• Open Access
    Review

    Feed your microbiome and improve sleep, stress resilience, and cognition

    Maciej Chichlowski 1*
    Jack Cotter 2
    Neil Fawkes 2
    Neeraj Pandey 2

    Explor Med. 2022;3:331–344 DOI: https://doi.org/10.37349/emed.2022.00097

    Received: May 31, 2022 Accepted: July 12, 2022 Published: August 29, 2022

    Academic Editor: Feng Tian, Shandong Provincial Hospital Affiliated to Shandong University; Shandong Provincial Hospital Affiliated to Shandong First Medical University, China

    This article belongs to the special issue The Role of Gut Microbiota and its Metabolites in Gastrointestinal Diseases

    Abstract

    The brain and gut are connected both physically and biochemically. The gut-brain axis includes the central nervous system, neuroendocrine and neuroimmune systems, the enteric nervous system and vagus nerve, and the gut microbiome. It can influence brain function and even behavior, suggesting that dietary interventions may help enhance and protect mental health and cognitive performance. This review focuses on the role of the microbiome and its metabolites in sleep regulation, neurodegenerative disorders, mechanisms of stress, and mood. It also provides examples of nutritional interventions which can restore healthy gut microbiota and aid with risk reduction and management of many disorders related to mental and cognitive health. Evidence suggests a shift in the gut microbiota towards a balanced composition could be a target to maintain brain health, reduce stress and improve quality of life.

    Keywords

    Gut-brain axis, microbiome, Alzheimer’s disease, Parkinson’s disease, biotics, sleep, stress, blood-brain barrier

    Introduction

    Mental health and neurodegenerative disorders, like Alzheimer’s disease (AD) and Parkinson’s disease (PD), are among the most common, costly, and debilitating health conditions worldwide [1]. Poor sleep and high stress are prevalent in the general population and may be causal factors in the onset of these issues [24]. Available pharmaceutical treatments are often slow or inadequate, leading to an interest in preventative, adjunctive, and alternative therapeutic approaches. The gut has been identified as a critical avenue for investigation, given structural gut-brain connections and the burgeoning literature linking gut microbiome disruptions to central nervous system (CNS) disorders. Anxiety, depression, and dementia are linked to gastrointestinal (GI) microbiome disturbances [5, 6]. Increasingly, preclinical studies support the role of the gut in shaping behavioral and cognitive outcomes [79]. The evidence points to nutrition as a feasible target to improve mental wellbeing and maintain healthy cognitive function.

    The gut stores neurotransmitters, such as serotonin, a signaling molecule and key hormone that stabilizes mood and feelings of well-being [10] and dopamine (up to half is stored in the gut) which is essential in processes linked to motivation, memory, and attention [11]. The gut-brain axis (GBA) refers to the bi-directional communication between brain and gut, which is often significantly impacted by microbiome [12].

    Interestingly, a microbiota-gut-immune-glia (MGIG) axis was described for depressive disorders. It is likely to be activated by gut-associated lymphoid tissue upon changes to gut barrier lining resulting in the aberrant cross-talk between glial-neurons, low-grade inflammation, and depression (reviewed by Rudzki and Maes [13]).

    Additionally, cross talk between gut microbiota and CNS has been implicated in several other mental disorders ranging from autism [14], attention-deficit/hyperactivity disorder (ADHD) [15] to alcoholism [1416]. A three-way interplay with microbiome intimately associated with sleep and cognition has been described [17].

    This review focuses on crucial evidence linking the microbiome to stress, sleep quality, and cognitive decline. Recent intervention studies demonstrating how the consumption of nutritional ingredients may offer benefits for associated health conditions are also highlighted.

    The GBA

    The role of microbiota

    The human microbiota consists of trillions of microbial cells in each person [18]. In more technical terms, microbiota refers to all microorganisms found in a selected environment (e.g., gut). However, another widely used term is microbiome which describes the collection of genomes from all the microorganisms. The gut microbiome is vital in the functioning of the nervous system. Gut microbiota produce neuroactive compounds, such as neurotransmitters, amino acids, and metabolites, which affect the brain or indirectly act through the enteric nervous system (ENS). The ENS is an autonomous part of the nervous system which controls mucosal secretions, and motor and endocrine functions [19]. It connects the gut microbiota, hypothalamic-pituitary-adrenal (HPA) axis, and vagus nerve transmitting information from the gut to the brain [19]. Bacterial fermentation products, such as butyrate [20] and gamma-aminobutyric acid (GABA) [21], have neuroprotective effects. For example, GABA-producing Lactobacillus species (Lacticaseibacillus according to new nomenclature) influence the neural network of the host [22]. Germ-free mice (born and raised in sterile conditions) had higher tryptophan (essential in serotonin production) compared to control mice but almost no serotonin [23]. The microbiota-depleted mice were also deficient in vitamin B6 metabolites, which are the molecules that accelerate the production of serotonin and dopamine [23]. The protective effects of butyrate include its capacity to upregulate the melatonergic (melatonin-producing) pathway [24]. At the same time, melatonin also upregulates gut microbiome-derived butyrate [25]. Thus the microbiota plays a vital role in the production of neurotransmitters from the diet through various pathways.

    The GI metabolism is closely connected to brain function via the circulatory system and vagus nerve, creating GBA. Ninety percent of the neural connection in GBA is through the vagus afferent nerve fibers, which travel from the gut to the brain [26]. Although conceptual borders between hormones, neurotransmitters, and cytokines are blurred (i.e., certain compounds can act as both hormone and neurotransmitter, such as adrenaline), GBA components are well-defined. They can work separately or in unison (Figure 1). Also, changes on either side of GBA communication may affect the other. For example, gut microbiota communicates with the brain through cytokines released by mucosal immune cells, serotonin produced by endocrine cells, and the vagus nerve. In contrast, efferent signals (away from the brain) related to stress, such as sympathetic neurotransmitters (e.g., GABA), influence gut microbiome and secretion [27]. For example, increased stress can cause changes in gut microbiome (e.g., lower levels of lactic acid bacteria) [28]. In contrast, modulation of the microbiome with probiotics or diet can help elevate mood and reduce anxiety [29].

