Next Article in Journal
Analysis of Expression Pattern of snoRNAs in Human Cells A549 Infected by Influenza A Virus
Next Article in Special Issue
Gut Microbiota and Alzheimer’s Disease: How to Study and Apply Their Relationship
Previous Article in Journal
Episignature Mapping of TRIP12 Provides Functional Insight into Clark–Baraitser Syndrome
Previous Article in Special Issue
The Interplay between Gut Microbiota and Parkinson’s Disease: Implications on Diagnosis and Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

What Are the Key Gut Microbiota Involved in Neurological Diseases? A Systematic Review

1
REVAL Rehabilitation Research Center, Faculty of Rehabilitation Sciences, Hasselt University, 3590 Diepenbeek, Belgium
2
Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(22), 13665; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232213665
Submission received: 31 August 2022 / Revised: 2 November 2022 / Accepted: 3 November 2022 / Published: 8 November 2022
(This article belongs to the Special Issue Gut-Brain Axis of Neurodegenerative Disease)

Abstract

:
There is a growing body of evidence highlighting there are significant changes in the gut microbiota composition and relative abundance in various neurological disorders. We performed a systematic review of the different microbiota altered in a wide range of neurological disorders (Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis, and stroke). Fifty-two studies were included representing 5496 patients. At the genus level, the most frequently involved microbiota are Akkermansia, Faecalibacterium, and Prevotella. The overlap between the pathologies was strongest for MS and PD, sharing eight genera (Akkermansia, Butyricicoccus, Bifidobacterium, Coprococcus, Dorea, Faecalibacterium, Parabacteroides, and Prevotella) and PD and stroke, sharing six genera (Enterococcus, Faecalibacterium, Lactobacillus, Parabacteroides, Prevotella, and Roseburia). The identification signatures overlapping for AD, PD, and MS raise the question of whether these reflect a common etiology or rather common consequence of these diseases. The interpretation is hampered by the low number and low power for AD, ALS, and stroke with ample opportunity for false positive and false negative findings.

1. Introduction

The role and importance of the bidirectional communications between the brain and the gut—also referred to as the brain–gut axis [1]—in the pathogenesis of various central nervous disorders [2] is receiving increasing interest in recent years and is, probably, one of the most promising areas of research [3,4,5]. The gut microbiome is composed of a vast number of microorganisms: about 1000 bacterial species and 7000 bacterial strains have been identified representing a total of 1013–1014 different microorganisms in the gut [6,7]. The gut–brain axis is a bidirectional communication axis involving the intestinal microbiome, the intestinal barrier, intestinal inflammation, and the intestinal/systemic/brain immune systems, among other components [8] (see the different communications and pathways in Figure 1). The gut–brain axis contributes to normal central nervous system function and pathology [9,10].
Modifications of the compositions of the gut microbiota have been identified in numerous pathologies and the different potential mechanisms of action have been described in a significant number of narrative reviews mostly focusing on Alzheimer’s disease (AD) [11,12,13,14,15,16], Parkinson’s disease (PD) [17,18,19,20,21,22,23], and multiple sclerosis (MS) [24,25,26,27]. Changes in the gut microbiota have also been found in stroke [28,29] and amyotrophic lateral sclerosis (ALS) [30,31].
A main field of research seeks to determine whether modification of the gut microbiota can decrease the risk of developing diseases or improve the health of the patients in AD [32], PD [33], MS [34], and ALS [35].
Figure 1. Diagram depicting the communication between the gut and the brain. Multiple pathways, including the autonomic nervous system, enteric nervous system, hypothalamic–pituitary–adrenal (HPA), immune pathways, endocrine pathways, and neural pathways, all have a strong influence on this bidirectional relationship, adapted from [36,37,38].
Figure 1. Diagram depicting the communication between the gut and the brain. Multiple pathways, including the autonomic nervous system, enteric nervous system, hypothalamic–pituitary–adrenal (HPA), immune pathways, endocrine pathways, and neural pathways, all have a strong influence on this bidirectional relationship, adapted from [36,37,38].
Ijms 23 13665 g001
Clinical studies have been fueled by the findings in transgenic animal models for AD and PD, which strongly support the interplay between gut microbiota and AD and PD in the brain and identified the gut microbiome as a key determinant of the risk of dementia and a potential target for preventive interventions. In AD transgenic mice, changes in the gut microbiome have been shown to alter amyloid deposition in the brain [39,40]. PD and other synucleinopathies are characterized by the aggregation of α-synuclein protein (αSyn) overexpression in mice, which results in motor dysfunction. It has been shown that gut microbiota influence motor deficits, microglial activation, and αSyn pathology and appear to be a necessary part in pathogenesis [41]. Antibiotic treatment ameliorates pathophysiology in adult animals, whereas microbial re-colonization promotes it, suggesting that postnatal signaling between the gut and the brain modulates disease [42].
In humans, recent evidence suggests that probiotic treatment can improve cognition of patients with mild cognitive impairment by improving the regulation of gut microbiota [43]. Modification of the gut microbiota can also be obtained by modifying the diet—particularly promising results have been found in various neurological conditions using a ketogenetic diet [44]. Despite a growing body of evidence [45], quantitative evidence about the various microbiota involved in neurological diseases is lacking. Many unanswered questions remain: from a physiopathological perspective, it is important to determine if some gut microbiota are consistently linked to a specific disorder across studies to understand their mechanism of action in a specific disease, while from a research perspective it is crucial to evaluate whether findings are consistent. It is also of interest to determine if the associations and modifications are associated with multiple neurological diseases. This may suggest either a common pathogenesis or a common progression of the disease, i.e., neurological pathology may lead to changes in the gut microbiome or vice-versa. Either pathway may be of interest. A causal pathway may open opportunities for prevention and intervention if the neurological disease leads to changes in the gut microbiome. It is, therefore, important to identify the changes in microbiota in the different pathologies, and to determine the direction of these changes in order to enhance management through personalized treatment and precision medicine [46].
In this review, we summarize the findings of the studies of microbiome association with neurodegenerative disorders including AD, PD, and ALS. We further compare these to the findings of microbiome studies on MS, which is a neuroinflammatory disease, and stroke, which often co-occurs with AD and PD.

2. Methods

2.1. Search Strategy

To review the relation of the microbiome in the gut to neurological disorders in the human population, we performed a literature review that included articles published prior to 1 January 2022 with a combination of terms “gut”, “microbiome”, “stool”, “fecal” and the different pathologies “Alzheimer’s disease”, “Parkinson’s disease”, “Multiple Sclerosis”, “Stroke”, “Amyotrophic lateral sclerosis”. Additional relevant articles were sought through a manual bibliography search. Inclusion criteria were: human research, focus on AD, PD, MS, AMS, and stroke that makes a comparison between patients and healthy controls, focus on gut microbiota quantified from stool samples, and articles published in English in peer-reviewed journals. A flow diagram of study selection is presented in Figure 2.

2.2. Data Extraction

From the included studies the following parameters were extracted: the country where the experiments have been carried out, general characteristics of the patients and controls (e.g., age, BMI, sex ratio), and microbiome analysis method. We then listed the different taxon in which relative abundance has been found statistically significantly different between patients and healthy controls and indicated the direction: relative abundance increased or decreased in the various diseases under study.

2.3. Statistical Analysis

We determined whether findings were consistent across studies of a single disorder based on statistical significance and the direction of association for the studies that showed significant results. If two studies were found to show the same direction of the relationship (e.g., microbiota taxus was found more often or less often in patients in both studies), findings were classified as consistent. As most studies were small, one expects heterogeneity across studies due to random fluctuation. We therefore also considered findings for which the consistency was more than 50% as consistent. We present the phylogenetic trees of the microbiota that have been consistently found in the literature for the different pathologies using the GraPhIAn package in Python. General information about the included studies and baseline data on patients were summarized in graphics. To compare the different microbiota involved in the studied pathologies we used Venn diagrams using the VennDiagram package in R [47].

3. Results

3.1. Review of the Gut Microbiome in PD, AD, MS, Stroke, and ALS

In total, 52 studies were included in the final analysis, representing 3177 patients and 2319 controls; the repartition of the patients within the different pathologies is presented in Supplementary Figure S1. The figure shows that PD is the most frequently studied disorder, comprising 54% of all patients and 44% of all studies. As the gut microbiome is known to depend on the environment and diet, which differ across societies and thus across studies, the distribution of the studies per country is presented in Supplementary Figure S2. It is of note that most of the studies were conducted in China (n = 16, 31%), Northern and Eastern Europe (n = 18, 35% for Germany, Italy, Finland, Luxembourg, and the Netherlands), and in the USA (n = 10, 19%).
The characteristics for for PD patients in Supplementary Table S1, for AD patients in Supplementary Table S2, for MS patients in Supplementary Table S3, for stroke patients in Supplementary Table S4, and in Supplementary Table S5 for ALS patients.
We found a total of 1709 PD patients and 1224 healthy controls [48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70]. At the genus level, 60 gut microbiota were identified in at least one study, 16 (25%) associations to PD were consistent in terms of significance and direction (Akkermansia, Alistipes, Anaerotroncus, Barnesiellaceae, Bifidobacterium, Blautia, Butyricicoccus, Christensenella, Dorea, Enterococcus, Escherichia/Shigella, Eubacterium, Faecalibacterium, Fusinibacter, Klebsiella, Oscilospira, Parabacteroides). The association between Akkermansia and PD is replicated in nine studies, that of Bifidobacterium and Faecalibacterium in seven studies. For seven microbiota (Bacteroides, Coprococcus, Lactobacillus, Parabacteroides, Prevotella, Roseburia, Streptococcus) at least two studies were found in the same direction, while for two (Clostridium, Ruminococcus) there were four studies suggesting opposite direction. A phylogenetic tree representing the results is presented in Figure 3, while the complete results of the included studies are presented in Supplementary Table S6.
For AD, a smaller number of studies were found (n = 7), with each study comprising only a limited number of patients. Due to the small sample size of individual studies, nine gut microbiota were reported to be associated with AD in only a single study. Integrating the data of 258 AD patients and 216 healthy controls [71,72,73,74,75,76,77], we find at the family level consistent findings for the Clostridiaceae, Enterococcaceae, and Lachnospiraceae. However, at the genus level, only one association (6%) was found to be consistent (Escherichia/Shigella). For four (25%) associations (Alistipes, Bacteroides, Bifidobacterium, Blautia) at least two studies were found in the same direction. A phylogenetic tree representing the results is presented in Figure 4, and the complete results are presented in Supplementary Table S7.
We found ten studies on MS, involving a total of 307 MS patients and 311 healthy controls [78,79,80,81,82,83,84,85,86,87]. Despite the small number of patients and controls, there was a relatively high consistency seen at the genus level involving 11 (18%) associations (Actinomyces, Akkermansia, Bifidobacterium, Coprococcus, Dialister, Dorea, Faecalibacterium, Haemophilus, Megasphaera, Paraprevotella, Slackia) and only six (10%) associations in the opposite direction (Butyricicoccus, Clostridium, Gemmiger, Parabacteroides, Phascolarctobacterium, Prevotella) (see Figure 5 and Supplementary Table S8).
Six studies on stroke were included in the review, involving a total of 744 stroke patients and 403 healthy controls [88,89,90,91,92,93]. At the genus level, 42 associations were seen in one study, seven (13%) associations to stroke were consistent in terms of significance and direction (Anaerostipes, Enterococcus, Faecalibacterium, Lachnospira, Lactobacillus, Parabacteroides, Roseburia) (see Figure 6 and Supplementary Table S9).
Six studies on ALS were found, involving a total of 159 ALS patients and 165 healthy controls [94,95,96,97,98,99]. One study did not present statistics but only bar plots to visually represent differences between patients and control so we did not include it in the analysis [100]. Twenty microbiota have been identified in at least one study and only five (20%) were found in two studies. The direction of the association was inconsistent in terms of direction for the phylum Firmicutes and consistent for Class Bacteroidia, Negativicutes, Order Bacteroidales, and Clostridiales. No consistency in association was observed at either the family or the genus level (see Figure 7 and Supplementary Table S10).

