Fcimb 13 1238005
Fcimb 13 1238005
KEYWORDS
FIGURE 1
Action mechanisms by which psychobiotics and fecal microbial transplantation exert the potential therapeutic effect on ASD and ADHD.
Abbreviations: g-aminobutyric acid (GABA), short-chain fatty acids (SCFAs).
ADHD. Therefore, we thoroughly summarized the latest reports on later-life health (Stiemsma and Michels, 2018; Niemarkt et al., 2019;
potential therapeutic mechanisms and promising perspectives, in Korpela et al., 2020).
addition to observed changes in the gut microbiome composition
and metabolites. Moreover, this review presents directions for
future treatments that could be employed to directly manipulate 2.2 Gut-brain axis and bidirectional
gut microbiota during early life stages in humans to prevent the communication
development of such diseases.
The past 15 years have seen the emergence of the microbiota as one
of the critical regulators of gut-brain function through a complex
2 Role of gut microbiome axis network of signaling pathways, which has led to the appreciation of the
importance of a distinct MGB axis (Cryan et al., 2019). There are
2.1 Early-life gut microbial colonization various bidirectional communicating pathways between the gut
and development microbiome and the brain, which include vagus nerve (VN),
immunity with tryptophan metabolism, endocrine system, and
Human microbial colonization begins in the fetus and enteric nervous system (ENS) with diverse bacterial byproducts, such
continues to develop and modulate species abundance for as peptidoglycans, short-chain fatty acids (SCFAs), and branched-chain
approximately three years until the gut microbiome becomes amino acids (Jena et al., 2020).
adult-like (Figure 2A) (Arrieta et al., 2014; Senn et al., 2020). Bidirectional communication between the microbiota and the
There is increasing evidence that the gut microbiota and its host through the gut-brain axis is an essential pathway for accessing
byproducts could play pivotal functions in the immune system the synergetic mechanism to modulate the host brain and behavior
maturation, development, and behavior of the host throughout the (Dinan and Cryan, 2017; Ronan et al., 2021). Studies to identify and
life cycle (Erkosar et al., 2013). examine the MGB axis have used different yet complementary
Microbiota development follows typical timely changes and the microbiota interventions, including germ-free rodents, antibiotic-
interplay between the gut microbiome and the rest of the human induced depletion, prebiotic/probiotic supplementation,
body that have been analyzed through metagenomics studies and gastrointestinal infection, and FMT (Cryan et al., 2019). Top-
recent strain-level profiling (Bäckhed et al., 2015; Selma-Royo et al., down signaling influences the motor, sensory, and secretory
2020). Especially microbial colonization of the newborn period is a functions of the gastrointestinal tract via the efferent fibers of the
critical process that affects long-term neurological outcomes and VN. Bottom-up communication affects the function of the brain,
FIGURE 2
(A) Microbial colonization and development from birth to child. (B) Various environmental factors could induce gut dysbiosis and result in the
pathogenesis of necrotizing enterocolitis, late onset sepsis, neurodevelopmental disorders, diarrhea, atopic disease, and type I diabetes.
especially the amygdala and hypothalamus, via the afferent roles in the production of substances, such as SCFAs, which are
vagal fibers. reported to have diverse neurobiological correlations in the context
of the MGB axis, and 4-ethylphenylsulfate, which is a dietary
tyrosine metabolite that is considered to induce ADS-like
3 Neurodevelopmental disorders and behavior among many others (Hsiao et al., 2013; Dalile et al.,
gut microbiome 2019). In an experiment with genetically engineered mice,
maternal interleukin-17a secreted by Th17 cells was observed to
3.1 Gut dysbiosis in neurodevelopmental induce behavioral and cortical issues in their offspring, suggesting a
disorders possible role for the cytokine receptor interleukin-17a in the
modulation of ASD (Choi et al., 2016).
Dysbiosis may play a role in the etiology and development of
neurodevelopment disorders (Nagpal and Cryan, 2021). Earlier
reports confirmed that the index of gut microbial a-diversity of 3.3 ADHD and gut microbiome
1-year-old children showed a close correlation with cognitive
functions at 2-year-old (Carlson et al., 2018). Furthermore, ADHD is a common childhood-onset neurodevelopmental
additional human studies have suggested that the first year of life disorder that persists into adulthood, with a worldwide prevalence
is the most authoritative period in human cognitive development of 5% (Polanczyk et al., 2007). ADHD is distinguished by symptom
(Figure 2B). These results indicate that the occurrence risk of domains of inattention, hyperactivity, and/or impulsivity. Although
diseases might be increased during fetal development and early some children might not reach the threshold of full diagnosis,
life stages. ADHD traits are continuously distributed throughout the
population (Brikell et al., 2021). ADHD is a complex genetic
disorder with a high heritability rate of 76%; however, this
3.2 ASD and gut microbiome estimate encompasses gene by environment interaction and such
interactions may account for much of the etiology of ADHD
In general, ASD patients frequently coincidently have various (Faraone and Larsson, 2019). Diverse environmental factors,
gastrointestinal disorders, and it might be due to the tight including perinatal factors (prematurity, low birth weight) and
association between gut microbial disturbances and mental psychosocial determinants (adoption, child neglect), have been
health. Moreover, these connections in ASD individuals have reported as reasonable factors to ADHD.
suggested increased gut permeability, called “leaky gut”. If gut However, there are few reports on the role of the gut
permeability arose, the gut barrier allows to cross the bacterial microbiome in ADHD patients. A recent systematic review found
metabolites into animal’s body, and it could negatively affect that all six included studies had distinct taxon findings between
neurodevelopment during early childhood through the gut-brain patients with ADHD and healthy controls. However, results varied
axis. Fowlie and colleagues demonstrated that psychobiotics between studies, and there was minimal consensus on which
treatment in ASD patients has a potential to relieve ASD bacterial taxa correlated most with ADHD (Sukmajaya et al., 2021).
symptoms by modulation of the gut microbiome (Fowlie In the case of ADHD, there have been multiple attempts to find
et al., 2018). relationships between clinical features and differences of this
An important feature of ASD is its marked comorbidity with disorder and healthy samples based on the gut-brain axis concept.
gastrointestinal symptoms. A high rate of ASD patients, ranging A study that attempted to associate gut microbiota and plasma
from 9% to 90%, report comorbid gastrointestinal symptoms such cytokine levels with ADHD showed a higher abundance of three
as constipation, abdominal pain, diarrhea, gas, and vomiting genera (Agathobacter, Anaerostipes, and Lachnospira) and
(Vuong and Hsiao, 2017). Moreover, the observed GI decreased levels of TNF-a in the ADHD group compared with
disturbances were strongly correlated with ASD severity. These that of the control group (Wang et al., 2022). However, the studies
GI problems suggest that the intestine plays an important role in conducted so far have not been able to show clear relationships
ASD pathogenesis. Previous studies on microbiota composition in between microbial taxa and ADHD, compared to the relatively
patients with ASD have shown highly heterogeneous results more established ASD studies (Bundgaard-Nielsen et al., 2020).
between studies, but the majority of them have found that the Considering the relative paucity of scientific literature, more
overall microbiota composition of ASD cases is different from that research efforts to clarify the possible relationship between
of the controls (Bundgaard-Nielsen et al., 2020). However, no ADHD and the gut-brain axis are required.
specific bacteria are consistently associated with ASD diagnosis or
severity in the literature.