    The interactions that occur at the GI mucosa level, and via neural, endocrine, or immune activity. Immunoregulatory pathway: a finely balanced system where microbiome interacts with immune cells to train and mature the components of the immune system while the immune system allows vital microbiome and host cell symbiosis. Neuroendocrine pathway: microbiome affects the HPA axis and the CNS via secretion of neurotransmitters, such as GABA, serotonin, melatonin, histamine, and acetylcholine. Vagus nerve pathway: vagus nerve establishes the connection between the brain and the GI tract and sends information about its state to the brain via afferent fibers

    Along with the increase in age, there is a shift in the microbiome. The microbiome changes are associated with impairment in sleep, development of anxiety, and depression disorders [30]. Aging also reshapes the gut microbiome structure with a high abundance of pro-inflammatory bacteria (e.g., Pseudomonadota and previous nomenclature Proteobacteria) and fewer anti-inflammatory bacteria (e.g., Lactocaseibacillus or Bifidobacterium species), causing local systemic inflammation and permeability of the GI tract [31]. This further impairs the blood-brain barrier (BBB) function, leading to neuroinflammation and neuronal damage.

    Crossing the BBB

    The BBB is composed of multiple cell types and is the protective system that restricts the entry of blood-derived molecules and pathogens into the CNS (Figure 2), which would be harmful to the brain. The BBB comprises endothelial cells (which regulate inflammatory reactions), glia (cells with various supporting functions in the brain, e.g., astrocytes), neurons, and extracellular matrix components that maintain the CNS. Neurons, glial cells, and immune cells share intercellular signals, while some signals can cross the BBB in both directions. Preclinical data demonstrated the role GBA plays in maintaining BBB integrity. For example, in mice deprived of microbiome (i.e., germ-free), the BBB is more permeable, with decreased tight junction protein expression and disorganized tight junctions [32]. Microbiota products, such as short-chain fatty acids (SCFAs), restore BBB stability [33]. Sleep loss and severe stress impact the function of BBB by disrupting the interaction of pericytes and endothelial cells [34, 35]. Sun and colleagues [36] demonstrated that learning and memory were impaired in rats with increased BBB permeability. Those authors described a strong correlation between sleep deprivation and BBB, suggesting a molecular basis for those changes.

    The BBB. The BBB is a structural and functional impediment to undesirable components or microorganisms that would harm the brain. It comprises endothelial cells (which regulate inflammatory reactions), astrocytes (with various supporting functions in the brain), neurons, and extracellular matrix components that function together in the maintenance of CNS

    Sleep

    An essential physiological process impacted by GBA is sleep. Sleep is regulated by 1) sleep pressure, the accumulation and binding of adenosine (nucleoside presents in all human cells) to its receptor and further signaling the need for sleep, and 2) circadian rhythm. Circadian rhythm is a biological process that follows an about 24-hour cycle based on the body’s internal clock, e.g., the sleep-wake cycle. Aging involves changes to sleep patterns, like an increase in the time it takes to fall asleep (sleep latency) and an overall decline in rapid eye movement (REM) sleep [37]. With age, people tend to have more difficulty falling asleep (sleep latency increases) and more trouble staying asleep (shallow and fragmented sleep). Sleep is critical in neural biological regulation by allowing the elimination of metabolites via BBB (reviewed by Cuddapah et al. [38]).

    Factors that alter sleep pressure can also cause sleep disturbances [39]. Dementia, impaired immunity, and stress are correlated with disrupted sleep, leading to cognitive decline (reviewed by Hughes et al. [40]). There is an intricate connection between sleep and mood states, and the relationship between sleep quality, and anxiety and depression has been described [4145]. Aging is one of the risk factors for insomnia, coupled with fragmented sleep [46] and gut dysbiosis [47]; there seems to be a confluence of risk factors that lead to insomnia and subsequent many mental issues seen in old age [30].

    Microbiome structure and diversity also exhibit diurnal rhythms [48]. In a preclinical study, depletion of microbes eliminated serotonin in the gut, affecting the sleep-wake cycles [23]. Stress exposure also disrupts sleep (circadian misalignment) and causes gut microbial imbalance (dysbiosis). Preclinical studies show that microbiota has a profound effect on sleep. Germ-free mice had more REM and non-REM sleep (compared to animals colonized with bacteria) during the time when mice are typically active. The same study also showed that the absence of bacteria caused less daytime non-REM sleep, in contrast to normal behavior when mice typically sleep [23]. Those sleep disturbances could be related to low levels of serotonin, a key neurotransmitter associated with the GBA [41]. In their recent review, Han et al. [49], conclude that abnormal sleep patterns and duration affect the composition and diversity of the microbiota.

    Several preclinical and clinical studies have demonstrated a link between sleep and cognitive disorders such as AD and PD [17] (further reviewed here [50]).

    Neurodegenerative disorders

    AD

    AD is a neurodegenerative CNS disorder accounting for 60–80% of all dementias [51]. Clinical features include a progressive decline in cognitive function (including a severe memory impairment) that adversely impacts daily functioning and quality of life. The hallmark pathology of AD is characterized by the extracellular deposition of beta-amyloid (a small piece of a larger protein) as neuritic plaques and intracellular hyperphosphorylated tau protein as tangles [52]. The BBB functioning is severely affected in AD patients, as shown by magnetic resonance imaging (MRI) studies [53].

    There is evidence of an aberrant gut microbiome resulting in inflammation, primarily through microglia, which impacts synapse and pathophysiology of AD (reviewed by Bairamian [54]). Studies increasingly suggest that microbes likely contribute to the progressive neurodegeneration observed in AD [55]. The microbiome of elderly patients with AD shows a lower proportion of bacteria producing butyrate, which promotes anti-inflammatory activity, and more bacterial taxa known to cause pro- inflammatory states [56, 57]. Cattaneo et al. [57] observed a shift towards pro-inflammatory and lower anti-inflammatory bacteria (e.g., Bacillus fragilis, Eubacterium rectale, Faecalibacterium prausnitzii, and Bacteroides fragilis) in amyloid-positive patients compared to healthy subjects. Interestingly, patients with depression, bipolar disorder, and anxiety also tend to have fewer butyrate-producing bacteria and more pro-inflammatory bacteria [58].