3.2. Integrating Data across Neurological Diseases

Figure 8 shows the microbiota that are associated with multiple disorders and the number of studies that found the association of the genus to a disease. Genus Akkermansia is associated with four disorders (AD, PD, MS, and stroke) and Faecalibacterium with three disorders (PD, MS, and stroke). While Prevotella is also found to be associated with four disorders (AD, PD, MS, and stroke), the direction of association is inconsistent across the pathologies and studies.
In Figure 9, we plotted Venn diagrams summarizing the association to multiple neurological diseases. For the microbiota where inconsistencies were observed across studies, we define consistency if more than 50% of the studies observed association in the same direction. Figure 8 shows that based on the data available to date there is no specific signature for AD or ALS, i.e., no genus involved only in AD or ALS. For PD at the genus level there are eight disease-specific microbiome signatures (Anaerotroncus, Barnesiellaceae, Eubacterium, Fusicatenibacter, Klebsiella, Oscillospira, and Streptococcus). For MS we found eight disease-specific signatures (Actinomyces, Clostridium, Coprococcus, Dialister, Haemophilus, Megamonas, Paraprevotella, and Slackia). Overlapping changes in the microbiome were also seen in PD and MS (Akkermansia, Butyricicoccus, Coprococcus, and Dorea); between PD and AD (Alistipes, Bacteroides, Blautia, and Escherichia/Shigella); between PD and stroke (Enterococcus, Lactobacillus, Roseburia); between PD, MS, and stroke (Faecalibacterium, Parabacteroides, and Prevotella); and between PD, MS, and AD (Bifidobacterium).
However, Figure 8 and Figure 9 do not take into account the direction of the association. Figure 10 and Figure 11 show the direction of the associations. At the genus level, Alistipes, Bacteroides, Bifidobacterium, and Escherichia/Shigella are increased in abundance in AD and PD, while the opposite direction is found for Blautia (relative increase in AD but decrease in PD). When comparing AD to MS there is an increase in the relative abundance of Bifidobacterium. For PD and MS, Akkermansia and Bifidobacterium are found to be consistently increased and Faecalibacterium and Prevotella decreased in abundance. Opposite directions are found for Coprococcus (decrease in PD and increase in MS), Dorea (decrease in PD and increase in MS), and Parabacteroides (increase in PD and decrease in MS). For MS and stroke, there is a decrease in the relative abundance of Faecalibacterium and Prevotella. Finally, when comparing MS to stroke we found a similar direction for Faecalibacterium and Prevotella (increase) and an opposite direction for Parabacteroides (decrease in MS).
The picture emerging is that there is an overlap between neurological disorders for Alistipes, Akkermansia, Blautia, Bifidobacterium, Dorea, Escherichia/Shigella, Faecalibacterium, Parabacteroides, and Prevotella.

4. Discussion

The past decade has seen a rapid rise in studies of the role of microbiota in various disorders. In this review, we focus on the consistency of the direction of the relationship found within neurological diseases and between neurologic diseases including PD, AD, ALS, MS, and stroke. The disease with the most consistent microbiome associations is PD. The findings are most reliable as they are based on the largest samples size. Amongst the microbiota associated most frequently, a clear pattern was observed for the genus Akkermansia which showed a consistent increase in nine studies in PD [50,54,55,57,60,61,62,67,69] and four in MS [79,82,83,86]. Akkermansia resides in the mucus layer of the large intestine [101], where they are involved in maintaining intestinal integrity and mucin degradation [102]. Akkermansia uses mucins as a carbon source [103] and can cleave sialic acids [104]. As shown in Figure 2, the increase in the relative abundance of Akkermansia in PD is confirmed at all the taxonomic levels presenting a strong signature of the disease. Such consistency along all the taxonomic levels was not seen for any other microbiota for the other diseases studied. Akkermansia can stimulate dendritic cells to produce TGFβ and interleukin 6 (IL6) and 1 (IL1), activating regulatory T Cells (Tregs) which may be relevant for the pathogenesis of MS as well as for PD [105]. Changes in the gut microbiota have been linked with an increase in inflammation in the central nervous system in various neurological diseases. Our first line of defense is the innate immune system, which consists of physical and chemical barriers, immune cells, and blood proteins (such as cytokines). Toll-like receptors (TLRs) located on the membranes of epithelial and lymphoid cells in the small intestine mediate this differential recognition of commensal and pathogenic bacteria [106]. The question to answer is how to interpret the change in Akkermansia seen in both disorders. MS is primarily a neuroinflammatory disorder and there is increasing interest in the role of neuroinflammation in PD as a therapeutic target [107,108,109]. A late complication of MS and PD is cognitive decline and there is increasing evidence that neuroinflammation is involved in dementia and cognitive decline. In this study, the evidence for the role of Akkermansia in dementia was not consistent. However, a problem interpreting the findings of the AD studies is that the power of these studies has been low and false negative findings may be frequent. Akkermansia have been associated with diabetes and cardiovascular disease, co-morbidity of which are often seen in neurodegenerative disorders. Recently, Akkermansia intervention has been approved by the European Food Safe authority [110]. Last but not least, the abundance of Akkermansia has also been associated with constipation, which is often seen in PD patients and is a major clinical problem [111]. Of note is that constipation is very common in MS patients and a hypothesis to be tested is whether this problem in MS is related to the increased abundance of Akkermansia in the gut.
The overlap with PD and MS is substantial with eight microbiota found in common: Akkermansia, Bifidobacterium, Butyricicoccus, Coprococcus, Dorea, Faecalibacterium, Parabacteroides, and Prevotella. Dorea is abundant in MS but seen to be reduced in PD. The finding of the increased relative abundance of Bifidobacterium across PD and MS studies is far from understood. The genus Bifidobacterium is known to metabolize lactose. The genus Bifidobacterium has previously been implicated in lactose tolerance [112]. Bifidobacterium was the first microbial to colonize humans and is seen as positive for health [113]. For PD, one may speculate that the relatively healthy lifestyle in some patients may partially explain these findings. PD patients have been found to smoke less [114]. The effects of Bifidobacterium and Lactobacillus species (also involved in PD) delivered as probiotics have been previously studied in PD [115]. The results indicated a decrease in negative mood in healthy persons [116]. Psychobiotics are probiotics believed to promote mental wellness through interaction with other commensal gut microbes [117]. In mice, probiotic administration leads to a drop in proinflammatory cytokines and an increase in anti-inflammatory cytokines, resulting in cognitive and emotional enhancement [118]. Furthermore, Dorea is constituent of healthy gut flora and has also been linked to irritable bowel syndrome [119].
The increased abundance of Enterococcus (consistent in all PD samples) and Lactobacillus (abundant in PD patients in most of the included studies) is of interest as these taxa may interfere with Levodopa medication of the patients [120] and thus impact the quality of life [121]. This genus is also implicated in tyrosine metabolism, which is explored as a new intervention in stroke [122]. Interestingly, experimental studies in mice show evidence that the inclusion of Akkermansia and Parabacteroides in the diet of mice confers seizure protection compared to a control diet [123].
Bacteroides was found to be altered in AD and PD. The bacteria are implicated in adaptive immunity, which is mediated by B cells and T cells, two primary lymphocyte populations. Through its polysaccharide molecules, Bacteroides fragilis induces a systemic Th1 response, which is essential for eradicating intracellular infections. On the other hand, segmented filamentous bacteria have been shown to be effective inducers of Th17 cells, and Clostridia have been found to increase the production of colonic Tregs, a crucial mediator of immunological tolerance whose dysfunction can lead to autoimmune diseases. [124]. They can affect the nervous system, contributing to the maturation of naive microglia in the absence of microbiota, which could be explained by the modifications observed in PD and AD.
AD and PD are the two most common neurodegenerative diseases in the elderly. Of note is that each of the microbiota that are consistently associated to AD are also consistently associated to PD. At the genus level, for AD and PD, changes of Alistipes, Bacteroides, Bifidobacterium, and Escherichia/Shigella are found in the same direction (an increase of relative abundance) while the opposite direction is found for Blautia (relative increase in AD but decrease in PD). Alistipes and Bacteroides have been implicated in cholesterol homeostasis. Escherichia coli has been implicated in amyloid (AD) and alpha-synuclein (PD) metabolism as well as gliosis (seen both in AD and PD [104]).
In addition to the pathways discussed above, there are several possible explanations for the mechanisms of action driving the relationship between gut microbiota and the different diseases. A recent review underlines the relationship between inflammation, pain, microbiota, and the different lipids, focusing on the possible involvement of the N-acylethanolamine family and short-chain fatty acids in the gut–brain axis and their role in the central nervous system diseases [125]. The lymphatic system could be the mediator of this communication between the gut microbiota and the brain [126].
In this review, we summarized the current evidence of the modification of gut microbiota in various pathologies. We limited our review to the main neurodegenerative diseases and MS but gut microbiota composition is also modified in other neurological disorders such as Rett syndrome [127] and in neurocritical ill patients [128]. However, their roles are not limited to neurological or neuropsychiatric diseases; they are also involved in hypertension [129,130], cirrhosis [131,132] or primary hepatocellular carcinoma [133], diabetes [134], autoimmune diseases [135], systemic lupus erythematosus [136], systemic immunity in allergic disease [137], Behcet’s disease [138], systemic sclerosis [139], rheumatoid arthritis [140], and could also potentially influence vitamin D production [141]. There are also correlations reported between some gut microbiota and personality in adults [142].
An essential part of the research in this field is the development of interventions to modify the microbiota. The gut microbiome is determined by diet [143,144]. For example, intermittent fasting led to increased gut bacteria richness, enrichment of the Lactobacillaceae, Bacteroidaceae, and Prevotellaceae families, and enhanced antioxidative microbial metabolic pathways [145]. The most promising results of clinical improvement after modification of the gut microbiota have been obtained in patients with epilepsy after a ketogenic diet [146,147,148]. There was an overall decrease in the mean species diversity after treatment and, importantly, a difference in the variation of species between responders and non-responders. Further analysis of species composition before and after treatment showed a significant increase in Bacteroides and a decrease in Firmicutes and Actinobacteria. When comparing responders and non-responders, Clostridiales, Clostridia, Ruminococcaeceae, Lachnospiraceae, Alistipes, and Tikenellacase were significantly increased in non-responders [149]. Interesting clinical results have also been obtained using fecal transplantation (FMT). FMT is the technique of administering feces from healthy donors to potential recipients in attempt to restore a stable microbiota in the stomach [150]. FMT has been shown to be both safe and effective in the treatment of recurrent Clostridium difficile infection [151]. This has prompted research into the impact of FMT on various illnesses brought on or aggravated by gut dysbiosis. Promising results have been found in both animal and human studies [152,153].
Remarkably, FMT reduced gut microbial dysbiosis, decreased fecal SCFAs, alleviated physical impairment, and increased striatal DA and 5-HT content of PD mice. Further, FMT reduced the activation of microglia and astrocytes in the substantia nigra, and reduced expression of TLR4/TNF-α signaling pathway components in gut and brain [152,154].
Another promising domain of research is the modification of gut microbiota composition using probiotics. Probiotics are microorganisms that are administered in the form of a medicine, food, dietary supplement, or infant formula [155]. The majority of a normal probiotic consists of gut bacteria that occur naturally in the human body. They may provide health benefits to the host when administered in sufficient doses and frequency. Probiotic administration is currently under development and validation in various disorders such as PD [156], AD [157], MS [158], and stroke [159].
A lot of research, mainly performed in AD and PD, has also been carried out using transgenic animals (mice) to try to better understand if the changes in abundance in microbiota may be a cause or consequence of pathogenesis as the communication between the microbiome in the gastrointestinal tract and the central nervous system is bi-directional [160]. In AD, different models have been tested: amyloid metabolism (APP/PS1) [39,161,162,163,164,165,166], senescence models (SAMP8) [40,167], and triple mutations (APP, MAPT, PSEN1) [168,169,170]. Of note is that there was no difference between the amyloid models. Studies using animal models also provided new potentially exciting opportunities for the prevention or management of these disorders. In APPswe/PS1dE9 transgenic mice with fecal microbiota transplantation, treatment can improve cognitive deficits and reduce the brain deposition of Aβ. As for translational studies in humans, other approaches have recently been successfully tested in mice to modify the gut microbiota of transgenic animals, such as oral administration of anti-inflammatory Bifidobacterium longum (NK46) from human gut microbiota. NK46 treatment suppressed amyloid-β, β/γ-secretases, and caspase-3 expression and amyloid-β accumulation in the hippocampus of 5XFAD-Tg mice. Suppression of gut dysbiosis by NK46 can mitigate cognitive decline through the regulation of microbiota [171]. Silibinin and silymarin administration tended to decrease the microbiota diversity and exhibited a regulative effect in abundance on several key bacterial species associated with AD development in APP/PS1 mice [172]. In PD, most of the studies have been performed with ASO mice (α-synuclein overexpressing) to stimulate the production of αSyn in the central nervous system [173] and with Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) [174]. From a therapeutical perspective, the protective effect of Ceftriaxone has been shown in MPTP intoxicated mouse model. Ceftriaxone had a neuroprotective effect on MPTP-induced PD mice, and its neuroprotective effect could be through regulating inflammation and intestinal microbiota [175]. Other approaches such as curcumin [176], Oligosaccharides [177], chicoric acid [178], and Mucuna pruriens [179] also show promising results.
The main limitation of the current studies are the populations investigated. We have seen that a majority of the studies have been done in China and in the USA but if we analyzed the results by pathologies, we observed that 7 of the 10 studies in MS have been done in the USA and Canada, and 5 out of the 6 studies on stroke in Asia (4 in China, 1 in Japan). It has also been shown that, even within the USA, the gut microbiome composition in PD variated in different states [180]. This high homogeneity generalizes the results for these pathologies, since it has been shown that genetics [181,182], socioeconomic status [183], and diet influence the composition of the microbiota [184,185,186,187].
We observed a major difference in the mean age of the participants in the different pathologies and adjustment for confounding, such as body mass index (BMI), medication, constipation [71,73,77], and/or diet and alcohol consumption [188,189], while it has been demonstrated that those factors influence microbiota composition and abundance [167,190,191,192]. There are also important differences in the types of the different studied diseases. For AD, for example, some studies include patients with very mild dementia and moderate dementia [71]; in one study, the authors compared AD patients with and without amyloidosis [72]. In PD, most of the studies are performed with sporadic PD but also in familial PD, while the genetic factors differ and, therefore, the potential relationship with the brain–gut axis [193]. For studies in MS, there is a mix of patients with relapsing-remitting MS and progressive MS. Last but not least, there are major differences in the severity and in the duration of the disease, two elements that have shown an important role in the alterations of the microbiome in the case of degenerative pathologies. The interpretation is hampered by the low sample sizes of the AD, ALS, and stroke studies leading to false positive and false negative findings.

