.
Drug treatment options have focused primarily on mid-to-late-stage AD and have historically targeted the cholinergic system implicated in memory decline or the Aβ cascade [7,8]. These drug treatments have been largely ineffective, demonstrating, at most, some improvement in cognition but still an overall decline as time progresses. In addition to this, new treatments are being explored targeting tau and tau mechanisms. Drugs targeting post-translational modifications of tau such as inhibition of tau hyperphosphorylation kinases (GSK3B, CDK5, Fyn), promoting the activity of tau dephosphorylation, or inhibiting tau acetylation are in phase 1 and 2 of various clinical trials [9,10,11]. These drugs include rapamune, nilotinib, lithium chloride, salsalate and MK8716. Drugs targeting tau aggregation (methylene blue, curcumin), microtubule stabilization (NAP, TPI 287), tau expression and tau clearance through immunotherapy (C2N-8E12, BMS-986168, AADvac-1) are currently undergoing trial [12]. Despite promising preclinical results, many trials targeting tau have failed due to adverse effects [13]. Methylene blue, which showed great promise in animal studies, was shown to be limited to inhibition of tau fibril formation and does not provide the same effect to tau oligomers [10]. Other therapeutic strategies such as anti-inflammation have been reviewed elsewhere [13]. From these drug trials, it is becoming evident that pathological processes begin long before clinical symptoms, and no significant clinical benefits have been observed. Given the gradual and progressive nature of AD, an emphasis must be put on finding targets for treatment that are present during the earlier, pre-clinical stages of AD progression. This window for treatment spans years to decades, therefore providing ample opportunity to change potential outcomes.

2. Stress, Enrichment and the Gut Microbiome: Potential Focus for a Cure

While there is evidence of AD being highly heritable, ranging from approximately 60–100% heritability depending on the timing of disease onset, the prevalence of genetically linked AD cases is very low (<1%) amongst all cases of the disease [14,15,16,17]. This suggests that there are other non-genetic, environmental risk factors that play a larger role in determining whether an individual develops AD and, therefore, may provide avenues for alternative treatment options.

2.1. Stress and AD

Up to 40% of Alzheimer’s risk can be attributed to environmental factors [18]. Disease development and progression can be strongly influenced by stress, although the presence of stress alone may not lead to Alzheimer’s disease. Stress may be one of the many factors that influence whether symptoms will appear earlier or later when a precursor to the disease (i.e., persistently phosphorylated tau) is present. Stress-induced physiological processes can adversely affect healing, coping, and maintaining a positive quality of life. Cognitive and motor functions are deteriorated by neurodegenerative diseases, which is by itself a form of stress. Furthermore, AD also leads to dysregulation of the stress response through the HPA axis [19].
Stress is well-known to play a negative role in the development of a variety of disorders such as cardiovascular disease, obesity, and gastrointestinal disorders, as well as psychiatric and neurodegenerative disorders. According to work on animal models, manipulating the glucocorticoid milieu can result in behavioural, molecular, and cellular changes similar to AD [20,21,22]. Specifically, stress or glucocorticoid administration increases Aβ precursor protein and tau phosphorylation, both of which are associated with synaptic dysfunction and neuronal death in AD [21,22].
Several studies have found direct links between stress and Alzheimer’s disease pathology. A study published by Sotiropoulos and colleagues found that stress resulted in abnormal phosphorylation of tau [18]. Stress upregulates at least two tau epitopes, which are strongly implicated in the pathophysiology of Alzheimer’s disease. In another study on more than thirteen thousand patients tracked over the course of fifty years, a two-fold greater risk of dementia in the late stages of life in patients who suffer from depression was observed [23]. An analysis of study results from the Rush Memory and Aging Project showed that patients who demonstrated a high level of “distress proneness” were three times more likely to develop dementia over the course of three years [23]. Neuroticism, which refers to the tendency to experience distress and negative emotions, has been linked to a wide variety of mental health conditions. It is known that individuals with high levels of neuroticism are more likely to suffer from depression, anxiety disorders, and post-traumatic stress disorder [24]. Also, evidence suggests a link between neuroticism and Alzheimer’s disease. An 800-member cohort of the Catholic clergy was followed for five years by Wilson and colleagues, in which participants were assessed for distress proneness at baseline [25]. They found that higher levels of neuroticism at baseline were associated with a higher risk of developing AD dementia over time, namely, people who were distressed had a 2.4 times higher risk of developing dementia [25].
It has been found that patients with AD possess increased cortisol levels in their biological fluids (e.g., plasma, saliva, and cerebrospinal fluid) [20], which correlates with the high association between anxiety-related mental illness and AD [26]. Researchers have found that those with mild cognitive impairment typically have higher average circulating cortisol levels than their age-matched counterparts at all diurnal time points [24]. Moreover, patients with dementia exhibit impaired hypothalamic–pituitary–adrenal (HPA) axis feedback when dexamethasone is given, indicating impaired HPA axis function. The release of cortisol is expected to increase with increased HPA activity, accelerating and intensifying disease progression.

2.2. Enrichment Effects on Cognition and AD

Environmental enrichment can also influence the development and progression of dementia. Enrichment is often examined as cognitive (i.e., mentally stimulating or requiring mental processing) or physical activities. Engaging in cognitively stimulating activities may alter the AD pathophysiological cascade in a positive manner among older adults. In a longitudinal study examining life-long cognitive enrichment, including education and other mentally stimulating activities, it was found that high cognitive enrichment scores throughout the lifespan reduced the risk of developing cognitive impairment, and delayed progression from mild cognitive impairment to dementia [27]. Among cognitively normal older adults, Wilson et al. [28] found that following a 6-year long, multi-interview assessment of frequent cognitive activity, with activities such as reading, watching television or listening to the radio, playing games or completing puzzles, and going to museums, each additional point of cognitive activity reduced cognitive decline by 52% during follow up assessments. Leisure activities that stimulate cognitive function have been associated with a reduced risk of dementia development. For example, crossword puzzles may reduce the risk of cognitive decline by improving cognitive reserve, by directly modifying the disease, or by integrating a variety of other healthy behaviours [29]. It has been reported that individuals engaged in cognitively stimulating leisure activities experienced slower rates of cognitive decline and a faster rate of post-onset cognitive decline—this is consistent with the cognitive reserve hypothesis, which holds that cognitively stimulating activities may delay the development of clinical cognitive impairments [30,31].
The results of psychological studies have suggested that conversation is a highly cognitively stimulating task. To understand others’ intentions and feelings, in addition to linguistic ability, conversations require attention, working memory, organizational and control of thoughts (executive functions), and social cognition. Across decades of research, isolation, which older adults are particularly susceptible to, has consistently been linked to cognitive decline and AD. Compared with their counterparts who had five or six social connections, elderly individuals with no social connections were 2.37 times more likely to experience cognitive decline in one cohort study [32]. Strengthening this, loneliness increases the risk of AD by more than double [33] and has been found to spike levels of Interleukin-6, an inflammatory agent implicated in numerous age-related diseases, including AD [34].
One enriching activity is dancing, which has grown in popularity amongst seniors in recent years as it integrates a variety of aspects, from audiovisual perception to physical perception, and emotional expression. This method of non-pharmacological cognitive intervention is beneficial to older adults because it improves episodic memory, executive performance, and global cognition [35]. Additionally, the use of music, which engages the entire brain, including the auditory system, syntactic system, semantic system, memory, and motor function, can contribute to the enhancement of cognitive function.
Studies have demonstrated that mild to moderate levels of physical activity can reduce the likelihood of dementia and AD. In a systematic review of 163,000 non-psychotic participants, researchers discovered that physical activity could reduce the risk of dementia by 28% and AD by 45% [36]. Various studies have linked exercise with hippocampus development as it reduces the decline of cortical tissue in the elderly. MRI findings showed that active individuals had a larger hippocampus, better spatial memory, and improved cognitive and physical health. There is a growing body of evidence that exercise reduces cognitive degeneration and dementia [37].

2.3. Microbiota–Gut–Brain Axis and AD

The microbiota–gut–brain axis represents the bidirectional communication between the gut and the brain. In AD, gut dysbiosis is becoming increasingly important due to the effects it can have on gut barrier function, blood–brain barrier (BBB) function, and neuroinflammation. Increased barrier permeability, as seen in AD studies, provides an opportunity for toxic amyloids, neuroinflammatory cytokines, lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs) to move into the brain and seed neurodegeneration [38]. The microbial community of the gut microbiota plays an active role in homeostasis and disease. In humans, aging contributes to the increase in pro-inflammatory bacteria, which has been found to induce systemic inflammation. Pro-inflammatory bacteria and, in turn, neuroinflammation has been found in amyloid-positive patients when compared to healthy elders [39]. Human studies aiming to classify differences in bacterial species in the gut makeup of healthy vs. AD patients have provided insight into the interplay between human health and disease. Vogt et al. discovered a significant decrease in Firmicutes and Bifidobacteria in the fecal samples of AD patients, accompanied by an increase in Bacteroidetes species [40]. This was validated through 16sRNA sequencing conducted by Zhuang et al. and Liu et al., finding similar results with the addition of the dysregulation of multiple other bacterial families [41,42]. Interestingly, a difference between Enterobacteriaceae was also found between MCI and AD patients, indicating a change in gut composition with disease progression [42]. A further understanding of the regulation of bacterial species through aging and disease is needed, as vast discrepancies can be found between patients, demographic regions, and ages.

2.4. Stress and Gut Dysbiosis

The complex system of the gut microbiota is constantly challenged by biological and lifestyle factors, including environmental factors like stress. Studies on military personnel reveal the detrimental effects of chronic stress in extreme situations and often multiple stressors in combination [43]. In this example of extreme chronic stress, many health decrements have been observed including musculoskeletal injury [44], endocrine disruption [45], inflammation [46], and illness and infection [47]. Military personnel are a common group to study changes from extreme stress in gut microbiota, however, various levels of stress are a ubiquitous part of daily life for many individuals.
Commonly, stress induces the release of hormones such as glucocorticoids and catecholamines which have been shown to modulate GI function and microbial growth [48,49,50]. Stress-induced changes along the vagus nerve have also been found to reduce digestive activity, likely altering substrate availability [50]. It has been shown that stress-induced changes in gut microbiota in humans can degrade the physical gut barrier and increase gut permeability [51].
In a longitudinal study on military personnel, 73 soldiers were placed in a situation of high stress and examined for microbiota composition changes and intestinal permeability [52]. It was found that 23% of metabolites were significantly altered after stress, and increased intestinal permeability was observed through an increase in sucralose excretion during stress, independent of diet [52]. Additional longitudinal studies were performed on psychological stress in students before and during exams [53,54]. In students, it was found that fecal levels of lactic acid bacteria were lower during a high-stress situation, associated with an increase in cortisol concentration [53]. Strengthening this, higher levels of salivary cortisol were observed before an exam [54]. The daily consumption of probiotic Lactobacillus casei significantly reduced gastrointestinal stress and symptoms and was found to preserve the diversity of the gut microbiota compared to non-probiotic-fed students [54].

3. Animal Models of Stress, Enrichment, and Gut Dysbiosis in AD

As aforementioned, environmental factors are implicated in the development of symptoms related to dementia and AD, including environmental enrichment, stress, and gut microbiome health [55,56,57]. Environmental enrichment, including enhanced living conditions, increased social encounters, and a variety of activities has been shown to improve cognitive abilities and reduced symptoms of stress and depression often comorbid with AD. Conversely, stress, particularly chronic stress, is one of the largest risk factors associated with AD and has been implicated in both the onset of the disease and in worsening disease progression. The bi-directional communication between the gut and the brain is also thought to play an important role in disease progression on its own and can be altered by both stress and enrichment. The effects of enrichment, stress, and gut health on AD are frequently studied in animal models and those models will be discussed below.

3.1. Roles of Enrichment in Animal Models of Alzheimer’s Disease

A large number of animal models of environmental (EE) and social (SE) mimicking cognitive enrichment in humans, along with physical (PE) enrichment, have shown that enrichment largely improves cognitive symptoms of AD and ameliorates underlying pathological processes and inflammatory responses and reduces neuronal death [58,59,60,61,62,63,64,65]. Transgenic mouse and Aβ-infused rat models of AD using PE, including voluntary running exercise and resistance ladder climbing, showed improvements in both short- and long-term memory, reduced inflammation, and reversed deficits in neurogenesis compared to non-physically enriched AD counterparts [60,61,65,66,67]. However, PE is not as effective as other forms of enrichment alone and is often used in combination with or as part of other enrichment paradigms. Similarly, SE, achieved through grouping animals together either in housing or for the duration of the paradigm, in AD models also shows promising results, with animals having intact social recognition, improved spatial learning, improvements in motor skills, and reduced anxiety-like symptoms compared to non-enriched transgenic or Aβ-infused AD model animals [60,62,63,68], like with PE, SE alone does not produce as strong of effects as more comprehensive or combined protocols of enrichment. EE typically involves providing subjects with a variety of items that can be interacted with, including toys, chews, hides, treats, and running wheels. In AD models, EE has been shown to improve recognition and spatial memory, reduce tau and Aβ pathology, improve neurogenesis, reverse effects of stress and high-fat or high-sucrose diets, reduce inflammation, increase synaptic plasticity, improve immune system activity, and reduce neuronal death seen in AD [58,59,60,61,62,63,66,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87]. These vast beneficial effects of EE are seen in both mouse and rat models, with enrichment protocols occurring over timelines beginning between 3 weeks of age to 18 months, and lasting between 1 month and 23 months. Results across varying timelines are relatively consistent, though some studies have found time-dependent effects of enrichment on AD pathology and symptom development. For example, Mirochnic and colleagues found that 6 months of enrichment was sufficient for EE, but not PE, to elicit beneficial effects, but both were beneficial after 18 months [66]. Similarly, Polito and collaborators [88], found that following 15 months of EE, Aβ deposition was reduced; however, 4 months was not long enough to produce the same effect despite being sufficient to reduce the spatial memory deficit seen in AD. Others have implemented EE protocols prior to AD pathology appearing and found that it can have protective or preventative effects [60,74,76,89].
Another strategy to examine the protective effects of EE has been to implement enrichment protocols during gestation or neonatal stages. Ziegler-Waldkirch and colleagues [90] found that pregnancy and lactation increased Aβ pathology in an APP/PS1 transgenic mouse model, but exposure to EE during pregnancy and the lactation period prevented such pathology, with those dams having as little Aβ and as much neurogenesis as non-pregnant counterparts. Furthermore, another study found that EE during pregnancy of AD transgenic left offspring with intact learning and memory and synaptic plasticity, suggesting generational protective effects [91]. Gentle handling during the neonatal period has produced some mixed results. One study found that this form of enrichment early in life was able to reduce behavioural traits of AD (i.e., increased swim time and reduced immobility time in a forced swim test) more effectively than EE later in life [92]. On the other hand, a study found neonate handling to only produce mild protective effects against spatial memory decline, though that effect was more pronounced in females than males [93].
There are also examples of rather unique enrichment protocols. One study, conducted by Yeung and colleagues [64], used repeated cognitive testing as intellectual or cognitive enrichment in their 3xTG-AD mouse model of AD and found that this regular training improved performance on a separate spatial test compared to non-enriched AD animals. Another study used olfactory enrichment, in which a different odor was presented in the animals’ home cage each day over 40 or 80 days [94]. They found that long-term olfactory enrichment reduced tau phosphorylation. Results from these studies demonstrate that a variety of activities and stimuli can be considered enriching and produce beneficial effects to protect against AD.
Enrichment does not always produce such promising results, however. A number of studies have shown that EE produces rather mild or mixed results, especially during the later stages of AD pathology. These findings include failing to reduce Aβ or tau pathology, cognitive decline, or neuronal loss, and not improving cognitive performance, inflammation levels, or neurogenesis [95,96,97,98,99,100,101,102,103,104,105,106]. These works have not necessarily been unsuccessful in demonstrating some improvements following EE, but the effects are not as robust as the studies discussed above. Additionally, some studies have found that EE worsens AD pathology, resulting in reduced neurogenesis, increased apoptosis, and more rapid and extensive accumulation of Aβ despite cognitive improvements in AD animals, while producing opposite results in healthy counterparts [107,108,109,110]. These studies suggest that enrichment may facilitate some pathological processes of AD due to genetic predisposition. Fulopova and collaborators [111], also provided a possible explanation for the varied results of enrichment studies by demonstrating that Aβ accumulates more so in the prefrontal cortex than in other brain regions, and while EE is able to improve cognitive function and reduce Aβ accumulation, the prefrontal cortex maintains high levels of plaques.
Some experimenters have recognized that EE alone is not sufficient to produce the desired widespread improvement in cognitive function and pathology in AD and have used other therapies alongside enrichment to assess their combined effect. Studies combining EE with drug therapies, such as memantine, an N-methyl-D-aspartate receptor antagonist, donepezil, an acetylcholinesterase inhibitor, magnesium-L-threonate, an ingestible method to increase essential magnesium in the brain, or Bifidobacterium breve probiotic, have shown that the drug or EE alone can produce some benefits, but when combined there is improvement in learning and memory or more long-term effects [112,113,114,115]. Combination therapy using EE and caffeine was not as robust, with individual therapies having nearly equal effects as combined therapies [116]. Alternatively, one study using an aluminum induction mouse model of AD combined EE with resveratrol treatment, an antioxidant and anti-inflammatory compound, and found that while both individually reduced Aβ deposition, when combined the Aβ burden worsened [117].
Ultimately, these enrichment studies provide a hopeful message that a variety of enriching stimuli, including enjoyable activities, a varied diet, physical activity, social interactions, mentally stimulating activities, and even olfactory stimulation, can reduce the risk and severity of AD pathology and symptom development. Enrichment may counteract some negative environmental influences, including stress and an unhealthy diet, that would worsen the risk of AD, and if not sufficient alone, can be used in combination with other therapies to combat symptom development in AD. These enrichment AD models are summarized in Table 1 below.