    In the mouse AD model, brain infection with Salmonella typhimurium promoted beta-amyloid plaque formation close to Salmonella attachment sites long before the natural occurrence of plaques in uninfected mice [59]. Bacteria such as Bacillus subtilis secrete large amounts of lipopolysaccharides (LPS, an endotoxin that can induce anxiety-like behavior) and amyloid proteins that cross the BBB [60]. Changes in gut microbial composition enhance the release of LPS and amyloid proteins, which likely contribute to AD pathogenesis, especially during aging when both the GI tract epithelium and BBB are more permeable. LPS and amyloid proteins directly pass through a compromised GI tract or BBB and trigger the passage of cytokines which likely drive AD [59].

    PD

    In PD, neurons producing dopamine in the mid-brain (substantia nigra and striatum) degenerate, resulting in the deficiency of this critical neurotransmitter [61] and the buildup of alpha-synuclein aggregates in the remaining neurons. Metabolites produced by bacteria trigger an immune response inducing inflammation and development of PD [62]. With lower levels of the intestinal hormone ghrelin, researchers observed increased Lactobacillaceae and decreased Prevotellaceae in the gut [61]. The microbiome of PD patients had fewer butyrate-producing bacteria and more pro-inflammatory Proteobacteria, which may trigger misfolding of alpha-synuclein and the development of PD [63].

    Nutritional interventions—modulating the gut microbiome

    With the demonstrated link between the microbiome and both AD and PD, modulating the gut microbiome with diet or biotics could lead to promising prophylactics and solutions. A preclinical study with fecal microbiota transfer therapy in AD mice demonstrated changes in microbiome which ameliorated amyloidosis, tau pathology, and cognitive impairment [64]. In another study, probiotic Bifidobacterium breve A1 increased the plasma acetate levels in mice; this probiotic also suppressed inflammation-related genes induced by beta-amyloid accumulation in the brain [65]. In mouse AD models, mice fed fructooligosaccharide (FOS) were resilient to induced cognitive deficits and performed similarly to controls in a water maze [66]. Dietary 2’-fucosyllactose (2’-FL), a human milk oligosaccharide, has a potential cognition-boosting effect [67]. 2’-FL contains a monosaccharide, fucose, found in the synapses and is vital in cognitive function [68]. Vagotomy prevented the effects of oral 2’-FL on long-term potentiation (key mechanism in learning and memory processes) in rats [69]. Wang and colleagues [70] reported that the subdiaphragmatic vagus nerve plays a vital role in the pathogenesis of depression and is a major modulatory pathway between the brain and gut microbiota. The same group showed that subdiaphragmatic vagotomy blocked the development of depression-like phenotypes in mice treated with antibiotics after fecal matter transfer [71].

    In clinical studies, supplementation with B. breve A1 helped improve cognitive function in older adults who experience mild cognitive impairment [7274]. In another clinical study with probiotics, Georgescu and colleagues [75] have shown that Lactobacillus acidophilus and Bifidobacterium infantis can reduce abdominal pain and bloating in PD patients. Tamtaji et al. [76] administered probiotic treatment or placebo for 12 weeks to PD patients and found decreases in motor symptoms, C-reactive protein, malondialdehyde (a marker for oxidative stress), and increased glutathione (antioxidant).

    SCFAs, the primary metabolites produced by bacteria in the gut, affect GBA and psychological function via the GI tract. Ho et al. [77] found that select SCFAs, especially butyric, valeric, and propionic acids, reduce beta-amyloid accumulation, which suggests the hope for SCFAs in managing AD. PD patients, too have fewer SCFA-producing bacteria and lower SCFA levels in the stool samples compared to healthy controls [78], suggesting that beneficial effects of SCFAs may extend to other neurodegenerative conditions.

    Stress

    Mechanisms of stress

    Stress impacts the body via GBA, and gut bacteria profoundly impact well-being and response to stress and anxiety [7981]. Daily or prolonged stress, with substantial increases during the COVID-19 pandemic [82], can detrimentally affect physical, mental, and cognitive health.

    When experiencing a stressful event, the amygdala (a brain area that controls emotions) sends a signal to the hypothalamus (brain command center) [83], which consequently alerts the nervous system. CNS then releases a flood of stress hormones, including adrenaline and cortisol (neuroendocrine stress marker) (Figure 3). Stress-induced corticotropin-releasing hormone (CRH) from the amygdala and hypothalamus initiates the HPA axis; it also acts on mucosal mast cells to increase the release of tumor necrosis factor-alpha which leads to increased gut permeability [84].

    Mechanisms of stress. In a stressful event, the amygdala sends a signal to the hypothalamus, which triggers an alert through the nervous system. This releases a flood of stress hormones, including adrenaline and cortisol. Fight-or-flight response slows down digestion and increases gut sensitivity. Stress harms the intestinal barrier and can initiate a vicious cycle of inflammation. ACTH: adrenocorticotropic hormone

    Chronic stress can manifest as physical symptoms such as headache, sleep loss, difficulty in concentrating, and skin issues [85]. In addition to the fight-or-flight response, repeated exposure to stress can initiate a vicious cycle of inflammation and negatively impacts the intestinal barrier [86, 87]. Stress hormones, such as cortisol, can affect intestinal barrier integrity and immune signaling within the gut and change gut microbiota composition. Indeed, Geng et al. [88] demonstrated how psychological stress alone had significant effects on the composition and diversity of gut microbiota in rats. Those researchers further characterized the negative impact of stress on intestinal and BBB, including decreased expression of tight junction proteins, looser tight junctions, and broken basement membrane.