5. Conclusions

Summarizing the evidence for a role of the gut microbiome in and across neurological diseases, we find that the findings are most consistent for PD and MS. We further find that the findings are often not unique to neurological diseases. There is a substantial overlap between PD and MS and AD and PD that needs further investigation about whether this reflects a common etiology of those disorders and shared consequence of neurological pathology. The most remarkable microbiota signature was observed in PD with an important increase in the relative abundance of the Akkermansia at all the different levels of the taxon. Additionally, for MS, findings were often consistent over studies, despite the small series of patients studied. The most similar signature, from the perspectives of microbiome composition, are PD and MS. For future clinical applications, baseline value and standard reporting templates have been developed that should ease and spread the analysis of gut microbiota in daily practice for clinicians [194,195,196]. Despite the limitations of the studies conducted to date, aggregating the data of the current studies strongly suggests that the microbiome is a promising target for preventive and/or restorative treatment. The findings beg for more research aiming to answer the question of whether targeted anti or probiotic interventions may be relevant for the prevention of neurological disorders, or may improve the quality of life of patients.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/ijms232213665/s1.

Author Contributions

The study was conceived by B.B. and C.v.D., N.A. and C.v.D. verified the analytical methods. B.B., N.A. and C.v.D. did the data interpretation. C.v.D. supervised the findings of this work. All authors discussed the results and contributed to the final manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Stilling, R.M.; Dinan, T.G.; Cryan, J.F. Microbial Genes, Brain & Behaviour—Epigenetic Regulation of the Gut-Brain Axis. Genes Brain Behav. 2014, 13, 69–86. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, Y.; Kasper, L.H. The Role of Microbiome in Central Nervous System Disorders. Brain Behav. Immun. 2014, 38, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Seguella, L.; Capuano, R.; Sarnelli, G.; Esposito, G. Play in Advance against Neurodegeneration: Exploring Enteric Glial Cells in Gut-Brain Axis during Neurodegenerative Diseases. Expert Rev. Clin. Pharmacol. 2019, 12, 555–564. [Google Scholar] [CrossRef] [PubMed]
  4. Sasmita, A.O. Modification of the Gut Microbiome to Combat Neurodegeneration. Rev. Neurosci. 2019, 30, 795–805. [Google Scholar] [CrossRef] [PubMed]
  5. Heiss, C.N.; Olofsson, L.E. The Role of the Gut Microbiota in Development, Function and Disorders of the Central Nervous System and the Enteric Nervous System. J. Neuroendocrinol. 2019, 31, e12684. [Google Scholar] [CrossRef]
  6. Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T.; Creasy, H.H.; Earl, A.M.; FitzGerald, M.G.; Fulton, R.S.; et al. Human Microbiome Project Consortium Structure, Function and Diversity of the Healthy Human Microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef] [Green Version]
  7. Westfall, S.; Lomis, N.; Kahouli, I.; Dia, S.Y.; Singh, S.P.; Prakash, S. Microbiome, Probiotics and Neurodegenerative Diseases: Deciphering the Gut Brain Axis. Cell. Mol. Life Sci. 2017, 74, 3769–3787. [Google Scholar] [CrossRef]
  8. Abdel-Haq, R.; Schlachetzki, J.C.M.; Glass, C.K.; Mazmanian, S.K. Microbiome–Microglia Connections via the Gut–Brain Axis. J. Exp. Med. 2019, 216, 41–59. [Google Scholar] [CrossRef] [Green Version]
  9. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  10. Socała, K.; Doboszewska, U.; Szopa, A.; Serefko, A.; Włodarczyk, M.; Zielińska, A.; Poleszak, E.; Fichna, J.; Wlaź, P. The Role of Microbiota-Gut-Brain Axis in Neuropsychiatric and Neurological Disorders. Pharmacol. Res. 2021, 172, 105840. [Google Scholar] [CrossRef]
  11. Kowalski, K.; Mulak, A. Brain-Gut-Microbiota Axis in Alzheimer’s Disease. J. Neurogastroenterol. Motil. 2019, 25, 48–60. [Google Scholar] [CrossRef] [Green Version]
  12. Angelucci, F.; Cechova, K.; Amlerova, J.; Hort, J. Antibiotics, Gut Microbiota, and Alzheimer’s Disease. J. Neuroinflamm. 2019, 16, 108. [Google Scholar] [CrossRef] [Green Version]
  13. Bostanciklioğlu, M. The Role of Gut Microbiota in Pathogenesis of Alzheimer’s Disease. J. Appl. Microbiol. 2019, 127, 954–967. [Google Scholar] [CrossRef]
  14. Sochocka, M.; Donskow-Łysoniewska, K.; Diniz, B.S.; 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, 1841–1851. [Google Scholar] [CrossRef] [Green Version]
  15. Doifode, T.; Giridharan, V.V.; Generoso, J.S.; Bhatti, G.; Collodel, A.; Schulz, P.E.; Forlenza, O.V.; Barichello, T. The Impact of the Microbiota-Gut-Brain Axis on Alzheimer’s Disease Pathophysiology. Pharmacol. Res. 2021, 164, 105314. [Google Scholar] [CrossRef]
  16. 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, 5026–5043. [Google Scholar] [CrossRef]
  17. Chapelet, G.; Leclair-Visonneau, L.; Clairembault, T.; Neunlist, M.; Derkinderen, P. Can the Gut Be the Missing Piece in Uncovering PD Pathogenesis? Parkinsonism Relat. Disord. 2019, 59, 26–31. [Google Scholar] [CrossRef]
  18. Santos, S.F.; de Oliveira, H.L.; Yamada, E.S.; Neves, B.C.; Pereira, A. The Gut and Parkinson’s Disease-A Bidirectional Pathway. Front. Neurol. 2019, 10, 574. [Google Scholar] [CrossRef] [Green Version]
  19. Chiang, H.-L.; Lin, C.-H. Altered Gut Microbiome and Intestinal Pathology in Parkinson’s Disease. J. Mov. Disord. 2019, 12, 67–83. [Google Scholar] [CrossRef]
  20. Sharma, S.; Awasthi, A.; Singh, S. Altered Gut Microbiota and Intestinal Permeability in Parkinson’s Disease: Pathological Highlight to Management. Neurosci. Lett. 2019, 712, 134516. [Google Scholar] [CrossRef]
  21. Miraglia, F.; Colla, E. Microbiome, Parkinson’s Disease and Molecular Mimicry. Cells 2019, 8, 222. [Google Scholar] [CrossRef] [Green Version]
  22. Nowak, J.M.; Kopczyński, M.; Friedman, A.; Koziorowski, D.; Figura, M. Microbiota Dysbiosis in Parkinson Disease-In Search of a Biomarker. Biomedicines 2022, 10, 2057. [Google Scholar] [CrossRef] [PubMed]
  23. Grant, H.; Anderton, R.; Gasson, N.; Lawrence, B.J. The Gut Microbiome and Cognition in Parkinson’s Disease: A Systematic Review. Nutr. Neurosci. 2022, 2022, 1–10. [Google Scholar] [CrossRef] [PubMed]
  24. Sato, W.; Yamamura, T. Multiple Sclerosis: Possibility of a Gut Environment-Induced Disease. Neurochem. Int. 2019, 130, 104475. [Google Scholar] [CrossRef] [PubMed]
  25. Buscarinu, M.C.; Fornasiero, A.; Romano, S.; Ferraldeschi, M.; Mechelli, R.; Reniè, R.; Morena, E.; Romano, C.; Pellicciari, G.; Landi, A.C.; et al. The Contribution of Gut Barrier Changes to Multiple Sclerosis Pathophysiology. Front. Immunol. 2019, 10, 1916. [Google Scholar] [CrossRef]
  26. Ghadiri, F.; Ebadi, Z.; Asadollahzadeh, E.; Naser Moghadasi, A. Gut Microbiome in Multiple Sclerosis-Related Cognitive Impairment. Mult. Scler. Relat. Disord. 2022, 67, 104165. [Google Scholar] [CrossRef]
  27. Correale, J.; Hohlfeld, R.; Baranzini, S.E. The Role of the Gut Microbiota in Multiple Sclerosis. Nat. Rev. Neurol. 2022, 18, 544–558. [Google Scholar] [CrossRef]
  28. Bao, Z.; Zhang, Z.; Zhou, G.; Zhang, A.; Shao, A.; Zhou, F. Novel Mechanisms and Therapeutic Targets for Ischemic Stroke: A Focus on Gut Microbiota. Front. Cell. Neurosci. 2022, 16, 871720. [Google Scholar] [CrossRef]
  29. Wang, J.; Zhang, H.; He, J.; Xiong, X. The Role of the Gut Microbiota in the Development of Ischemic Stroke. Front. Immunol. 2022, 13, 845243. [Google Scholar] [CrossRef]
  30. Boddy, S.L.; Giovannelli, I.; Sassani, M.; Cooper-Knock, J.; Snyder, M.P.; Segal, E.; Elinav, E.; Barker, L.A.; Shaw, P.J.; McDermott, C.J. The Gut Microbiome: A Key Player in the Complexity of Amyotrophic Lateral Sclerosis (ALS). BMC Med. 2021, 19, 13. [Google Scholar] [CrossRef]
  31. Martin, S.; Battistini, C.; Sun, J. A Gut Feeling in Amyotrophic Lateral Sclerosis: Microbiome of Mice and Men. Front. Cell. Infect. Microbiol. 2022, 12, 839526. [Google Scholar] [CrossRef]
  32. Mancuso, C.; Santangelo, R. Alzheimer’s Disease and Gut Microbiota Modifications: The Long Way between Preclinical Studies and Clinical Evidence. Pharmacol. Res. 2018, 129, 329–336. [Google Scholar] [CrossRef]
  33. Sun, M.-F.; Shen, Y.-Q. Dysbiosis of Gut Microbiota and Microbial Metabolites in Parkinson’s Disease. Ageing Res. Rev. 2018, 45, 53–61. [Google Scholar] [CrossRef]
  34. Mirza, A.; Mao-Draayer, Y. The Gut Microbiome and Microbial Translocation in Multiple Sclerosis. Clin. Immunol. 2017, 183, 213–224. [Google Scholar] [CrossRef]
  35. Zhang, Y.-G.; Wu, S.; Yi, J.; Xia, Y.; Jin, D.; Zhou, J.; Sun, J. Target Intestinal Microbiota to Alleviate Disease Progression in Amyotrophic Lateral Sclerosis. Clin. Ther. 2017, 39, 322–336. [Google Scholar] [CrossRef] [Green Version]
  36. Morais, L.H.; Schreiber, H.L.; Mazmanian, S.K. The Gut Microbiota-Brain Axis in Behaviour and Brain Disorders. Nat. Rev. Microbiol. 2021, 19, 241–255. [Google Scholar] [CrossRef]
  37. Suganya, K.; Koo, B.-S. Gut–Brain Axis: Role of Gut Microbiota on Neurological Disorders and How Probiotics/Prebiotics Beneficially Modulate Microbial and Immune Pathways to Improve Brain Functions. Int. J. Mol. Sci. 2020, 21, 7551. [Google Scholar] [CrossRef]
