Ijms 22 10028

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

International Journal of

Molecular Sciences

Review
The Role of Gut Microbiota and Gut–Brain Interplay in
Selected Diseases of the Central Nervous System
Julia Doroszkiewicz 1, * , Magdalena Groblewska 2 and Barbara Mroczko 1,3

1 Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland;


[email protected]
2 Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
[email protected]
3 Department of Biochemical Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland
* Correspondence: [email protected]; Tel.: +48-85-686-51-68

Abstract: The gut microbiome has attracted increasing attention from researchers in recent years.
The microbiota can have a specific and complex cross-talk with the host, particularly with the central
nervous system (CNS), creating the so-called “gut–brain axis”. Communication between the gut,
intestinal microbiota, and the brain involves the secretion of various metabolites such as short-chain
fatty acids (SCFAs), structural components of bacteria, and signaling molecules. Moreover, an
imbalance in the gut microbiota composition modulates the immune system and function of tissue
barriers such as the blood–brain barrier (BBB). Therefore, the aim of this literature review is to describe
how the gut–brain interplay may contribute to the development of various neurological disorders,
combining the fields of gastroenterology and neuroscience. We present recent findings concerning the

 effect of the altered microbiota on neurodegeneration and neuroinflammation, including Alzheimer’s
Citation: Doroszkiewicz, J.;
and Parkinson’s diseases, as well as multiple sclerosis. Moreover, the impact of the pathological shift
Groblewska, M.; Mroczko, B. The in the microbiome on selected neuropsychological disorders, i.e., major depressive disorders (MDD)
Role of Gut Microbiota and Gut–Brain and autism spectrum disorder (ASD), is also discussed. Future research on the effect of balanced gut
Interplay in Selected Diseases of the microbiota composition on the gut–brain axis would help to identify new potential opportunities for
Central Nervous System. Int. J. Mol. therapeutic interventions in the presented diseases.
Sci. 2021, 22, 10028. https://doi.org/
10.3390/ijms221810028 Keywords: gut microbiota; dysbiosis; gut–brain axis; central nervous system; enteric nervous system;
neuroinflammation; neurodegeneration; neuropsychiatric disorders
Academic Editor:
Antonella Scorziello

Received: 16 August 2021


1. Introduction
Accepted: 15 September 2021
Published: 17 September 2021
When we think of the intestines, we associate them with rather basic and somewhat
primitive functions that include the processing of food to deliver energy to the body.
Publisher’s Note: MDPI stays neutral
However, this type of understanding of the role of the gut appears to be inaccurate. The
with regard to jurisdictional claims in
gut is innervated by the enteric nervous system (ENS), which acts independently from the
published maps and institutional affil- central nervous system (CNS). However, there is a connection between these two parts of
iations. the human nervous system that allows for the exchange of information [1]. Knowledge of
the gut function and its connection to the CNS through the ENS has prompted researchers
to postulate the existence of a specific network called the “gut–brain axis”. The network
is based on a complex system, including the vagus nerve, with both sympathetic and
Copyright: © 2021 by the authors.
parasympathetic input, as well as certain gut hormones and cytokines [2,3].
Licensee MDPI, Basel, Switzerland.
The gut microbiota is a crucial part of the gastrointestinal tract. Recent data suggest
This article is an open access article
that the connection between the gut and the brain should also be extended to the intestinal
distributed under the terms and microbiome, creating the specific “microbiota–gut–brain axis”, as the latter role of bacterial
conditions of the Creative Commons flora within the gut has significantly emerged. Interestingly, it is postulated that microbes
Attribution (CC BY) license (https:// may participate in the development of the nervous system. This hypothesis has been con-
creativecommons.org/licenses/by/ firmed in germ-free mouse models, which are considered the gold standard for microbiota
4.0/). studies. These studies have demonstrated impaired neurogenesis and morphology in the

Int. J. Mol. Sci. 2021, 22, 10028. https://doi.org/10.3390/ijms221810028 https://www.mdpi.com/journal/ijms


Int. J. Mol. Sci. 2021, 22, 10028 2 of 19

hippocampus and amygdala of the tested animals [4,5]. Furthermore, a different study
conducted on germ-free mice revealed a disrupted microglia morphology and its processes,
resulting in disturbed activation and response to pathogens [6]. These results have led
to the establishment of a connection between pathological changes in the gut microbiota
and neurological diseases. This review summarizes recent findings connecting the areas of
microbiology, neurology, and the interplay between the gut microbiota and the brain in
selected neurological diseases.

2. Gut Microbiota
The gut microbiota can be defined as a collection of microorganisms, primarily bacte-
ria, which colonize the human gastrointestinal (GI) tract. The number of microorganisms
inhabiting the GI tract has been estimated to range between 1013 and 1014 , with an approx-
imate weight of 2 kg, outnumbering the cells in the whole human body [7]. The normal
human gut microbiota comprises two major anaerobic phyla, Bacteroidetes and Firmicutes.
Interestingly, the ratio between these two species may be a relevant biomarker of gut
dysbiosis in obesity [8]. Other identified types occur in small numbers and include some
species from the Proteobacteria, Cyanobacteria, Verrucomicrobia, and Actinobacteria phyla [9].
In order to explain the influence of microbiota on the human body, we can divide the
microorganisms that comprise the microbiota into “healthy” types, which exert a positive
impact on the body, and “unhealthy” types, which may cause harm to the host. By way of
illustration, it is believed that Actinobacteria, particularly Bifidobacterium sp., demonstrate
anti-inflammatory properties and exert a positive effect on the integrity of the intestinal
barrier [10]. By contrast, Clostridium difficile is the leading cause of diarrhea [11]. The
human gut microbiome contains around 4 million different genes, which is over 100 times
more than the total human genes present in the human genome [12].

2.1. Changes in the Composition of the Intestinal Microbiome through Life


It is believed that in normal conditions the fetus is germ-free, and the colonization
and further development of the microbiota commences at birth. Interestingly, the method
of delivery exerts a significant effect on the gut microbiota composition in newborns and
infants. A recent study revealed that children born through caesarian section display
diminished microbial diversity, primarily with decreased levels of Bacteroides or even
their absence, which perseveres throughout the years. Moreover, after some time, it is
only Bacteroides that differentiate the microbiota of babies delivered by C-section from the
microbiota of those delivered vaginally [13]. Furthermore, fecal microbiota in vaginally
delivered babies is broadly similar to that of their mothers, while in newborns delivered
through caesarian section there is only 41% resemblance [14].
Another factor that significantly affects the gut microbiota is the feeding method. It has
been demonstrated that breastfed infants show a higher level of dominance for Bifidobacteria,
while formula-fed babies may display an increased prevalence of Bacteroides [15]. New
studies reveal that environmental factors may cause modifications in the gut microbiota
throughout life. Due to these factors, considerable changes occur in a child’s microbiota
within the first 2–3 years of life. Following that, a certain degree of stabilization is achieved
in the third year of life [16,17], after which time the stability is gradually lost over the
years. Throughout life, the microbiota changes and diversifies, with a gradually increasing
proportion of Bacteroides and Clostridium. Centenarians, in particular, exhibit greater
abundance of Enterobacteriaceae [18,19].

2.2. Factors Influencing Gut Microbiota


Environmental factors such as diet, antibiotics, and infections may cause unhealthy
diversification of the microbiota and may lead to increased intestinal permeability known as
“leaky gut syndrome”. This might facilitate an improper and unnecessary immune response
that enhances inflammation that is already occurring in the digestive tract [20]. As diet has
a considerable impact on the gut microbiota composition, multiple studies have indicated
Int. J. Mol. Sci. 2021, 22, 10028 3 of 19

that it is the most important modifying factor [21,22]. The Western diet, characterized by an
increased dietary intake of saturated fat, sugar, and red meat, results in a shift to Firmicutes,
with a decreased abundance of Bacteroides and a disturbed Firmicutes/Bacteroidetes ratio [23].
In contrast, vegetarian or Mediterranean diets, characterized by the high consumption of
vegetables and fruit, healthy fats, and whole grains; moderate consumption of red wine;
and low consumption of red meat, result in healthier, more diverse microbiota. These
changes include an increase in Bacteroides, with a decrease in Firmicutes [24], which is
considered a neuroprotective factor associated with improved cognitive function and a
lower risk of Alzheimer’s disease (AD) [25–27].
Antibiotic consumption, and in particular antibiotic overuse, is one of the main factors
causing dysbiosis. As the majority of prescribed antibiotics are broad-spectrum antibiotics,
they eliminate both “good” and “bad” bacteria [28]. Furthermore, increased stress levels
caused by modern lifestyles may also exert a negative effect on the gut microbiota and its
diversity, and may result in an increased abundance of Clostridium [29]. However, there
are factors that can improve the gut microbiota such as exercise, which enhances microbial
diversity and promotes the presence of bacteria producing butyrate, known for its health-
promoting and anti-inflammatory properties, as well as its capacity to increase insulin
sensitivity [30–32]. Moreover, the consumption of polyphenols in green tea, red wine, and
other dietary products can also promote the growth of healthy and helpful bacteria [33].

2.3. Functions of the Gut Microflora


The main functions of the gut microbiota are nutrient metabolism and the synthesis
of vitamins. It also participates in the breakdown of drugs and other xenobiotics. During
these processes, the microbiota releases a wide range of metabolites and small molecules
that affect the body. The digestion of dietary fiber, a process that is possible only with the
participation of gut microbes, results in the production of short-chain fatty acids (SCFAs);
excretion of intestinal gases, such as methane and carbon dioxide; and the release of modest
amounts of lactate and alcohols [34]. Importantly, SCFAs are not only a source of energy
but they also affect the maturation of the microglia in the CNS. They may act as signaling
molecules within the CNS [6,35]. It has also been demonstrated that enteric microbes have
the ability to metabolize primary bile acids to secondary forms. These bile acids can also
perform metabolic activities, including their signaling role within the nervous system [36].
Any alterations in the composition, diversity, or excessive functionality of the gut
microbiota may constitute potential pathogenic factors. Importantly, normal gut microbiota
can stimulate the immune system and induce the release of proinflammatory cytokines
such as IL-1β, IL-6, or TNF-α [37]. As a result, the microbiota impacts the immune response,
thus protecting the host from pathogens [8]. Moreover, continual changes in epithelial
cells may serve as another protective mechanism within the gut that prevents enhanced
inflammation in the intestines [37]. Proliferation of intestinal epithelial cells is a clearing
mechanism that leads to the replacement of old cell layers and helps to isolate pathogens
from body cavities.

3. Gut–Brain Interplay
3.1. Metabolites Produced by the Gut Microbiota
The connection between the intestines and the CNS involves many pathways mediated
by various substances synthesized by microbes. The gut microbiota is capable of producing
and releasing some active metabolites that may serve as neuromediators participating in
communication with the CNS and affecting the brain. Short-chain fatty acids (SCFAs),
aromatic amino acids, and bile acids are the main substances from the microflora affecting
the brain. SCFAs consist mostly of acetate, butyrate, and propionate, which could be the
products of bacterial fermentation of carbohydrates. Interactions between these acids and
the gut may be mediated through binding to G-protein-coupled receptors [38].
(SCFAs), aromatic amino acids, and bile acids are the main substances from the
affecting the brain. SCFAs consist mostly of acetate, butyrate, and propion
could be the products of bacterial fermentation of carbohydrates. Interaction
these acids and the gut may be mediated through binding to G-protein-couple
Int. J. Mol. Sci. 2021, 22, 10028 [38]. 4 of 19

3.2. Direct and Indirect Effects of Gut Microbiota on the CNS


Neurotransmitters
3.2. Direct and Indirect and their
Effects of Gut Microbiota onprecursors
the CNS produced in the gut may also affe
els in the brain.
Neurotransmitters Besides
and their beingproduced
precursors obtained in from
the gutthe
may breakdown of food,
also affect their levels neurot
can also be produced by bacteria. By way of illustration, Escherichia
in the brain. Besides being obtained from the breakdown of food, neurotransmitters can coli can re
mine, serotonin,
also be produced by bacteria. and noradrenaline,
By way of illustration,while Lactobacilli
Escherichia produce
coli can release serotonin, GA
dopamine,
serotonin,choline,
and noradrenaline, while Lactobacilli produce serotonin, GABA, acetylcholine,
and histamine [39–41], which can influence the host brain. This mech
and histamine
been [39–41],
proposed which can influence
to play the host
an important rolebrain.
in theThis mechanism of
development hascertain
been neuro
proposed to play an important role in the development of certain neurological diseases,
eases, including Alzheimer’s disease, Parkinson’s disease (PD), depressive dis
including Alzheimer’s disease, Parkinson’s disease (PD), depressive disorders, and autism
autism spectrum disorders [42,43]. Furthermore, SCFAs are capable of indirect
spectrum disorders [42,43]. Furthermore, SCFAs are capable of indirectly affecting the gut–
brain axis the gut−brain
by inducing theaxis byofinducing
release some gutthe releasesuch
hormones, of some gut hormones,
as glucagon-like such as glu
peptide-1
(GLP-1) andpeptide-1 (GLP-1)
leptin, through and leptin, through
enteroendocrine enteroendocrine
cells. These enteric hormonescells.may
These enteric horm
interact
interact
with the vagus nervewith
and the
evenvagus nerve and
brain receptors even The
[44–46]. brain receptors
effects [44–46].
of the gut The on
microbiota effects of
the brain are presented in Figure 1.
crobiota on the brain are presented in Figure 1.

Figure 1. Pathways between


Figure the intestines
1. Pathways betweenand
the the central
intestines nervous
and system
the central (CNS).
nervous Figure
system illustrates
(CNS). how the gut mic
Figure illustrates
can interact with how
CNSthe through severalcan
gut microbiota pathways. Firstly,
interact with CNS through produced
through several metabolites
pathways. such asproduced
Firstly, through short-chain fatt
(SCFAs) [38]; secondly, directly
metabolites with
such neurotransmitters
as short-chain fatty acidsfor example
(SCFAs) [38];dopamine and serotonin
secondly, directly [39–41]; and lastly, ind
with neurotransmitters
influencing the release of enteric
for example hormones
dopamine [44–46].[39–41]; and lastly, indirectly influencing the release of enteric
and serotonin
hormones [44–46].
Interestingly, a number of studies indicate the contribution of SCFAs to t
Interestingly, a number of studies indicate the contribution of SCFAs to the mainte-
nance of physical barriers, such as the blood−brain barrier (BBB) or intestina
nance of physical barriers, such as the blood–brain barrier (BBB) or intestinal barrier by
impactingimpacting the tight
the tight junctions junctions
between between
cells [46–48]. cells [46–48].
Similarly, bile acidsSimilarly, bile
can activate acids can
recep-
tors in theceptors
host andin actthe host andparticles,
as signaling act as and
signaling particles,
can affect and can affect
barrier permeability barrier pe
[49,50].
[49,50].
Furthermore, Furthermore, lipopolysaccharide
lipopolysaccharide (LPS) produced
(LPS) produced by bacteria by bacteria
is able to influence BBBis able t
permeability
BBB bypermeability
inducing the release of inflammatory
by inducing cytokines
the release [51].
of inflammatory cytokines [51].
Int. J. Mol. Sci. 2021, 22, 10028 5 of 19

3.3. Relevance of Healthy Gut Microbiota


Experiments conducted on germ-free mice have confirmed the importance of normal
microbiota for many essential processes in the brain. One of them is the metabolism of
serotonin, a neurotransmitter responsible for mood and appetite, which is mostly produced
by enterochromaffin cells of the gastrointestinal tract [52]. Tryptophan is a precursor for
this neuromediator. Recent studies have demonstrated that germ-free mice had increased
tryptophan levels in plasma and decreased serotonin levels in the serum [53–55]. Moreover,
the rodents exhibited a decreased expression of a crucial neurotrophic factor, brain-derived
neurotrophic factor (BDNF), which is responsible for maintaining and promoting neuro-
genesis [56]. Furthermore, these animals also presented cognitive impairment, problems
with sociability, and depressive and anxious behaviors [55].
Although germ-free mouse models are the gold standard for gut microbiome re-
search, antibiotic-treated models constitute a more accessible and less expensive approach.
Antibiotics may be used to modify the gut bacteria, analyze changes in the microbiota,
and evaluate their impact on the brain [57]. It has been demonstrated that experimental
treatment with oral antibiotics not only induces intestinal dysbacteriosis, but also leads
to an imbalance in the gut–brain axis. Moreover, antibiotic-induced changes in microbial
composition also cause certain neurobehavioral alterations, such as increased anxiety and
“depressive-like” behaviors, as well as neuronal activation in different brain regions of
mice [58].