    Dietary supplements—modulating the gut microbiome

    Preclinical data

    In rats, prebiotics polydextrose (PDX; branched polymer of glucose and sorbitol) and galactooligosaccharides (GOS; linear polymer of galactose) were a promising treatment for behavior, using the model which mirrors symptoms of human depression and anxiety. These prebiotics also reduced the expression of a stress-evoked marker for neuronal activity (c-fos) in the dorsal raphe nucleus (a brainstem structure important for learned helplessness behaviors). It modulated gene expression in neural circuits essential in stress resistance [89]. Rats injected with LPS that received GOS in drinking water demonstrated fewer anxious behaviors than those not fed GOS [90]. Similarly, rats fed GOS were more exploratory, more social, and showed fewer depression- like behaviors compared to controls [91]. In young pigs, a diet with PDX impacted exploratory behavior, recognition memory, and neurochemistry [92].

    Sialylation contributes to the structural diversity of glycoconjugates (sugars linked to other biological molecules) which have a critical role in neural development, synapses functioning, cognition development, and memory formation [93]. In a mouse model, a social disruption stressor caused anxiety-like behavior in control mice but not in mice fed acidic oligosaccharides, 3’- or 6’-sialyllactose (3’-SL or 6’-SL) for two weeks before stress induction. Their results suggested that sialyllactose helped to maintain baseline behavior [7]. Diet with 3’-SL and 6’-SL also protected colonic microbiota from stress (e.g., maintained beta diversity) [7]. Rodents in the stress group fed 3’-SL and 6’-SL demonstrated behavior like non-stressed controls, pointing to a protective effect of sialyllactose.

    In a double-blind, randomized placebo-controlled study, participants received 5.5 g/day of GOS, FOS, or maltodextrin (placebo) for three weeks [94]. GOS intake reduced anxiety behavior, accompanied by physiologically reduced waking cortisol and fewer prominent rises and falls in cortisol. Sleep quality, stress, and gut microbiome were assessed in adults 30–50 years of age who consumed a dairy-based product with added GOS and vitamins and minerals over three weeks (1 h before bedtime) [95]. The test diet reduced salivary cortisol and stimulated beneficial bacteria. Another clinical study reported better cognitive flexibility and sustained attention performance in healthy adults receiving PDX supplement [96]. Finally, supplementation with oligofructose-enriched inulin improved memory tasks and mood [97].

    Patients with chronic fatigue syndrome (N = 35) showed improved anxiety symptoms after two months of supplementation with Lacticaseibacillus casei [98]. In another randomized, double-blind study (N = 40), a combination of L. acidophilus, L. casei, and Bifidobacterium bifidum for eight weeks improved the Beck Depression Inventory scores [99]. Recently, Nishida and colleagues [100] reported the stress-relieving effects of probiotic Lactobacillus gasseri CP2305 and suggested that bacterial cell components may contribute to this activity. Several studies have indicated that other probiotics may also have the potential to help reduce physiological markers of stress [101, 102].

    Conclusions

    Nutrition and diet can influence the gut microbiome and impact cognitive function and behavior. Unbalanced nutrition, aging, and a sedentary lifestyle cause disturbances in gut microbiota and may lead to chronic inflammation. This can further result in neuronal loss, with unfavorable changes potentially leading to the release of gut-derived inflammatory mediators and the pathogenesis of AD, PD, and other disorders. Restoration of healthy gut microbiota via nutritional supplementation might provide a practical approach for preventing and managing many disorders. In addition, the development of probiotic strains that modulate the GBA could be promising as a complementary approach for enhancing mental and cognitive health.

    Abbreviations

    2’-FL:

    2’-fucosyllactose

    3’-SL:

    3’-sialyllactose

    AD:

    Alzheimer’s disease

    BBB:

    blood-brain barrier

    CNS:

    central nervous system

    GABA:

    gamma-aminobutyric acid

    GI:

    gastrointestinal

    GBA:

    gut-brain axis

    GOS:

    galactooligosaccharides

    HPA:

    hypothalamic-pituitary-adrenal

    LPS:

    lipopolysaccharides

    PD:

    Parkinson’s disease

    PDX:

    polydextrose

    REM:

    rapid eye movement

    SCFA:

    short-chain fatty acids

    Declarations

    Acknowledgments

    We want to express our thanks to Dr. Jennifer Wampler for reviewing our draft. We thank the medical illustrators at www.haderermuller.com who helped us to visualize many concepts in this manuscript.

    Author contributions

    MC contributed conceptualization, original draft, and editing; JC, NF, NP review and editing. All authors contributed to the article and approved the submitted version.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2022.