3.2. Roles of Stress in Animal Models of Alzheimer’s Disease

Animal models examining the roles of stress, in its many forms, and stress-associated hormones in AD have demonstrated a link between environmental stress and pathological progression and symptom development of AD, often concluding that corticotrophin-releasing factor (CRF) is an essential mechanistic factor in the said link [18,62,119,120,121,122,123,124,125,126,127,128]. Further, this relationship is bidirectional, with stress acting as a driving force behind AD-like symptoms and pathology development, shown from studies finding AD pathology and symptoms in wild-type (WT) subjects post stress, but also making subjects more prone to stress and susceptible to its effects [129,130,131,132,133,134,135].
One common stress paradigm used in animal models of AD is chronic unpredictable stress (CUS), typically involving a number of different stressors applied in random—or unpredictable—order over three to six weeks [18,62,122,129,133,134,136,137,138,139,140,141]. Frequently used stressors include soiled bedding, a tilted cage, bright lights or disrupted light schedule, and changing litter mates. Studies using such methodology have shown that this leads to increased Aβ deposition, increased ptau, impaired spatial memory, reduced synaptic plasticity, increased impulsivity, and impaired olfactory discrimination learning.
Similarly, sleep deprivation (SD) is another prominent method used to elicit stress in AD models and is fairly effective at inducing AD pathophysiology, with SD in WT animals usually resulting in reduced cognitive function and accumulations of Aβ and ptau [130,131,142]. There is great variability in the duration of protocols of SD, with some as short as a single 4 h session, and others lasting up to 6 weeks [130,143]. The most common method to achieve SD in rodent models of AD is using a modified cage filled with water and only small platforms throughout so that animals are still able to move around but will fall into the water when they fall asleep, which results in complete SD or fragmented sleep. Lack of sleep has devastating effects on spatial memory, fear learning, and synaptic plasticity, and results in increased Aβ and ptau deposits [143,144,145,146,147,148,149,150,151,152,153,154]. Difficulty sleeping is also one of the ways in which the bidirectional link between stress and AD is demonstrated. As described, SD or fragmentation has been shown to increase AD pathology; however, Kincheski and collaborators [144] infused Aβ into the brains of subjects and found them to have more fragmented sleep. This two-way relationship between stress and AD can create a cycle, with stress triggering AD pathology, which, in turn, increases levels of stress.
Other common stress paradigms used include restraint and immobility (RIS) stress and social isolation (SI). Like with CUS and SD, RIS and SI also increased levels of Aβ and ptau, with one study finding this to be specific to female subjects after 5 days, poorer spatial and fear memory, increased inflammation, increased neurodegeneration, and CRF implicated as a mediating factor for these changes [120,121,124,155,156,157,158,159,160,161,162]. Despite this, some studies have found that RIS alone may not be sufficient to elicit strong effects, either with behavioural or pathological changes, suggesting that some stressors may not be as detrimental to health as others [163,164].
Studies examining the hormonal aspect of stress (HS) and its relationship to AD have concluded that glucocorticoids can lead to increased soluble Aβ and impair fear recall, while CRF and its receptor, corticotrophin-releasing factor receptor-1, are involved in the process of ptau phosphorylation [115,119,123,128,151]. Other studies have examined alternate styles of stressors, including maternal separation (MS) and psychosocial stress (PS), and found that these stressors lead to worsened spatial memory, increased levels of stress hormones, increased inflammation, and increased Aβ and ptau in AD animals [125,126,165,166,167].
These animal studies create a clear image that stress is linked to the pathological progression of AD and symptom development. Stressors have been applied at time points from during pregnancy, early in life, and into late adulthood, and largely have resulted in worsened cognition and increased AD pathology. These models, along with the models of enrichment discussed earlier, create a strong case encouraging people to avoid stress and live an enriched life. A summary of the stress models is provided in Table 2 below.

3.3. Stress and Gut Microbiota in Animal Models

The gut microbiota has been identified to play a key role in regulating brain function. The bidirectional network between the gut and brain is connected by various pathways including the vagus nerve, neurotransmitters, immune system, hypothalamic–pituitary–adrenal (HPA) axis, and metabolites [67,168,169]. The modification of the gut microbiota and gut community makeup has gained attention in recent years and positive outcomes have been shown in various diseases.
The gut undergoes periods of shift, where significant changes and regulation of bacterial makeup occur at specific time points in one’s life [170]. Interestingly, these periods can be targeted through stress paradigms, and the important gut establishment can be disrupted. Specific changes in gut microbiota communities due to stress-inducing paradigms are outlined below.

3.3.1. Gut Dysbiosis and Stress

Stress has been shown to have varying effects on gut dysbiosis and gut health at all stages of life. Studies in germ-free (GF) mice have provided evidence for clear communication between the gut and the brain, termed the “microbiota gut–brain axis” [171]. It was found that in GF mice, mild restraint stress produced an upregulation of corticosterone and adrenocorticotropic hormone, indicating a critical role of the microbiota in the stress response and HPA axis [171]. Recent research has provided strong evidence to show that gut microbiota makeup is related to stress in both animal models and humans. Further strengthening the relationship between stress and the gut, stressors presented in animal studies have been shown to negatively affect gut health and cause gut dysbiosis (Table 3). Together, these studies present evidence that stress can be both a cause and a result of gut dysbiosis.
It has been well documented that stress exposure can have a direct influence on the gut microbiota makeup, and can damage the balance between microbes. A plethora of bacteroids has been found to be regulated through chronic stress paradigms in animal models. Specifically, maternal separation (MS) is used as a common model of stress. MS is based on the work of Hofer, where the deleterious impact of early weaning was revealed, and shown to have implications on offspring physiology and intestinal health [172]. The model uses the early mother–infant separation in animals that most commonly occur during the first 1–3 weeks of life [173]. This development period is a critical window of both sensitivity to stress and microbiota community establishment [174,175]. A growing body of evidence suggests MS paradigms can alter the gut microbiota composition in both animal and human models. However, the quantification and direction of gut disturbance are not consistent across studies due to the use of different species, strains, sex, protocols, and microbiota analyses.
Levels of fecal and intestinal bacteria have been shown to be altered in many animal studies of MS. As shown in Table 3, studies in mice induced gut microbiota alterations between stressed animals and control groups. Specifically, the abundance of bacteria and diversity levels in the microbiome were negatively affected by the stress paradigm. In support of this, in one study on postnatal day (PD)-28, mice showed a reduced level of alpha diversity in the gut [176]. In contrast to this, Moya-Perez et al. [177] showed no change in diversity. MS has been shown to have the ability to increase systemic inflammation, and systemic microbial loads through gut barrier dysfunction [176,177]. Levels of toxic bacteria have also been found to be upregulated in mice [176], and symptoms of IBS (intestinal hyperpermeability, visceral hypersensitivity, microbial dysbiosis, and low-grade intestinal inflammation) were found as a result [178]. Gareau et al. [179] showed increased adhesion and penetration of total bacteria in the gut, along with significantly reduced levels of Lactobacillus species. Notably, levels of bacteria and SCFA-producing genera can be dysregulated due to MS. Significantly higher levels of SCFA-producing genera such as Fusobacterium and Clostridium have been found in the gut, and this was shown to be positively correlated with the degree of visceral hypersensitivity [180,181].
Physical stressors are another popular form of stress paradigm used in animal studies. In a study on germ-free mice, it was demonstrated that a stress paradigm induced an exacerbated release of corticosterone and adrenocorticotropic hormone when compared to controls [171]. In addition to this, Table 3 shows the effects on the gut microbiome of various RIS studies in various mouse and rat models. Alterations of gut microbiota including increased kynurenine toxicity, changes in bacterial makeup, downregulation of microbiome flora richness, and disrupted alpha and beta indices of diversity have been observed [182,183,184,185,186,187]. As seen in MS stress-induced studies, intestinal hyperpermeability was observed due to RIS and tight junction protein levels (ZO-1, Occludin, and Claudin1) were disrupted [188,189]. Levels of Akkermansia, a common mucin-degrading bacteria, were significantly decreased in stressed animals compared to controls. This bacteria is inversely correlated with inflammation, diabetes, and obesity [190]. Together, studies revealing stress-related gut dysbiosis provide support for the critical role the microbiota may have in the development of the stress response and the effect that stress has on neurodegenerative diseases, such as AD.
Interestingly, the HPA axis, the main stress axis in mammals, has been shown to be in close communication with the gut microbiome, and one of the important methods of communication between the gut and the brain [191]. Activation of the HPA axis causes a release of CRF from the hypothalamus, and CRF becomes one of the main stress-related neuropeptides involved with both the brain and gut [191]. Stress-induced activation of the HPA axis by MS increases levels of corticosterone, CRF, and has been shown to also affect the permeability of the intestinal wall [192,193,194]. Both CRF and the family of neuropeptides urocortin (Ucns) are shown to have proinflammatory and anti-inflammatory effects on the GI. Regulating the release of these peptides alters the inflammation in the GI tract and plays a role in increasing intestinal barrier leakage [193,195,196].
Table 3. Studies of stress effects on gut microbiome from common methods: maternal separation and restraint stress.
Table 3. Studies of stress effects on gut microbiome from common methods: maternal separation and restraint stress.
StressorAnimal ModelFindingsReference
Maternal SeparationC57BL/6J (male)Early life stress-induced gut microbiota alterations and lasting CNS inflammation
Rescued by probiotics
Probiotics improve glucocorticoid sensitivity
[177]
C57BL/6J (male)Significant reduction of serum TNF-a and increase in IL-6
Reduction of IL-6 levels after probiotic ingestion
[197]
C57BL/6J (male)Differential abundance of gut microbiota in maternal separation groups differed from control groups
Stress exposure reduced the alpha diversity and altered microbial community at PD28
[198]
C57BL/6J (male)Maternal separation combined with chronic unpredictable stress paradigms increased systemic microbial load through gut barrier dysfunctions
Stress-related increases in Clostridium were observed
[176]
C57BL/6N (both sexes)Maternal separation-induced changes in animals lead to intestinal dysbiosis[199]
NMRI mice (both sexes)Maternal separation altered the composition of gut microbiota[200]
C3H/HeN mice (male)Maternal separation induces microbiota dysbiosis in favor of pathobionts
IL-17 and IL-22 decreased in response to glucose intolerance in stressed animals compared to controls
[201]
C3H/HeN mice (both sexes)Maternal separation induced the main features of IBS (intestinal hyperpermeability, visceral hypersensitivity, microbiota dysbiosis, and low-grade intestinal inflammation[178]
SD (male)Maternal separation reduced swim behaviour and decreased mobility in forced swim test
Stress-related decrease in brain noradrenaline and increase in peripheral IL-6
Probiotic ingestion normalized these levels
[202]
Maternal SeparationSD (both sexes)Maternal separation increased adhesion/penetration of total bacteria in gut and significantly reduced Lactobacillus species
Stress-related elevation of corticosterone levels observed
Probiotic ingestion ameliorated gut penetration and restored corticosterone levels
[179]
SD (both sexes)Maternal separation pups showed an adult-like profile of long-lasting fear memories and fear relapse following extinction
Probiotic-treated pups exhibited age-appropriate infantile amnesia and resistance to relapse
[203]
SD (both sexes)Maternal separation disrupted the Firmicutes-to-Bacteroides ratio in the gut
Stress-related disruption of acetate, propionate, and butyrate in fecal samples was observed
This was restored by a probiotic mixture
[204]
SD (both sexes)Significant differences in microbial community of gut in Maternal separation group compared to control[205]
SD (both sexes)Maternal separation showed a difference in abundance of various bacteroids in fecal samples of both sexes
Levels of proinflammatory cytokines were increased in the colon and sera of male stressed rats
[206]
SD (both sexes)Maternal separation animals showed significantly lower levels of SCFA producers[180]
SD (both sexes)Maternal separation animals showed a difference in SCFA-producing genera, Fusobacterium and Clostridium compared to controls
Fusobacterium stress-related increase positively correlated with the degree of visceral hypersensitivity
[181]
SD (both sexes)Maternal separation triggered gut microbiota composition changes compared to control groups
Gut dysbiosis was reversed by probiotic ingestion (Lactobacillus)
[142]
Maternal SeparationSD (both sexes)Maternal separation produced alterations in the structure and composition of the gut microbiota
Significant differences in bacterial types in stressed animal microbiomes compared to controls
[207]
SD (both sexes)Maternal separation rats showed hypercorticosteronemia, enhanced intestinal permeability, and changes in gut microbiota structure
Probiotic feeding prevented these changes
[208]
Wistar rats (male)Significant gut barrier dysfunction in maternal separation groups compared to control
ML-7 (MLCK inhibitor) strengthened the intestinal barrier and restored levels of numerous bacteria in the gut
[173]
Physical RestraintC57BL/6J (male)Chronic restraint stress exacerbated kynurenine (Kyn) toxic signaling in the gut, especially the colon
Indoleamine 2,3-dioxygenase was upregulated in the brain and gut, promoting transfer of Tryptophan metabolic pathway to Kyn signaling
Stressed mice showed enhanced intestinal permeability compared to controls
[182]
C57BL/6J (male)Restraint stress increased intestinal hyperpermeability and disrupted tight junction proteins (ZO-1, Occludin, and Claudin1)
Increased inflammation in enteric glial cells
increased relative abundance of fecal Akkermansia (mucin-degrading Gram-negative bacteria)
[188]
C57BL/6J (male)Chronic restraint animals showed decreased levels of SCFA in feces compared to controls
Stressed animals showed gut dysbiosis, and decreased levels of Occludin and Claudin-1, correlating with decreased intestinal barrier function
[189]
C57BL/6J (both sexes)Chronic restraint stress revealed sex differences in fecal microbiota makeup
Fecal transplant of stressed mice to germ-free mice decreased pain threshold, and resulted in further sex differences in gut-microbiota makeup
[209]
Physical RestraintC57BL/6N (male)Chronic restraint mice exhibited alterations in microbiota composition, disruption of colonic mucus, and aggravation of colitis
Abundance of Akkermansia was significantly decreased in stressed mice compared to controls
[183]
C57BL/6 specific-pathogen free (both sexes)Lactobaccillus animalis was enriched in stress group, and positively correlated to behavioural deficiencies[210]
ICR mice (male)Microbiome flora richness was significantly lower in the stress group compared to controls[184]
Wistar rats (male)Chronic restrain stress increased gut dysbiosis compared to controls
Both alpha and beta diversity indexes were increased in stress animals
[185]
SD (male)Chronic restraint stress enhanced the abundance of bacteroids and altered gut microbiota and metabolites
Altered gut microbiota was correlated with PI3K/Akt/mTOR pathways
Chronic restraint stress decreased phosphorylation of PI3K/Akt/mTOR pathway in microglia and enhanced LPS-induced microglia activation
[211]
SD (male)Restraint-stressed rats exhibited dysregulated gut microbiota
Fecal B-d-glucosidase activity differed from control rats, leading to systemic exposure to ginsenoside RB1 and its metabolites
[186]
SD (both sexes)Chronic restraint stress-induced dysbiosis[187]

3.3.2. Role of Stress-Induced Gut Dysbiosis in AD

With stress having a lasting impact on gut health and contributing to gut dysbiosis, many physiological and behavioural consequences have also been highlighted. Stress-induced increased inflammation and impaired cognition have been found in animal models of various neurodegenerative diseases, depression, and irritable bowel syndrome (IBS) [212,213]. Specifically, the intestinal barrier has been found to be damaged in AD patients and animal models [214,215]. Gut microbes influenced by stress exposure can impact intestinal barrier function and ultimately lead to intestinal permeability [182,188,203,216]. Interestingly, when probiotic Lactobacillus was administered in rodents, this barrier leakiness was prevented, along with improved behavioural, cognitive, and biochemical parameters [213,217].
A correlation between increased dysbiosis and stress in the gut was shown to be related to the PI3K/Akt/mTOR pathways [185]. Chronic stress decreased phosphorylation of this pathway in microglia and enhanced LPS-induced microglial activation [185]. This is consistent with AD research, in that the activation of this pathway subsequently produces an increase in GSK-3B-induced tau phosphorylation [218].
Alterations in the gut–brain axis and the composition of gut microbial species are seen in aging. LPS, neuroinflammatory cytokines, and amyloids, which may contribute to the pathogenesis of AD, have been hypothesized to pass through the compromised gastrointestinal tract (GI) tract and BBB [219,220,221]. The increased leakiness of the intestinal barrier from gut dysbiosis can increase the abundance of toxic movement from the gut to the brain, and enhance disease progression. Stress paradigms such as MS have been associated with increased toll-like receptor 3,4 and 5 mRNA expression [222]. Implications of this increase can include higher susceptibility to infection, higher levels of pathogenic bacteria in the gut, and an increase in cytokine production, which can seed neuroinflammation in AD [222]. In addition to this, downregulation of levels of the gene expression encoding the glucocorticoid receptor, which is responsible for down-regulating inflammation in the cortisol response, was found in models of early life stress [118]. In the same study, it was also found that exposure to stress at a young age increased the expression of toll-like receptor (TLR) 4, activating the innate immune system response, and increasing inflammation [118].