4. Gut and Neurological Diseases


Recent data suggest that neuroinflammation may be a pathogenic factor in several
neurodegenerative disorders. In a neuroinflammatory state, activation of the microglia
and the release of proinflammatory proteins, such as TNF-α, IL-6, or MCP-1, as well as
reactive oxygen species by glial cells and resident macrophages, might result in chronic
neuroinflammation [20,59]. As the gut microbiota is believed to contribute to various
pathogenic pathways, there are a growing number of studies linking changes in the healthy
microbiome to the development of a number of neurological diseases, including those
with neurodegenerative etiology, as well as to certain neuropsychiatric disorders [3,60,61].
Likewise, diminished microbiota diversity throughout life may be connected with neurode-
generation [62].
Although loss of intestinal epithelial cell integrity and chronic inflammation seem to
be the main consequences of changes in the gut microbiota, neuroinflammation as well as
neurodegenerative and neuropsychiatric disorders may also be the result of these changes.
The importance of a healthy gut microbiota and its diverse composition for normal brain
function has been confirmed in studies on rodent models [63]. It has been demonstrated
that the presence of Lactobacillus bacteria exerts a positive effect on the brains of rats with
experimental cerebral ischemia reperfusion injury via the inhibition of neural cell apoptosis
and the reduction of oxidative stress through the downregulation of the TLR4/NF-kB
signaling pathway [63]. As described in previous sections, a number of factors, including
diet, stress, infections, and antibiotic use, may result in gut microbiota dysbiosis. The
connection between changes in the gut and CNS disorders is shown in Figure 2, with
further description in later sections.
Int. J.J.Mol.
Int. Mol.Sci.
Sci.2021, 22,x10028
2021,22, FOR PEER REVIEW 66 of
of 19
19

Figure 2.
Figure Schematic description
2. Schematic description of of disturbances
disturbancesin in the
the intestinal
intestinal microbiota
microbiota and
and diseases
diseases ofof the
the CNS.
CNS. The
The figure
figure depicts
depicts
major
major factors, such as stress [29], diet [21], infections, and antibiotic intake [28], which can promote dysbiosis in the gut
factors, such as stress [29], diet [21], infections, and antibiotic intake [28], which can promote dysbiosis in the gut
microbiota.
microbiota. Changes
Changes inin the
thecomposition
compositionofofthethemicrobiome
microbiome lead
lead to to
gutgut permeability,
permeability, influencing
influencing systemic
systemic inflammation
inflammation and,
and, as a result,
as a result, may may induce
induce diseases
diseases of theofCNS.
the CNS.

4.1.Alzheimer’s
4.1. Alzheimer’sDisease
Disease
Alzheimer’s disease is the
Alzheimer’s themost
mostprevalent
prevalentneurodegenerative
neurodegenerative disorder.
disorder. ADADis aisprogres-
a pro-
sive disease
gressive whose
disease whosefirst first
clinical symptoms
clinical symptoms appear decades
appear after after
decades the onset of pathological
the onset of patho-
changes
logical in the brain,
changes in thethus making
brain, thus older
making individuals the most affected
older individuals the mostage group.age
affected Hallmarks
group.
of the disease include progressive accumulation of amyloid-beta
Hallmarks of the disease include progressive accumulation of amyloid-beta (Aβ) plaques, (Aβ) plaques, formed
following
formed the cleavage
following of amyloid
the cleavage precursor
of amyloid protein (APP),
precursor proteinin(APP),
the brain andbrain
in the neurofibrillary
and neu-
tangles (NFTs),
rofibrillary which
tangles consist
(NFTs), of hyperphosphorylated
which tau protein tau
consist of hyperphosphorylated [64].protein [64].
The etiology
The etiologyof of AD
AD hashas not
not been
been fully
fully elucidated,
elucidated, butbut aa number
number of of factors
factors influence
influence
the pathogenesis of the disease. Proposed hypotheses on the origin
the pathogenesis of the disease. Proposed hypotheses on the origin of AD includes of AD includes gradual
grad-
accumulation of Aβ in the AD brain, followed by progressive
ual accumulation of Aβ in the AD brain, followed by progressive deposition of the deposition of the Tau protein.
Tau
AnotherAnother
protein. hypothesis suggests
hypothesis the rolethe
suggests of soluble oligomers
role of soluble of Aβ and/or
oligomers Tau protein
of Aβ and/or Tau pro- as
the most harmful factors affecting the brain tissue [65,66]. Moreover,
tein as the most harmful factors affecting the brain tissue [65,66]. Moreover, the contribu- the contribution
of the
tion of immune
the immune system to AD
system pathogenesis
to AD pathogenesis hashas
also been
also beensuggested.
suggested.Insoluble
Insolubledeposits
depos-
of of
its AβAβ may
may beberecognized
recognizedbybythe theimmune
immunesystem systemas asaa foreign
foreign material, triggering the
material, triggering the
inflammatory cascade, which leads to neuronal damage. Amyloid
inflammatory cascade, which leads to neuronal damage. Amyloid plaques and NFTs gen- plaques and NFTs
generate
erate inflammation
inflammation withinwithin the brain,
the brain, primarily
primarily throughout
throughout activation
activation of theofmicroglia
the microglia
and
and astrocytes.
astrocytes.
The neuroinflammation hypothesis in the pathogenesis of AD is associated with cer-
The neuroinflammation hypothesis in the pathogenesis of AD is associated with cer-
tain gut microbiota alterations. Neuroinflammatory processes occur with ongoing systemic
tain gut microbiota alterations. Neuroinflammatory processes occur with ongoing sys-
inflammation, which may further intensify neuroinflammation [20,67,68]. Furthermore, it
temic inflammation, which may further intensify neuroinflammation [20,67,68]. Further-
has recently been suggested that AD is initiated is the gut, and not the brain, from where
more, it has recently been suggested that AD is initiated is the gut, and not the brain, from
it subsequently proceeds to the brain. The hypothesis was confirmed in a study by Sun
where it subsequently proceeds to the brain. The hypothesis was confirmed in a study by
et al., in which Aβ oligomers were injected into the wall of the gastrointestinal tract of
Sun et al., in which 1–42Aβ1–42 oligomers were injected into the wall of the gastrointestinal tract
mice. Initially, the oligomers of Aβ1–42 were internalized into enteric cholinergic neurons,
Int. J. Mol. Sci. 2021, 22, 10028 7 of 19

but after one year of observation, relocated amyloid was found in the brain of rodents,
which exhibited gastrointestinal tract dysfunction and cognitive deficits [69]. The findings
confirmed that intra-gastrointestinal oligomers of Aβ1–42 could disturb not only enteric
function, but also induce AD in the studied animal model of AD. Moreover, as Aβ from the
periphery could contribute to the Aβ burden in the brain, these results may support the
hypothesis that Aβ has prion-like properties [70]. Furthermore, the discovery of amyloid
migration may prove the connection between the gut and neuroinflammation in AD.
Recent data also link changes in the gut microbiota with AD [71]. Changes in the
phyla Bacteroidetes and Firmicutes, unrelated to age, have been demonstrated in mouse
models of AD [72–74]. The results from animal models have also been confirmed in hu-
man studies using gene sequencing techniques. Human fecal samples showed similar
results, with alterations in the microbiome, including decreased Firmicutes and Bifidobac-
terium, but increased Bacteroidetes in AD patients [75]. Another study conducted among
cognitively impaired patients with brain amyloidosis revealed an increased abundance of
Escherichia/Shigella, known as pro-inflammatory bacteria, with a simultaneous decrease in
anti-inflammatory Eubacterium rectale in comparison with healthy controls and individuals
without amyloidosis in PET imaging. Additionally, in these patients, a positive correlation
was found between increased blood levels of pro-inflammatory cytokines, such as IL-1β
and CXCL2, and a component of the inflammasome complex NLR family pyrin domain
containing 3 (NLRP3), and abundance of Escherichia/Shigella [75,76]. When the CSF levels of
various AD biomarkers were investigated in a study by Vogt et al., a significant relationship
was revealed between YKL-40 concentrations, and an increased abundance of Bacteroides
and decreased presence of Turicibacter [75].
The connection between the gut and the brain in AD has also been confirmed in stud-
ies exploring the transfer of healthy microbiota from wild type mice to mouse models of
AD. Normalization of the gut microbiome led to the reduced formation of Aβ plaques and
neurofibrillary tangles, decreased glial reactivity, and improved cognitive performance [77].
Moreover, the results of the study are in line with human studies showing that transplan-
tation of healthy microbiota to AD patients suffering from Clostridium difficile infection
resulted in improved cognitive function, as demonstrated by the Mini-Mental State Exam
(MMSE) score [78,79]. On the other hand, it was revealed that pathological changes in
microbiota composition, e.g., a decrease in Bifidobacteria and the overgrowth of Clostridium
difficile, may stimulate a shift in the expression of proinflammatory molecules [3,18].
Interestingly, a specific kind of amyloid has also been identified on the bacterial
cell surface. The first discoveries of this protein were described in Escherichia coli curli,
the proteinaceous components of bacterial extracellular fibers [80]. Similar abilities to
produce bacterial amyloids have been discovered in other species, such as Staphylococcus,
Streptococcus, Salmonella, and Klebsiella. Bacterial amyloid plays an important role in the
building of a microbial biofilm, which prevents gut bacteria eradication [81]. Although the
primary structures of bacterial and CNS amyloids are not similar, their tertiary structures
reveal a significant similarity [82]. As a result, the presence of bacterial amyloid in the gut is
thought to affect the immune system, consequently intensifying reaction to the formation of
neuronal amyloids [81]. Knowing that bacterial amyloids can cross physiological barriers,
it has been suggested that they contribute to the development of AD [83].
Lipopolysaccharide (LPS), which is known for its pro-inflammatory properties, is
present in the membrane of Gram-negative bacteria such as Bacteroides. It has been proven
that LPS is capable of generating inflammation and may mediate the release of many proin-
flammatory cytokines through Toll-like receptor-4 (TLR-4) [84]. This receptor contributes
to the activation of the microglia at the earliest stages of Aβ deposition in the brain, which
has been demonstrated in mouse models of AD with TLR-4 deletion, showing enhanced
amyloidosis [85].
Another rodent study on bacterial LPS revealed that following the intraperitoneal
injection of LPS, mice showed elevated hippocampal levels of Aβ1-42 with simultane-
ous cognitive defects [86]. The importance of this bacterial endotoxin for amyloid fibril
Int. J. Mol. Sci. 2021, 22, 10028 8 of 19

formation has been confirmed by in vitro experiments that demonstrated that LPS from
Escherichia coli could potentiate Aβ organization in compact fibrils. These results confirm
that LPS is a key factor in the kinetics of Aβ fibrillogenesis [87]. Furthermore, a significant
increase in LPS levels has been observed in the brain samples obtained from AD patients
that were co-localized with Aβ plaques, suggesting that this bacterial molecule has the abil-
ity of to pass through physiological barriers into the brain [88,89]. Consequently, elevated
LPS levels have been found in the plasma of AD patients [90]. This finding is consistent
with the aforementioned hypothesis of the “leaky gut syndrome” and loss of integrity of
intestinal and BBB barriers with age, which contribute to neuroinflammation. Furthermore,
knowing that Bifidobacterium and Lactobacillus exert a beneficial effect on LPS levels and
barrier integrity, a decrease in the abundance of these bacteria may play a significant role
in the development of AD [91].

4.2. Parkinson’s Disease


Parkinson’s disease is the second most common neurodegenerative disorder [92],
which may also be linked to disturbances in the brain–gut–microbiota axis. PD is a chronic,
progressive disease characterized by both motor and non-motor features. It mostly occurs
in men and women over 40 years of age and its incidence increases with age [93,94]. Tremor,
bradykinesia, and postural instability are the primary motor symptoms, while cognitive
decline, sleep disturbances, depression, and anxiety are the main non-motor symptoms [95].
The causes of PD include deterioration of the dopaminergic neurons in the extrapyramidal
tract of the midbrain, which is believed to be responsible for motor dysfunction [96]. The
histopathological hallmark of PD is the presence of misfolded, insoluble α-synuclein,
which may aggregate into Lewy bodies in neurons, thus inducing neurodegeneration [97].
Similarly to AD, neuroinflammation also plays a role in the pathophysiology of PD, which
is evident in microgliosis and astrogliosis [98].
Non-motor symptoms attributable to the digestive system are particularly common
in patients with PD. Hypersalivation, which is the result of impaired swallowing, and
constipation, caused by motility changes, are the most common dysfunctions of the gas-
trointestinal tract [99]. These observations support the hypothesis that PD may originate
within the gut. Moreover, constipation may be linked to enteric nervous system degen-
eration caused by the aggregation of alpha-synuclein, increased intestinal permeability,
and local inflammation [100]. It has also been observed that there are strong associations
between changes in intestinal motility, which may predate neurological manifestations
of the disease by a number of years, a well as a subsequent diagnosis of PD. It has been
demonstrated that constipation significantly increases the risk of developing PD [101,102].
It has been suggested that disturbed gut microbiota, which is responsible for intestinal
motility, increased permeability, and chronic local inflammation, could be considered an
important factor in the pathophysiology of PD. As dysfunction of the gastrointestinal
system is a characteristic feature of PD, the gut microbiota composition in PD patients
has also been investigated. It has been shown that patients suffering from PD have an
increased abundance of Enterobacteriaceae, which was positively correlated with postural
instability [103]. In contrast, another study demonstrated that certain families of healthy
bacteria, such as Prevotellaceae and Lachnospiraceae, were reduced in the fecal samples in
PD patients [104]. Prevotellaceae are known for participating in the production of mucins,
which play a crucial role in maintaining intestinal permeability. Thus, their reduced
presence might be linked to the “leaky gut syndrome” [105]. Lachnospiraceae, such as
Blautia, Coprococcus, and Roseburia, participate in the production of SCFAs which are known
for their anti-inflammatory and gut protective properties, which may also contribute to
altered permeability [104,106].
Interestingly, studies on animal models of PD have demonstrated a connection be-
tween motor deficits, neuroinflammation, and the gut microbiota. Mouse models of PD
have shown a disturbed gut microbiota, with a decreased presence of Firmicutes and
Clostridiales and an increased abundance of Proteobacteria and Enterobacteriales. Experi-
Int. J. Mol. Sci. 2021, 22, 10028 9 of 19

mental transplantation of fecal microbiota not only ameliorated microbiota composition


and reduced SCFAs concentration in mouse feces, but also diminished the activation of
microglia and astrocytes within their brains [107].
A recent study demonstrated that providing germ-free mice with selected microbial
metabolites resulted in neuroinflammation and detectable PD-like physical symptoms [108].
Consequently, transplantation of the microbiota from healthy individuals to mice over-
expressing α-synuclein resulted in diminished motor impairments in comparison with
the animals treated with microbiota from PD patients [108]. Additionally, a recent study
conducted on aged rats with aggregated α-synuclein in the intestinal submucosal plexus,
which were exposed to transgenic E. coli that produced bacterial amyloids, displayed a
significant reaction to bacterial curli. It has been demonstrated that the production of
α-synuclein within the intestines and its accumulation in the brain of examined rodents is
intensified, which further prompts microgliosis and astrogliosis. Moreover, the rat brains
showed an increased expression of TLR-2, TNF-α, and IL-6, which may indicate that amy-
loids produced by bacteria provoke α-synuclein aggregation and, as a result, an innate
immune system response [109,110]. Another microbial metabolite, LPS, might be a crucial
factor in the pathophysiology of PD, similarly to AD. LPS-induced inflammation in rodent
models of PD activate the microglia, which results in dopaminergic neuron damage and
loss [99,111].
It is worth mentioning that a large number of PD patients are infected with Helicobacter
pylori. For a number of years, the presence of gastric ulcers, which are triggered by H. pylori,
has been linked to PD [112]. On the other hand, this bacteria is also known for impairing the
absorption of levodopa, a key drug in the treatment of motor aspects of PD [113]. It has been
demonstrated that comorbidity between PD and H. pylori infection is related to more severe
manifestations of PD and more significantly impaired motor function [114]. Interestingly,
the existence of H. pylori infection might also increase the incidence of PD [112,115].