  38. Collins, S.M.; Surette, M.; Bercik, P. The Interplay between the Intestinal Microbiota and the Brain. Nat. Rev. Microbiol. 2012, 10, 735–742. [Google Scholar] [CrossRef]
  39. Xin, Y.; Diling, C.; Jian, Y.; Ting, L.; Guoyan, H.; Hualun, L.; Xiaocui, T.; Guoxiao, L.; Ou, S.; Chaoqun, Z.; et al. Effects of Oligosaccharides from Morinda Officinalis on Gut Microbiota and Metabolome of APP/PS1 Transgenic Mice. Front. Neurol. 2018, 9, 412. [Google Scholar] [CrossRef]
  40. Peng, W.; Yi, P.; Yang, J.; Xu, P.; Wang, Y.; Zhang, Z.; Huang, S.; Wang, Z.; Zhang, C. Association of Gut Microbiota Composition and Function with a Senescence-Accelerated Mouse Model of Alzheimer’s Disease Using 16S RRNA Gene and Metagenomic Sequencing Analysis. Aging 2018, 10, 4054–4065. [Google Scholar] [CrossRef]
  41. Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480.e12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Dutta, S.K.; Verma, S.; Jain, V.; Surapaneni, B.K.; Vinayek, R.; Phillips, L.; Nair, P.P. Parkinson’s Disease: The Emerging Role of Gut Dysbiosis, Antibiotics, Probiotics, and Fecal Microbiota Transplantation. J. Neurogastroenterol. Motil. 2019, 25, 363–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. De Lara-Sánchez, S.S.; Sánchez-Pérez, A.M. Probiotics Treatment Can Improve Cognition in Patients with Mild Cognitive Impairment: A Systematic Review. J. Alzheimers Dis. 2022, 89, 1–19. [Google Scholar] [CrossRef] [PubMed]
  44. Lim, J.-M.; Letchumanan, V.; Tan, L.T.-H.; Hong, K.-W.; Wong, S.-H.; Ab Mutalib, N.-S.; Lee, L.-H.; Law, J.W.-F. Ketogenic Diet: A Dietary Intervention via Gut Microbiome Modulation for the Treatment of Neurological and Nutritional Disorders (a Narrative Review). Nutrients 2022, 14, 3566. [Google Scholar] [CrossRef] [PubMed]
  45. Vamanu, E.; Rai, S.N. The Link between Obesity, Microbiota Dysbiosis, and Neurodegenerative Pathogenesis. Diseases 2021, 9, 45. [Google Scholar] [CrossRef]
  46. Feng, W.; Liu, J.; Ao, H.; Yue, S.; Peng, C. Targeting Gut Microbiota for Precision Medicine: Focusing on the Efficacy and Toxicity of Drugs. Theranostics 2020, 10, 11278–11301. [Google Scholar] [CrossRef]
  47. Chen, H.; Boutros, P.C. VennDiagram: A Package for the Generation of Highly-Customizable Venn and Euler Diagrams in R. BMC Bioinform. 2011, 12, 35. [Google Scholar] [CrossRef] [Green Version]
  48. Scheperjans, F.; Aho, V.; Pereira, P.A.B.; Koskinen, K.; Paulin, L.; Pekkonen, E.; Haapaniemi, E.; Kaakkola, S.; Eerola-Rautio, J.; Pohja, M.; et al. Gut Microbiota Are Related to Parkinson’s Disease and Clinical Phenotype. Mov. Disord. 2015, 30, 350–358. [Google Scholar] [CrossRef]
  49. Hasegawa, S.; Goto, S.; Tsuji, H.; Okuno, T.; Asahara, T.; Nomoto, K.; Shibata, A.; Fujisawa, Y.; Minato, T.; Okamoto, A.; et al. Intestinal Dysbiosis and Lowered Serum Lipopolysaccharide-Binding Protein in Parkinson’s Disease. PLoS ONE 2015, 10, e0142164. [Google Scholar] [CrossRef] [Green Version]
  50. Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic Bacterial Composition in Parkinson’s Disease. Mov. Disord. 2015, 30, 1351–1360. [Google Scholar] [CrossRef]
  51. Unger, M.M.; Spiegel, J.; Dillmann, K.-U.; Grundmann, D.; Philippeit, H.; Bürmann, J.; Faßbender, K.; Schwiertz, A.; Schäfer, K.-H. Short Chain Fatty Acids and Gut Microbiota Differ between Patients with Parkinson’s Disease and Age-Matched Controls. Parkinsonism Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
  52. Petrov, V.A.; Saltykova, I.V.; Zhukova, I.A.; Alifirova, V.M.; Zhukova, N.G.; Dorofeeva, Y.B.; Tyakht, A.V.; Kovarsky, B.A.; Alekseev, D.G.; Kostryukova, E.S.; et al. Analysis of Gut Microbiota in Patients with Parkinson’s Disease. Bull. Exp. Biol. Med. 2017, 162, 734–737. [Google Scholar] [CrossRef]
  53. Li, W.; Wu, X.; Hu, X.; Wang, T.; Liang, S.; Duan, Y.; Jin, F.; Qin, B. Structural Changes of Gut Microbiota in Parkinson’s Disease and Its Correlation with Clinical Features. Sci. China Life Sci. 2017, 60, 1223–1233. [Google Scholar] [CrossRef]
  54. Hill-Burns, E.M.; Debelius, J.W.; Morton, J.T.; Wissemann, W.T.; Lewis, M.R.; Wallen, Z.D.; Peddada, S.D.; Factor, S.A.; Molho, E.; Zabetian, C.P.; et al. Parkinson’s Disease and Parkinson’s Disease Medications Have Distinct Signatures of the Gut Microbiome. Mov. Disord. 2017, 32, 739–749. [Google Scholar] [CrossRef] [Green Version]
  55. Bedarf, J.R.; Hildebrand, F.; Coelho, L.P.; Sunagawa, S.; Bahram, M.; Goeser, F.; Bork, P.; Wüllner, U. Functional Implications of Microbial and Viral Gut Metagenome Changes in Early Stage L-DOPA-Naïve Parkinson’s Disease Patients. Genome Med. 2017, 9, 39. [Google Scholar] [CrossRef] [Green Version]
  56. Hopfner, F.; Künstner, A.; Müller, S.H.; Künzel, S.; Zeuner, K.E.; Margraf, N.G.; Deuschl, G.; Baines, J.F.; Kuhlenbäumer, G. Gut Microbiota in Parkinson Disease in a Northern German Cohort. Brain Res. 2017, 1667, 41–45. [Google Scholar] [CrossRef]
  57. Heintz-Buschart, A.; Pandey, U.; Wicke, T.; Sixel-Döring, F.; Janzen, A.; Sittig-Wiegand, E.; Trenkwalder, C.; Oertel, W.H.; Mollenhauer, B.; Wilmes, P. The Nasal and Gut Microbiome in Parkinson’s Disease and Idiopathic Rapid Eye Movement Sleep Behavior Disorder. Mov. Disord. 2018, 33, 88–98. [Google Scholar] [CrossRef] [Green Version]
  58. Qian, Y.; Yang, X.; Xu, S.; Wu, C.; Song, Y.; Qin, N.; Chen, S.-D.; Xiao, Q. Alteration of the Fecal Microbiota in Chinese Patients with Parkinson’s Disease. Brain Behav. Immun. 2018, 70, 194–202. [Google Scholar] [CrossRef]
  59. Lin, A.; Zheng, W.; He, Y.; Tang, W.; Wei, X.; He, R.; Huang, W.; Su, Y.; Huang, Y.; Zhou, H.; et al. Gut Microbiota in Patients with Parkinson’s Disease in Southern China. Parkinsonism Relat. Disord. 2018, 53, 82–88. [Google Scholar] [CrossRef]
  60. Barichella, M.; Severgnini, M.; Cilia, R.; Cassani, E.; Bolliri, C.; Caronni, S.; Ferri, V.; Cancello, R.; Ceccarani, C.; Faierman, S.; et al. Unraveling Gut Microbiota in Parkinson’s Disease and Atypical Parkinsonism. Mov. Disord. 2019, 34, 396–405. [Google Scholar] [CrossRef]
  61. Li, C.; Cui, L.; Yang, Y.; Miao, J.; Zhao, X.; Zhang, J.; Cui, G.; Zhang, Y. Gut Microbiota Differs Between Parkinson’s Disease Patients and Healthy Controls in Northeast China. Front. Mol. Neurosci. 2019, 12, 171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Li, F.; Wang, P.; Chen, Z.; Sui, X.; Xie, X.; Zhang, J. Alteration of the Fecal Microbiota in North-Eastern Han Chinese Population with Sporadic Parkinson’s Disease. Neurosci. Lett. 2019, 707, 134297. [Google Scholar] [CrossRef] [PubMed]
  63. Aho, V.T.E.; Pereira, P.A.B.; Voutilainen, S.; Paulin, L.; Pekkonen, E.; Auvinen, P.; Scheperjans, F. Gut Microbiota in Parkinson’s Disease: Temporal Stability and Relations to Disease Progression. EBioMedicine 2019, 44, 691–707. [Google Scholar] [CrossRef] [Green Version]
  64. Weis, S.; Schwiertz, A.; Unger, M.M.; Becker, A.; Faßbender, K.; Ratering, S.; Kohl, M.; Schnell, S.; Schäfer, K.-H.; Egert, M. Effect of Parkinson’s Disease and Related Medications on the Composition of the Fecal Bacterial Microbiota. npj Parkinson’s Dis. 2019, 5, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Pietrucci, D.; Cerroni, R.; Unida, V.; Farcomeni, A.; Pierantozzi, M.; Mercuri, N.B.; Biocca, S.; Stefani, A.; Desideri, A. Dysbiosis of Gut Microbiota in a Selected Population of Parkinson’s Patients. Parkinsonism Relat. Disord. 2019, 65, 124–130. [Google Scholar] [CrossRef]
  66. Ren, T.; Gao, Y.; Qiu, Y.; Jiang, S.; Zhang, Q.; Zhang, J.; Wang, L.; Zhang, Y.; Wang, L.; Nie, K. Gut Microbiota Altered in Mild Cognitive Impairment Compared with Normal Cognition in Sporadic Parkinson’s Disease. Front. Neurol. 2020, 11, 137. [Google Scholar] [CrossRef]
  67. Vascellari, S.; Palmas, V.; Melis, M.; Pisanu, S.; Cusano, R.; Uva, P.; Perra, D.; Madau, V.; Sarchioto, M.; Oppo, V.; et al. Gut Microbiota and Metabolome Alterations Associated with Parkinson’s Disease. mSystems 2020, 5, e00561. [Google Scholar] [CrossRef]
  68. Nishiwaki, H.; Ito, M.; Ishida, T.; Hamaguchi, T.; Maeda, T.; Kashihara, K.; Tsuboi, Y.; Ueyama, J.; Shimamura, T.; Mori, H.; et al. Meta-Analysis of Gut Dysbiosis in Parkinson’s Disease. Mov. Disord. 2020, 35, 1626–1635. [Google Scholar] [CrossRef]
  69. Cirstea, M.S.; Yu, A.C.; Golz, E.; Sundvick, K.; Kliger, D.; Radisavljevic, N.; Foulger, L.H.; Mackenzie, M.; Huan, T.; Finlay, B.B.; et al. Microbiota Composition and Metabolism Are Associated with Gut Function in Parkinson’s Disease. Mov. Disord. 2020, 35, 1208–1217. [Google Scholar] [CrossRef]
  70. Cosma-Grigorov, A.; Meixner, H.; Mrochen, A.; Wirtz, S.; Winkler, J.; Marxreiter, F. Changes in Gastrointestinal Microbiome Composition in PD: A Pivotal Role of Covariates. Front. Neurol. 2020, 11, 1041. [Google Scholar] [CrossRef]
  71. Vogt, N.M.; Kerby, R.L.; Dill-McFarland, K.A.; Harding, S.J.; Merluzzi, A.P.; Johnson, S.C.; Carlsson, C.M.; Asthana, S.; Zetterberg, H.; Blennow, K.; et al. Gut Microbiome Alterations in Alzheimer’s Disease. Sci. Rep. 2017, 7, 13537. [Google Scholar] [CrossRef] [Green Version]
  72. Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. Association of Brain Amyloidosis with Pro-Inflammatory Gut Bacterial Taxa and Peripheral Inflammation Markers in Cognitively Impaired Elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef] [Green Version]
