Skip to main content
Erschienen in: Molecular Neurodegeneration 1/2022

Open Access 01.12.2022 | Review

Microbial-derived metabolites as a risk factor of age-related cognitive decline and dementia

verfasst von: Emily Connell, Gwenaelle Le Gall, Matthew G. Pontifex, Saber Sami, John F. Cryan, Gerard Clarke, Michael Müller, David Vauzour

Erschienen in: Molecular Neurodegeneration | Ausgabe 1/2022

Abstract

A consequence of our progressively ageing global population is the increasing prevalence of worldwide age-related cognitive decline and dementia. In the absence of effective therapeutic interventions, identifying risk factors associated with cognitive decline becomes increasingly vital. Novel perspectives suggest that a dynamic bidirectional communication system between the gut, its microbiome, and the central nervous system, commonly referred to as the microbiota-gut-brain axis, may be a contributing factor for cognitive health and disease. However, the exact mechanisms remain undefined. Microbial-derived metabolites produced in the gut can cross the intestinal epithelial barrier, enter systemic circulation and trigger physiological responses both directly and indirectly affecting the central nervous system and its functions. Dysregulation of this system (i.e., dysbiosis) can modulate cytotoxic metabolite production, promote neuroinflammation and negatively impact cognition. In this review, we explore critical connections between microbial-derived metabolites (secondary bile acids, trimethylamine-N-oxide (TMAO), tryptophan derivatives and others) and their influence upon cognitive function and neurodegenerative disorders, with a particular interest in their less-explored role as risk factors of cognitive decline.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
5-HT
Serotonin (5-hydroxytryptamine)
ACh
Acetylcholine
AD
Alzheimer’s disease
AhR
Aryl hydrocarbon receptor
Amyloid-β
BA
Bile acids
BBB
Blood–brain barrier
BDNF
Brain-derived neurotrophic factor
CA
Cholic acid
CDCA
Chenodeoxycholic acid
CNS
Central nervous system
CSF
Cerebrospinal fluid
CYP27A1
Sterol 27-hydroxylase
CYP46A1
Cholesterol 24-hydroxylase
CYP7A1
Cholesterol 7α-hydroxylase
DCA
Deoxycholic acid
DMN
Default mode network
ECC
Enterochromaffin cells
FMO1/3
Flavin-containing monooxygenase 1/ 3
GABA
γ-Aminobutyric acid
GF
Germ-free
GI
Gastrointestinal
GUDCA
Glycoursodeoxycholic acid
HVA
Homovanillic acid
IA
Indole-3- acrylic acid
IAA
Indole-3- acetic acid
IAld
Indole-3-aldehyde
IDO
Indoleamine 2, 3-dioxygenase
ILA
Indole-3-lactic acid
ImP
Imidazole propionate
IPA
Indole propionic acid
I3S
Indoxyl-3-sulfate
KP
Kynurenine pathway
LAT1
Large neutral amino acid transporters
LCA
Lithocholic acid
L-DOPA
L-3,4-Dihydroxyphenylalanine
LPS
Lipopolysaccharide
MCI
Mild cognitive impairment
MMSE
Mini-mental state examination
MRI
Magnetic resonance imaging
NMDA
N-methyl-D-aspartate
NR
Nicotinamide riboside
PBMCs
Peripheral blood mononuclear cells
PCS
p-Cresyl sulphate
PD
Parkinson’s disease
PET
Positron emission tomography
ROS
Reactive oxygen species
SCFAs
Short-chain fatty acids
TLR
Toll-like receptor
TMA
Trimethylamine
TMAO
Trimethylamine N-oxide
TUDCA
Tauroursodeoxycholic acid

Background

Age is the predominant risk factor for cognitive decline. Whilst some decline in cognition is considered an inevitable part of healthy ageing, deleterious changes in cognition, including mild cognitive impairment (MCI) and age-related dementias (e.g., Alzheimer’s disease, AD), are estimated to impact approximately 15% and 11% of the population over 65 years respectively [1, 2]. By 2050, the global elderly population is expected to increase by 21% [2], increasing incidences of cognitive decline [3]. Cognitive decline exacerbates broad social and economic issues, including depression, social withdrawal, difficulties performing everyday tasks, drastic reductions in quality of life and greater reliance on others (social care) [4]. Understanding how to promote healthy ageing whilst resisting aberrant changes in cognition is therefore becoming a priority.
Addressing modifiable risk factors can delay the onset, or even ameliorate cognitive decline [5], whilst assisting with the identification of asymptomatic individuals with an increased chance of developing the condition in the future [6]. Currently, hypertension [7], diabetes mellitus [8, 9], arteriosclerosis [10], obesity [11] and hypercholesterolemia [12] are the most significant risk factors associated with age-related cognitive decline among others [13]. Given the connection between cognition and these metabolic diseases, it is perhaps unsurprising that dietary factors can elicit a substantial influence upon cognitive function [14] through the modulation of a microbiota-gut-brain axis [15]. The microbiota-gut-brain axis is a complex communication system bridging the gut, liver and the central nervous system (CNS) that is modulated by the microbiome, a collection of 1014 microorganisms with an extensive functional gene repertoire [16]. These microorganisms predominantly reside in the gut, metabolising dietary compounds into a vast range of metabolites. Metabolites can cross the intestinal epithelial barrier; a structure connected by tight junction proteins, lamina propria and reinforced by mucosal secretions [17], primarily via active transport, and enter systemic circulation. From here, metabolites can directly initiate physiological responses by crossing the blood–brain barrier (BBB) and influencing the CNS [18], or indirectly via vagus nerve stimulation (Fig. 1) [19].
The capability of microbial-derived bioactive metabolites to influence the CNS provides a novel mechanistic pathway for cognitive decline, warranting its further exploration. Within the gastrointestinal (GI) tract, microbiota populations are in part reflective of their local physiological conditions. The small intestine, due to its proximity with the stomach, contains high concentrations of acids, oxygen and antimicrobials, thereby restricting bacterial growth to predominantly fast-growing anaerobes that can adhere to epithelia or mucus [20]. Conversely, colonic regions promote much denser bacterial communities, dominated by anaerobes such as Prevotellaceae and Lachnospiraceae, that can digest complex carbohydrates [21]. Numerous intrinsic factors (e.g., genetics, immune response, metabolites) and extrinsic factors (e.g., diet, lifestyle) also impact gut microbial composition, making it an attractive therapeutic intervention target [22]. The composition of these microbial communities determines the concentration of neurotransmitters or neuromodulators (including microbial-derived metabolites) released into circulation. Broad deviations in these microbial compositions, often referred to as “dysbiosis”, condition distinctly different metabolic profiles that may contribute to cognitive decline [23, 24]. Gut microbial composition is known to be significantly altered in patients with MCI, a transitional stage preceding AD, suggesting microbial changes may occur in the early stages of cognitive decline and influence its progression [2529].
Intestinal microbiota possess the capacity to produce hundreds of metabolites [30, 31], yet the influence of these compounds on cognitive health have not been uncovered. The present review details the roles of newly emerging microbial-derived metabolites that are less-explored in the current literature in the context of cognitive health and provide an additional in-depth discussion of their use as potential indicative factors of cognitive decline.
As we age, some of our cognitive abilities decline. Cognitive capabilities such as verbal skills, remain largely unaffected by brain ageing and can even improve over time [32]. Other essential capabilities, including mental reasoning, memory (in particular episodic, working and recognition memory) and processing speed, steadily deteriorate with age (See review [33] for further information). During ageing, the brain undergoes various structural and functional changes. The most apparent being a gradual shrinkage of the brain, alongside an increase in ventricular space and cerebrospinal fluid (CSF) [34, 35]. Brain atrophy increases in the elderly in an anterior–posterior gradient, with the most severe consequences taking place in the prefrontal regions [36, 37]. A reduction in white matter (the nerve fibres connecting different brain regions) integrity has been linked with normal cognitive ageing, impairing the transfer of information between cortical regions [38], an essential process for higher cognitive functioning [39].
Structural neuroimaging highlights differing trends in the neurobiology of pathological ageing and detrimental cognitive decline. Here, individuals are more likely to experience reductions in gray matter in the dorsolateral and medial prefrontal, parietal, and lateral temporal regions [40, 41], alongside a loss of white matter integrity in the cingulum, corpus callosum, and superior longitudinal fasciculus [4244]. This is instead of a decline in the frontal regions that typically occurs in healthy ageing.
AD is also associated with volume loss in the medial temporal lobe, a brain region highly associated with memory functions. Reduction typically starts in the anterolateral entorhinal cortex and advances medially across the remaining entorhinal cortex to the hippocampus [45, 46], with atrophy occurring at rates of 4.9–8.2%. In healthy ageing, atrophy in these regions occurs at a lower rate, diverging from pathological ageing, at 0.2–3.8% [46]. More recently, using longitudinal MRI and PET data, a similar divergence in volume loss has been noted in the locus coeruleus [47].
Finally, the default mode network (DMN), a resting-state network associated with cognitive processes of oneself (e.g., autobiographical memory), demonstrates connectivity patterns that distinguish healthy ageing from AD. Results from a task free-fMRI suggest AD patients have an accelerated ageing pattern of connectivity [48] and decreased resting-state activity in the posterior cingulate and hippocampus when compared with age-matched controls [49]. However, the biological mechanisms behind the heterogeneity of age-related cognitive decline are complex and not well understood.

The microbiota-gut-brain axis in the context of ageing and cognitive decline

The human gut microbiome represents a complex community of microbes that live in a mutualistic relationship with their host. Initially, these microorganisms were considered to be solely responsible for intestinal processes (fermentation of carbohydrates, synthesis of vitamins and xenobiotic metabolism) [50]. However, over the last 15 years, this notion has been revised, owing to increasing evidence of a bidirectional communication system between the CNS and the GI tract, more commonly referred to as the ‘gut-brain axis’.
The gut-brain axis encompasses the CNS, the autonomic and enteric nervous system, and peripheral nerves and is vital for maintaining homeostasis. Signals from the brain control the secretory and sensory function of the gut, whilst the brain and gut communicate via physiological channels including the neuroendocrine, autonomic nervous system, neuroimmune pathways and molecules synthesised from gut microbes [51]. Since the gut microbiota is integral to the modulation of this communication at different levels (from the gut lumen to the CNS) and chronologically as we age, many have broadened the term to ‘microbiota-gut-brain axis’ [52]. Indeed, the existence of the microbiota-gut-brain axis is supported by substantial preclinical and human evidence, highlighting its effect on different cognitive domains. Firstly, germ-free (GF) mice show that the brain is markedly affected by the absence of microbiota, exhibiting deficiencies in learning, memory recognition and emotional behaviours [5356]. These behavioural changes were accompanied by altered brain-derived neurotrophic factor (BDNF) expression in the hippocampus [54, 57, 58], a molecule inherently linked with synaptic plasticity and cognitive function [5961], and significant microbiota-associated changes in the quantity of dopamine and activation of serotonin synthesis pathways [6265], suggesting an important role of microbiota in memory, brain health and behaviour. Secondly, chronic antibiotic depletion of microbiota populations alters tryptophan metabolism and the expression of key cognitive signalling molecules in the brain such as BDNF, N-methyl-d-aspartate receptor subunit 2B (NR2B), serotonin transporter, neuropeptide Y system, oxytocin and vasopressin [66, 67]. These changes are associated with long-lasting effects on cognition and increases in anxiety-related behaviours [66, 68]. Finally, administering specific prebiotics/probiotics modulates behaviour in both rodents and humans, including changes in depression, anxiety and stress [57, 6973], alongside changes in immune markers, hippocampal synaptic efficacy and tryptophan metabolism [74, 75].
As we age, microbiota composition and function changes [76]. In humans, this has been associated with a decrease in species diversity, a reduction in Clostridiales and Bifidobacterium and a rise in Proteobacteria and pathobionts such as Enterobacteriaceae [76, 77]. However, abnormal alterations in intestinal microbiota composition, as seen in early cognitive decline and AD [25, 78], are associated with local and systemic inflammation, and dysregulation of the microbiota-gut-brain axis [79]. Advances in sequencing technologies have enabled us to investigate the association between cognitive decline and gut dysbiosis at the phylum level. These studies have highlighted differences in taxonomic levels of Bacteroides, Firmicutes, Actinobacteria, Ruminococcus, Lachnospiraceae, and Selenomonadales between AD patients in comparison to controls [25, 78, 8082].Such dysregulation has been associated with an increase in inflammatory markers, cytokines and the permeability of the gut epithelial barrier (‘leaky gut’), resulting in excessive leakage of bioactivate molecules, such as short-chain fatty acids (SCFAs), kynurenines, melatonin, histamine, bile acids, and neurotransmitters, into the blood. The resulting increase in neuroactive products can no longer efficiently be removed by the body’s next barrier; the liver, and therefore can cause a variety of physiological changes directly and indirectly affecting the CNS, including further decrease in BBB function. In the elderly population, this dysregulation becomes particularly relevant, as the BBB becomes more permeable with age [83]. A more permeable BBB allows an increased influx of harmful blood components, including microbial metabolites, into the brain; a feature seen in AD patients (reviewed by [84]). This process promotes neuroinflammation and macrophage dysfunction, leading to neural injury and ultimately a reduction in cognitive function [85, 86].
As previously outlined, the gut can also influence the brain indirectly through the vagus nerve activation. The vagus nerve consists of 80% afferent and 20% efferent fibres [87]. Afferent fibres connect to all four layers of the digestive tract, but do not cross the epithelial layer and therefore are not in direct contact with gut microbiota. As a result, the microbiota activates these fibres indirectly via the release of metabolites or bacterial products. Enteroendocrine cells (ECCs) make up approximately 1% of intestinal epithelial cells and can detect signals from the microbiota through toll-like receptors (TLR), capable of identifying bacterial compound such as lipopolysaccharides (LPS) [88], or through receptors activated by microbiota-derived metabolites such as SCFAs [89]. ECCs can subsequently interact with vagal afferent fibres through the release of serotonin or gut hormones [90, 91]. This indirect signalling between the gut microbiota and the brain via the vagus nerve can modulate certain cognitive functions. For example, rodents fed with the probiotic L. rhamnosus for 28 days had a decrease in anxiety-related behaviour, whilst inducing region-dependent alterations in γ-aminobutyric acid (GABA) receptor [92]. Importantly, this result only occurred with an intact vagus nerve, as mice undergoing a vagotomy did not display these behavioural and neurochemical changes. Similarly, in a colitis model, the normalisation of anxiety-like behaviours by the probiotic Bifidobacterium longum NCC3001 was found to be vagally dependent [93]. However, the total effects of the microbiome are not solely dependent on the vagus nerve stimulation, as mice orally receiving a mixed antimicrobial treatment had altered exploratory behaviour and hippocampal BDNF, independently of vagal integrity [94]. Together, these data emphasise that while the vagus nerve provides a crucial bridge allowing communication between the gut, its microbiome and brain, there are also other essential routes of communication comprising the microbiota-gut-brain axis, indicating its complex connectivity.

The microbiota-gut-liver-brain axis

The relationship between the gut, liver and brain has increasingly been highlighted in recent years due to a high prevalence of liver disease, which is commonly accompanied by clear and global cognitive impairment (hepatic encephalopathy) [95, 96]. The gut and liver are linked by the portal vein, biliary tract and systemic circulation, allowing microbial and host-derived metabolites to influence liver function. Conversely, the liver acts as a vital barrier, removing potentially harmful compounds from the blood using a range of hepatic immune cells, including Kupffer cells, hepatic stellate cells and natural killer cells [97], modulating the concentration of metabolites directly and indirectly influencing the CNS. The liver also controls unrestricted bacterial growth in the gut, maintaining gut eubiosis, through the transport of bile salts through the biliary tract into the intestinal lumen leading to the secretion of antimicrobial compounds [98, 99]. For example, bile acids can bind to the FXR receptor in enterocytes, initiating the production of antimicrobial peptides such as angiogenin 1 and RNAse family member 4, which can inhibit bacterial overgrowth in the gut and intestinal barrier dysfunction [100]. Gut dysbiosis causes an imbalance of microbial and host-derived products, reducing the epithelial barrier function and causing increased leakage in the system. Long-term, this process can initiate metabolic disorders in the liver, promoting liver damage (reviewed by [101]). As such, liver damage has been found to correlate with the severity of gut dysbiosis [102]. Since a diseased liver cannot effectively remove harmful products from the blood or inhibit the overgrowth of bacteria [103105], this process can accelerate microbiota-gut-brain axis dysregulation and ultimately cognitive decline. Thus, when considering the occurrence of cognitive decline associated with microbial-derived metabolites, the role and function of the liver cannot be ignored.

Microbial-derived metabolites and cognitive decline

Bile acids

Humans produce large, hydrophilic pools of primary bile acids (BA) from cholesterol in the liver that are secreted into bile (Fig. 2). BAs are largely synthesised via two biosynthetic pathways: the classical pathway and the alternative pathway [106]. The classic pathway produces the majority of BAs in humans (~ 90%) and is initiated by the cholesterol 7α-hydroxylase (CYP7A1) enzyme to synthesise the primary BAs cholic acid (CA) and chenodeoxycholic acid (CDCA) [107]. The alternative pathway contributes less than 10% of BA synthesis (with more minor pathways contributing the remainder) and is initiated by sterol 27-hydroxylase (CYP27A1) [108]. After synthesis in the liver, CA and CDCA can be conjugated with hydrophilic taurine or glycine residues before they are secreted from hepatocytes into the bile canaliculi. They are stored in the gallbladder ready to be distributed into the small intestine following a meal to expedite digestion and emulsify dietary lipids and fat-soluble vitamins. Once secreted into the small intestine, more hydrophobic secondary BAs are formed by gut bacteria and are subsequently excreted or reabsorbed in the ileum to enter the enterohepatic circulation and recycle back to the liver [109]. This efficient process ensures BAs are recycled between 4 to 12 times a day [106].
In the brain, cholesterol can be metabolised by a final pathway known as the neural cholesterol pathway. As the brain is one of the more sensitive organs to hypercholesterolemia, this cholesterol breakdown is essential to maintaining brain health [110]. Excess cholesterol becomes oxidised into 24- and 25-hydroxycholesterol by cholesterol 24-hydroxylase (CYP46A1), an enzyme primarily expressed in the brain [111]. Once 24(S)-hydroxycholesterol is formed, it can pass through the BBB and enter circulation. From here, 24(S)-hydroxycholesterol travels back to the liver to be metabolised by CYP39A1 and continue in BA synthesis [111]. In mice with mutated CYP46A1 function, 24(S)-hydroxycholesterol is not formed and is associated with impairments in spatial, associative and motor learning, highlighting the importance of this pathway for maintaining cognitive function [112].

BAs in the brain

Over 20 conjugated and unconjugated BAs and their receptors have been reported in both human and rodent brains [110, 113115], suggesting BAs can not only cross the BBB but also bind to nuclear receptors and initiate physiological responses [115, 116]. However, the mechanism by which BAs cross the BBB is still uncertain. Unconjugated BAs may diffuse across the BBB as CA, CDCA and deoxycholic acid (DCA) are known to diffuse across phospholipid bilayers [117] and concentrations in the brain correlate with serum levels [113]. On the other hand, conjugated bile salts must cross the BBB via active transport due to their hydrophilic anionic structure at physiological pH [118, 119]. Indeed, members of the solute carrier (SLC) family, such as the organic anion transporting polypeptides (OATP1A4 and 1C1) [120] and the apical sodium-dependent bile acid transporter (ASBT or SLC10A2) [121], and members of the ATP-binding cassette transporters (ABC) family such as ABCC2 and ABCC4 [122, 123] have been identified in the brain. Conversely, Baloni and colleagues through a large-scale transcriptomics analysis of 2,114 post-mortem brains identified only three BA transporters (ABCC1, ABCC4 and SLC51A/SLC51B) in the brain [110]. The primary role of these transporters is to reduce the concentration of cytotoxic molecules by transporting them into the bloodstream [124]. Yet, since these transporters occur on both the basolateral (blood-facing) and apical (brain-facing) side [125], they may also transport molecules into the CNS from systemic circulation, indicating a potential endogenous signalling role of BAs in the brain. However, there is still a lack of direct evidence of in vivo transport of BA over the BBB [125].

BAs and cognitive function

While BA function in the GI tract is well-characterised, significantly fewer studies investigate their effect in the brain, limiting our knowledge [111]. Accumulating evidence suggests that cognition can be influenced by the dysregulation of BA synthesis and metabolism. Indeed, BAs profiles are reportedly altered in cases of MCI and AD, with an increase in cytotoxic secondary BAs and a decrease in primary BAs, suggesting a role of the gut microbiome in the disease progression [126]. Specifically, increased serum concentrations of the secondary BA DCA have been observed in AD patients. DCA has been previously linked with the presence of cognitive symptoms [127] and can modulate mitochondrial pathways causing apoptosis in a variety of tissues and cell types [128]. BA dysbiosis, resulting from either liver or microbiota dysfunction, has been subsequently linked to changes in gut permeability, possibly through FXR and TGR5 receptor signalling, and inflammation, promoting further bacterial dysbiosis in the gut [129]. Inflammation is also a known trigger of microglial activation and reduced neuroplasticity [130], possibly through the production of reactive oxygen species [131], highly reactive chemical molecules that have been previously linked with cognitive decline and AD [132, 133]. Although, some have proposed an important physiological role of ROS in brain metabolic signalling [134].
Alternatively, some BAs have been reported to have neuroprotective effects in the brain (for summary see Table 1). The primary BA CA has been identified as an LXR ligand, which in turn promoted midbrain neural development and neurogenesis in zebrafish [135]. Tauroursodeoxycholic acid (TUDCA), a secondary conjugated BA, can suppress amyloid- β (Aβ) -induced apoptosis in neuronal cell cultures and rodent neurons through the inhibition of the E2F-1/p53/BAX pathway [136, 137]. Similarly, in APP/PS1 double-transgenic mice, providing a TUDCA enriched diet for 6 months reduced Aβ aggregates, neuronal apoptosis, memory deficits and phosphorylation of TAU [138140]. TUDCA has also been shown to induce anti-inflammatory effects in a mouse model of acute neuroinflammation through its binding and activation of G protein‐coupled bile acid receptor 1/Takeda G protein‐coupled receptor 5 (GPBAR1/TGR5), a receptor expressed on microglia [141]. Finally, in adult rats, TUDCA also enhanced neural stem cell proliferation and early neurogenesis [142], processes that are significantly diminished in AD (reviewed by [143]), with some research suggesting increasing neurogenesis may counteract AD pathological outcomes. Together these findings provide convincing evidence that cognition can be influenced by BAs. Yet further research is required to determine the involvement of specific BA transporters and receptors, as well as the subsequent mechanisms in their neuroprotective and detrimental effects.
Table 1
Bile acids and their impact on cognition and dementia
Bile Acid
In Vitro/ In Vivo (species)
Model
Findings
Reference
CA (Primary Bile Acid)
In Vivo (Male Sprague-Drawly rats)
Ibotenic Acid-Induced
Dementia Model
A combination of administering baicalin, jasminoidin and cholic acid improved cognitive performance through the promotion of pathways related to neuroprotection and neurogenesis
[144]
In Vivo (Zebrafish)
Zebrafish embryos exposed to a cholic acid-treated medium
Cholic acid was identified as a new Lxr ligand, which in turn promoted neural development and neurogenesis in the midbrain of zebrafish
[137]
CDCA (Primary Bile Acid)
In Vivo (Adult male Wistar rats)
AlCl3 induced AD
CDCA treatment reduces neurotoxicity and cognitive decline via increased insulin signalling
[145]
In Vitro
Primary dissociated cultures of the posterior hypothalamus
CDCA is an antagonist for NMDA and GABAA receptors and can significantly reduce neuronal firing
[146]
TCA (Primary Conjugated Bile Acid)
In Vivo (human)
Human brain tissue with AD pathology vs age-matched healthy controls
TCA was significantly lower (p = 0.01) in AD patients than in age-matched controls
[117]
DCA (Secondary Bile Acid)
In Vitro
BCS-TC2 human colon adenocarcinoma cells
DCA modulates mitochondrial pathways causing apoptosis
[130]
In Vivo (human)
Serum samples from AD patients, amnesic MCI patients and healthy controls
DCA was increased in amnesic MCI and AD in comparison to healthy controls and correlated with cognitive symptoms
[129]
LCA (Secondary Bile Acid)
In Vivo (human)
Plasma samples from patients with AD, MCI and healthy controls
LCA was significantly higher in AD patients (p = 0.004) compared to healthy controls
[147]
UDCA (Secondary Bile Acid)
In Vitro
BV-2 microglial cell line
UDCA can initiate an anti-inflammatory effect by inhibiting NF-κB activation
[148]
TUDCA (Secondary Conjugated Bile Acid)
In Vitro
Neuron cell cultures and primary rat neurons
Inhibition of the E2F-1/p53/Bax pathway, leading to suppression of Aβ-induced apoptosis
[138]
In Vitro
Primary cultures of rat cortical and hippocampal neurons
Reduction in synaptic deficits induced by Aβ through inhibiting the downregulation of postsynaptic density protein-95, leading to a reduction in neuronal death
[149]
In Vivo (mouse)
AD model: APP/PS1 double transgenic mice
Dietary TUDCA provided for 6 months decreased Aβ aggregation and enhanced memory retention
[141]
In Vivo (mouse)
AD model: APP/PS1 double transgenic mice
Dietary TUDCA provided for 6 months decreased hippocampal and prefrontal amyloid deposition and inhibited spatial, recognition and contextual memory deficiencies
[150]
In Vivo (mouse)
AD model: APP/PS1 double transgenic mice
Intraperitoneal injections of TUDCA decreased Aβ deposition, glycogen synthase kinase 3β activity, phosphorylation of τ, and neuroinflammation
[142]
In Vitro
Aβ-treated primary rat cortical neurons
TUDCA prevented Aβ induced cytochrome c release and neuronal death through the PI3K signalling pathway
[151]
In Vitro
Aβ-treated primary rat cortical neurons
TUDCA reduced Aβ induced apoptosis through the binding to mineralocorticoid receptors
[140]

BA as a risk factor of cognitive decline

The association between BAs and cognitive decline, in particular with known AD pathologies [152], has raised speculations that BA profiles could be used as a risk factor of cognitive decline. Currently, there is limited research into the topic. However, Olazarάn and colleagues investigated a large cohort of patients with MCI and AD and identified DCA as being independently associated with the presence of cognitive symptoms [127]. Mapstone et al. identified seven blood-based markers which included glycoursodeoxycholic acid (GUDCA) and could predict the onset of AD or amnestic MCI with 2–3 years with an accuracy of over 90% [153]. Similarly, Marksteiner and colleagues were able to differentiate between healthy controls and AD patients from the concentration of lithocholic acid (LCA) in plasma [154]. However, it should be noted this study utilised a relatively small sample size (n = 80) and did not control for the effects of varied diets between individuals, warranting further investigation into the use of BAs as risk factors of cognitive decline.

TMAO

Trimethylamine N-oxide (TMAO) is a microbial-dependent metabolite generated by the breakdown of dietary fish, meat and fat [147, 155]. Trimethylamine (TMA), the precursor to TMAO, is produced from the metabolism of choline, L-carnitine and phosphatidylcholine by anaerobic microbes in the gut, predominantly located in the small intestine (Fig. 2) [144, 145]. TMA subsequently travels through the portal vein to the liver where it is oxidised by flavin-containing monooxygenase 1 and 3 (FMO1 and FMO3) to form TMAO [146]. Once formed, TMAO can enter the systemic circulation, hence TMAO plasma levels (typically 3 μmol/L in healthy individuals [148]) have been found to correlate with the gut microbial composition [149].

TMAO and the brain

In vivo studies have identified TMAO in the CSF of both mice and humans, implying that circulating TMAO can influence the CNS [147, 150]. The high concentrations of TMAO detected in the human CSF suggests liver-derived TMAO can cross the BBB, however, the penetration mechanism is unclear [151]. It is also possible a portion of TMAO found in the brain may be synthesised de novo, as FMO3, the enzyme required to convert TMA to TMAO, has been detected in the adult brain [156].