4.3. Multiple Sclerosis


In contrast with AD and PD, multiple sclerosis (MS) is a disease affecting mostly young
adults, particularly women [116]. It is a demyelinating CNS disease with an inflammatory
component. MS is characterized by chronic inflammation both in white and grey matter
of the brain and the spinal cord, which causes destruction of the myelin that covers the
neurons [117]. Although the mechanisms underlying MS are not fully understood, it
is postulated that malfunction of the immune system is the most probable cause of the
disease [118]. Apart from chronic inflammation, altered selectivity of BBB in MS brain
has been observed. This state facilitates the migration of immune cells (mostly T-cells)
to the nervous system and penetration of the brain. Following infiltration into the CNS,
T-cells start to recognize myelin as a trigger to the immune system, which causes enhanced
inflammation and results in demyelination [117,119].
The pathophysiology of MS may be also linked to genetic and environmental fac-
tors [120]. Obesity in early life [121], decreased vitamin D levels in the blood, and in-
sufficient exposure to sunlight [122], as well as smoking [123], are the established and
most frequently described causes. All of these aspects can also indirectly affect the gut
microbiota. As microbes may control immunity by regulating T-cells, the gut microbiota
has received attention as an important factor in MS pathology.
It has been demonstrated that there are considerable differences between stool samples
from MS patients and those from healthy controls. MS samples revealed decreased levels
of Bacteroidetes, Clostridium, Fecalibacterium, and Prevotella taxa (the last one produces propi-
onate, which is a SCFA) [124–126]. Moreover, increased abundance of Methanobrevibacter
and Akkermansia muciniphila has been observed in different types of MS [127]. In addition,
transplantation of the microbiota from MS patients to germ-free mice resulted in intensified
experimental autoimmune encephalomyelitis (EAE), an animal type of demyelinating
diseases, in contrast with germ-free mice treated with healthy microbiota [128]. These
Int. J. Mol. Sci. 2021, 22, 10028 10 of 19

results suggest the potential involvement of the microbiota in the development of MS and
its impact on disease progression.
It has been revealed that various metabolites produced by commensal Clostridium,
such as SCFA butyrate, propionate, and acetate, may differentially induce T-regulatory
cells, affecting the balance between pro- and anti-inflammatory cells in MS patients [129].
Moreover, studies among MS patients treated with vitamin D have revealed an altered com-
position of the gut microbiota, with elevated levels of Faecalibacterium, an anti-inflammatory
bacteria that produces butyrate [125]. These results may also confirm the positive effect of
vitamin D supplementation for MS patients.

4.4. Major Depressive Disorder


Major depressive disorder (MDD) is a mental disorder and is one of the leading
causes of disability, morbidity, and mortality in developed countries. In 2017, more than
264 million people worldwide were affected by MDD [130]. The main symptoms of MDD
include low mood, difficulties in concentration, fatigue, appetite alteration, and digestive
and sleeping problems. For an appropriate diagnosis, the symptoms must be present
continuously for a minimum of a 2-week period [131]. The pathophysiology of MDD
is still not fully understood. However, it is suggested that deficiency in monoamine
neurotransmitters, such as serotonin, noradrenaline, and dopamine, may be the key cause
of the disorder [132]. Another cause of the disease may be systemic inflammation, with
elevated blood cytokine levels, which also demonstrates that depression is a systemic
disease. Systemic inflammation also leads to neuroinflammation and the activation of
microglia and astrocytes, contributing to the development of MDD, affecting behavior and
emotions [133–136].
As previously described, the gut and the brain communicate in a bidirectional manner
and interactions between these organs are important for the development of the CNS.
Moreover, the gut microbiota exerts a significant impact on the CNS and may act as a
mediator in communication between the gut and the brain. Studies on germ-free mice show
that changes in the gut microbes may promote anxiety-like behaviors in these animals,
whose effects persist after colonization with normal intestinal microbiota [137]. Interactions
between the gut and the brain are essential to the development of stress systems within
the CNS, with a possible critical time window, after which reconstitution of the microbiota
may not be able to normalize the behavioral phenotype. Another study in which the
microbiota from depressed patients was transferred to rats, revealed enhanced depression-
like behaviors in these animals, with disturbed tryptophan metabolism [138].
The existing body of knowledge of the gut microbiota and microbiota-released molecules
have prompted researchers to consider disturbances in the gut–brain axis as a new aspect of
MDD pathology. Some studies have revealed that gut microbiota composition in depressed
patients was significantly altered in comparison with healthy controls. MDD patients had
an increased abundance of Bacteroidetes and Proteobacteria, with a decrease in Firmicutes,
Bifidobacterium, and Lactobacillus [139–141]. Similar results have been obtained from ani-
mal studies, with an enhanced proportion of Bacteroidetes and a decreased proportion of
Firmicutes in various depression models [142–144].
Moreover, it has been identified that the presence of Coprococcus, a bacterial species
that produces a beneficial SCFA, butyrate, in the patient’s gut, is connected with indicators
of a higher quality of life, such as perceived health status, physical functioning, vitality,
emotional well-being, and social functioning. Interestingly, Coprococcus was found to
be diminished in MDD patients [145]. A recent meta-analysis demonstrated a decline
in depression scores in MDD patients after restoration of the microbiota with probiotics,
which also confirms essential connections between depression and the gut microbiota [146].

4.5. Autism Spectrum Disorder


Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized
by impairments in communication and social interactions, repetitive and stereotyped
Int. J. Mol. Sci. 2021, 22, 10028 11 of 19

behaviors, and restricted interests [147]. It is commonly diagnosed in infants, mostly


boys, between 1 and 2 years of age [148]. However, as ASD consists of a broad range
of conditions, it may be diagnosed later in life, also in adults [149]. Although the exact
cause of ASD is not fully understood and is highly complicated, it is linked to genetic and
environmental input [150].
Apart from psychological aspects, ASD patients exhibit gastrointestinal symptoms,
such as diarrhea and/or constipation, as well as abdominal pain [151]. Recognizing the
relationship between the gut and the brain, researchers started investigating the micro-
biota of ASD patients. It has been demonstrated that people suffering from ASD show
an increased abundance of Clostridium and Lactobacillus species [152,153]. Furthermore,
Bacteroidetes dominate in the intestines, with a concurrent decrease in Firmicutes [154].
Another study revealed decreased levels of beneficial species Prevotella and Coprococcus
in the intestinal microflora of autistic children in comparison with healthy controls [155].
Similar results have been obtained in animal studies. It has been demonstrated that the
valproic acid rat model of autism exhibits changes in the diversity and number of species,
and has a composition of gut bacteria similar to those observed in human autism [156].
Moreover, significantly increased serum levels of bacterial LPS, as well as IL-1beta and
IL-6, the biomarkers of inflammation, have been observed in adult autistic patients in com-
parison with healthy controls. Furthermore, these results were inversely correlated with
socialization scores [157]. These findings confirm the role of microbiota in ASD. However,
the significance of low-grade endotoxemia in the pathophysiology of autism needs further
investigation.
Importantly, it seems that exposure in utero to inflammation resulting from distur-
bances in the maternal gut microbiota may increase the probability of ASD occurrence
in children. This has been confirmed in studies on mouse models, which indicate that
changes in the maternal gut microbiome promote neurodevelopmental abnormalities in
mouse offspring [158].