  73. Zhuang, Z.-Q.; Shen, L.-L.; Li, W.-W.; Fu, X.; Zeng, F.; Gui, L.; Lü, Y.; Cai, M.; Zhu, C.; Tan, Y.-L.; et al. Gut Microbiota Is Altered in Patients with Alzheimer’s Disease. J. Alzheimers Dis. 2018, 63, 1337–1346. [Google Scholar] [CrossRef] [Green Version]
  74. Haran, J.P.; Bhattarai, S.K.; Foley, S.E.; Dutta, P.; Ward, D.V.; Bucci, V.; McCormick, B.A. Alzheimer’s Disease Microbiome Is Associated with Dysregulation of the Anti-Inflammatory P-Glycoprotein Pathway. MBio 2019, 10, e00632-19. [Google Scholar] [CrossRef] [Green Version]
  75. Lopizzo, N.; Provasi, S.; Marizzoni, M.; Borruso, L.; Andryszak, P.; Frisoni, G.B.; Cattaneo, A. Identification of Gut Microbiota Signature in Alzheimer’s Disease: Possible Role in Influencing Peripheral Inflammation. Eur. Neuropsychopharmacol. 2019, 29, S167–S168. [Google Scholar] [CrossRef]
  76. Liu, P.; Wu, L.; Peng, G.; Han, Y.; Tang, R.; Ge, J.; Zhang, L.; Jia, L.; Yue, S.; Zhou, K.; et al. Altered Microbiomes Distinguish Alzheimer’s Disease from Amnestic Mild Cognitive Impairment and Health in a Chinese Cohort. Brain Behav. Immun. 2019, 80, 633–643. [Google Scholar] [CrossRef]
  77. Li, B.; He, Y.; Ma, J.; Huang, P.; Du, J.; Cao, L.; Wang, Y.; Xiao, Q.; Tang, H.; Chen, S. Mild Cognitive Impairment Has Similar Alterations as Alzheimer’s Disease in Gut Microbiota. Alzheimer’s Dement. 2019, 15, 1357–1366. [Google Scholar] [CrossRef]
  78. Miyake, S.; Kim, S.; Suda, W.; Oshima, K.; Nakamura, M.; Matsuoka, T.; Chihara, N.; Tomita, A.; Sato, W.; Kim, S.-W.; et al. Dysbiosis in the Gut Microbiota of Patients with Multiple Sclerosis, with a Striking Depletion of Species Belonging to Clostridia XIVa and IV Clusters. PLoS ONE 2015, 10, e0137429. [Google Scholar] [CrossRef] [Green Version]
  79. Cantarel, B.L.; Waubant, E.; Chehoud, C.; Kuczynski, J.; DeSantis, T.Z.; Warrington, J.; Venkatesan, A.; Fraser, C.M.; Mowry, E.M. Gut Microbiota in Multiple Sclerosis: Possible Influence of Immunomodulators. J. Investig. Med. 2015, 63, 729–734. [Google Scholar] [CrossRef] [Green Version]
  80. Chen, J.; Chia, N.; Kalari, K.R.; Yao, J.Z.; Novotna, M.; Paz Soldan, M.M.; Luckey, D.H.; Marietta, E.V.; Jeraldo, P.R.; Chen, X.; et al. Multiple Sclerosis Patients Have a Distinct Gut Microbiota Compared to Healthy Controls. Sci. Rep. 2016, 6, 28484. [Google Scholar] [CrossRef]
  81. Tremlett, H.; Fadrosh, D.W.; Faruqi, A.A.; Zhu, F.; Hart, J.; Roalstad, S.; Graves, J.; Lynch, S.; Waubant, E. US Network of Pediatric MS Centers Gut Microbiota in Early Pediatric Multiple Sclerosis: A Case-Control Study. Eur. J. Neurol. 2016, 23, 1308–1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Jangi, S.; Gandhi, R.; Cox, L.M.; Li, N.; von Glehn, F.; Yan, R.; Patel, B.; Mazzola, M.A.; Liu, S.; Glanz, B.L.; et al. Alterations of the Human Gut Microbiome in Multiple Sclerosis. Nat. Commun. 2016, 7, 12015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Cekanaviciute, E.; Yoo, B.B.; Runia, T.F.; Debelius, J.W.; Singh, S.; Nelson, C.A.; Kanner, R.; Bencosme, Y.; Lee, Y.K.; Hauser, S.L.; et al. Gut Bacteria from Multiple Sclerosis Patients Modulate Human T Cells and Exacerbate Symptoms in Mouse Models. Proc. Natl. Acad. Sci. USA 2017, 114, 10713–10718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Forbes, J.D.; Chen, C.; Knox, N.C.; Marrie, R.-A.; El-Gabalawy, H.; de Kievit, T.; Alfa, M.; Bernstein, C.N.; Van Domselaar, G. A Comparative Study of the Gut Microbiota in Immune-Mediated Inflammatory Diseases—Does a Common Dysbiosis Exist? Microbiome 2018, 6, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kozhieva, M.; Naumova, N.; Alikina, T.; Boyko, A.; Vlassov, V.; Kabilov, M.R. Primary Progressive Multiple Sclerosis in a Russian Cohort: Relationship with Gut Bacterial Diversity. BMC Microbiol. 2019, 19, 309. [Google Scholar] [CrossRef]
  86. Ventura, R.E.; Iizumi, T.; Battaglia, T.; Liu, M.; Perez-Perez, G.I.; Herbert, J.; Blaser, M.J. Gut Microbiome of Treatment-Naïve MS Patients of Different Ethnicities Early in Disease Course. Sci. Rep. 2019, 9, 16396. [Google Scholar] [CrossRef] [Green Version]
  87. Oezguen, N.; Yalcinkaya, N.; Kücükali, C.I.; Dahdouli, M.; Hollister, E.B.; Luna, R.A.; Türkoglu, R.; Kürtüncü, M.; Eraksoy, M.; Savidge, T.C.; et al. Microbiota Stratification Identifies Disease-Specific Alterations in Neuro-Behçet’s Disease and Multiple Sclerosis. Clin. Exp. Rheumatol. 2019, 37 (Suppl. 121), 58–66. [Google Scholar]
  88. Yin, J.; Liao, S.-X.; He, Y.; Wang, S.; Xia, G.-H.; Liu, F.-T.; Zhu, J.-J.; You, C.; Chen, Q.; Zhou, L.; et al. Dysbiosis of Gut Microbiota with Reduced Trimethylamine-N-Oxide Level in Patients With Large-Artery Atherosclerotic Stroke or Transient Ischemic Attack. J. Am. Heart Assoc. 2015, 4, e002699. [Google Scholar] [CrossRef] [Green Version]
  89. Yamashiro, K.; Tanaka, R.; Urabe, T.; Ueno, Y.; Yamashiro, Y.; Nomoto, K.; Takahashi, T.; Tsuji, H.; Asahara, T.; Hattori, N. Gut Dysbiosis Is Associated with Metabolism and Systemic Inflammation in Patients with Ischemic Stroke. PLoS ONE 2017, 12, e0171521. [Google Scholar] [CrossRef] [Green Version]
  90. Xia, G.-H.; You, C.; Gao, X.-X.; Zeng, X.-L.; Zhu, J.-J.; Xu, K.-Y.; Tan, C.-H.; Xu, R.-T.; Wu, Q.-H.; Zhou, H.-W.; et al. Stroke Dysbiosis Index (SDI) in Gut Microbiome Are Associated with Brain Injury and Prognosis of Stroke. Front. Neurol. 2019, 10, 397. [Google Scholar] [CrossRef]
  91. Li, N.; Wang, X.; Sun, C.; Wu, X.; Lu, M.; Si, Y.; Ye, X.; Wang, T.; Yu, X.; Zhao, X.; et al. Change of Intestinal Microbiota in Cerebral Ischemic Stroke Patients. BMC Microbiol. 2019, 19, 191. [Google Scholar] [CrossRef]
  92. Haak, B.W.; Westendorp, W.F.; van Engelen, T.S.R.; Brands, X.; Brouwer, M.C.; Vermeij, J.-D.; Hugenholtz, F.; Verhoeven, A.; Derks, R.J.; Giera, M.; et al. Disruptions of Anaerobic Gut Bacteria Are Associated with Stroke and Post-Stroke Infection: A Prospective Case-Control Study. Transl. Stroke Res. 2020, 12, 581–592. [Google Scholar] [CrossRef]
  93. Li, H.; Zhang, X.; Pan, D.; Liu, Y.; Yan, X.; Tang, Y.; Tao, M.; Gong, L.; Zhang, T.; Woods, C.R.; et al. Dysbiosis Characteristics of Gut Microbiota in Cerebral Infarction Patients. Transl. Neurosci. 2020, 11, 124–133. [Google Scholar] [CrossRef]
  94. Fang, X.; Wang, X.; Yang, S.; Meng, F.; Wang, X.; Wei, H.; Chen, T. Evaluation of the Microbial Diversity in Amyotrophic Lateral Sclerosis Using High-Throughput Sequencing. Front. Microbiol. 2016, 7, 1479. [Google Scholar] [CrossRef] [Green Version]
  95. Brenner, D.; Hiergeist, A.; Adis, C.; Mayer, B.; Gessner, A.; Ludolph, A.C.; Weishaupt, J.H. The Fecal Microbiome of ALS Patients. Neurobiol. Aging 2018, 61, 132–137. [Google Scholar] [CrossRef]
  96. Mazzini, L.; Mogna, L.; De Marchi, F.; Amoruso, A.; Pane, M.; Aloisio, I.; Cionci, N.B.; Gaggìa, F.; Lucenti, A.; Bersano, E.; et al. Potential Role of Gut Microbiota in ALS Pathogenesis and Possible Novel Therapeutic Strategies. J. Clin. Gastroenterol. 2018, 52, S68–S70. [Google Scholar] [CrossRef]
  97. Zhai, C.-D.; Zheng, J.-J.; An, B.-C.; Huang, H.-F.; Tan, Z.-C. Intestinal Microbiota Composition in Patients with Amyotrophic Lateral Sclerosis: Establishment of Bacterial and Archaeal Communities Analyses. Chin. Med. J. 2019, 132, 1815–1822. [Google Scholar] [CrossRef]
  98. Di Gioia, D.; Bozzi Cionci, N.; Baffoni, L.; Amoruso, A.; Pane, M.; Mogna, L.; Gaggìa, F.; Lucenti, M.A.; Bersano, E.; Cantello, R.; et al. A Prospective Longitudinal Study on the Microbiota Composition in Amyotrophic Lateral Sclerosis. BMC Med. 2020, 18, 153. [Google Scholar] [CrossRef]
  99. Zeng, Q.; Shen, J.; Chen, K.; Zhou, J.; Liao, Q.; Lu, K.; Yuan, J.; Bi, F. The Alteration of Gut Microbiome and Metabolism in Amyotrophic Lateral Sclerosis Patients. Sci. Rep. 2020, 10, 12998. [Google Scholar] [CrossRef]
  100. Rowin, J.; Xia, Y.; Jung, B.; Sun, J. Gut Inflammation and Dysbiosis in Human Motor Neuron Disease. Physiol. Rep. 2017, 5, e13443. [Google Scholar] [CrossRef]
  101. Xing, J.; Li, X.; Sun, Y.; Zhao, J.; Miao, S.; Xiong, Q.; Zhang, Y.; Zhang, G. Comparative Genomic and Functional Analysis of Akkermansia Muciniphila and Closely Related Species. Genes Genom. 2019, 41, 1253–1264. [Google Scholar] [CrossRef] [PubMed]
  102. Geerlings, S.Y.; Kostopoulos, I.; de Vos, W.M.; Belzer, C. Akkermansia Muciniphila in the Human Gastrointestinal Tract: When, Where, and How? Microorganisms 2018, 6, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Derrien, M.; Vaughan, E.E.; Plugge, C.M.; de Vos, W.M. Akkermansia Muciniphila Gen. Nov., Sp. Nov., a Human Intestinal Mucin-Degrading Bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54, 1469–1476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Goodrich, J.K.; Davenport, E.R.; Beaumont, M.; Jackson, M.A.; Knight, R.; Ober, C.; Spector, T.D.; Bell, J.T.; Clark, A.G.; Ley, R.E. Genetic Determinants of the Gut Microbiome in UK Twins. Cell Host Microbe 2016, 19, 731–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Liu, S.; Rezende, R.M.; Moreira, T.G.; Tankou, S.K.; Cox, L.M.; Wu, M.; Song, A.; Dhang, F.H.; Wei, Z.; Costamagna, G.; et al. Oral Administration of MiR-30d from Feces of MS Patients Suppresses MS-like Symptoms in Mice by Expanding Akkermansia Muciniphila. Cell Host Microbe 2019, 26, 779–794.e8. [Google Scholar] [CrossRef]
  106. Nandwana, V.; Nandwana, N.K.; Das, Y.; Saito, M.; Panda, T.; Das, S.; Almaguel, F.; Hosmane, N.S.; Das, B.C. The Role of Microbiome in Brain Development and Neurodegenerative Diseases. Molecules 2022, 27, 3402. [Google Scholar] [CrossRef]