4. Therapeutic Potential of Probiotics on Gut Dysbiosis

Probiotics have been shown to maintain human intestinal health and are defined as live microorganisms which confer a health benefit on the host by the World Health Organization [223]. Alteration of the gut microbiota can be achieved through the supplementation of probiotics, although the viability when administered remains questionable in some cases. Two commonly used probiotic species, Lactobacillus and Bifidobacterium, have been shown to have promising effects on host health by changing the gut microbiota composition. In healthy subjects, probiotic supplementation of these reveals a decrease in anxiety, depression, and stress-related behaviors [224,225]. Both of these species have been shown to inhibit harmful bacteria, improve gastrointestinal barrier function, and suppress proinflammatory cytokines [226]. Certain probiotics have the potential of suppressing AD-related GSK3B overexpression, by modulating the PI3K/Akt signaling pathways in the gut or indirectly through metabolites and SCFAs to provide changes in the gut and brain [227,228,229]. Multiple probiotic strains have been shown to promote the survival and growth of existing neurons and have positive effects on learning, memory, and cognition through increased BDNF levels, neuropeptides, or neurotransmitters in animal models [230,231,232,233].
Further strengthening the role of the gut microbiota in the stress response, probiotic supplement feeding has shown promising results for restoring and normalizing levels of gut microbiota bacteria and strengthening gut barrier function. Studies highlighting the ability of probiotics to recuse stress-related deficiencies in behavioural and biological parameters are reviewed in Cryan et al. [187] and shown in Figure 1. As seen in Table 3, probiotics can rescue deficits in the gut caused by both MS and RIS. Gut microbiota alterations and dysbiosis, increased adhesion, permeability, and penetration of the gut barrier, and the disrupted Firmicutes-to-Bacteroides ratio in the gut were all rescued in models supplemented with Lactobacillus [177,179,204,208]. Levels of peripheral neuroinflammatory cytokines, notably IL-6, which have been observed to be increased in stress animals, can be normalized through probiotic ingestion [197,202]. Neuroinflammation both in the CNS and PNS is improved after feeding [177].
Probiotic supplementation has also been shown to improve AD pathologies, as reviewed by Peterson et al. [234]. Multiple studies on AD patients show promising therapeutic effects on AD progression, including improved learning and memory, decreased oxidative stress, modified insulin resistance, improved behavioural performance on visual-spatial and executive/language tasks, decreased inflammation, mitochondrial dysfunction, gut barrier permeability, and DNA damage [234,235]. These effects are repeated and in some cases strengthened when combined with herbs or selenium treatment [236,237].

5. Conclusions

Taken together, stress paradigms in animal studies are shown to affect AD development and gut microbiota makeup, causing dysbiosis. The order in which these events occur is unclear; however, the relationship between the two cannot be overlooked. Further strengthening the role of the gut microbiota in the stress response, probiotic supplement feeding has shown promising results for restoring and normalizing levels of gut microbiota bacteria and strengthening gut barrier function. Combined with this, studies using probiotic supplementation have also been shown to improve AD pathologies [233]. Additionally, enrichment may be useful as a complement to other treatment methods in AD, helping to reduce cognitive symptoms and possibly slow pathological progression and potentially reverse stress-induced gut dysbiosis, which in turn could lead to further improvement. Given that AD is often comorbid with other neuropsychiatric disorders, including depression, bipolar disorder, and schizophrenia as outlined by Garcez and colleagues [238], enrichment may also help better those comorbidities or lessen their severity alongside AD. Combining enrichment (cognitive and exercise) and gut health can provide an optimal lifestyle that can serve as a beneficial strategy to combat stress and AD.
Additional studies using 16sRNA sequencing are needed in this field to establish a clear picture of which gut bacteria have a direct effect on AD development and the order in which these changes occur. Additionally, higher importance must be placed on sex differences in gut microbiota stress and enrichment studies. Overall, the understanding of the roles enrichment, stress, and gut dysbiosis has in AD development provides a possibility for non-invasive therapeutic potential through both enrichment and probiotic supplementation. Behaviorual intervention and gut microbiota manipulation could provide exciting new avenues to slow, stop, or prevent AD progression in both pre-clinical and prodromal stages.

Author Contributions

S.E.T., C.M.F. and Q.Y. conceived the idea; S.E.T., C.M.F. and T.L.K. wrote the manuscript; S.E.T., C.M.F. and Q.Y. edited the final version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a Canadian Institutes of Health Research Project Fund (PJT-169197) awarded to Q.Y., and S.E.T. was supported by an Aging Research Centre Newfoundland and Labrador (ARC-NL) fellowship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

List of Abbreviations

β-amyloid
AD Alzheimer’s disease
BBB blood–brain barrier
CUSchronic unpredictable stress
CRFcorticotrophin-releasing factor
EE environmental enrichment
GI gastrointestinal tract
GF germ free
HS hormonal stress
HPAhypothalamic–pituitary–adrenal
IBS irritable bowel syndrome
LPS lipopolysaccharides
MCImild cognitive impairment
MS maternal separation
NFT neurofibrillary tangles
ptau phosphorylated tau
PD postnatal day
PE physical enrichment
PS psychosocial stress
RIS restraint and immobility stress
SCFAshort-chain fatty acids
SS sleep restriction stress
SE social enrichment