5. Conclusions
In recent years, the gut microbiota and its importance for the functioning of the human
body has generated considerable interest among researchers, although we still do not know
whether alerations in the microbiota trigger pathological changes or coexist with them.
However, a balanced composition of the gut microbiota and the production of various
bacterial metabolites have proven their profound significance for host health, including for
the CNS. The gut–brain axis, which may be defined as a complex interplay between the
function of the gastrointestinal system, including the enteric nervous system, the activity
of our intestinal microbes, and the CNS, may influence the development of various brain
diseases.
The body homeostasis may be affected by the pathological shift in the microbiome
and its altered metabolism, thus promoting the development of different neurological
and neuropsychiatric disorders. These gut microbiome-related diseases of the CNS in-
clude neurodegenerative disorders, such as Alzheimer’s disease, Parkinson’s disease, and
multiple sclerosis, as well as depression and autism spectrum disorders. Therefore, the
intestinal microflora can be seen as an important factor in the development of CNS and the
progression of various neurological diseases.
Although the gold standard in microbiota analysis is a fecal material examination, a
multiplatform analysis of its metabolites may also provide valuable information regarding
the state of the microbiome. Moreover, the gut microbiome could be a potential new target
for the treatment of these diseases. Prevention of gut dysbiosis by probiotics may also
provide protection from the disorders described above. A summary of the microbiota
changes observed in neurological diseases is presented in Table 1.
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 12 of 19
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 12 of 19
Although the gold standard in microbiota analysis is a fecal material examination, a
multiplatform
Although analysisthe gold of its metabolites
standard may also
in microbiota provide
analysis is a valuable information
fecal material regard-a
examination,
Although
ing the
multiplatform the
the gold
state ofanalysis standard
microbiome.
of in microbiota
Moreover,
its metabolites may analysis
thealso
gut is a valuable
microbiome
provide fecalcould
material
be aexamination,
potential
information newa
regard-
target
Int. J. Mol. Sci. 2021, 22, 10028 ing Although
multiplatform
thefor the
theoftreatment
state gold
analysis standard
of
ofits
the microbiome. these in
metabolitesmicrobiota
diseases.may
Moreover, analysis
thealso
gut provide
Prevention is a fecal
of gutvaluable
microbiome material examination,
couldinformation
dysbiosis by aprobiotics
be newa12 of 19
regard-
potential may
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
multiplatform analysis of 12 of 19
ing
alsothe
target state
provide
for oftreatment
theprotection ofits
the microbiome.
from metabolites
the
these diseases.may
Moreover,
disorders thealso
gut provide
described
Prevention microbiome
above.
of gutA valuable
couldinformation
summary
dysbiosis be
by the regard-
ofaprobiotics
potential new
microbiota
may
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
ing the state oftreatment
theinmicrobiome. Moreover, 12 of 19
target
changes
also
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
for theprotection
observed
provide of these
neurological
from diseases.
the disorders isthe
diseases Prevention gut microbiome
presented
described ofingut
above. Table could
Adysbiosis
1.
summary be
byofaprobiotics
potential
the new
may
microbiota
12 of 19
target forobserved
also provide
changes theprotection
treatment of these
from
in neurological diseases.
diseases Prevention
the disorders described
is presented ofingut
above. Adysbiosis
Tablesummary
1. byofprobiotics may
the microbiota
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 12 of 19
also
changes Table
provide 1.
observedChanges
protectionin in the
from microbiota
the
neurological in
disorders
diseases neurological
described
is diseases.
presentedabove.
in A
Tablesummary
1. of the microbiota
TableAlthough
1. Changesthe gold
in the standardininneurological
microbiota microbiotadiseases.
analysis is a fecal material examination, a
changes
Table observed
multiplatform
Although
1. Changes the
in in
analysis neurological
gold
the of its
standard
microbiota in diseases
metabolites
in microbiota
neurologicalis presented
may also
analysis
diseases. in
provideis Table
fecal1.material
a valuable information regard-
examination, a
Typeofof Disease Bacteria Direction of material
Changes Author
Table
ing the1. Type
Although
Changes
state
multiplatform Disease
the
ofanalysis
the gold
in the standard
microbiota
microbiome.
of in Bacteria
inneurological
microbiota
Moreover,
its metabolites may Direction
analysis
thediseases.
gut
also microbiome
provide of
is a valuable
fecal Changes
could Author
be aexamination,
potential
information newa
regard-
Table
target
ing the Type
Although
multiplatform
1.for
Changes
theof
state of Disease
the gold
analysis
in the
treatment
the standard
of
ofits
microbiota
these
microbiome. in in
metabolites Bacteria
microbiota
may
Bacteroidetes
neurological
Bacteroidetes
diseases.
Moreover, also Direction
analysis
thediseases.
Prevention
gut of isgut
provide
microbiome of
a valuable
fecal Changes
material
dysbiosis
could Author
examination,
information [75,76]
by aprobiotics
be potential newa
regard-
[75,76]
may
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
ing the Type
multiplatform
state of
of Disease
analysis
the of its
microbiome. metabolites Bacteria
Moreover, may the also
gut Direction
provide
microbiome of
valuable Changes
could information
be a Author
12new
of 19
regard-
potential
also provide
target for theprotection
treatment from
of these Bacteroidetes
the Escherichia/Shigella
disorders
diseases. described above.
Prevention of gutAdysbiosis
summary by [75,76]
ofprobiotics
the microbiota
[76] may
Type of Disease Bacteria
Bacteroidetes
Escherichia/Shigella Direction of Changes Author
[75,76]
ing
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW the
target
changes
also Alzheimer’s
state
forobserved
provide
Alzheimer’s theDisease
oftreatment
theprotection microbiome.
of these
inDisease
neurological
from Moreover,
diseases.
diseases isthe
the Escherichia/Shigella
disorders gut microbiome
Prevention
presented
described ofingut
above. Table could
Adysbiosis
1.
summary be
byofaprobiotics
the 12[76]
potential new
may
of 19
microbiota
[76]
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW Firmicutes
Bacteroidetes [75]
12 of 19
[75,76]
target
also Alzheimer’s
for
provide
changes the protection
observed DiseaseDisease
treatment of
from these
the diseases.
disorders Prevention
described
Escherichia/Shigella
in neurological diseases of
above.gut A
is presented in Table 1. dysbiosis
summary byof probiotics
the may
microbiota
[76]
Alzheimer’s Firmicutes
Firmicutes
Eubacterium [75]
12[75]
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
also
Table provide
changes
Although
1. Changesprotection
observed in in
the gold
the from thein
neurological
standard
microbiota disorders
diseases is rectale
described
Escherichia/Shigella
in microbiota
neurological
Firmicutes presentedabove.
analysis
diseases. in aA
is Tablesummary
fecal1.material [76]
of examination,
[76]
[75]
of 19
the microbiota a
changes Alzheimer’s
observed Disease
in neurological Eubacterium
Enterobacteriaceae
diseases is rectale
presented in Table 1. [76]
[103]
multiplatform
TableAlthough
1. Changesthe analysis
in the of its
microbiotametabolites
in may also provide valuable information regard-
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW gold standard inneurological
Firmicutes
Eubacterium
Eubacterium
microbiota diseases.
rectale
rectale
analysis is a fecal material [75]
12[76]
[76]
examination, a
of 19
Table
ing the1. Type
Changes
Although
state ofthe
of Disease
in the
the goldmicrobiota
standardin
microbiome. Bacteria
inneurological
microbiota
Enterobacteriaceae
Moreover,
Helicobacter the gut
pylori
Direction
diseases.
analysis is a fecal
microbiome of Changes
material
could be a Author
examination,
[103]
potential
[114] newa
multiplatform
Type analysis of its Eubacterium
metabolites may
Enterobacteriaceaerectale
also provide valuable information [76]
regard-
[103]
Table 1.for
targetAlthoughtheof
Parkinson’s
Changes
multiplatform Disease
in Disease
the
analysis
treatment
the goldmicrobiota
of
ofits
these
standard
Bacteria
Bacteroidetes
indiseases.
inneurological
metabolites may
Enterobacteriaceae
microbiota also
Prevention Direction
diseases.
provide
analysis
of Changes
a valuable
of isgut dysbiosis
fecal information Author
[75,76]
by probiotics
material regard-
12[103]
may
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
ing the Type
state of
Parkinson’sof Disease
the microbiome.
Disease
Helicobacter
Prevotellaceae
Bacteria
Moreover, pylori
the
Enterobacteriaceae gut Direction
microbiome of
could be aexamination,
Changes [114]
[104]
Author
potential
[103] new a
of 19
ing
alsothe state protection
provide
multiplatform ofanalysis
the microbiome.
from
of its the Helicobacter
Bacteroidetes
Moreover,
disorders pylori
thealso
gut provide
described
Escherichia/Shigella
metabolites may microbiome
above. A couldinformation
summary
valuable beofa the [114]
[75,76]
potential new
microbiota
[76]
regard-
Type
targetAlthough
for theof
Parkinson’s
Alzheimer’s Disease
treatment
Disease
Disease Prevotellaceae
of theseHelicobacterBacteria
diseases. Prevention
Bacteroidetes
Lachnospiraceae
pylori Direction
of is
gut of Changes
dysbiosis [104]
Author
by probiotics
[75,76]maya
[104][114]
target
changes
ing the forobserved
state
Parkinson’sthe
theoftreatment gold
in standard
neurological
theDisease
microbiome.
Disease in microbiota
Helicobacter
of these Escherichia/Shigella
diseases.
diseases
Moreover, isthe
Prevotellaceae analysis
pylori
Prevention
presented
gut a fecal
ofingut
Table
microbiome 1.material
dysbiosis
could be examination,
[114]
by aprobiotics
[104]
[76]
potential may
new
also provide protection
Parkinson’s from the Firmicutes
disorders described
Bacteroidetes
Lachnospiraceae above. A summary of the [75]
microbiota
[75,76]
[104]
also Alzheimer’s
multiplatform
provide protectionDisease
analysis of
fromits the Escherichia/Shigella
Methanobrevibacter
metabolites
disorders may also
described
Prevotellaceae provide
above. valuable
A summary information
of the [76]
[127]
regard-
microbiota
[104]
target
changes forobserved
the treatment
Alzheimer’s
Although Disease
in of these Eubacterium
neurological diseases. Prevention
Lachnospiraceae
Firmicutes
Prevotellaceae
diseases isthe
presented ofingut dysbiosis
Table by probiotics
1.material [104]
[75] maya
[76][104]
ing
Table
also the
changes 1. state of the
observed
Changes
provide the gold
in in
protectionthe
standard
microbiome.
neurological
microbiota
from the in
in microbiota
Moreover,
Akkermansia
diseases
neurological
Firmicutes
disorders
analysis
rectale
Escherichia/Shigella
Methanobrevibacter gut
ismucini-
presented
diseases.
described
is a fecal
microbiome
in Table
above. A could
1.
summary beofaexamination,
the [76]
[127]
potential
[75] new
microbiota
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW Alzheimer’s
multiplatform Disease
analysis of its Lachnospiraceae
Methanobrevibacter
metabolites
Eubacterium may also provide
rectale valuable information [104]
12
[127] of 19
regard-
[76]
target for the treatment of these Enterobacteriaceae
diseases.
Akkermansia Prevention
Firmicutes
phila
Lachnospiraceae mucini- of gut dysbiosis by [103]
probiotics
[75] may
[104]
changes
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
Table
ing the observed
1. Type
Changes of the in
in the
Diseaseneurological diseases
Eubacterium
microbiota Akkermansia
in neurological
Bacteriais
Methanobrevibacter presented
rectale
diseases. in Table
Direction 1.
of Changes [76]
12
Author
[127] of 19
also
Table
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 1. state
provide
Changesof
protection microbiome.
in the from thein
microbiota Moreover,
neurological
Helicobacter
phila
the
Enterobacteriaceae
disorders gut microbiome
mucini-
described
Bacteroidetes above.
diseases.
pylori
could
A summary beofa the
potential
[103] new
microbiota
[114]
12 of 19
[124]
target Multiple
for theof
Parkinson’s Sclerosis
treatment
Disease Eubacterium
Enterobacteriaceae
of theseAkkermansia
diseases. rectale
Prevention ofingut dysbiosis [127]
by probiotics[76]
[103][127]
may
changes
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW observed
Type
Table 1.Multiple
Changes in in
theneurological
Disease Bacteroidetes
diseasesphila
microbiotaMethanobrevibacter
Bacteria
inHelicobacter
neurologicalismucini-
presented
pylori Table
Direction
diseases. 1. Changes
of [75,76]
Author
[114]
12 of 19
Type
also provide of Sclerosis
Disease
protection Prevotellaceae
Bacteroidetes
Clostridium
Bacteria
Enterobacteriaceae Direction of Changes [104]
[124]
[127]
[124]
Author
[103]
Parkinson’s
Although gold from
theDisease the Escherichia/Shigella
standard disorders described
in Bacteroidetes
microbiota
Helicobacter
phila above.
analysis
pylori is a A summary
fecal material of examination,
the microbiota
[114]
[76]
[124] a
Multiple Sclerosis
Parkinson’s Disease Bacteroidetes
Prevotellaceae
Akkermansia
Lachnospiraceae
Clostridium [75,76]
[104]
[104]
[124]
changes
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
Table Alzheimer’s
observed
Type
multiplatform
Although
1.Multiple
Changes the
in Disease
in
ofanalysis
Diseaseneurological
gold
the of
standard
microbiota in diseases
its metabolites
in Bacteria
microbiota
Helicobacter
neurologicalis
Bacteroidetes
Fecalibacterium
may
Prevotellaceae presented
also
analysis
pylori
diseases. in
is Table
Direction
provide a fecal1.
of
valuable Changes
information
material [75,76]
Author
[125]
regard-
examination,
[114]
12
[104][127]
of a
19
Parkinson’s Sclerosis Bacteroidetes
muciniphila
Firmicutes analysis is a fecal material examination,
Clostridium [124]
[75]
[124]
Although
ing the
multiplatform
Alzheimer’s theDisease
the
state ofanalysis gold
Disease
standard
microbiome.
of Escherichia/Shigella
in microbiota
Lachnospiraceae
Methanobrevibacter
Moreover, thealso
Fecalibacterium
its metabolites may
Bacteroidetes
Escherichia/Shigella
Prevotella
Prevotellaceae gut provide
microbiome couldinformation
valuable [76]
[104]
[127]
be a potential
[125] newa
regard-
[75,76]
[76]
[126]
[104]
Type
Although
multiplatform
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
Table Alzheimer’s
1.for
Changes of Disease
the gold
analysis
in Disease
the standard
of its
microbiota Lachnospiraceae
in
metabolitesClostridium
Bacteria
microbiota
Fecalibacterium
Eubacterium
in may
neurological also
rectale Direction
analysis
provide
diseases. is a of
fecal
valuable Changes
material
information [104]
[124]
Author
examination,
[125]
regard-
[76]
12 a
of 19
target
ing the theoftreatment
state the of theseAkkermansia
microbiome. Firmicutes
Bacteroidetes
Methanobrevibacter
diseases.
Moreover, Prevention
Prevotella the of gut dysbiosis
gut microbiome
mucini- could be [75][124]
[127]
by aprobiotics
potential
[126] may
new
multiplatformMultiple Sclerosis
analysis of Escherichia/Shigella
Firmicutes
Bacteroidetes
Lachnospiraceae
Methanobrevibacter
Fecalibacterium [76]
[75]
[138]
[104]
[127]
[125]
ing
alsothe
target state
Alzheimer’s
provide
for
Type
Although
oftreatment
of
theDisease
theprotection
Disease
the gold ofits
microbiome.
from metabolites
the
these
standard
Moreover,
diseases.
Eubacterium
Akkermansia
in
may
Bacteroidetesthealso
Enterobacteriaceae
Prevotella
disorders gut provide
described
Prevention
phila
Bacteria
microbiota rectale
mucini-
microbiome
above.
of is aA
gut
Direction
analysis
valuable
couldinformation
summary
dysbiosis
of
fecal
be
by
Changes
material the regard-
ofaprobiotics
potential
[75,76]
[103]
[126] new
microbiota
[76] maya
Author
examination,
ing the
target state
for the of the
treatment microbiome.
of these Bacteroidetes
Firmicutes
Eubacterium
Clostridium
Proteobacteria
Methanobrevibacter
Moreover,
diseases. rectale
the gut
Prevention microbiome
of gut could
dysbiosis be
by a [138]
[75]
[76]
[138]
[127]
potential
probiotics [124]
new
may
changes
also observed
provide protectionin neurological
from the Akkermansia
Prevotella
diseases
disorders mucini-
is pylori
presented
described
Escherichia/Shigella
Helicobacter
Bacteroidetes
phila in Table
above. A 1. information
summary [126]
of the microbiota
[76]
[114]
[138]
multiplatform
target Multiple
Alzheimer’s
for analysis
Sclerosis
theprotection
treatment of
ofits
these Enterobacteriaceae
Bacteroidetes
metabolites may
Bacteroidetes
Proteobacteria
diseases. also provide
Prevention ofingut valuable
dysbiosis by [103]
[124]
[127]
regard-
[75,76]
[138]
also provide
changes Parkinson’s
observed
Although the Disease
in from the
neurological
gold standard disorders
Eubacterium
diseases
in phila
microbiotadescribed
Enterobacteriaceae
Akkermansia
Firmicutes
is rectale
mucini-
presentedabove.
analysis is A
Table
a summary
fecal1.material ofprobiotics
[76] maya
the microbiota
[103]
[138]
examination,
ing the
Major state of
Depressive
Multiple the microbiome.
Disorder
Sclerosis Bacteroidetes
Firmicutes
Fecalibacterium
Proteobacteria
Prevotellaceae
Bacteroidetes
Moreover, the gut microbiome could be a [138]
[75]
[138]
[104]
[124]
[127]
potential [125]
new
also
Tableprovide
changes
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 1.Multiple
Changesprotection
observed in from
theneurological
inDisease
microbiotathe inHelicobacter
disorders
diseases is pylori
Clostridium
described
Escherichia/Shigella
Firmicutes
phila
neurological presentedabove.
diseases. A
in Table summary
1. information [114]
[124]
of the microbiota
[76]
[138]
12 of 19
multiplatform
target Parkinson’s
Alzheimer’s
for the analysis
Sclerosis
Disease
treatment of
of its
these Enterobacteriaceae
Helicobacter
Bacteroidetes
Bifidobacterium
metabolites may
Proteobacteria
diseases. pylori
also provide
Prevention of gut valuable
dysbiosis by [103]
[114]
[124]
[140]
regard-
[138]
probiotics may
Major Depressive
Parkinson’s Disorder Eubacterium
Lachnospiraceae
Firmicutes
Clostridium rectale [76]
[104]
[138]
[124]
changes
Table
ing the
Major
observed
1.Multiple
Changes
state of
Depressive in Disease
the in
the neurological
microbiota in
microbiome.
Disorder
Sclerosis
diseases isthe
Prevotellaceae
Fecalibacterium
neurological
Firmicutes
Prevotella
Bacteroidetes
Moreover,
Bifidobacterium
Helicobacter
Prevotellaceae
presented
diseases. in Table could
gut microbiome
pylori
1. beofa the [104]
[125]
[75][126]
[124]
potential
[140]
[114]
[104] new
also
Tableprovide
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 1. Type
Changesprotection
of Disease
in the from thein
microbiota Clostridium
Lactobacillus
disorders
Bacteriadescribed
Firmicutes
neurological above.
Direction
diseases. A summary
of Changes [124]
[140]
microbiota
Author
[138]
12 of 19
target Parkinson’s
for the Disease
treatment of these Enterobacteriaceae
Methanobrevibacter
Bifidobacterium
Fecalibacterium
Lachnospiraceae
Prevotella
diseases. Prevention of gut dysbiosis by [103]
[127]
[140]
[125]
[104]
[126]
probiotics may
Major
changes Depressive
observed Disorder Eubacterium
Clostridium
Lactobacillus rectale [76]
[124]
[140]
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
Table 1. Type
Changes in in
theneurological
of Disease diseases is presented
Prevotellaceae
Lachnospiraceae
Bacteria
Fecalibacterium
Coprococcus
Bacteroidetes
microbiota Akkermansia
inHelicobacter
neurological
Bacteroidetes
Bifidobacterium diseases.
pylori
mucini-
in Tableof
Direction 1. Changes [104]
Author
[125]
[144]
[75,76]
12[138]
[140]
[114]
of 19
also provide
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW Type protection
Although ofthe gold from
Disease standard in Lactobacillus
Prevotella
the Methanobrevibacter
disorders
microbiota
Bacteriadescribed
Bacteroidetes above.
analysis is a A
Direction summary
fecal
ofmaterial
Changes [140]
[126]
of examination,
the microbiota
Author
[127]
[138]
12 of 19a
Parkinson’s Disease Enterobacteriaceae
Fecalibacterium
Coprococcus
Bacteroidetes
Lachnospiraceae
Methanobrevibacter
Prevotella [103]
[127]
[125]
[144]
[75,76]
[104]
[127]
[126]
[154]
changes observed
Type
multiplatform of in
Disease
analysis neurological
of its Escherichia/Shigella
diseases
metabolites phila
Lactobacillus
Bacteriais
Prevotellaceae
may
Coprococcus presented
also in Table
Direction
provide 1.
of
valuable Changes
information [76]
[140]
[104]
Author
regard-
[144]
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
Table Alzheimer’s
1. Changes in Disease Bacteroidetes
the microbiota Akkermansia
inHelicobacter
neurological
Proteobacteria
Proteobacteria diseases.
mucini-
pylori [138]
[75,76]
12[138]
[138]
[114]
of 19
Although the gold standard Prevotella
inBacteroidetes
Akkermansia
microbiota
Methanobrevibacter
Bacteroidetes
Clostridium