TMAO and cognitive decline

Over the last decade, TMAO has received increased attention in medical studies due to its links with cardiovascular diseases [157], obesity, diabetes [158], chronic kidney disease [159], metabolic syndrome [160], brain ageing and cognitive impairment [161] and neurodegenerative disorders such as AD [147]. However, the influence of TMAO on cognition is unclear. In fact, there is much controversy as to whether TMAO promotes a positive or detrimental effect on the brain.
Both experimental [161163] and clinical [164166] studies suggest high levels of TMAO may be causally linked to cognitive decline. Vogt and colleagues discovered an increase in CSF TMAO in AD patients in comparison to controls, suggesting the metabolite may contribute to decreasing neurological function [147]. However, a recent Mendelian randomisation study disputes this relationship [167].
The mechanisms by which TMAO may contribute to cognitive decline remain broad and unclear. TMAO reportedly modulates lipid and hormonal homeostasis [147], encourages platelet hyperreactivity via the enhancement of stimulus-dependent release of calcium ions [168], modifies cholesterol and sterol breakdown, reduces reverse cholesterol transport [169], and increases endothelial dysfunction through the induction of the NLRP3 inflammasome [170]. Rodents fed supraphysiological doses of TMAO also suggest the metabolite promotes neuronal senescence, oxidative stress, mitochondrial dysfunction and prevents mTOR signalling [161]. Furthermore, TMAO is known to upregulate macrophage scavenger receptors and induce CD68 expression [169, 171], a marker known to correlate with cognitive impairment in rodents [172].
High circulating TMAO may also promote neuroinflammation, a recognised mediator of cognitive ageing and neurological function [173, 174], by increasing brain NF-κB and proinflammatory cytokines, thereby promoting proinflammatory signalling pathways [164]. Brunt and colleagues suggested that elevated TMAO in plasma and the brain can stimulate astrocytes, neuroinflammation and reduce cognitive function, especially in the subdomain of memory [164]. High circulating concentrations of TMAO also downregulated the antioxidant enzyme methionine sulfoxide reductase A in the hippocampus of aged rats with induced cognitive impairment by sevoflurane exposure [175]. This downregulation is suggested to sensitise the hippocampus to oxidative stress, promoting microglial mediated neuroinflammation and cognitive impairment. Collectively, studies indicate a detrimental effect of TMAO when modulated above physiologically relevant concentrations.
In line with this, reducing TMAO has been shown to alleviate cognitive impairment. 3,3-Dimethyl-1-butanol, an inhibitor of microbial TMA formation, reduced cognitive decline, long term potentiation and pathological deterioration in AD transgenic mice [162]. Similarly, the probiotic L. Plantarum decreased circulating TMAO levels, alleviating cognitive impairments and pathological deterioration, exhibiting the potential modulation of the gut microbiome for therapeutic benefit [176].
In contrast to the substantial evidence supporting a detrimental effect of TMAO upon the brain, several studies suggest TMAO may exert a neuroprotective effect when within normal physiological ranges (plasma levels ~ 0.5–5 µM). Hoyles and colleagues, using a mixed in vitro endothelial cell culture and in vivo rodent model approach, discovered that TMAO can enhance and protect BBB integrity through modulation of the actin cytoskeleton and tight junctions [177]. Here, administering TMAO reduced paracellular permeability, likely due to an increase in annexin A1 expression. TMAO, therefore, may promote BBB function and help protect the brain from an influx of cytotoxic molecules. Interestingly, TMA, the precursor to TMAO, was found to have a deleterious effect on endothelial barrier integrity in rodents, inducing actin stress fibre formation and leading to increased presence in the CNS [178].
TMAO is a naturally occurring osmolyte and as such has been found to stimulate TAU-induced tubulin assembly in vitro [179]. TMAO, therefore, can promote and enhance microtubule assembly in hyperphosphorylated and most mutant TAU proteins, decreasing microtubule disassembly and neuronal death; two hallmark features of AD [180]. TMAO overcomes functional deficits caused by phosphorylation by lowering the critical concentration of tubulin required for assembly [181], with assembly occurring at a faster rate than wild-type TAU [180]. Therefore, as an osmolyte, and with its ability to favourably hydrate partially denatured proteins, TMAO has been suggested as a potential therapeutic approach in AD and other protein misfolding conditions [182].
Collectively, it seems plausible that TMAO affects the brain in a dose-dependent manner, as within a physiologically relevant range, TMAO possess neuroprotective potential. However, interpreting the relationship between systemic TMAO and cognition is further complicated by studies indicating wide inter and intra-individual variations in circulating TMAO levels [183]. TMAO concentrations vary with age [184], diet [169] and cholic acid levels (a BA known to induce FMO3 expression via FXR activation [185]); factors often not accounted for in association studies. In fact, plasma TMAO concentrations have been found to mirror an individual’s intake of whole grain, fish and vegetables [186]. TMAO levels are also influenced by renal clearance, as glomerular filtration rate is inversely related to plasma TMAO concentrations [159]. As a result, changes in plasma TMAO may be a consequence of an accumulation of factors unrelated to cognitive decline [187].

TMAO as a risk factor of cognitive decline

Due to TMAO’s high association with atherosclerosis and cardiovascular disease, TMAO has been considered a risk factor of vascular dementia [188]. However, a data-driven, hypothesis-free computational analysis into microbial metabolites and AD identified TMAO as one of the top potential biomarkers of neurodegeneration, successfully predicting changes in memory and fluid cognition in ageing individuals [189]. These results show promising potential for use of TMAO as a risk factor of cognitive decline. However, the current contrasting evidence surrounding the relationship necessitates further in vivo investigation.

Amino acid-microbiota-derived metabolites

Tryptophan

Tryptophan is an essential aromatic amino acid that cannot be synthesised by animal cells [190]. Humans, therefore, need to attain tryptophan through dietary sources such as fish, milk and chicken or, if vegetarian, seeds, soybeans and peas [191, 192]. Tryptophan is a biosynthetic precursor to numerous microbial and host metabolites, making it essential to human health [190]. Approximately 4–6% of tryptophan reaches the colon where gut microbiota metabolise it into a wide variety of molecules (Fig. 2), thereby limiting the availability of tryptophan for the host [193]. Evidence for the involvement of microbiota in tryptophan metabolism comes from GF mice, who display increased plasma tryptophan levels which are normalised after conventionalisation [62, 194].
Previous experimental reports implicate tryptophan and its derivatives in modulating human health and neurological function [195]. Gut microbiota can directly and indirectly modulate two major tryptophan metabolism pathways, the serotonin pathway and the kynurenine pathway (KP), affecting the concentration of various cognitively relevant metabolites and neurotransmitters [196198]. Conversely, the two pathways can negatively influence microbial proliferation and diversity [199]. Gut microbiota can also directly metabolise tryptophan into indole and its derivatives [200], which has also been associated with cognitive function [191].

The kynurenine pathway and cognitive decline

Around 90–95% of dietary tryptophan is metabolised by the KP, mainly taking place in the liver, forming the intermediates kynurenic acid, quinolinic acid, picolinic acid, 3-hydroxykynurenine (3-HK) and nicotinamide adenine dinucleotide, known as kynurenines [201]. Only tryptophan, 3-HK and kynurenine are known to readily cross the BBB. However, fluctuations in the systemic concentrations of these metabolites directly impacts KP metabolism in the CNS, including the synthesis of kynurenic acid and quinolinic acid in the brain [202]. Quinolinic acid, an endogenous neurotoxin, is known to activate N-methyl-D-aspartate (NMDA) receptors, increase neuronal activity, elevate intracellular calcium concentrations and modulate BBB integrity [203]. Quinolinic acid can also increase neuronal glutamate release whilst inhibiting its reuptake by astrocytes and inhibiting glutamate synthetase synthase (an enzyme playing a crucial role in the glutamate metabolism in astrocytes) to produce a cytotoxic response [204, 205]. Kynurenic acid, on the other hand, plays a neuroprotective role against quinolinic acid’s toxicity, acting as an antagonist on both glycine and glutamate modulatory sites of NMDA receptors at high and low concentrations respectively [198]. However, the abnormal build-up of kynurenic acid can induce glutamatergic hypofunction, possibly disturbing cognitive functioning [205].
Accumulating evidence implicates the KP in AD progression and inflammatory responses [206]. Increased plasma concentrations of the cytotoxic quinolinic acid (from 192 to 334 nM) and reduced concentrations of tryptophan (from 29.83 mM to 22.09 mM) and neuroprotective kynurenic acid (from 30.94 nM to 20.85 nM) has been associated with AD patients in comparison to healthy controls [207]. Unbalanced upregulation of the KP may trigger a degree of injury to the surrounding tissues, playing a role in neurodegeneration [208]. Previous studies have found an inverse relationship between KP activation and cognitive performance [209].
In a cognitively healthy population, increased inflammatory markers are related to poor cognitive performance [210]. In AD, indoleamine 2, 3-dioxygenase (IDO), the enzyme responsible for catabolising tryptophan into products that enter the KP, is stimulated through proinflammatory cytokine activity, including interferon-gamma (IFN-γ) [211], interleukin-12 (IL-12), interleukin-18 (IL-18) [212], and the Aβ 1–42 fragment [213]. Complex neuroinflammation in the CNS is linked with AD development. Microglia and astrocytes, which contain all of the enzymes necessary for the KP, are the primary effectors of neuroinflammation in AD [214]. The edge of senile plaques in the hippocampus of post-mortem AD brain tissue has the greatest amounts of IDO and quinolinic acid expressed by microglia and astrocytes [215]. Activated microglia are the main source of quinolinic acid throughout neuroinflammation [216]. Quinolinic acid produces hyperphosphorylation of TAU in human cortical neurones, cytotoxicity in astrocytes and neurons, astrocytic activation and astrogliosis [208, 217]. Together, these studies strongly suggest the involvement of IDO and KP metabolism in neuroinflammation and cognitive impairment.
Accordingly, the KP is a well-rationalised therapeutic target for improving cognition. Several proof-of-concept studies using known KP pathway modulators, such as the kynurenine monooxygenase (KMO) inhibitor JM6, prevents spatial memory deficits, anxiety-related behaviours, and synaptic loss in APP Tg mice [218]. In addition, the IDO-1 inhibitor, coptisine, decreases the activation of microglia and astrocytes in APP/PS1 mice, preventing neuronal loss and improving cognitive function [208]. However, the specific relationship between tryptophan depletion or supplementation and the modulation of KP intermediates remains unclear [219221].

Serotonin pathway and cognitive decline

Approximately 3% of dietary tryptophan is required to produce serotonin (5-hydroxytryptamine (5-HT)) and melatonin [193]. 5-HT is primarily found in the GI tract, blood platelets and the CNS and is synthesised via a two-stage enzymatic reaction involving tryptophan hydroxylase and aromatic amino acid decarboxylase. Serotonin synthesised in the GI tract cannot cross over the BBB under healthy conditions [222]. Tryptophan, on the other hand, can enter the CNS via carrier proteins [223]. Therefore, the gut microbiota importantly regulates tryptophan availability for serotonin synthesis in the CNS.
Enzymes such as tryptophan hydroxylase and IDO balance the ratio of tryptophan metabolism via the KP and serotonin pathways [224]. A shift in tryptophan metabolism to the KP decreases the availability of tryptophan in the serotonin pathway, consequently reducing serotonin availability for the host [225]. Serotonin plays a vital role in behaviours requiring high cognitive demand [196]. Reductions in serotonin, therefore, are frequently linked with declines in learning, memory consolidation [226] and long-term memory [227]. As such, serotonin is associated with neurological disorders such as depression [228] and AD [229], resulting in treatment options such as selective serotonin reuptake inhibitors (SSRI) to increase 5-HT neurotransmission and improve mood in the context of depression. In rodents, administering tryptophan orally, thereby increasing 5-HT neurotransmission, was found to improve memory acquisition, consolidation and storage [230], whilst daily tryptophan injections improved spatial memory [231]. Together, this evidence strongly suggests a link between cognitive decline and tryptophan through changes in tryptophan metabolism.

Other tryptophan metabolites

Numerous studies have identified abnormal tryptophan metabolism in patients with cognitive decline [71, 191, 232]. Although most studies link this association with the KP and its intermediates, other tryptophan metabolites, such as indole and its derivatives, may play a role. Bacterial tryptophan catabolites tryptamine, skatole, indole, indole-3- acetic acid (IAA), indole-3- acrylic acid (IA), indole-3-aldehyde (IAld), indole propionic acid (IPA), indoxyl-3-sulfate (I3S) and indole-3-lactic acid (ILA) are ligands of the aryl hydrocarbon receptor (AhR) [233238]. AhR is a transcription factor widely expressed by cells in the immune system and known to play a role in inflammation, a factor highly associated with ageing and cognitive decline [239]. Antibiotic-treated mice administered with indole, I3S, IPA and IAld were found to have reduced CNS inflammation via AhR activation in astrocytes [240]. Wei and colleagues discovered activation of the AhR by indole could promote neurogenesis in the adult mouse hippocampus [241]. Interestingly, this result was found to be ligand specific as kynurenine, another known AhR ligand, failed to replicate these findings.
Both in vitro and in vivo studies have associated indoles with enhancing intestinal barrier function by increasing gene expression associated with the maintenance of epithelial cell structure and function [242, 243], thereby decreasing the concentration of neuroactive products in circulation [79]. The activation of AhR also helps preserve epithelial barrier function by maintaining tight junction integrity [244]. IA may also have anti-inflammatory and anti-oxidative effects in LPS-activated human peripheral blood mononuclear cells (PBMCs) by reducing IL-6 and IL-1β secretion and activation of the NRF2-ARE pathway [245], a pathway suggested to ameliorate cognitive deficits [246, 247].

Tryptophan & derivatives as risk factors of cognitive decline

Although no research studies to date have exclusively investigated the use of tryptophan and its derivatives as a risk factor of cognitive decline, many reports have highlighted the potential use of tryptophan pathway imbalances to reveal signs of early cognitive decline [248]. Kaddurah-Daouk and colleagues concluded from studying CSF of AD patients that changes in tryptophan, as well as methionine, tyrosine, and purine metabolism occurred in MCI and AD, suggesting its potential use as a risk factor of cognitive decline [232]. However, the authors concluded that these changes may not be detectable in plasma, as the amount to which metabolic changes in blood mirror fluctuations in CSF remains to be investigated. Nevertheless, plasma metabolic profiling revealed changes in tryptophan metabolism in early cognitive decline, along with alterations in progesterone, lysophosphatidylcholine, L-phenylalanine, dihydrosphingosine and phytosphingosine [248]. Despite a lack of studies into the use of tryptophan and its derivates as a risk factor of cognitive decline, these studies highlight the possible future use of metabolomic profiling to detect early changes.

GABA

Through either direct access via the circulatory system, or via other communication routes, microbial metabolites may have the capacity to interfere and impact the function of the CNS [249, 250]. GABA is the main inhibitory neurotransmitter in the human brain and other parts of the body [251] and is reportedly unable to cross the BBB, although this statement is disputed [252]. This molecule was recently shown to be both a product of bacteria in the gut [250, 253, 254] and an important substrate for other gut community members [255]. It was also shown to have activity in rodent models of anxiety and visceral pain [256]. Bacterial strains, such as L. rhamnosus, can also modify GABA receptor expression and concentrations of glutamate (a precursor to GABA) and GABA in the brain [257].

Other bacterial amino acid metabolites

Amino acids present in dietary protein serve (particularly if overconsumed) as a fermentation substrate for bacteria in the large intestine. P—Cresol is the product of the microbial conversion of tyrosine, notably by the bacteria from the Coriobacteriaceae or Clostridium genera [258]. P—Cresol is a known uremic toxin and therefore can be further conjugated with sulphate by host cells to form p—cresyl sulphate (PCS) as part of the detoxification mechanism, promoting the removal of the metabolite by the kidneys.
P—Cresol is known to increase endothelial permeability in vitro through modulation of the actin cytoskeleton and adherens junctions [259], decreasing the gut’s barrier function. In the brain, p-cresol has been found to modulate dopamine turnover in Autism Spectrum Disorder BTBR mice, significantly increasing anxiety-like and hyperactive behaviours [260]. p-Cresol’s derivative, PCS, has been detected in the CSF of PD patients, suggesting the metabolite may cross the BBB and have a pathogenic effect in the CNS [261]. Although, this relationship may in part be due to the increased permeability of the BBB seen in PD [262]. PCS has been linked with cell death and dysfunction through oxidative stress, inflammation, impairment of mitochondrial dynamics and vascular disruption [263266]. Moreover, PCS administration in mice with nephrectomy contributed to neurological dysfunction through impairment of cell survival and neurogenesis, supporting its potential role in cognitive decline [267].
Imidazole propionate (ImP) has recently been uncovered as a microbially produced metabolite derived from the amino acid histidine [268]. Elevated serum concentrations of ImP are associated with low bacterial gene richness [269], a factor previously linked to low-grade inflammation, metabolic and inflammatory disorders [270]. ImP is also associated with a type 2 diabetes-related microbiome, stimulating impaired glucose metabolism through the initiation of the p38γ-mTOR1-S6K1 signalling pathway [268, 269, 271]. Type 2 diabetes is a well-characterised risk factor for dementia, with a 1.5–2.5-fold increase in dementia risk, suggesting an association between ImP, the gut microbiome and cognitive decline [272, 273].

Other emerging microbial-derived metabolites

During digestion, nutrients and bioactives (proteins, amino acids, polysaccharides, fibres, fats, polyphenols, etc.) are catabolised into host-derived and bacterial metabolites that have the ability to interact with the host’s cells and the resident gut microbiome [256, 274]. This continuous process results in the production of a wide array of chemicals representing a wealth of chemical classes. Dietary proteins are broken down into potentially active peptides [256] that are further transformed into bacterial products such as neurotransmitter amino acids like glutamate, glycine, aspartate, serine and GABA or polyamines [250, 275, 276]. Aromatic amino acids (tryptophan, tyrosine, and phenylalanine) and polyphenols yield a myriad of compounds during catabolism leading to the formation of simpler structures containing at least one phenol ring (phenols) which can then be further transformed by the host (sulfation, glucuronidation) before re-entering circulation [256, 274]. Additionally, dietary choline and niacin are substrates for the synthesis of molecules essential for cellular function in the brain namely acetylcholine and nicotinamide adenine dinucleotide (NAD +) precursors [277279], some of which have recently been shown to be synthesised by the gut microbiota [250, 254, 275, 278, 280]. These recent developments provide further evidence of the microbiota’s role in the production of beneficial signalling molecules that contribute to the maintenance of homeostasis during the ageing process.
As described earlier in the review, the BBB selectively allows circulating solutes to enter the CNS. Polyamines, polyphenols and some of their products (3-(3’-hydroxyphenyl)propionate and 3-hydroxybenzoate) [281] have been shown to cross the barrier even though the transfer seems somewhat limited [282286]. Meanwhile, nicotinamide and niacin, both precursors for NAD + , a coenzyme essential for the maintenance of the CNS, have the capacity to freely cross the BBB [278, 287]. More research on the topic is needed as the knowledge regarding their transport across BBB is in its infancy and partly based on in vitro models [282, 286, 288]. The question of whether the potential activity of those molecules on brain functions is either direct or based on interactions with peripheral systems remains open [289].

Short-chain fatty acids

Short-chain fatty acids (SCFAs) are small organic compounds primarily formed from microbial anaerobic fermentation of dietary fibres in the cecum and colon [290]. Accumulating evidence suggests SCFAs can attenuate cognitive decline, however, the underlying mechanisms remain unclear [291]. Recent studies suggest SCFAs can cross the BBB via monocarboxylate transporters present in endothelial cells within the brain tissue [292]. In fact, the uptake of SCFAs into the brain has formerly been exhibited in rodents following the injection of 14C-SCFAs into the carotid artery [293]. However, as well as crossing the BBB, SCFAs may also help preserve its integrity. GF mice with reduced SCFA levels were found to exhibit increased BBB permeability due to a reduction in the expression of tight junction proteins [294]. This BBB dysfunction was later reversed following conventionalisation with pathogen-free microbiota and monoculture strains producing SCFAs. Furthermore, in a rodent model of traumatic brain injury, the administration of sodium butyrate prevented BBB breakdown and promoted neurogenesis, highlighting a key role for SCFAs in not only maintaining CNS homeostasis but also possibly in preventing or reducing neural decline [295].
Select SCFAs can also manipulate epigenetic mechanisms, including DNA methylation, histone modification and their interactions, which may influence age-related cognitive changes [296]. Butyrate has been widely investigated due to its roles in receptor signalling and metabolic regulation. However, pharmacological studies also highlight butyrate as a histone deacetylase inhibitor, capable of increasing histone acetylation and inducing the expression of neurotrophic and anti-inflammatory genes [297, 298]. Accumulating evidence also suggests a role for butyrate in modifying DNA methylation [299301].
In the CNS, SCFAs have also been linked to reducing neuroinflammatory processes important for shaping brain function. Sodium butyrate has been linked to a decrease in microglial activation and pro-inflammatory cytokine secretion [297, 302]. Rodents supplemented with dietary acetate had a decrease in neuroglial activation by reducing the expression of pro-inflammatory cytokines and modulating brain histone acetylation [303]. Likewise, acetate also modulated inflammatory cytokines and signalling pathways in astrocyte primary culture [304]. In vivo and in vitro sodium butyrate administration was observed to have an anti-inflammatory role via protein kinase B (Akt)-RhoGTPase signalling and histone deacetylase inhibition, stimulating structural and functional changes in microglial towards a homeostatic profile [305]. SCFAs may also improve brain hypometabolism, a known contributor to neuronal dysfunction and AD, by providing an alternate substrate for energy metabolism [306, 307] highlighting a further potential method to mitigate and protect against neuroinflammatory processes. Nevertheless, the precise signalling underlying SCFA's influence within the CNS remains unclear, however, the inhibition of histone deacetylase has been put forward as the primary mechanism [308].
Select SCFAs may also moderate AD progression [309]. For example, valeric acid, butyric acid and propionic acid have been found in vitro to interfere with protein–protein interactions necessary for Aβ assemblies, potentially reducing the formation of toxic aggregates [290]. Yet, it remains unclear if SCFAs produced in the GI tract can play a role in protein misfolding in vivo [290]. However, Colombo and colleagues found GF AD mice display reduced circulatory SCFA concentrations and Aβ deposition, yet when supplemented with SCFAs, show an increase in Aβ plaque deposition, suggesting SCFA mediation [309]. In line with this, a clinical study into elderly individuals with ranging cognitive performance found an association between SCFA levels in the blood and brain amyloid deposition [310].
APOE genotype, the largest genetic risk factor of AD, has been associated with the composition of butyrate-producing microbiota in the gut [311]. Faecal samples from AD patients typically consist of an abundance of SCFAs, particularly butyrate-producing bacteria [312]. However, currently, there is no comparison of SCFA concentrations in age-matched healthy controls and therefore its use as a risk factor of cognitive decline is limited. This may in part be due to SCFA's volatile nature, making the compound difficult to detect in human samples, and current research also demonstrating low reproducibility. Notably, participant diet is rarely incorporated when quantifying SCFAs in research studies. Yet, the quantity and type of ingested fibre are known to have a large influence on microbial composition and, therefore, the concentration and type of SCFAs produced [306, 313]. Faecal SCFA concentrations also cannot fully signify production rates or accurate concentrations of SCFAs present in the colon, as significant percentages of SCFAs are immediately consumed locally in the gut [314, 315]. Changes in SCFA faecal concentrations, therefore, may be the result of either a fluctuation in its production or colonic absorption. Consequently, as of present, the knowledge on using SCFAs as a risk factor of cognitive decline is extremely limited.

Acetylcholine

Acetylcholine (ACh) is a common cholinergic neurotransmitter in the central and peripheral nervous systems. In the periphery, ACh can be produced from choline by numerous bacteria, including Lactobacillus plantarum, Bacillus subtilis, Escherichia coli, and Staphylococcus aureus [316, 317]. Within the microbiota-gut-brain axis, ACh can modulate intestinal motility, secretion and enteric neurotransmission. essential for the transmission of excitatory signals between neurons. Its dysregulation is closely linked with AD [318]. ACh cannot cross the BBB. Therefore, choline availability in the periphery importantly modulates the concentration of ACh in the CNS [319].

Dopamine

Dopamine is the leading catecholamine neurotransmitter in the mammalian CNS, playing a key role in a broad spectrum of cognitive abilities, including working memory, planning, selective attention abilities, motivation and reward processing [320, 321]. Dopaminergic transmission abnormalities have been linked to cognitive decline and numerous CNS disorders [reviewed by [322]]. Dopamine itself cannot cross the BBB. However, its precursor molecule L-3,4-dihydroxyphenylalanine (L-DOPA) can be transported across the BBB by large neutral amino acid transporters (LAT1) expressed on endothelial cells [323].
One approach to investigating the involvement of gut microbiota and their metabolites on cognitive decline is through the use of broad-spectrum antibiotics to induce gut dysbiosis by preventing the growth of select microorganisms. Administering an antibiotic cocktail of ampicillin, vancomycin, neomycin, metronidazole, and amphotericin B to the drinking water of male Swiss mice increased concentrations of L-DOPA and homovanillic acid (HVA), a dopamine-derived metabolite, in the amygdala in comparison to control mice [66]. However, no significant changes in dopamine levels were detected. Similarly, Hoban and colleagues observed increased concentrations of L-DOPA in the prefrontal cortex and hippocampus of adult male Sprague–Dawley rats after supplying an antibiotic cocktail of ampicillin, vancomycin, ciprofloxacin, imipenem, and metronidazole for 42 days. Together, these studies suggest antibiotic-induced dysbiosis can impact dopamine neurochemistry in the rodent brain.
As discussed earlier in this review, one mechanism in which intestinal bacteria can communicate with the brain is via stimulation of the vagus nerve. Interestingly, Han and colleagues found stimulation of vagal afferent fibres from the upper intestinal tract can promote dopamine release in the brain of mice [324]. Dopamine can also be synthesised in the intestinal lumen by gut microbes [325]. Indeed, gut microbes belonging to the genus Prevotella, Bacteroides, Lactobacillus, Bifidobacterium, Clostridium, Enterococcus, and Ruminococcus have been suggested to modulate dopaminergic activity and influence Parkinson’s disease (PD) pathophysiology (reviewed by [326]). Gut microbiota can also increase luminal dopamine bioavailability through enzymes such as β- glucuronidase [325] and tyrosine decarboxylase [327], demonstrating a key role of the gut microbiota in modulating peripheral dopamine levels. Interestingly, plasma L-DOPA levels were found to be significantly increased in probable AD patients in comparison to controls, whereas dopamine concentrations were decreased [328].

Polyphenols

Both plant-based foods, rich in polyphenols, and dietary proteins are substrates for colonic bacteria which produce phenolic compounds that potentially benefit human health [256, 274, 329331]. The current research on the impact of beneficial and harmful microbial phenolic compounds 3-(3’-hydroxyphenyl) propionate, 3-hydroxybenzoate, indoxyl sulfate, p-cresol sulfate on the brain is at an early stage but determining the role of those products on the gut-brain axis is a promising field of research [256, 274, 281, 330]. Frolinger and colleagues have recently demonstrated a link between polyphenolic products produced by gut microbiota and cognitive resilience in rats [332] and Esteban-Fernández et al. showed that 3-hydroxyphenylacetic acid and other microbial-derived phenolic compounds have a neuroprotective effect on a human neuroblastoma cell line [333]. Metabolomics on circulating metabolites also correlated levels of catabolites of the phenylalanine and tyrosine pathways to poorer mini-mental state examination (MMSE) scores in a cohort of hypertensive patients [334]. Mostly driven by in vitro and animal-based studies, research on the effect of phenolic compounds are nonetheless accruing evidence that microbial phenolic compounds could play a role in brain metabolism [286].

Polyamines

Polyamines were first described in 1677 by Antonie van Leewenhoek who reported the presence of crystals in human semen [284]. It was much later in 1924 that Dudley and colleagues characterised one of their components, isolating spermine from bovine brain [284]. Polyamines are small molecules essential to cell growth and ubiquitous to all life forms. Most of the polyamine pool is bound to RNA conferring an important role to polyamines in stabilising this molecule and contributing to the process of its translation [284]. Putrescine, spermidine and spermine are synthesized by plants, mammals and bacteria and represent the most abundant polyamines found in tissues [335337].
Levels of polyamines found in mice decline with brain ageing [65, 338] but in humans, only spermidine levels seem to change over time reaching their highest level at 40 years of age and remaining at similar levels thereafter [336]. Elevated levels have been reported in the brain from AD patients, where increased ornithine decarboxylase activity was found to be associated with AD processes [339].
They are abundant in food, quickly absorbed and distributed to all body tissues [340]. The polyamine content in the lower part of the intestine however is considered to be mostly of microbial origin [337]. Sustained circulating levels at an older age have been associated with enhanced longevity and the prevention of age-associated disease [340, 341]. Conversely, lower spermidine levels were found in blood from AD patients when compared to healthy individuals [342], a characteristic that was associated with lower MMSE scoring in another study conducted on older subjects in nursing homes [343].
Preserving adequate levels of polyamines could represent a valuable approach to maintaining the optimal functioning of cell metabolism and the prevention of chronic illnesses. Supplementation could be achieved by a regular intake of a polyamine-rich food diet or synthetic polyamines, or by the provision of microbial polyamine synthesis with probiotic supplements [344]. A recent study highlighted olive oil, fruits, cheese, and seafood as good sources of polyamines and that a steady intake may have a role in prolonging human life. The authors speculated that the mechanism involved could be a capacity for polyamines to counteract mild chronic inflammation and confer beneficial effects on vascular function [340]. Another study reported an association between spermidine intake estimated with a self-reported food frequency questionnaire and cortical thickness and hippocampal volume in older adults [345].
A study on mice fed arginine, a precursor for the synthesis of polyamines [276, 337] and probiotics LKM512 showed that long term administration offered protection against age-induced memory impairment via a mechanism involving the production of polyamines by microbiota [341]. The putative protective properties of polyamines are inhibition of cytokines release, inhibition of reactive oxygen species (ROS) production [346, 347], an impact on T-cell function and the maintenance of synaptic plasticity through the prevention of demyelination [347], thus presenting a defence against events that embody hallmarks of neurodegeneration [348, 349]. Polyamines also have the capacity to induce cytoprotective autophagy, a process involving the degradation of damaged organelles and biological debris [344, 350, 351]. They have a significant role in the maintenance of mitochondrial metabolic function. Indeed, spermidine is needed to chemically modify eukaryotic initiation factor 5A (eIF5A), an important enzyme involved in TCA cycle maintenance and electron transport chain in macrophages [352], highlighting an important role for this polyamine in the regulation of mitochondrial metabolism as any reduced activity can lead to neuroinflammation and neurodegeneration [348, 349].
As mentioned earlier, host bacterial production of polyamines was recently shown to delay senescence in mice [341]. Although the exact mechanisms were not elucidated, the authors speculated that autophagy [350, 351] may play a role in the preservation of memory capacity in ageing. This is further reinforced by recent studies on mice which showed that supplementation by spermidine and spermine may delay brain ageing and alleviate AD pathology via mechanisms involving autophagy, promotion of ATP, reduction of ROS [353, 354] and inflammation [355]. Maglione and colleagues showed that spermidine offered protection from synaptic alterations in the hippocampus of ageing mice extending their lifespan with a late treatment (starting at 18 months) [356].
The research on both autophagy and polyamines and cognitive health, which is getting traction, has recently translated into human trials. A long-term spermidine-rich treatment (dosage: 1.2 mg/day) was given to participants at risk of developing AD and found to be safe and well-tolerated [357]. This 3‐month randomized, placebo-controlled, double‐blind Phase II trial was shown to moderately improve the memory performance and to enhance the mnemonic discrimination ability of the treated individuals compared to the placebo-treated group [358]. The authors have designed a new trial using the same treatment that will expand the intervention period to 12 months and will include a larger cohort (n = 100 as opposed to n = 30) and a follow-up assessment 18 months after the start of the study [359]. In parallel, another group supplied older adults in nursing homes with spermidine added to bread for 3 months and evaluated the cognitive performance of the subjects with the CERAD-Plus test which consists of seven tests including an MMSE, a learn, recall and recognize a word list and phonemic fluid [360]. They reported a significant correlation between an intake of spermidine and improvement in cognitive performance, particularly in subjects with mild and moderate dementia. Their preliminary results offer hope for the possible mitigation of cognitive decline by enabling sustainable levels of polyamines in the body.