  107. Hirsch, E.C.; Hunot, S. Neuroinflammation in Parkinson’s Disease: A Target for Neuroprotection? Lancet Neurol. 2009, 8, 382–397. [Google Scholar] [CrossRef]
  108. Simon, D.K.; Tanner, C.M.; Brundin, P. Parkinson Disease Epidemiology, Pathology, Genetics, and Pathophysiology. Clin. Geriatr. Med. 2020, 36, 1–12. [Google Scholar] [CrossRef]
  109. De Virgilio, A.; Greco, A.; Fabbrini, G.; Inghilleri, M.; Rizzo, M.I.; Gallo, A.; Conte, M.; Rosato, C.; Ciniglio Appiani, M.; de Vincentiis, M. Parkinson’s Disease: Autoimmunity and Neuroinflammation. Autoimmun. Rev. 2016, 15, 1005–1011. [Google Scholar] [CrossRef] [Green Version]
  110. EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA); Turck, D.; Bohn, T.; Castenmiller, J.; De Henauw, S.; Hirsch-Ernst, K.I.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; Naska, A.; et al. Safety of Pasteurised Akkermansia Muciniphila as a Novel Food Pursuant to Regulation (EU) 2015/2283. EFS2 2021, 19, e06780. [Google Scholar] [CrossRef]
  111. Romano, S.; Savva, G.M.; Bedarf, J.R.; Charles, I.G.; Hildebrand, F.; Narbad, A. Meta-Analysis of the Parkinson’s Disease Gut Microbiome Suggests Alterations Linked to Intestinal Inflammation. npj Parkinsons Dis. 2021, 7, 27. [Google Scholar] [CrossRef]
  112. Szilagyi, A.; Shrier, I.; Heilpern, D.; Je, J.; Park, S.; Chong, G.; Lalonde, C.; Cote, L.-F.; Lee, B. Differential Impact of Lactose/Lactase Phenotype on Colonic Microflora. Can. J. Gastroenterol. 2010, 24, 373–379. [Google Scholar] [CrossRef] [Green Version]
  113. O’Callaghan, A.; van Sinderen, D. Bifidobacteria and Their Role as Members of the Human Gut Microbiota. Front. Microbiol. 2016, 7, 925. [Google Scholar] [CrossRef] [Green Version]
  114. Li, X.; Li, W.; Liu, G.; Shen, X.; Tang, Y. Association between Cigarette Smoking and Parkinson’s Disease: A Meta-Analysis. Arch. Gerontol. Geriatr. 2015, 61, 510–516. [Google Scholar] [CrossRef]
  115. Sun, H.; Zhao, F.; Liu, Y.; Ma, T.; Jin, H.; Quan, K.; Leng, B.; Zhao, J.; Yuan, X.; Li, Z.; et al. Probiotics Synergized with Conventional Regimen in Managing Parkinson’s Disease. npj Parkinsons Dis. 2022, 8, 62. [Google Scholar] [CrossRef]
  116. Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.-F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of Psychotropic-like Properties of a Probiotic Formulation (Lactobacillus Helveticus R0052 and Bifidobacterium Longum R0175) in Rats and Human Subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [Green Version]
  117. Sarkar, A.; Lehto, S.M.; Harty, S.; Dinan, T.G.; Cryan, J.F.; Burnet, P.W.J. Psychobiotics and the Manipulation of Bacteria-Gut-Brain Signals. Trends Neurosci. 2016, 39, 763–781. [Google Scholar] [CrossRef] [Green Version]
  118. Savignac, H.M.; Couch, Y.; Stratford, M.; Bannerman, D.M.; Tzortzis, G.; Anthony, D.C.; Burnet, P.W.J. Prebiotic Administration Normalizes Lipopolysaccharide (LPS)-Induced Anxiety and Cortical 5-HT2A Receptor and IL1-β Levels in Male Mice. Brain Behav. Immun. 2016, 52, 120–131. [Google Scholar] [CrossRef] [Green Version]
  119. Rajilić-Stojanović, M.; Biagi, E.; Heilig, H.G.H.J.; Kajander, K.; Kekkonen, R.A.; Tims, S.; de Vos, W.M. Global and Deep Molecular Analysis of Microbiota Signatures in Fecal Samples from Patients with Irritable Bowel Syndrome. Gastroenterology 2011, 141, 1792–1801. [Google Scholar] [CrossRef]
  120. Maini Rekdal, V.; Bess, E.N.; Bisanz, J.E.; Turnbaugh, P.J.; Balskus, E.P. Discovery and Inhibition of an Interspecies Gut Bacterial Pathway for Levodopa Metabolism. Science 2019, 364, eaau6323. [Google Scholar] [CrossRef] [Green Version]
  121. Van Kessel, S.P.; Frye, A.K.; El-Gendy, A.O.; Castejon, M.; Keshavarzian, A.; van Dijk, G.; El Aidy, S. Gut Bacterial Tyrosine Decarboxylases Restrict Levels of Levodopa in the Treatment of Parkinson’s Disease. Nat. Commun. 2019, 10, 310. [Google Scholar] [CrossRef] [PubMed]
  122. Connil, N.; Le Breton, Y.; Dousset, X.; Auffray, Y.; Rincé, A.; Prévost, H. Identification of the Enterococcus Faecalis Tyrosine Decarboxylase Operon Involved in Tyramine Production. Appl. Environ. Microbiol. 2002, 68, 3537–3544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Olson, C.A.; Vuong, H.E.; Yano, J.M.; Liang, Q.Y.; Nusbaum, D.J.; Hsiao, E.Y. The Gut Microbiota Mediates the Anti-Seizure Effects of the Ketogenic Diet. Cell 2018, 173, 1728–1741.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kamada, N.; Seo, S.-U.; Chen, G.Y.; Núñez, G. Role of the Gut Microbiota in Immunity and Inflammatory Disease. Nat. Rev. Immunol. 2013, 13, 321–335. [Google Scholar] [CrossRef] [PubMed]
  125. Russo, R.; Cristiano, C.; Avagliano, C.; De Caro, C.; La Rana, G.; Raso, G.M.; Canani, R.B.; Meli, R.; Calignano, A. Gut-Brain Axis: Role of Lipids in the Regulation of Inflammation, Pain and CNS Diseases. Curr. Med. Chem. 2018, 25, 3930–3952. [Google Scholar] [CrossRef]
  126. Tsunoda, I. Lymphatic System and Gut Microbiota Affect Immunopathology of Neuroinflammatory Diseases, Including Multiple Sclerosis, Neuromyelitis Optica and Alzheimer’s Disease. Clin. Exp. Neuroimmunol. 2017, 8, 177–179. [Google Scholar] [CrossRef] [Green Version]
  127. Strati, F.; Cavalieri, D.; Albanese, D.; De Felice, C.; Donati, C.; Hayek, J.; Jousson, O.; Leoncini, S.; Pindo, M.; Renzi, D.; et al. Altered Gut Microbiota in Rett Syndrome. Microbiome 2016, 4, 41. [Google Scholar] [CrossRef] [Green Version]
  128. Xu, R.; Tan, C.; Zhu, J.; Zeng, X.; Gao, X.; Wu, Q.; Chen, Q.; Wang, H.; Zhou, H.; He, Y.; et al. Dysbiosis of the Intestinal Microbiota in Neurocritically Ill Patients and the Risk for Death. Crit. Care 2019, 23, 195. [Google Scholar] [CrossRef] [Green Version]
  129. Li, J.; Zhao, F.; Wang, Y.; Chen, J.; Tao, J.; Tian, G.; Wu, S.; Liu, W.; Cui, Q.; Geng, B.; et al. Gut Microbiota Dysbiosis Contributes to the Development of Hypertension. Microbiome 2017, 5, 14. [Google Scholar] [CrossRef] [Green Version]
  130. Yan, Q.; Gu, Y.; Li, X.; Yang, W.; Jia, L.; Chen, C.; Han, X.; Huang, Y.; Zhao, L.; Li, P.; et al. Alterations of the Gut Microbiome in Hypertension. Front. Cell. Infect. Microbiol. 2017, 7, 381. [Google Scholar] [CrossRef] [Green Version]
  131. Bajaj, J.S.; Hylemon, P.B.; Ridlon, J.M.; Heuman, D.M.; Daita, K.; White, M.B.; Monteith, P.; Noble, N.A.; Sikaroodi, M.; Gillevet, P.M. Colonic Mucosal Microbiome Differs from Stool Microbiome in Cirrhosis and Hepatic Encephalopathy and Is Linked to Cognition and Inflammation. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G675–G685. [Google Scholar] [CrossRef]
  132. Bajaj, J.S.; Heuman, D.M.; Hylemon, P.B.; Sanyal, A.J.; White, M.B.; Monteith, P.; Noble, N.A.; Unser, A.B.; Daita, K.; Fisher, A.R.; et al. Altered Profile of Human Gut Microbiome Is Associated with Cirrhosis and Its Complications. J. Hepatol. 2014, 60, 940–947. [Google Scholar] [CrossRef] [Green Version]
  133. Ni, J.; Huang, R.; Zhou, H.; Xu, X.; Li, Y.; Cao, P.; Zhong, K.; Ge, M.; Chen, X.; Hou, B.; et al. Analysis of the Relationship Between the Degree of Dysbiosis in Gut Microbiota and Prognosis at Different Stages of Primary Hepatocellular Carcinoma. Front. Microbiol. 2019, 10, 1458. [Google Scholar] [CrossRef]
  134. Chen, P.-C.; Chien, Y.-W.; Yang, S.-C. The Alteration of Gut Microbiota in Newly Diagnosed Type 2 Diabetic Patients. Nutrition 2019, 63, 51–56. [Google Scholar] [CrossRef]
  135. Chen, B.; Sun, L.; Zhang, X. Integration of Microbiome and Epigenome to Decipher the Pathogenesis of Autoimmune Diseases. J. Autoimmun. 2017, 83, 31–42. [Google Scholar] [CrossRef]
  136. Luo, X.M.; Edwards, M.R.; Mu, Q.; Yu, Y.; Vieson, M.D.; Reilly, C.M.; Ahmed, S.A.; Bankole, A.A. Gut Microbiota in Human Systemic Lupus Erythematosus and a Mouse Model of Lupus. Appl. Environ. Microbiol. 2018, 84, e02288-17. [Google Scholar] [CrossRef] [Green Version]
  137. Ipci, K.; Altıntoprak, N.; Muluk, N.B.; Senturk, M.; Cingi, C. The Possible Mechanisms of the Human Microbiome in Allergic Diseases. Eur. Arch. Otorhinolaryngol. 2017, 274, 617–626. [Google Scholar] [CrossRef]
  138. Ye, Z.; Zhang, N.; Wu, C.; Zhang, X.; Wang, Q.; Huang, X.; Du, L.; Cao, Q.; Tang, J.; Zhou, C.; et al. A Metagenomic Study of the Gut Microbiome in Behcet’s Disease. Microbiome 2018, 6, 135. [Google Scholar] [CrossRef] [Green Version]
  139. Patrone, V.; Puglisi, E.; Cardinali, M.; Schnitzler, T.S.; Svegliati, S.; Festa, A.; Gabrielli, A.; Morelli, L. Gut Microbiota Profile in Systemic Sclerosis Patients with and without Clinical Evidence of Gastrointestinal Involvement. Sci. Rep. 2017, 7, 14874. [Google Scholar] [CrossRef] [Green Version]
  140. Liu, X.; Zou, Q.; Zeng, B.; Fang, Y.; Wei, H. Analysis of Fecal Lactobacillus Community Structure in Patients with Early Rheumatoid Arthritis. Curr. Microbiol. 2013, 67, 170–176. [Google Scholar] [CrossRef]
  141. Zuo, K.; Li, J.; Xu, Q.; Hu, C.; Gao, Y.; Chen, M.; Hu, R.; Liu, Y.; Chi, H.; Yin, Q.; et al. Dysbiotic Gut Microbes May Contribute to Hypertension by Limiting Vitamin D Production. Clin. Cardiol. 2019, 42, 710–719. [Google Scholar] [CrossRef] [PubMed]
  142. Kim, H.-N.; Yun, Y.; Ryu, S.; Chang, Y.; Kwon, M.-J.; Cho, J.; Shin, H.; Kim, H.-L. Correlation between Gut Microbiota and Personality in Adults: A Cross-Sectional Study. Brain Behav. Immun. 2018, 69, 374–385. [Google Scholar] [CrossRef] [PubMed]