Firmicutes mucini-
Escherichia/Shigella analysis is a fecalcould
material [126]
[154]
[76]
examination,
[75] newa
[127]
[138]
[153]
ing the state
Parkinson’s
Multiple
Alzheimer’sof the microbiome.
Disease
Sclerosis
Disease Bacteroidetes
Moreover,
Coprococcus
Bacteroidetes
Lachnospiraceae
phila the
Proteobacteria
Escherichia/Shigella gut microbiome be a [124]
potential
[144]
[75,76]
[104]
[127]
[154]
[138]
[76]
multiplatformMajor
Type of Depressive
Disease
analysis of Firmicutes
Prevotellaceae
Bacteria Direction ofmaterial
Changes [138]
[104]
Author
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW
Table
targetAlthough
Major1.for
Changes
Alzheimer’s the
theDisorder
treatment
Depressive gold
in Disease
the ofits
microbiota
Disorder metabolites
standard
these in
Akkermansia may
inneurological
microbiota
phila
Bacteroidetes
Clostridium
Firmicutes
Firmicutes
Proteobacteria
Lactobacillus
diseases.
Eubacterium
Clostridium
also provide
analysis
diseases.
mucini-
Prevention
rectale
Escherichia/Shigella
Bacteroidetes
Methanobrevibacter
a valuable
of isgut fecal
dysbiosis information
by probiotics regard-
examination,
[138]
[153]
12[138]
[75]
[138]
[153]
[76]
[124]
[154]
[76]
[124]
[127] may a
of 19
ing Although
Autism
the Multiple
Spectrum
state
multiplatform of the
the gold
Sclerosis
analysis standard
Disorder
microbiome.
of its in microbiota
Clostridium
Firmicutes
Bifidobacterium
Moreover,
metabolites may the analysis
gut
also is a
microbiome
provide fecal material
could
valuable be aexamination,
[153]
[138]
[75]
[140]
potential
information new
regard- a
also
MajorAlzheimer’s
provide
Multipleprotection
Depressive Disease
Sclerosis from
Disorder Bacteroidetes
Lachnospiraceae
disorders phila
Proteobacteria
Lactobacillus
the Eubacterium described
Firmicutes rectale above. A summary [75,76]
[104]
[124]
[138]
[153]
of examination,
the microbiota
[76]
[138]
[154]
Type
Although
multiplatform of Disease
the gold
analysis standard
of its Enterobacteriaceae
in Bacteria
microbiota
Fecalibacterium
metabolites may also Direction
analysis
provideis a of
fecal
valuable Changes
material
information [103]
Author
[125]
regard-
[153] a
ingAutism
target
thefor
Major
changes
Spectrum
the
state oftreatment
Depressive
observed of theseAkkermansia
Disorder
theinmicrobiome.
Disorder
neurological
Firmicutes
Clostridium
Bifidobacterium
Lactobacillus
diseases.
Moreover,
Eubacterium
diseases
mucini-
Prevention
Bifidobacterium
isthe
Lactobacillus
Escherichia/Shigella
Bacteroidetes
Methanobrevibacter
Clostridium ofingut
gut microbiome
rectale
presented dysbiosis
Table could
1. be [75]
[124]
[76][140]
[153]
by aprobiotics
[140]
potential
[140]
[76]
[124]
[127] may
new
ingAutism
the Multiple
multiplatform Spectrum
state of Sclerosis
analysis
the Disorder
of its
microbiome. metabolites Firmicutes
Enterobacteriaceae
Bifidobacterium
Prevotella
Moreover,
Helicobacter may
Bacteroidetesthe also
gut
pylori provide
microbiome valuable
could information
be a [138]
[154]
[103]
[140]
[155]
regard-
potential
[114]
[75,76] new
also Alzheimer’s
provide theprotection Diseasefrom the Eubacterium
disorders phila
Prevotella
Lactobacillus
described above. [126]
[153]
target
ing
Major
Autism
the
target
forDepressive
Although
Parkinson’s
for
treatment
Spectrum
state
the of the
the
treatmentgold
Disease
of these
Disorder
standard
Disorder
microbiome.
of these
diseases.
in Prevention
Fecalibacterium
Firmicutes
Lactobacillus
Enterobacteriaceae
microbiota
Coprococcus
Akkermansia
Firmicutes
Clostridium
Lactobacillus
Fecalibacterium
Bifidobacterium
Moreover,
diseases.
rectale
the analysis
mucini-
gut
Prevention
Prevotella
of is aA
gut
microbiome
of gut
summary
dysbiosis
fecalcould
dysbiosis
by
material
be
by
ofprobiotics
a
the microbiota
[76] may
[125]
[154]
[140]
[103]
examination,
[144]
[75][140]
[124]
[125]
[140]
potential
probiotics
[155] new
may a
changes
also
Table observed
provide
1.Multiple
Changesprotectionin
theneurological
Sclerosis
inDisease from
microbiotathe inHelicobacter
Lactobacillus
Coprococcus
diseases
disorders
Prevotellaceaepylori
is presented
described
Escherichia/Shigella
Bacteroidetes
Firmicutes
neurological in Table
above.
diseases. A 1. information
summary [114]
[140]
[155]
of the microbiota
[104]
[76]
[124]
[127]
[138]
[154]
multiplatform
Parkinson’s
Alzheimer’s analysis
Diseaseof its Enterobacteriaceae
metabolites Prevotella
may
Coprococcus
Helicobacter
phila
Bacteroidetes also
pylori provide valuable [103]
[126]
regard-
[155]
[144]
[114]
[154]
target
changes forobserved
also provide theprotection
treatment of these
from
inmicrobiome.
neurological diseases.
the Eubacterium
disorders Prevention
described
Fecalibacterium
Prevotella
Lactobacillus
Coprococcus
diseases rectale
isthe
Prevotellaceae presented ofingut
above. Adysbiosis
Tablesummary
1. byofprobiotics
[76] may
the microbiota
[125]
[126]
[140]
[155]
[104]
ing theParkinson’s
state of the Disease Coprococcus
Lachnospiraceae
Firmicutes
Clostridium
Coprococcus
Proteobacteria
Moreover,
Prevotella gut microbiome could beofa the [144]
[104]
[75][144]
[124]
[138]
potential
[155] new
also provide
changes Type
1.Multipleprotection
observed
of in
Disease
Changes Sclerosis
Table Parkinson’s inDisease from the
neurological Helicobacter
the conceptualization, Bacteroidetes
Coprococcus
disorders
diseases
Prevotellaceae
Bacteria
Bacteroidetes
Clostridium
microbiota inEnterobacteriaceae
neurological
pylori
described
is presentedabove.
in A
Table
Direction summary
1.
of Changes [114]
[138]
[155]
[154]
microbiota
[104]
Author
[124]
[153] J.D.;
Author Contributions: J.D. and diseases.
Prevotella
Bacteroidetes
Coprococcus
Lachnospiraceae B.M.; writing—original draft preparation, [103]
[126]
[138]
[144]
[104]
target
changes for the
observedtreatment of these Bacteroidetes
diseases. Prevention
Methanobrevibacter
Eubacterium is rectale
Fecalibacterium
Firmicutes
Coprococcus of gut dysbiosis by [154]
probiotics
[127]
[125]
[138]
[155] may
[76]acqui-
writing—review
Table
Author
Major1.Contributions:
Changes
Depressive in in
and the neurological
editing, M.G. and
microbiota
Disorder
diseases
Prevotellaceae
Proteobacteria
B.M.;
inHelicobacter
conceptualization, neurological
Clostridium
Bacteroidetesand
Lactobacillus
Bacteroidetes
Proteobacteria
presented
J.D.visualization,
Bacteroidetes
Lachnospiraceae diseases. in Table
J.D.;
B.M.; writing—original
pylori
1. draft
supervision, M.G.; [104]
[138]
[153]
funding
[75,76]
[104]
[124]
preparation,
[153]
[114]
[138][154]
J.D.;
also
Table
Authorprovide
1.
Autism Type
Changesprotection
of
Spectrum
Contributions:Disease
in the from
microbiota
Disorder the disorders
in Bacteria
neurologicaldescribed
Methanobrevibacter
Clostridium
Akkermansia
Enterobacteriaceae
conceptualization, J.D. and
Prevotella mucini- above.
Direction
diseases.
B.M.; A summary
writing—originalof Changes
draftof the [154]
microbiota
Author
[127]
[153]
[103]
preparation,
[126] J.D.;
sition, Parkinson’s
B.M. All authors
writing—review have read
andDisease
editing, M.G.andand Bifidobacterium
agreed
B.M.; to the published
visualization,
Lachnospiraceae
Firmicutes
Lactobacillus
Escherichia/Shigella
Methanobrevibacter
version
J.D.; of the
supervision, manuscript.
M.G.; [140]
funding
[104]
[138]
[153]
[76]
[127]acqui-
changes observed
Type of in
Diseaseneurological Fecalibacterium
Firmicutes
diseases is
Prevotellaceae
Proteobacteria
Firmicutes
Bacteria
Clostridium presented in Table
Direction 1.
of Changes [125]
[154]
[104]
[138]
Author
[153]
Major
Table 1.
Autism
Author
sition, Depressive
writing—review
Changes
Spectrum and
Contributions:
Alzheimer’s
B.M. in
Allresearch
authors theDisorder
editing, M.G.
microbiota and
in
conceptualization,
Disease
have read and B.M.; visualization,
Bacteroidetes
Akkermansia
neurological
philathe
Clostridium
J.D. and
Lactobacillus
agreed mucini-
diseases.
B.M.; J.D.; supervision,
writing—original M.G.;
draft funding
preparation,
[153]acqui-
[75,76]
[153]
J.D.;
Major
Funding:
Autism Depressive
TypeAllof
This
Spectrum Disorder
Diseasewas
havesupported
Disorder by the to
Helicobacter
Bacteroidetes
Lactobacillus
Bacteria
Methanobrevibacter
Medical
Bifidobacterium
Firmicutes
Akkermansia
Prevotella
published
pyloriUniversity
mucini-
version
Direction
of the manuscript.
of
of Changes
of Bialystok, [114]
[138]
[140]
Author
[127]
Poland. Grant
[140]
[154] num-
[75]acqui-
[126]
sition, Parkinson’s
B.M.
writing—review andDisease
authors editing, read
M.G.andand agreed
B.M.; to
Bacteroidetes
Lachnospiraceae
Firmicutes the
Bifidobacterium
phila
Lactobacillus
Escherichia/Shigella
Bacteroidetes published
visualization, version
J.D.; the manuscript.
supervision, M.G.; funding[155]
[75,76]
[104]
[138]
[140]
[153]
[76]num-
[124]
[127]
bers
Funding:
Major Multiple
Autism
Type
This of Sclerosis
Spectrum
SUB/1/DN/21/006/1198
Disease
research
Depressive was and
supported
theDisorder byFirmicutes
SUB/1/DN/21/001/1198.
Prevotellaceae
Proteobacteria
Bacteria
the Medical
Coprococcus
Bacteroidetes Direction
University of of Changes
Bialystok, Poland. [154]
[104]
[138]
Author
Grant
[144]
Table
sition, 1.
Autism Changes
Spectrum
Alzheimer’s
B.M. in Disease
All authors microbiota
have read andAkkermansia
in neurological
agreed
Eubacteriumphila
Lactobacillus
Lactobacillus
to the
Prevotella
Bacteroidetes
Coprococcus published version of the manuscript.[75,76]
mucini-
diseases.
rectale [140]
[76][153]
[155]
[138]
[155]
Funding:
bers This Disorder
Multiple research was and
Sclerosis
SUB/1/DN/21/006/1198 supported Escherichia/Shigella
Bacteroidetes
Methanobrevibacter
Bifidobacterium
by the Medical
Lactobacillus
Firmicutes
Clostridium
SUB/1/DN/21/001/1198.
Prevotella University of Bialystok, Poland. Grant [76]
[124]
[127]
[140]
[75]num-
[154]
[124]
[155]
Alzheimer’s
Institutional Review
Multiple Disease
Board
Sclerosis Statement: Bacteroidetes
Lachnospiraceae
Firmicutes
Not phila
applicable.
Escherichia/Shigella
Bacteroidetes
Coprococcus [75,76]
[104]
[138]
[154]
[76]num-
[124]
[144]
bers SUB/1/DN/21/006/1198
Funding:
Major Type
This of
Depressive Disease
research and
was supported
Disorder Enterobacteriaceae
SUB/1/DN/21/001/1198.
Proteobacteria
by Bacteria
the Medical
Akkermansia
Firmicutes
Clostridium
Lactobacillus
Firmicutes Direction
University
mucini- of Changes
of Bialystok, [155]
[103]
[138]
Author
Poland. Grant[75]
[124]
[140]
Alzheimer’s
Institutional Review Disease
Board Statement: Coprococcus
Prevotella
Fecalibacterium
Eubacterium
Coprococcus
Not rectale
applicable.
Escherichia/Shigella
Bacteroidetes
Methanobrevibacter
Bifidobacterium
Clostridium [76][154]
[144]
[155]
[125]
[155]
[76]
[124]
[127]
[140]
[153]
Author
bers Multiple
Contributions:Sclerosis
SUB/1/DN/21/006/1198
Informed Consent conceptualization,
Statement: andNot Firmicutes
J.D. and
SUB/1/DN/21/001/1198.
applicable.
Bacteroidetes
Helicobacter
Bacteroidetes
Firmicutes B.M.;
pylori writing—original draft [75] J.D.;
[124]
preparation,
[154]
[114]
[75,76]
[138]
Alzheimer’s
Institutional ReviewDiseaseBoard Statement: Notphila
Eubacterium applicable.
Fecalibacterium
Coprococcus
Bacteroidetes rectale J.D.; supervision, M.G.; funding [125]
[76]acqui-
[144]
[154]
[155]
MajorParkinson’s
Depressive
writing—review and Disease
Disorder
editing, M.G. Enterobacteriaceae
and Prevotella
B.M.;
Not Akkermansia visualization,
Firmicutes
Clostridium
Lactobacillus
Prevotella mucini- [103]
[126]
[75]
[124]
[140]
Author Contributions:
Informed
Institutional
Data
Consent
Review
Availability
conceptualization,
Statement:
Board
Statement:
applicable.
Statement:
Not
J.D. and
Fecalibacterium
Eubacterium
Clostridium
NotJ.D.
applicable.
B.M.; writing—original draft preparation,
rectale
applicable.
Prevotellaceae
Escherichia/Shigella
Bacteroidetes
Bifidobacterium [76][155]
[153]
[125]
[153]
[104]
[76]
[124]
[127]
[140]
J.D.;
Autism
Author
Informed
sition, Multiple
B.M.
writing—review Spectrum
Contributions:
Consent Sclerosis
All authors
and Disorder
conceptualization,
Statement:
have
editing, Notand
read
M.G. and agreed
Helicobacter toand
Enterobacteriaceae
Prevotella
Bacteroidetes
applicable.
Clostridium
Bacteroidetes
B.M.; the B.M.; writing—original
published
pylori
visualization, version
J.D.; of thedraft
supervision, preparation,
manuscript.
M.G.; [103]
[126]
[154]
[153]
[114]
[138]
funding J.D.;
acqui-
Alzheimer’s Disease phila
Fecalibacterium
Eubacterium
Coprococcus
Firmicutes rectale J.D.; supervision, M.G.; funding [125]
[76]acqui-
[144]
[154]
Data
Author Availability
InformedParkinson’s
writing—review
Conflicts
sition, Contributions:
B.M. Consent
of Interest:
Allresearch
Statement:
and
authors Disease
editing,
The
Not
M.G. Enterobacteriaceae
and
conceptualization,
Statement: Not
authors
have read
Prevotella
applicable.
Lactobacillus
B.M.;
applicable.
declare J.D.
no visualization,
Lachnospiraceae
Firmicutes
Clostridium
Lactobacillus
Coprococcus and
conflict B.M.;
of writing—original
interest. draft [103]
[126]
[153]
[104]
[75]
[124]
[140]
preparation, [155]
J.D.;
Autism
Data Spectrum
Availability
Funding: This
Multiple Disorder
Statement:
was
Sclerosis Not and agreed
applicable.
supported the to
Helicobacter
Bacteroidetes
Clostridium
Lactobacillus
Bacteroidetes
by the
Proteobacteria
Prevotellaceae
Medical
Enterobacteriaceae
Prevotella
published
pyloriUniversityversion of the manuscript.
of Bialystok, [114]
[138]
[153]
[138]
[104]
[124]
Poland. Grant
[103]
[126] num-
sition, B.M.
Autism
writing—review All
Parkinson’s authors
Spectrum and have
Disease
Disorder
editing, read
M.G.and Bacteroidetes
agreed
Helicobacter
and to
Bacteroidetes
B.M.; the published
pylori
visualization, version
J.D.; of the manuscript.
supervision, M.G.; funding[154]
[155]
[114]
[138]
Conflicts
Funding:
of Interest:
bers SUB/1/DN/21/006/1198
Data Availability
This
Parkinson’s
The authors
Statement:
research was
Disease and
Not declare Firmicutes
applicable.
supported
no conflict
Methanobrevibacter
Fecalibacterium
Eubacterium
Coprococcus
SUB/1/DN/21/001/1198.
Proteobacteria
Prevotellaceae
Lactobacillus
byFirmicutes
the Medical
of
rectaleinterest.
University of Bialystok, Poland. [76]acqui-
[154]
[127]
[125]
[144]
[138]
[104]
[153]
[154]
Grant num-
Conflicts
sition, B.M. ofSpectrum
Interest:
All authors The authors
have read and Lachnospiraceae
declare Clostridium
no conflict
agreed
Helicobacter to the
Bacteroidetes
Clostridium
Coprococcus of interest.version of the manuscript.[114]
published
pylori [104]
[138]
[124]
[138]
[153]
[155]num-
Autism
Funding:
Major
bers This
Depressive
SUB/1/DN/21/006/1198 Disorder
researchDisorder
was and
supported Proteobacteria
Prevotellaceae
by the Medical
Prevotella
Akkermansia
Enterobacteriaceae
Prevotella
SUB/1/DN/21/001/1198.
Bacteroidetes University
mucini- of Bialystok, Poland. [138]
[104]
draftGrant
[155]
[103]
[126]
[154]
Parkinson’s
Author
Institutional
Conflicts of Review
Interest: Disease
Contributions:
Board
The Conceptualization,
Statement:
authors Lachnospiraceae
declare Firmicutes
Prevotella
Notno J.D.ofand
applicable.
conflict
Methanobrevibacter
Bifidobacterium
Fecalibacterium
B.M.; writing—original
interest. preparation,
[104]
[138]
[154]
[155]
[127]
[140]
[125]
J.D.;
bers SUB/1/DN/21/006/1198
Funding:
Major This research
Depressive Disorderwas and SUB/1/DN/21/001/1198.
supported Proteobacteria
Prevotellaceae
Lactobacillus
by the Medical
Lachnospiraceae
Firmicutes
phila University of Bialystok, Poland. [138]
[104]
[153]
Grant
[104]
[138] num-acquisi-
writing—review
Autism
Institutional Spectrum
Review Board and editing,
Disorder
Statement:M.G. Coprococcus
and
Helicobacter B.M.;
Bacteroidetes
Clostridium
Not visualization,
pylori
applicable. J.D.; supervision, M.G.; [155]
funding
[114]
[138]
[153]
Major
Author Depressive
Contributions: Disorder Methanobrevibacter
Bifidobacterium
Prevotella
Coprococcus
Akkermansia
conceptualization, Lactobacillus
Prevotella
J.D. andmucini- [127]
[140]
[155]
[140] J.D.;
[126]
bers SUB/1/DN/21/006/1198
Informed
InstitutionalConsent
Parkinson’s
tion, B.M.
ReviewAllStatement:
Disease
authors andNot
have SUB/1/DN/21/001/1198.
applicable.
read
Board Statement: Lachnospiraceae
Firmicutes
and
Not agreed toB.M.;
applicable.
Bacteroidetes
Methanobrevibacter
Bifidobacterium
writing—original
the published version ofdraft
the preparation,
[104]
[138]
manuscript.
[154]
[124]
[127]
[140]
Major Multiple
Depressive
writing—review Sclerosis
and Disorder
editing, M.G. and Proteobacteria
Prevotellaceae
Lactobacillus
B.M.;
Akkermansia visualization,
mucini- J.D.; supervision, M.G.; [138]
[104]
[153]
funding acqui-
Informed
Autism
Institutional
Author
Data
Consent
Spectrum
Review
Contributions:
Availability
Statement:
Disorder
Board
Statement:
Not applicable.
Statement:
conceptualization,
Not Not
applicable.
phila
Lactobacillus
Coprococcus
Coprococcus
Bacteroidetes
applicable.
J.D. and B.M.; writing—original draft
[140]
[155]
[144]
[138]
preparation, J.D.; Grant
InformedFunding:
sition, B.M.
Author Consent This
All authors
Contributions: research
Statement:
have was
Notand
read Methanobrevibacter
Bifidobacterium
Prevotella
Akkermansia
Clostridium
supported
applicable.
conceptualization, agreedJ.D.to by
Lactobacillus mucini-
the
and the Medical
published
B.M.; University
version
writing—original of theof Bialystok,
manuscript.
draft
[127]
[140]
[155]
[124]
Poland.
[140]
preparation, J.D.;
Multiple
writing—review Sclerosis
and editing, M.G. and Lachnospiraceae
Firmicutes
B.M.;phila
Bacteroidetes
Coprococcus
visualization, J.D.; supervision, M.G.; funding [104]
[138]
[154]
[124]
[127]
[144]acqui-
Data Availability
Major
Informed
Conflicts Depressive
numbers
Consent
of
writing—review Interest:
Statement:
and Disorder
Statement:
The
editing,
Not
SUB/1/DN/21/006/1198
Not
authors
M.G. Bacteroidetes
Proteobacteria
applicable.and
Akkermansia
applicable.
declare
and no
B.M.; SUB/1/DN/21/001/1198.
phila
Lactobacillus
Coprococcus mucini-
conflict of interest.
visualization, J.D.; supervision, M.G.; [154]
[138]
[140]
[155]
funding acqui-
Author
Data
Funding:
sition, Contributions:
Availability
B.M. This
Allresearch
authors conceptualization,
Statement:
was
have Not and
supported
read Fecalibacterium
Coprococcus
applicable.
by J.D.to
the and
Bacteroidetes
Methanobrevibacter
Bifidobacteriumthe B.M.;
Medical
Prevotella
agreed writing—original
University
published of thedraft
of Bialystok,
version Poland.
manuscript. [125]
[144]num-
preparation,
[124]
[127]
[140]
Grant
[155] J.D.;
Multiple Sclerosis Clostridium
Bacteroidetes
Clostridium
Firmicutes [124]
[154]
[153]
[138]
sition, B.M.
Conflicts
bers ofAll
Multiple
writing—review authors
Interest: have
The
Sclerosis
and
SUB/1/DN/21/006/1198 editing, read
authors
M.G.
and
and
declare
and nophila
agreed
B.M.; to the published
conflict
Bacteroidetes
Coprococcus of interest.
visualization,
Prevotella
SUB/1/DN/21/001/1198.
version
J.D.; of the manuscript.
supervision, M.G.; [124]
[144]
funding
[126]acqui-
Data Availability
Major Depressive
Conflicts of Interest:
Statement:
Disorder
The
Not
authors declare Bacteroidetes
applicable.
Akkermansia
Clostridium
Lactobacillus
Coprococcus
no mucini-
conflict ofUniversity
interest. [154]
[124]
[140]
[155]
Funding:
Author
sition, B.M. This
Allresearch
Contributions:
authors was
have supported
conceptualization, Fecalibacterium
by the
read andBifidobacterium
agreed Medical
Clostridium
J.D.toand
Bacteroidetes
Lactobacillusthe B.M.; of Bialystok,
writing—original
published version Poland.
of thedraft [125]
Grant
[153]num-
[153]
preparation,
manuscript. [124]
[127]
[140] J.D.;
Funding: Multiple
This Sclerosis
research was supported Clostridium
by the Medical
Bacteroidetes
Bacteroidetes
Clostridium University of Bialystok, Poland. [124]
Grant
[154]
[138]
[153] num-
Autism
Institutional
bers ofSpectrum
writing—review Review
SUB/1/DN/21/006/1198
Conflicts Interest:
and Disorder
Board
The
editing,Statement:
and
authors
M.G. declare
and Notno
B.M.;phila
applicable.
SUB/1/DN/21/001/1198.
conflict
Fecalibacterium
Coprococcus of interest.
visualization, J.D.; supervision, M.G.; [125]
[144]
funding acqui-
bers SUB/1/DN/21/006/1198 and Prevotella
Lactobacillus
SUB/1/DN/21/001/1198.
Clostridium
Firmicutes
Lactobacillus [126]
[153]
[124]
[154]num-
[140]
Funding:
Autism
Author
sition, B.M. This
All research
Spectrum
Contributions:
authors was
have supported
Disorder
conceptualization,
read and Fecalibacterium
by the Medical
Clostridium
J.D.toand
Lactobacillus
agreed University
the B.M.;
Proteobacteria
Bacteroidetes of Bialystok,
writing—original
published version Poland.
of thedraft [125]
Grant
[153]
preparation,
manuscript. [153] J.D.;
[138]
[124]
Informed Multiple
InstitutionalConsent
Review Sclerosis
Statement:
Board Not Prevotella
Bacteroidetes
applicable.
Statement: Not applicable.
Bacteroidetes [126]
[154]
[138]
Autism
bers Spectrum
SUB/1/DN/21/006/1198
Institutional Review
writing—review Disorder
and
Board Statement:
and editing, Firmicutes
SUB/1/DN/21/001/1198.
Fecalibacterium
Prevotella
Coprococcus
Not applicable.
M.G. and Lactobacillus
B.M.; visualization, J.D.; supervision, M.G.; funding [154]
[125]
[155]
[144]acqui-
Prevotella
Firmicutes [126]
[153]
[138]
Int. J. Mol. Sci. 2021, 22, 10028 13 of 19