Nicotinamide

Energy and niacin and nicotinamide pathways are under tight homeostasis as shown by a lack of change in ATP and nicotinamides levels in the brain of colonised ex germ-free mice [65]. There is evidence that the levels of these molecules which are essential for the development and maintenance of CNS neurons decline with age and in neurogenerative states [277]. Promising results from an AD animal model led to a 24-week double-blind, placebo-controlled randomized clinical trial of nicotinamide in subjects with mild to moderate AD [279]. Unfortunately, this study failed to show an improvement in cognitive function in those volunteers. A similar study provided a 10-week supplementation with nicotinamide riboside (NR) to older individuals with MCI [361]. This trial resulted in demonstrating a positive effect on certain functions in the brain and frailty measures but like in the previous study, ultimately ended in a lack of change in cognitive measures [361]. This illustrates the complexity of translating results from animal studies to human trials with the dose, duration of the supplementation and environmental factors affecting the likelihood of a successful outcome. Nonetheless, recent studies showed that the gut microbiota can assist in the production of nicotinamide and other NAD + precursors [254] as demonstrated by Kim and colleagues who showed that treatment with nicotinamide mononucleotide in mice not only led to the microbial production of the deamidated product nicotinic acid mononucleotide, but also tripled the endogenous levels of NR, showing an important connection between the gut microbiome and the niacin and nicotinamide pathway [280].

Vitamin K

Vitamin K is a vital micronutrient that can be derived directly from our diet (phylloquinone) or intestinal microbiota (menaquinone) [362]. Vitamin K’s role is well-defined in blood coagulation and its beneficial effects on myelin integrity in the brain [363, 364]. Recent studies outline a positive relationship between vitamin K levels and cognitive performance [365, 366], and the administration of vitamin K antagonists to rats alters cognitive performance [367]. Increased dietary vitamin K intake is linked to a decrease in subjective memory complaints in an elderly cohort [368], whilst low concentrations of vitamin K in the blood have been correlated with the APOE-ε4 allele; the largest genetic risk factor of AD [363]. However, the direct relationship between microbial-derived vitamin K and cognition, and hence its use as a risk factor of cognitive decline, is yet to be uncovered.

Conclusions and future directions

The concept of microbial-derived metabolites influencing cognitive decline is gaining traction, with implications in the field of neuroscience, metabolomics and hepatology. However, due to the complexity of this relationship, the specific myriad of mechanisms responsible remain largely unknown, whilst defined roles of individual metabolites are only characterised for a select few (for a summary see Fig. 3). Therefore, amid this ambiguity, there remains a real need for additional research to highlight and validate key pathways, metabolites and mechanisms to further elucidate the influence of the microbiota-gut-brain axis on cognition [189].
There remain many challenges facing this growing field. Firstly, a lack of specificity limits our ability to distinguish between host vs microbiota-derived metabolite contribution as particularly if there is known co-metabolism, true microbial involvement may be masked or exaggerated. Secondly, as demonstrated by TMAO [362], some inconsistencies still exist among certain metabolites under context-specific vs dose-specific conditions. This may in part be due to heterogeneity between studies, with variations in study designs, methods of assessing cognitive performance and/or quantifying metabolites. As a result, further research ought to be collated via a more standardised methodology to increase comparability. Thirdly, the influence of the microbiome on cognition is not the totality of microbial metabolites produced in the gut as the varying capabilities of these metabolites to penetrate the BBB play a key role [79]. Consequently, the mechanisms used by many metabolites to cross the BBB are still unknown and some may even be synthesised de novo. Fourthly, from a translational perspective, the described research has largely been conducted in animals. Establishing whether these findings translate to humans will be crucial yet challenging due to the greater complexity and environmental exposure humans encounter, in turn shaping each individual’s microbiome [369]. Finally, understanding these highly complex systems, particularly as we move more towards human studies, requires the continued advancement of computational and statistical methods to obtain and implement multi-omics and longitudinal data necessary for a comprehensive approach [330]. Together, these challenges render it difficult to outline specific host-microbiota interactions in a mechanistic manner, which is needed to advance the field past associations towards implementable microbiota-driven targets.
Nevertheless, the wealth of association studies highlight a positive future for the use of microbiota-derived metabolites as risk factors of cognitive decline [370]. Future studies should progress using robust and replicable metabolic phenotyping across various stages of cognitive decline in humans. Recent advancements using this approach are underway, utilising metabolic phenotyping of urine [371] and blood [153, 372] to predict incipient AD with high degrees of accuracy. However, several studies using comparable approaches have not been able to replicate these findings [373, 374]. This may be due to intrinsic difficulties surrounding the heterogeneity of cognitive decline seen in neurodegenerative diseases and the variety of analytical methods used in metabolic profiling, ranging from 1H-NMR, LC–MS/MS, GC–MS, UHPLC-MS and CE-MS [371]. Hence, currently, the literature is too scarce to support the implementation of metabolite-derived risk factors in clinical practice.
In conclusion, although significant work remains to fully understand the role of microbial-derived metabolites as key mediators of cognitive decline, identifying modifiable factors that promote healthy ageing and cognition will have vital clinical implications in today’s growing elderly population, whilst also helping to identify novel underlying mechanism.