References

  1. Alzheimer’s Association. What Is Alzheimer’s Disease? Alzheimer’s Disease and Dementia. 2023. Available online: https://www.alz.org/alzheimers-dementia/what-is-alzheimers (accessed on 2 March 2023).
  2. Alzheimer Society of Canada. What Is Alzheimer’s disease? Alzheimer Society of Canada. 2023. Available online: https://alzheimer.ca/en/about-dementia/what-alzheimers-disease (accessed on 2 March 2023).
  3. Weller, J.; Budson, A. Current understanding of Alzheimer’s disease diagnosis and treatment. F1000 Res. 2018, 7, 1161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Dickson, D.W. The Pathogenesis of Senile Plaques. J. Neuropathol. Exp. Neurol. 1997, 56, 321–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Selkoe, D.J. Alzheimer’s disease: Genes, Proteins, and Therapy. Physiol. Rev. 2001, 81, 741–766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Ossenkoppele, R.; van der Kant, R.; Hansson, O. Tau biomarkers in Alzheimer’s disease: Towards implementation in clinical practice and trials. Lancet Neurol. 2022, 21, 726–734. [Google Scholar] [CrossRef] [PubMed]
  7. Crous-Bou, M.; Minguillón, C.; Gramunt, N.; Molinuevo, J.L. Alzheimer’s disease prevention: From risk factors to early intervention. Alzheimer’s Res. Ther. 2017, 9, 71. [Google Scholar] [CrossRef] [Green Version]
  8. Schneider, L.S.; Mangialasche, F.; Andreasen, N.; Feldman, H.; Giacobini, E.; Jones, R.; Mantua, V.; Mecocci, P.; Pani, L.; Winbald, B.; et al. Clinical trials and late-stage drug development for Alzheimer’s disease: An appraisal from 1984 to 2014. J. Intern. Med. 2015, 275, 251–283. [Google Scholar] [CrossRef]
  9. Congdon, E.E.; Sigurdsson, E.M. Tau-targeting therapies for Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 399–415. [Google Scholar] [CrossRef]
  10. Soeda, Y.; Saito, M.; Maeda, S.; Ishida, K.; Nakamura, A.; Kojima, S.; Takashima, A. Methylene Blue Inhibits Formation of Tau Fibrils but not of Granular Tau Oligomers: A Plausible Key to Understanding Failure of a Clinical Trial for Alzheimer’s disease. J. Alzheimer’s Dis. JAD 2019, 68, 1677–1686. [Google Scholar] [CrossRef]
  11. Jadhav, S.; Avila, J.; Schöll, M.; Kovacs, G.G.; Kövari, E.; Skrabana, R.; Evans, L.D.; Kontsekova, E.; Malawska, B.; de Silva, R.; et al. A walk through tau therapeutic strategies. Acta Neuropathol. Commun. 2019, 7, 22. [Google Scholar] [CrossRef]
  12. Varidaki, A.; Hong, Y.; Coffey, E.T. Repositioning Microtubule Stabilizing Drugs for Brain Disorders. Front. Cell. Neurosci. 2018, 12, 226. [Google Scholar] [CrossRef]
  13. Ozben, T.; Ozben, S. Neuro-inflammation and anti-inflammatory treatment options for Alzheimer’s disease. Clin. Biochem. 2019, 72, 87–89. [Google Scholar] [CrossRef] [PubMed]
  14. Blennow, K.; de Leon, M.J.; Zetterberg, H. Alzheimer’s disease. Lancet 2006, 368, 387–403. [Google Scholar] [CrossRef] [PubMed]
  15. Cacace, R.; Sleegers, K.; Van Broeckhoven, C. Molecular genetics of early-onset Alzheimer’s disease revisited. Alzheimer’s Dement. 2016, 12, 733–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Gatz, M.; Reynolds, C.A.; Fratiglioni, L.; Johansson, B.; Mortimer, J.A.; Berg, S.; Fiske, A.; Pedersen, N.L. Role of Genes and Environments for Explaining Alzheimer Disease. Arch. Gen. Psychiatry 2006, 63, 168–174. [Google Scholar] [CrossRef] [Green Version]
  17. Wingo, T.S.; Lah, J.J.; Levey, A.I.; Cutler, D.J. Autosomal Recessive Causes Likely in Early-Onset Alzheimer Disease. Arch. Neurol. 2012, 69, 59–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Sotiropoulos, I.; Catania, C.; Pinto, L.G.; Silva, R.; Pollerberg, G.E.; Takashima, A.; Sousa, N.; Almeida, O.F.X. Stress Acts Cumulatively to Precipitate Alzheimer’s disease-Like Tau Pathology and Cognitive Deficits. J. Neurosci. 2011, 31, 7840–7847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Dodt, C.; Dittman, J.; Hruby, J.; Späth-Schwalbe, E.; Born, J.; Schüttler, R.; Fehm, H.L. Cortisol Secretion in Young, Mentally Healthy Elderly and Patients with Senile Dementia of Alzheimer’s Type. J. Clin. Endocrinol. Metab. 1991, 72, 272–276. [Google Scholar] [CrossRef]
  20. Caruso, A.; Nicoletti, F.; Mango, D.; Saidi, A.; Orlando, R.; Scaccianoce, S. Stress as risk factor for Alzheimer’s disease. Pharmacol. Res. 2018, 132, 130–134. [Google Scholar] [CrossRef]
  21. Budas, G.; Coughlan, C.M.; Seckl, J.R.; Breen, K.C. The effect of corticosteroids on amyloid beta precursor protein/amyloid precursor-like protein expression and processing in vivo. Neurosci. Lett. 1999, 276, 61–64. [Google Scholar] [CrossRef]
  22. Green, K.N.; Billings, L.M.; Roozendaal, B.; McGaugh, J.L.; LaFerla, F.M. Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer’s disease. J. Neurosci. 2006, 26, 9047–9056. [Google Scholar] [CrossRef] [Green Version]
  23. Justice, N.J. The relationship between stress and Alzheimer’s disease. Neurobiol. Stress 2018, 8, 127–133. [Google Scholar] [CrossRef] [PubMed]
  24. Escher, C.M.; Sannemann, L.; Jessen, F. Stress and Alzheimer’s disease. J. Neural Transm. 2019, 126, 1155–1161. [Google Scholar] [CrossRef] [PubMed]
  25. Wilson, R.S.; Evans, D.A.; Bienias, J.L.; Mendes de Leon, C.F.; Schneider, J.A.; Bennett, D.A. Proneness to psychological distress is associated with risk of Alzheimer’s disease. Neurology 2003, 61, 1479–1485. [Google Scholar] [CrossRef] [PubMed]
  26. Mendez, M.F. The relationship between anxiety and Alzheimer’s disease. J. Alzheimer’s Dis. Rep. 2021, 5, 171–177. [Google Scholar] [CrossRef]
  27. Xu, H.; Yang, R.; Dintica, C.; Qi, X.; Song, R.; Bennett, D.A.; Xu, W. Association of lifespan cognitive reserve indicator with the risk of mild cognitive impairment and its progression to dementia. Alzheimer’s Dement. 2020, 16, 873–882. [Google Scholar] [CrossRef]
  28. Wilson, R.S.; Barnes, L.L.; Aggarwal, N.T.; Boyle, P.A.; Hebert, L.E.; de Leon, C.F.M.; Evans, D.A. Cognitive activity and the cognitive morbidity of Alzheimer’s disease. Neurology 2010, 75, 990–996. [Google Scholar] [CrossRef] [Green Version]
  29. Pillai, J.A.; Hall, C.B.; Dickson, D.W.; Buschke, H.; Lipton, R.B.; Verghese, J. Association of Crossword Puzzle Participation with memory decline in persons who develop dementia. J. Int. Neuropsychol. Soc. 2011, 17, 1006–1013. [Google Scholar] [CrossRef] [Green Version]
  30. Hall, C.B.; Lipton, R.B.; Sliwinski, M.; Katz, M.J.; Derby, C.A.; Verghese, J. Cognitive activities delay onset of memory decline in persons who develop dementia. Neurology 2009, 73, 356–361. [Google Scholar] [CrossRef] [Green Version]
  31. Roe, C.M.; Mintun, M.A.; D’Angelo, G.; Xiong, C.; Grant, E.A.; Morris, J.C. Alzheimer disease and cognitive reserve. Arch. Neurol. 2008, 65, 1467. [Google Scholar] [CrossRef] [Green Version]
  32. Bassuk, S.S.; Glass, T.A.; Berkman, L.F. Social Disengagement and Incident Cognitive Decline in Community-Dwelling Elderly Persons. Ann. Intern. Med. 1999, 131, 165–173. [Google Scholar] [CrossRef]
  33. Wilson, R.S.; Krueger, K.R.; Arnold, S.E.; Schneider, J.A.; Kelly, J.F.; Barnes, L.L.; Tang, Y.; Bennett, D.A. Loneliness and risk of Alzheimer disease. Arch. Gen. Psychiatry 2007, 64, 234. [Google Scholar] [CrossRef] [Green Version]
  34. Friedman, E.M.; Hayney, M.S.; Love, G.D.; Urry, H.L.; Rosenkranz, M.A.; Davidson, R.J.; Singer, B.H.; Ryff, C.D. Social relationships, sleep quality, and interleukin-6 in aging women. Proc. Natl. Acad. Sci. USA 2005, 102, 18757–18762. [Google Scholar] [CrossRef]
  35. Yuan, Y.; Li, X.; Liu, W. Dance activity interventions targeting cognitive functioning in older adults with mild cognitive impairment: A meta-analysis. Front. Psychol. 2022, 13, 966675. [Google Scholar] [CrossRef] [PubMed]
  36. 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] [PubMed] [Green Version]
  37. Ahlskog, J.E.; Geda, Y.E.; Graff-Radford, N.R.; Peterson, R.C. Physical exercise as a preventive or disease-modifying treatment of dementia and brain aging. Mayo Clin. Proc. 2011, 86, 876–884. [Google Scholar] [CrossRef] [Green Version]
  38. Varesi, A.; Pierella, E.; Romeo, M.; Piccini, G.B.; Alfano, C.; Bjørklund, G.; Oppong, A.; Ricevuti, G.; Esposito, C.; Chirumbolo, S.; et al. The Potential Role of Gut Microbiota in Alzheimer’s disease: From Diagnosis to Treatment. Nutrients 2022, 14, 668. [Google Scholar] [CrossRef] [PubMed]
  39. Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. INDIA-FBP Group 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] [Green Version]
  40. 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] [Green Version]
  41. Zhuang, Z.Q.; Shen, L.L.; Li, W.W.; Fu, X.; Zeng, F.; Gui, L.; Lü, Y.; Cai, M.; Zhu, C.; Tan, Y.L.; et al. Gut Microbiota is altered in Patients with Alzheimer’s disease. J. Alzheimer’s Dis. JAD 2018, 63, 1337–1346. [Google Scholar] [CrossRef] [Green Version]
  42. Liu, P.; Wu, L.; Peng, G.; Han, Y.; Tang, R.; Ge, J.; Zhang, L.; Jia, L.; Yue, S.; Zhou, K.; et al. Altered microbiomes distinguish Alzheimer’s disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain Behav. Immun. 2019, 80, 633–643. [Google Scholar] [CrossRef]
  43. Weeks, S.R.; McAuliffe, C.L.; Durussel, D.; Pasquina, P.F. Physiological and psychological fatigue in extreme conditions: The military example. J. Inj. Funct. Rehabil. 2010, 2, 438–441. [Google Scholar] [CrossRef] [PubMed]
  44. Jacobs, J.M.; Cameron, K.L.; Bojescul, J.A. Lower extremity stress fractures in the military. Clin. Sports Med. 2014, 33, 591–613. [Google Scholar] [CrossRef] [PubMed]
  45. Henning, P.C.; Park, B.S.; Kim, J.S. Physiological decrements during sustained military operational stress. Mil. Med. 2011, 176, 991–997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Pasiakos, S.M.; Margolis, L.M.; Murphy, N.E.; McClung, H.L.; Martini, S.; Gundersen, Y.; Castellani, J.W.; Karl, J.P.; Teien, H.K.; Madslien, E.H.; et al. Effects of exercise mode, energy, and macronutrient interventions on inflammation during military training. Physiol. Rep. 2016, 4, e12820. [Google Scholar] [CrossRef] [Green Version]
  47. Sanchez, J.L.; Cooper, M.J.; Myers, C.A.; Cummings, J.F.; Vest, K.G.; Russell, K.L.; Sanchez, J.L.; Hiser, M.J.; Gaydos, C.A. Respiratory Infections in the U.S. Military: Recent Experience and Control. Clin. Microbiol. Rev. 2015, 28, 743–800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Lyte, M.; Ernst, S. Catecholamine induced growth of gram negative bacteria. Life Sci. 1992, 50, 203–212. [Google Scholar] [CrossRef] [PubMed]
  49. Ulrich-Lai, Y.M.; Herman, J.P. Neural regulation of endocrine and autonomic stress responses. Nat. Rev. Neurosci. 2009, 10, 397–409. [Google Scholar] [CrossRef] [Green Version]
  50. Galley, J.D.; Bailey, M.T. Impact of stressor exposure on the interplay between commensal microbiota and host inflammation. Gut Microbes 2014, 5, 390–396. [Google Scholar] [CrossRef] [Green Version]
  51. van Wijck, K.; Lenaerts, K.; Grootjans, J.; Wijnands, K.A.; Poeze, M.; van Loon, L.J.; Dejong, C.H.; Buurman, W.A. Physiology and pathophysiology of splanchnic hypoperfusion and intestinal injury during exercise: Strategies for evaluation and prevention. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G155–G168. [Google Scholar] [CrossRef] [Green Version]
  52. Karl, J.P.; Margolis, L.M.; Madslien, E.H.; Murphy, N.E.; Castellani, J.W.; Gundersen, Y.; Hoke, A.V.; Levangie, M.W.; Kumar, R.; Chakraborty, N.; et al. Changes in intestinal microbiota composition and metabolism coincide with increased intestinal permeability in young adults under prolonged physiological stress. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 312, G559–G571. [Google Scholar] [CrossRef] [Green Version]
  53. Knowles, S.R.; Nelson, E.A.; Palombo, E.A. Investigating the role of perceived stress on bacterial flora activity and salivary cortisol secretion: A possible mechanism underlying susceptibility to illness. Biol. Psychol. 2008, 77, 132–137. [Google Scholar] [CrossRef] [PubMed]
  54. Kato-Kataoka, A.; Nishida, K.; Takada, M.; Kawai, M.; Kikuchi-Hayakawa, H.; Suda, K.; Ishikawa, H.; Gondo, Y.; Shimizu, K.; Matsuki, T.; et al. Fermented Milk Containing Lactobacillus casei Strain Shirota Preserves the Diversity of the Gut Microbiota and Relieves Abdominal Dysfunction in Healthy Medical Students Exposed to Academic Stress. Appl. Environ. Microbiol. 2016, 82, 3649–3658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Doifode, T.; Giridharan, V.V.; Generoso, J.S.; Bhatti, G.; Collodel, A.; Schulz, P.E.; Forlenza, O.V.; Barichello, T. The impact of the microbiota-gut-brain axis on Alzheimer’s disease pathophysiology. Pharmacol. Res. 2021, 164, 105314. [Google Scholar] [CrossRef] [PubMed]
  56. Liew, A.K.Y.; Teo, C.H.; Soga, T. The Molecular Effects of Environmental. Enrichment on Alzheimer’s disease. Mol. Neurobiol. 2022, 59, 7095–7118. [Google Scholar] [CrossRef]
  57. Mohammadi, S.; Zandi, M.; Kataj, P.D.; Zandi, L.K. Chronic stress and Alzheimer’s disease. Biotechnol. Appl. Biochem. 2021, 69, 1451–1458. [Google Scholar] [CrossRef] [PubMed]
  58. Cao, M.; Hu, P.-P.; Zhang, Y.-L.; Yan, Y.-X.; Shields, C.B.; Zhang, Y.-P.; Hu, G.; Xiao, M. Enriched physical environment reverses spatial cognitive impairment of socially isolated APPswe/PS1dE9 transgenic mice before amyloidosis onset. CNS Neurosci. Ther. 2018, 24, 202–211. [Google Scholar] [CrossRef] [Green Version]
  59. Lahiani-Cohen, I.; Lourbopoulos, A.; Haber, E.; Rozenstein-Tsalkovich, L.; Abramsky, O.; Grigoriadis, N.; Rosenmann, H. Moderate Environmental Enrichment Mitigates Tauopathy in a Neurofibrillary Tangle Mouse Model. J. Neuropathol. Exp. Neurol. 2011, 70, 610–621. [Google Scholar] [CrossRef] [Green Version]
  60. Prado Lima, M.G.; Schimidt, H.L.; Garcia, A.; Daré, L.R.; Carpes, F.P.; Izquierdo, I.; Mello-Carpes, P.B. Environmental enrichment and exercise are better than social enrichment to reduce memory deficits in amyloid beta neurotoxicity. Proc. Natl. Acad. Sci. USA 2018, 115, 2403–2409. [Google Scholar] [CrossRef] [Green Version]
  61. Rodríguez, J.J.; Noristani, H.N.; Olabarria, M.; Fletcher, J.; Somerville, T.D.D.; Yeh, C.Y.; Verkhratsky, A. Voluntary running and environmental enrichment restores impaired hippocampal neurogenesis in a triple transgenic mouse model of Alzheimer’s disease. Curr. Alzheimer Res. 2011, 8, 707–717. [Google Scholar] [CrossRef]
  62. Torraville, S. Understanding Differential Roles of Stress and Enrichment in Pathogenesis of Alzheimer’s Disease in a Novel Rat Tau Model. Master’s Thesis, Memorial University of Newfoundland, St. John, NL, Canada, 2022. Publication No. 15659. MUN Research Repository. Available online: http://research.library.mun.ca/id/eprint/15659 (accessed on 31 October 2022).
  63. Valero, J.; España, J.; Parra-Damas, A.; Martín, E.; Rodríguez-Álvarez, J.; Saura, C.A. Short-term environmental enrichment rescues adult neurogenesis and memory deficits in APP(Sw,Ind) transgenic mice. PLoS ONE 2011, 6, e16832. [Google Scholar] [CrossRef] [Green Version]
  64. Yeung, S.T.; Martinez-Coria, H.; Ager, R.R.; Rodriguez-Ortiz, C.J.; Baglietto-Vargas, D.; LaFerla, F.M. Repeated cognitive stimulation alleviates memory impairments in an Alzheimer’s disease mouse model. Brain Res. Bull. 2015, 117, 10–15. [Google Scholar] [CrossRef] [PubMed]
  65. 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]