    References

    GBD 2019 Diseases and Injuries Collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet. 2020;396:120422. [DOI] [PubMed] [PMC]
    Freeman D, Sheaves B, Waite F, Harvey AG, Harrison PJ. Sleep disturbance and psychiatric disorders. Lancet Psychiatry. 2020;7:62837. [DOI] [PubMed]
    Johansson L, Guo X, Waern M, Ostling S, Gustafson D, Bengtsson C, et al. Midlife psychological stress and risk of dementia: a 35-year longitudinal population study. Brain. 2010;133:221724. [DOI] [PubMed]
    Sabia S, Fayosse A, Dumurgier J, van Hees VT, Paquet C, Sommerlad A, et al. Association of sleep duration in middle and old age with incidence of dementia. Nat Commun. 2021;12:2289. [DOI] [PubMed] [PMC]
    Collins SM, Kassam Z, Bercik P. The adoptive transfer of behavioral phenotype via the intestinal microbiota: experimental evidence and clinical implications. Curr Opin Microbiol. 2013;16:2405. [DOI] [PubMed]
    Foster JA, McVey Neufeld KA. Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci. 2013;36:30512. [DOI] [PubMed]
    Tarr AJ, Galley JD, Fisher SE, Chichlowski M, Berg BM, Bailey MT. The prebiotics 3’sialyllactose and 6’sialyllactose diminish stressor-induced anxiety-like behavior and colonic microbiota alterations: evidence for effects on the gut-brain axis. Brain Behav Immun. 2015;50:16677. [DOI] [PubMed] [PMC]
    Thompson RS, Vargas F, Dorrestein PC, Chichlowski M, Berg BM, Fleshner M. Dietary prebiotics alter novel microbial dependent fecal metabolites that improve sleep. Sci Rep. 2020;10:3848. [DOI] [PubMed] [PMC]
    Thompson RS, Roller R, Mika A, Greenwood BN, Knight R, Chichlowski M, et al. Dietary prebiotics and bioactive milk fractions improve NREM sleep, enhance REM sleep rebound and attenuate the stress-induced decrease in diurnal temperature and gut microbial alpha diversity. Front Behav Neurosci. 2017;10:240. [DOI] [PubMed] [PMC]
    Bornstein JC. Serotonin in the gut: what does it do? Front Neurosci. 2012;6:16. [DOI]
    Eisenhofer G, Aneman A, Friberg P, Hooper D, Fåndriks L, Lonroth H, et al. Substantial production of dopamine in the human gastrointestinal tract. J Clin Endocrinol Metab. 1997;82:386471. [DOI] [PubMed]
    Teichman EM, O’Riordan KJ, Gahan CGM, Dinan TG, Cryan JF. When rhythms meet the Blues: circadian interactions with the microbiota-gut-brain axis. Cell Metab. 2020;31:44871. [DOI] [PubMed]
    Rudzki L, Maes M. The microbiota-gut-immune-glia (MGIG) axis in major depression. Mol Neurobiol. 2020;57:426995. [DOI] [PubMed]
    Tomova A, Soltys K, Repiska G, Palkova L, Filcikova D, Minarik G, et al. Specificity of gut microbiota in children with autism spectrum disorder in Slovakia and its correlation with astrocytes activity marker and specific behavioural patterns. Physiol Behav. 2020;214:112745. [DOI] [PubMed]
    Cenit MC, Nuevo IC, Codoñer-Franch P, Dinan TG, Sanz Y. Gut microbiota and attention deficit hyperactivity disorder: new perspectives for a challenging condition. Eur Child Adolesc Psychiatry. 2017;26:108192. [DOI] [PubMed]
    Hillemacher T, Bachmann O, Kahl KG, Frieling H. Alcohol, microbiome, and their effect on psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2018;85:10515. [DOI] [PubMed]
    Roh JH, Jiang H, Finn MB, Stewart FR, Mahan TE, Cirrito JR, et al. Potential role of orexin and sleep modulation in the pathogenesis of Alzheimer’s disease. J Exp Med. 2014;211:248796. [DOI] [PubMed] [PMC]
    Ursell LK, Metcalf JL, Parfrey LW, Knight R. Defining the human microbiome. Nutr Rev. 2012;70 Suppl 1:S3844. [DOI] [PubMed] [PMC]
    Costa M, Brookes SJ, Hennig GW. Anatomy and physiology of the enteric nervous system. Gut. 2000;47 Suppl 4:iv159. [DOI] [PubMed] [PMC]
    Kim HJ, Leeds P, Chuang DM. The HDAC inhibitor, sodium butyrate, stimulates neurogenesis in the ischemic brain. J Neurochem. 2009;110:122640. [DOI] [PubMed] [PMC]
    Cho YR, Chang JY, Chang HC. Production of gamma-aminobutyric acid (GABA) by Lactobacillus buchneri isolated from kimchi and its neuroprotective effect on neuronal cells. J Microbiol Biotechnol. 2007;17:1049. [PubMed]
    Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A. 2011;108:160505. [DOI] [PubMed] [PMC]
    Ogawa Y, Miyoshi C, Obana N, Yajima K, Hotta-Hirashima N, Ikkyu A, et al. Gut microbiota depletion by chronic antibiotic treatment alters the sleep/wake architecture and sleep EEG power spectra in mice. Sci Rep. 2020;10:19554. [DOI] [PubMed] [PMC]
    Jin CJ, Engstler AJ, Sellmann C, Ziegenhardt D, Landmann M, Kanuri G, et al. Sodium butyrate protects mice from the development of the early signs of non-alcoholic fatty liver disease: role of melatonin and lipid peroxidation. Br J Nutr. 2016;116:168293. [DOI] [PubMed]
    Gao T, Wang Z, Dong Y, Cao J, Chen Y. Melatonin-mediated colonic microbiota metabolite butyrate prevents acute sleep deprivation-induced colitis in mice. Int J Mol Sci. 2021;22:11894. [DOI] [PubMed] [PMC]
    Breit S, Kupferberg A, Rogler G, Hasler G. Vagus nerve as modulator of the brain-gut axis in psychiatric and inflammatory disorders. Front Psychiatry. 2018;9:44. [DOI] [PubMed] [PMC]
    Flowers SA, Evans SJ, Ward KM, McInnis MG, Ellingrod VL. Interaction between atypical antipsychotics and the gut microbiome in a bipolar disease cohort. Pharmacotherapy. 2017;37:2617. [DOI] [PubMed]