Acknowledgements

Figures created with BioRender.com.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Alzheimer’s Association. Alzheimer’s disease facts and figures. Alzheimer’s & Dementia. 2019;2019(15):321–87. Alzheimer’s Association. Alzheimer’s disease facts and figures. Alzheimer’s & Dementia. 2019;2019(15):321–87.
4.
Zurück zum Zitat Tiwari S, Atluri V, Kaushik A, Yndart A, Nair M. Alzheimer’s disease: pathogenesis, diagnostics, and therapeutics. Int J Nanomedicine. 2019;14:5541–54.PubMedPubMedCentralCrossRef Tiwari S, Atluri V, Kaushik A, Yndart A, Nair M. Alzheimer’s disease: pathogenesis, diagnostics, and therapeutics. Int J Nanomedicine. 2019;14:5541–54.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Kivipelto M, Helkala E-L, Laakso MP, Hänninen T, Hallikainen M, Alhainen K, et al. Midlife vascular risk factors and Alzheimer’s disease in later life: longitudinal, population based study. BMJ British Medical Journal Publishing Group. 2001;322:1447–51.CrossRef Kivipelto M, Helkala E-L, Laakso MP, Hänninen T, Hallikainen M, Alhainen K, et al. Midlife vascular risk factors and Alzheimer’s disease in later life: longitudinal, population based study. BMJ British Medical Journal Publishing Group. 2001;322:1447–51.CrossRef
6.
Zurück zum Zitat Balagopal PB, de Ferranti SD, Cook S, Daniels SR, Gidding SS, Hayman LL, et al. Nontraditional risk factors and biomarkers for cardiovascular disease: mechanistic, research, and clinical considerations for youth. Circulation. American Heart Association. 2011;123:2749–69. Balagopal PB, de Ferranti SD, Cook S, Daniels SR, Gidding SS, Hayman LL, et al. Nontraditional risk factors and biomarkers for cardiovascular disease: mechanistic, research, and clinical considerations for youth. Circulation. American Heart Association. 2011;123:2749–69.
7.
8.
Zurück zum Zitat Profenno LA, Porsteinsson AP, Faraone SV. Meta-Analysis of Alzheimer’s Disease Risk with Obesity, Diabetes, and Related Disorders. Biol Psychiat. 2010;67:505–12.PubMedCrossRef Profenno LA, Porsteinsson AP, Faraone SV. Meta-Analysis of Alzheimer’s Disease Risk with Obesity, Diabetes, and Related Disorders. Biol Psychiat. 2010;67:505–12.PubMedCrossRef
10.
Zurück zum Zitat Knopman D, Boland LL, Mosley T, Howard G, Liao D, Szklo M, et al. Cardiovascular risk factors and cognitive decline in middle-aged adults. Neurology. Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology. 2001;56:42–8. Knopman D, Boland LL, Mosley T, Howard G, Liao D, Szklo M, et al. Cardiovascular risk factors and cognitive decline in middle-aged adults. Neurology. Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology. 2001;56:42–8.
11.
Zurück zum Zitat Dye L, Boyle NB, Champ C, Lawton C. The relationship between obesity and cognitive health and decline. Proceedings of the Nutrition Society. Cambridge University Press. 2017;76:443–54. Dye L, Boyle NB, Champ C, Lawton C. The relationship between obesity and cognitive health and decline. Proceedings of the Nutrition Society. Cambridge University Press. 2017;76:443–54.
12.
Zurück zum Zitat Goldstein FC, Ashley AV, Endeshaw Y, Hanfelt J, Lah JJ, Levey AI. Effects of Hypertension and Hypercholesterolemia on Cognitive Functioning in Patients with Alzheimer’s Disease. Alzheimer Dis Assoc Disord. 2008;22:336–42.PubMedPubMedCentralCrossRef Goldstein FC, Ashley AV, Endeshaw Y, Hanfelt J, Lah JJ, Levey AI. Effects of Hypertension and Hypercholesterolemia on Cognitive Functioning in Patients with Alzheimer’s Disease. Alzheimer Dis Assoc Disord. 2008;22:336–42.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet. 2020;396:413–46.PubMedPubMedCentralCrossRef Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet. 2020;396:413–46.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Nutaitis AC, Tharwani SD, Serra MC, Goldstein FC, Zhao L, Sher SS, et al. Diet as a Risk Factor for Cognitive Decline in African Americans and Caucasians with a Parental History of Alzheimer’s Disease: A Cross-Sectional Pilot Study Dietary Patterns. J Prev Alzheimers Dis. 2019;6:50–5.PubMed Nutaitis AC, Tharwani SD, Serra MC, Goldstein FC, Zhao L, Sher SS, et al. Diet as a Risk Factor for Cognitive Decline in African Americans and Caucasians with a Parental History of Alzheimer’s Disease: A Cross-Sectional Pilot Study Dietary Patterns. J Prev Alzheimers Dis. 2019;6:50–5.PubMed
15.
Zurück zum Zitat Gareau MG. Microbiota-Gut-Brain Axis and Cognitive Function. In: Lyte M, Cryan JF, editors. Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Health and Disease. New York, NY: Springer; 2014. p. 357–71.CrossRef Gareau MG. Microbiota-Gut-Brain Axis and Cognitive Function. In: Lyte M, Cryan JF, editors. Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Health and Disease. New York, NY: Springer; 2014. p. 357–71.CrossRef
16.
Zurück zum Zitat Berg G, Rybakova D, Fischer D, Cernava T, Vergès M-CC, Charles T, et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8:103. Berg G, Rybakova D, Fischer D, Cernava T, Vergès M-CC, Charles T, et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8:103.
17.
Zurück zum Zitat Chelakkot C, Ghim J, Ryu SH. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med Nature Publishing Group. 2018;50:1–9.CrossRef Chelakkot C, Ghim J, Ryu SH. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med Nature Publishing Group. 2018;50:1–9.CrossRef
18.
Zurück zum Zitat Mayer EA, Knight R, Mazmanian SK, Cryan JF, Tillisch K. Gut Microbes and the Brain: Paradigm Shift in Neuroscience. J Neurosci. 2014;34:15490–6.PubMedPubMedCentralCrossRef Mayer EA, Knight R, Mazmanian SK, Cryan JF, Tillisch K. Gut Microbes and the Brain: Paradigm Shift in Neuroscience. J Neurosci. 2014;34:15490–6.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol. 2016;14:20–32.PubMedCrossRef Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol. 2016;14:20–32.PubMedCrossRef
21.
Zurück zum Zitat Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol Nature Publishing Group. 2016;14:20–32.CrossRef Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol Nature Publishing Group. 2016;14:20–32.CrossRef
22.
Zurück zum Zitat Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem T, Zeevi D, et al. Environment dominates over host genetics in shaping human gut microbiota. Nature Nature Publishing Group. 2018;555:210–5. Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem T, Zeevi D, et al. Environment dominates over host genetics in shaping human gut microbiota. Nature Nature Publishing Group. 2018;555:210–5.
23.
Zurück zum Zitat Ling Z, Zhu M, Yan X, Cheng Y, Shao L, Liu X, et al. Structural and Functional Dysbiosis of Fecal Microbiota in Chinese Patients With Alzheimer’s Disease. Frontiers in Cell and Developmental Biology. 2021;8: 634069.PubMedPubMedCentralCrossRef Ling Z, Zhu M, Yan X, Cheng Y, Shao L, Liu X, et al. Structural and Functional Dysbiosis of Fecal Microbiota in Chinese Patients With Alzheimer’s Disease. Frontiers in Cell and Developmental Biology. 2021;8: 634069.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Liu S, Gao J, Zhu M, Liu K, Zhang H-L. Gut Microbiota and Dysbiosis in Alzheimer’s Disease: Implications for Pathogenesis and Treatment. Mol Neurobiol. 2020;57:5026–43.PubMedPubMedCentralCrossRef Liu S, Gao J, Zhu M, Liu K, Zhang H-L. Gut Microbiota and Dysbiosis in Alzheimer’s Disease: Implications for Pathogenesis and Treatment. Mol Neurobiol. 2020;57:5026–43.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Li B, He Y, Ma J, Huang P, Du J, Cao L, et al. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimer’s & Dementia. 2019;15:1357–66.CrossRef Li B, He Y, Ma J, Huang P, Du J, Cao L, et al. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimer’s & Dementia. 2019;15:1357–66.CrossRef
26.
Zurück zum Zitat Liu P, Wu L, Peng G, Han Y, Tang R, Ge J, et al. Altered microbiomes distinguish Alzheimer’s disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain Behav Immun. 2019;80:633–43.PubMedCrossRef Liu P, Wu L, Peng G, Han Y, Tang R, Ge J, et al. Altered microbiomes distinguish Alzheimer’s disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain Behav Immun. 2019;80:633–43.PubMedCrossRef
27.
Zurück zum Zitat Zhang X, Wang Y, Liu W, Wang T, Wang L, Hao L, et al. Diet quality, gut microbiota, and microRNAs associated with mild cognitive impairment in middle-aged and elderly Chinese population. Am J Clin Nutr. 2021;114:429–40.PubMedCrossRef Zhang X, Wang Y, Liu W, Wang T, Wang L, Hao L, et al. Diet quality, gut microbiota, and microRNAs associated with mild cognitive impairment in middle-aged and elderly Chinese population. Am J Clin Nutr. 2021;114:429–40.PubMedCrossRef
28.
Zurück zum Zitat Nagpal R, Neth BJ, Wang S, Mishra SP, Craft S, Yadav H. Gut mycobiome and its interaction with diet, gut bacteria and alzheimer’s disease markers in subjects with mild cognitive impairment: A pilot study. EBioMedicine. 2020;59: 102950.PubMedPubMedCentralCrossRef Nagpal R, Neth BJ, Wang S, Mishra SP, Craft S, Yadav H. Gut mycobiome and its interaction with diet, gut bacteria and alzheimer’s disease markers in subjects with mild cognitive impairment: A pilot study. EBioMedicine. 2020;59: 102950.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Saji N, Murotani K, Hisada T, Tsuduki T, Sugimoto T, Kimura A, et al. The relationship between the gut microbiome and mild cognitive impairment in patients without dementia: a cross-sectional study conducted in Japan. Sci Rep. Nature Publishing Group. 2019;9:19227. Saji N, Murotani K, Hisada T, Tsuduki T, Sugimoto T, Kimura A, et al. The relationship between the gut microbiome and mild cognitive impairment in patients without dementia: a cross-sectional study conducted in Japan. Sci Rep. Nature Publishing Group. 2019;9:19227.
30.
Zurück zum Zitat Bostanciklioğlu M. The role of gut microbiota in pathogenesis of Alzheimer’s disease. J Appl Microbiol. 2019;127:954–67.PubMedCrossRef Bostanciklioğlu M. The role of gut microbiota in pathogenesis of Alzheimer’s disease. J Appl Microbiol. 2019;127:954–67.PubMedCrossRef
31.
Zurück zum Zitat Visconti A, Le Roy CI, Rosa F, Rossi N, Martin TC, Mohney RP, et al. Interplay between the human gut microbiome and host metabolism. Nat Commun. 2019;10:4505.PubMedPubMedCentralCrossRef Visconti A, Le Roy CI, Rosa F, Rossi N, Martin TC, Mohney RP, et al. Interplay between the human gut microbiome and host metabolism. Nat Commun. 2019;10:4505.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Lubinski R. Dementia and communication. Singular. 1995. Lubinski R. Dementia and communication. Singular. 1995.
34.
Zurück zum Zitat Mortamet B, Zeng D, Gerig G, Prastawa M, Bullitt E. Effects of Healthy Aging Measured By Intracranial Compartment Volumes Using a Designed MR Brain Database. Med Image Comput Comput Assist Interv. 2005;8:383–91.PubMedPubMedCentral Mortamet B, Zeng D, Gerig G, Prastawa M, Bullitt E. Effects of Healthy Aging Measured By Intracranial Compartment Volumes Using a Designed MR Brain Database. Med Image Comput Comput Assist Interv. 2005;8:383–91.PubMedPubMedCentral
35.
Zurück zum Zitat Scahill RI, Frost C, Jenkins R, Whitwell JL, Rossor MN, Fox NC. A longitudinal study of brain volume changes in normal aging using serial registered magnetic resonance imaging. Arch Neurol. 2003;60:989–94.PubMedCrossRef Scahill RI, Frost C, Jenkins R, Whitwell JL, Rossor MN, Fox NC. A longitudinal study of brain volume changes in normal aging using serial registered magnetic resonance imaging. Arch Neurol. 2003;60:989–94.PubMedCrossRef
36.
Zurück zum Zitat Deary IJ, Corley J, Gow AJ, Harris SE, Houlihan LM, Marioni RE, et al. Age-associated cognitive decline. Br Med Bull. 2009;92:135–52.PubMedCrossRef Deary IJ, Corley J, Gow AJ, Harris SE, Houlihan LM, Marioni RE, et al. Age-associated cognitive decline. Br Med Bull. 2009;92:135–52.PubMedCrossRef
37.
38.
Zurück zum Zitat Sullivan EV, Pfefferbaum A. Diffusion tensor imaging and aging. Neurosci Biobehav Rev. 2006;30:749–61.PubMedCrossRef Sullivan EV, Pfefferbaum A. Diffusion tensor imaging and aging. Neurosci Biobehav Rev. 2006;30:749–61.PubMedCrossRef
39.
Zurück zum Zitat Bolandzadeh N, Davis JC, Tam R, Handy TC, Liu-Ambrose T. The association between cognitive function and white matter lesion location in older adults: a systematic review. BMC Neurol. 2012;12:126.PubMedPubMedCentralCrossRef Bolandzadeh N, Davis JC, Tam R, Handy TC, Liu-Ambrose T. The association between cognitive function and white matter lesion location in older adults: a systematic review. BMC Neurol. 2012;12:126.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Alexander GE, Bergfield KL, Chen K, Reiman EM, Hanson KD, Lin L, et al. Gray matter network associated with risk for Alzheimer’s disease in young to middle-aged adults. Neurobiol Aging. 2012;33:2723–32.PubMedPubMedCentralCrossRef Alexander GE, Bergfield KL, Chen K, Reiman EM, Hanson KD, Lin L, et al. Gray matter network associated with risk for Alzheimer’s disease in young to middle-aged adults. Neurobiol Aging. 2012;33:2723–32.PubMedPubMedCentralCrossRef
41.
42.
Zurück zum Zitat Adluru N, Destiche DJ, Lu SY-F, Doran ST, Birdsill AC, Melah KE, et al. White matter microstructure in late middle-age: Effects of apolipoprotein E4 and parental family history of Alzheimer’s disease. Neuroimage Clin. 2014;4:730–42. Adluru N, Destiche DJ, Lu SY-F, Doran ST, Birdsill AC, Melah KE, et al. White matter microstructure in late middle-age: Effects of apolipoprotein E4 and parental family history of Alzheimer’s disease. Neuroimage Clin. 2014;4:730–42.
43.
Zurück zum Zitat Persson J, Lind J, Larsson A, Ingvar M, Cruts M, Van Broeckhoven C, et al. Altered brain white matter integrity in healthy carriers of the APOE epsilon4 allele: a risk for AD? Neurology. 2006;66:1029–33.PubMedCrossRef Persson J, Lind J, Larsson A, Ingvar M, Cruts M, Van Broeckhoven C, et al. Altered brain white matter integrity in healthy carriers of the APOE epsilon4 allele: a risk for AD? Neurology. 2006;66:1029–33.PubMedCrossRef
44.
Zurück zum Zitat Heise V, Filippini N, Ebmeier KP, Mackay CE. The APOE ɛ4 allele modulates brain white matter integrity in healthy adults. Mol Psychiatry. 2011;16:908–16.PubMedCrossRef Heise V, Filippini N, Ebmeier KP, Mackay CE. The APOE ɛ4 allele modulates brain white matter integrity in healthy adults. Mol Psychiatry. 2011;16:908–16.PubMedCrossRef
45.
Zurück zum Zitat Miller MI, Ratnanather JT, Tward DJ, Brown T, Lee DS, Ketcha M, et al. Network Neurodegeneration in Alzheimer’s Disease via MRI Based Shape Diffeomorphometry and High-Field Atlasing. Frontiers in Bioengineering and Biotechnology. 2015;3:54.PubMedPubMedCentralCrossRef Miller MI, Ratnanather JT, Tward DJ, Brown T, Lee DS, Ketcha M, et al. Network Neurodegeneration in Alzheimer’s Disease via MRI Based Shape Diffeomorphometry and High-Field Atlasing. Frontiers in Bioengineering and Biotechnology. 2015;3:54.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Thompson PM, Hayashi KM, De Zubicaray GI, Janke AL, Rose SE, Semple J, et al. Mapping hippocampal and ventricular change in Alzheimer disease. Neuroimage. 2004;22:1754–66.PubMedCrossRef Thompson PM, Hayashi KM, De Zubicaray GI, Janke AL, Rose SE, Semple J, et al. Mapping hippocampal and ventricular change in Alzheimer disease. Neuroimage. 2004;22:1754–66.PubMedCrossRef
47.
Zurück zum Zitat Jacobs HIL, Becker JA, Kwong K, Engels-Domínguez N, Prokopiou PC, Papp KV, et al. In vivo and neuropathology data support locus coeruleus integrity as indicator of Alzheimer’s disease pathology and cognitive decline. Sci Transl Med. 2021;13:612.CrossRef Jacobs HIL, Becker JA, Kwong K, Engels-Domínguez N, Prokopiou PC, Papp KV, et al. In vivo and neuropathology data support locus coeruleus integrity as indicator of Alzheimer’s disease pathology and cognitive decline. Sci Transl Med. 2021;13:612.CrossRef
48.
Zurück zum Zitat Jones DT, Machulda MM, Vemuri P, McDade EM, Zeng G, Senjem ML, et al. Age-related changes in the default mode network are more advanced in Alzheimer disease. Neurology. 2011;77:1524–31.PubMedPubMedCentralCrossRef Jones DT, Machulda MM, Vemuri P, McDade EM, Zeng G, Senjem ML, et al. Age-related changes in the default mode network are more advanced in Alzheimer disease. Neurology. 2011;77:1524–31.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Greicius MD, Srivastava G, Reiss AL, Menon V. Default-mode network activity distinguishes Alzheimer’s disease from healthy aging: Evidence from functional MRI. Proc Natl Acad Sci U S A. 2004;101:4637–42.PubMedPubMedCentralCrossRef Greicius MD, Srivastava G, Reiss AL, Menon V. Default-mode network activity distinguishes Alzheimer’s disease from healthy aging: Evidence from functional MRI. Proc Natl Acad Sci U S A. 2004;101:4637–42.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, et al. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr. 2018;57:1–24.PubMedCrossRef Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, et al. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr. 2018;57:1–24.PubMedCrossRef
51.
Zurück zum Zitat Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015;28:203–9.PubMedPubMedCentral Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015;28:203–9.PubMedPubMedCentral
52.
Zurück zum Zitat Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The Microbiota-Gut-Brain Axis. Physiol Rev. 2019;99:1877–2013.PubMedCrossRef Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The Microbiota-Gut-Brain Axis. Physiol Rev. 2019;99:1877–2013.PubMedCrossRef
54.
Zurück zum Zitat Gareau MG, Wine E, Rodrigues DM, Cho JH, Whary MT, Philpott DJ, et al. Bacterial infection causes stress-induced memory dysfunction in mice. Gut BMJ Publishing Group. 2011;60:307–17. Gareau MG, Wine E, Rodrigues DM, Cho JH, Whary MT, Philpott DJ, et al. Bacterial infection causes stress-induced memory dysfunction in mice. Gut BMJ Publishing Group. 2011;60:307–17.
55.
Zurück zum Zitat Heijtz RD, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, et al. Normal gut microbiota modulates brain development and behavior. PNAS National Academy of Sciences. 2011;108:3047–52.CrossRef Heijtz RD, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, et al. Normal gut microbiota modulates brain development and behavior. PNAS National Academy of Sciences. 2011;108:3047–52.CrossRef
56.
Zurück zum Zitat Luczynski P, McVey Neufeld K-A, Oriach CS, Clarke G, Dinan TG, Cryan JF. Growing up in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior. Int J Neuropsychopharmacol. 2016;19:8.CrossRef Luczynski P, McVey Neufeld K-A, Oriach CS, Clarke G, Dinan TG, Cryan JF. Growing up in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior. Int J Neuropsychopharmacol. 2016;19:8.CrossRef
57.
Zurück zum Zitat Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, et al. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. 2011;141(599–609):609.e1-3. Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, et al. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. 2011;141(599–609):609.e1-3.
58.
Zurück zum Zitat Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011;108:3047–52.PubMedCrossRef Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011;108:3047–52.PubMedCrossRef
59.
Zurück zum Zitat Baj G, Carlino D, Gardossi L, Tongiorgi E. Toward a unified biological hypothesis for the BDNF Val66Met-associated memory deficits in humans: a model of impaired dendritic mRNA trafficking. Front Neurosci. 2013;7:188.PubMedPubMedCentralCrossRef Baj G, Carlino D, Gardossi L, Tongiorgi E. Toward a unified biological hypothesis for the BDNF Val66Met-associated memory deficits in humans: a model of impaired dendritic mRNA trafficking. Front Neurosci. 2013;7:188.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Lu K, Abo RP, Schlieper KA, Graffam ME, Levine S, Wishnok JS, et al. Arsenic exposure perturbs the gut microbiome and its metabolic profile in mice: an integrated metagenomics and metabolomics analysis. Environ Health Perspect. 2014;122:284–91.PubMedPubMedCentralCrossRef Lu K, Abo RP, Schlieper KA, Graffam ME, Levine S, Wishnok JS, et al. Arsenic exposure perturbs the gut microbiome and its metabolic profile in mice: an integrated metagenomics and metabolomics analysis. Environ Health Perspect. 2014;122:284–91.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil. 2011;23:255–64.PubMedCrossRef Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil. 2011;23:255–64.PubMedCrossRef
62.
Zurück zum Zitat Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney RD, Shanahan F, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Molecular Psychiatry Nature Publishing Group. 2013;18:666–73.CrossRef Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney RD, Shanahan F, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Molecular Psychiatry Nature Publishing Group. 2013;18:666–73.CrossRef
63.
Zurück zum Zitat Lukić I, Getselter D, Koren O, Elliott E. Role of Tryptophan in Microbiota-Induced Depressive-Like Behavior: Evidence From Tryptophan Depletion Study. Front Behav Neurosci. 2019;13:123.PubMedPubMedCentralCrossRef Lukić I, Getselter D, Koren O, Elliott E. Role of Tryptophan in Microbiota-Induced Depressive-Like Behavior: Evidence From Tryptophan Depletion Study. Front Behav Neurosci. 2019;13:123.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Lyte JM, Gheorghe CE, Goodson MS, Kelley-Loughnane N, Dinan TG, Cryan JF, et al. Gut-brain axis serotonergic responses to acute stress exposure are microbiome-dependent. Neurogastroenterol Motil. 2020;32: e13881.PubMedCrossRef Lyte JM, Gheorghe CE, Goodson MS, Kelley-Loughnane N, Dinan TG, Cryan JF, et al. Gut-brain axis serotonergic responses to acute stress exposure are microbiome-dependent. Neurogastroenterol Motil. 2020;32: e13881.PubMedCrossRef
65.
Zurück zum Zitat Matsumoto M, Kibe R, Ooga T, Aiba Y, Sawaki E, Koga Y, et al. Cerebral low-molecular metabolites influenced by intestinal microbiota: a pilot study. Front Syst Neurosci. 2013;7:9.PubMedPubMedCentralCrossRef Matsumoto M, Kibe R, Ooga T, Aiba Y, Sawaki E, Koga Y, et al. Cerebral low-molecular metabolites influenced by intestinal microbiota: a pilot study. Front Syst Neurosci. 2013;7:9.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Desbonnet L, Clarke G, Traplin A, O’Sullivan O, Crispie F, Moloney RD, et al. Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour. Brain Behav Immun. 2015;48:165–73.PubMedCrossRef Desbonnet L, Clarke G, Traplin A, O’Sullivan O, Crispie F, Moloney RD, et al. Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour. Brain Behav Immun. 2015;48:165–73.PubMedCrossRef
67.
Zurück zum Zitat Fröhlich EE, Farzi A, Mayerhofer R, Reichmann F, Jačan A, Wagner B, et al. Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication. Brain Behav Immun. 2016;56:140–55.PubMedPubMedCentralCrossRef Fröhlich EE, Farzi A, Mayerhofer R, Reichmann F, Jačan A, Wagner B, et al. Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication. Brain Behav Immun. 2016;56:140–55.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Verdu EF, Bercik P, Huang XX, Lu J, Al-Mutawaly N, Sakai H, et al. The role of luminal factors in the recovery of gastric function and behavioral changes after chronic Helicobacter pylori infection. American Journal of Physiology-Gastrointestinal and Liver Physiology. American Physiological Society; 2008;295:4. Verdu EF, Bercik P, Huang XX, Lu J, Al-Mutawaly N, Sakai H, et al. The role of luminal factors in the recovery of gastric function and behavioral changes after chronic Helicobacter pylori infection. American Journal of Physiology-Gastrointestinal and Liver Physiology. American Physiological Society; 2008;295:4.
69.
Zurück zum Zitat Azpiroz F, Dubray C, Bernalier-Donadille A, Cardot J-M, Accarino A, Serra J, et al. Effects of scFOS on the composition of fecal microbiota and anxiety in patients with irritable bowel syndrome: a randomized, double blind, placebo controlled study. Neurogastroenterol Motil. 2017;29:e12911. Azpiroz F, Dubray C, Bernalier-Donadille A, Cardot J-M, Accarino A, Serra J, et al. Effects of scFOS on the composition of fecal microbiota and anxiety in patients with irritable bowel syndrome: a randomized, double blind, placebo controlled study. Neurogastroenterol Motil. 2017;29:e12911.
70.
Zurück zum Zitat 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;82:472–87.PubMedCrossRef 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;82:472–87.PubMedCrossRef
71.
Zurück zum Zitat Liu X, Cao S, Zhang X. Modulation of Gut Microbiota–Brain Axis by Probiotics, Prebiotics, and Diet. J Agric Food Chem. American Chemical Society; 2015;63:7885–95. Liu X, Cao S, Zhang X. Modulation of Gut Microbiota–Brain Axis by Probiotics, Prebiotics, and Diet. J Agric Food Chem. American Chemical Society; 2015;63:7885–95.
72.
Zurück zum Zitat Marx W, Scholey A, Firth J, D’Cunha NM, Lane M, Hockey M, et al. Prebiotics, probiotics, fermented foods and cognitive outcomes: A meta-analysis of randomized controlled trials. Neurosci Biobehav Rev. 2020;118:472–84.PubMedCrossRef Marx W, Scholey A, Firth J, D’Cunha NM, Lane M, Hockey M, et al. Prebiotics, probiotics, fermented foods and cognitive outcomes: A meta-analysis of randomized controlled trials. Neurosci Biobehav Rev. 2020;118:472–84.PubMedCrossRef
73.
Zurück zum Zitat Pinto-Sanchez MI, Hall GB, Ghajar K, Nardelli A, Bolino C, Lau JT, et al. Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain Activity: A Pilot Study in Patients With Irritable Bowel Syndrome. Gastroenterology Elsevier. 2017;153:448–59.CrossRef Pinto-Sanchez MI, Hall GB, Ghajar K, Nardelli A, Bolino C, Lau JT, et al. Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain Activity: A Pilot Study in Patients With Irritable Bowel Syndrome. Gastroenterology Elsevier. 2017;153:448–59.CrossRef
74.
Zurück zum Zitat Farhangi MA, Javid AZ, Sarmadi B, Karimi P, Dehghan P. A randomized controlled trial on the efficacy of resistant dextrin, as functional food, in women with type 2 diabetes: Targeting the hypothalamic-pituitary-adrenal axis and immune system. Clin Nutr. 2018;37:1216–23.PubMedCrossRef Farhangi MA, Javid AZ, Sarmadi B, Karimi P, Dehghan P. A randomized controlled trial on the efficacy of resistant dextrin, as functional food, in women with type 2 diabetes: Targeting the hypothalamic-pituitary-adrenal axis and immune system. Clin Nutr. 2018;37:1216–23.PubMedCrossRef
75.
Zurück zum Zitat Jia S, Lu Z, Gao Z, An J, Wu X, Li X, et al. Chitosan oligosaccharides alleviate cognitive deficits in an amyloid-β1-42-induced rat model of Alzheimer’s disease. Int J Biol Macromol. 2016;83:416–25.PubMedCrossRef Jia S, Lu Z, Gao Z, An J, Wu X, Li X, et al. Chitosan oligosaccharides alleviate cognitive deficits in an amyloid-β1-42-induced rat model of Alzheimer’s disease. Int J Biol Macromol. 2016;83:416–25.PubMedCrossRef
76.
Zurück zum Zitat Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao J, et al. Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol. 2016;16:90.PubMedPubMedCentralCrossRef Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao J, et al. Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol. 2016;16:90.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut microbiome alterations in Alzheimer’s disease. Scientific Reports Nature Publishing Group. 2017;7:13537.CrossRef Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut microbiome alterations in Alzheimer’s disease. Scientific Reports Nature Publishing Group. 2017;7:13537.CrossRef
79.
Zurück zum Zitat Sochocka M, Donskow-Łysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The Gut Microbiome Alterations and Inflammation-Driven Pathogenesis of Alzheimer’s Disease—a Critical Review. Mol Neurobiol. 2019;56:1841–51.PubMedCrossRef Sochocka M, Donskow-Łysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The Gut Microbiome Alterations and Inflammation-Driven Pathogenesis of Alzheimer’s Disease—a Critical Review. Mol Neurobiol. 2019;56:1841–51.PubMedCrossRef
80.
Zurück zum Zitat Zhuang Z-Q, Shen L-L, Li W-W, Fu X, Zeng F, Gui L, et al. Gut Microbiota is Altered in Patients with Alzheimer’s Disease. Journal of Alzheimer’s Disease IOS Press. 2018;63:1337–46.CrossRef Zhuang Z-Q, Shen L-L, Li W-W, Fu X, Zeng F, Gui L, et al. Gut Microbiota is Altered in Patients with Alzheimer’s Disease. Journal of Alzheimer’s Disease IOS Press. 2018;63:1337–46.CrossRef
81.
Zurück zum Zitat 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;49:60–8.PubMedCrossRef 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;49:60–8.PubMedCrossRef
82.
Zurück zum Zitat Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci V, et al. Alzheimer’s Disease Microbiome Is Associated with Dysregulation of the Anti-Inflammatory P-Glycoprotein Pathway. mBio. 2019;10:3. Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci V, et al. Alzheimer’s Disease Microbiome Is Associated with Dysregulation of the Anti-Inflammatory P-Glycoprotein Pathway. mBio. 2019;10:3.
83.