Specifically for ASD, the specific composition of microbial taxa 4 Psychobiotics: potential roles on
in the human gut, including Firmicutes/Bacteroidetes ratio, is MGB axis as treatment target
reported to differ between the control and patient groups, with
Fusobacteria and Verrucomicrobia abundances being lower in the Psychobiotics, next-generation probiotics (NGPs) for the brain,
patient group (De Angelis et al., 2013). Microbial taxa have specific are a special class of probiotics that positively affect neurological
function and can be applied for the treatment of psychiatric diseases spectrum from stress alleviation to being an adjuvant in the
(Cheng et al., 2019). They are different from typical probiotics in treatment of diverse neuro-developmental and degenerative
their ability to affect the gut-brain axis by modulating microbial diseases (ADHD, ASD, Parkinson’s disease, and Alzheimer’s
composition, immune activation, VN signaling, and production of disease). Generally, conventional psychobiotic bacteria belong to
neuroactive metabolites, such as neurotransmitters, cytokines, the family Lactobacilli, and Bifidobacteria (Sharma et al., 2021).
SCFAs, and enteroendocrine hormones (Bermú dez-Humará n A summarized overview of clinical studies on the use of
et al., 2019; Kwak et al., 2021; Morais et al., 2021). Considering psychobiotics and FMT in individuals with ASD or ADHD is
this potential, psychobiotics have a wide-ranging application shown in Table 1.
TABLE 1 Treatment trials with psychobiotics and fecal microbial transportation in ASD and ADHD patients1.
ASD symptoms
35 patients 10 ↓
ASD Probiotics L. plantarum PS128 6.0 × 10 CFU (Kong et al., 2021)
(3–25 years) Veillonella, ↑
Streptococcus ↑
ASD symptoms
22 patients 9 ↓ (Kałużna-Czapliń ska and
ASD Probiotics L. acidophilus 5.0 × 10 CFU
(4–10 years) Urinary Błaszczyk, 2012)
arabinitol ↓
ASD symptoms
↓
Gut symptoms
30 patients B. longum, L. rhamnosus,
ASD Probiotics 5.0 × 108 CFU ↓ (Shaaban et al., 2018)
(5–9 years) L. acidophilus
Bifidobacterium
↑
Lactobacillus ↑
Behavior
41 patients improved
ASD Prebiotics Galactooligosaccharide 1.8 g for 6 months (Grimaldi et al., 2018)
(4–11 years) Bifidobacterium
↑
Behavior
8 patients B. infantis 2.0 × 1011 CFU improved
ASD Synbiotics (Sanctuary et al., 2019)
(2–11 years) Bovine colostrum 5.0– 10.0 g/day Gut symptoms
↓
(Continued)
TABLE 1 Continued
Standardized human
18 children ASD symptoms
ASD FMT gut microbiota (Hamilton, Weingarden 2.5 × 1012 cells/day (Kang et al., 2017)
(7–17 years) ↓
et al., 2012)
ASD symptoms
Standardized human
18 children ↓
ASD FMT gut microbiota (Hamilton, Weingarden 2.5 × 1012 cells/day (Kang et al., 2019)
(7–17 years) Improved
et al., 2012)
behavior
ASD symptoms
Standardized human
40 children 12 ↓
ASD FMT gut microbiota (Hamilton, Weingarden 2.5 × 10 cells/day (Li et al., 2021)
(3–17 years) Improved
et al., 2012)
behavior
Improve QoL
35 patients
ADHD Probiotics L. rhamnosus GG 1.0 × 1010 CFU Improve (Kumperscak et al., 2020)
(4–17 years)
cytokines
ADHD
30 patients
ADHD Probiotics B. bifidum Bf-688 5.0 × 109 CFU symptom ↓ (Wang et al., 2022)
(4–16 years)
Weight gain ↑
L. mesenteroides, L. paracasei,
66 patients 4.0 × 1011 CFU ADHD
ADHD Synbiotics L. plantarum (Skott et al., 2020)
(5–55 years) 2.5 g of prebiotics symptom ↓
B-glucan, inulin, pectin, starch
ADHD
symptoms ↓
34 patients L. reuteri, L. acidophilus,
ADHD Probiotics 8.0 × 109 CFU Inflammation ↓ (Sepehrmanesh et al., 2021)
(8–12 years) L. fermentum, B. bifidum
Oxidative stress
↓
ADHD
1 patient symptom ↓
ADHD FMT Healthy doner’s microbiota Not applicable (Hooi et al., 2022)
(22 years) F. prausnitzii ↑
B. longum ↓
1
Abbreviations: autism spectrum disorders (ASD), attention deficit/hyperactivity disorder (ADHD), fecal microbial transplantation (FMT).
administration on gut microbiota composition in children with plantarum, L. acidophilus, and L. paracacei) attenuated the
ASD (Tomova et al., 2015). The abundance of Clostridia and inflammatory responses in a VPA-induced rodent ASD model. In
Desulfovibrio and Bacteroidetes/Firmicutes ratio were related to particular, this study also showed that psychobiotic treatment
the severity of ASD and gastrointestinal symptoms. After decreased serum pro-inflammatory cytokine, IL-6 levels, and
probiotics treatment, the amount of Firmicutes significantly increased anti-inflammatory cytokine, IL-10 levels, with the
decreased, which resulted in an increase in the Bacteroidetes/ improved status of diverse behavior tests including social
Firmicutes ratio to a level similar to that observed in healthy interaction, anxiety, and repetitive behaviors (Adıgüzel et al.,
children, Bifidobacterium increased, and Desulfovibrio decreased 2022). Alonazi also demonstrated that dietary psychobiotic
significantly. Moreover, a study of a mixture of Lactobacillus spp. supplementation could reduce levels of various serum
and Bifidobacterium spp. in two different rodent ASD models inflammatory cytokines, such as IL-1b, IL-8, IL-10, and IFN-g, in
indicated that a probiotic mixture could improve social behavioral an ASD rat model induced by a neurotoxic dose of propionic acid
symptoms by modulating the gut microbial population (Mintá l (Alonazi et al., 2022).
et al., 2022).
4.1.3 Neural pathway and chemical signaling
4.1.2 Anti-inflammation and immunomodulation 4.1.3.1 Changing microbial signals (neuroendocrine
4.1.2.1 Psychobiotics as immunomodulators signaling)
Psychobiotics have a potential to not only reconstruct the gut The gut microbiota influences the brain directly through neural
barrier function by resisting harmful bacteria, but also exert an pathways, including the VN and ENS. The VN connects the ENS
immunomodulatory effect by reducing circulating hormones and and CNS and it could be activated by cytokines, which could be
pro-inflammatory cytokines in serum. The gut microbiota has been modulated by bacteria, and byproducts from bacteria including
demonstrated to serve as a regulator of intestinal, systemic, and endotoxins and peptides. In particular, the neuropeptide could be
CNS resident immune cell function (Zheng D et al., 2020). Gut sensed by receptors associated with dendritic cells in the gut, which
microbiota can communicate with the CNS by regulating intestinal could transfer the signals to the brain (Perez-Burgos et al., 2013).
and peripheral immune cells and peripheral immune responses via Psychobiotics modulate CNS-related behaviors through the VN
circulating cytokines (Arrieta and Finlay, 2012). pathway and the physiological response of various metabolites,
including SCFAs, enteroendocrine hormones, cytokines, and
4.1.2.2 Psychobiotics reduce inflammation neurotransmitters (Bravo et al., 2011; Dinan et al., 2014; Sgritta
Any peripheral inflammatory event induces VN to cause the et al., 2019).
suppression of the release of proinflammatory cytokines from
intestinal macrophages (Daliri et al., 2016). Probiotics reduce gut 4.1.3.2 Hormones and metabolic changes
inflammation through various mechanisms, such as reducing The levels of oxytocin and DHEA-S, which have been
inflammatory cytokines and other immunomodulatory effects. For considered to be etiologies of ASD, were significantly lower in the
example, anti-inflammatory cytokines (IL-4 and IL-10) and plasma of children with ASD in a clinical study, and there was a
proinflammatory cytokines (TNF-a, IL-1b, IL-2, IL-6, IL-8, IL-12, trend towards a correlation between decreased DHEA-S levels and a
IL-17, and IL-18) are significantly changed by Lactobacillus lower Bacteroidetes/Firmicutes ratio which increased after probiotic
rhamnosus GG (LGG) (Miyazawa et al., 2015; Cicenia et al., 2016; implementation (Tomova et al., 2015). According to Grimaldi,
Fong et al., 2016; Wang et al., 2016; Aoki-Yoshida et al., 2017; increases in butyrate production, potentially positively affecting
Clarke, 2018; Cai et al., 2019). ASD, were detected in children with ASD following exclusion
diets (Nankova et al., 2014; Grimaldi et al., 2018). Additionally,
4.1.2.3 Inflammatory cytokines change in ASD lower levels of amino acids (isoleucine, leucine, valine, alanine, and
In clinical studies, Tomova et al. found that TNF-a levels were glutamine) and lactate were detected in the B-GOS group. The
strongly correlated with GI symptoms and showed a trend toward presence of amino acids in feces is associated with problems in gut
correlation with ASD severity (Tomova et al., 2015). Probiotic barrier function (Marchesi et al., 2007).
supplementation significantly decreased TNF-a levels in the feces
of children with ASD. Similar to this study, Sanctuary demonstrated 4.1.3.3 Gut microbiome modulation and
that psychobiotic supplementation could reduce the intracellular GABA metabolism
expression of certain cytokines in CD4+ T cells (Sanctuary et al., A study of psychobiotics in a rodent ASD model, which was
2019). The frequency of CD4+/IL-13+ T cells was significantly induced by oral propionic acid ingestion, proposed that
lower after the treatment. Lactobacillus bulgaricus and Bifidobacterium infantis could
ameliorate glutamate excitotoxicity, a major autistic feature in this
4.1.2.4 Animal studies on the anti-inflammatory effects model. The therapeutic effect of these psychobiotics might be due to
of psychobiotics the reduction of oxidative stress, restoration of the depleted GABA
Adıgüzel et al. demonstrated that dietary treatment with signaling pathway, and upregulation of the GABA receptor’s gene
multispecies probiotics formulations (Sptreptococcus thermophilus, expression (Bin-Khattaf et al., 2022). Ingestion of Lactobacillus
Bifidobacterium breve, B. animalis, Lactobacillus helveticus, L. rhamnosus could connect bidirectional communication of the
gut-brain axis, and it could regulate emotional behaviors by 4.2.1.3 Synthesis of serotonin beyond BBB
controlling the GABA receptor expression in the VN (Bravo Serotonin also plays a role in ADHD pathogenesis, however, it
et al., 2011). In 2018, the specific bacterial species of ASD were affects brain function not directly, but via the nervous system
identified in Shank3 knock-out mice, and this study suggested that (Banerjee and Nandagopal, 2015; Hou et al., 2018). On the other
oral Lactobacillus reuteri ingestion could decrease repetitive hand, gut microbiota directly act a biological role on the brain by
behaviors by up-regulation of the g-aminobutyric acid (GABA)- modulation tryptophan’s peripheral availability because tryptophan
related metabolism (Tabouy et al., 2018). can cross the BBB and affect serotonin synthesis in CNS (Richard
et al., 2009; Schwarcz and Stone, 2017).
4.2.3 Immune pathway and anti-inflammation the healthy group. Especially, the FMT response significantly
4.2.3.1 Anti-inflammatory effects of probiotics reduces the abundance of Eubacterium coprostanoligenes (Li et al.,
Lactobacillus rhamnosus GG is known to strengthen the gut 2021). These data also indicated that decrement in the population of
permeability barrier by fortifying intestinal tight junctions, mucin Eubacterium coprostanoligenes by FMT might be a curative
layer thickness, and antigen-specific immunoglobulin A production technique for ASD symptoms and behaviors.
(Asano et al., 2012). In particular, L. rhamnosus GG administrated
participants showed a significant decrease in the serum levels of the 5.1.2 Modulating neurotransmitters
pro-inflammatory cytokines (IL-6, IL-12 p70, and TNF-a). 5.1.2.1 FMT alters the serum levels of neurotransmitters
(Kumperscak et al., 2020). Unlike probiotics, FMT refers to the transfer of the full spectrum of
gut microbial communities containing more than 1,000 bacterial strains,
and it might be more effective than psychobiotics in aspects of
5 FMT: rebuilding gut microecology physiological regulation in the nervous system, endocrine system, and
host behavior (Chen et al., 2022). In recent studies, FMT could exhibit a
The definition of FMT is the transfer technique of a healthy donor’s recovery effect on the serum levels of serotonin, GABA, and DA in the
fecal specimen to the GI tract of a recipient patient to reestablish the ASD cohort, which means that FMT might be an effective technique in
normal gut microbiome. This technique has been focused in recent years regulating neurotransmitters via the MGB axis (Li et al., 2021). Moreover,
because of the technical advances in metagenomics sequencing and the FMT in the ASD cohort could decrease GABA and serotonin in serum,
growing understanding of its function. FMT has been demonstrated to but the dopamine level was increased by FMT. It could be assumed that
be able to reconstruct a normally functioning microbial community, FMT may be an efficient approach to modulate neurotransmitter
making it an accepted therapy with biological plausibility. Considering secretion for regulation of the central nerve via the MGB axis.
the effect of FMT on the reorganization of gut microbiota, it is considered
to have the potential for the treatment of neurodevelopmental diseases 5.1.3 Regulating immune responses
such as ASD through the interaction of the MGB axis. It is necessary to 5.1.3.1 Chemokines and microbiome
determine the optimal composition of the microbiome to be used for Alterations in the gut microbiota composition after FMT could
FMT by clarifying the structure or functional profile of the microbes significantly improve behavioral impairments and regulate immune
associated with improved clinical outcomes (Zhuang et al., 2019). responses in ASD. Chen and colleagues demonstrated that treatment
Bacterial diversity and health-associated functions, such as colonization using FMT with in vitro cultured healthy donor’s intestinal microbiota
resistance, can be restored using FMT. In addition to bioactive had a positive effect on ASD symptoms in mouse ASD model (Chen
compounds, FMT is also a source of microbes, such as phages. These et al., 2020). They observed amelioration of anxiety actions and
components come together in a symbiotic community, allowing better repetitive performance with lower serum levels of metabolites, such
colonization of the GI tract (Goldenberg et al., 2018). In terms of the gut as GRO-a and MIP-1a, and a conversely higher level of MCP-3,
microbiota, FMT is considered an untargeted intervention. RANTES, and Eotaxin. Additionally, family or genus levels of S24-7,
Clostridiaceae, Prevotella, and Candidatus Arthromitus were key
microbial taxa in FMT treatment, and serum levels of chemokines
5.1 Therapeutic mechanisms and their were related to the relative abundance of these taxa.
effects on ASD
5.1.3.2 Original donor vs in vitro cultured
5.1.1 Altering gut ecosystem In this study, both original donor microbiota transplantation
5.1.1.1 Bacterial diversity and cultured microbiota transplantation improved behavioral
FMT could serve as a protective treatment for reconstructing the abnormalities and chemokine disorders in an ASD mouse model
gut microbiota at both the phylum and genus levels and has a and were effective in the modification of several key differential taxa
therapeutic effect on ASD symptoms and gastrointestinal disorders in the gut microbial composition (Chen et al., 2020). These results
(Li et al., 2021). A modified FMT protocol for children with ASD, of cultured microbiota transplantation suggest the possibility of
termed microbiota transfer therapy, appears to be a promising using “donor-free FMT” and regulating the donor gut microbiota
approach to alter the gut microbiome and improve GI and structure before transplantation during in vitro culture. The batch
behavioral symptoms of ASD (Kang et al., 2017; Kang et al., 2019). methods are fast, easy, and repeatable culturing techniques.
This protocol improved GI and ASD symptoms, and the microbiome
persisted for two years after treatment, suggesting a long-term impact.
Important changes in the gut microbiota at the end of treatment were 5.2 Therapeutic mechanisms/effects on
observed during follow-ups, including significant increases in bacterial ADHD
diversity and relative abundance of Bifidobacteria and Prevotella.
5.2.1 Neuroprotective effects of the
5.1.1.2 Engraftment of the donor microbiome transplanted microbiome
Li et al. showed that the gut microbial population of ASD A case report provides preliminary evidence regarding the use of
children was altered by FMT with donor microbiota toward that of FMT in a patient with C. difficile infection and ADHD. The authors
suggested that gut microbiome modulation, particularly the gain or loss microbiome to improve the GI and behavioral symptoms of patients
of specific microbial species and pathways involving the metabolism of with ASD or ADHD. Development of the gut microbiome in early life
SCFAs, tryptophan, and GABA, may merit further exploration as a plays an important role in the overall well-being of humans.
potential therapeutic strategy for ADHD (Hooi et al., 2022). Among Numerous studies have demonstrated that early alterations in the
bacteria engrafted through FMT, F. prausnitzii may reduce gut microbiome are closely related to neurodevelopmental disorders,
neuroinflammation and alleviate ADHD symptoms. F. prausnitzii such as ASD and ADHD. Nevertheless, the ambiguous and equivocal
exhibits anti-inflammatory effects by increasing anti-inflammatory evidence of clinical studies makes it difficult to believe the therapeutic
cytokines and decreasing inflammatory cytokines that promote method targeting the MGB axis. To better understand the role of the
neuroinflammation and development of ADHD. L. ruminis possesses gut microbiome in heterogeneous and complex ASD/ADHD
genes contributing to the pentose phosphate pathway, which pathogenesis, double-blind, randomized, controlled trials and
contributes to SCFA production. Engraftment of Lactobacillus genus treatments tailored to individual characteristics and the host
may exert neuroprotection by producing anti-inflammatory SCFAs. microbiome are recommended. In particular, the process of
(Basen and Kurrer, 2021). intestinal microbiota colonization and establishment in the early
stage of life is crucially affected by maternal conditions/diseases, mode
of delivery, and exposure to antibiotics. Therefore, future studies are
6 Future perspectives and needed to determine more accurate therapeutic targets in immune,
concluding remarks metabolic, endocrine, and neural pathways by mechanism validation
through culturomics experiments of mainly modulated microbial
Although the application of pre- and probiotics as psychobiotics populations and metabolomic analysis of the mother’s skin, vagina,
remains promising, it is feasible that the effect of psychobiotics will gut microbiota, and infant gut environments.
be decreased over time due to the significant influence of
environmental reasons occurred in child development (Grimaldi
et al., 2018). Further studies should be needed to address the drug Author contributions
administration timing, the effect of different strain combinations,
safety, and efficacy of probiotics. Furthermore, the novel therapeutic All authors researched the data for this article, made substantial
functions of the psychobiotics and commensal bacteria will be contributions to discussions of the content, wrote the article, and
investigated in synthetic biology fields (Bin-Khattaf et al., 2022). reviewed and/or edited the manuscript prior to submission.
For example, Korpela proposed that oral-fecal transplantation with
diluted fecal samples from the maternal gut microbiome could
restore normal gut microbiota in Cesarean-born infants (Korpela Funding
et al., 2020). Furthermore, a study demonstrated that using E. coli
native to the target murine host to knock-in specific functions and This research was supported by the research fund of Hanyang
apply them back to the host enabled the perpetual engraftment of University MEB (Global Center for Developmental Disorders, HY-
transgenic bacteria in the intestine, which was demonstrated until 201900000003070) and the National Research Foundation of Korea
the transformation stage of human borne E. coli (Asano et al., 2012). (NRF) funded by the Ministry of Science and ICT (RS-2023-
As such, various studies have focused on fortifying the modulating 00219983).
effects on the gut microbiome via adjustment at the molecular level.
The use of probiotics is feasible in children, and short-term
supplementation has been shown to be safe. However, the long-term Conflict of interest
effects of repeated applications on the gut microbiome and the safety
concerns of treatment are unknown. The actual efficacy of FMT has The authors declare that the research was conducted in the
been proved by various studies using diverse animal models. However, absence of any commercial or financial relationships that could be
Safety is the most important aspect in the FMT study because most construed as a potential conflict of interest.
ASD patients are children. In previous studies, the oral ingestion of
human fecal suspensions was considered an unpleasant experience for
patients and might cause side effects, including extra ingestion with Publisher’s note
acid inhibitors. As it has been known that a colon-release capsule
coated with acid-resistant hydroxypropyl cellulose is the best All claims expressed in this article are solely those of the authors
formulation for patients (Kang et al., 2019). and do not necessarily represent those of their affiliated organizations,
This review summarizes the current knowledge on the positive or those of the publisher, the editors and the reviewers. Any product
effects and potential pathways of promising therapeutic interventions, that may be evaluated in this article, or claim that may be made by its
including psychobiotic supplementation and modulation of the gut manufacturer, is not guaranteed or endorsed by the publisher.
References
Aarts, E., Ederveen, T. H., Naaijen, J., Zwiers, M. P., Boekhorst, J., Timmerman, H. Cheroni, C., Caporale, N., and Testa, G. (2020). Autism spectrum disorder at the
M., et al. (2017). Gut microbiome in ADHD and its relation to neural reward crossroad between genes and environment: contributions, convergences, and
anticipation. PloS One 12 (9), e0183509. doi: 10.1371/journal.pone.0183509 interactions in ASD developmental pathophysiology. Mol. Autism. 11 (1), 69.
Adıgüzel, E., Ç içek, B., Ünal, G., Aydın, M. F., and Barlak-Keti, D. (2022). Probiotics doi: 10.1186/s13229-020-00370-1
and prebiotics alleviate behavioral deficits, inflammatory response, and gut dysbiosis in Choi, G. B., Yim, Y. S., Wong, H., Kim, S., Kim, H., Kim, S. V., et al. (2016). The
prenatal VPA-induced rodent model of autism. Physiol. Behav. 256, 113961. maternal interleukin-17a pathway in mice promotes autism-like phenotypes in
doi: 10.1016/j.physbeh.2022.113961 offspring. Science 351 (6276), 933–939. doi: 10.1126/science.aad0314
Alonazi, M., Ben Bacha, A., Al Suhaibani, A., Almnaizel, A. T., Aloudah, H. S., and Cicenia, A., Santangelo, F., Gambardella, L., Pallotta, L., Iebba, V., Scirocco, A., et al.
El-Ansary, A. (2022). Psychobiotics improve propionic acid-induced (2016). Protective role of postbiotic mediators secreted by lactobacillus rhamnosus GG
neuroinflammation in juvenile rats, rodent model of autism. Transl. Neurosci. 13 (1), versus lipopolysaccharide-induced damage in human colonic smooth muscle cells. J.
292–300. doi: 10.1515/tnsci-2022-0226 Clin. Gastroenterol. 50, S140–S144. doi: 10.1097/MCG.0000000000000681
Aoki-Yoshida, A., Saito, S., Tsuruta, T., Ohsumi, A., Tsunoda, H., and Sonoyama, K. Clarke, T. C. (2018). The use of complementary health approaches among US adults
(2017). Exosomes isolated from sera of mice fed lactobacillus strains affect with a recent cancer diagnosis. J. Altern. Complement. Med. 24 (2), 139–145.
inflammatory cytokine production in macrophages iná vitro. Biochem. Biophys. Res. doi: 10.1089/acm.2016.0182
Commun. 489 (2), 248–254. doi: 10.1016/j.bbrc.2017.05.152 Cryan, J. F., O'Riordan, K. J., Cowan, C. S., Sandhu, K. V., Bastiaanssen, T. F.,
Arnold, L. E., Luna, R. A., Williams, K., Chan, J., Parker, R. A., Wu, Q., et al. (2019). Boehme, M., et al. (2019). The microbiota-gut-brain axis. Physiol. Rev. 99 (4), 1877–
Probiotics for gastrointestinal symptoms and quality of life in autism: a placebo-controlled 2013. doi: 10.1152/physrev.00018.2018
pilot trial. J. Child Adolesc. Psychopharmacol. 29 (9), 659–669. doi: 10.1089/cap.2018.0156 Dalile, B., Van Oudenhove, L., Vervliet, B., and Verbeke, K. (2019). The role of short-
Arrieta, M. C., and Finlay, B. B. (2012). The commensal microbiota drives immune chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol.
homeostasis. Front. Immunol. 3. doi: 10.3389/fimmu.2012.00033 Hepatol. 16 (8), 461–478. doi: 10.1038/s41575-019-0157-3
Arrieta, M. C., Stiemsma, L. T., Amenyogbe, N., Brown, E. M., and Finlay, B. (2014). Daliri, E. B. M., Oh, D. H., and Lee, B. H. (2016). Psychobiotics; a promise for
The intestinal microbiome in early life: health and disease. Front. Immunol. 5. neurodevelopmental therapy. J. Probiotics Health 4, 1e4. doi: 10.4172/2329-
doi: 10.3389/fimmu.2014.00427 8901.1000146
Asano, Y., Hiramoto, T., Nishino, R., Aiba, Y., Kimura, T., Yoshihara, K., et al. De Angelis, M., Piccolo, M., Vannini, L., Siragusa, S., De Giacomo, A., Serrazzanetti,
(2012). Critical role of gut microbiota in the production of biologically active, free D. I., et al. (2013). Fecal microbiota and metabolome of children with autism and
catecholamines in the gut lumen of mice. Am. J. Physiol. Gastrointest. Liver Physiol. 303 pervasive developmental disorder not otherwise specified. PloS One 8 (10), e76993.
(11), G1288–G1295. doi: 10.1152/ajpgi.00341.2012 doi: 10.1371/journal.pone.0076993
Bäckhed, F., Roswall, J., Peng, Y., Feng, Q., Jia, H., Kovatcheva-Datchary, P., et al. Del Campo, N., Chamberlain, S. R., Sahakian, B. J., and Robbins, T. W. (2011). The
(2015). Dynamics and stabilization of the human gut microbiome during the first year roles of dopamine and noradrenaline in the pathophysiology and treatment of
of life. Cell Host Microbe 17 (5), 690–703. doi: 10.1016/j.chom.2015.04.004 attention-deficit/hyperactivity disorder. Biol. Psychiatry 69 (12), e145–e157.
Banerjee, E., and Nandagopal, K. (2015). Does serotonin deficit mediate doi: 10.1016/j.biopsych.2011.02.036
susceptibility to ADHD? Neurochem. Int. 82, 52–68. doi: 10.1016/j.neuint.2015.02.001 Del Toro-Barbosa, M., Hurtado-Romero, A., Garcia-Amezquita, L. E., and Garcı́a-
Basen, M., and Kurrer, S. E. (2021). A close look at pentose metabolism of gut Cayuela, T. (2020). Psychobiotics: mechanisms of action, evaluation methods and
bacteria. FEBS J. 288 (6), 1804–1808. doi: 10.1111/febs.15575 effectiveness in applications with food products. Nutrients 12 (12), 3896. doi: 10.3390/
nu12123896
Bermú dez-Humará n, L. G., Salinas, E., Ortiz, G. G., Ramirez-Jirano, L. J., Morales, J.
A., and Bitzer-Quintero, O. K. (2019). From probiotics to psychobiotics: live beneficial Dinan, T. G., Borre, Y. E., and Cryan, J. F. (2014). Genomics of schizophrenia: time
bacteria which act on the brain-gut axis. Nutrients 11 (4), 890. doi: 10.3390/nu11040890 to consider the gut microbiome? Mol. Psychiatry 19 (12), 1252–1257. doi: 10.1038/
mp.2014.93
Bin-Khattaf, R. M., Alonazi, M. A., Al-Dbass, A. M., Almnaizel, A. T., Aloudah, H. S.,
Soliman, D. A., et al. (2022). Probiotic ameliorating effects of altered GABA/glutamate Dinan, T. G., and Cryan, J. F. (2017). Brain–gut–microbiota axis–mood, metabolism
signaling in a rodent model of autism. Metabolites 12 (8), 720. doi: 10.3390/ and behaviour. Nat. Rev. Gastroenterol. Hepatol. 14 (2), 69–70. doi: 10.1038/
metabo12080720 nrgastro.2016.200
Bravo, J. A., Forsythe, P., Chew, M. V., Escaravage, E., Savignac, H. M., Dinan, T. G., Enticott, P. G., Rinehart, N. J., Tonge, B. J., Bradshaw, J. L., and Fitzgerald, P. B.
et al. (2011). Ingestion of lactobacillus strain regulates emotional behavior and central (2010). A preliminary transcranial magnetic stimulation study of cortical inhibition
GABA receptor expression in a mouse via the vagus nerve. Proc. Nat. Acad. Sci. 108 and excitability in high-functioning autism and asperger disorder. Dev. Med. Child
(38), 16050–16055. doi: 10.1073/pnas.110299910 Neurol. 52 (8), e179–e183. doi: 10.1111/j.1469-8749.2010.03665.x
Brikell, I., Burton, C., Mota, N. R., and Martin, J. (2021). Insights into attention- Erkosar, B., Storelli, G., Defaye, A., and Leulier, F. (2013). Host-intestinal microbiota
deficit/hyperactivity disorder from recent genetic studies. Psychol. Med. 51 (13), 2274– mutualism:”learning on the fly”. Cell Host Microbe 13 (1), 8–14. doi: 10.1016/
2286. doi: 10.1017/S0033291721000982 j.chom.2012.12.004
Bundgaard-Nielsen, C., Knudsen, J., Leutscher, P. D., Lauritsen, M. B., Nyegaard, M., Faraone, S. V., and Larsson, H. (2019). Genetics of attention deficit hyperactivity
Hagstrøm, S., et al. (2020). Gut microbiota profiles of autism spectrum disorder and disorder. Mol. Psychiatry 24 (4), 562–575. doi: 10.1038/s41380-018-0070-0
attention deficit/hyperactivity disorder: a systematic literature review. Gut Microbes 11 Fong, F. L. Y., Kirjavainen, P. V., and El-Nezami, H. (2016). Immunomodulation of
(5), 1172–1187. doi: 10.1080/19490976.2020.1748258 lactobacillus rhamnosus GG (LGG)-derived soluble factors on antigen-presenting cells
Bundgaard-Nielsen, C., Lauritsen, M. B., Knudsen, J. K., Rold, L. S., Larsen, M. H., of healthy blood donors. Sci. Rep. 6 (1), 22845. doi: 10.1038/srep22845
Hindersson, P., et al. (2023). Children and adolescents with attention deficit Fowlie, G., Cohen, N., and Ming, X. (2018). The perturbance of microbiome and gut-
hyperactivity disorder and autism spectrum disorder share distinct microbiota brain axis in autism spectrum disorders. Int. J. Mol. Sci. 19 (8), 2251. doi: 10.3390/
compositions. Gut Microbes 15 (1), 2211923. doi: 10.1080/19490976.2023.2211923 ijms19082251
Cai, Z., He, W., Zhuang, F. J., and Chen, Y. (2019). The role of high high-sensitivity Galland, L. (2014). The gut microbiome and the brain. J. Med. Food 17 (12), 1261–
c-reactive protein levels at admission on poor prognosis after acute ischemic stroke. Int. 1272. doi: 10.1089/jmf.2014.7000
J. Neurosci. 129 (5), 423–429. doi: 10.1080/00207454.2018.1538139 Ghanaatgar, M., Taherzadeh, S., Ariyanfar, S., Jahromi, S. R., Martami, F., Gharaei, J.
Carlson, A. L., Xia, K., Azcarate-Peril, M. A., Goldman, B. D., Ahn, M., Styner, M. A., M., et al. (2022). Probiotic supplement as an adjunctive therapy with Ritalin for
et al. (2018). Infant gut microbiome associated with cognitive development. Biol. treatment of attention-deficit hyperactivity disorder symptoms in children: a double-
Psychiatry 83 (2), 148–159. doi: 10.1016/j.biopsych.2017.06.021 blind placebo-controlled randomized clinical trial. Nutr. Food Sci. 53 (1), 19–34.
Chen, K., Fu, Y., Wang, Y., Liao, L., Xu, H., Zhang, A., et al. (2020). Therapeutic doi: 10.1108/NFS-12-2021-0388
effects of the in vitro cultured human gut microbiota as transplants on altering gut Goldenberg, S. D., Batra, R., Beales, I., Digby-Bell, J. L., Irving, P. M., Kellingray, L.,
microbiota and improving symptoms associated with autism spectrum disorder. et al. (2018). Comparison of different strategies for providing fecal microbiota
Microb. Ecol. 80, 475–486. doi: 10.1007/s00248-020-01494-w transplantation to treat patients with recurrent clostridium difficile infection in two
Chen, Y., Xueying, Z., Jiaqu, C., Qiyi, C., Huanlong, Q., Ning, L., et al. (2022). English hospitals: a review. Infect. Dis. Ther. 7, 71–86. doi: 10.1007/s40121-018-0189-y
FTACMT study protocol: a multicentre, double-blind, randomised, placebo-controlled Grimaldi, R., Gibson, G. R., Vulevic, J., Giallourou, N., Castro-Mejı́a, J. L., Hansen, L.
trial of faecal microbiota transplantation for autism spectrum disorder. BMJ Open 12 H., et al. (2018). A prebiotic intervention study in children with autism spectrum
(1), e051613. doi: 10.1136/bmjopen-2021-051613 disorders (ASDs). Microbiome 6 (1), 1–13. doi: 10.1186/s40168-018-0523-3
Cheng, L. H., Liu, Y. W., Wu, C. C., Wang, S., and Tsai, Y. C. (2019). Psychobiotics in Grossi, E., Melli, S., Dunca, D., and Terruzzi, V. (2016). Unexpected improvement in
mental health, neurodegenerative and neurodevelopmental disorders. J. Food Drug core autism spectrum disorder symptoms after long-term treatment with probiotics.
Anal. 27 (3), 632–648. doi: 10.1016/j.jfda.2019.01.002 SAGE open med. Case Rep. 4, 2050313X16666231. doi: 10.1177/2050313X16666231
Guidetti, C., Salvini, E., Viri, M., Deidda, F., Amoruso, A., Visciglia, A., et al. (2022). Ming, X., Chen, N., Ray, C., Brewer, G., Kornitzer, J., and Steer, R. A. (2018). A gut
Randomized double-blind crossover study for evaluating a probiotic mixture on feeling: a hypothesis of the role of the microbiome in attention-deficit/hyperactivity
gastrointestinal and behavioral symptoms of autistic children. J.Clin. Med. 11 (18), disorders. Child Neurol. Open 5, 2329048X18786799. doi: 10.1177/2329048X18786799
5263. doi: 10.3390/jcm11185263 Mintá l, K., Tó th, A., Hormay, E., Ková cs, A., Lá szló , K., Bufa, A., et al. (2022). Novel
Hooi, S. L., Dwiyanto, J., Rasiti, H., Toh, K. Y., Wong, R. K. M., and Lee, J. W. J. probiotic treatment of autism spectrum disorder associated social behavioral symptoms
(2022). A case report of improvement on ADHD symptoms after fecal microbiota in two rodent models. Sci. Rep. 12 (1), 5399. doi: 10.1038/s41598-022-09350-2
transplantation with gut microbiome profiling pre-and post-procedure. Curr. Med. Res. Miyazawa, K., Yoda, K., Kawase, M., Harata, G., and He, F. (2015). Influence of
Opin. 38 (11), 1977–1982. doi: 10.1080/03007995.2022.2129232 orally administered lactobacillus GG on respiratory immune response in a murine
Hou, Y. W., Xiong, P., Gu, X., Huang, X., Wang, M., and Wu, J. (2018). Association model of diet-induced obesity. Microbiol. Immunol. 59 (2), 99–103. doi: 10.1111/1348-
of serotonin receptors with attention deficit hyperactivity disorder: a systematic review 0421.12226
and meta-analysis. Curr. Med. Sci. 38, 538–551. doi: 10.1007/s11596-018-1912-3 Morais, L. H., Schreiber, H. L.IV, and Mazmanian, S. K. (2021). The gut microbiota–
Hsiao, E. Y., McBride, S. W., Hsien, S., Sharon, G., Hyde, E. R., McCue, T., et al. brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 19 (4), 241–255.
(2013). Microbiota modulate behavioral and physiological abnormalities associated doi: 10.1038/s41579-020-00460-0
with neurodevelopmental disorders. Cell. 155 (7), 1451–1463. doi: 10.1016/ Nagpal, J., and Cryan, J. F. (2021). Microbiota-brain interactions: moving toward
j.cell.2013.11.024 mechanisms in model organisms. Neuron 109 (24), 3930–3953. doi: 10.1016/
Jena, A., Montoya, C. A., Mullaney, J. A., Dilger, R. N., Young, W., McNabb, W. C., j.neuron.2021.09.036
et al. (2020). Gut-brain axis in the early postnatal years of life: a developmental Nankova, B. B., Agarwal, R., MacFabe, D. F., and La Gamma, E. F. (2014). Enteric
perspective. Front. Iinteg. Neurosci. 14. doi: 10.3389/fnint.2020.00044 bacterial metabolites propionic and butyric acid modulate gene expression, including
Jung, T. H., Hwang, H. J., and Han, K. S. (2022). Correlation of attention deficit CREB-dependent catecholaminergic neurotransmission, in PC12 cells-possible
hyperactivity disorder with gut microbiota according to the dietary intake of Korean relevance to autism spectrum disorders. PloS One 9 (8), e103740. doi: 10.1371/
elementary school students. PloS One 17 (9), e0275520. doi: 10.1371/ journal.pone.0103740
journal.pone.0275520 Niemarkt, H. J., De Meij, T. G., van Ganzewinkel, C. J., de Boer, N. K., Andriessen, P.,
Kalenik, A., Kardaś, K., Rahnama, A., Sirojć , K., and Wolań czyk, T. (2021). Gut Hütten, M. C., et al. (2019). Necrotizing enterocolitis, gut microbiota, and brain development:
microbiota and probiotic therapy in ADHD: a review of current knowledge. Prog. role of the brain-gut axis. Neonatology 115 (4), 423–431. doi: 10.1159/000497420
N eu ro ps yc ho ph ar m a col . B i o l . P s yc hi a tr y 1 1 0, 11 02 7 7. do i: 10 .1 01 6 / O’Mahony, S. M., Clarke, G., Borre, Y. E., Dinan, T. G., and Cryan, J. F. (2015).
j.pnpbp.2021.110277 Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain
Kałużna-Czapliń ska, J., and Błaszczyk, S. (2012). The level of arabinitol in autistic Res. 277, 32–48. doi: 10.1016/j.bbr.2014.07.027
children after probiotic therapy. Nutrition. 28 (2), 124–126. doi: 10.1016/ Pärtty, A., Kalliomäki, M., Wacklin, P., Salminen, S., and Isolauri, E. (2015). A
j.nut.2011.08.002 possible link between early probiotic intervention and the risk of neuropsychiatric
Kang, D. W., Adams, J. B., Coleman, D. M., Pollard, E. L., Maldonado, J., disorders later in childhood: a randomized trial. Pediatr. Res. 77 (6), 823–828.
McDonough-Means, S., et al. (2019). Long-term benefit of microbiota transfer doi: 10.1038/pr.2015.51
therapy on autism symptoms and gut microbiota. Sci. Rep. 9 (1), 5821. doi: 10.1038/ Perez-Burgos, A., Wang, B., Mao, Y. K., Mistry, B., Neufeld, K. A. M., Bienenstock, J.,
s41598-019-42183-0 et al. (2013). Psychoactive bacteria Lactobacillus rhamnosus (JB-1) elicits rapid
Kang, D. W., Adams, J. B., Gregory, A. C., Borody, T., Chittick, L., Fasano, A., et al. frequency facilitation in vagal afferents. Am. J. Physiol. Gastrointest. Liver Physiol.
(2017). Microbiota transfer therapy alters gut ecosystem and improves gastrointestinal 304 (2), G211–G220. doi: 10.1152/ajpgi.00128.2012
and autism symptoms: an open-label study. Microbiome 5 (1), 1–16. doi: 10.1186/ Polanczyk, G., De Lima, M. S., Horta, B. L., Biederman, J., and Rohde, L. A. (2007).
s40168-016-0225-7 The worldwide prevalence of ADHD: a systematic review and metaregression analysis.
Kho, Z. Y., and Lal, S. K. (2018). The human gut microbiome–a potential controller Am. J. Psychiatry 164 (6), 942–948. doi: 10.1176/appi.ajp.164.6.942
of wellness and disease. Front. Microbiol. 9, 1835. doi: 10.3389/fmicb.2018.01835 Prosperi, M., Santocchi, E., Guiducci, L., Frinzi, J., Morales, M. A., Tancredi, R., et al.
Kong, X. J., Liu, J., Liu, K., Koh, M., Sherman, H., Liu, S., et al. (2021). Probiotic and (2022). Interventions on microbiota: where do we stand on a gut–brain link in autism?
oxytocin combination therapy in patients with autism spectrum disorder: a A Systematic Review. Nutrients. 14 (3), 462. doi: 10.3390/nu14030462
randomized, double-blinded, placebo-controlled pilot trial. Nutrients. 13 (5), 1552. Rianda, D., Agustina, R., Setiawan, E. A., and Manikam, N. R. M. (2019). Effect of
doi: 10.3390/nu13051552 probiotic supplementation on cognitive function in children and adolescents: a
Korpela, K., Helve, O., Kolho, K. L., Saisto, T., Skogberg, K., Dikareva, E., et al. systematic review of randomised trials. Benef. Microbes 10 (8), 873–882.
(2020). Maternal fecal microbiota transplantation in cesarean-born infants rapidly doi: 10.3920/BM2019.0068
restores normal gut microbial development: a proof-of-concept study. Cell 183 (2), Richard, D. M., Dawes, M. A., Mathias, C. W., Acheson, A., Hill-Kapturczak, N., and
324–334. doi: 10.1016/j.cell.2020.08.047 Dougherty, D. M. (2009). L-tryptophan: basic metabolic functions, behavioral research
Kumperscak, H. G., Gricar, A., Ülen, I., and Micetic-Turk, D. (2020). A pilot and therapeutic indications. Int. J. Tryptophan Res. 2, IJTR–S2129. doi: 10.4137/ijtr.s2129
randomized control trial with the probiotic strain lactobacillus rhamnosus GG (LGG) Rommelse, N. N., Franke, B., Geurts, H. M., Hartman, C. A., and Buitelaar, J. K. (2010).
in ADHD: children and adolescents report better health-related quality of life. Front. Shared heritability of attention-deficit/hyperactivity disorder and autism spectrum
Psychiatry 11. doi: 10.3389/fpsyt.2020.00181 disorder. Eur. Child Adolesc. Psychiatry 19, 281–295. doi: 10.1007/s00787-010-0092-x
Kwak, M. J., Tan, P. L., Oh, J. K., Chae, K. S., Kim, J., Kim, S. H., et al (2021). The Ronan, V., Yeasin, R., and Claud, E. C. (2021). Childhood development and the
effects of multispecies probiotic formulations on growth performance, hepatic microbiome–the intestinal microbiota in maintenance of health and development of
metabolism, intestinal integrity and fecal microbiota in growing-finishing pigs. disease during childhood development. Gastroenterology. 160 (2), 495–506.
Animal Feed Science and Technology 274, 114833. doi: 10.1053/j.gastro.2020.08.065
LeBlanc, J. G., Chain, F., Martı́n, R., Bermú dez-Humará n, L. G., Courau, S., and Sanctuary, M. R., Kain, J. N., Chen, S. Y., Kalanetra, K., Lemay, D. G., Rose, D. R.,
Langella, P. (2017). Beneficial effects on host energy metabolism of short-chain fatty et al. (2019). Pilot study of probiotic/colostrum supplementation on gut function in
acids and vitamins produced by commensal and probiotic bacteria. Microb. Cell children with autism and gastrointestinal symptoms. PloS One 14 (1), e0210064.
Factories 16 (1), 1–10. doi: 10.1186/s12934-017-0691-z doi: 10.1371/journal.pone.0210064
Li, N., Chen, H., Cheng, Y., Xu, F., Ruan, G., Ying, S., et al. (2021). Fecal microbiota Santocchi, E., Guiducci, L., Prosperi, M., Calderoni, S., Gaggini, M., Apicella, F., et al.
transplantation relieves gastrointestinal and autism symptoms by improving the gut (2020). Effects of probiotic supplementation on gastrointestinal, sensory and core
microbiota in an open-label study. Front. Cell. Infect. Microbiol. 11. doi: 10.3389/ symptoms in autism spectrum disorders: a randomized controlled trial. Front.
fcimb.2021.759435 Psychiatry 11. doi: 10.3389/fpsyt.2020.550593
Liu, Y. W., Liong, M. T., Chung, Y. C. E., Huang, H. Y., Peng, W. S., Cheng, Y. F., Sarkar, A., Lehto, S. M., Harty, S., Dinan, T. G., Cryan, J. F., and Burnet, P. W.
et al. (2019). Effects of lactobacillus plantarum PS128 on children with autism spectrum (2016). Psychobiotics and the manipulation of bacteria–gut–brain signals. Trends
disorder in Taiwan: a randomized, double-blind, placebo-controlled trial. Nutrients. 11 Neurosci. 39 (11), 763–781. doi: 10.1016/j.tins.2016.09.002
(4), 820. doi: 10.3390/nu11040820
Satti, S., Palepu, M. S. K., Singh, A. A., Jaiswal, Y., Dash, S. P., Gajula, S. N. R., et al.
Liu, S., Rao, S., Xu, Y., Li, J., Huang, H., Zhang, X., et al. (2020). Identifying common (2023). Anxiolytic-and antidepressant-like effects of bacillus coagulans unique IS-2
genome-wide risk genes for major psychiatric traits. Hum. Genet. 139, 185–198. mediate via reshaping of microbiome gut-brain axis in rats. Neurochem. Int. 163,
doi: 10.1007/s00439-019-02096-4 105483. doi: 10.1016/j.neuint.2023.105483
Marchesi, J. R., Holmes, E., Khan, F., Kochhar, S., Scanlan, P., Shanahan, F., et al. Schwarcz, R., and Stone, T. W. (2017). The kynurenine pathway and the brain:
(2007). Rapid and noninvasive metabonomic characterization of inflammatory bowel challenges, controversies and promises. Neuropharmacology 112, 237–247.
disease. J. Proteome Res. 6 (2), 546–551. doi: 10.1021/pr060470d doi: 10.1016/j.neuropharm.2016.08.003
Mensi, M. M., Rogantini, C., Marchesi, M., Borgatti, R., and Chiappedi, M. (2021). Selma-Royo, M., Calatayud Arroyo, M., Garcı́a-Mantrana, I., Parra-Llorca, A.,
Lactobacillus plantarum PS128 and other probiotics in children and adolescents with Escuriet, R., Martı́nez-Costa, C., et al. (2020). Perinatal environment shapes
autism spectrum disorder: a real-world experience. Nutrients. 13 (6), 2036. microbiota colonization and infant growth: impact on host response and intestinal
doi: 10.3390/nu13062036 function. Microbiome 8 (1), 1–19. doi: 10.1186/s40168-020-00940-8
Senn, V., Bassler, D., Choudhury, R., Scholkmann, F., Righini-Grunder, F., Vuille- Tick, B., Bolton, P., Happé , F., Rutter, M., and Rijsdijk, F. (2016). Heritability of
dit-Bille, R. N., et al. (2020). Microbial colonization from the fetus to early childhood–a autism spectrum disorders: a meta-analysis of twin studies. J. Child Psychol. Psychiatry
comprehensive review. Front. Cell. Infect. Microbiol. 10. doi: 10.3389/ 57 (5), 585–595. doi: 10.1111/jcpp.12499
fcimb.2020.573735 Tomova, A., Husarova, V., Lakatosova, S., Bakos, J., Vlkova, B., Babinska, K., et al.
Sepehrmanesh, Z., Shahzeidi, A., Mansournia, M. A., Ghaderi, A., and Ahmadvand, (2015). Gastrointestinal microbiota in children with autism in Slovakia. Physiol. Behav.
A. (2021). Clinical and metabolic reaction to probiotic supplement in children suffering 138, 179–187. doi: 10.1016/j.physbeh.2014.10.033
attention-deficit hyperactivity disorder: a randomized, double-blind, placebo- Vuong, H. E., and Hsiao, E. Y. (2017). Emerging roles for the gut microbiome in
controlled experiment. Int. Arch. Health Sci. 8 (2), 90. doi: 10.4103/iahs.iahs_112_20 autism spectrum disorder. Biol. Psychiatry 81 (5), 411–423. doi: 10.1016/
Sgritta, M., Dooling, S. W., Buffington, S. A., Momin, E. N., Francis, M. B., Britton, R. j.biopsych.2016.08.024
A., et al. (2019). Mechanisms underlying microbial-mediated changes in social Wang, H., Gao, K., Wen, K., Allen, I. C., Li, G., Zhang, W., et al. (2016). Lactobacillus
behavior in mouse models of autism spectrum disorder. Neuron 101 (2), 246–259. rhamnosus GG modulates innate signaling pathway and cytokine responses to
doi: 10.1016/j.neuron.2018.11.018 rotavirus vaccine in intestinal mononuclear cells of gnotobiotic pigs transplanted
Shaaban, S. Y., El Gendy, Y. G., Mehanna, N. S., El-Senousy, W. M., El-Feki, H. S., Saad, K., with human gut microbiota. BMC Microbiol. 16 (1), 1–14. doi: 10.1186/s12866-016-
et al. (2018). The role of probiotics in children with autism spectrum disorder: a prospective, 0727-2
open-label study. Nutr. Neurosci. 21 (9), 676–681. doi: 10.1080/1028415X.2017.1347746 Wang, L. J., Yang, C. Y., Kuo, H. C., Chou, W. J., Tsai, C. S., and Lee, S. Y. (2022).
Sharma, R., Gupta, D., Mehrotra, R., and Mago, P. (2021). Psychobiotics: the next-generation Effect of bifidobacterium bifidum on clinical characteristics and gut microbiota in
probiotics for the brain. Curr. Microbiol. 78, 449–463. doi: 10.1007/s00284-020-02289-5 attention-Deficit/Hyperactivity disorder. J. Pers. Med. 12 (2), 227. doi: 10.3390/
Shaw, W., Kassen, E., and Chaves, E. (1995). Increased urinary excretion of analogs jpm12020227
of Krebs cycle metabolites and arabinose in two brothers with autistic features. Clin. Yunes, R. A., Poluektova, E. U., Dyachkova, M. S., Klimina, K. M., Kovtun, A. S.,
Chem. 41 (8), 1094–1104. doi: 10.1093/clinchem/41.8.1094 Averina, O. V., et al. (2016). GABA production and structure of gadB/gadC genes in
Silva, Y. P., Bernardi, A., and Frozza, R. L. (2020). The role of short-chain fatty acids lactobacillus and bifidobacterium strains from human microbiota. Anaerobe 42, 197–
from gut microbiota in gut-brain communication. Front. Endocrinol. 11. doi: 10.3389/ 204. doi: 10.1016/j.anaerobe.2016.10.011
fendo.2020.00025 Zheng, D., Liwinski, T., and Elinav, E. (2020). Interaction between microbiota and
Skott, E., Yang, L. L., Stiernborg, M., Söderström, Å., Rüegg, J., Schalling, M., et al. immunity in health and disease. Cell Res. 30 (6), 492–506. doi: 10.1038/s41422-020-
(2020). Effects of a synbiotic on symptoms, and daily functioning in attention deficit 0332-7
hyperactivity disorder–a double-blind randomized controlled trial. Brain Behav. Zheng, Y., Verhoeff, T. A., Perez Pardo, P., Garssen, J., and Kraneveld, A. D.
Immun. 89, 9–19. doi: 10.1016/j.bbi.2020.05.056 (2020). The gut-brain axis in autism spectrum disorder: a focus on the
Stiemsma, L. T., and Michels, K. B. (2018). The role of the microbiome in the developmental metalloproteases ADAM10 and ADAM17. Int. J. Mol. Sci. 22 (1), 118.
origins of health and disease. Pediatrics 141 (4), e20172437. doi: 10.1542/peds.2017-2437 doi: 10.3390/ijms22010118
Sukmajaya, A. C., Lusida, M. I., and Setiawati, Y. (2021). Systematic review of gut Zhou, A., Cao, X., Mahaganapathy, V., Azaro, M., Gwin, C., Wilson, S., et al. (2023).
microbiota and attention-deficit hyperactivity disorder (ADHD). Ann. Gen. Psychiatry Common genetic risk factors in ASD and ADHD co-occurring families. Hum. Genet.
20 (1), 1–12. doi: 10.1186/s12991-021-00330-w 142 (2), 217–230. doi: 10.1007/s00439-022-02496-z
Tabouy, L., Getselter, D., Ziv, O., Karpuj, M., Tabouy, T., Lukic, I., et al. (2018). Zhuang, L., Chen, H., Zhang, S., Zhuang, J., Li, Q., and Feng, Z. (2019). Intestinal
Dysbiosis of microbiome and probiotic treatment in a genetic model of autism microbiota in early life and its implications on childhood health. Genomics Proteomics
spectrum disorders. Brain Behav. Immun. 73, 310–319. doi: 10.1016/j.bbi.2018.05.015 Bioinf. 17 (1), 13–25. doi: 10.1016/j.gpb.2018.10.002