    Knowles SR, Nelson EA, Palombo EA. Investigating the role of perceived stress on bacterial flora activity and salivary cortisol secretion: a possible mechanism underlying susceptibility to illness. Biol Psychol. 2008;77:1327. [DOI] [PubMed]
    Kazemi A, Noorbala AA, Azam K, Eskandari MH, Djafarian K. Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: A randomized clinical trial. Clin Nutr. 2019;38:5228. [DOI] [PubMed]
    Zhang H, Liu L, Cheng S, Jia Y, Wen Y, Yang X, et al. Assessing the joint effects of brain aging and gut microbiota on the risks of psychiatric disorders. Brain Imaging Behav. 2022;16:150415. [DOI] [PubMed]
    Boehme M, Guzzetta KE, Bastiaanssen TFS, van de Wouw M, Moloney GM, Gual-Grau A, et al. Microbiota from young mice counteracts selective age-associated behavioral deficits. Nature Aging. 2021;1:66676. [DOI]
    Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth M, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 2014;6:263ra158. [DOI] [PubMed] [PMC]
    Silva YP, Bernardi A, Frozza RL. The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol (Lausanne). 2020;11:25. [DOI] [PubMed] [PMC]
    Medina-Flores F, Hurtado-Alvarado G, Contis-Montes de Oca A, López-Cervantes SP, Konigsberg M, Deli MA, et al. Sleep loss disrupts pericyte-brain endothelial cell interactions impairing blood- brain barrier function. Brain Behav Immun. 2020;89:11832. [DOI] [PubMed]
    Sharma A, Muresanu DF, Lafuente JV, Patnaik R, Tian ZR, Buzoianu AD, et al. Sleep deprivation-induced blood-brain barrier breakdown and brain dysfunction are exacerbated by size-related exposure to Ag and Cu nanoparticles. Neuroprotective effects of a 5-HT3 receptor antagonist ondansetron. Mol Neurobiol. 2015;52:86781. [DOI] [PubMed]
    Sun J, Wu J, Hua F, Chen Y, Zhan F, Xu G. Sleep deprivation induces cognitive impairment by increasing blood-brain barrier permeability via CD44. Front Neurol. 2020;11:563916. [DOI] [PubMed] [PMC]
    Kaneshwaran K, Olah M, Tasaki S, Yu L, Bradshaw EM, Schneider JA, et al. Sleep fragmentation, microglial aging, and cognitive impairment in adults with and without Alzheimer’s dementia. Sci Adv. 2019;5:eaax7331. [DOI] [PubMed] [PMC]
    Cuddapah VA, Zhang SL, Sehgal A. Regulation of the blood-brain barrier by circadian rhythms and sleep. Trends Neurosci. 2019;42:50010. [DOI] [PubMed] [PMC]
    Muth CC. Sleep-wake disorders. JAMA. 2016;316:2322. [DOI] [PubMed]
    Hughes AJ, Dunn KM, Chaffee T. Sleep disturbance and cognitive dysfunction in multiple sclerosis: a systematic review. Curr Neurol Neurosci Rep. 2018;18:2. [DOI] [PubMed]
    Oh CM, Kim HY, Na HK, Cho KH, Chu MK. The effect of anxiety and depression on sleep quality of individuals with high risk for insomnia: a population-based study. Front Neurol. 2019;10:849. [DOI] [PubMed] [PMC]
    Short MA, Louca M. Sleep deprivation leads to mood deficits in healthy adolescents. Sleep Med. 2015;16:98793. [DOI] [PubMed]
    Sarchiapone M, Mandelli L, Carli V, Iosue M, Wasserman C, Hadlaczky G, et al. Hours of sleep in adolescents and its association with anxiety, emotional concerns, and suicidal ideation. Sleep Med. 2014;15:24854. [DOI] [PubMed]
    Ho YT, Tsai YC, Kuo TBJ, Yang CCH. Effects of Lactobacillus plantarum PS128 on depressive symptoms and sleep quality in self-reported insomniacs: a randomized, double-blind, placebo-controlled pilot trial. Nutrients. 2021;13:2820. [DOI] [PubMed] [PMC]
    Short MA, Booth SA, Omar O, Ostlundh L, Arora T. The relationship between sleep duration and mood in adolescents: a systematic review and meta-analysis. Sleep Med Rev. 2020;52:101311. [DOI] [PubMed]
    Morin CM, Gramling SE. Sleep patterns and aging: comparison of older adults with and without insomnia complaints. Psychol Aging. 1989;4:2904. [DOI] [PubMed]
    Li Y, Hao Y, Fan F, Zhang B. The role of microbiome in insomnia, circadian disturbance and depression. Front Psychiatry. 2018;9:669. [DOI] [PubMed] [PMC]
    Brooks JF2nd, Behrendt CL, Ruhn KA, Lee S, Raj P, Takahashi JS, et al. The microbiota coordinates diurnal rhythms in innate immunity with the circadian clock. Cell. 2021;184:415467.e12. [DOI] [PubMed] [PMC]
    Han M, Yuan S, Zhang J. The interplay between sleep and gut microbiota. Brain Res Bull. 2022;180:13146. [DOI] [PubMed]
    Diederich NJ, McIntyre DJ. Sleep disorders in Parkinson’s disease: many causes, few therapeutic options. J Neurol Sci. 2012;314:129. [DOI] [PubMed]
    Li Y, Shao L, Mou Y, Zhang Y, Ping Y. Sleep, circadian rhythm and gut microbiota: alterations in Alzheimer’s disease and their potential links in the pathogenesis. Gut Microbes. 2021;13:1957407. [DOI] [PubMed] [PMC]
    Duyckaerts C, Delatour B, Potier MC. Classification and basic pathology of Alzheimer disease. Acta Neuropathol. 2009;118:536. [DOI] [PubMed]
    Montagne A, Barnes SR, Sweeney MD, Halliday MR, Sagare AP, Zhao Z, et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron. 2015;85:296302. [DOI] [PubMed] [PMC]
    Bairamian D, Sha S, Rolhion N, Sokol H, Dorothée G, Lemere CA, et al. Microbiota in neuroinflammation and synaptic dysfunction: a focus on Alzheimer’s disease. Mol Neurodegener. 2022;17:19. [DOI] [PubMed] [PMC]