Zurück zum Zitat Verheggen ICM, de Jong JJA, van Boxtel MPJ, Gronenschild EHBM, Palm WM, Postma AA, et al. Increase in blood–brain barrier leakage in healthy, older adults. GeroScience. 2020;42:1183–93.PubMedPubMedCentralCrossRef Verheggen ICM, de Jong JJA, van Boxtel MPJ, Gronenschild EHBM, Palm WM, Postma AA, et al. Increase in blood–brain barrier leakage in healthy, older adults. GeroScience. 2020;42:1183–93.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Sweeney MD, Sagare AP, Zlokovic BV. Blood–brain barrier breakdown in Alzheimer’s disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14:133–50.PubMedPubMedCentralCrossRef Sweeney MD, Sagare AP, Zlokovic BV. Blood–brain barrier breakdown in Alzheimer’s disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14:133–50.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Wu M-L, Yang X-Q, Xue L, Duan W, Du J-R. Age-related cognitive decline is associated with microbiota-gut-brain axis disorders and neuroinflammation in mice. Behav Brain Res. 2021;402: 113125.PubMedCrossRef Wu M-L, Yang X-Q, Xue L, Duan W, Du J-R. Age-related cognitive decline is associated with microbiota-gut-brain axis disorders and neuroinflammation in mice. Behav Brain Res. 2021;402: 113125.PubMedCrossRef
86.
Zurück zum Zitat Ticinesi A, Tana C, Nouvenne A, Prati B, Lauretani F, Meschi T. Gut microbiota, cognitive frailty and dementia in older individuals: a systematic review. Clin Interv Aging. 2018;13:1497–511.PubMedPubMedCentralCrossRef Ticinesi A, Tana C, Nouvenne A, Prati B, Lauretani F, Meschi T. Gut microbiota, cognitive frailty and dementia in older individuals: a systematic review. Clin Interv Aging. 2018;13:1497–511.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Agostoni E, Chinnock JE, Daly MDB, Murray JG. Functional and histological studies of the vagus nerve and its branches to the heart, lungs and abdominal viscera in the cat. J Physiol. 1957;135:182–205.PubMedPubMedCentralCrossRef Agostoni E, Chinnock JE, Daly MDB, Murray JG. Functional and histological studies of the vagus nerve and its branches to the heart, lungs and abdominal viscera in the cat. J Physiol. 1957;135:182–205.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Abreu MT, Fukata M, Arditi M. TLR Signaling in the Gut in Health and Disease. The Journal of Immunology. American Association of Immunologists; 2005;174:4453–60. Abreu MT, Fukata M, Arditi M. TLR Signaling in the Gut in Health and Disease. The Journal of Immunology. American Association of Immunologists; 2005;174:4453–60.
89.
Zurück zum Zitat Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proceedings of the National Academy of Sciences. Proceedings of the National Academy of Sciences; 2008;105:16767–72. Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proceedings of the National Academy of Sciences. Proceedings of the National Academy of Sciences; 2008;105:16767–72.
90.
Zurück zum Zitat Li Y, Hao Y, Zhu J, Owyang C. Serotonin released from intestinal enterochromaffin cells mediates luminal non–cholecystokinin-stimulated pancreatic secretion in rats. Gastroenterology. 2000;118:1197–207.PubMedCrossRef Li Y, Hao Y, Zhu J, Owyang C. Serotonin released from intestinal enterochromaffin cells mediates luminal non–cholecystokinin-stimulated pancreatic secretion in rats. Gastroenterology. 2000;118:1197–207.PubMedCrossRef
91.
Zurück zum Zitat Strader AD, Woods SC. Gastrointestinal hormones and food intake. Gastroenterology. 2005;128:175–91.PubMedCrossRef Strader AD, Woods SC. Gastrointestinal hormones and food intake. Gastroenterology. 2005;128:175–91.PubMedCrossRef
92.
Zurück zum Zitat 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. Proceedings of the National Academy of Sciences. Proceedings of the National Academy of Sciences; 2011;108:16050–5. 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. Proceedings of the National Academy of Sciences. Proceedings of the National Academy of Sciences; 2011;108:16050–5.
93.
Zurück zum Zitat Bercik P, Park AJ, Sinclair D, Khoshdel A, Lu J, Huang X, et al. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut–brain communication. Neurogastroenterol Motil. 2011;23:1132–9.PubMedPubMedCentralCrossRef Bercik P, Park AJ, Sinclair D, Khoshdel A, Lu J, Huang X, et al. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut–brain communication. Neurogastroenterol Motil. 2011;23:1132–9.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, et al. The Intestinal Microbiota Affect Central Levels of Brain-Derived Neurotropic Factor and Behavior in Mice. Gastroenterology. 2011;141:599–609.PubMedCrossRef Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, et al. The Intestinal Microbiota Affect Central Levels of Brain-Derived Neurotropic Factor and Behavior in Mice. Gastroenterology. 2011;141:599–609.PubMedCrossRef
95.
Zurück zum Zitat Bajaj JS, Ridlon JM, Hylemon PB, Thacker LR, Heuman DM, Smith S, et al. Linkage of gut microbiome with cognition in hepatic encephalopathy. American Journal of Physiology-Gastrointestinal and Liver Physiology. American Physiological Society. 2012;302:168–75. Bajaj JS, Ridlon JM, Hylemon PB, Thacker LR, Heuman DM, Smith S, et al. Linkage of gut microbiome with cognition in hepatic encephalopathy. American Journal of Physiology-Gastrointestinal and Liver Physiology. American Physiological Society. 2012;302:168–75.
96.
Zurück zum Zitat Bajaj JS, Hylemon PB, Ridlon JM, Heuman DM, Daita K, White MB, et al. Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. American Journal of Physiology-Gastrointestinal and Liver Physiology. American Physiological Society. 2012;303:675–85. Bajaj JS, Hylemon PB, Ridlon JM, Heuman DM, Daita K, White MB, et al. Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. American Journal of Physiology-Gastrointestinal and Liver Physiology. American Physiological Society. 2012;303:675–85.
97.
Zurück zum Zitat Racanelli V, Rehermann B. The liver as an immunological organ. Hepatology. 2006;43:54–62.CrossRef Racanelli V, Rehermann B. The liver as an immunological organ. Hepatology. 2006;43:54–62.CrossRef
98.
Zurück zum Zitat Binder HJ, Filburn B, Floch M. Bile acid inhibition of intestinal anaerobic organisms. Am J Clin Nutr. 1975;28:119–25.PubMedCrossRef Binder HJ, Filburn B, Floch M. Bile acid inhibition of intestinal anaerobic organisms. Am J Clin Nutr. 1975;28:119–25.PubMedCrossRef
100.
Zurück zum Zitat Inagaki T, Moschetta A, Lee Y-K, Peng L, Zhao G, Downes M, et al. Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci U S A. 2006;103:3920–5.PubMedPubMedCentralCrossRef Inagaki T, Moschetta A, Lee Y-K, Peng L, Zhao G, Downes M, et al. Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci U S A. 2006;103:3920–5.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Bajaj JS, Heuman DM, Hylemon PB, Sanyal AJ, White MB, Monteith P, et al. Altered profile of human gut microbiome is associated with cirrhosis and its complications. J Hepatol. 2014;60:940–7.PubMedCrossRef Bajaj JS, Heuman DM, Hylemon PB, Sanyal AJ, White MB, Monteith P, et al. Altered profile of human gut microbiome is associated with cirrhosis and its complications. J Hepatol. 2014;60:940–7.PubMedCrossRef
103.
Zurück zum Zitat Pande C, Kumar A, Sarin SK. Small-intestinal bacterial overgrowth in cirrhosis is related to the severity of liver disease. Aliment Pharmacol Ther. 2009;29:1273–81.PubMedCrossRef Pande C, Kumar A, Sarin SK. Small-intestinal bacterial overgrowth in cirrhosis is related to the severity of liver disease. Aliment Pharmacol Ther. 2009;29:1273–81.PubMedCrossRef
104.
Zurück zum Zitat Kuang L, Zhou W, Jiang Y. Association of small intestinal bacterial overgrowth with nonalcoholic fatty liver disease in children: A meta-analysis. PLOS ONE. Public Library of Science. 2021;16:e0260479. Kuang L, Zhou W, Jiang Y. Association of small intestinal bacterial overgrowth with nonalcoholic fatty liver disease in children: A meta-analysis. PLOS ONE. Public Library of Science. 2021;16:e0260479.
105.
Zurück zum Zitat Ding J-H, Jin Z, Yang X-X, Lou J, Shan W-X, Hu Y-X, et al. Role of gut microbiota via the gut-liver-brain axis in digestive diseases. World J Gastroenterol. 2020;26:6141–62.PubMedPubMedCentralCrossRef Ding J-H, Jin Z, Yang X-X, Lou J, Shan W-X, Hu Y-X, et al. Role of gut microbiota via the gut-liver-brain axis in digestive diseases. World J Gastroenterol. 2020;26:6141–62.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Šarenac TM, Mikov M. Bile Acid Synthesis: From Nature to the Chemical Modification and Synthesis and Their Applications as Drugs and Nutrients. Front Pharmacol. 2018;9:939.PubMedPubMedCentralCrossRef Šarenac TM, Mikov M. Bile Acid Synthesis: From Nature to the Chemical Modification and Synthesis and Their Applications as Drugs and Nutrients. Front Pharmacol. 2018;9:939.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Spichak S, Bastiaanssen TFS, Berding K, Vlckova K, Clarke G, Dinan TG, et al. Mining microbes for mental health: Determining the role of microbial metabolic pathways in human brain health and disease. Neurosci Biobehav Rev. 2021;125:698–761.PubMedCrossRef Spichak S, Bastiaanssen TFS, Berding K, Vlckova K, Clarke G, Dinan TG, et al. Mining microbes for mental health: Determining the role of microbial metabolic pathways in human brain health and disease. Neurosci Biobehav Rev. 2021;125:698–761.PubMedCrossRef
110.
Zurück zum Zitat Baloni P, Funk CC, Yan J, Yurkovich JT, Kueider-Paisley A, Nho K, et al. Metabolic Network Analysis Reveals Altered Bile Acid Synthesis and Metabolism in Alzheimer’s Disease. Cell Rep Med. 2020;1: 100138.PubMedPubMedCentralCrossRef Baloni P, Funk CC, Yan J, Yurkovich JT, Kueider-Paisley A, Nho K, et al. Metabolic Network Analysis Reveals Altered Bile Acid Synthesis and Metabolism in Alzheimer’s Disease. Cell Rep Med. 2020;1: 100138.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Kotti TJ, Ramirez DMO, Pfeiffer BE, Huber KM, Russell DW. Brain cholesterol turnover required for geranylgeraniol production and learning in mice. Proc Natl Acad Sci U S A. 2006;103:3869–74.PubMedPubMedCentralCrossRef Kotti TJ, Ramirez DMO, Pfeiffer BE, Huber KM, Russell DW. Brain cholesterol turnover required for geranylgeraniol production and learning in mice. Proc Natl Acad Sci U S A. 2006;103:3869–74.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Higashi T, Watanabe S, Tomaru K, Yamazaki W, Yoshizawa K, Ogawa S, et al. Unconjugated bile acids in rat brain: Analytical method based on LC/ESI-MS/MS with chemical derivatization and estimation of their origin by comparison to serum levels. Steroids. 2017;125:107–13.PubMedCrossRef Higashi T, Watanabe S, Tomaru K, Yamazaki W, Yoshizawa K, Ogawa S, et al. Unconjugated bile acids in rat brain: Analytical method based on LC/ESI-MS/MS with chemical derivatization and estimation of their origin by comparison to serum levels. Steroids. 2017;125:107–13.PubMedCrossRef
114.
Zurück zum Zitat Mano N, Goto T, Uchida M, Nishimura K, Ando M, Kobayashi N, et al. Presence of protein-bound unconjugated bile acids in the cytoplasmic fraction of rat brain. J Lipid Res. 2004;45:295–300.PubMedCrossRef Mano N, Goto T, Uchida M, Nishimura K, Ando M, Kobayashi N, et al. Presence of protein-bound unconjugated bile acids in the cytoplasmic fraction of rat brain. J Lipid Res. 2004;45:295–300.PubMedCrossRef
115.
Zurück zum Zitat Pan X, Elliott CT, McGuinness B, Passmore P, Kehoe PG, Hölscher C, et al. Metabolomic Profiling of Bile Acids in Clinical and Experimental Samples of Alzheimer’s Disease. Metabolites. 2017;7:28. Pan X, Elliott CT, McGuinness B, Passmore P, Kehoe PG, Hölscher C, et al. Metabolomic Profiling of Bile Acids in Clinical and Experimental Samples of Alzheimer’s Disease. Metabolites. 2017;7:28.
116.
Zurück zum Zitat Quinn M, McMillin M, Galindo C, Frampton G, Pae HY, DeMorrow S. Bile acids permeabilize the blood brain barrier after bile duct ligation in rats via Rac1-dependent mechanisms. Dig Liver Dis. 2014;46:527–34.PubMedPubMedCentralCrossRef Quinn M, McMillin M, Galindo C, Frampton G, Pae HY, DeMorrow S. Bile acids permeabilize the blood brain barrier after bile duct ligation in rats via Rac1-dependent mechanisms. Dig Liver Dis. 2014;46:527–34.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Kamp F, Hamilton JA, Kamp F, Westerhoff HV, Hamilton JA. Movement of fatty acids, fatty acid analogues, and bile acids across phospholipid bilayers. Biochemistry. 1993;32:11074–86.PubMedCrossRef Kamp F, Hamilton JA, Kamp F, Westerhoff HV, Hamilton JA. Movement of fatty acids, fatty acid analogues, and bile acids across phospholipid bilayers. Biochemistry. 1993;32:11074–86.PubMedCrossRef
118.
Zurück zum Zitat Benedetti A, Di Sario A, Marucci L, Svegliati-Baroni G, Schteingart CD, Ton-Nu HT, et al. Carrier-mediated transport of conjugated bile acids across the basolateral membrane of biliary epithelial cells. Am J Physiol. 1997;272:1416–24. Benedetti A, Di Sario A, Marucci L, Svegliati-Baroni G, Schteingart CD, Ton-Nu HT, et al. Carrier-mediated transport of conjugated bile acids across the basolateral membrane of biliary epithelial cells. Am J Physiol. 1997;272:1416–24.
119.
Zurück zum Zitat Bron B, Waldram R, Silk DB, Williams R. Serum, cerebrospinal fluid, and brain levels of bile acids in patients with fulminant hepatic failure. Gut. 1977;18:692–6.PubMedPubMedCentralCrossRef Bron B, Waldram R, Silk DB, Williams R. Serum, cerebrospinal fluid, and brain levels of bile acids in patients with fulminant hepatic failure. Gut. 1977;18:692–6.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Cheng X, Maher J, Chen C, Klaassen CD. Tissue Distribution and Ontogeny of Mouse Organic Anion Transporting Polypeptides (oatps). Drug Metab Dispos. American Society for Pharmacology and Experimental Therapeutics. 2005;33:1062–73. Cheng X, Maher J, Chen C, Klaassen CD. Tissue Distribution and Ontogeny of Mouse Organic Anion Transporting Polypeptides (oatps). Drug Metab Dispos. American Society for Pharmacology and Experimental Therapeutics. 2005;33:1062–73.
121.
Zurück zum Zitat Nizamutdinov D, DeMorrow S, McMillin M, Kain J, Mukherjee S, Zeitouni S, et al. Hepatic alterations are accompanied by changes to bile acid transporter-expressing neurons in the hypothalamus after traumatic brain injury. Scientific Reports. Nature Publishing Group. 2017;7:40112. Nizamutdinov D, DeMorrow S, McMillin M, Kain J, Mukherjee S, Zeitouni S, et al. Hepatic alterations are accompanied by changes to bile acid transporter-expressing neurons in the hypothalamus after traumatic brain injury. Scientific Reports. Nature Publishing Group. 2017;7:40112.
122.
Zurück zum Zitat Soontornmalai A, Vlaming MLH, Fritschy J-M. Differential, strain-specific cellular and subcellular distribution of multidrug transporters in murine choroid plexus and blood–brain barrier. Neuroscience. 2006;138:159–69.PubMedCrossRef Soontornmalai A, Vlaming MLH, Fritschy J-M. Differential, strain-specific cellular and subcellular distribution of multidrug transporters in murine choroid plexus and blood–brain barrier. Neuroscience. 2006;138:159–69.PubMedCrossRef
123.
Zurück zum Zitat Roberts LM, Black DS, Raman C, Woodford K, Zhou M, Haggerty JE, et al. Subcellular localization of transporters along the rat blood–brain barrier and blood–cerebral-spinal fluid barrier by in vivo biotinylation. Neuroscience. 2008;155:423–38.PubMedCrossRef Roberts LM, Black DS, Raman C, Woodford K, Zhou M, Haggerty JE, et al. Subcellular localization of transporters along the rat blood–brain barrier and blood–cerebral-spinal fluid barrier by in vivo biotinylation. Neuroscience. 2008;155:423–38.PubMedCrossRef
124.
Zurück zum Zitat Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Begley DJ. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37:13–25.PubMedCrossRef Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Begley DJ. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37:13–25.PubMedCrossRef
125.
Zurück zum Zitat Mertens KL, Kalsbeek A, Soeters MR, Eggink HM. Bile Acid Signaling Pathways from the Enterohepatic Circulation to the Central Nervous System. Front Neurosci. 2017;11:617. Mertens KL, Kalsbeek A, Soeters MR, Eggink HM. Bile Acid Signaling Pathways from the Enterohepatic Circulation to the Central Nervous System. Front Neurosci. 2017;11:617.
126.
Zurück zum Zitat MahmoudianDehkordi S, Arnold M, Nho K, Ahmad S, Jia W, Xie G, et al. Altered bile acid profile associates with cognitive impairment in Alzheimer’s disease-An emerging role for gut microbiome. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association. 2019;15:76–92.CrossRef MahmoudianDehkordi S, Arnold M, Nho K, Ahmad S, Jia W, Xie G, et al. Altered bile acid profile associates with cognitive impairment in Alzheimer’s disease-An emerging role for gut microbiome. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association. 2019;15:76–92.CrossRef
127.
Zurück zum Zitat Olazarán J, Gil-de-Gómez L, Rodríguez-Martín A, Valentí-Soler M, Frades-Payo B, Marín-Muñoz J, et al. A blood-based, 7-metabolite signature for the early diagnosis of Alzheimer’s disease. J Alzheimers Dis. 2015;45:1157–73.PubMedCrossRef Olazarán J, Gil-de-Gómez L, Rodríguez-Martín A, Valentí-Soler M, Frades-Payo B, Marín-Muñoz J, et al. A blood-based, 7-metabolite signature for the early diagnosis of Alzheimer’s disease. J Alzheimers Dis. 2015;45:1157–73.PubMedCrossRef
128.
Zurück zum Zitat Ignacio Barrasa J, Olmo N, Pérez-Ramos P, Santiago-Gómez A, Lecona E, Turnay J, et al. Deoxycholic and chenodeoxycholic bile acids induce apoptosis via oxidative stress in human colon adenocarcinoma cells. Apoptosis. 2011;16:1054–67.PubMedCrossRef Ignacio Barrasa J, Olmo N, Pérez-Ramos P, Santiago-Gómez A, Lecona E, Turnay J, et al. Deoxycholic and chenodeoxycholic bile acids induce apoptosis via oxidative stress in human colon adenocarcinoma cells. Apoptosis. 2011;16:1054–67.PubMedCrossRef
129.
Zurück zum Zitat Tran CD, Grice DM, Wade B, Kerr CA, Bauer DC, Li D, et al. Gut permeability, its interaction with gut microflora and effects on metabolic health are mediated by the lymphatics system, liver and bile acid. Future Microbiol. 2015;10:1339–53.PubMedCrossRef Tran CD, Grice DM, Wade B, Kerr CA, Bauer DC, Li D, et al. Gut permeability, its interaction with gut microflora and effects on metabolic health are mediated by the lymphatics system, liver and bile acid. Future Microbiol. 2015;10:1339–53.PubMedCrossRef
130.
Zurück zum Zitat Jena PK, Sheng L, Di Lucente J, Jin L-W, Maezawa I, Wan Y-JY. Dysregulated bile acid synthesis and dysbiosis are implicated in Western diet-induced systemic inflammation, microglial activation, and reduced neuroplasticity. FASEB J. 2018;32:2866–77. Jena PK, Sheng L, Di Lucente J, Jin L-W, Maezawa I, Wan Y-JY. Dysregulated bile acid synthesis and dysbiosis are implicated in Western diet-induced systemic inflammation, microglial activation, and reduced neuroplasticity. FASEB J. 2018;32:2866–77.
131.
Zurück zum Zitat Bernstein H, Bernstein C, Payne CM, Dvorak K. Bile acids as endogenous etiologic agents in gastrointestinal cancer. World J Gastroenterol. 2009;15:3329–40.PubMedPubMedCentralCrossRef Bernstein H, Bernstein C, Payne CM, Dvorak K. Bile acids as endogenous etiologic agents in gastrointestinal cancer. World J Gastroenterol. 2009;15:3329–40.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Dröge W, Schipper HM. Oxidative stress and aberrant signaling in aging and cognitive decline. Aging Cell. 2007;6:361–70.PubMedCrossRef Dröge W, Schipper HM. Oxidative stress and aberrant signaling in aging and cognitive decline. Aging Cell. 2007;6:361–70.PubMedCrossRef
133.
Zurück zum Zitat Moreira PI, Carvalho C, Zhu X, Smith MA, Perry G. Mitochondrial dysfunction is a trigger of Alzheimer’s disease pathophysiology. Biochim Biophys Acta. 2010;1802:2–10.PubMedCrossRef Moreira PI, Carvalho C, Zhu X, Smith MA, Perry G. Mitochondrial dysfunction is a trigger of Alzheimer’s disease pathophysiology. Biochim Biophys Acta. 2010;1802:2–10.PubMedCrossRef
134.
Zurück zum Zitat Leloup C, Magnan C, Benani A, Bonnet E, Alquier T, Offer G, et al. Mitochondrial reactive oxygen species are required for hypothalamic glucose sensing. Diabetes. 2006;55:2084–90.PubMedCrossRef Leloup C, Magnan C, Benani A, Bonnet E, Alquier T, Offer G, et al. Mitochondrial reactive oxygen species are required for hypothalamic glucose sensing. Diabetes. 2006;55:2084–90.PubMedCrossRef
135.
Zurück zum Zitat Theofilopoulos S, Wang Y, Kitambi SS, Sacchetti P, Sousa KM, Bodin K, et al. Brain endogenous liver X receptor ligands selectively promote midbrain neurogenesis. Nat Chem Biol Nature Publishing Group. 2013;9:126–33.CrossRef Theofilopoulos S, Wang Y, Kitambi SS, Sacchetti P, Sousa KM, Bodin K, et al. Brain endogenous liver X receptor ligands selectively promote midbrain neurogenesis. Nat Chem Biol Nature Publishing Group. 2013;9:126–33.CrossRef
136.
Zurück zum Zitat Ramalho RM, Ribeiro PS, Solá S, Castro RE, Steer CJ, Rodrigues CMP. Inhibition of the E2F–1/p53/Bax pathway by tauroursodeoxycholic acid in amyloid beta-peptide-induced apoptosis of PC12 cells. J Neurochem. 2004;90:567–75.PubMedCrossRef Ramalho RM, Ribeiro PS, Solá S, Castro RE, Steer CJ, Rodrigues CMP. Inhibition of the E2F–1/p53/Bax pathway by tauroursodeoxycholic acid in amyloid beta-peptide-induced apoptosis of PC12 cells. J Neurochem. 2004;90:567–75.PubMedCrossRef
137.
Zurück zum Zitat Rodrigues CMP, Spellman SR, Solá S, Grande AW, Linehan-Stieers C, Low WC, et al. Neuroprotection by a Bile Acid in an Acute Stroke Model in the Rat. J Cereb Blood Flow Metab. SAGE Publications Ltd STM. 2002;22:463–71. Rodrigues CMP, Spellman SR, Solá S, Grande AW, Linehan-Stieers C, Low WC, et al. Neuroprotection by a Bile Acid in an Acute Stroke Model in the Rat. J Cereb Blood Flow Metab. SAGE Publications Ltd STM. 2002;22:463–71.
138.
Zurück zum Zitat Solá S, Amaral JD, Borralho PM, Ramalho RM, Castro RE, Aranha MM, et al. Functional modulation of nuclear steroid receptors by tauroursodeoxycholic acid reduces amyloid beta-peptide-induced apoptosis. Mol Endocrinol. 2006;20:2292–303.PubMedCrossRef Solá S, Amaral JD, Borralho PM, Ramalho RM, Castro RE, Aranha MM, et al. Functional modulation of nuclear steroid receptors by tauroursodeoxycholic acid reduces amyloid beta-peptide-induced apoptosis. Mol Endocrinol. 2006;20:2292–303.PubMedCrossRef
139.
Zurück zum Zitat Nunes AF, Amaral JD, Lo AC, Fonseca MB, Viana RJS, Callaerts-Vegh Z, et al. TUDCA, a bile acid, attenuates amyloid precursor protein processing and amyloid-β deposition in APP/PS1 mice. Mol Neurobiol. 2012;45:440–54.PubMedCrossRef Nunes AF, Amaral JD, Lo AC, Fonseca MB, Viana RJS, Callaerts-Vegh Z, et al. TUDCA, a bile acid, attenuates amyloid precursor protein processing and amyloid-β deposition in APP/PS1 mice. Mol Neurobiol. 2012;45:440–54.PubMedCrossRef
140.
Zurück zum Zitat Dionísio PA, Amaral JD, Ribeiro MF, Lo AC, D’Hooge R, Rodrigues CMP. Amyloid-β pathology is attenuated by tauroursodeoxycholic acid treatment in APP/PS1 mice after disease onset. Neurobiol Aging. 2015;36:228–40.PubMedCrossRef Dionísio PA, Amaral JD, Ribeiro MF, Lo AC, D’Hooge R, Rodrigues CMP. Amyloid-β pathology is attenuated by tauroursodeoxycholic acid treatment in APP/PS1 mice after disease onset. Neurobiol Aging. 2015;36:228–40.PubMedCrossRef
141.
Zurück zum Zitat Yanguas-Casás N, Barreda-Manso MA, Nieto-Sampedro M, Romero-Ramírez L. TUDCA: An Agonist of the Bile Acid Receptor GPBAR1/TGR5 With Anti-Inflammatory Effects in Microglial Cells. J Cell Physiol. 2017;232:2231–45.PubMedCrossRef Yanguas-Casás N, Barreda-Manso MA, Nieto-Sampedro M, Romero-Ramírez L. TUDCA: An Agonist of the Bile Acid Receptor GPBAR1/TGR5 With Anti-Inflammatory Effects in Microglial Cells. J Cell Physiol. 2017;232:2231–45.PubMedCrossRef
142.
Zurück zum Zitat Soares R, Ribeiro FF, Xapelli S, Genebra T, Ribeiro MF, Sebastião AM, et al. Tauroursodeoxycholic Acid Enhances Mitochondrial Biogenesis, Neural Stem Cell Pool, and Early Neurogenesis in Adult Rats. Mol Neurobiol. 2018;55:3725–38.PubMed Soares R, Ribeiro FF, Xapelli S, Genebra T, Ribeiro MF, Sebastião AM, et al. Tauroursodeoxycholic Acid Enhances Mitochondrial Biogenesis, Neural Stem Cell Pool, and Early Neurogenesis in Adult Rats. Mol Neurobiol. 2018;55:3725–38.PubMed
143.
Zurück zum Zitat Cosacak MI, Bhattarai P, Kizil C. Alzheimer’s disease, neural stem cells and neurogenesis: cellular phase at single-cell level. Neural Regen Res. 2019;15:824–7.PubMedCentral Cosacak MI, Bhattarai P, Kizil C. Alzheimer’s disease, neural stem cells and neurogenesis: cellular phase at single-cell level. Neural Regen Res. 2019;15:824–7.PubMedCentral
144.
Zurück zum Zitat Craciun S, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc Natl Acad Sci. 2012;109:21307–12.PubMedPubMedCentralCrossRef Craciun S, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc Natl Acad Sci. 2012;109:21307–12.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Stremmel W, Schmidt KV, Schuhmann V, Kratzer F, Garbade SF, Langhans C-D, et al. Blood Trimethylamine-N-Oxide Originates from Microbiota Mediated Breakdown of Phosphatidylcholine and Absorption from Small Intestine. PLoS One. 2017;12:e0170742. Stremmel W, Schmidt KV, Schuhmann V, Kratzer F, Garbade SF, Langhans C-D, et al. Blood Trimethylamine-N-Oxide Originates from Microbiota Mediated Breakdown of Phosphatidylcholine and Absorption from Small Intestine. PLoS One. 2017;12:e0170742.
146.
Zurück zum Zitat Wang S-Z, Yu Y-J, Adeli K. Role of Gut Microbiota in Neuroendocrine Regulation of Carbohydrate and Lipid Metabolism via the Microbiota-Gut-Brain-Liver Axis. Microorganisms. 2020;8:527.PubMedCentralCrossRef Wang S-Z, Yu Y-J, Adeli K. Role of Gut Microbiota in Neuroendocrine Regulation of Carbohydrate and Lipid Metabolism via the Microbiota-Gut-Brain-Liver Axis. Microorganisms. 2020;8:527.PubMedCentralCrossRef
147.
Zurück zum Zitat Vogt NM, Romano KA, Darst BF, Engelman CD, Johnson SC, Carlsson CM, et al. The gut microbiota-derived metabolite trimethylamine N-oxide is elevated in Alzheimer’s disease. Alzheimer’s Research & Therapy. 2018;10:124.CrossRef Vogt NM, Romano KA, Darst BF, Engelman CD, Johnson SC, Carlsson CM, et al. The gut microbiota-derived metabolite trimethylamine N-oxide is elevated in Alzheimer’s disease. Alzheimer’s Research & Therapy. 2018;10:124.CrossRef
148.
Zurück zum Zitat Ufnal M, Zadlo A, Ostaszewski R. TMAO: A small molecule of great expectations. Nutrition. 2015;31:1317–23.PubMedCrossRef Ufnal M, Zadlo A, Ostaszewski R. TMAO: A small molecule of great expectations. Nutrition. 2015;31:1317–23.PubMedCrossRef
149.
Zurück zum Zitat Org E, Blum Y, Kasela S, Mehrabian M, Kuusisto J, Kangas AJ, et al. Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the METSIM cohort. Genome Biol. 2017;18:70.PubMedPubMedCentralCrossRef Org E, Blum Y, Kasela S, Mehrabian M, Kuusisto J, Kangas AJ, et al. Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the METSIM cohort. Genome Biol. 2017;18:70.PubMedPubMedCentralCrossRef
150.
Zurück zum Zitat Del Rio D, Zimetti F, Caffarra P, Tassotti M, Bernini F, Brighenti F, et al. The Gut Microbial Metabolite Trimethylamine-N-Oxide Is Present in Human Cerebrospinal Fluid. Nutrients. 2017;9:1053.PubMedCentralCrossRef Del Rio D, Zimetti F, Caffarra P, Tassotti M, Bernini F, Brighenti F, et al. The Gut Microbial Metabolite Trimethylamine-N-Oxide Is Present in Human Cerebrospinal Fluid. Nutrients. 2017;9:1053.PubMedCentralCrossRef
151.
Zurück zum Zitat Vernetti L, Gough A, Baetz N, Blutt S, Broughman JR, Brown JA, et al. Functional Coupling of Human Microphysiology Systems: Intestine, Liver, Kidney Proximal Tubule, Blood-Brain Barrier and Skeletal Muscle. Scientific Reports. Nature Publishing Group. 2017;7:42296. Vernetti L, Gough A, Baetz N, Blutt S, Broughman JR, Brown JA, et al. Functional Coupling of Human Microphysiology Systems: Intestine, Liver, Kidney Proximal Tubule, Blood-Brain Barrier and Skeletal Muscle. Scientific Reports. Nature Publishing Group. 2017;7:42296.
152.
Zurück zum Zitat Nho K, Kueider-Paisley A, MahmoudianDehkordi S, Arnold M, Risacher SL, Louie G, et al. Altered Bile Acid Profile in Mild Cognitive Impairment and Alzheimer’s Disease: Relationship to Neuroimaging and CSF Biomarkers. Alzheimers Dement. 2019;15:232–44.PubMedCrossRef Nho K, Kueider-Paisley A, MahmoudianDehkordi S, Arnold M, Risacher SL, Louie G, et al. Altered Bile Acid Profile in Mild Cognitive Impairment and Alzheimer’s Disease: Relationship to Neuroimaging and CSF Biomarkers. Alzheimers Dement. 2019;15:232–44.PubMedCrossRef
153.
Zurück zum Zitat Mapstone M, Cheema AK, Fiandaca MS, Zhong X, Mhyre TR, MacArthur LH, et al. Plasma phospholipids identify antecedent memory impairment in older adults. Nature Medicine Nature Publishing Group. 2014;20:415–8. Mapstone M, Cheema AK, Fiandaca MS, Zhong X, Mhyre TR, MacArthur LH, et al. Plasma phospholipids identify antecedent memory impairment in older adults. Nature Medicine Nature Publishing Group. 2014;20:415–8.
154.
Zurück zum Zitat Marksteiner J, Blasko I, Kemmler G, Koal T, Humpel C. Bile acid quantification of 20 plasma metabolites identifies lithocholic acid as a putative biomarker in Alzheimer’s disease. Metabolomics. 2017;14:1.PubMedPubMedCentralCrossRef Marksteiner J, Blasko I, Kemmler G, Koal T, Humpel C. Bile acid quantification of 20 plasma metabolites identifies lithocholic acid as a putative biomarker in Alzheimer’s disease. Metabolomics. 2017;14:1.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Mitchell SC, Zhang AQ, Smith RL. Chemical and Biological Liberation of Trimethylamine from Foods. J Food Compos Anal. 2002;15:277–82.CrossRef Mitchell SC, Zhang AQ, Smith RL. Chemical and Biological Liberation of Trimethylamine from Foods. J Food Compos Anal. 2002;15:277–82.CrossRef