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.

Abbreviations

AD Alzheimer’s disease
ASD Autism spectrum disorder
Aβ Amyloid beta
BDNF Brain-derived neutrophic factor
CNS Central nervous system
ENS Enteric nervous system
GLP-1 Glucagon-like peptide
LPS Lipopolysaccharide
MDD Major depressive disorder
MS Multiple sclerosis
NFTs Neurofibrillary tangles
NLRP3 NLR family pyrin domain containing 3
PD Parkinson’s disease
SCFA Short-chain fatty acid
TLR-4 Toll-like receptor-4

References
1. Rao, M.; Gershon, M.D. The bowel and beyond: The enteric nervous system in neurological disorders. Nat. Rev. Gastroenterol.
Hepatol. 2016, 13, 517. [CrossRef]
2. Gibbons, C.H. Basics of autonomic nervous system function. Handb. Clin. Neurol. 2019, 160, 407–418. [CrossRef]
3. 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. [CrossRef]
4. Ogbonnaya, E.S.; Clarke, G.; Shanahan, F.; Dinan, T.G.; Cryan, J.F.; O’Leary, O.F. Adult Hippocampal Neurogenesis Is Regulated
by the Microbiome. Biol. Psychiatry 2015, 78, e7–e9. [CrossRef]
5. Luczynski, P.; Whelan, S.O.; O’Sullivan, C.; Clarke, G.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. Adult microbiota-deficient mice
have distinct dendritic morphological changes: Differential effects in the amygdala and hippocampus. Eur. J. Neurosci. 2016, 44,
2654–2666. [CrossRef] [PubMed]
6. Erny, D.; de Angelis, A.L.H.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.;
Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965.
[CrossRef] [PubMed]
7. Flint, H.J. The impact of nutrition on the human microbiome. Nutr. Rev. 2012, 70, S10–S13. [CrossRef] [PubMed]
8. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Reddy, D.N. Role of the normal gut microbiota. World
J. Gastroenterol. 2015, 21, 8787. [CrossRef]
9. Sommer, F.; Bäckhed, F. The gut microbiota-masters of host development and physiology. Nat. Rev. Microbiol. 2013, 11, 227–238.
[CrossRef]
10. Underwood, M.A.; German, J.B.; Lebrilla, C.B.; Mills, D.A. Bifidobacterium longum subspecies infantis: Champion colonizer of
the infant gut. Pediatr. Res. 2015, 77, 229. [CrossRef]
11. Peng, Z.; Ling, L.; Stratton, C.W.; Li, C.; Polage, C.R.; Wu, B.; Tang, Y.-W. Advances in the diagnosis and treatment of Clostridium
difficile infections. Emerg. Microbes Infect. 2018, 7, 15. [CrossRef]
12. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A
human gut microbial gene catalog established by metagenomic sequencing. Nature 2010, 464, 59. [CrossRef]
13. Shao, Y.; Forster, S.C.; Tsaliki, E.; Vervier, K.; Strang, A.; Simpson, N.; Kumar, N.; Stares, M.D.; Rodger, A.; Brocklehurst, P.; et al.
Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth. Nature 2019, 574, 117–121. [CrossRef]
14. Bäckhed, F.; Roswall, J.; Peng, Y.; Feng, Q.; Jia, H.; Kovatcheva-Datchary, P.; Li, Y.; Xia, Y.; Xie, H.; Zhong, H.; et al. Dynamics and
Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe 2015, 17, 690–703. [CrossRef]
15. Fallani, M.; Young, D.; Scott, J.; Norin, E.; Amarri, S.; Adam, R.; Aguilera, M.; Khanna, S.; Gil, A.; Edwards, C.A.; et al. Intestinal
Microbiota of 6-week-old Infants Across Europe: Geographic Influence Beyond Delivery Mode, Breast-feeding, and Antibiotics. J.
Pediatr. Gastroenterol. Nutr. 2010, 51, 77–84. [CrossRef]
16. García-Peña, C.; Álvarez-Cisneros, T.; Quiroz-Baez, R.; Friedland, R.P. Microbiota and Aging. A Review and Commentary. Arch.
Med. Res. 2017, 48, 681–689. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2021, 22, 10028 14 of 19