    Bulgart HR, Neczypor EW, Wold LE, Mackos AR. Microbial involvement in Alzheimer disease development and progression. Mol Neurodegener. 2020;15:42. [DOI] [PubMed] [PMC]
    Hammond CJ, Hallock LR, Howanski RJ, Appelt DM, Little CS, Balin BJ. Immunohistological detection of Chlamydia pneumoniae in the Alzheimer’s disease brain. BMC Neurosci. 2010;11:121. [DOI] [PubMed] [PMC]
    Cattaneo A, Cattane N, Galluzzi S, Provasi S Lopizzo N Festari C, et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging. 2017;49:608. [DOI] [PubMed]
    Kumar DK, Choi SH, Washicosky KJ, Eimer WA, Tucker S, Ghofrani J, et al. Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med. 2016;8:340ra72. [DOI] [PubMed] [PMC]
    Nikolova VL, Hall MRB, Hall LJ, Cleare AJ, Stone JM, Young AH. Perturbations in gut microbiota composition in psychiatric disorders: a review and meta-analysis. JAMA Psychiatry. 2021;78:134354. [DOI] [PubMed] [PMC]
    Mancuso C, Santangelo R. Alzheimer’s disease and gut microbiota modifications: The long way between preclinical studies and clinical evidence. Pharmacol Res. 2018;129:32936. [DOI] [PubMed]
    Caputi V, Giron MC. Microbiome-gut-brain axis and Toll-like receptors in Parkinson’s disease. Int J Mol Sci. 2018;19:1689. [DOI] [PubMed] [PMC]
    Campos-Acuña J, Elgueta D, Pacheco R. T-cell-driven inflammation as a mediator of the gut-brain axis involved in Parkinson’s disease. Front Immunol. 2019;10:239. [DOI] [PubMed] [PMC]
    Keshavarzian A, Green SJ, Engen PA, Voigt RM, Naqib A, Forsyth CB, et al. Colonic bacterial composition in Parkinson’s disease. Mov Disord. 2015;30:135160. [DOI] [PubMed]
    Kim MS, Kim Y, Choi H, Kim W, Park S, Lee D, et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut. 2020;69:28394. [DOI] [PubMed]
    Kobayashi Y, Sugahara H, Shimada K, Mitsuyama E, Kuhara T, Yasuoka A, et al. Therapeutic potential of Bifidobacterium breve strain A1 for preventing cognitive impairment in Alzheimer’s disease. Sci Rep. 2017;7:13510. [DOI] [PubMed] [PMC]
    Yen CH, Wang CH, Wu WT, Chen HL. Fructo-oligosaccharide improved brain β-amyloid, β-secretase, cognitive function, and plasma antioxidant levels in D-galactose-treated Balb/cJ mice. Nutr Neurosci. 2017;20:22837. [DOI] [PubMed]
    Vázquez E, Barranco A, Ramirez M, Gruart A, Delgado-Garcia JM, Jimenez ML, et al. Dietary 2’-fucosyllactose enhances operant conditioning and long-term potentiation via gut-brain communication through the vagus nerve in rodents. PLoS One. 2016;11:e0166070. [DOI] [PubMed] [PMC]
    Sukumar R, Rose SP, Burgoyne RD. Increased incorporation of [3H]fucose into chick brain glycoproteins following training on a passive avoidance task. J Neurochem. 1980;34:10006. [DOI] [PubMed]
    Vázquez E, Barranco A, Ramírez M, Gruart A, Delgado-García JM, Martínez-Lara E, et al. Effects of a human milk oligosaccharide, 2’-fucosyllactose, on hippocampal long-term potentiation and learning capabilities in rodents. J Nutr Biochem. 2015;26:45565. [DOI] [PubMed]
    Wang S, Ishima T, Zhang J, Qu Y, Chang L, Pu Y, et al. Ingestion of Lactobacillus intestinalis and Lactobacillus reuteri causes depression- and anhedonia-like phenotypes in antibiotic-treated mice via the vagus nerve. J Neuroinflammation. 2020;17:241. [DOI] [PubMed] [PMC]
    Pu Y, Tan Y, Qu Y, Chang L, Wang S, Wei Y, et al. A role of the subdiaphragmatic vagus nerve in depression-like phenotypes in mice after fecal microbiota transplantation from Chrna7 knock-out mice with depression-like phenotypes. Brain Behav Immun. 2021;94:31826. [DOI] [PubMed]
    Kobayashi Y, Kinoshita T, Matsumoto A, Yoshino K, Saito I, Xiao JZ. Bifidobacterium breve A1 supplementation improved cognitive decline in older adults with mild cognitive impairment: an open-label, single-arm study. J Prev Alzheimers Dis. 2019;6:705. [DOI] [PubMed]
    Kobayashi Y, Kuhara T, Oki M, Xiao JZ. Effects of Bifidobacterium breve A1 on the cognitive function of older adults with memory complaints: a randomised, double-blind, placebo-controlled trial. Benef Microbes. 2019;10:51120. [DOI] [PubMed]
    Xiao J, Katsumata N, Bernier F, Ohno K, Yamauchi Y, Odamaki T, et al. Probiotic Bifidobacterium breve in improving cognitive functions of older adults with suspected mild cognitive impairment: a randomized, double-blind, placebo-controlled trial. J Alzheimers Dis. 2020;77:13947. [DOI] [PubMed] [PMC]
    Georgescu D, Ancusa OE, Georgescu LA, Ionita I, Reisz D. Nonmotor gastrointestinal disorders in older patients with Parkinson’s disease: is there hope? Clin Interv Aging. 2016;11:16018. [DOI] [PubMed] [PMC]
    Tamtaji OR, Taghizadeh M, Daneshvar Kakhaki R, Kouchaki E, Bahmani F, Borzabadi S, et al. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: a randomized, double-blind, placebo-controlled trial. Clin Nutr. 2019;38:10315. [DOI] [PubMed]
    Ho L, Ono K, Tsuji M, Mazzola P, Singh R, Pasinetti GM. Protective roles of intestinal microbiota derived short chain fatty acids in Alzheimer’s disease-type beta-amyloid neuropathological mechanisms. Expert Rev Neurother. 2018;18:8390. [DOI] [PubMed] [PMC]
    Unger MM, Spiegel J, Dillmann KU, Grundmann D, Philippeit H, Bürmann J, et al. Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat Disord. 2016;32:6672. [DOI] [PubMed]