156.
Zurück zum Zitat Zhang J, Cashman JR. Quantitative analysis of FMO gene mRNA levels in human tissues. Drug Metab Dispos. 2006;34:19–26.PubMedCrossRef Zhang J, Cashman JR. Quantitative analysis of FMO gene mRNA levels in human tissues. Drug Metab Dispos. 2006;34:19–26.PubMedCrossRef
157.
Zurück zum Zitat Tang WHW, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM, et al. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol. 2014;64:1908–14.PubMedCrossRef Tang WHW, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM, et al. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol. 2014;64:1908–14.PubMedCrossRef
158.
Zurück zum Zitat Gao X, Liu X, Xu J, Xue C, Xue Y, Wang Y. Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet. J Biosci Bioeng. 2014;118:476–81.PubMedCrossRef Gao X, Liu X, Xu J, Xue C, Xue Y, Wang Y. Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet. J Biosci Bioeng. 2014;118:476–81.PubMedCrossRef
159.
Zurück zum Zitat Missailidis C, Hällqvist J, Qureshi AR, Barany P, Heimbürger O, Lindholm B, et al. Serum Trimethylamine-N-Oxide Is Strongly Related to Renal Function and Predicts Outcome in Chronic Kidney Disease. PLOS ONE. Public Library of Science. 2016;11:e0141738. Missailidis C, Hällqvist J, Qureshi AR, Barany P, Heimbürger O, Lindholm B, et al. Serum Trimethylamine-N-Oxide Is Strongly Related to Renal Function and Predicts Outcome in Chronic Kidney Disease. PLOS ONE. Public Library of Science. 2016;11:e0141738.
160.
Zurück zum Zitat Barrea L, Annunziata G, Muscogiuri G, Di Somma C, Laudisio D, Maisto M, et al. Trimethylamine-N-oxide (TMAO) as Novel Potential Biomarker of Early Predictors of Metabolic Syndrome. Nutrients. Multidisciplinary Digital Publishing Institute. 2018;10:1971. Barrea L, Annunziata G, Muscogiuri G, Di Somma C, Laudisio D, Maisto M, et al. Trimethylamine-N-oxide (TMAO) as Novel Potential Biomarker of Early Predictors of Metabolic Syndrome. Nutrients. Multidisciplinary Digital Publishing Institute. 2018;10:1971.
161.
Zurück zum Zitat Li D, Ke Y, Zhan R, Liu C, Zhao M, Zeng A, et al. Trimethylamine‐N‐oxide promotes brain aging and cognitive impairment in mice. Aging Cell. 2018;17:e12768. Li D, Ke Y, Zhan R, Liu C, Zhao M, Zeng A, et al. Trimethylamine‐N‐oxide promotes brain aging and cognitive impairment in mice. Aging Cell. 2018;17:e12768.
162.
Zurück zum Zitat Gao Q, Wang Y, Wang X, Zhang X, Wang R-T. Decreased levels of circulating trimethylamine N-oxide alleviate cognitive and pathological deterioration in transgenic mice: a potential therapeutic approach for Alzheimer’s disease. Aging. 2019;11:8642–63.PubMedPubMedCentralCrossRef Gao Q, Wang Y, Wang X, Zhang X, Wang R-T. Decreased levels of circulating trimethylamine N-oxide alleviate cognitive and pathological deterioration in transgenic mice: a potential therapeutic approach for Alzheimer’s disease. Aging. 2019;11:8642–63.PubMedPubMedCentralCrossRef
163.
Zurück zum Zitat Govindarajulu M, Pinky PD, Steinke I, Bloemer J, Ramesh S, Kariharan T, et al. Gut Metabolite TMAO Induces Synaptic Plasticity Deficits by Promoting Endoplasmic Reticulum Stress. Front Mol Neurosci Frontiers. 2020;13:138.CrossRef Govindarajulu M, Pinky PD, Steinke I, Bloemer J, Ramesh S, Kariharan T, et al. Gut Metabolite TMAO Induces Synaptic Plasticity Deficits by Promoting Endoplasmic Reticulum Stress. Front Mol Neurosci Frontiers. 2020;13:138.CrossRef
164.
Zurück zum Zitat Brunt VE, LaRocca TJ, Bazzoni AE, Sapinsley ZJ, Miyamoto-Ditmon J, Gioscia-Ryan RA, et al. The gut microbiome–derived metabolite trimethylamine N-oxide modulates neuroinflammation and cognitive function with aging. GeroScience. 2020;43:377–94.PubMedPubMedCentralCrossRef Brunt VE, LaRocca TJ, Bazzoni AE, Sapinsley ZJ, Miyamoto-Ditmon J, Gioscia-Ryan RA, et al. The gut microbiome–derived metabolite trimethylamine N-oxide modulates neuroinflammation and cognitive function with aging. GeroScience. 2020;43:377–94.PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat He W, Luo Y, Liu J-P, Sun N, Guo D, Cui L-L, et al. Trimethylamine N-Oxide, a Gut Microbiota-Dependent Metabolite, is Associated with Frailty in Older Adults with Cardiovascular Disease. Clin Interv Aging. 2020;15:1809–20.PubMedPubMedCentralCrossRef He W, Luo Y, Liu J-P, Sun N, Guo D, Cui L-L, et al. Trimethylamine N-Oxide, a Gut Microbiota-Dependent Metabolite, is Associated with Frailty in Older Adults with Cardiovascular Disease. Clin Interv Aging. 2020;15:1809–20.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Zhu C, Li G, Lv Z, Li J, Wang X, Kang J, et al. Association of plasma trimethylamine-N-oxide levels with post-stroke cognitive impairment: a 1-year longitudinal study. Neurol Sci. 2020;41:57–63.PubMedCrossRef Zhu C, Li G, Lv Z, Li J, Wang X, Kang J, et al. Association of plasma trimethylamine-N-oxide levels with post-stroke cognitive impairment: a 1-year longitudinal study. Neurol Sci. 2020;41:57–63.PubMedCrossRef
167.
Zurück zum Zitat Zhuang Z, Gao M, Yang R, Liu Z, Cao W, Huang T. Causal relationships between gut metabolites and Alzheimer’s disease: a bidirectional Mendelian randomization study. Neurobiol Aging. 2020;100:119.PubMed Zhuang Z, Gao M, Yang R, Liu Z, Cao W, Huang T. Causal relationships between gut metabolites and Alzheimer’s disease: a bidirectional Mendelian randomization study. Neurobiol Aging. 2020;100:119.PubMed
168.
Zurück zum Zitat Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, et al. Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk. Cell. 2016;165:111–24.PubMedPubMedCentralCrossRef Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, et al. Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk. Cell. 2016;165:111–24.PubMedPubMedCentralCrossRef
169.
Zurück zum Zitat Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–85.PubMedPubMedCentralCrossRef Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–85.PubMedPubMedCentralCrossRef
170.
Zurück zum Zitat Chen M-L, Zhu X-H, Ran L, Lang H-D, Yi L, Mi M-T. Trimethylamine-N-Oxide Induces Vascular Inflammation by Activating the NLRP3 Inflammasome Through the SIRT3-SOD2-mtROS Signaling Pathway. J Am Heart Assoc. 2017;6:e006347. Chen M-L, Zhu X-H, Ran L, Lang H-D, Yi L, Mi M-T. Trimethylamine-N-Oxide Induces Vascular Inflammation by Activating the NLRP3 Inflammasome Through the SIRT3-SOD2-mtROS Signaling Pathway. J Am Heart Assoc. 2017;6:e006347.
171.
Zurück zum Zitat Seldin MM, Meng Y, Qi H, Zhu W, Wang Z, Hazen SL, et al. Trimethylamine N-Oxide Promotes Vascular Inflammation Through Signaling of Mitogen-Activated Protein Kinase and Nuclear Factor-κB. J Am Heart Assoc. 2016;5:e002767. Seldin MM, Meng Y, Qi H, Zhu W, Wang Z, Hazen SL, et al. Trimethylamine N-Oxide Promotes Vascular Inflammation Through Signaling of Mitogen-Activated Protein Kinase and Nuclear Factor-κB. J Am Heart Assoc. 2016;5:e002767.
172.
Zurück zum Zitat Farso M, Ménard C, Colby-Milley J, Quirion R. The immune marker CD68 correlates with cognitive impairment in normally aged rats. Neurobiol Aging. 2013;34:1971–6.PubMedCrossRef Farso M, Ménard C, Colby-Milley J, Quirion R. The immune marker CD68 correlates with cognitive impairment in normally aged rats. Neurobiol Aging. 2013;34:1971–6.PubMedCrossRef
173.
174.
Zurück zum Zitat Simen AA, Bordner KA, Martin MP, Moy LA, Barry LC. Cognitive dysfunction with aging and the role of inflammation. Ther Adv Chronic Dis. 2011;2:175–95.PubMedPubMedCentralCrossRef Simen AA, Bordner KA, Martin MP, Moy LA, Barry LC. Cognitive dysfunction with aging and the role of inflammation. Ther Adv Chronic Dis. 2011;2:175–95.PubMedPubMedCentralCrossRef
175.
Zurück zum Zitat Zhao L, Zhang C, Cao G, Dong X, Li D, Jiang L. Higher Circulating Trimethylamine N-oxide Sensitizes Sevoflurane-Induced Cognitive Dysfunction in Aged Rats Probably by Downregulating Hippocampal Methionine Sulfoxide Reductase A. Neurochem Res. 2019;44:2506–16.PubMedCrossRef Zhao L, Zhang C, Cao G, Dong X, Li D, Jiang L. Higher Circulating Trimethylamine N-oxide Sensitizes Sevoflurane-Induced Cognitive Dysfunction in Aged Rats Probably by Downregulating Hippocampal Methionine Sulfoxide Reductase A. Neurochem Res. 2019;44:2506–16.PubMedCrossRef
176.
Zurück zum Zitat Wang Q-J, Shen Y-E, Wang X, Fu S, Zhang X, Zhang Y-N, et al. Concomitant memantine and Lactobacillus plantarum treatment attenuates cognitive impairments in APP/PS1 mice. Aging (Albany NY). 2020;12:628–49.CrossRef Wang Q-J, Shen Y-E, Wang X, Fu S, Zhang X, Zhang Y-N, et al. Concomitant memantine and Lactobacillus plantarum treatment attenuates cognitive impairments in APP/PS1 mice. Aging (Albany NY). 2020;12:628–49.CrossRef
177.
Zurück zum Zitat Hoyles L, Pontifex MG, Rodriguez-Ramiro I, Anis-Alavi MA, Jelane KS, Snelling T, et al. Regulation of blood-brain barrier integrity by microbiome-associated methylamines and cognition by trimethylamine N-oxide. Microbiome. 2021;9:235.PubMedPubMedCentralCrossRef Hoyles L, Pontifex MG, Rodriguez-Ramiro I, Anis-Alavi MA, Jelane KS, Snelling T, et al. Regulation of blood-brain barrier integrity by microbiome-associated methylamines and cognition by trimethylamine N-oxide. Microbiome. 2021;9:235.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat McArthur S, Carvalho A, Fonseca S, Snelling T, Nicholson J, Glen R, et al. Effects of gut-derived trimethylamines on the blood-brain barrier. 2018. McArthur S, Carvalho A, Fonseca S, Snelling T, Nicholson J, Glen R, et al. Effects of gut-derived trimethylamines on the blood-brain barrier. 2018.
179.
Zurück zum Zitat Tseng H-C, Graves DJ. Natural Methylamine Osmolytes, Trimethylamine N-Oxide and Betaine, Increase Tau-Induced Polymerization of Microtubules. Biochem Biophys Res Commun. 1998;250:726–30.PubMedCrossRef Tseng H-C, Graves DJ. Natural Methylamine Osmolytes, Trimethylamine N-Oxide and Betaine, Increase Tau-Induced Polymerization of Microtubules. Biochem Biophys Res Commun. 1998;250:726–30.PubMedCrossRef
180.
Zurück zum Zitat Smith M, Crowther RA, Goedert M. The natural osmolyte trimethylamine N-oxide (TMAO) restores the ability of mutant tau to promote microtubule assembly. FEBS Lett. 2000;484:265–70.PubMedCrossRef Smith M, Crowther RA, Goedert M. The natural osmolyte trimethylamine N-oxide (TMAO) restores the ability of mutant tau to promote microtubule assembly. FEBS Lett. 2000;484:265–70.PubMedCrossRef
181.
182.
Zurück zum Zitat Bose S, Cho J. Targeting chaperones, heat shock factor-1, and unfolded protein response: Promising therapeutic approaches for neurodegenerative disorders. Ageing Res Rev. 2017;35:155–75.PubMedCrossRef Bose S, Cho J. Targeting chaperones, heat shock factor-1, and unfolded protein response: Promising therapeutic approaches for neurodegenerative disorders. Ageing Res Rev. 2017;35:155–75.PubMedCrossRef
183.
Zurück zum Zitat Velasquez MT, Ramezani A, Manal A, Raj DS. Trimethylamine N-Oxide: The Good, the Bad and the Unknown. Toxins (Basel). 2016;8:326.PubMedCentralCrossRef Velasquez MT, Ramezani A, Manal A, Raj DS. Trimethylamine N-Oxide: The Good, the Bad and the Unknown. Toxins (Basel). 2016;8:326.PubMedCentralCrossRef
184.
Zurück zum Zitat Wang Z, Levison BS, Hazen JE, Donahue L, Li X-M, Hazen SL. Measurement of trimethylamine-N-oxide by stable isotope dilution liquid chromatography tandem mass spectrometry. Anal Biochem. 2014;455:35–40.PubMedPubMedCentralCrossRef Wang Z, Levison BS, Hazen JE, Donahue L, Li X-M, Hazen SL. Measurement of trimethylamine-N-oxide by stable isotope dilution liquid chromatography tandem mass spectrometry. Anal Biochem. 2014;455:35–40.PubMedPubMedCentralCrossRef
185.
Zurück zum Zitat Janeiro MH, Ramírez MJ, Milagro FI, Martínez JA, Solas M. Implication of Trimethylamine N-Oxide (TMAO) in Disease: Potential Biomarker or New Therapeutic Target. Nutrients. 2018;10:10.CrossRef Janeiro MH, Ramírez MJ, Milagro FI, Martínez JA, Solas M. Implication of Trimethylamine N-Oxide (TMAO) in Disease: Potential Biomarker or New Therapeutic Target. Nutrients. 2018;10:10.CrossRef
186.
Zurück zum Zitat Costabile G, Vetrani C, Bozzetto L, Giacco R, Bresciani L, Del Rio D, et al. Plasma TMAO increase after healthy diets: results from 2 randomized controlled trials with dietary fish, polyphenols, and whole-grain cereals. Am J Clin Nutr. 2021;114:1342–50.PubMedCrossRef Costabile G, Vetrani C, Bozzetto L, Giacco R, Bresciani L, Del Rio D, et al. Plasma TMAO increase after healthy diets: results from 2 randomized controlled trials with dietary fish, polyphenols, and whole-grain cereals. Am J Clin Nutr. 2021;114:1342–50.PubMedCrossRef
187.
Zurück zum Zitat DiNicolantonio JJ, McCarty M, OKeefe J. Association of moderately elevated trimethylamine N-oxide with cardiovascular risk: is TMAO serving as a marker for hepatic insulin resistance. Open Heart. Archives of Disease in childhood; 2019;6:e000890. DiNicolantonio JJ, McCarty M, OKeefe J. Association of moderately elevated trimethylamine N-oxide with cardiovascular risk: is TMAO serving as a marker for hepatic insulin resistance. Open Heart. Archives of Disease in childhood; 2019;6:e000890.
188.
Zurück zum Zitat Li S, Shao Y, Li K, HuangFu C, Wang W, Liu Z, et al. Vascular Cognitive Impairment and the Gut Microbiota. J Alzheimers Dis. 2018;63:1209–22.PubMedCrossRef Li S, Shao Y, Li K, HuangFu C, Wang W, Liu Z, et al. Vascular Cognitive Impairment and the Gut Microbiota. J Alzheimers Dis. 2018;63:1209–22.PubMedCrossRef
190.
Zurück zum Zitat Agus A, Planchais J, Sokol H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe. 2018;23:716–24.PubMedCrossRef Agus A, Planchais J, Sokol H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe. 2018;23:716–24.PubMedCrossRef
191.
Zurück zum Zitat Clarke G, Villalobos-Manriquez F, Marin DC. Tryptophan Metabolism and the Microbiome-Gut-Brain Axis. In: Burnet PW, editors. The Oxford Handbook of the Microbiome-Gut-Brain Axis. 2020. Clarke G, Villalobos-Manriquez F, Marin DC. Tryptophan Metabolism and the Microbiome-Gut-Brain Axis. In: Burnet PW, editors. The Oxford Handbook of the Microbiome-Gut-Brain Axis. 2020.
192.
Zurück zum Zitat Richard DM, Dawes MA, Mathias CW, Acheson A, Hill-Kapturczak N, Dougherty DM. L-Tryptophan: Basic Metabolic Functions, Behavioral Research and Therapeutic Indications. Int J Tryptophan Res. 2009;2:45–60.PubMedPubMedCentralCrossRef Richard DM, Dawes MA, Mathias CW, Acheson A, Hill-Kapturczak N, Dougherty DM. L-Tryptophan: Basic Metabolic Functions, Behavioral Research and Therapeutic Indications. Int J Tryptophan Res. 2009;2:45–60.PubMedPubMedCentralCrossRef
193.
Zurück zum Zitat Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, et al. Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism. Front Cell Infect Microbiol. 2018;8:13.PubMedPubMedCentralCrossRef Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, et al. Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism. Front Cell Infect Microbiol. 2018;8:13.PubMedPubMedCentralCrossRef
194.
Zurück zum Zitat Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. PNAS. 2009;106:3698–703.PubMedPubMedCentralCrossRef Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. PNAS. 2009;106:3698–703.PubMedPubMedCentralCrossRef
196.
Zurück zum Zitat Jenkins TA, Nguyen JCD, Polglaze KE, Bertrand PP. Influence of Tryptophan and Serotonin on Mood and Cognition with a Possible Role of the Gut-Brain Axis. Nutrients. 2016;8:56. Jenkins TA, Nguyen JCD, Polglaze KE, Bertrand PP. Influence of Tryptophan and Serotonin on Mood and Cognition with a Possible Role of the Gut-Brain Axis. Nutrients. 2016;8:56.
197.
198.
Zurück zum Zitat Stone TW, Darlington LG. The kynurenine pathway as a therapeutic target in cognitive and neurodegenerative disorders. Br J Pharmacol. 2013;169:1211–27.PubMedPubMedCentralCrossRef Stone TW, Darlington LG. The kynurenine pathway as a therapeutic target in cognitive and neurodegenerative disorders. Br J Pharmacol. 2013;169:1211–27.PubMedPubMedCentralCrossRef
199.
Zurück zum Zitat Dehhaghi M, Kazemi Shariat Panahi H, Guillemin GJ. Microorganisms, Tryptophan Metabolism, and Kynurenine Pathway: A Complex Interconnected Loop Influencing Human Health Status. Int J Tryptophan Res. 2019;12:1178646919852996. Dehhaghi M, Kazemi Shariat Panahi H, Guillemin GJ. Microorganisms, Tryptophan Metabolism, and Kynurenine Pathway: A Complex Interconnected Loop Influencing Human Health Status. Int J Tryptophan Res. 2019;12:1178646919852996.
200.
Zurück zum Zitat Agus A, Clément K, Sokol H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut BMJ Publishing Group. 2021;70:1174–82. Agus A, Clément K, Sokol H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut BMJ Publishing Group. 2021;70:1174–82.
201.
Zurück zum Zitat Badawy AA-B. Kynurenine Pathway of Tryptophan Metabolism: Regulatory and Functional Aspects. Int J Tryptophan Res. 2017;10:1178646917691938. Badawy AA-B. Kynurenine Pathway of Tryptophan Metabolism: Regulatory and Functional Aspects. Int J Tryptophan Res. 2017;10:1178646917691938.
202.
Zurück zum Zitat Schwarcz R, Bruno JP, Muchowski PJ, Wu H-Q. Kynurenines in the mammalian brain: when physiology meets pathology. Nat Rev Neurosci. 2012;13:465–77.PubMedPubMedCentralCrossRef Schwarcz R, Bruno JP, Muchowski PJ, Wu H-Q. Kynurenines in the mammalian brain: when physiology meets pathology. Nat Rev Neurosci. 2012;13:465–77.PubMedPubMedCentralCrossRef
203.
Zurück zum Zitat Lugo-Huitrón R, Ugalde Muñiz P, Pineda B, Pedraza-Chaverrí J, Ríos C, Pérez-de la Cruz V. Quinolinic Acid: An Endogenous Neurotoxin with Multiple Targets. Oxid Med Cell Longev. 2013;2013:104024. Lugo-Huitrón R, Ugalde Muñiz P, Pineda B, Pedraza-Chaverrí J, Ríos C, Pérez-de la Cruz V. Quinolinic Acid: An Endogenous Neurotoxin with Multiple Targets. Oxid Med Cell Longev. 2013;2013:104024.
204.
Zurück zum Zitat Ting KK, Brew BJ, Guillemin GJ. Effect of quinolinic acid on human astrocytes morphology and functions: implications in Alzheimer’s disease. J Neuroinflammation. 2009;6:36.PubMedPubMedCentralCrossRef Ting KK, Brew BJ, Guillemin GJ. Effect of quinolinic acid on human astrocytes morphology and functions: implications in Alzheimer’s disease. J Neuroinflammation. 2009;6:36.PubMedPubMedCentralCrossRef
205.
Zurück zum Zitat Fujigaki H, Yamamoto Y, Saito K. L-Tryptophan-kynurenine pathway enzymes are therapeutic target for neuropsychiatric diseases: Focus on cell type differences. Neuropharmacology. 2017;112:264–74.PubMedCrossRef Fujigaki H, Yamamoto Y, Saito K. L-Tryptophan-kynurenine pathway enzymes are therapeutic target for neuropsychiatric diseases: Focus on cell type differences. Neuropharmacology. 2017;112:264–74.PubMedCrossRef
206.
Zurück zum Zitat Hwu P, Du MX, Lapointe R, Do M, Taylor MW, Young HA. Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J Immunol. 2000;164:3596–9.PubMedCrossRef Hwu P, Du MX, Lapointe R, Do M, Taylor MW, Young HA. Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J Immunol. 2000;164:3596–9.PubMedCrossRef
207.
Zurück zum Zitat Gulaj E, Pawlak K, Bien B, Pawlak D. Kynurenine and its metabolites in Alzheimer’s disease patients. Adv Med Sci. 2010;55:204–11.PubMedCrossRef Gulaj E, Pawlak K, Bien B, Pawlak D. Kynurenine and its metabolites in Alzheimer’s disease patients. Adv Med Sci. 2010;55:204–11.PubMedCrossRef
208.
Zurück zum Zitat Yu D, Tao B-B, Yang Y-Y, Du L-S, Yang S-S, He X-J, et al. The IDO Inhibitor Coptisine Ameliorates Cognitive Impairment in a Mouse Model of Alzheimer’s Disease. Journal of Alzheimer’s Disease IOS Press. 2015;43:291–302.CrossRef Yu D, Tao B-B, Yang Y-Y, Du L-S, Yang S-S, He X-J, et al. The IDO Inhibitor Coptisine Ameliorates Cognitive Impairment in a Mouse Model of Alzheimer’s Disease. Journal of Alzheimer’s Disease IOS Press. 2015;43:291–302.CrossRef
209.
Zurück zum Zitat Ramos-Chávez LA, Roldán-Roldán G, García-Juárez B, González-Esquivel D, Pérez de la Cruz G, Pineda B, et al. Low Serum Tryptophan Levels as an Indicator of Global Cognitive Performance in Nondemented Women over 50 Years of Age. Oxid Med Cell Longev. 2018;2018:2018:8604718. Ramos-Chávez LA, Roldán-Roldán G, García-Juárez B, González-Esquivel D, Pérez de la Cruz G, Pineda B, et al. Low Serum Tryptophan Levels as an Indicator of Global Cognitive Performance in Nondemented Women over 50 Years of Age. Oxid Med Cell Longev. 2018;2018:2018:8604718.
210.
Zurück zum Zitat Smith JA, Das A, Ray SK, Banik NL. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull. 2012;87:10–20.PubMedCrossRef Smith JA, Das A, Ray SK, Banik NL. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull. 2012;87:10–20.PubMedCrossRef
211.
Zurück zum Zitat Yamada A, Akimoto H, Kagawa S, Guillemin GJ, Takikawa O. Proinflammatory cytokine interferon-γ increases induction of indoleamine 2,3-dioxygenase in monocytic cells primed with amyloid β peptide 1–42: implications for the pathogenesis of Alzheimer’s disease. J Neurochem. 2009;110:791–800.PubMedCrossRef Yamada A, Akimoto H, Kagawa S, Guillemin GJ, Takikawa O. Proinflammatory cytokine interferon-γ increases induction of indoleamine 2,3-dioxygenase in monocytic cells primed with amyloid β peptide 1–42: implications for the pathogenesis of Alzheimer’s disease. J Neurochem. 2009;110:791–800.PubMedCrossRef
212.
Zurück zum Zitat Liebau C, Baltzer AWA, Schmidt S, Roesel C, Karreman C, Prisack JB, et al. Interleukin-12 and interleukin-18 induce indoleamine 2,3-dioxygenase (IDO) activity in human osteosarcoma cell lines independently from interferon-gamma. Anticancer Res. 2002;22:931–6.PubMed Liebau C, Baltzer AWA, Schmidt S, Roesel C, Karreman C, Prisack JB, et al. Interleukin-12 and interleukin-18 induce indoleamine 2,3-dioxygenase (IDO) activity in human osteosarcoma cell lines independently from interferon-gamma. Anticancer Res. 2002;22:931–6.PubMed
213.
Zurück zum Zitat Solvang S-EH, Nordrehaug JE, Tell GS, Nygård O, McCann A, Ueland PM, et al. The kynurenine pathway and cognitive performance in community-dwelling older adults. The Hordaland Health Study. Brain Behav Immun. 2019;75:155–62. Solvang S-EH, Nordrehaug JE, Tell GS, Nygård O, McCann A, Ueland PM, et al. The kynurenine pathway and cognitive performance in community-dwelling older adults. The Hordaland Health Study. Brain Behav Immun. 2019;75:155–62.
214.
Zurück zum Zitat Mithaiwala MN, Santana-Coelho D, Porter GA, O’Connor JC. Neuroinflammation and the Kynurenine Pathway in CNS Disease: Molecular Mechanisms and Therapeutic Implications. Cells. 2021;10:1548.PubMedPubMedCentralCrossRef Mithaiwala MN, Santana-Coelho D, Porter GA, O’Connor JC. Neuroinflammation and the Kynurenine Pathway in CNS Disease: Molecular Mechanisms and Therapeutic Implications. Cells. 2021;10:1548.PubMedPubMedCentralCrossRef
215.
Zurück zum Zitat Guillemin GJ, Brew BJ, Noonan CE, Takikawa O, Cullen KM. Indoleamine 2,3 dioxygenase and quinolinic acid immunoreactivity in Alzheimer’s disease hippocampus. Neuropathol Appl Neurobiol. 2005;31:395–404.PubMedCrossRef Guillemin GJ, Brew BJ, Noonan CE, Takikawa O, Cullen KM. Indoleamine 2,3 dioxygenase and quinolinic acid immunoreactivity in Alzheimer’s disease hippocampus. Neuropathol Appl Neurobiol. 2005;31:395–404.PubMedCrossRef
216.
Zurück zum Zitat Heyes MP, Morrison PF. Quantification of local de novo synthesis versus blood contributions to quinolinic acid concentrations in brain and systemic tissues. J Neurochem. 1997;68:280–8.PubMedCrossRef Heyes MP, Morrison PF. Quantification of local de novo synthesis versus blood contributions to quinolinic acid concentrations in brain and systemic tissues. J Neurochem. 1997;68:280–8.PubMedCrossRef
217.
Zurück zum Zitat Rahman A, Ting K, Cullen KM, Braidy N, Brew BJ, Guillemin GJ. The Excitotoxin Quinolinic Acid Induces Tau Phosphorylation in Human Neurons. PLOS ONE. Public Library of Science; 2009;4:e6344. Rahman A, Ting K, Cullen KM, Braidy N, Brew BJ, Guillemin GJ. The Excitotoxin Quinolinic Acid Induces Tau Phosphorylation in Human Neurons. PLOS ONE. Public Library of Science; 2009;4:e6344.
218.
Zurück zum Zitat Zwilling D, Huang S-Y, Sathyasaikumar KV, Notarangelo FM, Guidetti P, Wu H-Q, et al. Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell. 2011;145:863–74.PubMedPubMedCentralCrossRef Zwilling D, Huang S-Y, Sathyasaikumar KV, Notarangelo FM, Guidetti P, Wu H-Q, et al. Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell. 2011;145:863–74.PubMedPubMedCentralCrossRef
219.
Zurück zum Zitat Crockett MJ, Clark L, Roiser JP, Robinson OJ, Cools R, Chase HW, et al. Converging evidence for central 5-HT effects in acute tryptophan depletion. Mol Psychiatry. 2012;17:121–3.PubMedCrossRef Crockett MJ, Clark L, Roiser JP, Robinson OJ, Cools R, Chase HW, et al. Converging evidence for central 5-HT effects in acute tryptophan depletion. Mol Psychiatry. 2012;17:121–3.PubMedCrossRef
220.
Zurück zum Zitat Hughes MM, Carballedo A, McLoughlin DM, Amico F, Harkin A, Frodl T, et al. Tryptophan depletion in depressed patients occurs independent of kynurenine pathway activation. Brain Behav Immun. 2012;26:979–87.PubMedCrossRef Hughes MM, Carballedo A, McLoughlin DM, Amico F, Harkin A, Frodl T, et al. Tryptophan depletion in depressed patients occurs independent of kynurenine pathway activation. Brain Behav Immun. 2012;26:979–87.PubMedCrossRef
221.
Zurück zum Zitat van Donkelaar EL, Blokland A, Ferrington L, Kelly P a. T, Steinbusch HWM, Prickaerts J. Mechanism of acute tryptophan depletion: is it only serotonin? Mol Psychiatry. 2011;16:695–713. van Donkelaar EL, Blokland A, Ferrington L, Kelly P a. T, Steinbusch HWM, Prickaerts J. Mechanism of acute tryptophan depletion: is it only serotonin? Mol Psychiatry. 2011;16:695–713.
222.
Zurück zum Zitat El-Merahbi R, Löffler M, Mayer A, Sumara G. The roles of peripheral serotonin in metabolic homeostasis. FEBS Lett. 2015;589:1728–34.PubMedCrossRef El-Merahbi R, Löffler M, Mayer A, Sumara G. The roles of peripheral serotonin in metabolic homeostasis. FEBS Lett. 2015;589:1728–34.PubMedCrossRef
223.
Zurück zum Zitat Höglund E, Øverli Ø, Winberg S. Tryptophan Metabolic Pathways and Brain Serotonergic Activity: A Comparative Review. Front Endocrinol. 2019;10:158.CrossRef Höglund E, Øverli Ø, Winberg S. Tryptophan Metabolic Pathways and Brain Serotonergic Activity: A Comparative Review. Front Endocrinol. 2019;10:158.CrossRef
224.
Zurück zum Zitat Lovelace MD, Varney B, Sundaram G, Lennon MJ, Lim CK, Jacobs K, et al. Recent evidence for an expanded role of the kynurenine pathway of tryptophan metabolism in neurological diseases. Neuropharmacology. 2017;112:373–88.PubMedCrossRef Lovelace MD, Varney B, Sundaram G, Lennon MJ, Lim CK, Jacobs K, et al. Recent evidence for an expanded role of the kynurenine pathway of tryptophan metabolism in neurological diseases. Neuropharmacology. 2017;112:373–88.PubMedCrossRef
225.
226.
Zurück zum Zitat Cowen P, Sherwood A. The role of serotonin in cognitive function: Evidence from recent studies and implications for understanding depression. Journal of psychopharmacology (Oxford, England). 2013;27:575–83.CrossRef Cowen P, Sherwood A. The role of serotonin in cognitive function: Evidence from recent studies and implications for understanding depression. Journal of psychopharmacology (Oxford, England). 2013;27:575–83.CrossRef
227.
Zurück zum Zitat Schmitt JAJ, Wingen M, Ramaekers JG, Evers EAT, Riedel WJ. Serotonin and Human Cognitive Performance. Curr Pharm Des. 2006;12:2473–86.PubMedCrossRef Schmitt JAJ, Wingen M, Ramaekers JG, Evers EAT, Riedel WJ. Serotonin and Human Cognitive Performance. Curr Pharm Des. 2006;12:2473–86.PubMedCrossRef
229.
Zurück zum Zitat Porter RJ, Lunn BS, Walker LLM, Gray JM, Ballard CG, O’Brien JT. Cognitive Deficit Induced by Acute Tryptophan Depletion in Patients With Alzheimer’s Disease. AJP American Psychiatric Publishing. 2000;157:638–40.CrossRef Porter RJ, Lunn BS, Walker LLM, Gray JM, Ballard CG, O’Brien JT. Cognitive Deficit Induced by Acute Tryptophan Depletion in Patients With Alzheimer’s Disease. AJP American Psychiatric Publishing. 2000;157:638–40.CrossRef
230.
Zurück zum Zitat Haider S, Khaliq S, Haleem DJ. Enhanced serotonergic neurotransmission in the hippocampus following tryptophan administration improves learning acquisition and memory consolidation in rats. Pharmacol Rep. 2007;59:53–7.PubMed Haider S, Khaliq S, Haleem DJ. Enhanced serotonergic neurotransmission in the hippocampus following tryptophan administration improves learning acquisition and memory consolidation in rats. Pharmacol Rep. 2007;59:53–7.PubMed
231.
Zurück zum Zitat Levkovitz Y, Richter-Levin G, Segal M. Effect of 5-hydroxytryptophane on behavior and hippocampal physiology in young and old rats. Neurobiol Aging. 1994;15:635–41.PubMedCrossRef Levkovitz Y, Richter-Levin G, Segal M. Effect of 5-hydroxytryptophane on behavior and hippocampal physiology in young and old rats. Neurobiol Aging. 1994;15:635–41.PubMedCrossRef
232.