17. Palmer, C.; Bik, E.M.; DiGiulio, D.B.; Relman, D.A.; Brown, P.O. Development of the Human Infant Intestinal Microbiota. PLoS
Biol. 2007, 5, e50177. [CrossRef]
18. Claesson, M.J.; Cusack, S.; O’Sullivan, O.; Greene-Diniz, R.; de Weerd, H.; Flannery, E.; Marchesi, J.R.; Falush, D.; Dinan, T.;
Fitzgerald, G.; et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl. Acad.
Sci. USA 2011, 108, 4586–4591. [CrossRef] [PubMed]
19. Odamaki, T.; Kato, K.; Sugahara, H.; Hashikura, N.; Takahashi, S.; Xiao, J.; Abe, F.; Osawa, R. Age-related changes in gut
microbiota composition from newborn to centenarian: A cross-sectional study. BMC Microbiol. 2016, 16, 90. [CrossRef] [PubMed]
20. Spielman, L.J.; Gibson, D.L.; Klegeris, A. Unhealthy gut, unhealthy brain: The role of the intestinal microbiota in neurodegenera-
tive diseases. Neurochem. Int. 2018, 120, 149–163. [CrossRef] [PubMed]
21. Flint, H.J.; Scott, K.P.; Louis, P.; Duncan, S.H. The role of the gut microbiota in nutrition and health. Nat. Rev. Gastroenterol. Hepatol.
2012, 9, 577–589. [CrossRef] [PubMed]
22. Martin, F.-P.J.; Sprenger, N.; Montoliu, I.; Rezzi, S.; Kochhar, S.; Nicholson, J.K. Dietary Modulation of Gut Functional Ecology
Studied by Fecal Metabonomics. J. Proteome Res. 2010, 9, 5284–5295. [CrossRef]
23. Turnbaugh, P.J.; Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Knight, R.; Gordon, J.I. The Effect of Diet on the Human Gut Microbiome:
A Metagenomic Analysis in Humanized Gnotobiotic Mice. Sci. Transl. Med. 2009, 1, 6ra14. [CrossRef] [PubMed]
24. Merra, G.; Noce, A.; Marrone, G.; Cintoni, M.; Tarsitano, M.G.; Capacci, A.; Lorenzo, A. De Influence of Mediterranean Diet on
Human Gut Microbiota. Nutrients 2021, 13, 7. [CrossRef]
25. Scarmeas, N.; Stern, Y.; Mayeux, R.; Manly, J.J.; Schupf, N.; Luchsinger, J.A. Mediterranean Diet and Mild Cognitive Impairment.
Arch. Neurol. 2009, 66, 216–225. [CrossRef]
26. McEvoy, C.T.; Guyer, H.; Langa, K.M.; Yaffe, K. Neuroprotective Diets Are Associated with Better Cognitive Function: The Health
and Retirement Study. J. Am. Geriatr. Soc. 2017, 65, 1857–1862. [CrossRef]
27. Gardener, S.; Gu, Y.; Rainey-Smith, S.R.; Keogh, J.B.; Clifton, P.M.; Mathieson, S.L.; Taddei, K.; Mondal, A.; Ward, V.K.; Scarmeas,
N.; et al. Adherence to a Mediterranean diet and Alzheimer’s disease risk in an Australian population. Transl. Psychiatry 2012, 2,
e164. [CrossRef]
28. Ianiro, G.; Tilg, H.; Gasbarrini, A. Antibiotics as deep modulators of gut microbiota: Between good and evil. Gut 2016, 65,
1906–1915. [CrossRef]
29. Molina-Torres, G.; Rodriguez-Arrastia, M.; Roman, P.; Sanchez-Labraca, N.; Cardona, D. Stress and the gut microbiota-brain axis.
Behav. Pharmacol. 2019, 30, 187–200. [CrossRef] [PubMed]
30. Allen, J.M.; Mailing, L.J.; Niemiro, G.M.; Moore, R.; Cook, M.D.; White, B.A.; Holscher, H.D.; Woods, J.A. Exercise Alters Gut
Microbiota Composition and Function in Lean and Obese Humans. Med. Sci. Sports Exerc. 2018, 50, 747–757. [CrossRef] [PubMed]
31. Gubert, C.; Kong, G.; Renoir, T.; Hannan, A.J. Exercise, diet and stress as modulators of gut microbiota: Implications for
neurodegenerative diseases. Neurobiol. Dis. 2020, 134, 104621. [CrossRef] [PubMed]
32. Magne, F.; Gotteland, M.; Gauthier, L.; Zazueta, A.; Pesoa, S.; Navarrete, P.; Balamurugan, R. The Firmicutes/Bacteroidetes Ratio:
A Relevant Marker of Gut Dysbiosis in Obese Patients? Nutrients 2020, 12, 1474. [CrossRef]
33. Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and
implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [CrossRef]
34. Adak, A.; Khan, M.R. An insight into gut microbiota and its functionalities. Cell. Mol. Life Sci. 2018, 76, 473–493. [CrossRef]
35. 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. [CrossRef]
36. Nie, Y.; Hu, J.; Yan, X. Cross-talk between bile acids and intestinal microbiota in host metabolism and health. J. Zhejiang Univ. Sci.
B 2015, 16, 436. [CrossRef]
37. Semin, I.; Ninnemann, J.; Bondareva, M.; Gimaev, I.; Kruglov, A.A. Interplay Between Microbiota, Toll-Like Receptors and
Cytokines for the Maintenance of Epithelial Barrier Integrity. Front. Med. 2021, 8, 644333. [CrossRef]
38. Priori, D.; Colombo, M.; Clavenzani, P.; Jansman, A.J.M.; Lallès, J.-P.; Trevisi, P.; Bosi, P. The Olfactory Receptor OR51E1 Is Present
along the Gastrointestinal Tract of Pigs, Co-Localizes with Enteroendocrine Cells and Is Modulated by Intestinal Microbiota.
PLoS ONE 2015, 10, e0129501. [CrossRef] [PubMed]
39. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [CrossRef] [PubMed]
40. Tsavkelova, E.; Botvinko, I.V.; Kudrin, V.; Oleskin, A. Detection of neurotransmitter amines in microorganisms with the use of
high-performance liquid chromatography. Dokl. Biochem. 2000, 372, 115–117.
41. Stanaszek, P.M.; Snell, J.F.; O’Neill, J.J. Isolation, extraction, and measurement of acetylcholine from Lactobacillus plantarum. Appl.
Environ. Microbiol. 1977, 34, 237–239. [CrossRef]
42. Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological
Disorders. Nutrients 2021, 13, 2099. [CrossRef] [PubMed]
43. Jameson, K.G.; Olson, C.A.; Kazmi, S.A.; Hsiao, E.Y. Toward Understanding Microbiome-Neuronal Signaling. Mol. Cell 2020, 78,
577–583. [CrossRef]
44. Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble,
F.M. Short-Chain Fatty Acids Stimulate Glucagon-Like Peptide-1 Secretion via the G-Protein–Coupled Receptor FFAR2. Diabetes
2012, 61, 364–371. [CrossRef]
Int. J. Mol. Sci. 2021, 22, 10028 15 of 19

45. Everard, A.; Lazarevic, V.; Gaïa, N.; Johansson, M.; Ståhlman, M.; Backhed, F.; Delzenne, N.M.; Schrenzel, J.; François, P.; Cani, P.D.
Microbiome of prebiotic-treated mice reveals novel targets involved in host response during obesity. ISME J. 2014, 8, 2116–2130.
[CrossRef] [PubMed]
46. Caspani, G.; Swann, J. Small talk: Microbial metabolites involved in the signaling from microbiota to brain. Curr. Opin. Pharmacol.
2019, 48, 99–106. [CrossRef] [PubMed]
47. Peng, L.; He, Z.; Chen, W.; Holzman, I.R.; Lin, J. Effects of Butyrate on Intestinal Barrier Function in a Caco-2 Cell Monolayer
Model of Intestinal Barrier. Pediatr. Res. 2007, 61, 37–41. [CrossRef]
48. Plöger, S.; Stumpff, F.; Penner, G.B.; Schulzke, J.-D.; Gäbel, G.; Martens, H.; Shen, Z.; Günzel, D.; Aschenbach, J.R. Microbial
butyrate and its role for barrier function in the gastrointestinal tract. Ann. N. Y. Acad. Sci. 2012, 1258, 52–59. [CrossRef] [PubMed]
49. Swann, J.R.; Want, E.J.; Geier, F.M.; Spagou, K.; Wilson, I.D.; Sidaway, J.E.; Nicholson, J.K.; Holmes, E. Systemic gut microbial
modulation of bile acid metabolism in host tissue compartments. Proc. Natl. Acad. Sci. USA 2011, 108, 4523–4530. [CrossRef]
50. Quinn, M.; McMillin, M.; Galindo, C.; Frampton, G.; Pae, H.Y.; DeMorrow, S. Bile acids permeabilize the blood brain barrier after
bile duct ligation in rats via Rac1-dependent mechanisms. Dig. Liver Dis. 2014, 46, 527–534. [CrossRef]
51. Goyal, D.; Ali, S.A.; Singh, R.K. Emerging role of gut microbiota in modulation of neuroinflammation and neurodegeneration
with emphasis on Alzheimer’s disease. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 106, 110112. [CrossRef] [PubMed]
52. Berger, M.; Gray, J.A.; Roth, B.L. The Expanded Biology of Serotonin. Annu. Rev. Med. 2009, 60, 355–366. [CrossRef]
53. Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain
axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673.
[CrossRef] [PubMed]
54. Ridaura, V.; Belkaid, Y. Gut Microbiota: The Link to Your Second Brain. Cell 2015, 161, 193–194. [CrossRef]
55. Luczynski, P.; Neufeld, K.-A.M.; Oriach, C.S.; Clarke, G.; Dinan, T.G.; Cryan, J.F. Growing up in a Bubble: Using Germ-Free
Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior. Int. J. Neuropsychopharmacol. 2016, 19, 1–17.
[CrossRef] [PubMed]
56. Heijtz, R.D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut
microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047. [CrossRef]
57. Kennedy, E.A.; King, K.Y.; Baldridge, M.T. Mouse Microbiota Models: Comparing Germ-Free Mice and Antibiotics Treatment as
Tools for Modifying Gut Bacteria. Front. Physiol. 2018, 9, 1534. [CrossRef]
58. Wang, P.; Tu, K.; Cao, P.; Yang, Y.; Zhang, H.; Qiu, X.-T.; Zhang, M.-M.; Wu, X.-J.; Yang, H.; Chen, T. Antibiotics-induced intestinal
dysbacteriosis caused behavioral alternations and neuronal activation in different brain regions in mice. Mol. Brain 2021, 14, 49.
[CrossRef]
59. Block, M.L.; Hong, J.-S. Microglia and inflammation-mediated neurodegeneration: Multiple triggers with a common mechanism.
Prog. Neurobiol. 2005, 76, 77–98. [CrossRef] [PubMed]
60. Catanzaro, R.; Anzalone, M.G.; Calabrese, F.; Milazzo, M.; Capuana, M.L.; Italia, A.; Occhipinti, S.; Marotta, F. The gut microbiota
and its correlations with the central nervous system disorders. Panminerva Med. 2015, 57, 127–143.
61. Jyothi, H.J.; Vidyadhara, D.J.; Mahadevan, A.; Philip, M.; Parmar, S.K.; Manohari, S.G.; Shankar, S.K.; Raju, T.R.; Alladi, P.A.
Aging causes morphological alterations in astrocytes and microglia in human substantia nigra pars compacta. Neurobiol. Aging
2015, 36, 3321–3333. [CrossRef] [PubMed]
62. Dinan, T.G.; Cryan, J.F. Gut instincts: Microbiota as a key regulator of brain development, ageing and neurodegeneration. J.
Physiol. 2017, 595, 489–503. [CrossRef]
63. Wanchao, S.; Chen, M.; Zhiguo, S.; Futang, X.; Mengmeng, S. Protective effect and mechanism of Lactobacillus on cerebral
ischemia reperfusion injury in rats. Braz. J. Med. Biol. Res. 2018, 51, e7172. [CrossRef] [PubMed]
64. Blennow, K.; Zetterberg, H. Biomarkers for Alzheimer’s disease: Current status and prospects for the future. J. Intern. Med. 2018,
284, 643–663. [CrossRef] [PubMed]
65. Mroczko, B.; Groblewska, M.; Litman-Zawadzka, A.; Kornhuber, J.; Lewczuk, P. Amyloid β oligomers (AβOs) in Alzheimer’s
disease. J. Neural Transm. 2018, 125, 177–191. [CrossRef] [PubMed]
66. Mroczko, B.; Groblewska, M.; Litman-Zawadzka, A. The Role of Protein Misfolding and Tau Oligomers (TauOs) in Alzheimer0 s
Disease (AD). Int. J. Mol. Sci. 2019, 20, 4661. [CrossRef] [PubMed]
67. Calsolaro, V.; Edison, P. Neuroinflammation in Alzheimer’s disease: Current evidence and future directions. Alzheimer’s Dement.
2016, 12, 719–732. [CrossRef] [PubMed]
68. Rao, J.S.; Rapoport, S.I.; Kim, H.-W. Altered neuroinflammatory, arachidonic acid cascade and synaptic markers in postmortem
Alzheimer’s disease brain. Transl. Psychiatry 2011, 1, e31. [CrossRef]
69. Sun, Y.; Sommerville, N.R.; Liu, J.Y.H.; Ngan, M.P.; Poon, D.; Ponomarev, E.D.; Lu, Z.; Kung, J.S.C.; Rudd, J.A. Intra-gastrointestinal
amyloid-β1–42 oligomers perturb enteric function and induce Alzheimer’s disease pathology. J. Physiol. 2020, 598, 4209–4223.
[CrossRef] [PubMed]
70. Walker, L.C.; Schelle, J.; Jucker, M. The Prion-Like Properties of Amyloid-β Assemblies: Implications for Alzheimer’s Disease.
Cold Spring Harb. Perspect. Med. 2016, 6, a024398. [CrossRef]
71. Tan, L.Y.; Yeo, X.Y.; Bae, H.-G.; Lee, D.P.S.; Ho, R.C.; Kim, J.E.; Jo, D.-G.; Jung, S. Association of Gut Microbiome Dysbiosis with
Neurodegeneration: Can Gut Microbe-Modifying Diet Prevent or Alleviate the Symptoms of Neurodegenerative Diseases? Life
2021, 11, 698. [CrossRef]
Int. J. Mol. Sci. 2021, 22, 10028 16 of 19

72. 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. Alzheimer’s Dis. 2017, 56,
775–788. [CrossRef]
73. Bostanciklioğlu, M. The role of gut microbiota in pathogenesis of Alzheimer’s disease. J. Appl. Microbiol. 2019, 127, 954–967.
[CrossRef] [PubMed]
74. Sun, B.-L.; Li, W.-W.; Wang, J.; Xu, Y.-L.; Sun, H.-L.; Tian, D.-Y.; Wang, Y.-J.; Yao, X.-Q. Gut Microbiota Alteration and Its Time
Course in a Tauopathy Mouse Model. J. Alzheimer’s Dis. 2019, 70, 399–412. [CrossRef]
75. 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. [CrossRef] [PubMed]
76. 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. [CrossRef]
77. Kim, M.-S.; Kim, Y.; Choi, H.H.; Kim, W.; Park, S.; Lee, D.D.-S.; Kim, D.K.; Kim, H.J.; Choi, H.H.; Hyun, D.-W.; et al. Transfer
of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut 2020, 69, 283–294.
[CrossRef]
78. Park, S.-H.; Lee, J.H.; Shin, J.; Kim, J.-S.; Cha, B.; Lee, S.; Kwon, K.S.; Shin, Y.W.; Choi, S.H. Cognitive function improvement after
fecal microbiota transplantation in Alzheimer’s dementia patient: A case report. Curr. Med. Res. Opin. 2021, 1–6. [CrossRef]
79. Hazan, S. Rapid improvement in Alzheimer’s disease symptoms following fecal microbiota transplantation: A case report. J. Int.
Med. Res. 2020, 48, 030006052092593. [CrossRef]
80. Chapman, M.R. Role of Escherichia coli Curli Operons in Directing Amyloid Fiber Formation. Science 2002, 295, 851–855.
[CrossRef]
81. Friedland, R.P.; Chapman, M.R. The role of microbial amyloid in neurodegeneration. PLoS Pathog. 2017, 13, e1006654. [CrossRef]
82. Friedland, R.P. Mechanisms of Molecular Mimicry Involving the Microbiota in Neurodegeneration. J. Alzheimer’s Dis. 2015, 45,
349–362. [CrossRef]
83. Hill, J.M.; Lukiw, W.J. Microbial-generated amyloids and Alzheimer’s disease (AD). Front. Aging Neurosci. 2015, 7, 9. [CrossRef]
84. Li, Z.; Zhu, H.; Zhang, L.; Qin, C. The intestinal microbiome and Alzheimer’s disease: A review. Anim. Model. Exp. Med. 2018, 1,
180–188. [CrossRef]
85. Song, M.; Jin, J.; Lim, J.-E.; Kou, J.; Pattanayak, A.; Rehman, J.A.; Kim, H.-D.; Tahara, K.; Lalonde, R.; Fukuchi, K. TLR4 mutation
reduces microglial activation, increases Aβ deposits and exacerbates cognitive deficits in a mouse model of Alzheimer’s disease.
J. Neuroinflamm. 2011, 8, 92. [CrossRef] [PubMed]
86. Kahn, M.S.; Kranjac, D.; Alonzo, C.A.; Haase, J.H.; Cedillos, R.O.; McLinden, K.A.; Boehm, G.W.; Chumley, M.J. Prolonged
elevation in hippocampal Aβ and cognitive deficits following repeated endotoxin exposure in the mouse. Behav. Brain Res. 2012,
229, 176–184. [CrossRef]
87. Asti, A.; Gioglio, L. Can a Bacterial Endotoxin be a Key Factor in the Kinetics of Amyloid Fibril Formation? J. Alzheimer’s Dis.
2014, 39, 169–179. [CrossRef] [PubMed]
88. Zhao, Y.; Jaber, V.; Lukiw, W.J. Secretory Products of the Human GI Tract Microbiome and Their Potential Impact on Alzheimer’s
Disease (AD): Detection of Lipopolysaccharide (LPS) in AD Hippocampus. Front. Cell. Infect. Microbiol. 2017, 7, 318. [CrossRef]
[PubMed]
89. Zhan, X.; Stamova, B.; Jin, L.-W.; DeCarli, C.; Phinney, B.; Sharp, F.R. Gram-negative bacterial molecules associate with Alzheimer
disease pathology. Neurology 2016, 87, 2324. [CrossRef] [PubMed]
90. Zhang, R.; Miller, R.G.; Gascon, R.; Champion, S.; Katz, J.; Lancero, M.; Narvaez, A.; Honrada, R.; Ruvalcaba, D.; McGrath, M.S.
Circulating endotoxin and systemic immune activation in sporadic Amyotrophic Lateral Sclerosis (sALS). J. Neuroimmunol. 2009,
206, 121. [CrossRef] [PubMed]
91. Yang, X.; Yu, D.; Xue, L.; Li, H.; Du, J. Probiotics modulate the microbiota–gut–brain axis and improve memory deficits in aged
SAMP8 mice. Acta Pharm. Sin. B 2020, 10, 475–487. [CrossRef] [PubMed]
92. Elbaz, A.; Carcaillon, L.; Kab, S.; Moisan, F. Epidemiology of Parkinson’s disease. Rev. Neurol. 2016, 172, 14–26. [CrossRef]
93. Beitz, J.M. Parkinson’s disease: A review. Front. Biosci. 2014, S6, 65–74. [CrossRef] [PubMed]
94. Hayes, M.T. Parkinson’s Disease and Parkinsonism. Am. J. Med. 2019, 132, 802–807. [CrossRef] [PubMed]
95. Jankovic, J. Parkinson’s disease: Clinical features and diagnosis. J. Neurol. Neurosurg. Psychiatry 2008, 79, 368–376. [CrossRef]
96. Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [CrossRef]
97. Chartier, S.; Duyckaerts, C. Is Lewy pathology in the human nervous system chiefly an indicator of neuronal protection or of
toxicity? Cell Tissue Res. 2018, 373, 149–160. [CrossRef]
98. Pajares, M.; Rojo, A.I.; Manda, G.; Boscá, L.; Cuadrado, A. Inflammation in Parkinson’s Disease: Mechanisms and Therapeutic
Implications. Cells 2020, 9, 1687. [CrossRef]
99. Mulak, A.; Bonaz, B. Brain-gut-microbiota axis in Parkinson’s disease. World J. Gastroenterol. 2015, 21, 10609. [CrossRef]
100. Parashar, A.; Udayabanu, M. Gut microbiota: Implications in Parkinson’s disease. Park. Relat. Disord. 2017, 38, 1. [CrossRef]
101. Noyce, A.J.; Bestwick, J.P.; Silveira-Moriyama, L.; Hawkes, C.H.; Giovannoni, G.; Lees, A.J.; Schrag, A. Meta-analysis of early
nonmotor features and risk factors for Parkinson disease. Ann. Neurol. 2012, 72, 893–901. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2021, 22, 10028 17 of 19