  143. Libbey, J.E.; Sanchez, J.M.; Doty, D.J.; Sim, J.T.; Cusick, M.F.; Cox, J.E.; Fischer, K.F.; Round, J.L.; Fujinami, R.S. Variations in Diet Cause Alterations in Microbiota and Metabolites That Follow Changes in Disease Severity in a Multiple Sclerosis Model. Benef. Microbes 2018, 9, 495–513. [Google Scholar] [CrossRef] [PubMed]
  144. Rice, M.W.; Pandya, J.D.; Shear, D.A. Gut Microbiota as a Therapeutic Target to Ameliorate the Biochemical, Neuroanatomical, and Behavioral Effects of Traumatic Brain Injuries. Front. Neurol. 2019, 10, 875. [Google Scholar] [CrossRef] [PubMed]
  145. Cignarella, F.; Cantoni, C.; Ghezzi, L.; Salter, A.; Dorsett, Y.; Chen, L.; Phillips, D.; Weinstock, G.M.; Fontana, L.; Cross, A.H.; et al. Intermittent Fasting Confers Protection in CNS Autoimmunity by Altering the Gut Microbiota. Cell Metab. 2018, 27, 1222–1235.e6. [Google Scholar] [CrossRef] [Green Version]
  146. Zhang, Y.; Zhou, S.; Zhou, Y.; Yu, L.; Zhang, L.; Wang, Y. Altered Gut Microbiome Composition in Children with Refractory Epilepsy after Ketogenic Diet. Epilepsy Res. 2018, 145, 163–168. [Google Scholar] [CrossRef]
  147. Lindefeldt, M.; Eng, A.; Darban, H.; Bjerkner, A.; Zetterström, C.K.; Allander, T.; Andersson, B.; Borenstein, E.; Dahlin, M.; Prast-Nielsen, S. The Ketogenic Diet Influences Taxonomic and Functional Composition of the Gut Microbiota in Children with Severe Epilepsy. npj Biofilms Microbiomes 2019, 5, 5. [Google Scholar] [CrossRef] [Green Version]
  148. Fan, Y.; Wang, H.; Liu, X.; Zhang, J.; Liu, G. Crosstalk between the Ketogenic Diet and Epilepsy: From the Perspective of Gut Microbiota. Mediat. Inflamm. 2019, 2019, 8373060. [Google Scholar] [CrossRef]
  149. Spinelli, E.; Blackford, R. Gut Microbiota, the Ketogenic Diet and Epilepsy. Pediatr. Neurol. Briefs 2018, 32, 10. [Google Scholar] [CrossRef]
  150. Cui, B.; Li, P.; Xu, L.; Peng, Z.; Xiang, J.; He, Z.; Zhang, T.; Ji, G.; Nie, Y.; Wu, K.; et al. Step-up Fecal Microbiota Transplantation (FMT) Strategy. Gut Microbes 2016, 7, 323–328. [Google Scholar] [CrossRef] [Green Version]
  151. Kao, D.; Roach, B.; Silva, M.; Beck, P.; Rioux, K.; Kaplan, G.G.; Chang, H.-J.; Coward, S.; Goodman, K.J.; Xu, H.; et al. Effect of Oral Capsule- vs Colonoscopy-Delivered Fecal Microbiota Transplantation on Recurrent Clostridium Difficile Infection: A Randomized Clinical Trial. JAMA 2017, 318, 1985–1993. [Google Scholar] [CrossRef]
  152. Sun, M.-F.; Zhu, Y.-L.; Zhou, Z.-L.; Jia, X.-B.; Xu, Y.-D.; Yang, Q.; Cui, C.; Shen, Y.-Q. Neuroprotective Effects of Fecal Microbiota Transplantation on MPTP-Induced Parkinson’s Disease Mice: Gut Microbiota, Glial Reaction and TLR4/TNF-α Signaling Pathway. Brain Behav. Immun. 2018, 70, 48–60. [Google Scholar] [CrossRef]
  153. Evrensel, A.; Ceylan, M.E. Fecal Microbiota Transplantation and Its Usage in Neuropsychiatric Disorders. Clin. Psychopharmacol. Neurosci. 2016, 14, 231–237. [Google Scholar] [CrossRef] [Green Version]
  154. Zhao, Z.; Ning, J.; Bao, X.-Q.; Shang, M.; Ma, J.; Li, G.; Zhang, D. Fecal Microbiota Transplantation Protects Rotenone-Induced Parkinson’s Disease Mice via Suppressing Inflammation Mediated by the Lipopolysaccharide-TLR4 Signaling Pathway through the Microbiota-Gut-Brain Axis. Microbiome 2021, 9, 226. [Google Scholar] [CrossRef]
  155. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert Consensus Document. The International Scientific Association for Probiotics and Prebiotics Consensus Statement on the Scope and Appropriate Use of the Term Probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  156. Gazerani, P. Probiotics for Parkinson’s Disease. Int. J. Mol. Sci. 2019, 20, 74121. [Google Scholar] [CrossRef] [Green Version]
  157. Kesika, P.; Suganthy, N.; Sivamaruthi, B.S.; Chaiyasut, C. Role of Gut-Brain Axis, Gut Microbial Composition, and Probiotic Intervention in Alzheimer’s Disease. Life Sci. 2021, 264, 118627. [Google Scholar] [CrossRef]
  158. Ghezzi, L.; Cantoni, C.; Pinget, G.V.; Zhou, Y.; Piccio, L. Targeting the Gut to Treat Multiple Sclerosis. J. Clin. Investig. 2021, 131, 143774. [Google Scholar] [CrossRef]
  159. Zhong, D.-Y.; Li, L.; Ma, R.-M.; Deng, Y.-H. The Effect of Probiotics in Stroke Treatment. Evid. Based Complement. Altern. Med. 2021, 2021, 4877311. [Google Scholar] [CrossRef]
  160. Quigley, E.M.M. Microbiota-Brain-Gut Axis and Neurodegenerative Diseases. Curr. Neurol. Neurosci. Rep. 2017, 17, 94. [Google Scholar] [CrossRef]
  161. Shen, L.; Liu, L.; Ji, H.-F. Alzheimer’s Disease Histological and Behavioral Manifestations in Transgenic Mice Correlate with Specific Gut Microbiome State. J. Alzheimers Dis. 2017, 56, 385–390. [Google Scholar] [CrossRef] [PubMed]
  162. Harach, T.; Marungruang, N.; Duthilleul, N.; Cheatham, V.; Mc Coy, K.D.; Frisoni, G.; Neher, J.J.; Fåk, F.; Jucker, M.; Lasser, T.; et al. Reduction of Abeta Amyloid Pathology in APPPS1 Transgenic Mice in the Absence of Gut Microbiota. Sci. Rep. 2017, 7, 41802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Bonfili, L.; Cecarini, V.; Berardi, S.; Scarpona, S.; Suchodolski, J.S.; Nasuti, C.; Fiorini, D.; Boarelli, M.C.; Rossi, G.; Eleuteri, A.M. Microbiota Modulation Counteracts Alzheimer’s Disease Progression Influencing Neuronal Proteolysis and Gut Hormones Plasma Levels. Sci. Rep. 2017, 7, 2426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Sun, J.; Xu, J.; Ling, Y.; Wang, F.; Gong, T.; Yang, C.; Ye, S.; Ye, K.; Wei, D.; Song, Z.; et al. Fecal Microbiota Transplantation Alleviated Alzheimer’s Disease-like Pathogenesis in APP/PS1 Transgenic Mice. Transl. Psychiatry 2019, 9, 189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Wang, M.; Amakye, W.K.; Guo, L.; Gong, C.; Zhao, Y.; Yao, M.; Ren, J. Walnut-Derived Peptide PW5 Ameliorates Cognitive Impairments and Alters Gut Microbiota in APP/PS1 Transgenic Mice. Mol. Nutr. Food Res. 2019, 63, 1900326. [Google Scholar] [CrossRef]
  166. Tran, T.T.T.; Corsini, S.; Kellingray, L.; Hegarty, C.; Le Gall, G.; Narbad, A.; Müller, M.; Tejera, N.; O’Toole, P.W.; Minihane, A.-M.; 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, 8221–8231. [Google Scholar] [CrossRef] [Green Version]
  167. Zhan, J.; Liang, Y.; Liu, D.; Ma, X.; Li, P.; Liu, C.; Liu, X.; Wang, P.; Zhou, Z. Antibiotics May Increase Triazine Herbicide Exposure Risk via Disturbing Gut Microbiota. Microbiome 2018, 6, 224. [Google Scholar] [CrossRef] [Green Version]
  168. Brandscheid, C.; Schuck, F.; Reinhardt, S.; Schäfer, K.-H.; Pietrzik, C.U.; Grimm, M.; Hartmann, T.; Schwiertz, A.; Endres, K. Altered Gut Microbiome Composition and Tryptic Activity of the 5xFAD Alzheimer’s Mouse Model. J. Alzheimers Dis. 2017, 56, 775–788. [Google Scholar] [CrossRef]
  169. Maldonado Weng, J.; Parikh, I.; Naqib, A.; York, J.; Green, S.J.; Estus, S.; LaDu, M.J. Synergistic Effects of APOE and Sex on the Gut Microbiome of Young EFAD Transgenic Mice. Mol. Neurodegener. 2019, 14, 47. [Google Scholar] [CrossRef]
  170. Hoffman, J.D.; Yanckello, L.M.; Chlipala, G.; Hammond, T.C.; McCulloch, S.D.; Parikh, I.; Sun, S.; Morganti, J.M.; Green, S.J.; Lin, A.-L. Dietary Inulin Alters the Gut Microbiome, Enhances Systemic Metabolism and Reduces Neuroinflammation in an APOE4 Mouse Model. PLoS ONE 2019, 14, e0221828. [Google Scholar] [CrossRef] [Green Version]
  171. Lee, H.-J.; Lee, K.-E.; Kim, J.-K.; Kim, D.-H. Suppression of Gut Dysbiosis by Bifidobacterium Longum Alleviates Cognitive Decline in 5XFAD Transgenic and Aged Mice. Sci. Rep. 2019, 9, 11814. [Google Scholar] [CrossRef]
  172. Shen, L.; Liu, L.; Li, X.-Y.; Ji, H.-F. Regulation of Gut Microbiota in Alzheimer’s Disease Mice by Silibinin and Silymarin and Their Pharmacological Implications. Appl. Microbiol. Biotechnol. 2019, 103, 7141–7149. [Google Scholar] [CrossRef]
  173. Jackson, A.; Engen, P.A.; Forsyth, C.B.; Shaikh, M.; Naqib, A.; Wilber, S.; Frausto, D.M.; Raeisi, S.; Green, S.J.; Bradaric, B.D.; et al. Intestinal Barrier Dysfunction in the Absence of Systemic Inflammation Fails to Exacerbate Motor Dysfunction and Brain Pathology in a Mouse Model of Parkinson’s Disease. Front. Neurol. 2022, 13, 882628. [Google Scholar] [CrossRef]
  174. Lai, F.; Jiang, R.; Xie, W.; Liu, X.; Tang, Y.; Xiao, H.; Gao, J.; Jia, Y.; Bai, Q. Intestinal Pathology and Gut Microbiota Alterations in a Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine (MPTP) Mouse Model of Parkinson’s Disease. Neurochem. Res. 2018, 43, 1986–1999. [Google Scholar] [CrossRef]
  175. Zhou, X.; Lu, J.; Wei, K.; Wei, J.; Tian, P.; Yue, M.; Wang, Y.; Hong, D.; Li, F.; Wang, B.; et al. Neuroprotective Effect of Ceftriaxone on MPTP-Induced Parkinson’s Disease Mouse Model by Regulating Inflammation and Intestinal Microbiota. Oxid. Med. Cell. Longev. 2021, 2021, 9424582. [Google Scholar] [CrossRef]
  176. Cui, C.; Han, Y.; Li, H.; Yu, H.; Zhang, B.; Li, G. Curcumin-Driven Reprogramming of the Gut Microbiota and Metabolome Ameliorates Motor Deficits and Neuroinflammation in a Mouse Model of Parkinson’s Disease. Front. Cell. Infect. Microbiol. 2022, 12, 887407. [Google Scholar] [CrossRef]
  177. Liu, M.-M.; Zhou, N.; Jiang, N.; Lu, K.-M.; Wu, C.-F.; Bao, J.-K. Neuroprotective Effects of Oligosaccharides from Periplaneta Americana on Parkinson’s Disease Models In Vitro and In Vivo. Front. Pharmacol. 2022, 13, 936818. [Google Scholar] [CrossRef]