  66. Mirochnic, S.; Wolf, S.; Staufenbiel, M.; Kempermann, G. Age Effects on the Regulation of Adult Hippocampal Neurogenesis by Physical Activity and Environmental Enrichment in the APP23 Mouse Model of Alzheimer Disease. Hippocampus 2009, 19, 1008–1018. [Google Scholar] [CrossRef] [PubMed]
  67. Rodríguez, J.M.; Murphy, K.; Stanton, C.; Ross, R.P.; Kober, O.I.; Juge, N.; Avershina, E.; Rudi, K.; Narbad, A.; Jenmalm, M.C.; et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb. Ecol. Health Dis. 2015, 26, 26050. [Google Scholar] [CrossRef]
  68. Robison, L.S.; Francis, N.; Popescu, D.L.; Anderson, M.E.; Hatfield, J.; Xu, F.; Anderson, B.J.; Van Nostrand, W.E.; Robinson, J.K. Environmental Enrichment: Disentangling the Influence of Novelty, Social, and Physical Activity on Cerebral Amyloid Angiopathy in a Transgenic Mouse Model. Int. J. Mol. Sci. 2020, 21, 843. [Google Scholar] [CrossRef] [Green Version]
  69. Hu, Y.-S.; Xu, P.; Pigino, G.; Brady, S.T.; Larson, J.; Lazarov, O. Complex environment experience rescues impaired neurogenesis, enhances synaptic plasticity, and attenuates neuropathology in familial Alzheimer’s disease-linked APPswe/PS1ΔE9 mice. J. Fed. Am. Soc. Exp. Biol. 2010, 24, 1667–1681. [Google Scholar] [CrossRef]
  70. Herring, A.; Ambrée, O.; Tomm, M.; Habermann, H.; Sachser, N.; Paulus, W.; Keyvani, K. Environmental enrichment enhances cellular plasticity in transgenic mice with Alzheimer-like pathology. Exp. Neurol. 2009, 216, 184–192. [Google Scholar] [CrossRef]
  71. Salmin, V.V.; Komleva, Y.K.; Kuvacheva, N.V.; Morgun, A.V.; Khilazheva, E.D.; Lopatina, O.L.; Pozhilenkova, E.A.; Shapovalov, K.A.; Uspenskaya, Y.A.; Salmina, A.B. Differential Roles of Environmental Enrichment in Alzheimer’s Type Neurodegeneration and Physiological Aging. Front. Aging Neurosci. 2017, 9, 245. [Google Scholar] [CrossRef] [Green Version]
  72. Ziegler-Waldkirch, S.; d’Errico, P.; Sauer, J.-F.; Erny, D.; Savanthrapadian, S.; Loreth, D.; Katzmarski, N.; Blank, T.; Bartos, M.; Prinz, M.; et al. Seed-induced Aβ deposition is modulated by microglia under environmental enrichment in a mouse model of Alzheimer’s disease. EMBO J. 2018, 37, 167–182. [Google Scholar] [CrossRef]
  73. Jeong, Y.H.; Kim, J.M.; Yoo, J.; Lee, S.H.; Kim, H.-S.; Suh, Y.-H. Environmental enrichment compensates for the effects of stress on disease progression in Tg2576 mice, an Alzheimer’s disease model. J. Neurochem. 2011, 119, 1282–1293. [Google Scholar] [CrossRef]
  74. Stazi, M.; Wirths, O. Physical activity and cognitive stimulation ameliorate learning and motor deficits in a transgenic mouse model of Alzheimer’s disease. Behav. Brain Res. 2021, 397, 112951. [Google Scholar] [CrossRef] [PubMed]
  75. Berardi, N.; Braschi, C.; Capsoni, S.; Cattaneo, A.; Maffei, L. Environmental Enrichment Delays the Onset of Memory Deficits and Reduces Neuropathological Hallmarks in a Mouse Model of Alzheimer-Like Neurodegeneration. J. Alzheimer’s Dis. 2007, 11, 359–370. [Google Scholar] [CrossRef] [PubMed]
  76. Xu, H.; Gelyana, E.; Rajsombath, M.; Yang, T.; Li, S.; Selkoe, D. Environmental Enrichment Potently Prevents Microglia-Mediated Neuroinflammation by Human Amyloid β-Protein Oligomers. J. Neurosci. 2016, 36, 9041–9056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Balthazar, J.; Schöwe, N.M.; Cipolli, G.C.; Buck, H.S.; Viel, T.A. Enriched Environment Significantly Reduced Senile Plaques in a Transgenic Mice Model of Alzheimer’s Disease, Improving Memory. Front. Aging Neurosci. 2018, 10, 288. [Google Scholar] [CrossRef] [Green Version]
  78. Cattaud, V.; Bezzina, C.; Rey, C.C.; Lejards, C.; Dahan, L.; Verret, L. Early disruption of parvalbumin expression and perineuronal nets in the hippocampus of the Tg2576 mouse model of Alzheimer’s disease can be rescued by enriched environment. Neurobiol. Aging 2018, 72, 147–158. [Google Scholar] [CrossRef] [PubMed]
  79. Li, J.-Z.; Hao, X.-H.; Wu, H.-P.; Li, M.; Liu, X.-M.; Wu, Z.-B. An enriched environment delays the progression from mild cognitive impairment to Alzheimer’s disease in senescence-accelerated mouse prone 8 mice. Exp. Ther. Med. 2021, 22, 1320. [Google Scholar] [CrossRef]
  80. Song, M.K.; Kim, Y.J.; Lee, J.-M.; Kim, Y.-J. Neurovascular integrative effects of long-term environmental enrichment on chronic hypoperfusion rat model. Brain Res. Bull. 2020, 163, 160–169. [Google Scholar] [CrossRef]
  81. Griñán-Ferré, C.; Izquierdo, V.; Otero, E.; Puigoriol-Illamola, D.; Corpas, R.; Sanfeliu, C.; Ortuño-Sahagún, D.; Pallàs, M. Environmental Enrichment Improves Cognitive Deficits, AD Hallmarks and Epigenetic Alterations Presented in 5xFAD Mouse Model. Front. Cell. Neurosci. 2018, 12, 224. [Google Scholar] [CrossRef] [Green Version]
  82. Lazarov, O.; Robinson, J.; Tang, Y.-P.; Hairston, I.S.; Korade-Mirnics, Z.; Lee, V.M.-Y.; Hersh, L.B.; Sapolsky, R.M.; Mirnics, K. Environmental Enrichment Reduces Aβ Levels and Amyloid Deposition in Transgenic Mice. Cell 2005, 120, 701–713. [Google Scholar] [CrossRef] [Green Version]
  83. Costa, D.A.; Cracchiolo, J.R.; Bachstetter, A.D.; Hughes, T.F.; Bales, K.R.; Paul, S.M.; Mervis, R.F.; Arendash, G.W.; Potter, H. Enrichment improves cognition in AD mice by amyloid-related and unrelated mechanisms. Neurobiol. Aging 2007, 28, 831–844. [Google Scholar] [CrossRef]
  84. Selvi, Y.; Gergerlioglu, H.S.; Akbaba, N.; Oz, M.; Kandeger, A.; Demir, E.A.; Yerlikaya, F.H.; Nurullahoglu-Atalikm, K.E. Impact of enriched environment on production of tau, amyloid precursor protein and, amyloid-β peptide in high-fat and high-sucrose-fed rats. Acta Neuropsychiatr. 2016, 29, 291–298. [Google Scholar] [CrossRef] [PubMed]
  85. Maesako, M.; Uemura, K.; Kubota, M.; Kuzuya, A.; Sasaki, K.; Asada, M.; Watanabe, K.; Hayashida, N.; Ihara, M.; Ito, H.; et al. Environmental enrichment ameliorated high-fat diet-induced Aβ deposition and memory deficit in APP transgenic mice. Neurobiol. Aging 2012, 33, 1011.e11–1011.e23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Cracchiolo, J.R.; Mori, T.; Nazian, S.J.; Tan, J.; Potter, H.; Arendash, G.W. Enhanced cognitive activity—Over and above social or physical activity—Is required to protect Alzheimer’s mice against cognitive impairment, reduce Aβ deposition, and increase synaptic immunoreactivity. Neurobiol. Learn. Mem. 2007, 88, 277–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Arranz, L.; De Castro, N.M.; Baeza, I.; Giménez-Llort, L.; De la Fuente, M. Effect of Environmental Enrichment on the Immunoendocrine Aging of Male and Female Triple-Transgenic 3xTg-AD Mice for Alzheimer’s disease. J. Alzheimer’s Dis. 2011, 25, 727–737. [Google Scholar] [CrossRef]
  88. Polito, L.; Chierchia, A.; Tunesi, M.; Bouybayoune, I.; Kehoe, P.G.; Albani, D.; Forloni, G. Environmental Enrichment Lessens Cognitive Decline in APP23 Mice Without Affecting Brain Sirtuin Expression. J. Alzheimer’s Dis. 2014, 42, 851–864. [Google Scholar] [CrossRef]
  89. Verret, L.; Krezymon, A.; Halley, H.; Trouche, S.; Zerwas, M.; Lazouret, M.; Lassalle, J.-M.; Rampon, C. Transient enriched housing before amyloidosis onset sustains cognitive improvement in Tg2576 mice. Neurobiol. Aging 2013, 34, 211–225. [Google Scholar] [CrossRef]
  90. Ziegler-Waldkirch, S.; Marksteiner, K.; Stoll, J.; d’Errico, P.; Friesen, M.; Eiler, D.; Neudel, L.; Sturn, V.; Opper, I.; Datta, M.; et al. Environmental enrichment reverses Aβ pathology during pregnancy in a mouse model of Alzheimer’s disease. Acta Neuropathol. Commun. 2018, 6, 44. [Google Scholar] [CrossRef] [Green Version]
  91. Liu, Y.; Cao, L.; Zhang, X.; Liang, Y.; Xu, Y.; Zhu, C. Memantine Differentially Regulates Tau Phosphorylation Induced by Chronic Restraint Stress of Varying Duration in Mice. Neural Plast. 2019, 2019, 4168472. [Google Scholar] [CrossRef]
  92. Torres-Lista, V.; Giménez-Llort, L. Early postnatal handling and environmental enrichment improve the behavioral responses of 17-month-old 3xTg-AD and non-transgenic mice in the Forced Swim Test in a gender-dependent manner. Behav. Process. 2015, 120, 120–127. [Google Scholar] [CrossRef]
  93. Cañete, T.; Blázquez, G.; Tobeňa, A.; Giménez-Llort, L.; Fernández-Teruel, A. Cognitive and emotional alterations in young Alzheimer’s disease (3xTgAD) mice: Effects of neonatal handling stimulation and sexual dimorphism. Behav. Brain Res. 2015, 281, 156–171. [Google Scholar] [CrossRef]
  94. Liao, K.; Liu, D.; Zhu, L.-Q. Enriched odor exposure decrease tau phosphorylation in the rat hippocampus and cortex. Neurosci. Lett. 2012, 507, 22–26. [Google Scholar] [CrossRef] [PubMed]
  95. Stuart, K.E.; King, A.E.; Fernandez-Martos, C.M.; Dittmann, J.; Summers, M.J.; Vickers, J.C. Mid-life environmental enrichment increases synaptic density in CA1 in a mouse model of Aβ-associated pathology and positively influences synaptic and cognitive health in healthy aging. J. Comp. Neurol. 2017, 525, 1797–1810. [Google Scholar] [CrossRef] [PubMed]
  96. Nakano, M.; Kubota, K.; Hashizume, S.; Kobayashi, E.; Chikenji, T.S.; Saito, Y.; Fujimiya, M. An enriched environment prevents cognitive impairment in an Alzheimer’s disease model by enhancing the secretion of exosomal microRNA-146a from the choroid plexus. Brain Behav. Immun. Health 2020, 9, 100149. [Google Scholar] [CrossRef] [PubMed]
  97. Pietropaolo, S.; Feldon, J.; Yee, B.K. Environmental enrichment eliminates the anxiety phenotypes in a triple transgenic mouse model of Alzheimer’s disease. Cogn. Affect. Behav. Neurosci. 2014, 14, 996–1008. [Google Scholar] [CrossRef] [PubMed]
  98. Görtz, N.; Lewejohann, L.; Tomm, M.; Ambrée, O.; Keyvani, K.; Paulus, W.; Sachser, N. Effects of environmental enrichment on exploration, anxiety, and memory in female TgCRND8 Alzheimer mice. Behav. Brain Res. 2008, 191, 43–48. [Google Scholar] [CrossRef]
  99. Hüttenrauch, M.; Walter, S.; Kaufmann, M.; Weggen, S.; Wirths, O. Limited Effects of Prolonged Environmental Enrichment on the Pathology of 5xFAD Mice. Mol. Neurobiol. 2016, 54, 6542–6555. [Google Scholar] [CrossRef]
  100. Stuart, K.E.; King, A.E.; King, N.E.; Collins, J.M.; Vickers, J.C.; Ziebell, J.M. Late-life environmental enrichment preserves short-term memory and may attenuate microglia in male APP/PS1 mice. Neuroscience 2019, 408, 282–292. [Google Scholar] [CrossRef]
  101. Cotel, M.-C.; Jawhar, S.; Christensen, D.Z.; Bayer, T.A.; Wirths, O. Environmental enrichment fails to rescue working memory deficits, neuron loss, and neurogenesis in APP/PS1K1 mice. Neurobiol. Aging 2012, 33, 96–107. [Google Scholar] [CrossRef]
  102. Stozicka, Z.; Korenova, M.; Uhrinova, I.; Cubinkova, V.; Cente, M.; Kovacech, B.; Babindakova, N.; Matyasova, K.; Vargova, G.; Novak, M.; et al. Environmental Enrichment Rescues Functional Deficit and Alters Neuroinflammation in Transgenic Model of Tauopathy. J. Alzheimer’s Dis. 2020, 74, 951–964. [Google Scholar] [CrossRef]
  103. Arrendash, G.W.; Garcia, M.F.; Costa, D.A.; Cracchiolo, J.R.; Wefes, I.M.; Potter, H. Environmental enrichment improves cognition in aged Alzheimer’s transgenic mice despite stable β-amyloid deposition. Neuro Rep. 2004, 15, 1751–1754. [Google Scholar] [CrossRef]
  104. Beauquis, J.; Pavía, P.; Pomilio, C.; Vinuesa, A.; Podlutskaya, N.; Galvan, V.; Saravia, F. Environmental enrichment prevents astroglial pathological changes in the hippocampus of APP transgenic mice, model of Alzheimer’s disease. Exp. Neurol. 2013, 239, 28–37. [Google Scholar] [CrossRef] [PubMed]
  105. Blázquez, G.; Cañete, T.; Tobeña, A.; Giménez-Llort, L.; Fernández-Teruel, A. Cognitive and emotional profiles of aged Alzheimer’s disease (3xTgAD) mice: Effects of environmental enrichment and sexual dimorphism. Behav. Brain Res. 2014, 268, 185–201. [Google Scholar] [CrossRef] [PubMed]
  106. Mate, V.; Smolek, T.; Kazmerova, Z.V.; Jadhav, S.; Brezovakova, V.; Jurkanin, B.; Uhrinova, I.; Basheer, N.; Zilka, N.; Katina, S.; et al. Enriched environment ameliorates propagation of tau pathology and improves cognition in rat model of tauopathy. Front. Aging Neurosci. 2022, 14, 935973. [Google Scholar] [CrossRef]
  107. Jankowsky, J.L.; Xu, G.; Fromholt, D.; Gonzales, V.; Borchelt, D.R. Environmental Enrichment Exacerbates Amyloid Plaque Formation in a Transgenic Mouse Model of Alzheimer Disease. J. Neuropathol. Exp. Neurol. 2003, 62, 1220–1227. [Google Scholar] [CrossRef]
  108. Levi, O.; Michaelson, D.M. Environmental enrichment stimulates neurogenesis in apolipoprotein E3 and neuronal apoptosis in apolipoprotein E4 transgenic mice. J. Neurochem. 2006, 100, 202–210. [Google Scholar] [CrossRef] [PubMed]
  109. Jankowsky, J.L.; Melnikova, T.; Fadale, D.J.; Xu, G.M.; Slunt, H.H.; Gonzales, V.; Younkin, L.H.; Younkin, S.G.; Borchelt, D.R.; Savonenko, A.V. Environmental Enrichment Mitigates Cognitive Deficits in a Mouse Model of Alzheimer’s disease. J. Neurosci. 2005, 25, 5217–5224. [Google Scholar] [CrossRef] [Green Version]
  110. Montarolo, F.; Parolisi, R.; Hoxha, E.; Boda, E.; Tempia, F. Early Enriched Environment Exposure Protects Spatial Memory and Accelerates Amyloid Plaque Formation in APPSwe/PS1L166P Mice. Public Libr. Sci. One 2013, 8, e69381. [Google Scholar] [CrossRef] [Green Version]
  111. Fulopova, B.; Stuart, K.E.; Bennett, W.; Bindoff, A.; King, A.E.; Vickers, J.C.; Canty, A.J. Regional differences in beta amyloid plaque deposition and variable response to midlife environmental enrichment in the cortex of APP/PS1 mice. J. Comp. Neurol. 2021, 529, 1849–1862. [Google Scholar] [CrossRef]
  112. Huang, Y.; Huang, X.; Zhang, L.; Han, F.; Pang, K.-L.; Li, X.; Shen, J.-Y. Magnesium boosts the memory restorative effect of environmental enrichment in Alzheimer’s disease mice. CNS Neurosci. Ther. 2018, 24, 70–79. [Google Scholar] [CrossRef] [Green Version]
  113. Zhu, G.; Guo, M.; Zhao, J.; Zhang, H.; Wang, G.; Chen, W. Bifidobacterium breve intervention combined with environmental enrichment alleviates cognitive impairment by regulating the gut microbiota and microbial metabolites in Alzheimer’s disease mice. Front. Immunol. 2022, 13, 1013664. [Google Scholar] [CrossRef]
  114. Dong, J.; Zhou, M.; Wu, X.; Du, M.; Wang, X. Memantine combined with environmental enrichment improves spatial memory and alleviates Alzheimer’s disease-like pathology in senescence-accelerated prone-8 (SAMP8) mice. J. Biomed. Res. 2012, 26, 439–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Gholami, J.; Nagah, S.S.; Rajabian, A.; Saburi, E.; Hajali, V. The effect of combination pretreatment of donepezil and environmental enrichment on memory deficits in amyloid-beta-induced Alzheimer-like rat model. Biochem. Biophys. Rep. 2022, 32, 101392. [Google Scholar] [CrossRef] [PubMed]
  116. Stazi, M.; Zampar, S.; Klafki, H.-W.; Meyer, T.; Wirths, O. A Combination of Caffeine Supplementation and Enriched Environment in an Alzheimer’s disease Mouse Model. Int. J. Mol. Sci. 2023, 24, 2155. [Google Scholar] [CrossRef] [PubMed]
  117. Muhammad, M.S.; Ayo, J.O.; Danjuma, N.M.; AbdulWahab, A.; Isa, A.S.; Umar, A.H. Modulation of Memory and Neurochemical Changes by Resveratrol and Environmental Enrichment in Rodent Model of Alzheimer’s disease. Niger. J. Physiol. Sci. 2022, 37, 59–67. [Google Scholar] [CrossRef]
  118. Liu, E.; Zhou, Q.; Xie, A.-J.; Li, M.; Zhang, S.; Huang, H.; Liuyang, Z.; Wang, Y.; Liu, B.; Li, X.; et al. Enriched gestation activates the IGF pathway to evoke embryo-adult benefits to prevent Alzheimer’s disease. Transl. Neurodegener. 2019, 8, 8. [Google Scholar] [CrossRef]