102. Caputi, V.; Giron, M.C. Microbiome-Gut-Brain Axis and Toll-Like Receptors in Parkinson’s Disease. Int. J. Mol. Sci. 2018, 19, 1689.
[CrossRef]
103. 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.
[CrossRef]
104. 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, e00561-20. [CrossRef]
105. Forsyth, C.B.; Shannon, K.M.; Kordower, J.H.; Voigt, R.M.; Shaikh, M.; Jaglin, J.A.; Estes, J.D.; Dodiya, H.B.; Keshavarzian, A.
Increased Intestinal Permeability Correlates with Sigmoid Mucosa alpha-Synuclein Staining and Endotoxin Exposure Markers in
Early Parkinson’s Disease. PLoS ONE 2011, 6, e28032. [CrossRef]
106. 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. [CrossRef]
107. 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. [CrossRef]
108. 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.
[CrossRef] [PubMed]
109. Chen, S.G.; Stribinskis, V.; Rane, M.J.; Demuth, D.R.; Gozal, E.; Roberts, A.M.; Jagadapillai, R.; Liu, R.; Choe, K.; Shivakumar, B.;
et al. Exposure to the Functional Bacterial Amyloid Protein Curli Enhances Alpha-Synuclein Aggregation in Aged Fischer 344
Rats and Caenorhabditis elegans. Sci. Rep. 2016, 6, 34477. [CrossRef] [PubMed]
110. Megur, A.; Baltriukienė, D.; Bukelskienė, V.; Burokas, A. The Microbiota–Gut–Brain Axis and Alzheimer’s Disease: Neuroinflam-
mation Is to Blame? Nutrients 2021, 13, 37. [CrossRef] [PubMed]
111. Bodea, L.-G.; Wang, Y.; Linnartz-Gerlach, B.; Kopatz, J.; Sinkkonen, L.; Musgrove, R.; Kaoma, T.; Muller, A.; Vallar, L.; Di Monte,
D.A.; et al. Neurodegeneration by Activation of the Microglial Complement-Phagosome Pathway. J. Neurosci. 2014, 34, 8546–8556.
[CrossRef]
112. Nielsen, H.H.; Qiu, J.; Friis, S.; Wermuth, L.; Ritz, B. Treatment of Helicobacter Pylori Infection and Risk of Parkinson’s Disease in
Denmark. Eur. J. Neurol. 2012, 19, 864. [CrossRef]
113. Çamcı, G.; Oğuz, S. Association between Parkinson’s Disease and Helicobacter Pylori. J. Clin. Neurol. 2016, 12, 147. [CrossRef]
114. Tan, A.H.; Mahadeva, S.; Marras, C.; Thalha, A.M.; Kiew, C.K.; Yeat, C.M.; Ng, S.W.; Ang, S.P.; Chow, S.K.; Loke, M.F.; et al.
Helicobacter pylori infection is associated with worse severity of Parkinson’s disease. Park. Relat. Disord. 2015, 21, 221–225.
[CrossRef]
115. Gorlé, N.; Bauwens, E.; Haesebrouck, F.; Smet, A.; Vandenbroucke, R.E. Helicobacter and the Potential Role in Neurological
Disorders: There Is More than Helicobacter pylori. Front. Immunol. 2020, 11, 11. [CrossRef]
116. Harbo, H.F.; Gold, R.; Tintoré, M. Sex and gender issues in multiple sclerosis. Ther. Adv. Neurol. Disord. 2013, 6, 237–248.
[CrossRef] [PubMed]
117. Schepici, G.; Silvestro, S.; Bramanti, P.; Mazzon, E. The Gut Microbiota in Multiple Sclerosis: An Overview of ClinicalTrials. Cell
Transplant. 2019, 28, 1507. [CrossRef] [PubMed]
118. Nakahara, J.; Maeda, M.; Aiso, S.; Suzuki, N. Current Concepts in Multiple Sclerosis: Autoimmunity Versus Oligodendrogliopathy.
Clin. Rev. Allergy Immunol. 2011, 42, 26–34. [CrossRef]
119. Compston, A.; Coles, A. Multiple sclerosis. Lancet 2008, 372, 1502–1517. [CrossRef]
120. Olsson, T.; Barcellos, L.F.; Alfredsson, L. Interactions between genetic, lifestyle and environmental risk factors for multiple
sclerosis. Nat. Rev. Neurol. 2017, 13, 25–36. [CrossRef] [PubMed]
121. Guerrero-García, J.d.J.; Carrera-Quintanar, L.; López-Roa, R.I.; Márquez-Aguirre, A.L.; Rojas-Mayorquín, A.E.; Ortuño-Sahagún,
D. Multiple Sclerosis and Obesity: Possible Roles of Adipokines. Mediat. Inflamm. 2016, 2016, 1–24. [CrossRef]
122. Pierrot-Deseilligny, C.; Souberbielle, J.-C. Vitamin D and multiple sclerosis: An update. Mult. Scler. Relat. Disord. 2017, 14, 35–45.
[CrossRef]
123. Arneth, B. Multiple Sclerosis and Smoking. Am. J. Med. 2020, 133, 783–788. [CrossRef]
124. 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. [CrossRef]
125. 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. J. Investig. Med. 2015, 63, 729–734. [CrossRef]
126. Chen, J.; Chia, N.; Kalari, K.R.; Yao, J.Z.; Novotna, M.; Soldan, M.M.P.; 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. [CrossRef]
127. Mirza, A.; Forbes, J.D.; Zhu, F.; Bernstein, C.N.; Van Domselaar, G.; Graham, M.; Waubant, E.; Tremlett, H. The multiple sclerosis
gut microbiota: A systematic review. Mult. Scler. Relat. Disord. 2020, 37, 101427. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2021, 22, 10028 18 of 19

128. 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. [CrossRef] [PubMed]
129. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; DeRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al.
Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455.
[CrossRef] [PubMed]
130. WHO. Risk Reduction of Cognitive Decline and Dementia; World Health Organization: Geneva, Switzerland, 2019; ISBN
9789241550543.
131. Maurer, D.M.; Raymond, T.J.; Davis, B.N. Depression: Screening and Diagnosis. Am. Fam. Physician 2018, 98, 508–515.
132. Ruhé, H.G.; Mason, N.S.; Schene, A.H. Mood is indirectly related to serotonin, norepinephrine and dopamine levels in humans:
A meta-analysis of monoamine depletion studies. Mol. Psychiatry 2007, 12, 331–359. [CrossRef]
133. Ménard, C.; Hodes, G.E.; Russo, S.J. Pathogenesis of depression: Insights from human and rodent studies. Neuroscience 2016, 321,
138. [CrossRef] [PubMed]
134. Delpech, J.C.; Wei, L.; Hao, J.; Yu, X.; Madore, C.; Butovsky, O.; Kaffman, A. Early life stress perturbs the maturation of microglia
in the developing hippocampus. Brain. Behav. Immun. 2016, 57, 79–93. [CrossRef] [PubMed]
135. Johnson, F.K.; Kaffman, A. Early life stress perturbs the function of microglia in the developing rodent brain: New insights and
future challenges. Brain. Behav. Immun. 2018, 69, 18–27. [CrossRef]
136. Carlessi, A.S.; Borba, L.A.; Zugno, A.I.; Quevedo, J.; Réus, G.Z. Gut microbiota–brain axis in depression: The role of neuroinflam-
mation. Eur. J. Neurosci. 2021, 53, 222–235. [CrossRef] [PubMed]
137. Neufeld, K.-A.M.; Kang, N.; Bienenstock, J.; Foster, J.A. Effects of intestinal microbiota on anxiety-like behavior. Commun. Integr.
Biol. 2011, 4, 492–494. [CrossRef]
138. Kelly, J.R.; Borre, Y.; O’ Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Kennedy, P.J.; Beers, S.; Scott, K.; Moloney, G.; et al.
Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016,
82, 109–118. [CrossRef]
139. Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota
composition in patients with major depressive disorder. Brain. Behav. Immun. 2015, 48, 186–194. [CrossRef]
140. Liu, Y.; Zhang, L.; Wang, X.; Wang, Z.; Zhang, J.; Jiang, R.; Wang, X.; Wang, K.; Liu, Z.; Xia, Z.; et al. Similar Fecal Microbiota
Signatures in Patients with Diarrhea-Predominant Irritable Bowel Syndrome and Patients with Depression. Clin. Gastroenterol.
Hepatol. 2016, 14, 1602–1611. [CrossRef] [PubMed]
141. Aizawa, E.; Tsuji, H.; Asahara, T.; Takahashi, T.; Teraishi, T.; Yoshida, S.; Ota, M.; Koga, N.; Hattori, K.; Kunugi, H. Possible
association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder. J. Affect. Disord.
2016, 202, 254–257. [CrossRef] [PubMed]
142. Park, A.J.; Collins, J.; Blennerhassett, P.A.; Ghia, J.E.; Verdu, E.F.; Bercik, P.; Collins, S.M. Altered colonic function and microbiota
profile in a mouse model of chronic depression. Neurogastroenterol. Motil. 2013, 25, 733. [CrossRef]
143. O’Mahony, S.M.; Marchesi, J.R.; Scully, P.; Codling, C.; Ceolho, A.-M.; Quigley, E.M.M.; Cryan, J.F.; Dinan, T.G. Early Life Stress
Alters Behavior, Immunity, and Microbiota in Rats: Implications for Irritable Bowel Syndrome and Psychiatric Illnesses. Biol.
Psychiatry 2009, 65, 263–267. [CrossRef]
144. Yu, M.; Jia, H.; Zhou, C.; Yang, Y.; Zhao, Y.; Yang, M.; Zou, Z. Variations in gut microbiota and fecal metabolic phenotype
associated with depression by 16S rRNA gene sequencing and LC/MS-based metabolomics. J. Pharm. Biomed. Anal. 2017, 138,
231–239. [CrossRef]
145. Valles-Colomer, M.; Falony, G.; Darzi, Y.; Tigchelaar, E.F.; Wang, J.; Tito, R.Y.; Schiweck, C.; Kurilshikov, A.; Joossens, M.;
Wijmenga, C.; et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat. Microbiol. 2019,
4, 623–632. [CrossRef]
146. Huang, R.; Wang, K.; Hu, J. Effect of Probiotics on Depression: A Systematic Review and Meta-Analysis of Randomized Controlled
Trials. Nutrients 2016, 8, 483. [CrossRef]
147. Fattorusso, A.; Di Genova, L.; Dell’Isola, G.B.; Mencaroni, E.; Esposito, S. Autism Spectrum Disorders and the Gut Microbiota.
Nutrients 2019, 11, 521. [CrossRef] [PubMed]
148. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders; American Psychiatric Association:
Washington, DC, USA, 2013; ISBN 0-89042-555-8.
149. Geurts, H.M.; Jansen, M.D. A retrospective chart study: The pathway to a diagnosis for adults referred for ASD assessment.
Autism 2012, 16, 299–305. [CrossRef]
150. Risch, N.; Hoffmann, T.J.; Anderson, M.; Croen, L.A.; Grether, J.K.; Windham, G.C. Familial Recurrence of Autism Spectrum
Disorder: Evaluating Genetic and Environmental Contributions. Am. J. Psychiatry 2014, 171, 1206–1213. [CrossRef] [PubMed]
151. McElhanon, B.O.; McCracken, C.; Karpen, S.; Sharp, W.G. Gastrointestinal Symptoms in Autism Spectrum Disorder: A Meta-
analysis. Pediatrics 2014, 133, 872–883. [CrossRef] [PubMed]
152. Finegold, S.M.; Molitoris, D.; Song, Y.; Liu, C.; Vaisanen, M.; Bolte, E.; McTeague, M.; Sandler, R.; Wexler, H.; Marlowe, E.M.; et al.
Gastrointestinal Microflora Studies in Late-Onset Autism. Clin. Infect. Dis. 2002, 35, S6–S16. [CrossRef]
153. Liu, F.; Li, J.; Wu, F.; Zheng, H.; Peng, Q.; Zhou, H. Altered composition and function of intestinal microbiota in autism spectrum
disorders: A systematic review. Transl. Psychiatry 2019, 9, 43. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2021, 22, 10028 19 of 19

154. Finegold, S.M.; Dowd, S.E.; Gontcharova, V.; Liu, C.; Henley, K.E.; Wolcott, R.D.; Youn, E.; Summanen, P.H.; Granpeesheh, D.;
Dixon, D.; et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe 2010, 16, 444–453. [CrossRef]
[PubMed]
155. Kang, D.-W.; Park, J.G.; Ilhan, Z.E.; Wallstrom, G.; LaBaer, J.; Adams, J.B.; Krajmalnik-Brown, R. Reduced Incidence of Prevotella
and Other Fermenters in Intestinal Microflora of Autistic Children. PLoS ONE 2013, 8, e68322. [CrossRef]
156. Liu, F.; Horton-Sparks, K.; Hull, V.; Li, R.W.; Martínez-Cerdeño, V. The valproic acid rat model of autism presents with gut
bacterial dysbiosis similar to that in human autism. Mol. Autism 2018, 9, 61. [CrossRef] [PubMed]
157. Emanuele, E.; Orsi, P.; Boso, M.; Broglia, D.; Brondino, N.; Barale, F.; di Nemi, S.U.; Politi, P. Low-grade endotoxemia in patients
with severe autism. Neurosci. Lett. 2010, 471, 162–165. [CrossRef]
158. Kim, S.; Kim, H.; Yim, Y.S.; Ha, S.; Atarashi, K.; Tan, T.G.; Longman, R.S.; Honda, K.; Littman, D.R.; Choi, G.B.; et al. Maternal gut
bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature 2017, 549, 528–532. [CrossRef]

You might also like