    Allen AP, Dinan TG, Clarke G, Cryan JF. A psychology of the human brain-gut-microbiome axis. Soc Personal Psychol Compass. 2017;11:e12309. [DOI] [PubMed] [PMC]
    Sarkar A, Harty S, Lehto SM, Moeller AH, Dinan TG, Dunbar RIM, et al. The microbiome in psychology and cognitive neuroscience. Trends Cogn Sci. 2018;22:61136. [DOI] [PubMed]
    Rea K, Dinan TG, Cryan JF. The microbiome: a key regulator of stress and neuroinflammation. Neurobiol Stress. 2016;4:2333. [DOI] [PubMed] [PMC]
    Holman EA, Thompson RR, Garfin DR, Silver RC. The unfolding COVID-19 pandemic: a probability-based, nationally representative study of mental health in the United States. Sci Adv. 2020;6:eabd5390. [DOI] [PubMed] [PMC]
    Chrousos GP. The role of stress and the hypothalamic-pituitary-adrenal axis in the pathogenesis of the metabolic syndrome: neuro-endocrine and target tissue-related causes. Int J Obes Relat Metab Disord. 2000;24 Suppl 2:S505. [DOI] [PubMed]
    Vanuytsel T, van Wanrooy S, Vanheel H, Vanormelingen C Verschueren S Houben E, et al. Psychological stress and corticotropin-releasing hormone increase intestinal permeability in humans by a mast cell-dependent mechanism. Gut. 2014;63:12939. [DOI] [PubMed]
    Suchecki D, Tiba PA, Machado RB. REM sleep rebound as an adaptive response to stressful situations. Front Neurol. 2012;3:41. [DOI] [PubMed] [PMC]
    Yarandi SS, Peterson DA, Treisman GJ, Moran TH, Pasricha PJ. Modulatory effects of gut microbiota on the central nervous system: how gut could play a role in neuropsychiatric health and diseases. J Neurogastroenterol Motil. 2016;22:20112. [DOI] [PubMed] [PMC]
    Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci. 2015;9:392. [DOI] [PubMed] [PMC]
    Geng S, Yang L, Cheng F, Zhang Z, Li J, Liu W, et al. Gut microbiota are associated with psychological stress-induced defections in intestinal and blood-brain barriers. Front Microbiol. 2020;10:3067. [DOI] [PubMed] [PMC]
    Mika A, Day HE, Martinez A, Rumian NL, Greenwood BN, Chichlowski M, et al. Early life diets with prebiotics and bioactive milk fractions attenuate the impact of stress on learned helplessness behaviours and alter gene expression within neural circuits important for stress resistance. Eur J Neurosci. 2017;45:34257. [DOI] [PubMed]
    Savignac HM, Couch Y, Stratford M, Bannerman DM, Tzortzis G, Anthony DC, et al. Prebiotic administration normalizes lipopolysaccharide (LPS)-induced anxiety and cortical 5-HT2A receptor and IL1-β levels in male mice. Brain Behav Immun. 2016;52:12031. [DOI] [PubMed] [PMC]
    Burokas A, Arboleya S, Moloney RD, Peterson VL, Murphy K, Clarke G, et al. Targeting the microbiota- gut-brain axis: prebiotics have anxiolytic and antidepressant-like effects and reverse the impact of chronic stress in mice. Biol Psychiatry. 2017;82:47287. [DOI] [PubMed]
    Fleming SA, Monaikul S, Patsavas AJ, Waworuntu RV, Berg BM, Dilger RN. Dietary polydextrose and galactooligosaccharide increase exploratory behavior, improve recognition memory, and alter neurochemistry in the young pig. Nutr Neurosci. 2019;22:499512. [DOI] [PubMed]
    Wang B. Molecular mechanism underlying sialic acid as an essential nutrient for brain development and cognition. Adv Nutr. 2012;3:465S72S. [DOI] [PubMed] [PMC]
    Schmidt K, Cowen PJ, Harmer CJ, Tzortzis G, Errington S, Burnet PW. Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology (Berl). 2015;232:1793801. [DOI] [PubMed] [PMC]
    Schaafsma A, Mallee L, van den Belt M, Floris E, Kortman G, Veldman J, et al. The effect of a whey- protein and galacto-oligosaccharides based product on parameters of sleep quality, stress, and gut microbiota in apparently healthy adults with moderate sleep disturbances: a randomized controlled cross-over study. Nutrients. 2021;13:2204. [DOI] [PubMed] [PMC]
    Berding K, Long-Smith CM, Carbia C, Bastiaanssen TFS, van de Wouw M, Wiley N, et al. A specific dietary fibre supplementation improves cognitive performance-an exploratory randomised, placebo- controlled, crossover study. Psychopharmacology (Berl). 2021;238:14963. [DOI] [PubMed]
    Smith AP, Sutherland D, Hewlett P. An investigation of the acute effects of oligofructose-enriched inulin on subjective wellbeing, mood and cognitive performance. Nutrients. 2015;7:888796. [DOI] [PubMed] [PMC]
    Rao AV, Bested AC, Beaulne TM, Katzman MA, Iorio C, Berardi JM, et al. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog. 2009;1:6. [DOI] [PubMed] [PMC]
    Akkasheh G, Kashani-Poor Z, Tajabadi-Ebrahimi M, Jafari P, Akbari H, Taghizadeh M, et al. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: a randomized, double-blind, placebo-controlled trial. Nutrition. 2016;32:31520. [DOI] [PubMed]
    Nishida K, Sawada D, Kawai T, Kuwano Y, Fujiwara S, Rokutan K. Para-psychobiotic Lactobacillus gasseri CP2305 ameliorates stress-related symptoms and sleep quality. J Appl Microbiol. 2017;123:156170. [DOI] [PubMed]
    Wallace CJK, Milev RV. The efficacy, safety, and tolerability of probiotics on depression: clinical results from an open-label pilot study. Front Psychiatry. 2021;12:618279. [DOI] [PubMed] [PMC]
    Messaoudi M, Lalonde R, Violle N, Javelot H, Desor D, NejdiA, et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br J Nutr. 2011;105:75564. [DOI] [PubMed]