  119. Campbell, S.N.; Zhang, C.; Monte, L.; Roe, A.D.; Rice, K.C.; Taché, Y.; Masliah, E.; Rissman, R.A. Increased Tau Phosphorylation and Aggregation in the Hippocampus of Mice Overexpressing Corticotropin-Releasing Factor. J. Alzheimer’s Dis. 2015, 43, 967–976. [Google Scholar] [CrossRef] [PubMed]
  120. Carroll, J.C.; Iba, M.; Bangasser, D.A.; Valentino, R.J.; James, M.J.; Brunden, K.R.; Lee, V.M.-Y.; Trojanowski, J.Q. Chronic Stress Exacerbates Tau Pathology, Neurodegeneration, and Cognitive Performance through a Corticotropin-Releasing Factor Receptor-Dependent Mechanism in a Transgenic Mouse Model of Tauopathy. J. Neurosci. 2011, 31, 14436–14449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Devi, L.; Alldred, M.J.; Ginsberg, S.D.; Ohno, M. Sex- and brain region-specific acceleration of b-amyloidogenesis following behavioral stress in a mouse model of Alzheimer’s disease. Mol. Brain 2010, 3, 34. [Google Scholar] [CrossRef] [Green Version]
  122. Han, B.; Wang, J.-H.; Geng, Y.; Shen, L.; Wang, H.-L.; Wang, Y.-Y.; Wang, M.-W. Chronic Stress Contributes to Cognitive Dysfunction and Hippocampal Metabolic Abnormalities in APP/PS1 Mice. Cell. Physiol. Biochem. 2017, 41, 1766–1776. [Google Scholar] [CrossRef]
  123. Joshi, Y.B.; Chu, J.; Praticò, D. Stress Hormone Leads to Memory Deficits and Altered Tau Phosphorylation in a Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2012, 31, 167–176. [Google Scholar] [CrossRef]
  124. Justice, N.J.; Huang, L.; Tian, J.-B.; Cole, A.; Pruski, M.; Hunt, A.J.J.; Flores, R.; Zhu, M.X.; Arenkiel, B.R.; Zheng, H. Posttraumatic Stress Disorder-Like Induction Elevates b-Amyloid Levels, Which Directly Activates Corticotropin-Releasing Factor Neurons to Exacerbate Stress Responses. J. Neurosci. 2015, 35, 2612–2623. [Google Scholar] [CrossRef] [Green Version]
  125. Rothman, S.M.; Herdener, N.; Frankola, K.A.; Mughal, M.R.; Mattson, M.P. Chronic mild sleep restriction accentuates contextual memory impairments, and accumulations of cortical Ab and pTau in a mouse model of Alzheimer’s disease. Brain Res. 2013, 1529, 200–208. [Google Scholar] [CrossRef] [Green Version]
  126. Srivareerat, M.; Tran, T.T.; Alzoubi, K.H.; Alkadhi, K.A. Chronic Psychosocial Stress Exacerbates Impairment of Cognition and Long-Term Potentiation in b-Amyloid Rat Model of Alzheimer’s disease. Biol. Psychiatry 2009, 65, 918–926. [Google Scholar] [CrossRef]
  127. Dong, H.; Yuede, C.M.; Yoo, H.-S.; Martin, M.V.; Deal, C.; Mace, A.G.; Csernansky, J.G. Corticosterone and Related Receptor Expression are Associated with Increased β-Amyloid Plaques in Isolated Tg2576 Mice. Neuroscience 2008, 155, 154–163. [Google Scholar] [CrossRef] [Green Version]
  128. Lee, K.-W.; Kim, J.-B.; Seo, J.-S.; Kim, T.-K.; Im, J.-Y.; Baek, I.-S.; Kim, K.-S.; Lee, J.-K.; Han, P.-L. Behavioral stress accelerates plaque pathogenesis in the brain of Tg2576 mice via generation of metabolic oxidative stress. J. Neurochem. 2008, 108, 165–175. [Google Scholar] [CrossRef] [PubMed]
  129. AbdAlla, S.; el Hakim, A.; Abdelbaset, A.; Elfaramawy, Y.; Quitterer, U. Inhibition of ACE Retards Tau Hyperphosphorylation and Signs of Neuronal Degeneration in Aged Rats Subjected to Chronic Mild Stress. BioMed Res. Int. 2015, 2015, 917156. [Google Scholar] [CrossRef] [Green Version]
  130. Brice, K.N.; Hagen, C.W.; Peterman, J.L.; Figg, J.W.; Braden, P.N.; Chumley, M.J.; Boehm, G.W. Chronic sleep restriction increases soluble hippocampal Aβ-42 and impairs cognitive performance. Physiol. Behav. 2020, 226, 113128. [Google Scholar] [CrossRef] [PubMed]
  131. Chen, L.; Huang, J.; Yang, L.; Zeng, X.-A.; Zhang, Y.; Wang, X.; Chen, M.; Li, X.; Zhang, Y.; Zhang, M. Sleep deprivation accelerates the progression of Alzheimer’s disease by influencing Aβ-related metabolism. Neurosci. Lett. 2017, 650, 146–152. [Google Scholar] [CrossRef] [PubMed]
  132. Wu, C.; Gong, W.-G.; Wang, Y.-J.; Sun, J.-J.; Zhou, H.; Zhang, Z.-J.; Ren, Q.-G. Escitalopram alleviates stress-induced Alzheimer’s disease-like tau pathologies and cognitive deficits by reducing HPA-axis reactivity and insulin/GSK-3β signal pathway activity. Neurobiol. Aging 2018, 67, 137–147. [Google Scholar] [CrossRef]
  133. Yang, C.; Guo, X.; Wang, G.H.; Wang, H.L.; Liu, Z.C.; Liu, H.; Zhu, Z.X.; Li, Y. Changes in tau phosphorylation levels in the hippocampus and frontal cortex following chronic stress. Braz. J. Med. Biol. Res. 2014, 47, 237–244. [Google Scholar] [CrossRef] [Green Version]
  134. Yang, Y.; Wang, X.; Xiao, A.; Han, J.; Wang, Z.; Wen, M. Ketogenic diet prevents chronic sleep deprivation-induced Alzheimer’s disease by inhibiting iron dyshomeostasis and promoting repair via Sirt1/Nrf2 pathway. Front. Aging Neurosci. 2022, 14, 998292. [Google Scholar] [CrossRef]
  135. Cuadrado-Tejedor, M.; García-Osta, A. Chronic Mild Stress Assay Leading to Early Onset and Propagation of Alzheimer’s Disease Phenotype in Mouse Models. Syst. Biol. Alzheimer’s Dis. 2016, 1303, 241–246. [Google Scholar] [CrossRef]
  136. Cuadrado-Tejedor, M.; Ricobaraza, A.; Frechilla, D.; Franco, R.; Pérez-Mediavilla, A.; García-Osta, A. Chronic Mild Stress Accelerates the Onset and Progression of Alzheimer’s Disease Phenotype in Tg2576 Mice. J. Alzheimer’s Dis. 2012, 28, 567–578. [Google Scholar] [CrossRef] [Green Version]
  137. Wang, J.; Yuan, J.; Pang, J.; Ma, J.; Han, B.; Geng, Y.; Shen, L.; Wang, H.; Ma, Q.; Wang, Y.; et al. Effects of Chronic Stress on Cognition in Male SAMP8 Mice. Cell. Physiol. Biochem. 2016, 39, 1078–1086. [Google Scholar] [CrossRef] [PubMed]
  138. Cortese, A.; Delgado-Morales, R.; Almeida, O.F.X.; Romberg, C. The Arctic/Swedish APP mutation alters the impact of chronic stress on cognition in mice. Eur. J. Neurosci. 2019, 50, 2773–2785. [Google Scholar] [CrossRef] [Green Version]
  139. Han, B.; Yu, L.; Geng, Y.; Shen, L.; Wang, H.; Wang, Y.; Wang, J.; Wang, M. Chronic Stress Aggravates Cognitive Impairment and Suppresses Insulin Associated Signaling Pathway in APP/PS1 Mice. J. Alzheimer’s Dis. 2016, 53, 1539–1552. [Google Scholar] [CrossRef]
  140. Zhang, J.; Lin, L.; Dai, X.; Xiao, N.; Ye, Q.; Chen, X. ApoE4 increases susceptibility to stress-induced age-dependent depression-like behavior and cognitive impairment. J. Psychiatr. Res. 2021, 143, 292–301. [Google Scholar] [CrossRef] [PubMed]
  141. Yang, Y.; Zhao, S.; Yang, X.; Li, W.; Si, J.; Yang, X. The antidepressant potential of lactobacillus casei in the postpartum depression rat model mediated by the microbiota-gut-brain axis. Neurosci. Lett. 2022, 774, 136474. [Google Scholar] [CrossRef]
  142. Sethi, M.; Joshi, S.S.; Webb, R.L.; Beckett, T.L.; Donohue, K.D.; Murphy, M.P.; O’Hara, B.F.; Duncan, M.J. Increased fragmentation of sleep-wake cycles in the 5xFAD mouse model of Alzheimer’s disease. Neuroscience 2015, 290, 80–89. [Google Scholar] [CrossRef] [Green Version]
  143. Kincheski, G.C.; Valentim, I.S.; Clarke, J.R.; Cozachenco, D.; Castelo-Branco, M.T.L.; Ramos-Lobo, A.M.; Rumjanek, V.M.B.D.; Donato, J.J.; De Felice, F.G.; Ferreira, S.T. Chronic sleep restriction promotes brain inflammation and synapse loss, and potentiates memory impairment induced by amyloid-β oligomers in mice. Brain Behav. Immun. 2017, 64, 140–151. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, S.; Meng, Y.; Wang, N.; Lei, Q.; He, L.; Hong, X.; Li, Z. Disturbance of REM sleep exacerbates microglial activation in APP/PS1 mice. Neurobiol. Learn. Mem. 2023, 200, 107737. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, C.; Gao, W.-R.; Yin, J.; Wang, Z.-J.; Qi, J.-S.; Cai, H.-Y.; Wu, M.-N. Chronic sleep deprivation exacerbates cognitive and synaptic plasticity impairments in APP/PS1 transgenic mice. Behav. Brain Res. 2021, 412, 113400. [Google Scholar] [CrossRef] [PubMed]
  146. Holth, J.K.; Mahan, T.E.; Robinson, G.O.; Rocha, A.; Holtzman, D.M. Altered sleep and EEG power in the P301S Tau transgenic mouse model. Ann. Clin. Transl. Neurol. 2017, 4, 180–190. [Google Scholar] [CrossRef]
  147. Liu, S.; Lei, Q.; Liu, Y.; Zhang, X.; Li, Z. Acoustic Stimulation Improves Memory and Reverses the Contribution of Chronic Sleep Deprivation to Pathology in 3xTgAD Mice. Brain Sci. 2022, 12, 1509. [Google Scholar] [CrossRef] [PubMed]
  148. Di Meco, A.; Joshi, Y.B.; Praticò, D. Sleep deprivation impairs memory, tau metabolism, and synaptic integrity of a mouse model of Alzheimer’s disease with plaques and tangles. Neurobiol. Aging 2014, 35, 1813–1820. [Google Scholar] [CrossRef]
  149. Roh, J.H.; Jiang, H.; Finn, M.B.; Stewart, F.R.; Mahan, T.E.; Cirrito, J.R.; Heda, A.; Snider, B.J.; Li, M.; Yanagisawa, M.; et al. Potential role of orexin and sleep modulation in the pathogenesis of Alzheimer’s disease. J. Exp. Med. 2014, 211, 2487–2496. [Google Scholar] [CrossRef]
  150. Kang, J.-E.; Lim, M.M.; Bateman, R.J.; Lee, J.J.; Smyth, L.P.; Cirrito, J.R.; Fujiki, N.; Nishino, S.; Holtzman, D.M. Amyloid-β Dynamics are Regulated by Orexin and the Sleep-Wake Cycle. Science 2009, 326, 1005–1007. [Google Scholar] [CrossRef] [Green Version]
  151. Qiu, H.; Zhong, R.; Liu, H.; Zhang, F.; Li, S.; Le, W. Chronic Sleep Deprivation Exacerbates Learning-Memory Disability and Alzheimer’s Disease-Like Pathologies in AβPPSWE/PS1ΔE9 Mice. J. Alzheimer’s Dis. 2016, 50, 669–685. [Google Scholar] [CrossRef]
  152. Minakawa, E.N.; Miyazaki, K.; Maruo, K.; Yagihara, H.; Fujita, H.; Wada, K.; Nagai, Y. Chronic sleep fragmentation exacerbates amyloid β deposition in Alzheimer’s disease model mice. Neurosci. Lett. 2017, 653, 362–369. [Google Scholar] [CrossRef]
  153. Niu, L.; Zhang, F.; Xu, X.; Yang, Y.; Li, S.; Liu, H.; Le, W. Chronic sleep deprivation altered the expression of circadian clock genes and aggravated Alzheimer’s disease neuropathology. Brain Pathol. 2021, 32, e13028. [Google Scholar] [CrossRef]
  154. Sierksma, A.S.R.; Prickaerts, J.; Chouliaras, L.; Rostamian, S.; Delbroek, L.; Rutten, B.P.F.; Steinbusch, H.W.M.; van den Hove, D.L.A. Behavioral and neurobiological effects of prenatal stress exposure in male and female APPswe/PS1dE9 mice. Neurobiol. Aging 2013, 34, 319–337. [Google Scholar] [CrossRef] [PubMed]
  155. Wang, Y.; Zhu, T.; Wang, M.; Zhang, F.; Zhang, G.; Zhao, J.; Zhang, Y.; Wu, E.; Li, X. Icariin Attenuates M1 Activation of Microglia and Aβ Plaque Accumulation in the Hippocampus and Prefrontal Cortex by Up-Regulating PPARγ in Restraint/Isolation-Stressed APP/PS1 Mice. Front. Neurosci. 2019, 13, 291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Kunimoto, S.; Nakamura, S.; Wada, K.; Inoue, T. Chronic stress-mutated presenilin 1 gene interaction perturbs neurogenesis and accelerates neurodegeneration. Exp. Neurol. 2010, 221, 175–185. [Google Scholar] [CrossRef] [PubMed]
  157. Kang, J.-E.; Cirrito, J.R.; Dong, H.; Holtzman, D.M. Acute stress increases intersticial fluid amyloid-β via corticotropin-releasing factor and neuronal activity. Proc. Natl. Acad. Sci. USA 2007, 104, 10673–10678. [Google Scholar] [CrossRef]
  158. Perez Nievas, B.G.; Hammerschmidt, T.; Kummer, M.P.; Terwel, D.; Leza, J.C.; Heneka, M.T. Restraint stress increases neuroinflammation independently of amyloid β levels in amyloid precursor protein/PS1 transgenic mice. J. Neurochem. 2010, 116, 43–52. [Google Scholar] [CrossRef]
  159. Joshi, Y.B.; Giannopoulos, P.F.; Chu, J.; Sperow, M.; Kirby, L.G.; Abood, M.E.; Praticò, D. Absence of ALOX5 gene prevents stress-induced memory deficits, synaptic dysfunction and tauopathy in a mouse model of Alzheimer’s disease. Hum. Mol. Genet. 2014, 23, 6894–6902. [Google Scholar] [CrossRef] [Green Version]
  160. Jeong, Y.H.; Park, C.H.; Yoo, J.; Shin, K.Y.; Ahn, S.-M.; Kim, H.-S.; Lee, S.H.; Emson, P.C.; Suh, Y.H. Chronic stress accelerates learning and memory impairments and increases amyloid deposition in APPV7171-CT100 transgenic mice, an Alzheimer’s disease mom. FASEB J. 2006, 20, 587–802. [Google Scholar] [CrossRef] [Green Version]
  161. Vicens, P.; Heredia, L.; Torrente, M.; Domingo, J.L. Behavioural effects of PNU-282987 and stress in an animal model of Alzheimer’s disease. Psychogeriatrics 2006, 17, 33–42. [Google Scholar] [CrossRef]
  162. Yuan, Q.; Su, H.; Chau, W.-H.; Ng, C.T.; Huang, J.-D.; Wu, W.; Lin, Z.-X. Behavioral Stress Fails to Accelerate the Onset and Progression of Plaque Pathology in the Brain of a Mouse Model of Alzheimer’s disease. Public Libr. Sci. One 2013, 8, e53480. [Google Scholar] [CrossRef] [Green Version]
  163. Pentkowski, N.S.; Bouquin, S.J.; Maestas-Olguin, C.R.; Villasenor, Z.M.; Clark, B.J. Differential effects of chronic stress on anxiety-like behavior and contextual fear conditioning in the TgF344-AD rat model of Alzheimer’s disease. Behav. Brain Res. 2022, 418, 113661. [Google Scholar] [CrossRef]
  164. Hui, J.; Feng, G.; Zheng, C.; Jin, H.; Jia, N. Maternal separation exacerbates Alzheimer’s disease-like behavioral and pathological changes in adult APPswe/PS1dE9 mice. Behav. Brain Res. 2017, 318, 18–23. [Google Scholar] [CrossRef]
  165. Tanaka, T.; Hirai, S.; Hosokawa, M.; Saito, T.; Sakuma, H.; Saido, T.; Hasegawa, M.; Okado, H. Early-life stress induces the development of Alzheimer’s disease pathology via angiopathy. Exp. Neurol. 2021, 337, 113552. [Google Scholar] [CrossRef]
  166. Bachiller, S.; Hidalgo, I.; Garcia, M.G.; Boza-Serrano, A.; Paulus, A.; Denis, Q.; Haikal, C.; Manouchehrian, O.; Klementieva, O.; Li, J.Y.; et al. Early-life stress elicits peripheral and brain immune activation differently in wild type and 5xFAD mice in a sex-specific manner. J. Neuroinflammation 2022, 19, 151. [Google Scholar] [CrossRef]
  167. de Jonge, W.J. The gut’s little brain in control of intestinal immunity. Int. Sch. Res. Not. 2013, 2013, 630159. [Google Scholar] [CrossRef] [Green Version]
  168. Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [Green Version]
  169. Sommer, F.; Bäckhed, F. The gut microbiota—Masters of host development and physiology. Nat. Rev. Microbiol. 2013, 11, 227–238. [Google Scholar] [CrossRef]
  170. Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.N.; Kubo, C.; Koga, Y. Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. J. Physiol. 2004, 558, 263–275. [Google Scholar] [CrossRef] [PubMed]
  171. Ackerman, S.H.; Hofer, M.A.; Weiner, H. Early maternal separation increases gastric ulcer risk in rats by producing a latent thermoregulatory disturbance. Science 1978, 201, 373–376. [Google Scholar] [CrossRef] [PubMed]
  172. Borre, Y.E.; O’Keeffe, G.W.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Microbiota and neurodevelopmental windows: Implications for brain disorders. Trends Mol. Med. 2014, 20, 509–518. [Google Scholar] [CrossRef] [PubMed]
  173. Deng, Y.; Zhou, M.; Wang, J.; Yao, J.; Yu, J.; Liu, W.; Wu, L.; Wang, J.; Gao, R. Involvement of the microbiota-gut-brain axis in chronic restraint stress: Disturbances of the kynurenine metabolic pathway in both the gut and brain. Gut Microbes 2021, 13, 1869501. [Google Scholar] [CrossRef]
  174. 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] [PubMed] [Green Version]
  175. Zhou, K. Strategies to promote abundance of Akkermansia muciniphila, an emerging probiotics in the gut, evidence from dietary intervention studies. J. Funct. Foods 2017, 33, 194–201. [Google Scholar] [CrossRef] [PubMed]
  176. De Palma, G.; Blennerhassett, P.; Lu, J.; Deng, Y.; Park, A.J.; Green, W.; Denou, E.; Silva, M.A.; Santacruz, A.; Sanz, Y.; et al. Microbiota and host determinants of behavioural phenotype in maternally separated mice. Nat. Commun. 2015, 6, 7735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Moya-Pérez, A.; Perez-Villalba, A.; Benítez-Páez, A.; Campillo, I.; Sanz, Y. Bifidobacterium CECT 7765 modulates early stress-induced immune neuroendocrine and behavioral alterations in mice. Brain Behav. Immun. 2017, 65, 43–56. [Google Scholar] [CrossRef]
  178. Desbonnet, L.; Garrett, L.; Clarke, G.; Kiely, B.; Cryan, J.F.; Dinan, T.G. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience 2010, 170, 1179–1188. [Google Scholar] [CrossRef]
  179. Cowan, C.S.; Callaghan, B.L.; Richardson, R. The effects of a probiotic formulation (Lactobacillus rhamnosus and L. helveticus) on developmental trajectories of emotional learning in stressed infant rats. Transl. Psychiatry 2016, 6, e823. [Google Scholar] [CrossRef] [Green Version]
  180. Zhou, X.Y.; Li, M.; Li, X.; Long, X.; Zuo, X.L.; Hou, X.H.; Cong, Y.Z.; Li, Y.Q. Visceral hypersensitive rats share common dysbiosis features with irritable bowel syndrome patients. World J. Gastroenterol. 2016, 22, 5211–5227. [Google Scholar] [CrossRef]
  181. Lingpeng, P.; Jingzhu, S.; Wei, L.; Enqi, W.; Yaqin, L. Effect of water extracts from Cynanchum thesioides (Freyn) K. Schum. On visceral hypersensitivity and gut microbiota profile in maternally separated rats. J. Ethnopharmacol. 2021, 264, 113352. [Google Scholar] [CrossRef]
  182. Dodiya, H.B.; Forsyth, C.B.; Voigt, R.M.; Engen, P.A.; Patel, J.; Shaikh, M.; Green, S.J.; Naqib, A.; Roy, A.; Kordower, J.H.; et al. Chronic stress-induced gut dysfunction exacerbates Parkinson’s disease phenotype and pathology in a rotenone-induced mouse model of Parkinson’s disease. Neurobiol. Dis. 2020, 135, 104352. [Google Scholar] [CrossRef]
  183. Ji, S.; Han, S.; Yu, L.; Du, L.; You, Y.; Chen, J.; Wang, M.; Wu, S.; Li, S.; Sun, X.; et al. Jia Wei Xiao Yao San ameliorates chronic stress-induced depression-like behaviors in mice by regulating the gut microbiome and brain metabolome in relation to purine metabolism. Phytomedicine 2022, 98, 153940. [Google Scholar] [CrossRef]
  184. Vorvul, A.O.; Bobyntsev, I.I.; Medvedeva, O.A.; Mukhina, A.Y.; Svishcheva, M.V.; Azarova, I.E.; Andreeva, L.A.; Myasoedov, N.F. ACTH (6-9)-Pro-Gly-Pro ameliorates anxiety-like and depressive-like behaviour and gut mucosal microbiota composition in rats under conditions of chronic restraint stress. Neuropeptides 2022, 93, 102247. [Google Scholar] [CrossRef] [PubMed]
  185. Li, J.; Li, Y.; Duan, W.; Zhao, Z.; Yang, L.; Wei, W.; Li, J.; Li, Y.; Yu, Y.; Dai, B.; et al. Shugan granule contributes to the improvement of depression-like behaviors in chronic restraint stress-stimulated rats by altering gut microbiota. CNS Neurosci. Ther. 2022, 28, 1409–1424. [Google Scholar] [CrossRef]
  186. Medel-Matus, J.S.; Shin, D.; Dorfman, E.; Sankar, R.; Mazarati, A. Facilitation of kindling epileptogenesis by chronic stress may be mediated by intestinal microbiome. Epilepsia Open 2018, 3, 290–294. [Google Scholar] [CrossRef]
  187. 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] [PubMed]
  188. Xiao, Q.; Shu, R.; Wu, C.; Tong, Y.; Xiong, Z.; Zhou, J.; Yu, C.; Xie, X.; Fu, Z. Crocin-I alleviates the depression-like behaviors probably via modulating “microbiota-gut-brain” axis in mice exposed to chronic restraint stress. J. Affect. Disord. 2020, 276, 476–486. [Google Scholar] [CrossRef] [PubMed]
  189. Xu, X.; Zhan, G.; Chen, R.; Wang, D.; Guan, S.; Xu, H. Gut microbiota and its role in stress-induced hyperalgesia: Gender-specific responses linked to different changes in serum metabolites. Pharmacol. Res. 2022, 177, 106129. [Google Scholar] [CrossRef]
  190. Gispen-de Wied, C.C.; D’Haenen, H.; Verhoeven, W.M.; Wynne, H.J.; Westenberg, H.G.; Thijssen, J.H.; Van Ree, J.M. Inhibition of the pituitary-adrenal axis with dexamethasone and cortisol in depressed patients and healthy subjects: A dose-response study. Psychoneuroendocrinology 1993, 18, 191–204. [Google Scholar] [CrossRef]
  191. Barreau, F.; Cartier, C.; Leveque, M.; Ferrier, L.; Moriez, R.; Laroute, V.; Rosztoczy, A.; Fioramonti, J.; Bueno, L. Pathways involved in gut mucosal barrier dysfunction induced in adult rats by maternal deprivation: Corticotropin-releasing factor and nerve growth factor interplay. J. Physiol. 2007, 580 Pt 1, 347–356. [Google Scholar] [CrossRef]
  192. la Fleur, S.E.; Wick, E.C.; Idumalla, P.S.; Grady, E.F.; Bhargava, A. Role of peripheral corticotropin-releasing factor and urocortin II in intestinal inflammation and motility in terminal ileum. Proc. Natl. Acad. Sci. USA 2005, 102, 7647–7652. [Google Scholar] [CrossRef]
  193. Plotsky, P.M.; Thrivikraman, K.V.; Meaney, M.J. Central and feedback regulation of hypothalamic corticotropin-releasing factor secretion. Ciba Found. Symp. 1993, 172, 59–84. [Google Scholar] [CrossRef]
  194. Chatzaki, E.; Charalampopoulos, I.; Leontidis, C.; Mouzas, I.A.; Tzardi, M.; Tsatsanis, C.; Margioris, A.N.; Gravanis, A. Urocortin in human gastric mucosa: Relationship to inflammatory activity. J. Clin. Endocrinol. Metab. 2003, 88, 478–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Kohno, M.; Kawahito, Y.; Tsubouchi, Y.; Hashiramoto, A.; Yamada, R.; Inoue, K.I.; Kusaka, Y.; Kubo, T.; Elenkov, I.J.; Chrousos, G.P.; et al. Urocortin expression in synovium of patients with rheumatoid arthritis and osteoarthritis: Relation to inflammatory activity. J. Clin. Endocrinol. Metab. 2001, 86, 4344–4352. [Google Scholar] [CrossRef] [PubMed]
  196. Bailey, M.T.; Dowd, S.E.; Galley, J.D.; Hufnagle, A.R.; Allen, R.G.; Lyte, M. Exposure to a social stressor alters the structure of the intestinal microbiota: Implications for stressor-induced immunomodulation. Brain Behav. Immun. 2011, 25, 397–407. [Google Scholar] [CrossRef] [Green Version]
  197. Liu, Y.W.; Liu, W.H.; Wu, C.C.; Juan, Y.C.; Wu, Y.C.; Tsai, H.P.; Wang, S.; Tsai, Y.C. Psychotropic effects of Lactobacillus plantarum PS128 in early life-stressed and naïve adult mice. Brain Res. 2016, 1631, 1–12. [Google Scholar] [CrossRef] [Green Version]
  198. Kuti, D.; Winkler, Z.; Horváth, K.; Juhász, B.; Paholcsek, M.; Stágel, A.; Gulyás, G.; Czeglédi, L.; Ferenczi, S.; Kovács, K.J. Gastrointestinal (non-systemic) antibiotic rifaximin differentially affects chronic stress-induced changes in colon microbiome and gut permeability without effect on behavior. Brain Behav. Immun. 2020, 84, 218–228. [Google Scholar] [CrossRef]
  199. Amini-Khoei, H.; Haghani-Samani, E.; Beigi, M.; Soltani, A.; Mobini, G.R.; Balali-Dehkordi, S.; Haj-Mirzaian, A.; Rafieian-Kopaei, M.; Alizadeh, A.; Hojjati, M.R.; et al. On the role of corticosterone in behavioral disorders, microbiota composition alteration and neuroimmune response in adult male mice subjected to maternal separation stress. Int. Immunopharmacol. 2019, 66, 242–250. [Google Scholar] [CrossRef]
  200. Ilchmann-Diounou, H.; Olier, M.; Lencina, C.; Riba, A.; Barretto, S.; Nankap, M.; Sommer, C.; Guillou, H.; Ellero-Simatos, S.; Guzylack-Piriou, L.; et al. Early life stress induces type 2 diabetes-like features in ageing mice. Brain Behav. Immun. 2019, 80, 452–463. [Google Scholar] [CrossRef] [PubMed]
  201. Riba, A.; Olier, M.; Lacroix-Lamandé, S.; Lencina, C.; Bacquié, V.; Harkat, C.; Van Langendonck, N.; Gillet, M.; Cartier, C.; Baron, M.; et al. Early life stress in mice is a suitable model for Irritable Bowel Syndrome but does not predispose to colitis nor increase susceptibility to enteric infections. Brain Behav. Immun. 2018, 73, 403–415. [Google Scholar] [CrossRef] [Green Version]
  202. Gareau, M.G.; Jury, J.; MacQueen, G.; Sherman, P.M.; Perdue, M.H. Probiotic treatment of rat pups normalizes corticosterone release and ameliorates colonic dysfunction induced by maternal separation. Gut 2007, 56, 1522–1528. [Google Scholar] [CrossRef] [Green Version]
  203. Dandekar, M.P.; Palepu, M.S.K.; Satti, S.; Jaiswal, Y.; Singh, A.A.; Dash, S.P.; Gajula, S.N.R.; Sonti, R. Multi-strain Probiotic Formulation Reverses Maternal Separation and Chronic Unpredictable Mild Stress-Generated Anxiety- and Depression-like Phenotypes by Modulating Gut Microbiome-Brain Activity in Rats. ACS Chem. Neurosci. 2022, 13, 1948–1965. [Google Scholar] [CrossRef]
  204. Enqi, W.; Jingzhu, S.; Lingpeng, P.; Yaqin, L. Comparison of the Gut Microbiota Disturbance in Rat Models of Irritable Bowel Syndrome Induced by Maternal Separation and Multiple Early-Life Adversity. Front. Cell. Infect. Microbiol. 2021, 10, 581974. [Google Scholar] [CrossRef]
  205. Park, H.J.; Kim, S.A.; Kang, W.S.; Kim, J.W. Early-Life Stress Modulates Gut Microbiota and Peripheral and Central Inflammation in a Sex-Dependent Manner. Int. J. Mol. Sci. 2021, 22, 1899. [Google Scholar] [CrossRef]
  206. Egerton, S.; Donoso, F.; Fitzgerald, P.; Gite, S.; Fouhy, F.; Whooley, J.; Dinan, T.G.; Cryan, J.F.; Culloty, S.C.; Ross, R.P.; et al. Investigating the potential of fish oil as a nutraceutical in an animal model of early life stress. Nutr. Neurosci. 2022, 25, 356–378. [Google Scholar] [CrossRef]
  207. Fukui, H.; Oshima, T.; Tanaka, Y.; Oikawa, Y.; Makizaki, Y.; Ohno, H.; Tomita, T.; Watari, J.; Miwa, H. Effect of probiotic Bifidobacterium bifidum G9-1 on the relationship between gut microbiota profile and stress sensitivity in maternally separated rats. Sci. Rep. 2018, 8, 12384. [Google Scholar] [CrossRef] [PubMed]
  208. Rincel, M.; Darnaudéry, M. Maternal separation in rodents: A journey from gut to brain and nutritional perspectives. Proc. Nutr. Soc. 2019, 79, 113–132. [Google Scholar] [CrossRef]
  209. Chen, T.; Wang, R.; Duan, Z.; Yuan, X.; Ding, Y.; Feng, Z.; Bu, F.; Liu, L.; Wang, Q.; Zhou, J.; et al. Akkermansia muciniphila Protects Against Psychological Disorder-Induced Gut Microbiota-Mediated Colonic Mucosal Barrier Damage and Aggravation of Colitis. Front. Cell. Infect. Microbiol. 2021, 11, 723856. [Google Scholar] [CrossRef]
  210. Lin, S.; Li, Q.; Jiang, S.; Xu, Z.; Jiang, Y.; Liu, L.; Jiang, J.; Tong, Y.; Wang, P. Crocetin ameliorates chronic restraint stress-induced depression-like behaviors in mice by regulating MEK/ERK pathways and gut microbiota. J. Ethnopharmacol. 2021, 268, 113608. [Google Scholar] [CrossRef] [PubMed]
  211. Kang, A.; Zhang, S.; Zhu, D.; Dong, Y.; Shan, J.; Xie, T.; Wen, H.; Di, L. Gut microbiota in the pharmacokinetics and colonic deglycosylation metabolism of ginsenoside Rb1 in rats: Contrary effects of antimicrobials treatment and restraint stress. Chem. Biol. Interact. 2016, 258, 187–196. [Google Scholar] [CrossRef] [PubMed]
  212. Gareau, M.G.; Wine, E.; Rodrigues, D.M.; Cho, J.H.; Whary, M.T.; Philpott, D.J.; Macqueen, G.; Sherman, P.M. Bacterial infection causes stress-induced memory dysfunction in mice. Gut 2011, 60, 307–317. [Google Scholar] [CrossRef]
  213. Lin, L.; Zheng, L.J.; Zhang, L.J. Neuroinflammation, gut microbiome, and Alzheimer’s disease. Mol. Neurobiol. 2018, 55, 8243–8250. [Google Scholar] [CrossRef] [PubMed]
  214. Pellegrini, C.; Fornai, M.; D’Antongiovanni, V.; Antonioli, L.; Bernardini, N.; Derkinderen, P. The intestinal barrier in disorders of the central nervous system. Lancet Gastroenterol. Hepatol. 2023, 8, 66–80. [Google Scholar] [CrossRef] [PubMed]
  215. Bailey, M.T.; Coe, C.L. Maternal separation disrupts the integrity of the intestinal microflora in infant rhesus monkeys. Dev. Psychobiol. 1999, 35, 146–155. [Google Scholar] [CrossRef]
  216. Liang, S.; Wang, T.; Hu, X.; Luo, J.; Li, W.; Wu, X.; Jin, F. Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience 2015, 310, 561–577. [Google Scholar] [CrossRef] [PubMed]
  217. Sayas, C.L.; Avila, J. GSK-3 and tau: A key duet in Alzheimer’s disease. Cells 2021, 10, 721. [Google Scholar] [CrossRef]
  218. 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] [PubMed]
  219. Generoso, J.S.; Giridharan, V.V.; Lee, J.; Macedo, D.; Barichello, T. The role of the microbiota-gut-brain axis in neuropsychiatric disorders. Rev. Bras. De Psiquiatr. 2021, 43, 293–305. [Google Scholar] [CrossRef]
  220. Zhao, Y.; Lukiw, W.J. Microbiome-generated amyloid and potential impact on amyloidogenesis in Alzheimer’s disease (AD). J. Nat. Sci. 2015, 1, e138. [Google Scholar]
  221. McKernan, D.P.; Nolan, A.; Brint, E.K.; O’Mahony, S.M.; Hyland, N.P.; Cryan, J.F.; Dinan, T.G. Toll-like receptor mRNA expression is selectively increased in the colonic mucosa of two animal models relevant to irritable bowel syndrome. PLoS ONE 2009, 4, e8226. [Google Scholar] [CrossRef] [Green Version]
  222. Sanders, M.E.; Heimbach, J.T.; Pot, B.; Tancredi, D.J.; Lenoir-Wijnkoop, I.; Lahteenmaki-Uutela, A.; Gueimonde, M.; Bañares, S. Health claims substantiation for probiotic and prebiotic products. Gut Microbes 2011, 2, 127–133. [Google Scholar] [CrossRef] [Green Version]
  223. Smith, A.P.; Sutherland, D.; Hewlett, P. An Investigation of the Acute Effects of Oligofructose-Enriched Inulin on Subjective Wellbeing, Mood and Cognitive Performance. Nutrients 2015, 7, 8887–8896. [Google Scholar] [CrossRef]
  224. Xue, L.; He, J.; Gao, N.; Lu, X.; Li, M.; Wu, X.; Liu, Z.; Jin, Y.; Liu, J.; Xu, J.; et al. Probiotics may delay the progression of nonalcoholic fatty liver disease by restoring the gut microbiota structure and improving intestinal endotoxemia. Sci. Rep. 2017, 7, 45176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693 Pt B, 128–133. [Google Scholar] [CrossRef]
  226. Arseneault-Bréard, J.; Rondeau, I.; Gilbert, K.; Girard, S.A.; Tompkins, T.A.; Godbout, R.; Rousseau, G. Combination of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 reduces post-myocardial infarction depression symptoms and restores intestinal permeability in a rat model. Br. J. Nutr. 2012, 107, 1793–1799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Tillisch, K.; Labus, J.; Kilpatrick, L.; Jiang, Z.; Stains, J.; Ebrat, B.; Guyonnet, D.; Legrain-Raspaud, S.; Trotin, B.; Naliboff, B.; et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013, 144, 1394–1401. [Google Scholar] [CrossRef] [Green Version]
  228. Flynn, C.M.; Yuan, Q. Probiotic supplement as a promising strategy in early tau pathology prevention: Focusing on GSK-3β? Front. Neurosci. 2023, 17, 1159314. [Google Scholar] [CrossRef]
  229. Savignac, H.M.; Corona, G.; Mills, H.; Chen, L.; Spencer, J.P.; Tzortzis, G.; Burnet, P.W. Prebiotic feeding elevates central brain derived neurotrophic factor, N-methyl-D-aspartate receptor subunits and D-serine. Neurochem. Int. 2013, 63, 756–764. [Google Scholar] [CrossRef] [Green Version]
  230. O’Sullivan, E.; Barrett, E.; Grenham, S.; Fitzgerald, P.; Stanton, C.; Ross, R.P.; Quigley, E.M.; Cryan, J.F.; Dinan, T.G. BDNF expression in the hippocampus of maternally separated rats: Does Bifidobacterium breve 6330 alter BDNF levels? Benef. Microbes 2011, 2, 199–207. [Google Scholar] [CrossRef]
  231. Roy Sarkar, S.; Banerjee, S. Gut microbiota in neurodegenerative disorders. J. Neuroimmunol. 2019, 328, 98–104. [Google Scholar] [CrossRef]
  232. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [PubMed]
  233. Peterson, C.T. Dysfunction of the Microbiota-Gut-Brain Axis in Neurodegenerative Disease: The Promise of Therapeutic Modulation with Prebiotics, Medicinal Herbs, Probiotics, and Synbiotics. J. Evid. Based Integr. Med. 2020, 25, 2515690x20957225. [Google Scholar] [CrossRef]
  234. Ton, A.M.M.; Campagnaro, B.P.; Alves, G.A.; Aires, R.; Côco, L.Z.; Arpini, C.M.; Guerra, E.; Oliveira, T.; Campos-Toimil, M.; Meyrelles, S.S.; et al. Oxidative Stress and Dementia in Alzheimer’s Patients: Effects of Synbiotic Supplementation. Oxidative Med. Cell. Longev. 2020, 2020, 2638703. [Google Scholar] [CrossRef] [Green Version]
  235. Kapoor, R.; Srivastava, S.; Kakkar, P. Bacopa monnieri modulates antioxidant responses in brain and kidney of diabetic rats. Environ. Toxicol. Pharmacol. 2009, 27, 62–69. [Google Scholar] [CrossRef] [PubMed]
  236. Kuboyama, T.; Tohda, C.; Komatsu, K. Effects of Ashwagandha (roots of Withania somnifera) on neurodegenerative diseases. Biol. Pharm. Bull. 2014, 37, 892–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Bhattacharya, S.K.; Bhattacharya, A.; Kumar, A.; Ghosal, S. Antioxidant activity of Bacopa monniera in rat frontal cortex, striatum and hippocampus. Phytother. Res. PTR 2000, 14, 174–179. [Google Scholar] [CrossRef]
  238. Garcez, M.L.; Falchetti, A.C.B.; Mina, F.; Budni, J. Alzheimer’s disease associated with Psychiatric Comorbidities. Ann. Braz. Acad. Sci. 2015, 87, 1461–1473. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Effects of stress and probiotic manipulation on the gut microbiota and health.
Figure 1. Effects of stress and probiotic manipulation on the gut microbiota and health.
Biomedicines 11 01884 g001
Table 1. Studies examining the effect of enrichment on Alzheimer’s disease development and progression.
Table 1. Studies examining the effect of enrichment on Alzheimer’s disease development and progression.
Enrichment ParadigmAnimal ModelFindingsReference
Cognitive Enrichment3xTg-AD (both sexes—mouse)Repeated spatial learning training reduced cognitive decline in a separate spatial task[64]
Environmental EnrichmentE257K/P301S-Tau-Tg (both sexes—mice)Increased neurogenesis
Improved cognition
Reduced tau pathology
[59]
apoE4 (male—mouse)Improved neurogenesis in apoE3 controls
Worsened in apoE4 animals
[108]
APPswe/PS1ΔE9 (female—mice)Increased Aβ burden[109]
APPSw,Ind (female—mouse)Improved cognitive performance
Increased neurogenesis
[63]
APPswe/PS1ΔE9 (male—mice)Increased neurogenesis
Reduced Aβ and ptau
[69]
Environmental EnrichmentTgCRND8 (female—mouse)Increased neurogenesis
Reduced Aβ burden
[70]
Aβ1-42 infusion at CA1 (male—rat)Improved neurogenesis in Aβ animals[71]
5xFAD (male—mouse)Reversed cell death and reduced neurogenesis of Aβ seeded animals[72]
Tg4-42hom (both sexes—mouse)Preserved spatial and recognition memory[74]
AD11 (both sexes—mouse)Improved memory and reduced Aβ burden[75]
oAβ intraventricular injection (male—mouse)Prevented Aβ-mediated changes in microglia and inflammatory gene mRNA[76]
PDBFG-APPSwlnd (both sexes—mouse)Improved spatial memory
Reduced Aβ burden
[77]
Tg2576 (female—mouse)Reversed parvalbumin deficit[78]
SAMP8 (male—mouse)Increased spatial memory
Increased synaptic plasticity
Reduced apoptosis
Reduced Aβ burden
[79]
Chronic cerebral hypoperfusion (male—rat)Improved spatial memory
Reduced inflammation
[80]
5xFAD (female—mouse)Increased exploration
Reduced cognitive deficits
Reduced AD pathological markers
[81]
APPswe/PS1ΔE9 (male—mouse)Reduced Aβ deposition
Increased Aβ-degrading enzymes
[82]
PS1/PDAPP (both sexes—mouse)Increased cognitive performance
Reduced Aβ burden
[83]
APP23 (male—mouse)Improved spatial memory at 7–8 mo.
Reduced Aβ deposition by 18 mo.
[88]
High sucrose or high-fat diet (male—rat)High sucrose and fat diets increased AD pathology
Enrichment returned to control levels
[84]
APPSwe,Ind, high-fat diet (both sexes—mouse)High-fat diet worsened memory and pathology
Ameliorated by enrichment
[85]
Environmental EnrichmentTg2576 (female—mouse)Protective against cognitive decline and Aβ but did not rescue later[89]
3xTg-AD (both sexes—mouse)Improved immune system activity for males[87]
APPswe/PS1ΔE9 (male—mouse)Improved synaptic health but not cognitive abilities[95]
5xFAD (male—mouse)Prevented learning impairment
Increased synaptic plasticity
Reduced inflammation
[96]
3xTg-AD (female—mouse)Reduced anxiety-like behaviour[97]
TgCRND8 (female—mouse)Improved exploratory behaviour
Reduced anxiety-like behaviour
No memory improvement
[98]
5xFAD (female—mouse)Increased survival rate
Improved motor skills
No improvement of anxiety, Aβ, or inflammation
[99]
APPswe/PS1ΔE9 (male—mouse)Improved short-term spatial memory
Prevented increases in inflammation
[100]
APP/PS1KI (female—mouse)Mild improvement in motor skills
No changes to memory, Aβ, or neuronal loss
[101]
SHR72 (male—rat)Improved motor skills
Mildly improved inflammation
Did not influence ptau
[102]
APPsw (both sexes—mouse)Increased spatial memory despite continued Aβ deposition[103]
PDAPP-J20 (female—mouse)Reduced Aβ
Improved astroglial response toward plaques
[104]
3xTg-AD (both sexes—mouse)Mild improvements to spatial learning in advanced stages[105]
SHR72 (male—rat)Increased spatial memory performance
Reduced ptau in mild cases
[106]
APPswe/PS1ΔE9 (female—mouse)Improved spatial memory
Reduced Aβ deposition
[107]
APPSwe/PS1L166P (female—mouse)Reduced spatial memory impairment
Worsened Aβ deposition
[110]
APPswe/PS1ΔE9 (male—mouse)Improved spatial memory
Reduced Aβ in motor and sensory cortices
[111]
Physical EnrichmentAPPswe/PS1ΔE9 (male—mouse)Improved cognition
Reduced inflammation
[65]
Environmental and Physical EnrichmentAPP23 (female—mouse)At 18 mo. time point, both enrichment types reduced Aβ and increased neurogenesis[66]
3xTg-AD (both sexes—mouse)EE alone reduced inflammation
Combined EE and PE reduced to control levels
[67]
3xTg-AD (both sexes—mouse)Both enrichment types reversed deficits in neurogenesis[61]
Environmental, Physical, and Social EnrichmentAβ infusion at CA1 (male—rat)EE and PE prevent memory impairment
SE prevents social recognition impairment
[60]
APP/PS1 (both sexes—mouse)EE alone prevented cognitive impairment, reduced Aβ, and increased synaptic plasticity[86]
Environmental Enrichment with B. breveAβ1-42 infusion at hippocampus (male—mouse)Combined treatment rescues impaired cognitive performance[113]
Environmental Enrichment with MagnesiumAPPswe/PS1ΔE9 (female–mouse)EE alone improved short-term memory
Combined treatment with MgT improved long-term memory
[112]
Environmental Enrichment with ResveratrolAlCl3 (male—mouse)Both treatments individually reduced Aβ
Combined treatment worsened
[117]
Environmental Enrichment with CaffeineTg4-42 (both sexes—mouse)Combined treatment improved spatial memory, but did not add to individual effects of EE or Caf alone on motor skills, recognition, or neurogenesis[116]
Environmental Enrichment with DonepezilAβ1-42 infusion at hippocampus (male—rat)Combined treatment only was able to restore spatial memory and elevate BDNF to control levels[115]
Environmental Enrichment with MemantineSAMP8 (male—mouse)Both treatment options effective, but more so when combined, at improving spatial memory and reducing ptau and Aβ[114]
Gestational Enrichment3xTg-AD (both sexes—mouse)Offspring of enriched dams had preserved synaptic plasticity and memory[118]
5xFAD (female—mouse)While pregnancy and lactation worsen Aβ and neurogenesis, EE rescues to non-pregnant levels[90]
Neonatal Handling3xTg-AD (both sexes—mouse)Handling provided mild protective effects against spatial memory decline, especially in females[93]
3xTg-AD (both sexes—mouse)Handling improved performance in forced swim test [92]
Environmental Enrichment and Maternal SeparationhtauE14 infusion at locus coeruleus (both sexes—rat)EE reduced anxiety-like behaviour
Both types of enrichment improved spatial recall
[62]
Environmental Enrichment and StressTg2576 (female—mouse)EE counteracted spatial memory deficits induced by stress, reduced ptau, and improved neurogenesis[73]
Olfactory EnrichmentWT (male—rat)Long-term, but not short-term enrichment reduced ptau[94]
Environmental and Social Enrichment Tg-SwDI (female—mouse)SE, but especially EE, improved motor skills and cognitive performance[68]
Environmental Enrichment in Social IsolationAPPswe/PS1ΔE9 (male—mouse)EE reversed cognitive decline seen in isolation, reduced apoptosis and inflammation[58]
Table 2. Studies examining the effect of stress on Alzheimer’s disease development and progression.
Table 2. Studies examining the effect of stress on Alzheimer’s disease development and progression.
Stress ParadigmAnimal ModelFindingsReference
Chronic Unpredictable StressAPPswe/PS1ΔE9 (male—mouse)Increased Aβ deposition[122]
Aβ infusion in lateral ventricle (male—rat)Increased ptau accumulation[18]
WT (male—rat)Induced ptau, especially with repeat exposure, in hippocampus and PFC[134]
Tg2576 (female—mouse)Worsened spatial memory
Increased ptau and Aβ
[136]
Tg2576 (female—mouse)Accelerated spatial memory decline
Increased ptau and Aβ
[137]
SAMP8 (male—mouse)Worsens spatial memory deficit
Reduces synaptic plasticity
[138]
arcAβ (male—mouse)In WT controls, stress impaired attention and impulsivity
In Aβ mice, stress reduced impulsivity
[139]
APPswe/PS1ΔE9 (male—mouse)Increased Aβ deposition
Increased glucocorticoids
[140]
apoE4-TR (male—mouse)apoE4 mice were more susceptible to stress-induced cognitive decline and depression than controls[141]
Chronic Unpredictable Stress with Ace InhibitorWT (male—rat)Stress-induced ptau development, mediated through ACE enzyme activity[129]
Chronic Unpredictable Stress with EscitalopramWT (male—rat)Stress increased ptau burden
Escitalopram reduced ptau
[133]
Chronic Unpredictable Stress and Maternal SeparationhtauE14 infusion at locus coeruleus (both sexes—rat)CUS worsened, while maternal separation improved, discrimination learning in htauE14 animals[62]
Maternal SeparationAPPswe/PS1ΔE9 (male—mouse)Reduced spatial memory
Increased Aβ deposition
[165]
APPNL-G-F (male—mouse)Induced Aβ deposition
Impaired spatial memory
Increased inflammation
[166]
Model Mimics Stress Responsecorticotropin-releasing factor overexpression (female—mouse)CRF-OE increases ptau
Treatment with CRFR-1 antagonist reduces phosphorylation
[119]
Dexamethasone AdministrationTg2576 (female—mouse)Impaired fear recall
Increased Aβ
[123]
IsolationTg2576 (both sexes-mouse)Increased corticosterone and CRFR-1 expression
Increased Aβ deposition
[127]
Isolation and Environmental Enrichment5xFAD (male—mouse)Increased Aβ
Reduced fear learning
EE did not rescue those effects
[130]
Isolation and RestraintTg2576 (both sexes—mouse)Long-term isolation and short-term restraint increased Aβ, mediated through CRF[158]
Psychosocial StressAβ1-42 infusion at left ventricle (male—rat)Stress worsened spatial memory[126]
Gestational RestraintAPPswe/PS1ΔE9 (both sexes—mouse)Males had reduced spatial memory and HPA response
Females had increased spatial memory and depressive-like symptoms and reduced Aβ
[155]
RestraintTgF344-AD (male—rat)No significant changes in anxiety- or fear-like behaviour[164]
TgCRND8 (female—mouse)Restraint did not worsen Aβ pathology[163]
APP-CT100, Tg2576 (both sexes—mouse)Accelerated cognitive decline
Increased Aβ and ptau pathology
[161]
APPswe/PS1ΔE9 (male—mouse)Increased inflammation[159]
L/V-Tg (male—mouse)Short-term stress-induced neurodegeneration
Long-term also reduced neurogenesis
[157]
5xFAD (both sexes—mouse)Increased Aβ deposition in females[121]
APP/hAβ/PS1 (male—mouse)AD model animals more susceptible to effects of stress
Stress exacerbated AD pathology
[124]
Chronic Unpredictable Stress and RestraintTg2576 (female), P301S (male), CRF-OE (female) (mouse)Restraint increased Aβ and ptau pathology and worsened memory in AD models, not in CRF-OE[120]
Restraint with ALOX5 Knockout3xTg-AD (female—mouse)Stress worsened ptau and impaired fear memory but not in ALOX5 KO animals[123]
Restraint with IcariinAPP/PS1 (male—mouse)Stress-induced depressive-like behaviour, worsened spatial memory, and increased Aβ. Icariin reversed memory impairment and reduced Aβ[156]
Restraint with MemantineWT (male—mouse)Restraint-induced ptau by day 16
MEM dose-dependently reduced ptau
On day 28 MEM worsened ptau
[91]
Restraint with
CRFR NBI 27914
Tg2576 (both sexes—mouse)Restraint increased Aβ and ptau pathology
NBI prevented such effects
[128]
Restraint with PNU-282987APPswe/PS1ΔE9 (male—mouse)Stress-impaired spatial memory
PNU did not rescue
[162]
Sleep DeprivationWT (male—rat)Reduced spatial learning
Increased Aβ
[131]
WT (male—mouse)Reduced fear learning
Increased Aβ
[130]
APPswe/PS1ΔE9 (male—mouse)Reduced spatial memory
Increased Aβ and ptau
[154]
APPswe/PS1ΔE9 (female—mouse)Fragmented sleep increased Aβ
Severity of sleep deprivation correlated with amount of Aβ
[153]
APPswe/PS1ΔE9 (both sexes—mouse)Impaired spatial memory
Increased Aβ and ptau
[152]
5xFAD (both sexes—mouse)AD model animals had more fragmented sleep and were more susceptible to effects of sleep deprivation[143]
3xTg-AD (both sexes—mouse)Poorer spatial memory
Reduced synaptic plasticity
[149]
P301S (male—mouse)AD model mice had reduced sleep which worsened with age
Reduced sleep correlated with increased ptau
[147]
APPswe/PS1ΔE9 (male—mouse)Reduced spatial and fear memory
Reduced synaptic plasticity
Increased Aβ
Increased inflammation
[146]
Sleep DeprivationAPP/PS1 (both sexes—mouse)Poorer spatial memory
Increased inflammation
Reduced synaptic plasticity
Increased Aβ
[145]
Aβ1-42 infusion at lateral ventricle (male—mouse)Aβ infusion disrupted sleep patterns
Sleep-deprived animals more prone to Aβ effects on memory
[144]
3xTg-AD (male—mouse)Increased corticosterone
Reduced spatial memory
Increased Aβ and ptau
[125]
Sleep Deprivation and Acoustic Stimulation3xTg-AD (both sexes—mouse)Sleep deprivation worsened spatial memory and increased Aβ and ptau
Acoustic stimulation reversed such effects
[148]
Sleep Deprivation and Ketogenic DietWT (female—mouse)Sleep deprivation worsened spatial memory and increased Aβ and ptau
Keto diet reversed such effects
[135]
Sleep Deprivation and Orexin Receptor AntagonistTg2576 (sex not specified—mouse)Sleep deprivation increased Aβ deposition
Aβ reduced following almorexant treatment
[151]
Sleep Deprivation and Orexin KnockoutAPPswe/PS1ΔE9 (both sexes—mouse)Orexin KO mice had reduced Aβ and increased sleep time
Rescuing orexin neurons increased Aβ
[150]
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

Torraville, S.E.; Flynn, C.M.; Kendall, T.L.; Yuan, Q. Life Experience Matters: Enrichment and Stress Can Influence the Likelihood of Developing Alzheimer’s Disease via Gut Microbiome. Biomedicines 2023, 11, 1884. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines11071884

AMA Style

Torraville SE, Flynn CM, Kendall TL, Yuan Q. Life Experience Matters: Enrichment and Stress Can Influence the Likelihood of Developing Alzheimer’s Disease via Gut Microbiome. Biomedicines. 2023; 11(7):1884. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines11071884

Chicago/Turabian Style

Torraville, Sarah E., Cassandra M. Flynn, Tori L. Kendall, and Qi Yuan. 2023. "Life Experience Matters: Enrichment and Stress Can Influence the Likelihood of Developing Alzheimer’s Disease via Gut Microbiome" Biomedicines 11, no. 7: 1884. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines11071884

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