  178. Wang, N.; Feng, B.-N.; Hu, B.; Cheng, Y.-L.; Guo, Y.-H.; Qian, H. Neuroprotection of Chicoric Acid in a Mouse Model of Parkinson’s Disease Involves Gut Microbiota and TLR4 Signaling Pathway. Food Funct. 2022, 13, 2019–2032. [Google Scholar] [CrossRef]
  179. Zahra, W.; Birla, H.; Singh, S.S.; Rathore, A.S.; Dilnashin, H.; Singh, R.; Keshri, P.K.; Gautam, P.; Singh, S.P. Neuroprotection by Mucuna Pruriens in Neurodegenerative Diseases. Neurochem. Res. 2022, 47, 1816–1829. [Google Scholar] [CrossRef]
  180. Wallen, Z.D.; Appah, M.; Dean, M.N.; Sesler, C.L.; Factor, S.A.; Molho, E.; Zabetian, C.P.; Standaert, D.G.; Payami, H. Characterizing Dysbiosis of Gut Microbiome in PD: Evidence for Overabundance of Opportunistic Pathogens. npj Parkinsons Dis. 2020, 6, 11. [Google Scholar] [CrossRef]
  181. Simões, C.D.; Maukonen, J.; Kaprio, J.; Rissanen, A.; Pietiläinen, K.H.; Saarela, M. Habitual Dietary Intake Is Associated with Stool Microbiota Composition in Monozygotic Twins. J. Nutr. 2013, 143, 417–423. [Google Scholar] [CrossRef] [PubMed]
  182. Matsuki, T.; Yahagi, K.; Mori, H.; Matsumoto, H.; Hara, T.; Tajima, S.; Ogawa, E.; Kodama, H.; Yamamoto, K.; Yamada, T.; et al. A Key Genetic Factor for Fucosyllactose Utilization Affects Infant Gut Microbiota Development. Nat. Commun. 2016, 7, 11939. [Google Scholar] [CrossRef] [PubMed]
  183. Bowyer, R.C.E.; Jackson, M.A.; Le Roy, C.I.; Ni Lochlainn, M.; Spector, T.D.; Dowd, J.B.; Steves, C.J. Socioeconomic Status and the Gut Microbiome: A TwinsUK Cohort Study. Microorganisms 2019, 7, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Bibbò, S.; Ianiro, G.; Giorgio, V.; Scaldaferri, F.; Masucci, L.; Gasbarrini, A.; Cammarota, G. The Role of Diet on Gut Microbiota Composition. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 4742–4749. [Google Scholar] [PubMed]
  185. Chassaing, B.; Vijay-Kumar, M.; Gewirtz, A.T. How Diet Can Impact Gut Microbiota to Promote or Endanger Health. Curr. Opin. Gastroenterol. 2017, 33, 417–421. [Google Scholar] [CrossRef]
  186. Proctor, C.; Thiennimitr, P.; Chattipakorn, N.; Chattipakorn, S.C. Diet, Gut Microbiota and Cognition. Metab. Brain Dis. 2017, 32, 1–17. [Google Scholar] [CrossRef]
  187. Sandhu, K.V.; Sherwin, E.; Schellekens, H.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Feeding the Microbiota-Gut-Brain Axis: Diet, Microbiome, and Neuropsychiatry. Transl. Res. 2017, 179, 223–244. [Google Scholar] [CrossRef]
  188. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What Is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [Green Version]
  189. Temko, J.E.; Bouhlal, S.; Farokhnia, M.; Lee, M.R.; Cryan, J.F.; Leggio, L. The Microbiota, the Gut and the Brain in Eating and Alcohol Use Disorders: A “Ménage à Trois”? Alcohol Alcohol. 2017, 52, 403–413. [Google Scholar] [CrossRef] [Green Version]
  190. Gao, R.; Zhang, X.; Huang, L.; Shen, R.; Qin, H. Gut Microbiota Alteration After Long-Term Consumption of Probiotics in the Elderly. Probiotics Antimicrob. Proteins 2019, 11, 655–666. [Google Scholar] [CrossRef]
  191. Kim, J.E.; Park, J.J.; Lee, M.R.; Choi, J.Y.; Song, B.R.; Park, J.W.; Kang, M.J.; Son, H.J.; Hong, J.T.; Hwang, D.Y. Constipation in Tg2576 Mice Model for Alzheimer’s Disease Associated with Dysregulation of Mechanism Involving the MAChR Signaling Pathway and ER Stress Response. PLoS ONE 2019, 14, e0215205. [Google Scholar] [CrossRef]
  192. Sánchez, B.; Delgado, S.; Blanco-Míguez, A.; Lourenço, A.; Gueimonde, M.; Margolles, A. Probiotics, Gut Microbiota, and Their Influence on Host Health and Disease. Mol. Nutr. Food Res. 2017, 61, 1600240. [Google Scholar] [CrossRef] [Green Version]
  193. Roe, K. An Alternative Explanation for Alzheimer’s Disease and Parkinson’s Disease Initiation from Specific Antibiotics, Gut Microbiota Dysbiosis and Neurotoxins. Neurochem. Res. 2022, 47, 517–530. [Google Scholar] [CrossRef]
  194. King, C.H.; Desai, H.; Sylvetsky, A.C.; LoTempio, J.; Ayanyan, S.; Carrie, J.; Crandall, K.A.; Fochtman, B.C.; Gasparyan, L.; Gulzar, N.; et al. Baseline Human Gut Microbiota Profile in Healthy People and Standard Reporting Template. PLoS ONE 2019, 14, e0206484. [Google Scholar] [CrossRef] [Green Version]
  195. Xiang, L.; Lou, Y.; Liu, L.; Liu, Y.; Zhang, W.; Deng, J.; Guan, Y.; She, M.; You, X.; Liu, M.; et al. Gut Microbiotic Features Aiding the Diagnosis of Acute Ischemic Stroke. Front. Cell. Infect. Microbiol. 2020, 10, 587284. [Google Scholar] [CrossRef]
  196. Ling, Y.; Gong, T.; Zhang, J.; Gu, Q.; Gao, X.; Weng, X.; Liu, J.; Sun, J. Gut Microbiome Signatures Are Biomarkers for Cognitive Impairment in Patients with Ischemic Stroke. Front. Aging Neurosci. 2020, 12, 511562. [Google Scholar] [CrossRef]
Figure 2. PRISMA flow diagram of the studies selection.
Figure 2. PRISMA flow diagram of the studies selection.
Ijms 23 13665 g002
Figure 3. Phylogenetic distributions of the microbiota involved in Parkinson’s. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70].
Figure 3. Phylogenetic distributions of the microbiota involved in Parkinson’s. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70].
Ijms 23 13665 g003
Figure 4. Phylogenetic distributions of the microbiota involved in Alzheimer’s disease. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [71,72,73,74,75,76,77].
Figure 4. Phylogenetic distributions of the microbiota involved in Alzheimer’s disease. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [71,72,73,74,75,76,77].
Ijms 23 13665 g004
Figure 5. Phylogenetic distributions of the microbiota involved in multiple sclerosis. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [78,79,80,81,82,83,84,85,86,87].
Figure 5. Phylogenetic distributions of the microbiota involved in multiple sclerosis. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [78,79,80,81,82,83,84,85,86,87].
Ijms 23 13665 g005
Figure 6. Phylogenetic distributions of the microbiota involved in stroke. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [88,89,90,91,92,93].
Figure 6. Phylogenetic distributions of the microbiota involved in stroke. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [88,89,90,91,92,93].
Ijms 23 13665 g006
Figure 7. Phylogenetic distributions of the microbiota involved in amyotrophic lateral sclerosis. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [94,95,96,97,98,99].
Figure 7. Phylogenetic distributions of the microbiota involved in amyotrophic lateral sclerosis. Orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. These results are the summary of consistent findings of the included studies [94,95,96,97,98,99].
Ijms 23 13665 g007
Figure 8. List of the microbiota most frequently found and their distributions according to pathologies.
Figure 8. List of the microbiota most frequently found and their distributions according to pathologies.
Ijms 23 13665 g008
Figure 9. Venn diagram for the genera consistently found in the literature for neurodegenerative and neurological diseases.
Figure 9. Venn diagram for the genera consistently found in the literature for neurodegenerative and neurological diseases.
Ijms 23 13665 g009
Figure 10. Microbiota that have been identified in at least two studies for the same pathologies at the phylum, class, order and family levels. Grey is used for one study, orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. The numbers of the circle indicate the number of studies that identify the microbiota and the direction of the inconsistencies.
Figure 10. Microbiota that have been identified in at least two studies for the same pathologies at the phylum, class, order and family levels. Grey is used for one study, orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. The numbers of the circle indicate the number of studies that identify the microbiota and the direction of the inconsistencies.
Ijms 23 13665 g010
Figure 11. Microbiota that have been identified in at least two studies for the same pathologies at the genus level. Grey is used for one study, orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. The numbers of the circle indicate the number of studies that identify the microbiota and the direction of the inconsistencies.
Figure 11. Microbiota that have been identified in at least two studies for the same pathologies at the genus level. Grey is used for one study, orange indicates inconsistent results, blue indicates a decrease in relative abundance, and red indicates an increase in relative abundance. The numbers of the circle indicate the number of studies that identify the microbiota and the direction of the inconsistencies.
Ijms 23 13665 g011
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bonnechère, B.; Amin, N.; van Duijn, C. What Are the Key Gut Microbiota Involved in Neurological Diseases? A Systematic Review. Int. J. Mol. Sci. 2022, 23, 13665. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232213665

AMA Style

Bonnechère B, Amin N, van Duijn C. What Are the Key Gut Microbiota Involved in Neurological Diseases? A Systematic Review. International Journal of Molecular Sciences. 2022; 23(22):13665. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232213665

Chicago/Turabian Style

Bonnechère, Bruno, Najaf Amin, and Cornelia van Duijn. 2022. "What Are the Key Gut Microbiota Involved in Neurological Diseases? A Systematic Review" International Journal of Molecular Sciences 23, no. 22: 13665. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232213665

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop