You are on page 1of 11

Journal of Microbiology (2018) Vol. 56, No. 3, pp.

172–182 eISSN 1976-3794


DOI 10.1007/s12275-018-8032-4 pISSN 1225-8873

REVIEW

Mind-altering with the gut: Modulation of the gut-brain axis with


probiotics

al., 2011). The scaffolding of the gut-brain axis includes the


Namhee Kim, Misun Yun, Young Joon Oh, gastrointestinal tract, central nervous system (CNS), auto-
and Hak-Jong Choi* nomic nervous system (ANS), enteric nervous system (ENS),
neuroendocrine system, and immune system (Grenham et al.,
Microbiology and Functionality Research Group, World Institute of 2011). Recent studies have shown that the gut microbiota is
Kimchi, Gwangju 61755, Republic of Korea
involved in the neurodevelopment and diverse brain func-
tions through regulating the gut-brain axis (Carabotti et al.,
(Received Jan 21, 2018 / Revised Feb 7, 2018 / Accepted Feb 12, 2018)
2015; Erny et al., 2015). Gastrointestinal symptoms, such as
constipation, diarrhea, and abdominal pain, are common co-
It is increasingly evident that bidirectional interactions exist
morbidities in many neurological diseases (Westfall et al.,
among the gastrointestinal tract, the enteric nervous system,
2017). Moreover, recent advances in metagenomic sequenc-
and the central nervous system. Recent preclinical and clinical
ing have revealed that dysregulation in the composition of
trials have shown that gut microbiota plays an important role
gut microbiota (gut dysbiosis) is present in a variety of neu-
in these gut-brain interactions. Furthermore, alterations in gut
rological diseases. Consequently, the importance of main-
microbiota composition may be associated with pathogenesis
taining a healthy microbiota community (gut symbiosis) in
of various neurological disorders, including stress, autism, de-
the regulation of the gut-brain axis cannot be overly empha-
pression, Parkinson’s disease, and Alzheimer’s disease. There-
sized. The term microbiota-gut-brain (MGB) axis was intro-
fore, the concepts of the microbiota-gut-brain axis is emer-
duced to highlight the role of the microbiota in the gut-brain
ging. Here, we review the role of gut microbiota in bidirec-
axis.
tional interactions between the gut and the brain, including
Probiotics are defined as living microorganisms that, when
neural, immune-mediated, and metabolic mechanisms. We
ingested in adequate quantities, confer a health benefit on
highlight recent advances in the understanding of probiotic
the host; these microorganisms have been reported to exert a
modulation of neurological and neuropsychiatric disorders
wide range of effects (Hill et al., 2014). Although their mech-
via the gut-brain axis.
anisms in modulating host physiology are not yet fully elu-
cidated, probiotics might be able to modulate host immune
Keywords: probiotics, gut microbiota, nervous system, gut-
system (Bermudez-Brito et al., 2012). For example, Weissella
brain axis, gut dysbiosis, neurological disorders
cibaria WIKIM28 isolated from kimchi ameliorates atopic
dermatitis symptoms by regulating dendritic cell functions
and inducing regulatory T cell responses (Lim et al., 2017).
Introduction
Probiotic bacteria not only modulate host immune responses,
but also create a healthy gut environment through balanc-
The human gastrointestinal tract is inhabited by nearly 100
ing of the intestinal microflora. Ingestion of probiotics may
trillion microorganisms that are believed to play important
restore the composition of the gut microflora to a state more
roles in physiology (Gill et al., 2006). These microorganisms,
favorable for beneficial microorganisms (Choi et al., 2015;
collectively known as gut microbiota, are proposed as an es-
Mountzouris et al., 2007). Probiotics recently have attracted
sential “organ” in the maintenance of immune function, car-
attention in the context of brain function and health because
bohydrate metabolism, and metabolic homeostasis (Tremaroli
they serve to alter gut microflora toward a beneficial state,
and Backhed, 2012).
which could affect the gut-brain axis (Bravo et al., 2012).
Bidirectional communication between the brain and the gut,
In this review, we will focus on the role of gut microbiota in
known as the gut-brain axis, has long been recognized: the
crosstalk between the gut and brain, as well as how the mi-
brain modulates the gastrointestinal tract by regulating of mo-
crobiota affects a variety of neurological disorders. Finally,
tility, secretion, absorption, and blood flow; concurrently,
we will discuss recent findings in probiotic modulation of
the gut can affect brain function and behavior (Grenham et
brain function and neurological diseases.

*For correspondence. E-mail: hjchoi@wikim.re.kr; Tel.: +82-62-610-1729;


Fax: +82-62-610-1850
Copyright G2018, The Microbiological Society of Korea
Probiotics modulate the gut-brain axis 173

Interactions within the gut-brain axis constitutes a key indirect route for communication between
the gut microbiota and the nervous system (Macpherson and
Gut and brain regulate each other bidirectionally via multiple Uhr, 2004; Bengmark, 2013). The gut houses the gut-asso-
mechanisms and pathways, including neural, neuroendocrine, ciated lymphoid tissues (GALT) which are the largest collec-
and immunological signals. Here, we highlight key commu- tion of lymphoid tissues in the human body; theses tissues
nication pathways for a comprehensive understanding of the protect the body from microbial invasion via the gut. A va-
gut-brain axis. riety of gut and GALT immune cells, such as T cells, macro-
phages, and dendritic cells (DCs) can cross the blood-brain-
Neural communication between the gut and brain barrier (BBB) and affect neurons and glia in the brain (Dia-
Neural communication is mainly conducted within the ENS mond et al., 2011). In the brain, there are also resident im-
in the gastrointestinal tract. The ENS is a main division of the mune cells, such as macrophages and DCs in the choroid
ANS and regulates gastrointestinal tract functions such as plexus, microglia in the parenchymal region of brain, and
intestinal motility, mucous secretion, and blood flow (Brown- leukocytes in the cerebrospinal fluid (CSF) (Wang and Kas-
ing et al., 2017). The ENS interacts with the ANS and CNS via per, 2014). Therefore, gut microbiota, known to shape the
neurotransmitters (adrenaline, noradrenaline, and acetyl- host immune system, can also modulate the activity of these
choline), as well as sensory and motor neurons, all of which resident immune cells, in addition to their effects on neuro-
convey signals from the gut to the brain. nal cells in the CNS (Berer and Krishnamoorthy, 2012). More-
The intestinal microbiota regulates electrophysiological thre- over, a recent study of germ-free mice has revealed that gut
sholds in ENS neurons (Sarkar et al., 2016). For example, a microbiota can alter the immune system of the CNS by regu-
strain of Lactobacillus reuteri was shown to activate a calcium- lating microglial activation and homeostasis (Erny et al., 2015).
dependent potassium channel in neurons within rat colon Microglia in germ-free mice exhibited global defects in cel-
lular proportions and in maturation, thereby leading to di-
myenteric plexus (Kunze et al., 2009). Metabolic compounds
minished innate immune responses (Erny et al., 2015).
by Bifidobacterium longum NCC3001 elicited reduction of
Systemic circulation of immune factors, cytokines, and che-
action potential spikes in myenteric neurons, following elec-
mokines influences the brain via the vagus nerve and circum-
trical stimulation (Bercik et al., 2011b). In germ-free mice,
ventricular organs (Hosoi et al., 2002). In addition, another
myenteric sensory neurons exhibited decreased excitability,
pathway consists of direct transport by saturable transport
compared with normal mice (McVey Neufeld et al., 2013).
systems along the BBB (Banks, 2005). In the brain, pro-in-
Moreover, these neurons in conventionalized germ-free mice
flammatory cytokines can trigger further neuro-inflamma-
(conventionalized with intestinal bacteria from convention-
tion in the nervous system, thereby causing increased per-
ally-raised mice) revealed increased excitability following hy-
meability of the BBB (McCusker and Kelley, 2013). Leakage
perpolarization (McVey Neufeld et al., 2013). In addition to
of the BBB leads to immune cell infiltration, exacerbation of
their actions on myenteric neurons, gut bacteria also play a
inflammatory responses, and reactive gliosis (reactive changes
crucial role in initial colonization and homeostasis of glial
of glial cells in response to damage); causing neurodegenera-
cells in the intestinal mucosa (Kabouridis et al., 2015).
tion eventually (Obermeier et al., 2013). In addition, cyto-
Another neuronal pathway in gut-brain communication uti-
kines alter the concentrations of several neurotransmitters
lizes the vagus nerve (cranial nerve X that runs from brain-
in the brain, including serotonin, dopamine, and glutamate
stem to gastrointestinal tract) which has both efferent and
(Miller et al., 2013). Non-inflammatory cytokines also serve
afferent roles (Cryan and Dinan, 2012). Approximately 80%
as mediators for intestinal microbes to regulate brain func-
of vagal fibers are sensory, conveying information regarding
tion. For example, antibiotic exposure in neonatal mice pro-
the state of the body’s organs to the CNS (Thayer and Stern-
duced reductions in the level of plasma granulocyte colony
berg, 2009). The vagus nerve regulates several vital functions,
stimulating factor (G-CSF) (Deshmukh et al., 2014). G-CSF
including bronchial constriction, heart rate, and gut motility.
can stimulate neurogenesis in the brain by crossing the BBB;
Many known effects of gut microbiota or probiotics strains
further, it has a protective role in ischemic injury, as well as
are dependent on vagus nerve activity (Goehler et al., 2008;
in certain models of Parkinson’s disease and Alzheimer’s di-
Bercik et al., 2011b; Bravo et al., 2011). For example, chronic
sease (Shyu et al., 2004; Meuer et al., 2006; Prakash et al.,
treatment with L. rhamnosus JB-1 induced increases in γ-
2013; Wallner et al., 2015). Thus, by promoting the produc-
Aminobutyric acid (GABA) mRNA expression within spe-
tion of G-CSF, the gut microbiota may provide a potential
cific brain regions, in addition to reducing stress-induced
therapeutic agent for normal neurodevelopment, as well as
corticosterone and anxiety-and depression-related behavior
to combat the progression of neurodegenerative diseases.
(Bravo et al., 2011). Moreover, based on experiments in vago-
Additionally, recent findings regarding drainage of the lym-
tomized mice, the vagus nerve was identified as a major mo-
phatic system into the brain provide more decisive evidence
dulator in the gut-brain communication pathway (Bravo et
of direct cytokine entry into the brain, enabling interaction
al., 2011). However, vagus nerve-independent mechanisms
with neural tissues (Louveau et al., 2015; Sun et al., 2017).
also exist in gut–brain communication, as antimicrobial treat-
ments failed to show gut-brain dependence on vagus nerve
Effects of neuroactive compounds and metabolites from gut mi-
activity (Bercik et al., 2011a).
crobes on the central nervous system
Immune system mediated interaction Gut microbiota is capable of producing a spectrum of neu-
rotransmitters, neuroactive compounds, and metabolites.
Gut microbiota can directly affect the immune system; this
Neurotransmitters and neuroactive compounds that have
174 Kim et al.

been isolated from bacteria include GABA, serotonin, cate- cells expressing free fatty acid receptors (Psichas et al., 2015).
cholamine, and acetylcholine (Table 1). For example, Lacto- Secreted gut peptides can act in the hypothalamus, a pri-
bacillus and Bifidobacterium species can produce GABA; mary brain center for regulation of food intake and energy
Escherichia, Bacillus, and Saccharomyces spp. can produce homeostasis (Goldstone, 2006).
norepinephrine; Escherichia, Bacillus, Lactococcus, Lacto- Recent findings have revealed that commensal bacterial
bacillus, and Serratia can produce dopamine. Gut microbiota metabolites called N-acyl amides are enable to interact with
also indirectly controls the production of neurotransmitters G-protein-coupled receptor 119 (GPR119) which is highly
by regulating available precursors of neuroactive chemicals expressed in the small intestinal enteroendocrine cells that
or by stimulating host enteroendocrine and neuroendocrine secrete gut hormone peptides. In vivo mouse and in vitro
cells (Desbonnet et al., 2010; Yano et al., 2015). B. infantis cell-based analyses demonstrated that these lipid compounds
has been reported to increase the availability of tryptophan, can serve as endogenous ligand to regulate metabolic hor-
a precursor of 5-hydroxytryptamine (5-HT; a synonym of mones and glucose homeostasis (Cohen et al., 2017). Besides
serotonin), in the plasma, thereby raising 5-HT concentra- these ligands, commensal and pathogenic bacteria produce
tion in the brain (Desbonnet et al., 2008; Desbonnet et al., neurotoxins, formyl peptides, pathogen-associated molec-
2010). Spore-forming gut microbiota stimulates host enter- ular patterns (PAMPs), and lipopolysaccharides (LPS), all
ochromaffin (EC) cells to produce 5-HT; the majority of 5- of which are closely tied to the function of the enteric and
HT in the body is directly synthesized by EC cells from tryp- central nervous system, as well as the pathogenesis of neu-
tophan (Yano et al., 2015). Thus, probiotics producing neu- rological disorders (Yang and Chiu, 2017).
rotransmitters or promoting host cells to synthesize these
neurochemicals may be used as delivery vehicles for neuro- CNS influences gastrointestinal microbiota composition
active compounds (Lyte, 2011).
The CNS can alter gut microbiota composition and total bio-
Other well-known bacterial neuroactive metabolites include mass through regulation of satiety. Changes in dietary pat-
short-chain fatty acids (SCFAs), such as acetate, butyrate, lac- terns (e.g., through central nervous system regulation) may
tate, and propionate (Horn and Klein, 2013; Overduin et al.,
affect the ability of intestinal microbiota to access required
2013). SCFAs are produced via the fermentation of complex nutrients, ultimately influencing their composition (Wang
polysaccharides and demonstrated to possess immunomo- and Kasper, 2014). In addition, the nervous system plays
dulatory effects (Macfarlane and Macfarlane, 2003). SCFAs
an important role in the regulation of gut functions, such as
can directly interact with the nervous system, thereby activat- gastrointestinal mobility, as well as secretion of acid, bicar-
ing sympathetic neurons, and can also pass through the BBB, bonate, and mucus, all of which are prominent in the main-
thereby influencing behavior and neural signaling (Kimura
tenance of the mucous layer and biofilm (Rhee et al., 2009).
et al., 2013; Frost et al., 2014; Rios-Covian et al., 2016). For Thus, by altering the normal mucosal habitat, the central ner-
instance, butyrate can act as a potential epigenetic modifier vous system can affect gut microbiota composition (Rhee
by inhibiting histone deacetylases (HDACs) and stimulate
et al., 2009).
the hypothalamus-pituitary-adrenal (HPA) axis (Gagliano This direct influence is mediated by neurotransmitters, which
et al., 2014; Stilling et al., 2014). By inhibiting of HDACs, are produced by host neurons, and neurotransmitter recep-
butyrate can act as an anti-inflammatory agent by inhibit-
tors, which are expressed on gut microbiota. Binding of host-
ing nuclear factor κB (NF-κB) activation in colon cells (Inan generated neurotransmitters with bacteria affects the func-
et al., 2000). Further, butyrate inhibits intestinal inflamma- tion of microbial components, thereby promoting increased
tion by relieving HDAC inhibition of Foxp3, a transcription
susceptibility to inflammatory and infectious stimuli (Ca-
factor exclusively expressing in regulatory T cells (Tregs), thus rabotti et al., 2015). For instance, Pseudomonas fluorescens
promoting the generation of Tregs (Furusawa et al., 2013). internalizes GABA with neurotransmitter-binding charac-
Moreover, glial cells, especially in the developing brain, up-
teristics (Guthrie and Nicholson-Guthrie, 1989); for another
take SCFAs as an energy source for cellular metabolism (Ra- example, Escherichia coli O157:H7 contains host-derived ad-
fiki et al., 2003). SCFAs also stimulate the secretion of gut renergic receptors that activate virulence genes in response
hormone peptides, such as glucagon-like peptide-1 (GLP-1)
to inter-kingdom cross-signaling (quorum sensing) (Clarke
and peptide tyrosine tyrosine (PYY), from enteroendocrine et al., 2006).

Table 1. List of neuroactive compounds detected within various bacteria


Gut microbiota Neurochemical References
Lactobacillus, Bifidobacterium spp. GABA Barrett et al. (2012)
Bifidobacterium infantis, Streptococcus, Escherichia, Enterococcus,
Serotonin (5-HT) Özogul (2011), Holzer and Farzi (2014)
Lactococcus, Lactobacillus, Candida,
Clostridium sporogenes, Ruminococcus gnavus Tryptamine Williams et al. (2014)
Escherichia, Bacillus, Saccharomyces Norepinephrine Holzer and Farzi (2014)
Escherichia, Bacillus, Lactococcus, Lactobacillus, Serratia Dopamine Özogul (2011), Holzer and Farzi (2014)
Lactobacillus, Bacillus Acetylcholine Kawashima et al. (2007)
Landete et al. (2008), Thomas et al. (2012),
Lactococcus, Lactobacillus, Streptococcus, Enterococcus Histamine
Hemarajata et al. (2013)
Bacillus sp. JPJ L-dopa Surwase and Jadhav (2011)
Probiotics modulate the gut-brain axis 175

The CNS also directly or indirectly regulates release of other social and cognitive functions. Gastrointestinal disturbances,
signaling molecules, cytokines, and antimicrobial peptides including constipation, diarrhea, increased intestinal perme-
into the gut lumen by neurons, enteroendocrine cells, im- ability, and abdominal pain, are frequently reported in ASD
mune cells, and Paneth cells which are in charge of α-de- children (Coury et al., 2012). This evidence suggests that gas-
fensin secretion. These secreted products affect microbial trointestinal disturbances, perhaps linked to gut dysbiosis,
survival and their surrounding environment (Carabotti et may be associated with ASD (Cryan and Dinan, 2012). Con-
al., 2015). sistent with this hypothesis, both an animal model of ASD
The nervous system also regulates microbiota composition and clinical studies with ASD patients showed reductions in
by altering epithelial permeability, thereby allowing the pen- Bacteroides and increased levels of Clostridium species (Fine-
etration of bacteria and facilitating host-microbiome inter- gold et al., 2010; Hsiao et al., 2013). A similar imbalance in
action in the mucosa. Under stress, the HPA axis releases cor- Bacteroidetes and Firmicutes has also been reported in ASD
tisol, a stress hormone, thereby regulating gut movement and patients (Finegold et al., 2010; Strati et al., 2017). Moreover,
immune system responses (via cells, cytokines, and secretory altered levels of other gut microbiota have been found in stool
immunoglobulin A). Stress increases intestinal permeability samples from children with ASD; alteration affected the rel-
via corticotropin-releasing hormone (CRH)-mediated mast ative abundances of Bifidobacterium, Lactobacillus, Sutterella,
cell activation. Elevated intestinal permeability facilitates mi- Prevotella, and Ruminococcus genera (Adams et al., 2011; Kang
crobial access to immune cells and neuronal cells of the ENS; et al., 2013; Wang and Kasper, 2014). A recent study of an
consequently, this stress-based permeability alters the in- autistic cohort has revealed that the constipation in autistic
testinal microbial profile. Restraint and social stress in rodents patients is associated with increase in Escherichia/Shigella and
as well as maternal separation in non-human primates, lead Clostridium cluster XVIII (Strati et al., 2017).
to disrupted gut microbial composition (Bailey and Coe, 1999; According to several studies, propionic acid and related
Bailey et al., 2004, 2010; O’Mahony et al., 2009; Tillisch, SCFAs are generated by ASD-associated bacteria (Bactero-
2014). idetes, Clostridia, and Desulfovibrio), and these SCFAs can
induce ASD-linked neurochemical changes and behavior de-
ficits in rat, including abnormal motor movements, repeti-
The gut dysbiosis in CNS disorders tive interests, cognitive deficits, and impaired social interac-
tions (Macfabe, 2012).
Given that the gut microbiota can affect the function of the Depression is a stress-related mood disorder that is associ-
brain via many complex pathways and components, it is not ated with alterations in the HPA axis and immune responses.
surprising that many neurological diseases are associated In a maternal separation rodent model of depression, a di-
with dysbiosis in the gut (Table 2). Although autoimmune- versity of gut bacteria were decreased, along with hyperacti-
mediated CNS disorders, such as multiple sclerosis (MS), vation of the HPA axis and increased expression of pro-in-
experimental autoimmune encephalomyelitis (an animal mo- flammatory cytokines (O’Mahony et al., 2009; Vetulani, 2013).
del of MS), and neuromyelitis optica, are also closely related Infant monkeys undergoing maternal separation showed
with gut microbiota, we specifically focus on gut dysbiosis decreased fecal Lactobacillus (Bailey and Coe, 1999). Unlike
within neuro-psychiatric disorders and neurodegenerative animal models, there has been little evidence of intestinal
diseases. microbial changes in depressed patients. However, in a re-
Autism spectrum disorder (ASD) is a class of neurodevel- cent study, fecal pyrosequencing comparing patients with
opmental disorders that are characterized by deficits in both depression against healthy controls revealed increased fecal

Table 2. Links between altered gut microbiota composition and a variety of neurological and psychiatric disorders
Disease Altered gut microbiota References
Porphyromonadaceae  Bailey et al. (2010)
Stress Clostridium , Bacteroides  Bailey et al. (2011)
Oscillibacter , Anaerotruncus , Peptococcus, Lactobacillus  Golubeva et al. (2015)
Bifidobacterium , Lactobacillus  Aizawa et al. (2016)
Depression
Bacteroidetes , Proteobacteria , Actinobacteria , Firmicutes  Jiang et al. (2015)
Clostridium  Song et al. (2004), Parracho et al. (2005)
Sutterella spp. , Ruminococcus torques , Akkermansia muciniphila  Wang et al. (2011, 2013)
Clostridium , Sutterellaceae , Enterobacteriaceae , Bifidobacterium  De Angelis et al. (2013)
Autism
Collinsella , Corynebacterium , Dorea , Lactobacillus , Alistipes , Bilophila ,
Strati et al. (2017)
Dialister , Parabacteroides , Veillonella 
Desulfovibrio , Bacteroides vulgatus , Ruminococcus , Bifidobacterium  Finegold et al. (2010)
Association with bacterial and viral infection Bu et al. (2015)
Bacteroidetes , Tenericutes , Firmicutes , Verrucomicrobia , Proteobacteria G
Alzheimer’s disease Harach et al. (2017)
Actinobacteria , Allobaculum , Akkermansia G
Bacteroidetes , Firmicutes , Bifidobacterium  Vogt et al. (2017)
Ralstonia , Blautia , Coprococcus , Roseburia , Faecalibacterium  Keshavarzian et al. (2015)
Parkinson’s disease
Enterobacteriaceae , Prevotellaceae G Scheperjans et al. (2015)
176 Kim et al.

bacterial alpha diversity in depressed patients. In particular, compared with healthy brain. During 16S rRNA gene sequen-
relative abundances of the phyla Bacteroidetes, Proteobacteria, cing, read counts of Actinobacteria, primarily those of the
and Actinobacteria were increased, with concurrent signi- Propionibacteriaceae family, were significantly increased in
ficant reduction in the level of Firmicutes (Jiang et al., 2015). AD samples, compared with controls. These findings sup-
Alzheimer’s disease (AD) is a prevalent neurodegenerative port the idea that bacterial migration into the CNS could con-
disease and the most common form of dementia. According tribute to the onset of neurological disease, or facilitate the
to the amyloid hypothesis, the aggregation and cerebral de- pathogenesis of disease (Emery et al., 2017).
position of amyloid-β (Aβ) peptides into plaques is an ini- Parkinson’s disease (PD) is recognized as the second most
tial influence that drives AD pathogenesis (Hardy and Selkoe, common neurodegenerative disorder and mainly affects the
2002; Harach et al., 2017). A recent study using the Aβ pre- central motor system. Degeneration of dopamine neurons in
cursor protein (APP) transgenic mouse model has suggested the substantia nigra region results in decreased dopamine
that alterations in gut microbiota may contribute to amyloid levels in the brain, thereby causing abnormal brain activity.
deposition (Harach et al., 2017). APP transgenic mice exhibit Most PD patients experience constipation even before the
reduction in Firmicutes, Verrucomicrobia, Proteobacteria, onset of movement defects (Boursi et al., 2016). In clinical
and Actinobacteria, with concurrent increases in Bacteroi- studies, fecal pyrosequencing results showed the reduced
detes. A study using germ-free APP transgenic mice demon- relative abundance of Prevotellaceae and increased that of
strates reductions in cerebral Aβ amyloid pathology, com- Enterobacteriaceae in PD patients, compared with controls
pared with control transgenic mice. Moreover, colonization (Scheperjans et al., 2015). Furthermore, Enterobacteriaceae
with microbiota from conventional APP transgenic mice in- was positively associated with the severity of motor symp-
creased cerebral Aβ pathology in germ-free transgenic mice, toms, including postural instability and gait difficulty (For-
whereas control colonization using wild-type microbiota was syth et al., 2011; Cassani et al., 2015; Scheperjans et al., 2015).
less effective in increasing cerebral Aβ levels (Harach et al., Another clinical study, using fecal and mucosal samples
2017). Furthermore, clinical studies of the gut microbiota of from PD patients, showed that butyrate-producing bacteria,
AD patients reveal decreased microbial diversity and changes including the Blautia, Coprococcus, and Roseburia genera,
in composition compared with controls; these changes in- were significantly reduced in PD patients (Keshavarzian et
clude decreased levels of Firmicutes and Bifidobacterium and al., 2015). As mentioned above, butyrate exhibits anti-inflam-
increased levels of Bacteroidetes (Vogt et al., 2017). matory effect (Furusawa et al., 2013). The presence of the
A remarkable recent study has revealed that post-mortem putative pro-inflammatory bacteria Ralstonia in PD patients
brains from AD patients exhibit increased bacterial popul- suggested that pro-inflammatory dysbiosis in gut microbiota
ations, as well as different proportions of specific bacteria, may be part of the constellation of symptoms in PD patients.

Table 3. Effects of probiotics on brain function and behavior


Probiotic strain Model Duration Effects References
Rodent (maternal separation Normalized the immune response, Desbonnet et al.
Bifidobacterium infantis 45 days
model of depression) reduced behavioral deficits (2008, 2010)
Rodent (maternal immune Reversed ASD-related behavior,
Bacteroides fragilis 6 days Hsiao et al. (2013)
activation) improved gut barrier integrity
A mixture of Lactobacillus helveticus R0052
Rodent (healthy) 2 weeks Reduced anxiety-like behavior Messaoudi et al. (2011a)
and Bifidobacterium longum R0175
Bifidobacterium longum 1714, Bifidobacterium Rodent (innately anxious
6 weeks Reduced anxiety-like behavior Savignac et al. (2014)
breve 1205 BALB/c)
A mixture of Bifidobacterium animalis subsp. lactis,
Influenced brain activity in emotional
Streptococcus thermophilus, Lactobacillus bulgaricus, Human (healthy) 4 weeks Tillisch et al. (2013)
centers
and Lactococcus lactis subsp. lactis
Lactobacillus helveticus R0052 and Bifidobacterium Messaoudi et al.
Human (healthy) 30 days Less psychological distress
longum R0175 (2011a, 2011b)
Human (healthy, exposed Reduced stress–related gastrointestinal
Lactobacillus casei Shirota 11 weeks Kato-Kataoka et al. (2016)
to academic stress) symptoms
Lactobacillus casei Shirota Human (healthy) 3 weeks Improved mood Benton et al. (2007)
A mixture of Bifidobacterium bifidum W23, Bifido-
bacterium lactis W52, Lactobacillus acidophilus W37,
Reduced rumination and aggressive
Lactobacillus brevis W63, Lactobacillus casei W56, Human (healthy) 4 weeks Steenbergen et al. (2015)
cognition
Lactobacillus salivarius W24, and Lactococcus lactis
(W19 and W58)
Reduced D-arabinitol level in urine,
Kaluzna-Czaplinska
Lactobacillus acidophilus Human (autistic children) 2 months improvement in ability of concen-
and Blaszczyk (2012)
tration and carrying out orders
VSL#3 (VSL Pharmaceuticals Inc.), a multi-strain Human (ASD child, case Improved autistic core symptoms,
4 weeks Grossi et al. (2016)
mixture of ten probiotics report) relieved gastrointestinal symptoms
Lactobacillus casei Shirota Human (PD) 5 weeks Reduced constipation Cassani et al. (2011)
Lactobacillus acidophilus, Lactobacillus casei, Bifido-
Human (AD) 12 weeks Improved cognition Akbari et al. (2016)
bacterium bifidum, and Lactobacillus fermentum
Probiotics modulate the gut-brain axis 177

Further, this dysbiosis may trigger systemic inflammation, mechanism by which probiotics act on the gut-brain axis.
thereby facilitating the development of PD pathology (Kesha- One of approach examined the alleviation of inflammation
varzian et al., 2015). by probiotics. B. infantis 35624 and L. rhamnosus were found
Together, these studies highlight that dysbiosis in gut mi- to increase the concentration of IL-10, an anti-inflamma-
crobiota may play important roles in the initiation or pro- tory cytokine, and to decrease the concentration of inter-
gression of various diseases, including neurological disease. feron (IFN)-γ and tumor necrosis factor (TNF)-α, in plasma
Although the exact mechanism remains to be elucidated, (O’Mahony et al., 2005; Donato et al., 2010). Modulation of
these studies suggest that microbiota may modulate the gut- inflammation by probiotics could be related to improvement
brain axis. of BBB integrity and concomitant reduction of neuroin-
flammation in patients with neurological disorders (Felger
and Lotrich, 2013; Miller et al., 2013). Additionally, admin-
Modulation of gut-brain axis by probiotics istration of B. infantis to germ-free mice induced brain-de-
rived neurotrophic factor (BDNF) and N-methyl-D-aspar-
The connections between many neurological diseases and tate (NMDA) receptor subunit 2a (NR2a) expression; these
the composition of the intestinal microbiota has led to an molecules play a crucial role in learning and memory pro-
interest in improvement of intestinal microbiota through cesses in the cortex and hippocampus, and exhibit reduced
probiotics. A growing body of evidence supports the idea expression in the germ-free mouse (Sudo et al., 2004).
that certain probiotics may positively impact the pathoge-
nesis of neurological disorders. Clinical data is less compel- Human studies
ling than the animal model data, but is rapidly emerging
Although there are fewer human clinical studies than animal
(Table 3).
studies, there is increasing evidence that probiotics are effec-
tive in reducing depression and anxiety-like symptoms.
Animal studies
Oral administration of a probiotic formulation, consisting
A variety of probiotics have been investigated in animal mo- of L. helveticus R0052 and B. longum R0175, over 30 days
dels of neurological disorders. Bifidobacterium and Lacto- was shown to improve mood in generally healthy volunteers.
bacillus are the main genera that have shown beneficial ef- Compared with the placebo group, probiotic-treated volun-
fects on neurological diseases (e.g., anxiety, depression, stress, teers exhibited alleviation of anxiety and depressive symp-
and ASD). toms, as measured by the global scores of Hospital Anxiety
Recent studies used a maternal separation model to induce and Depression scale, and the global severity index of the
early-life stress in infant rats. Maternally separated rat pups Hopkins symptoms checklist. Urinary free cortisol was also
showed a poor performance in the forced swim test, as well decreased in probiotic formulation-treated participants, sug-
as increased inflammation and mRNA expression of corti- gesting a decreased level of stress (Messaoudi et al., 2011a,
cotrophin release factor in the amygdala region of brain. 2011b). These studies indicate the beneficial effects of pro-
Treatment with probiotic B. infantis in this animal model biotics treatment on anxiety and depression-related beha-
resulted in reversal of behavior deficits and inflammation viors in human volunteers (Liu et al., 2015).
(indexed by interleukin [IL]-6 release), as well as showed The stress-reducing effects of L. casei strain Shirota have
elevation of plasma tryptophan, a serotonergic precursor also been investigated under academic stress. Healthy me-
(Desbonnet et al., 2008, 2010). dical students, who were approaching an examination for
The maternal immune activation (MIA) mouse model of academic advancement, consumed either L. casei-fermented
ASD exhibited gastrointestinal barrier defects and altered milk or a placebo milk for 8 weeks, followed by the exam. The
microbial composition, both of which are known features of administration of L. casei Shirota significantly reduced sali-
ASD. Oral treatment with the human commensal bacterium, vary cortisol levels before the examination and alleviated gas-
Bacteroides fragilis, corrected the abnormalities in gut per- trointestinal symptoms. Moreover, the probiotics group had
meability and microbial composition, as well as ASD-related significantly higher numbers of gut microbial species in their
behaviors including communication defects, anxiety-like be- feces, compared with the placebo group, and the percentage
haviors, and sensorimotor behaviors. Normal levels of se- of Bacteroidaceae was significantly lowered in the probiotics
veral metabolites were also restored upon treatment with group (Kato-Kataoka et al., 2016).
Bacteroides fragilis. Among those restored metabolites, changes Taken together, these animal and human studies show that
in 4-ethylphenylsulfate are sufficient to cause anxiety-like probiotics may play an important role in the bidirectional
behavior (Hsiao et al., 2013). communication between the gut and the brain, and that they
A recent study has provided direct evidence for modulation can modulate brain function (Fig. 1). Although many research
of neurotransmitter concentrations by probiotics treatment, groups have attempted to demonstrate a mechanism on how
using magnetic resonance spectroscopy. Mice treated with probiotics modulate the gut-brain axis in normal and diseased
L. rhamnosus JB-1 showed increased levels of glutamate, conditions, however, the exact mechanisms have not been
N-acetyl aspartate, and GABA in the brain, indicating that yet well-defined. Therefore, for effective usage of probiotics
probiotics could regulate brain activity via metabolic path- in neurological disorders, it would be necessary to elucidate
ways and suggesting possibility for clinical translation of this their veiled interactions in the gut-brain axis.
probiotic approach into therapies for neurological disorders
(Janik et al., 2016).
Many other studies have been performed to elucidate the
178 Kim et al.

Fig. 1. Modulation of the gut-brain axis


by probiotics. The routes of communica-
tion between the gut and the brain include
neuronal, immune-mediated, and meta-
bolite-mediated pathways. Gut dysbiosis
leads to increased inflammation, as well
as, activation of the HPA axis, and altered
levels of neurotransmitters and bacterial
metabolites; these may contribute to ab-
normal signaling through the vagus nerve.
Reduced integrity of the gastrointestinal
barrier triggers bacterial migration (“leaky
gut”) and inflammation. Inflammatory
cytokines induce the disruption of blood-
brain barrier integrity. Probiotics have
the potential to normalize such processes
(Abbreviations: HPA axis, hypothalamus-
pituitary gland-adrenal gland axis; NE,
norepinephrine; GABA, γ-aminobutyric
acid; BBB, blood brain barrier; EEC, en-
teroendocrine cell; EC, enterochroma-
ffin cell); GLP-1, glucagon-like peptide-1;
PYY, peptide tyrosine tyrosine; 5-HT,
5-hydroxytryptamine; SCFAs, short-chain
fatty acids.

Conclusion Acknowledgements

Accumulated information from animal and human research This research was supported by grants from the World Ins-
suggests that the gastrointestinal microbiota has an impor- titute of Kimchi (KE1802-1 and KE1802-1-1), funded by
tant role in bidirectional communication between the gut the Ministry of Science and ICT, Republic of Korea.
and brain. It interacts with the nervous system via compli-
cated direct and indirect pathways including neuronal, im-
mune-mediated and neuro-endocrine-mediated signaling. References
Imbalance in the gut microbiota is associated with neuro-
developmental defects and neurological disorders, includ- Adams, J.B., Johansen, L.J., Powell, L.D., Quig, D., and Rubin, R.A.
ing stress response, anxiety, depression, and neurodegener- 2011. Gastrointestinal flora and gastrointestinal status in children
ative diseases. Thus, a variety of laboratory and clinical stu- with autism-comparisons to typical children and correlation
dies N-methyl-D-have been conducted to determine how with autism severity. BMC Gastroenterol. 11, 22.
Aizawa, E., Tsuji, H., Asahara, T., Takahashi, T., Teraishi, T., Yoshida,
alterations in gut microbiota can serve as indicators certain S., Ota, M., Koga, N., Hattori, K., and Kunugi, H. 2016. Possible
neurological diseases. Especially, administration of probiotics association of Bifidobacterium and Lactobacillus in the gut mi-
has shown to partially or completely restore perturbations in crobiota of patients with major depressive disorder. J. Affect.
commensal microbiota, alleviating a variety of neurological Disord. 202, 254–257.
disease symptoms Akbari, E., Asemi, Z., Daneshvar Kakhaki, R., Bahmani, F., Kouchaki,
Further studies are definitely needed to clarify the mecha- E., Tamtaji, O.R., Hamidi, G.A., and Salami, M. 2016. Effect of
nism for regulation of brain function by gut microbiota, as probiotic supplementation on cognitive function and metabolic
well as to determine whether changes in commensal bacteria status in Alzheimer’s disease: A randomized, double-blind and
composition cause the disease development or are the result controlled trial. Front. Aging Neurosci. 8, 256.
Bailey, M.T. and Coe, C.L. 1999. Maternal separation disrupts the
of disease progression. Moreover, to use probiotics for pre- integrity of the intestinal microflora in infant rhesus monkeys.
vention and treatment of neurological disorders, there must Dev. Psychobiol. 35, 146–155.
be greater investment in clinical trials of probiotics. Bailey, M.T., Dowd, S.E., Galley, J.D., Hufnagle, A.R., Allen, R.G.,
and Lyte, M. 2011. Exposure to a social stressor alters the struc-
ture of the intestinal microbiota: implications for stressor-in-
duced immunomodulation. Brain Behav. Immun. 25, 397–407.
Bailey, M.T., Dowd, S.E., Parry, N.M., Galley, J.D., Schauer, D.B.,
Probiotics modulate the gut-brain axis 179

and Lyte, M. 2010. Stressor exposure disrupts commensal mi- Proc. Natl. Acad. Sci. USA 103, 10420–10425.
crobial populations in the intestines and leads to increased col- Cohen, L.J., Esterhazy, D., Kim, S.H., Lemetre, C., Aguilar, R.R., Gor-
onization by Citrobacter rodentium. Infect. Immun. 78, 1509–1519. don, E.A., Pickard, A.J., Cross, J.R., Emiliano, A.B., Han, S.M.,
Bailey, M.T., Lubach, G.R., and Coe, C.L. 2004. Prenatal stress alters et al. 2017. Commensal bacteria make GPCR ligands that mimic
bacterial colonization of the gut in infant monkeys. J. Pediatr. human signalling molecules. Nature 549, 48–53.
Gastroenterol. Nutr. 38, 414–421. Coury, D.L., Ashwood, P., Fasano, A., Fuchs, G., Geraghty, M., Kaul,
Banks, W.A. 2005. Blood-brain barrier transport of cytokines: a me- A., Mawe, G., Patterson, P., and Jones, N.E. 2012. Gastrointestinal
chanism for neuropathology. Curr. Pharm. Des. 11, 973–984. conditions in children with autism spectrum disorder: develop-
Barrett, E., Ross, R.P., O’Toole, P.W., Fitzgerald, G.F., and Stanton, ing a research agenda. Pediatrics 130 Suppl 2, S160–S168.
C. 2012. γ-Aminobutyric acid production by culturable bacteria Cryan, J.F. and Dinan, T.G. 2012. Mind-altering microorganisms:
from the human intestine. J. Appl. Microbiol. 113, 411–417. the impact of the gut microbiota on brain and behaviour. Nat.
Bengmark, S. 2013. Gut microbiota, immune development and func- Rev. Neurosci. 13, 701–712.
tion. Pharmacol. Res. 69, 87–113. De Angelis, M., Piccolo, M., Vannini, L., Siragusa, S., De Giacomo,
Benton, D., Williams, C., and Brown, A. 2007. Impact of consuming A., Serrazzanetti, D.I., Cristofori, F., Guerzoni, M.E., Gobbetti,
a milk drink containing a probiotic on mood and cognition. Eur. M., and Francavilla, R. 2013. Fecal microbiota and metabolome
J. Clin. Nutr. 61, 355–361. of children with autism and pervasive developmental disorder
Bercik, P., Denou, E., Collins, J., Jackson, W., Lu, J., Jury, J., Deng, Y., not otherwise specified. PLoS One 8, e76993.
Blennerhassett, P., Macri, J., McCoy, K.D., et al. 2011a. The in- Desbonnet, L., Garrett, L., Clarke, G., Bienenstock, J., and Dinan,
testinal microbiota affect central levels of brain-derived neuro- T.G. 2008. The probiotic Bifidobacteria infantis: An assessment
tropic factor and behavior in mice. Gastroenterology 141, 599–609. of potential antidepressant properties in the rat. J. Psychiatr.
Bercik, P., Park, A.J., Sinclair, D., Khoshdel, A., Lu, J., Huang, X., Res. 43, 164–174.
Deng, Y., Blennerhassett, P.A., Fahnestock, M., Moine, D., et al. Desbonnet, L., Garrett, L., Clarke, G., Kiely, B., Cryan, J.F., and Dinan,
2011b. The anxiolytic effect of Bifidobacterium longum NCC3001 T.G. 2010. Effects of the probiotic Bifidobacterium infantis in
involves vagal pathways for gut-brain communication. Neuro- the maternal separation model of depression. Neuroscience 170,
gastroenterol. Motil. 23, 1132–1139. 1179–1188.
Berer, K. and Krishnamoorthy, G. 2012. Commensal gut flora and Deshmukh, H.S., Liu, Y., Menkiti, O.R., Mei, J., Dai, N., O’Leary,
brain autoimmunity: A love or hate affair? Acta Neuropathol. C.E., Oliver, P.M., Kolls, J.K., Weiser, J.N., and Worthen, G.S.
123, 639–651. 2014. The microbiota regulates neutrophil homeostasis and host
Bermudez-Brito, M., Plaza-Diaz, J., Munoz-Quezada, S., Gomez- resistance to Escherichia coli K1 sepsis in neonatal mice. Nat.
Llorente, C., and Gil, A. 2012. Probiotic mechanisms of action. Med. 20, 524–530.
Ann. Nutr. Metab. 61, 160–174. Diamond, B., Huerta, P.T., Tracey, K., and Volpe, B.T. 2011. It takes
Boursi, B., Mamtani, R., Haynes, K., and Yang, Y.X. 2016. Parkin- guts to grow a brain: Increasing evidence of the important role
son’s disease and colorectal cancer risk-A nested case control of the intestinal microflora in neuro- and immune-modulatory
study. Cancer Epidemiol. 43, 9–14. functions during development and adulthood. Bioessays 33,
Bravo, J.A., Forsythe, P., Chew, M.V., Escaravage, E., Savignac, H.M., 588–591.
Dinan, T.G., Bienenstock, J., and Cryan, J.F. 2011. Ingestion of Donato, K.A., Gareau, M.G., Wang, Y.J., and Sherman, P.M. 2010.
Lactobacillus strain regulates emotional behavior and central Lactobacillus rhamnosus GG attenuates interferon-γ and tumour
GABA receptor expression in a mouse via the vagus nerve. Proc. necrosis factor-α-induced barrier dysfunction and pro-inflam-
Natl. Acad. Sci. USA 108, 16050–16055. matory signalling. Microbiology 156, 3288–3297.
Bravo, J.A., Julio-Pieper, M., Forsythe, P., Kunze, W., Dinan, T.G., Emery, D.C., Shoemark, D.K., Batstone, T.E., Waterfall, C.M., Cog-
Bienenstock, J., and Cryan, J.F. 2012. Communication between hill, J.A., Cerajewska, T.L., Davies, M., West, N.X., and Allen, S.J.
gastrointestinal bacteria and the nervous system. Curr. Opin. Phar- 2017. 16S rRNA next generation sequencing analysis shows bac-
macol. 12, 667–672. teria in Alzheimer’s post-mortem brain. Front. Aging Neurosci.
Browning, K.N., Verheijden, S., and Boeckxstaens, G.E. 2017. The 9, 195.
vagus nerve in appetite regulation, mood, and intestinal inflam- Erny, D., de Angelis, A.L.H., Jaitin, D., Wieghofer, P., Staszewski, O.,
mation. Gastroenterology 152, 730–744. David, E., Keren-Shaul, H., Mahlakoiv, T., Jakobshagen, K., and
Bu, X.L., Yao, X.Q., Jiao, S.S., Zeng, F., Liu, Y.H., Xiang, Y., Liang, C.R., Buch, T. 2015. Host microbiota constantly control maturation
Wang, Q.H., Wang, X., Cao, H.Y., et al. 2015. A study on the and function of microglia in the CNS. Nat. Neurosci. 18, 965–977.
association between infectious burden and Alzheimer’s disease. Felger, J.C. and Lotrich, F.E. 2013. Inflammatory cytokines in de-
Eur. J. Neurol. 22, 1519–1525. pression: neurobiological mechanisms and therapeutic implica-
Carabotti, M., Scirocco, A., Maselli, M.A., and Severi, C. 2015. The tions. Neuroscience 246, 199–229.
gut-brain axis: interactions between enteric microbiota, central Finegold, S.M., Dowd, S.E., Gontcharova, V., Liu, C., Henley, K.E.,
and enteric nervous systems. Ann.Gastroenterol. 28, 203–209. Wolcott, R.D., Youn, E., Summanen, P.H., Granpeesheh, D., Di-
Cassani, E., Barichella, M., Cancello, R., Cavanna, F., Iorio, L., Ce- xon, D., et al. 2010. Pyrosequencing study of fecal microflora of
reda, E., Bolliri, C., Zampella Maria, P., Bianchi, F., Cestaro, B., autistic and control children. Anaerobe 16, 444–453.
et al. 2015. Increased urinary indoxyl sulfate (indican): New in- Forsyth, C.B., Shannon, K.M., Kordower, J.H., Voigt, R.M., Shaikh,
sights into gut dysbiosis in Parkinson’s disease. Parkinsonism. M., Jaglin, J.A., Estes, J.D., Dodiya, H.B., and Keshavarzian, A.
Relat. Disord. 21, 389–393. 2011. Increased intestinal permeability correlates with sigmoid
Cassani, E., Privitera, G., Pezzoli, G., Pusani, C., Madio, C., Iorio, L., mucosa α-synuclein staining and endotoxin exposure markers
and Barichella, M. 2011. Use of probiotics for the treatment of in early Parkinson’s disease. PLoS One 6, e28032.
constipation in Parkinson’s disease patients. Minerva Gastro- Frost, G., Sleeth, M.L., Sahuri-Arisoylu, M., Lizarbe, B., Cerdan, S.,
enterol. Dietol. 57, 117–121. Brody, L., Anastasovska, J., Ghourab, S., Hankir, M., Zhang, S.,
Choi, H.J., Lee, N.K., and Paik, H.D. 2015. Health benefits of lactic et al. 2014. The short-chain fatty acid acetate reduces appetite
acid bacteria isolated from kimchi, with respect to immunomo- via a central homeostatic mechanism. Nat. Commun. 5, 3611.
dulatory effects. Food Sci. Biotechnol. 24, 783–789. Furusawa, Y., Obata, Y., Fukuda, S., Endo, T.A., Nakato, G., Takaha-
Clarke, M.B., Hughes, D.T., Zhu, C., Boedeker, E.C., and Sperandio, shi, D., Nakanishi, Y., Uetake, C., Kato, K., Kato, T., et al. 2013.
V. 2006. The QseC sensor kinase: a bacterial adrenergic receptor. Commensal microbe-derived butyrate induces the differentiation
180 Kim et al.

of colonic regulatory T cells. Nature 504, 446–450. Janik, R., Thomason, L.A.M., Stanisz, A.M., Forsythe, P., Bienenstock,
Gagliano, H., Delgado-Morales, R., Sanz-Garcia, A., and Armario, A. J., and Stanisz, G.J. 2016. Magnetic resonance spectroscopy re-
2014. High doses of the histone deacetylase inhibitor sodium veals oral Lactobacillus promotion of increases in brain GABA,
butyrate trigger a stress-like response. Neuropharmacology 79, N-acetyl aspartate and glutamate. Neuroimage 125, 988–995.
75–82. Jiang, H., Ling, Z., Zhang, Y., Mao, H., Ma, Z., Yin, Y., Wang, W.,
Gill, S.R., Pop, M., Deboy, R.T., Eckburg, P.B., Turnbaugh, P.J., Sa- Tang, W., Tan, Z., Shi, J., et al. 2015. Altered fecal microbiota com-
muel, B.S., Gordon, J.I., Relman, D.A., Fraser-Liggett, C.M., and position in patients with major depressive disorder. Brain Behav.
Nelson, K.E. 2006. Metagenomic analysis of the human distal gut Immun. 48, 186–194.
microbiome. Science 312, 1355–1359. Kabouridis, P.S., Lasrado, R., McCallum, S., Chng, S.H., Snippert,
Goehler, L.E., Park, S.M., Opitz, N., Lyte, M., and Gaykema, R.P. H.J., Clevers, H., Pettersson, S., and Pachnis, V. 2015. Microbiota
2008. Campylobacter jejuni infection increases anxiety-like beha- controls the homeostasis of glial cells in the gut lamina propria.
vior in the holeboard: possible anatomical substrates for viscero- Neuron 85, 289–295.
sensory modulation of exploratory behavior. Brain Behav. Immun. Kaluzna-Czaplinska, J. and Blaszczyk, S. 2012. The level of arabinitol
22, 354–366. in autistic children after probiotic therapy. Nutrition 28, 124–126.
Goldstone, A.P. 2006. The hypothalamus, hormones, and hunger: Kang, D.W., Park, J.G., Ilhan, Z.E., Wallstrom, G., Labaer, J., Adams,
alterations in human obesity and illness. Prog. Brain Res. 153, J.B., and Krajmalnik-Brown, R. 2013. Reduced incidence of Pre-
57–73. votella and other fermenters in intestinal microflora of autistic
Golubeva, A.V., Crampton, S., Desbonnet, L., Edge, D., O’Sullivan, children. PLoS One 8, e68322.
O., Lomasney, K.W., Zhdanov, A.V., Crispie, F., Moloney, R.D., Kato-Kataoka, A., Nishida, K., Takada, M., Kawai, M., Kikuchi-
Borre, Y.E., et al. 2015. Prenatal stress-induced alterations in ma- Hayakawa, H., Suda, K., Ishikawa, H., Gondo, Y., Shimizu, K.,
jor physiological systems correlate with gut microbiota compo- Matsuki, T., et al. 2016. Fermented milk containing Lactobacillus
sition in adulthood. Psychoneuroendocrinology 60, 58–74. casei strain Shirota preserves the diversity of the gut microbiota
Grenham, S., Clarke, G., Cryan, J.F., and Dinan, T.G. 2011. Brain-gut- and relieves abdominal dysfunction in healthy medical stu-
microbe communication in health and disease. Front. Physiol. dents exposed to academic stress. Appl. Environ. Microbiol. 82,
2, 94. 3649–3658.
Grossi, E., Melli, S., Dunca, D., and Terruzzi, V. 2016. Unexpected Kawashima, K., Misawa, H., Moriwaki, Y., Fujii, Y.X., Fujii, T.,
improvement in core autism spectrum disorder symptoms after Horiuchi, Y., Yamada, T., Imanaka, T., and Kamekura, M. 2007.
long-term treatment with probiotics. SAGE Open Med. Case Ubiquitous expression of acetylcholine and its biological func-
Rep. 4, 2050313x16666231. tions in life forms without nervous systems. Life Sci. 80, 2206–
Guthrie, G.D. and Nicholson-Guthrie, C.S. 1989. γ-Aminobutyric 2209.
acid uptake by a bacterial system with neurotransmitter bind- Keshavarzian, A., Green, S.J., Engen, P.A., Voigt, R.M., Naqib, A.,
ing characteristics. Proc. Natl. Acad. Sci. USA 86, 7378–7381. Forsyth, C.B., Mutlu, E., and Shannon, K.M. 2015. Colonic bac-
Harach, T., Marungruang, N., Duthilleul, N., Cheatham, V., Mc Coy, terial composition in Parkinson’s disease. Mov. Disord. 30, 1351–
K.D., Frisoni, G., Neher, J.J., Fak, F., Jucker, M., Lasser, T., et al. 1360.
2017. Reduction of Aβ amyloid pathology in APPPS1 transgenic Kimura, I., Ozawa, K., Inoue, D., Imamura, T., Kimura, K., Maeda,
mice in the absence of gut microbiota. Sci. Rep. 7, 41802. T., Terasawa, K., Kashihara, D., Hirano, K., Tani, T., et al. 2013.
Hardy, J. and Selkoe, D.J. 2002. The amyloid hypothesis of Al- The gut microbiota suppresses insulin-mediated fat accumula-
zheimer’s disease: progress and problems on the road to thera- tion via the short-chain fatty acid receptor GPR43. Nat. Commun.
peutics. Science 297, 353–356. 4, 1829.
Hemarajata, P., Gao, C., Pflughoeft, K.J., Thomas, C.M., Saulnier, Kunze, W.A., Mao, Y.K., Wang, B., Huizinga, J.D., Ma, X., Forsythe,
D.M., Spinler, J.K., and Versalovic, J. 2013. Lactobacillus reu- P., and Bienenstock, J. 2009. Lactobacillus reuteri enhances ex-
teri-specific immunoregulatory gene rsiR modulates histamine citability of colonic AH neurons by inhibiting calcium-dependent
production and immunomodulation by Lactobacillus reuteri. J. potassium channel opening. J. Cell Mol. Med. 13, 2261–2270.
Bacteriol. 195, 5567–5576. Landete, J.M., De las Rivas, B., Marcobal, A., and Munoz, R. 2008.
Hill, C., Guarner, F., Reid, G., Gibson, G.R., Merenstein, D.J., Pot, Updated molecular knowledge about histamine biosynthesis by
B., Morelli, L., Canani, R.B., Flint, H.J., Salminen, S., et al. 2014. bacteria. Crit. Rev. Food Sci. Nutr. 48, 697–714.
Expert consensus document. The international scientific asso- Lim, S.K., Kwon, M.S., Lee, J., Oh, Y.J., Jang, J.Y., Lee, J.H., Park,
ciation for probiotics and prebiotics consensus statement on the H.W., Nam, Y.D., Seo, M.J., Roh, S.W., et al. 2017. Weissella ci-
scope and appropriate use of the term probiotic. Nat. Rev. Gast- baria WIKIM28 ameliorates atopic dermatitis-like skin lesions
roenterol. Hepatol. 11, 506–514. by inducing tolerogenic dendritic cells and regulatory T cells in
Holzer, P. and Farzi, A. 2014. Neuropeptides and the microbiota- BALB/c mice. Sci. Rep. 7, 40040.
gut-brain axis. Adv. Exp. Med. Biol. 817, 195–219. Liu, X., Cao, S., and Zhang, X. 2015. Modulation of gut microbiota-
Horn, T. and Klein, J. 2013. Neuroprotective effects of lactate in brain brain axis by probiotics, prebiotics, and diet. J. Agric. Food Chem.
ischemia: dependence on anesthetic drugs. Neurochem. Int. 62, 63, 7885–7895.
251–257. Louveau, A., Smirnov, I., Keyes, T.J., Eccles, J.D., Rouhani, S.J., Peske,
Hosoi, T., Okuma, Y., and Nomura, Y. 2002. The mechanisms of im- J.D., Derecki, N.C., Castle, D., Mandell, J.W., Lee, K.S., et al. 2015.
mune-to-brain communication in inflammation as a drug target. Structural and functional features of central nervous system
Curr. Drug Targets Inflamm. Allergy 1, 257–262. lymphatic vessels. Nature 523, 337–341.
Hsiao, E.Y., McBride, S.W., Hsien, S., Sharon, G., Hyde, E.R., McCue, Lyte, M. 2011. Probiotics function mechanistically as delivery ve-
T., Codelli, J.A., Chow, J., Reisman, S.E., Petrosino, J.F., et al. hicles for neuroactive compounds: Microbial endocrinology in
2013. Microbiota modulate behavioral and physiological abnor- the design and use of probiotics. BioEssays 33, 574–581.
malities associated with neurodevelopmental disorders. Cell 155, Macfabe, D.F. 2012. Short-chain fatty acid fermentation products of
1451–1463. the gut microbiome: implications in autism spectrum disorders.
Inan, M.S., Rasoulpour, R.J., Yin, L., Hubbard, A.K., Rosenberg, D.W., Microb. Ecol. Health Dis. 23, 19260.
and Giardina, C. 2000. The luminal short-chain fatty acid buty- Macfarlane, S. and Macfarlane, G.T. 2003. Regulation of short-chain
rate modulates NF-κB activity in a human colonic epithelial cell fatty acid production. Proc. Nutr. Soc. 62, 67–72.
line. Gastroenterology 118, 724–734. Macpherson, A.J. and Uhr, T. 2004. Compartmentalization of the
Probiotics modulate the gut-brain axis 181