Zurück zum Zitat Kaddurah-Daouk R, Zhu H, Sharma S, Bogdanov M, Rozen SG, Matson W, et al. Alterations in metabolic pathways and networks in Alzheimer’s disease. Transl Psychiatry. 2013;3: e244.PubMedPubMedCentralCrossRef Kaddurah-Daouk R, Zhu H, Sharma S, Bogdanov M, Rozen SG, Matson W, et al. Alterations in metabolic pathways and networks in Alzheimer’s disease. Transl Psychiatry. 2013;3: e244.PubMedPubMedCentralCrossRef
233.
Zurück zum Zitat Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39:372–85.PubMedCrossRef Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39:372–85.PubMedCrossRef
234.
Zurück zum Zitat Hubbard TD, Murray IA, Bisson WH, Lahoti TS, Gowda K, Amin SG, et al. Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles. Sci Rep. 2015;5:12689.PubMedPubMedCentralCrossRef Hubbard TD, Murray IA, Bisson WH, Lahoti TS, Gowda K, Amin SG, et al. Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles. Sci Rep. 2015;5:12689.PubMedPubMedCentralCrossRef
235.
Zurück zum Zitat Cheng Y, Jin U-H, Allred CD, Jayaraman A, Chapkin RS, Safe S. Aryl Hydrocarbon Receptor Activity of Tryptophan Metabolites in Young Adult Mouse Colonocytes. Drug Metab Dispos. American Society for Pharmacology and Experimental Therapeutics; 2015;43:1536–43. Cheng Y, Jin U-H, Allred CD, Jayaraman A, Chapkin RS, Safe S. Aryl Hydrocarbon Receptor Activity of Tryptophan Metabolites in Young Adult Mouse Colonocytes. Drug Metab Dispos. American Society for Pharmacology and Experimental Therapeutics; 2015;43:1536–43.
236.
Zurück zum Zitat Cervantes-Barragan L, Chai JN, Tianero MD, Di Luccia B, Ahern PP, Merriman J, et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science. 2017;357:806–10.PubMedPubMedCentralCrossRef Cervantes-Barragan L, Chai JN, Tianero MD, Di Luccia B, Ahern PP, Merriman J, et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science. 2017;357:806–10.PubMedPubMedCentralCrossRef
237.
Zurück zum Zitat Schroeder JC, DiNatale BC, Murray IA, Flaveny CA, Liu Q, Laurenzana EM, et al. The Uremic Toxin 3-Indoxyl Sulfate Is a Potent Endogenous Agonist for the Human Aryl Hydrocarbon Receptor. Biochemistry American Chemical Society. 2010;49:393–400. Schroeder JC, DiNatale BC, Murray IA, Flaveny CA, Liu Q, Laurenzana EM, et al. The Uremic Toxin 3-Indoxyl Sulfate Is a Potent Endogenous Agonist for the Human Aryl Hydrocarbon Receptor. Biochemistry American Chemical Society. 2010;49:393–400.
238.
Zurück zum Zitat Hubbard T, Murray I, Bisson W, Lahoti T, Gowda K, Amin S, et al. Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles. Scientific Reports. 2015;5:12689. Hubbard T, Murray I, Bisson W, Lahoti T, Gowda K, Amin S, et al. Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles. Scientific Reports. 2015;5:12689.
239.
Zurück zum Zitat Ramos-García NA, Orozco-Ibarra M, Estudillo E, Elizondo G, Gómez Apo E, Chávez Macías LG, et al. Aryl Hydrocarbon Receptor in Post-Mortem Hippocampus and in Serum from Young, Elder, and Alzheimer’s Patients. International Journal of Molecular Sciences. Multidisciplinary Digital Publishing Institute; 2020;21:1983. Ramos-García NA, Orozco-Ibarra M, Estudillo E, Elizondo G, Gómez Apo E, Chávez Macías LG, et al. Aryl Hydrocarbon Receptor in Post-Mortem Hippocampus and in Serum from Young, Elder, and Alzheimer’s Patients. International Journal of Molecular Sciences. Multidisciplinary Digital Publishing Institute; 2020;21:1983.
240.
Zurück zum Zitat Rothhammer V, Mascanfroni ID, Bunse L, Takenaka MC, Kenison JE, Mayo L, et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and CNS inflammation via the aryl hydrocarbon receptor. Nat Med. 2016;22:586–97.PubMedPubMedCentralCrossRef Rothhammer V, Mascanfroni ID, Bunse L, Takenaka MC, Kenison JE, Mayo L, et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and CNS inflammation via the aryl hydrocarbon receptor. Nat Med. 2016;22:586–97.PubMedPubMedCentralCrossRef
241.
Zurück zum Zitat Wei GZ, Martin KA, Xing PY, Agrawal R, Whiley L, Wood TK, et al. Tryptophan-metabolizing gut microbes regulate adult neurogenesis via the aryl hydrocarbon receptor. Proceedings of the National Academy of Sciences. Proceedings of the National Academy of Sciences; 2021;118:e2021091118. Wei GZ, Martin KA, Xing PY, Agrawal R, Whiley L, Wood TK, et al. Tryptophan-metabolizing gut microbes regulate adult neurogenesis via the aryl hydrocarbon receptor. Proceedings of the National Academy of Sciences. Proceedings of the National Academy of Sciences; 2021;118:e2021091118.
242.
Zurück zum Zitat Bansal T, Alaniz RC, Wood TK, Jayaraman A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc Natl Acad Sci U S A. 2010;107:228–33.PubMedCrossRef Bansal T, Alaniz RC, Wood TK, Jayaraman A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc Natl Acad Sci U S A. 2010;107:228–33.PubMedCrossRef
243.
Zurück zum Zitat Shimada Y, Kinoshita M, Harada K, Mizutani M, Masahata K, Kayama H, et al. Commensal bacteria-dependent indole production enhances epithelial barrier function in the colon. PLoS ONE. 2013;8: e80604.PubMedPubMedCentralCrossRef Shimada Y, Kinoshita M, Harada K, Mizutani M, Masahata K, Kayama H, et al. Commensal bacteria-dependent indole production enhances epithelial barrier function in the colon. PLoS ONE. 2013;8: e80604.PubMedPubMedCentralCrossRef
244.
Zurück zum Zitat Yu M, Wang Q, Ma Y, Li L, Yu K, Zhang Z, et al. Aryl Hydrocarbon Receptor Activation Modulates Intestinal Epithelial Barrier Function by Maintaining Tight Junction Integrity. Int J Biol Sci. 2018;14:69–77.PubMedPubMedCentralCrossRef Yu M, Wang Q, Ma Y, Li L, Yu K, Zhang Z, et al. Aryl Hydrocarbon Receptor Activation Modulates Intestinal Epithelial Barrier Function by Maintaining Tight Junction Integrity. Int J Biol Sci. 2018;14:69–77.PubMedPubMedCentralCrossRef
245.
Zurück zum Zitat Wlodarska M, Luo C, Kolde R, d’Hennezel E, Annand JW, Heim CE, et al. Indoleacrylic Acid Produced by Commensal Peptostreptococcus Species Suppresses Inflammation. Cell Host Microbe. 2017;22:25-37.e6.PubMedPubMedCentralCrossRef Wlodarska M, Luo C, Kolde R, d’Hennezel E, Annand JW, Heim CE, et al. Indoleacrylic Acid Produced by Commensal Peptostreptococcus Species Suppresses Inflammation. Cell Host Microbe. 2017;22:25-37.e6.PubMedPubMedCentralCrossRef
246.
Zurück zum Zitat Joshi G, Johnson JA. The Nrf2-ARE pathway: a valuable therapeutic target for the treatment of neurodegenerative diseases. Recent Pat CNS Drug Discov. 2012;7:218–29.PubMedPubMedCentralCrossRef Joshi G, Johnson JA. The Nrf2-ARE pathway: a valuable therapeutic target for the treatment of neurodegenerative diseases. Recent Pat CNS Drug Discov. 2012;7:218–29.PubMedPubMedCentralCrossRef
247.
Zurück zum Zitat Tian Y, Wang W, Xu L, Li H, Wei Y, Wu Q, et al. Activation of Nrf2/ARE pathway alleviates the cognitive deficits in PS1V97L-Tg mouse model of Alzheimer’s disease through modulation of oxidative stress. J Neurosci Res. 2019;97:492–505.PubMedCrossRef Tian Y, Wang W, Xu L, Li H, Wei Y, Wu Q, et al. Activation of Nrf2/ARE pathway alleviates the cognitive deficits in PS1V97L-Tg mouse model of Alzheimer’s disease through modulation of oxidative stress. J Neurosci Res. 2019;97:492–505.PubMedCrossRef
248.
Zurück zum Zitat Liu Y, Li N, Zhou L, Li Q, Li W. Plasma metabolic profiling of mild cognitive impairment and Alzheimer’s disease using liquid chromatography/mass spectrometry. Cent Nerv Syst Agents Med Chem. 2014;14:113–20.PubMedCrossRef Liu Y, Li N, Zhou L, Li Q, Li W. Plasma metabolic profiling of mild cognitive impairment and Alzheimer’s disease using liquid chromatography/mass spectrometry. Cent Nerv Syst Agents Med Chem. 2014;14:113–20.PubMedCrossRef
249.
Zurück zum Zitat Fülling C, Dinan TG, Cryan JF. Gut microbe to brain signaling: what happens in vagus. Neuron. 2019;101:998–1002.PubMedCrossRef Fülling C, Dinan TG, Cryan JF. Gut microbe to brain signaling: what happens in vagus. Neuron. 2019;101:998–1002.PubMedCrossRef
252.
Zurück zum Zitat Boonstra E, de Kleijn R, Colzato LS, Alkemade A, Forstmann BU, Nieuwenhuis S. Neurotransmitters as food supplements: the effects of GABA on brain and behavior. Front Psychol. 2015;6:1520.PubMedPubMedCentralCrossRef Boonstra E, de Kleijn R, Colzato LS, Alkemade A, Forstmann BU, Nieuwenhuis S. Neurotransmitters as food supplements: the effects of GABA on brain and behavior. Front Psychol. 2015;6:1520.PubMedPubMedCentralCrossRef
253.
Zurück zum Zitat Matsumoto M, Kibe R, Ooga T, Aiba Y, Kurihara S, Sawaki E, et al. Impact of Intestinal Microbiota on Intestinal Luminal Metabolome. Sci Rep. Nature Publishing Group; 2012;2:233. Matsumoto M, Kibe R, Ooga T, Aiba Y, Kurihara S, Sawaki E, et al. Impact of Intestinal Microbiota on Intestinal Luminal Metabolome. Sci Rep. Nature Publishing Group; 2012;2:233.
254.
Zurück zum Zitat Deng P, Valentino T, Flythe MD, Moseley HNB, Leachman JR, Morris AJ, et al. Untargeted stable isotope probing of the gut microbiota metabolome using 13C-labeled dietary fibers. J Proteome Res American Chemical Society. 2021;20:2904–13.CrossRef Deng P, Valentino T, Flythe MD, Moseley HNB, Leachman JR, Morris AJ, et al. Untargeted stable isotope probing of the gut microbiota metabolome using 13C-labeled dietary fibers. J Proteome Res American Chemical Society. 2021;20:2904–13.CrossRef
255.
Zurück zum Zitat Strandwitz P, Kim KH, Terekhova D, Liu JK, Sharma A, Levering J, et al. GABA-modulating bacteria of the human gut microbiota. Nat Microbiol Nature Publishing Group. 2019;4:396–403.CrossRef Strandwitz P, Kim KH, Terekhova D, Liu JK, Sharma A, Levering J, et al. GABA-modulating bacteria of the human gut microbiota. Nat Microbiol Nature Publishing Group. 2019;4:396–403.CrossRef
256.
Zurück zum Zitat Needham BD, Kaddurah-Daouk R, Mazmanian SK. Gut microbial molecules in behavioural and neurodegenerative conditions. Nat Rev Neurosci Nature Publishing Group. 2020;21:717–31.CrossRef Needham BD, Kaddurah-Daouk R, Mazmanian SK. Gut microbial molecules in behavioural and neurodegenerative conditions. Nat Rev Neurosci Nature Publishing Group. 2020;21:717–31.CrossRef
257.
Zurück zum Zitat Janik R, Thomason LAM, Stanisz AM, Forsythe P, Bienenstock J, Stanisz GJ. Magnetic resonance spectroscopy reveals oral lactobacillus promotion of increases in brain GABA. N-acetyl aspartate and glutamate NeuroImage. 2016;125:988–95.PubMed Janik R, Thomason LAM, Stanisz AM, Forsythe P, Bienenstock J, Stanisz GJ. Magnetic resonance spectroscopy reveals oral lactobacillus promotion of increases in brain GABA. N-acetyl aspartate and glutamate NeuroImage. 2016;125:988–95.PubMed
258.
Zurück zum Zitat Saito Y, Sato T, Nomoto K, Tsuji H. Identification of phenol- and p-cresol-producing intestinal bacteria by using media supplemented with tyrosine and its metabolites. FEMS Microbiol Ecol. 2018;94:9.CrossRef Saito Y, Sato T, Nomoto K, Tsuji H. Identification of phenol- and p-cresol-producing intestinal bacteria by using media supplemented with tyrosine and its metabolites. FEMS Microbiol Ecol. 2018;94:9.CrossRef
259.
Zurück zum Zitat Cerini C, Dou L, Anfosso F, Sabatier F, Moal V, Glorieux G, et al. P-cresol, a uremic retention solute, alters the endothelial barrier function in vitro. Thromb Haemost. 2004;92:140–50.PubMedCrossRef Cerini C, Dou L, Anfosso F, Sabatier F, Moal V, Glorieux G, et al. P-cresol, a uremic retention solute, alters the endothelial barrier function in vitro. Thromb Haemost. 2004;92:140–50.PubMedCrossRef
260.
Zurück zum Zitat Pascucci T, Colamartino M, Fiori E, Sacco R, Coviello A, Ventura R, et al. P-cresol Alters Brain Dopamine Metabolism and Exacerbates Autism-Like Behaviors in the BTBR Mouse. Brain Sciences. Multidisciplinary Digital Publishing Institute; 2020;10:233. Pascucci T, Colamartino M, Fiori E, Sacco R, Coviello A, Ventura R, et al. P-cresol Alters Brain Dopamine Metabolism and Exacerbates Autism-Like Behaviors in the BTBR Mouse. Brain Sciences. Multidisciplinary Digital Publishing Institute; 2020;10:233.
261.
Zurück zum Zitat Sankowski B, Księżarczyk K, Raćkowska E, Szlufik S, Koziorowski D, Giebułtowicz J. Higher cerebrospinal fluid to plasma ratio of p-cresol sulfate and indoxyl sulfate in patients with Parkinson’s disease. Clin Chim Acta. 2020;501:165–73.PubMedCrossRef Sankowski B, Księżarczyk K, Raćkowska E, Szlufik S, Koziorowski D, Giebułtowicz J. Higher cerebrospinal fluid to plasma ratio of p-cresol sulfate and indoxyl sulfate in patients with Parkinson’s disease. Clin Chim Acta. 2020;501:165–73.PubMedCrossRef
262.
Zurück zum Zitat Al-Bachari S, Naish JH, Parker GJM, Emsley HCA, Parkes LM. Blood-brain barrier leakage is increased in parkinson’s disease. Front Physiol. 2020;11: 593026.PubMedPubMedCentralCrossRef Al-Bachari S, Naish JH, Parker GJM, Emsley HCA, Parkes LM. Blood-brain barrier leakage is increased in parkinson’s disease. Front Physiol. 2020;11: 593026.PubMedPubMedCentralCrossRef
263.
Zurück zum Zitat Azevedo MLV, Bonan NB, Dias G, Brehm F, Steiner TM, Souza WM, et al. p-Cresyl sulfate affects the oxidative burst, phagocytosis process, and antigen presentation of monocyte-derived macrophages. Toxicol Lett. 2016;263:1–5.PubMedCrossRef Azevedo MLV, Bonan NB, Dias G, Brehm F, Steiner TM, Souza WM, et al. p-Cresyl sulfate affects the oxidative burst, phagocytosis process, and antigen presentation of monocyte-derived macrophages. Toxicol Lett. 2016;263:1–5.PubMedCrossRef
264.
Zurück zum Zitat Edamatsu T, Fujieda A, Itoh Y. Phenyl sulfate, indoxyl sulfate and p-cresyl sulfate decrease glutathione level to render cells vulnerable to oxidative stress in renal tubular cells. PLOS ONE. Public Library of Science; 2018;13:e0193342. Edamatsu T, Fujieda A, Itoh Y. Phenyl sulfate, indoxyl sulfate and p-cresyl sulfate decrease glutathione level to render cells vulnerable to oxidative stress in renal tubular cells. PLOS ONE. Public Library of Science; 2018;13:e0193342.
265.
Zurück zum Zitat Sun C-Y, Cheng M-L, Pan H-C, Lee J-H, Lee C-C. Protein-bound uremic toxins impaired mitochondrial dynamics and functions. Oncotarget Impact Journals. 2017;8:77722–33.CrossRef Sun C-Y, Cheng M-L, Pan H-C, Lee J-H, Lee C-C. Protein-bound uremic toxins impaired mitochondrial dynamics and functions. Oncotarget Impact Journals. 2017;8:77722–33.CrossRef
266.
Zurück zum Zitat Tang W-H, Wang C-P, Yu T-H, Tai P-Y, Liang S-S, Hung W-C, et al. Protein-bounded uremic toxin p-cresylsulfate induces vascular permeability alternations. Histochem Cell Biol. 2018;149:607–17.PubMedCrossRef Tang W-H, Wang C-P, Yu T-H, Tai P-Y, Liang S-S, Hung W-C, et al. Protein-bounded uremic toxin p-cresylsulfate induces vascular permeability alternations. Histochem Cell Biol. 2018;149:607–17.PubMedCrossRef
267.
Zurück zum Zitat Sun C-Y, Li J-R, Wang Y-Y, Lin S-Y, Ou Y-C, Lin C-J, et al. p-Cresol sulfate caused behavior disorders and neurodegeneration in mice with unilateral nephrectomy involving oxidative stress and neuroinflammation. Int J Mol Sci. 2020;21:E6687.PubMedCrossRef Sun C-Y, Li J-R, Wang Y-Y, Lin S-Y, Ou Y-C, Lin C-J, et al. p-Cresol sulfate caused behavior disorders and neurodegeneration in mice with unilateral nephrectomy involving oxidative stress and neuroinflammation. Int J Mol Sci. 2020;21:E6687.PubMedCrossRef
268.
Zurück zum Zitat Koh A, Molinaro A, Ståhlman M, Khan MT, Schmidt C, Mannerås-Holm L, et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell. 2018;175:947-961.e17.PubMedCrossRef Koh A, Molinaro A, Ståhlman M, Khan MT, Schmidt C, Mannerås-Holm L, et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell. 2018;175:947-961.e17.PubMedCrossRef
269.
Zurück zum Zitat Molinaro A, Bel Lassen P, Henricsson M, Wu H, Adriouch S, Belda E, et al. Imidazole propionate is increased in diabetes and associated with dietary patterns and altered microbial ecology. Nat Commun. 2020;11:5881.PubMedPubMedCentralCrossRef Molinaro A, Bel Lassen P, Henricsson M, Wu H, Adriouch S, Belda E, et al. Imidazole propionate is increased in diabetes and associated with dietary patterns and altered microbial ecology. Nat Commun. 2020;11:5881.PubMedPubMedCentralCrossRef
270.
Zurück zum Zitat Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–6.PubMedCrossRef Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–6.PubMedCrossRef
271.
Zurück zum Zitat Koh A, Mannerås-Holm L, Yunn N-O, Nilsson PM, Ryu SH, Molinaro A, et al. Microbial imidazole propionate affects responses to metformin through p38γ-dependent inhibitory AMPK phosphorylation. Cell Metab. 2020;32:643-653.e4.PubMedPubMedCentralCrossRef Koh A, Mannerås-Holm L, Yunn N-O, Nilsson PM, Ryu SH, Molinaro A, et al. Microbial imidazole propionate affects responses to metformin through p38γ-dependent inhibitory AMPK phosphorylation. Cell Metab. 2020;32:643-653.e4.PubMedPubMedCentralCrossRef
272.
Zurück zum Zitat Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM. Diabetes mellitus and the risk of dementia: the rotterdam study. Neurology. 1999;53:1937–42.PubMedCrossRef Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM. Diabetes mellitus and the risk of dementia: the rotterdam study. Neurology. 1999;53:1937–42.PubMedCrossRef
273.
Zurück zum Zitat Peila R, Rodriguez BL, Launer LJ. Type 2 diabetes, APOE gene, and the risk for dementia and related pathologies: the honolulu-asia aging study. Diabetes American Diabetes Association. 2002;51:1256–62. Peila R, Rodriguez BL, Launer LJ. Type 2 diabetes, APOE gene, and the risk for dementia and related pathologies: the honolulu-asia aging study. Diabetes American Diabetes Association. 2002;51:1256–62.
274.
Zurück zum Zitat Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7.PubMedCrossRef Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7.PubMedCrossRef
275.
Zurück zum Zitat Luan H, Wang X, Cai Z. Mass spectrometry-based metabolomics: targeting the crosstalk between gut microbiota and brain in neurodegenerative disorders. Mass Spectrom Rev. 2019;38:22–33.PubMedCrossRef Luan H, Wang X, Cai Z. Mass spectrometry-based metabolomics: targeting the crosstalk between gut microbiota and brain in neurodegenerative disorders. Mass Spectrom Rev. 2019;38:22–33.PubMedCrossRef
276.
Zurück zum Zitat Smith EA, Macfarlane GT. Dissimilatory amino acid metabolism in human colonic bacteria. Anaerobe. 1997;3:327–37.PubMedCrossRef Smith EA, Macfarlane GT. Dissimilatory amino acid metabolism in human colonic bacteria. Anaerobe. 1997;3:327–37.PubMedCrossRef
277.
Zurück zum Zitat Ferreira-Vieira TH, Guimaraes IM, Silva FR, Ribeiro FM. Alzheimer’s disease: targeting the cholinergic system. Curr Neuropharmacol. 2016;14:101–15.PubMedPubMedCentralCrossRef Ferreira-Vieira TH, Guimaraes IM, Silva FR, Ribeiro FM. Alzheimer’s disease: targeting the cholinergic system. Curr Neuropharmacol. 2016;14:101–15.PubMedPubMedCentralCrossRef
278.
Zurück zum Zitat Fricker RA, Green EL, Jenkins SI, Griffin SM. The Influence of Nicotinamide on Health and Disease in the Central Nervous System. Int J Tryptophan Res. 2018;11:1178646918776658. Fricker RA, Green EL, Jenkins SI, Griffin SM. The Influence of Nicotinamide on Health and Disease in the Central Nervous System. Int J Tryptophan Res. 2018;11:1178646918776658.
279.
Zurück zum Zitat Phelan M, Phase II. Clinical Trial of Nicotinamide for the Treatment of Mild to Moderate Alzheimer’s Disease. Journal of Geriatric Medicine and Gerontology. 2017;3:1–7.CrossRef Phelan M, Phase II. Clinical Trial of Nicotinamide for the Treatment of Mild to Moderate Alzheimer’s Disease. Journal of Geriatric Medicine and Gerontology. 2017;3:1–7.CrossRef
280.
Zurück zum Zitat Kim L-J, Chalmers TJ, Smith GC, Das A, Poon EWK, Wang J, et al. Nicotinamide mononucleotide (NMN) deamidation by the gut microbiome and evidence for indirect upregulation of the NAD+ metabolome. bioRxiv. Cold Spring Harbor Laboratory; 2020;289561:1–85. Kim L-J, Chalmers TJ, Smith GC, Das A, Poon EWK, Wang J, et al. Nicotinamide mononucleotide (NMN) deamidation by the gut microbiome and evidence for indirect upregulation of the NAD+ metabolome. bioRxiv. Cold Spring Harbor Laboratory; 2020;289561:1–85.
281.
Zurück zum Zitat Wang D, Ho L, Faith J, Ono K, Janle EM, Lachcik PJ, et al. Role of intestinal microbiota in the generation of polyphenol-derived phenolic acid mediated attenuation of Alzheimer’s disease β-amyloid oligomerization. Mol Nutr Food Res. 2015;59:1025–40.PubMedPubMedCentralCrossRef Wang D, Ho L, Faith J, Ono K, Janle EM, Lachcik PJ, et al. Role of intestinal microbiota in the generation of polyphenol-derived phenolic acid mediated attenuation of Alzheimer’s disease β-amyloid oligomerization. Mol Nutr Food Res. 2015;59:1025–40.PubMedPubMedCentralCrossRef
282.
Zurück zum Zitat Faria A, Mateus N, Calhau C. Flavonoid transport across blood-brain barrier: implication for their direct neuroprotective actions. Nutrition and Aging IOS Press. 2012;1:89–97.CrossRef Faria A, Mateus N, Calhau C. Flavonoid transport across blood-brain barrier: implication for their direct neuroprotective actions. Nutrition and Aging IOS Press. 2012;1:89–97.CrossRef
283.
Zurück zum Zitat Faundes V, Jennings MD, Crilly S, Legraie S, Withers SE, Cuvertino S, et al. Impaired eIF5A function causes a Mendelian disorder that is partially rescued in model systems by spermidine. Nat Commun. Nature Publishing Group; 2021;12:833. Faundes V, Jennings MD, Crilly S, Legraie S, Withers SE, Cuvertino S, et al. Impaired eIF5A function causes a Mendelian disorder that is partially rescued in model systems by spermidine. Nat Commun. Nature Publishing Group; 2021;12:833.
284.
Zurück zum Zitat Guerra GP, Rubin MA, Mello CF. Modulation of learning and memory by natural polyamines. Pharmacol Res. 2016;112:99–118.PubMedCrossRef Guerra GP, Rubin MA, Mello CF. Modulation of learning and memory by natural polyamines. Pharmacol Res. 2016;112:99–118.PubMedCrossRef
285.
Zurück zum Zitat Leclerc M, Dudonné S, Calon F. Can Natural Products Exert Neuroprotection without Crossing the Blood-Brain Barrier? Int J Mol Sci. 2021;22:3356.PubMedPubMedCentralCrossRef Leclerc M, Dudonné S, Calon F. Can Natural Products Exert Neuroprotection without Crossing the Blood-Brain Barrier? Int J Mol Sci. 2021;22:3356.PubMedPubMedCentralCrossRef
286.
Zurück zum Zitat Szwajgier D, Borowiec K, Pustelniak K. The Neuroprotective Effects of Phenolic Acids: Molecular Mechanism of Action. Nutrients. 2017;9:477.PubMedCentralCrossRef Szwajgier D, Borowiec K, Pustelniak K. The Neuroprotective Effects of Phenolic Acids: Molecular Mechanism of Action. Nutrients. 2017;9:477.PubMedCentralCrossRef
287.
Zurück zum Zitat Hankes LV, Coenen HH, Rota E, Langen KJ, Herzog H, Wutz W, et al. Effect of huntington’s and alzheimer’s diseases on the transport of nicotinic acid or nicotinamide across the human blood-brain barrier. Adv Exp Med Biol. 1991;294:675–8.PubMedCrossRef Hankes LV, Coenen HH, Rota E, Langen KJ, Herzog H, Wutz W, et al. Effect of huntington’s and alzheimer’s diseases on the transport of nicotinic acid or nicotinamide across the human blood-brain barrier. Adv Exp Med Biol. 1991;294:675–8.PubMedCrossRef
288.
Zurück zum Zitat Hole KL, Williams RJ. Flavonoids as an intervention for alzheimer’s disease: progress and hurdles towards defining a mechanism of action. Brain Plasticity IOS Press. 2020;6:167–92.CrossRef Hole KL, Williams RJ. Flavonoids as an intervention for alzheimer’s disease: progress and hurdles towards defining a mechanism of action. Brain Plasticity IOS Press. 2020;6:167–92.CrossRef
289.
Zurück zum Zitat Carecho R, Carregosa D, dos Santos CN. Low molecular weight (poly)phenol metabolites across the blood-brain barrier: the underexplored journey. Brain Plasticity IOS Press. 2020;6:193–214.CrossRef Carecho R, Carregosa D, dos Santos CN. Low molecular weight (poly)phenol metabolites across the blood-brain barrier: the underexplored journey. Brain Plasticity IOS Press. 2020;6:193–214.CrossRef
290.
Zurück zum Zitat 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 Review of Neurotherapeutics. Taylor & Francis; 2018;18:83–90. 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 Review of Neurotherapeutics. Taylor & Francis; 2018;18:83–90.
291.
Zurück zum Zitat Berding K, Carbia C, Cryan JF. Going with the grain: Fiber, cognition, and the microbiota-gut-brain-axis. Exp Biol Med (Maywood). SAGE Publications; 2021;246:796–811. Berding K, Carbia C, Cryan JF. Going with the grain: Fiber, cognition, and the microbiota-gut-brain-axis. Exp Biol Med (Maywood). SAGE Publications; 2021;246:796–811.
292.
293.
Zurück zum Zitat Oldendorf W. Carrier-mediated blood-brain barrier transport of short-chain monocarboxylic organic acids. American Journal of Physiology-Legacy Content. American Physiological Society; 1973;224:1450–3. Oldendorf W. Carrier-mediated blood-brain barrier transport of short-chain monocarboxylic organic acids. American Journal of Physiology-Legacy Content. American Physiological Society; 1973;224:1450–3.
294.
Zurück zum Zitat 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. 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.
295.
Zurück zum Zitat Li H, Sun J, Wang F, Ding G, Chen W, Fang R, et al. Sodium butyrate exerts neuroprotective effects by restoring the blood-brain barrier in traumatic brain injury mice. Brain Res. 2016;1642:70–8.PubMedCrossRef Li H, Sun J, Wang F, Ding G, Chen W, Fang R, et al. Sodium butyrate exerts neuroprotective effects by restoring the blood-brain barrier in traumatic brain injury mice. Brain Res. 2016;1642:70–8.PubMedCrossRef
296.
Zurück zum Zitat Harman MF, Martín MG. Epigenetic mechanisms related to cognitive decline during aging. J Neurosci Res. 2020;98:234–46.PubMedCrossRef Harman MF, Martín MG. Epigenetic mechanisms related to cognitive decline during aging. J Neurosci Res. 2020;98:234–46.PubMedCrossRef
297.
Zurück zum Zitat Patnala R, Arumugam TV, Gupta N, Dheen ST. HDAC Inhibitor sodium butyrate-mediated epigenetic regulation enhances neuroprotective function of microglia during ischemic stroke. Mol Neurobiol. 2017;54:6391–411.PubMedCrossRef Patnala R, Arumugam TV, Gupta N, Dheen ST. HDAC Inhibitor sodium butyrate-mediated epigenetic regulation enhances neuroprotective function of microglia during ischemic stroke. Mol Neurobiol. 2017;54:6391–411.PubMedCrossRef
298.
Zurück zum Zitat Huuskonen J, Suuronen T, Nuutinen T, Kyrylenko S, Salminen A. Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids. Br J Pharmacol. 2004;141:874–80.PubMedPubMedCentralCrossRef Huuskonen J, Suuronen T, Nuutinen T, Kyrylenko S, Salminen A. Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids. Br J Pharmacol. 2004;141:874–80.PubMedPubMedCentralCrossRef
299.
Zurück zum Zitat Wei YB, Melas PA, Wegener G, Mathé AA, Lavebratt C. Antidepressant-Like Effect of Sodium Butyrate is Associated with an Increase in TET1 and in 5-Hydroxymethylation Levels in the Bdnf Gene. International Journal of Neuropsychopharmacology. 2015;18:pyu032. Wei YB, Melas PA, Wegener G, Mathé AA, Lavebratt C. Antidepressant-Like Effect of Sodium Butyrate is Associated with an Increase in TET1 and in 5-Hydroxymethylation Levels in the Bdnf Gene. International Journal of Neuropsychopharmacology. 2015;18:pyu032.
300.
Zurück zum Zitat Shin H, Kim J-H, Lee YS, Lee YC. Change in gene expression profiles of secreted frizzled-related proteins (SFRPs) by sodium butyrate in gastric cancers: Induction of promoter demethylation and histone modification causing inhibition of Wnt signaling. International Journal of Oncology Spandidos Publications. 2012;40:1533–42. Shin H, Kim J-H, Lee YS, Lee YC. Change in gene expression profiles of secreted frizzled-related proteins (SFRPs) by sodium butyrate in gastric cancers: Induction of promoter demethylation and histone modification causing inhibition of Wnt signaling. International Journal of Oncology Spandidos Publications. 2012;40:1533–42.
301.
Zurück zum Zitat Sarkar S, Abujamra AL, Loew JE, Forman LW, Perrine SP, Faller DV. Histone Deacetylase Inhibitors Reverse CpG Methylation by Regulating DNMT1 through ERK Signaling. Anticancer Research. International Institute of Anticancer Research; 2011;31:2723–32. Sarkar S, Abujamra AL, Loew JE, Forman LW, Perrine SP, Faller DV. Histone Deacetylase Inhibitors Reverse CpG Methylation by Regulating DNMT1 through ERK Signaling. Anticancer Research. International Institute of Anticancer Research; 2011;31:2723–32.
302.
Zurück zum Zitat Yamawaki Y, Yoshioka N, Nozaki K, Ito H, Oda K, Harada K, et al. Sodium butyrate abolishes lipopolysaccharide-induced depression-like behaviors and hippocampal microglial activation in mice. Brain Res. 2018;1680:13–38.PubMedCrossRef Yamawaki Y, Yoshioka N, Nozaki K, Ito H, Oda K, Harada K, et al. Sodium butyrate abolishes lipopolysaccharide-induced depression-like behaviors and hippocampal microglial activation in mice. Brain Res. 2018;1680:13–38.PubMedCrossRef
303.
Zurück zum Zitat Soliman ML, Smith MD, Houdek HM, Rosenberger TA. Acetate supplementation modulates brain histone acetylation and decreases interleukin-1β expression in a rat model of neuroinflammation. J Neuroinflammation. 2012;9:51.PubMedPubMedCentralCrossRef Soliman ML, Smith MD, Houdek HM, Rosenberger TA. Acetate supplementation modulates brain histone acetylation and decreases interleukin-1β expression in a rat model of neuroinflammation. J Neuroinflammation. 2012;9:51.PubMedPubMedCentralCrossRef
304.
Zurück zum Zitat Soliman ML, Combs CK, Rosenberger TA. Modulation of inflammatory cytokines and mitogen-activated protein kinases by acetate in primary astrocytes. J Neuroimmune Pharmacol. 2013;8:287–300.PubMedCrossRef Soliman ML, Combs CK, Rosenberger TA. Modulation of inflammatory cytokines and mitogen-activated protein kinases by acetate in primary astrocytes. J Neuroimmune Pharmacol. 2013;8:287–300.PubMedCrossRef
305.
Zurück zum Zitat Wang P, Zhang Y, Gong Y, Yang R, Chen Z, Hu W, et al. Sodium butyrate triggers a functional elongation of microglial process via Akt-small RhoGTPase activation and HDACs inhibition. Neurobiol Dis. 2018;111:12–25.PubMedCrossRef Wang P, Zhang Y, Gong Y, Yang R, Chen Z, Hu W, et al. Sodium butyrate triggers a functional elongation of microglial process via Akt-small RhoGTPase activation and HDACs inhibition. Neurobiol Dis. 2018;111:12–25.PubMedCrossRef
306.
Zurück zum Zitat Den Besten G, Van Eunen K, Groen AK, Venema K, Reijngoud D-J, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013;54:2325–40.CrossRef Den Besten G, Van Eunen K, Groen AK, Venema K, Reijngoud D-J, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013;54:2325–40.CrossRef
307.
Zurück zum Zitat Zilberter Y, Zilberter M. The vicious circle of hypometabolism in neurodegenerative diseases: ways and mechanisms of metabolic correction. J Neurosci Res. 2017;95:2217–35.PubMedCrossRef Zilberter Y, Zilberter M. The vicious circle of hypometabolism in neurodegenerative diseases: ways and mechanisms of metabolic correction. J Neurosci Res. 2017;95:2217–35.PubMedCrossRef
308.
309.
Zurück zum Zitat Colombo AV, Sadler RK, Llovera G, Singh V, Roth S, Heindl S, et al. Microbiota-derived short chain fatty acids modulate microglia and promote Aβ plaque deposition. Elife. 2021;10: e59826.PubMedPubMedCentralCrossRef Colombo AV, Sadler RK, Llovera G, Singh V, Roth S, Heindl S, et al. Microbiota-derived short chain fatty acids modulate microglia and promote Aβ plaque deposition. Elife. 2021;10: e59826.PubMedPubMedCentralCrossRef
310.
Zurück zum Zitat Marizzoni M, Cattaneo A, Mirabelli P, Festari C, Lopizzo N, Nicolosi V, et al. Short-chain fatty acids and lipopolysaccharide as mediators between gut dysbiosis and amyloid pathology in alzheimer’s disease. J Alzheimers Dis. 2020;78:683–97.PubMedCrossRef Marizzoni M, Cattaneo A, Mirabelli P, Festari C, Lopizzo N, Nicolosi V, et al. Short-chain fatty acids and lipopolysaccharide as mediators between gut dysbiosis and amyloid pathology in alzheimer’s disease. J Alzheimers Dis. 2020;78:683–97.PubMedCrossRef
311.
Zurück zum Zitat Tran TTT, Corsini S, Kellingray L, Hegarty C, Le Gall G, Narbad A, et al. APOE genotype influences the gut microbiome structure and function in humans and mice: relevance for alzheimer’s disease pathophysiology. Faseb J. 2019;33:8221–31.PubMedPubMedCentralCrossRef Tran TTT, Corsini S, Kellingray L, Hegarty C, Le Gall G, Narbad A, et al. APOE genotype influences the gut microbiome structure and function in humans and mice: relevance for alzheimer’s disease pathophysiology. Faseb J. 2019;33:8221–31.PubMedPubMedCentralCrossRef
312.
Zurück zum Zitat Nguyen TTT, Fujimura Y, Mimura I, Fujii Y, Nguyen NL, Arakawa K, et al. Cultivable butyrate-producing bacteria of elderly japanese diagnosed with alzheimer’s disease. J Microbiol. 2018;56:760–71.PubMedCrossRef Nguyen TTT, Fujimura Y, Mimura I, Fujii Y, Nguyen NL, Arakawa K, et al. Cultivable butyrate-producing bacteria of elderly japanese diagnosed with alzheimer’s disease. J Microbiol. 2018;56:760–71.PubMedCrossRef
313.
Zurück zum Zitat Liebisch G, Ecker J, Roth S, Schweizer S, Öttl V, Schött H-F, et al. Quantification of Fecal Short Chain Fatty Acids by Liquid Chromatography Tandem Mass Spectrometry—Investigation of Pre-Analytic Stability. Biomolecules. Multidisciplinary Digital Publishing Institute; 2019;9:121. Liebisch G, Ecker J, Roth S, Schweizer S, Öttl V, Schött H-F, et al. Quantification of Fecal Short Chain Fatty Acids by Liquid Chromatography Tandem Mass Spectrometry—Investigation of Pre-Analytic Stability. Biomolecules. Multidisciplinary Digital Publishing Institute; 2019;9:121.
314.
Zurück zum Zitat Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ, Brummer R-J. Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther. 2008;27:104–19.PubMedCrossRef Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ, Brummer R-J. Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther. 2008;27:104–19.PubMedCrossRef
315.
Zurück zum Zitat Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG, Cryan JF. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem Int. 2016;99:110–32.PubMedCrossRef Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG, Cryan JF. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem Int. 2016;99:110–32.PubMedCrossRef
316.
Zurück zum Zitat Horiuchi Y, Kimura R, Kato N, Fujii T, Seki M, Endo T, et al. Evolutional study on acetylcholine expression. Life Sci. 2003;72:1745–56.PubMedCrossRef Horiuchi Y, Kimura R, Kato N, Fujii T, Seki M, Endo T, et al. Evolutional study on acetylcholine expression. Life Sci. 2003;72:1745–56.PubMedCrossRef
317.
Zurück zum Zitat Koussoulas K, Swaminathan M, Fung C, Bornstein JC, Foong JPP. Neurally released GABA acts via GABAC receptors to modulate Ca2+ transients evoked by trains of synaptic inputs, but not responses evoked by single stimuli, in myenteric neurons of mouse ileum. Front Physiol. 2018;9:97.PubMedPubMedCentralCrossRef Koussoulas K, Swaminathan M, Fung C, Bornstein JC, Foong JPP. Neurally released GABA acts via GABAC receptors to modulate Ca2+ transients evoked by trains of synaptic inputs, but not responses evoked by single stimuli, in myenteric neurons of mouse ileum. Front Physiol. 2018;9:97.PubMedPubMedCentralCrossRef
318.
Zurück zum Zitat Hampel H, Mesulam MM, Cuello AC, Farlow MR, Giacobini E, Grossberg GT, et al. The cholinergic system in the pathophysiology and treatment of alzheimer’s disease. Brain. 2018;141:1917–33.PubMedPubMedCentralCrossRef Hampel H, Mesulam MM, Cuello AC, Farlow MR, Giacobini E, Grossberg GT, et al. The cholinergic system in the pathophysiology and treatment of alzheimer’s disease. Brain. 2018;141:1917–33.PubMedPubMedCentralCrossRef
319.
Zurück zum Zitat Cohen EL, Wurtman RJ. Brain acetylcholine: increase after systemic choline administration. Life Sci. 1975;16:1095–102.PubMedCrossRef Cohen EL, Wurtman RJ. Brain acetylcholine: increase after systemic choline administration. Life Sci. 1975;16:1095–102.PubMedCrossRef
321.
Zurück zum Zitat Nieoullon A. Dopamine and the regulation of cognition and attention. Prog Neurobiol. 2002;67:53–83.PubMedCrossRef Nieoullon A. Dopamine and the regulation of cognition and attention. Prog Neurobiol. 2002;67:53–83.PubMedCrossRef
322.
Zurück zum Zitat Klein MO, Battagello DS, Cardoso AR, Hauser DN, Bittencourt JC, Correa RG. Dopamine: functions, signaling, and association with neurological diseases. Cell Mol Neurobiol. 2019;39:31–59.PubMedCrossRef Klein MO, Battagello DS, Cardoso AR, Hauser DN, Bittencourt JC, Correa RG. Dopamine: functions, signaling, and association with neurological diseases. Cell Mol Neurobiol. 2019;39:31–59.PubMedCrossRef
323.
Zurück zum Zitat Kageyama T, Nakamura M, Matsuo A, Yamasaki Y, Takakura Y, Hashida M, et al. The 4F2hc/LAT1 complex transports L-DOPA across the blood-brain barrier. Brain Res. 2000;879:115–21.PubMedCrossRef Kageyama T, Nakamura M, Matsuo A, Yamasaki Y, Takakura Y, Hashida M, et al. The 4F2hc/LAT1 complex transports L-DOPA across the blood-brain barrier. Brain Res. 2000;879:115–21.PubMedCrossRef
324.
325.
Zurück zum Zitat Asano Y, Hiramoto T, Nishino R, Aiba Y, Kimura T, Yoshihara K, et al. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. American Journal of Physiology-Gastrointestinal and Liver Physiology. American Physiological Society; 2012;303:1288–95. Asano Y, Hiramoto T, Nishino R, Aiba Y, Kimura T, Yoshihara K, et al. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. American Journal of Physiology-Gastrointestinal and Liver Physiology. American Physiological Society; 2012;303:1288–95.
326.
Zurück zum Zitat Hamamah S, Aghazarian A, Nazaryan A, Hajnal A, Covasa M. Role of Microbiota-Gut-Brain Axis in Regulating Dopaminergic Signaling. Biomedicines. Multidisciplinary Digital Publishing Institute; 2022;10:436. Hamamah S, Aghazarian A, Nazaryan A, Hajnal A, Covasa M. Role of Microbiota-Gut-Brain Axis in Regulating Dopaminergic Signaling. Biomedicines. Multidisciplinary Digital Publishing Institute; 2022;10:436.
327.
Zurück zum Zitat van Kessel SP, Frye AK, El-Gendy AO, Castejon M, Keshavarzian A, van Dijk G, et al. Gut bacterial tyrosine decarboxylases restrict levels of levodopa in the treatment of Parkinson’s disease. Nat Commun. Nature Publishing Group; 2019;10:310. van Kessel SP, Frye AK, El-Gendy AO, Castejon M, Keshavarzian A, van Dijk G, et al. Gut bacterial tyrosine decarboxylases restrict levels of levodopa in the treatment of Parkinson’s disease. Nat Commun. Nature Publishing Group; 2019;10:310.
328.
Zurück zum Zitat Fonteh AN, Harrington RJ, Tsai A, Liao P, Harrington MG. Free amino acid and dipeptide changes in the body fluids from alzheimer’s disease subjects. Amino Acids. 2007;32:213–24.PubMedCrossRef Fonteh AN, Harrington RJ, Tsai A, Liao P, Harrington MG. Free amino acid and dipeptide changes in the body fluids from alzheimer’s disease subjects. Amino Acids. 2007;32:213–24.PubMedCrossRef
329.
Zurück zum Zitat Flanagan E, Müller M, Hornberger M, Vauzour D. Impact of flavonoids on cellular and molecular mechanisms underlying age-related cognitive decline and neurodegeneration. Curr Nutr Rep. 2018;7:49–57.PubMedPubMedCentralCrossRef Flanagan E, Müller M, Hornberger M, Vauzour D. Impact of flavonoids on cellular and molecular mechanisms underlying age-related cognitive decline and neurodegeneration. Curr Nutr Rep. 2018;7:49–57.PubMedPubMedCentralCrossRef
330.
Zurück zum Zitat Krautkramer KA, Fan J, Bäckhed F. Gut microbial metabolites as multi-kingdom intermediates. Nature Reviews Microbiology Nature Publishing Group. 2021;19:77–94.CrossRef Krautkramer KA, Fan J, Bäckhed F. Gut microbial metabolites as multi-kingdom intermediates. Nature Reviews Microbiology Nature Publishing Group. 2021;19:77–94.CrossRef
331.
Zurück zum Zitat Russell WR, Duncan SH, Scobbie L, Duncan G, Cantlay L, Calder AG, et al. Major phenylpropanoid-derived metabolites in the human gut can arise from microbial fermentation of protein. Mol Nutr Food Res. 2013;57:523–35.PubMedCrossRef Russell WR, Duncan SH, Scobbie L, Duncan G, Cantlay L, Calder AG, et al. Major phenylpropanoid-derived metabolites in the human gut can arise from microbial fermentation of protein. Mol Nutr Food Res. 2013;57:523–35.PubMedCrossRef
332.
Zurück zum Zitat Frolinger T, Sims S, Smith C, Wang J, Cheng H, Faith J, et al. The gut microbiota composition affects dietary polyphenols-mediated cognitive resilience in mice by modulating the bioavailability of phenolic acids. Sci Rep. Nature Publishing Group; 2019;9:3546. Frolinger T, Sims S, Smith C, Wang J, Cheng H, Faith J, et al. The gut microbiota composition affects dietary polyphenols-mediated cognitive resilience in mice by modulating the bioavailability of phenolic acids. Sci Rep. Nature Publishing Group; 2019;9:3546.
333.
Zurück zum Zitat Esteban-Fernández A, Rendeiro C, Spencer JPE, del Coso DG, de Llano MDG, Bartolomé B, et al. Neuroprotective effects of selected microbial-derived phenolic metabolites and aroma compounds from wine in human sh-sy5y neuroblastoma cells and their putative mechanisms of action. Front Nutr. 2017;4:3.PubMedPubMedCentralCrossRef Esteban-Fernández A, Rendeiro C, Spencer JPE, del Coso DG, de Llano MDG, Bartolomé B, et al. Neuroprotective effects of selected microbial-derived phenolic metabolites and aroma compounds from wine in human sh-sy5y neuroblastoma cells and their putative mechanisms of action. Front Nutr. 2017;4:3.PubMedPubMedCentralCrossRef
334.
Zurück zum Zitat Huang Y, Zheng H, Tan K, Sun X, Ye J, Zhang Y. Circulating metabolomics profiling reveals novel pathways associated with cognitive decline in patients with hypertension. Ther Adv Neurol Disord. SAGE Publications Ltd STM; 2020;13:1756286420947973. Huang Y, Zheng H, Tan K, Sun X, Ye J, Zhang Y. Circulating metabolomics profiling reveals novel pathways associated with cognitive decline in patients with hypertension. Ther Adv Neurol Disord. SAGE Publications Ltd STM; 2020;13:1756286420947973.
335.
336.
Zurück zum Zitat Morrison LD, Becker L, Ang LC, Kish SJ. Polyamines in human brain: regional distribution and influence of aging. J Neurochem. 1995;65:636–42.PubMedCrossRef Morrison LD, Becker L, Ang LC, Kish SJ. Polyamines in human brain: regional distribution and influence of aging. J Neurochem. 1995;65:636–42.PubMedCrossRef
337.
Zurück zum Zitat Tofalo R, Cocchi S, Suzzi G. Polyamines and Gut Microbiota Front Nutr. 2019;6:16.PubMed Tofalo R, Cocchi S, Suzzi G. Polyamines and Gut Microbiota Front Nutr. 2019;6:16.PubMed
338.
Zurück zum Zitat Swann JR, Spitzer SO, Diaz HR. Developmental signatures of microbiota-derived metabolites in the mouse brain. Metabolites. 2020;10:172.PubMedCentralCrossRef Swann JR, Spitzer SO, Diaz HR. Developmental signatures of microbiota-derived metabolites in the mouse brain. Metabolites. 2020;10:172.PubMedCentralCrossRef
339.
Zurück zum Zitat Inoue K, Tsutsui H, Akatsu H, Hashizume Y, Matsukawa N, Yamamoto T, et al. Metabolic profiling of Alzheimer’s disease brains. Sci Rep. Nature Publishing Group; 2013;3:2364. Inoue K, Tsutsui H, Akatsu H, Hashizume Y, Matsukawa N, Yamamoto T, et al. Metabolic profiling of Alzheimer’s disease brains. Sci Rep. Nature Publishing Group; 2013;3:2364.
340.
Zurück zum Zitat Soda K, Kawakami M. Mediterranean diet and polyamine intake: possible contribution of increased polyamine intake to inhibition of age-associated disease. Nutr Diet Suppl. 2010;3:1. Soda K, Kawakami M. Mediterranean diet and polyamine intake: possible contribution of increased polyamine intake to inhibition of age-associated disease. Nutr Diet Suppl. 2010;3:1.
341.
Zurück zum Zitat Kibe R, Kurihara S, Sakai Y, Suzuki H, Ooga T, Sawaki E, et al. Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Sci Rep. Nature Publishing Group; 2014;4:4548. Kibe R, Kurihara S, Sakai Y, Suzuki H, Ooga T, Sawaki E, et al. Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Sci Rep. Nature Publishing Group; 2014;4:4548.
342.
Zurück zum Zitat Joaquim HPG, Costa AC, Forlenza OV, Gattaz WF, Talib LL. Decreased plasmatic spermidine and increased spermine in mild cognitive impairment and Alzheimer’s disease patients. Arch Clin Psychiatry (São Paulo). Faculdade de Medicina da Universidade de São Paulo; 2019;46:120–4. Joaquim HPG, Costa AC, Forlenza OV, Gattaz WF, Talib LL. Decreased plasmatic spermidine and increased spermine in mild cognitive impairment and Alzheimer’s disease patients. Arch Clin Psychiatry (São Paulo). Faculdade de Medicina da Universidade de São Paulo; 2019;46:120–4.
343.
Zurück zum Zitat Pekar T, Wendzel A, Flak W, Kremer A, Pauschenwein-Frantsich S, Gschaider A, et al. Spermidine in dementia : relation to age and memory performance. Wien Klin Wochenschr. 2020;132:42–6.PubMedCrossRef Pekar T, Wendzel A, Flak W, Kremer A, Pauschenwein-Frantsich S, Gschaider A, et al. Spermidine in dementia : relation to age and memory performance. Wien Klin Wochenschr. 2020;132:42–6.PubMedCrossRef
344.
Zurück zum Zitat Madeo F, Bauer MA, Carmona-Gutierrez D, Kroemer G. Spermidine: a physiological autophagy inducer acting as an anti-aging vitamin in humans? Autophagy. 2018;15:165–8.PubMedPubMedCentralCrossRef Madeo F, Bauer MA, Carmona-Gutierrez D, Kroemer G. Spermidine: a physiological autophagy inducer acting as an anti-aging vitamin in humans? Autophagy. 2018;15:165–8.PubMedPubMedCentralCrossRef
345.
Zurück zum Zitat Schwarz C, Horn N, Benson G, Wrachtrup Calzado I, Wurdack K, Pechlaner R, et al. Spermidine intake is associated with cortical thickness and hippocampal volume in older adults. Neuroimage. 2020;221: 117132.PubMedCrossRef Schwarz C, Horn N, Benson G, Wrachtrup Calzado I, Wurdack K, Pechlaner R, et al. Spermidine intake is associated with cortical thickness and hippocampal volume in older adults. Neuroimage. 2020;221: 117132.PubMedCrossRef
346.
Zurück zum Zitat Gruendler R, Hippe B, Sendula Jengic V, Peterlin B, Haslberger AG. Nutraceutical Approaches of Autophagy and Neuroinflammation in Alzheimer’s Disease: A Systematic Review. Molecules. 2020;25:6018.PubMedCentralCrossRef Gruendler R, Hippe B, Sendula Jengic V, Peterlin B, Haslberger AG. Nutraceutical Approaches of Autophagy and Neuroinflammation in Alzheimer’s Disease: A Systematic Review. Molecules. 2020;25:6018.PubMedCentralCrossRef
347.
Zurück zum Zitat Madeo F, Eisenberg T, Pietrocola F, Kroemer G. Spermidine in health and disease. Science. 2018;359:eaan2788. Madeo F, Eisenberg T, Pietrocola F, Kroemer G. Spermidine in health and disease. Science. 2018;359:eaan2788.
348.
Zurück zum Zitat Misrani A, Tabassum S, Yang L. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer’s Disease. Front Aging Neurosci. 2021;13:617588. Misrani A, Tabassum S, Yang L. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer’s Disease. Front Aging Neurosci. 2021;13:617588.
349.
Zurück zum Zitat Simpson DSA, Oliver PL. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants (Basel). 2020;9:743.PubMedCentralCrossRef Simpson DSA, Oliver PL. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants (Basel). 2020;9:743.PubMedCentralCrossRef
350.
Zurück zum Zitat De Risi M, Torromino G, Tufano M, Moriceau S, Pignataro A, Rivagorda M, et al. Mechanisms by which autophagy regulates memory capacity in ageing. Aging Cell. 2020;19: e13189.PubMedPubMedCentral De Risi M, Torromino G, Tufano M, Moriceau S, Pignataro A, Rivagorda M, et al. Mechanisms by which autophagy regulates memory capacity in ageing. Aging Cell. 2020;19: e13189.PubMedPubMedCentral
352.
Zurück zum Zitat Puleston DJ, Buck MD, Klein Geltink RI, Kyle RL, Caputa G, O’Sullivan D, et al. Polyamines and eIF5A hypusination modulate mitochondrial respiration and macrophage activation. Cell Metab. 2019;30:352-363.e8.PubMedPubMedCentralCrossRef Puleston DJ, Buck MD, Klein Geltink RI, Kyle RL, Caputa G, O’Sullivan D, et al. Polyamines and eIF5A hypusination modulate mitochondrial respiration and macrophage activation. Cell Metab. 2019;30:352-363.e8.PubMedPubMedCentralCrossRef
353.
Zurück zum Zitat Schroeder S, Hofer SJ, Zimmermann A, Pechlaner R, Dammbrueck C, Pendl T, et al. Dietary spermidine improves cognitive function. Cell Rep. 2021;35: 108985.PubMedCrossRef Schroeder S, Hofer SJ, Zimmermann A, Pechlaner R, Dammbrueck C, Pendl T, et al. Dietary spermidine improves cognitive function. Cell Rep. 2021;35: 108985.PubMedCrossRef
354.
Zurück zum Zitat Xu T-T, Li H, Dai Z, Lau GK, Li B-Y, Zhu W-L, et al. Spermidine and spermine delay brain aging by inducing autophagy in SAMP8 mice. Aging (Albany NY). 2020;12:6401–14.CrossRef Xu T-T, Li H, Dai Z, Lau GK, Li B-Y, Zhu W-L, et al. Spermidine and spermine delay brain aging by inducing autophagy in SAMP8 mice. Aging (Albany NY). 2020;12:6401–14.CrossRef
355.
Zurück zum Zitat Freitag K, Sterczyk N, Schulz J, Houtman J, Fleck L, Sigrist SJ, et al. The autophagy activator Spermidine ameliorates Alzheimer’s disease pathology and neuroinflammation in mice. bioRxiv. Cold Spring Harbor Laboratory; 2020;2020.12.27.424477. Freitag K, Sterczyk N, Schulz J, Houtman J, Fleck L, Sigrist SJ, et al. The autophagy activator Spermidine ameliorates Alzheimer’s disease pathology and neuroinflammation in mice. bioRxiv. Cold Spring Harbor Laboratory; 2020;2020.12.27.424477.
356.
Zurück zum Zitat Maglione M, Kochlamazashvili G, Eisenberg T, Rácz B, Michael E, Toppe D, et al. Spermidine protects from age-related synaptic alterations at hippocampal mossy fiber-CA3 synapses. Sci Rep. Nature Publishing Group; 2019;9:19616. Maglione M, Kochlamazashvili G, Eisenberg T, Rácz B, Michael E, Toppe D, et al. Spermidine protects from age-related synaptic alterations at hippocampal mossy fiber-CA3 synapses. Sci Rep. Nature Publishing Group; 2019;9:19616.
357.
Zurück zum Zitat Schwarz C, Stekovic S, Wirth M, Benson G, Royer P, Sigrist SJ, et al. Safety and tolerability of spermidine supplementation in mice and older adults with subjective cognitive decline. Aging (Albany NY). 2018;10:19–33.CrossRef Schwarz C, Stekovic S, Wirth M, Benson G, Royer P, Sigrist SJ, et al. Safety and tolerability of spermidine supplementation in mice and older adults with subjective cognitive decline. Aging (Albany NY). 2018;10:19–33.CrossRef
358.
Zurück zum Zitat Wirth M, Benson G, Schwarz C, Köbe T, Grittner U, Schmitz D, et al. The effect of spermidine on memory performance in older adults at risk for dementia: a randomized controlled trial. Cortex. 2018;109:181–8.PubMedCrossRef Wirth M, Benson G, Schwarz C, Köbe T, Grittner U, Schmitz D, et al. The effect of spermidine on memory performance in older adults at risk for dementia: a randomized controlled trial. Cortex. 2018;109:181–8.PubMedCrossRef
359.
Zurück zum Zitat Wirth M, Schwarz C, Benson G, Horn N, Buchert R, Lange C, et al. Effects of spermidine supplementation on cognition and biomarkers in older adults with subjective cognitive decline (SmartAge)-study protocol for a randomized controlled trial. Alzheimers Res Ther. 2019;11:36.PubMedPubMedCentralCrossRef Wirth M, Schwarz C, Benson G, Horn N, Buchert R, Lange C, et al. Effects of spermidine supplementation on cognition and biomarkers in older adults with subjective cognitive decline (SmartAge)-study protocol for a randomized controlled trial. Alzheimers Res Ther. 2019;11:36.PubMedPubMedCentralCrossRef
360.
Zurück zum Zitat Pekar T, Bruckner K, Pauschenwein-Frantsich S, Gschaider A, Oppliger M, Willesberger J, et al. The positive effect of spermidine in older adults suffering from dementia: first results of a 3-month trial. Wien Klin Wochenschr. 2021;133:484–91.PubMedCrossRef Pekar T, Bruckner K, Pauschenwein-Frantsich S, Gschaider A, Oppliger M, Willesberger J, et al. The positive effect of spermidine in older adults suffering from dementia: first results of a 3-month trial. Wien Klin Wochenschr. 2021;133:484–91.PubMedCrossRef
361.
Zurück zum Zitat Orr ME, Kotkowski E, Bair-Kelps D, Romo T, Espinoza S, Musi N, et al. Results from a pilot study: the effects of nicotinamide riboside on mild cognitive impairment. Alzheimer’s & Dementia. 2020;16: e044746.CrossRef Orr ME, Kotkowski E, Bair-Kelps D, Romo T, Espinoza S, Musi N, et al. Results from a pilot study: the effects of nicotinamide riboside on mild cognitive impairment. Alzheimer’s & Dementia. 2020;16: e044746.CrossRef
362.
Zurück zum Zitat Parker A, Fonseca S, Carding SR. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes. 2019;11:135–57.PubMedPubMedCentralCrossRef Parker A, Fonseca S, Carding SR. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes. 2019;11:135–57.PubMedPubMedCentralCrossRef
363.
Zurück zum Zitat Allison AC. The possible role of vitamin K deficiency in the pathogenesis of alzheimer’s disease and in augmenting brain damage associated with cardiovascular disease. Med Hypotheses. 2001;57:151–5.PubMedCrossRef Allison AC. The possible role of vitamin K deficiency in the pathogenesis of alzheimer’s disease and in augmenting brain damage associated with cardiovascular disease. Med Hypotheses. 2001;57:151–5.PubMedCrossRef
364.
Zurück zum Zitat McCann A, Jeffery IB, Ouliass B, Ferland G, Fu X, Booth SL, et al. Exploratory analysis of covariation of microbiota-derived vitamin K and cognition in older adults. Am J Clin Nutr. 2019;110:1404–15.PubMedPubMedCentralCrossRef McCann A, Jeffery IB, Ouliass B, Ferland G, Fu X, Booth SL, et al. Exploratory analysis of covariation of microbiota-derived vitamin K and cognition in older adults. Am J Clin Nutr. 2019;110:1404–15.PubMedPubMedCentralCrossRef
365.
Zurück zum Zitat Alisi L, Cao R, De Angelis C, Cafolla A, Caramia F, Cartocci G, et al. The relationships between vitamin K and cognition: a review of current evidence. Front Neurol. 2019;10:239.PubMedPubMedCentralCrossRef Alisi L, Cao R, De Angelis C, Cafolla A, Caramia F, Cartocci G, et al. The relationships between vitamin K and cognition: a review of current evidence. Front Neurol. 2019;10:239.PubMedPubMedCentralCrossRef
366.
Zurück zum Zitat Kiely A, Ferland G, Ouliass B, O’Toole PW, Purtill H, O’Connor EM. Vitamin K status and inflammation are associated with cognition in older Irish adults. Nutr Neurosci. 2020;23:591–9.PubMedCrossRef Kiely A, Ferland G, Ouliass B, O’Toole PW, Purtill H, O’Connor EM. Vitamin K status and inflammation are associated with cognition in older Irish adults. Nutr Neurosci. 2020;23:591–9.PubMedCrossRef
367.
Zurück zum Zitat Tamadon-Nejad S, Ouliass B, Rochford J, Ferland G. vitamin K deficiency induced by warfarin is associated with cognitive and behavioral perturbations, and alterations in brain sphingolipids in rats. Front Aging Neurosci. 2018;10:213.PubMedPubMedCentralCrossRef Tamadon-Nejad S, Ouliass B, Rochford J, Ferland G. vitamin K deficiency induced by warfarin is associated with cognitive and behavioral perturbations, and alterations in brain sphingolipids in rats. Front Aging Neurosci. 2018;10:213.PubMedPubMedCentralCrossRef
368.
Zurück zum Zitat Soutif-Veillon A, Ferland G, Rolland Y, Presse N, Boucher K, Féart C, et al. Increased dietary vitamin K intake is associated with less severe subjective memory complaint among older adults. Maturitas. 2016;93:131–6.PubMedCrossRef Soutif-Veillon A, Ferland G, Rolland Y, Presse N, Boucher K, Féart C, et al. Increased dietary vitamin K intake is associated with less severe subjective memory complaint among older adults. Maturitas. 2016;93:131–6.PubMedCrossRef
369.
Zurück zum Zitat Bray N. The microbiota–gut–brain axis. Nature Research: Nature Publishing Group; 2019. Bray N. The microbiota–gut–brain axis. Nature Research: Nature Publishing Group; 2019.
370.
Zurück zum Zitat Ticinesi A, Nouvenne A, Tana C, Prati B, Meschi T. Gut microbiota and microbiota-related metabolites as possible biomarkers of cognitive aging. Adv Exp Med Biol. 2019;1178:129–54.PubMedCrossRef Ticinesi A, Nouvenne A, Tana C, Prati B, Meschi T. Gut microbiota and microbiota-related metabolites as possible biomarkers of cognitive aging. Adv Exp Med Biol. 2019;1178:129–54.PubMedCrossRef
371.
Zurück zum Zitat Kurbatova N, Garg M, Whiley L, Chekmeneva E, Jiménez B, Gómez-Romero M, et al. Urinary metabolic phenotyping for Alzheimer’s disease. Scientific Reports. Nature Publishing Group; 2020;10:21745. Kurbatova N, Garg M, Whiley L, Chekmeneva E, Jiménez B, Gómez-Romero M, et al. Urinary metabolic phenotyping for Alzheimer’s disease. Scientific Reports. Nature Publishing Group; 2020;10:21745.
372.
Zurück zum Zitat Fiandaca MS, Zhong X, Cheema AK, Orquiza MH, Chidambaram S, Tan MT, et al. Plasma 24-metabolite panel predicts preclinical transition to clinical stages of alzheimer’s disease. Front Neurol. 2015;6:237.PubMedPubMedCentralCrossRef Fiandaca MS, Zhong X, Cheema AK, Orquiza MH, Chidambaram S, Tan MT, et al. Plasma 24-metabolite panel predicts preclinical transition to clinical stages of alzheimer’s disease. Front Neurol. 2015;6:237.PubMedPubMedCentralCrossRef
373.
Zurück zum Zitat Casanova R, Varma S, Simpson B, Kim M, An Y, Saldana S, et al. Blood metabolite markers of preclinical alzheimer’s disease in two longitudinally followed cohorts of older individuals. Alzheimer’s & Dementia. 2016;12:815–22.CrossRef Casanova R, Varma S, Simpson B, Kim M, An Y, Saldana S, et al. Blood metabolite markers of preclinical alzheimer’s disease in two longitudinally followed cohorts of older individuals. Alzheimer’s & Dementia. 2016;12:815–22.CrossRef
374.
Zurück zum Zitat de Leeuw FA, Peeters CFW, Kester MI, Harms AC, Struys EA, Hankemeier T, et al. Blood-based metabolic signatures in alzheimer’s disease. Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring. 2017;8:196–207. de Leeuw FA, Peeters CFW, Kester MI, Harms AC, Struys EA, Hankemeier T, et al. Blood-based metabolic signatures in alzheimer’s disease. Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring. 2017;8:196–207.
Metadaten
Titel
Microbial-derived metabolites as a risk factor of age-related cognitive decline and dementia
verfasst von
Emily Connell
Gwenaelle Le Gall
Matthew G. Pontifex
Saber Sami
John F. Cryan
Gerard Clarke
Michael Müller
David Vauzour
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Molecular Neurodegeneration / Ausgabe 1/2022
Elektronische ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-022-00548-6

Weitere Artikel der Ausgabe 1/2022

Molecular Neurodegeneration 1/2022 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Schwindelursache: Massagepistole lässt Otholiten tanzen

14.05.2024 Benigner Lagerungsschwindel Nachrichten

Wenn jüngere Menschen über ständig rezidivierenden Lagerungsschwindel klagen, könnte eine Massagepistole der Auslöser sein. In JAMA Otolaryngology warnt ein Team vor der Anwendung hochpotenter Geräte im Bereich des Nackens.

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.