mucosal immune responses to commensal intestinal bacteria. Rafiki, A., Boulland, J.L., Halestrap, A.P., Ottersen, O.P., and Ber-
Ann. N. Y. Acad. Sci. 1029, 36–43. gersen, L. 2003. Highly differential expression of the monocar-
McCusker, R.H. and Kelley, K.W. 2013. Immune-neural connections: boxylate transporters MCT2 and MCT4 in the developing rat
how the immune system’s response to infectious agents influ- brain. Neuroscience 122, 677–688.
ences behavior. J. Exp. Biol. 216, 84–98. Rhee, S.H., Pothoulakis, C., and Mayer, E.A. 2009. Principles and
McVey Neufeld, K.A., Mao, Y.K., Bienenstock, J., Foster, J.A., and clinical implications of the brain-gut-enteric microbiota axis.
Kunze, W.A. 2013. The microbiome is essential for normal gut Nat. Rev. Gastroenterol. Hepatol. 6, 306–314.
intrinsic primary afferent neuron excitability in the mouse. Neu- Rios-Covian, D., Ruas-Madiedo, P., Margolles, A., Gueimonde, M.,
rogastroenterol. Motil. 25, e183–e188. de Los Reyes-Gavilan, C.G., and Salazar, N. 2016. Intestinal short
Messaoudi, M., Lalonde, R., Violle, N., Javelot, H., Desor, D., Nejdi, chain fatty acids and their link with diet and human health. Front.
A., Bisson, J.F., Rougeot, C., Pichelin, M., Cazaubiel, M., et al. Microbiol. 7, 185.
2011a. Assessment of psychotropic-like properties of a probiotic Sarkar, A., Lehto, S.M., Harty, S., Dinan, T.G., Cryan, J.F., and Burnet,
formulation (Lactobacillus helveticus R0052 and Bifidobacterium P.W. 2016. Psychobiotics and the manipulation of bacteria-gut-
longum R0175) in rats and human subjects. Br. J. Nutr. 105, 755– brain signals. Trends Neurosci. 39, 763–781.
764. Savignac, H.M., Kiely, B., Dinan, T.G., and Cryan, J.F. 2014. Bifido-
Messaoudi, M., Violle, N., Bisson, J.F., Desor, D., Javelot, H., and bacteria exert strain-specific effects on stress-related behavior
Rougeot, C. 2011b. Beneficial psychological effects of a probiotic and physiology in BALB/c mice. Neurogastroenterol. Motil. 26,
formulation (Lactobacillus helveticus R0052 and Bifidobacterium 1615–1627.
longum R0175) in healthy human volunteers. Gut microbes 2, Scheperjans, F., Aho, V., Pereira, P.A., Koskinen, K., Paulin, L., Pek-
256–261. konen, E., Haapaniemi, E., Kaakkola, S., Eerola-Rautio, J., Pohja,
Meuer, K., Pitzer, C., Teismann, P., Kruger, C., Goricke, B., Laage, R., M., et al. 2015. Gut microbiota are related to Parkinson’s disease
Lingor, P., Peters, K., Schlachetzki, J.C., Kobayashi, K., et al. 2006. and clinical phenotype. Mov. Disord. 30, 350–358.
Granulocyte-colony stimulating factor is neuroprotective in a Shyu, W.C., Lin, S.Z., Yang, H.I., Tzeng, Y.S., Pang, C.Y., Yen, P.S.,
model of Parkinson’s disease. J. Neurochem. 97, 675–686. and Li, H. 2004. Functional recovery of stroke rats induced by
Miller, A.H., Haroon, E., Raison, C.L., and Felger, J.C. 2013. Cyto- granulocyte colony-stimulating factor-stimulated stem cells. Cir-
kine targets in the brain: impact on neurotransmitters and neu- culation 110, 1847–1854.
rocircuits. Depress. Anxiety 30, 297–306. Song, Y., Liu, C., and Finegold, S.M. 2004. Real-time PCR quanti-
Mountzouris, K.C., Tsirtsikos, P., Kalamara, E., Nitsch, S., Schatz- tation of clostridia in feces of autistic children. Appl. Environ.
mayr, G., and Fegeros, K. 2007. Evaluation of the efficacy of a Microbiol. 70, 6459–6465.
probiotic containing Lactobacillus, Bifidobacterium, Enterococcus, Steenbergen, L., Sellaro, R., van Hemert, S., Bosch, J.A., and Colzato,
and Pediococcus strains in promoting broiler performance and L.S. 2015. A randomized controlled trial to test the effect of mul-
modulating cecal microflora composition and metabolic activi- tispecies probiotics on cognitive reactivity to sad mood. Brain
ties. Poult. Sci. 86, 309–317. Behav. Immun. 48, 258–264.
O’Mahony, L., McCarthy, J., Kelly, P., Hurley, G., Luo, F., Chen, K., Stilling, R.M., Dinan, T.G., and Cryan, J.F. 2014. Microbial genes,
O’Sullivan, G.C., Kiely, B., Collins, J.K., Shanahan, F., et al. 2005. brain & behaviour - epigenetic regulation of the gut-brain axis.
Lactobacillus and Bifidobacterium in irritable bowel syndrome: Genes Brain Behav. 13, 69–86.
symptom responses and relationship to cytokine profiles. Gastro- Strati, F., Cavalieri, D., Albanese, D., De Felice, C., Donati, C., Hayek,
enterology 128, 541–551. J., Jousson, O., Leoncini, S., Renzi, D., Calabro, A., et al. 2017.
O’Mahony, S.M., Marchesi, J.R., Scully, P., Codling, C., Ceolho, A.M., New evidences on the altered gut microbiota in autism spectrum
Quigley, E.M., Cryan, J.F., and Dinan, T.G. 2009. Early life stress disorders. Microbiome 5, 24.
alters behavior, immunity, and microbiota in rats: implications Sudo, N., Chida, Y., Aiba, Y., Sonoda, J., Oyama, N., Yu, X.N., Kubo,
for irritable bowel syndrome and psychiatric illnesses. Biol. Psy- C., and Koga, Y. 2004. Postnatal microbial colonization programs
chiatry 65, 263–267. the hypothalamic-pituitary-adrenal system for stress response
Obermeier, B., Daneman, R., and Ransohoff, R.M. 2013. Develop- in mice. J. Physiol. 558, 263–275.
ment, maintenance and disruption of the blood-brain barrier. Sun, B.L., Wang, L.H., Yang, T., Sun, J.Y., Mao, L.L., Yang, M.F.,
Nat. Med. 19, 1584–1596. Yuan, H., Colvin, R.A., and Yang, X.Y. 2017. Lymphatic drainage
Overduin, J., Schoterman, M.H., Calame, W., Schonewille, A.J., and system of the brain: A novel target for intervention of neuro-
Ten Bruggencate, S.J. 2013. Dietary galacto-oligosaccharides and logical diseases. Prog. Neurobiol. doi: 10.1016/j.pneurobio.2017.
calcium: effects on energy intake, fat-pad weight and satiety-re- 08.007 (in press).
lated, gastrointestinal hormones in rats. Br. J. Nutr. 109, 1338– Surwase, S.N. and Jadhav, J.P. 2011. Bioconversion of L-tyrosine
1348. to L-DOPA by a novel bacterium Bacillus sp. JPJ. Amino Acids
Özogul, F. 2011. Effects of specific lactic acid bacteria species on 41, 495–506.
biogenic amine production by foodborne pathogen. Int. J. Food Thayer, J.F. and Sternberg, E.M. 2009. Neural concomitants of im-
Sci. Technol. 46, 478–484. munity-focus on the vagus nerve. Neuroimage 47, 908–910.
Parracho, H.M., Bingham, M.O., Gibson, G.R., and McCartney, Thomas, C.M., Hong, T., van Pijkeren, J.P., Hemarajata, P., Trinh,
A.L. 2005. Differences between the gut microflora of children D.V., Hu, W., Britton, R.A., Kalkum, M., and Versalovic, J. 2012.
with autistic spectrum disorders and that of healthy children. J. Histamine derived from probiotic Lactobacillus reuteri suppresses
Med. Microbiol. 54, 987–991. TNF via modulation of PKA and ERK signaling. PLoS One 7,
Prakash, A., Medhi, B., and Chopra, K. 2013. Granulocyte colony e31951.
stimulating factor (GCSF) improves memory and neurobehavior Tillisch, K. 2014. The effects of gut microbiota on CNS function
in an amyloid-beta induced experimental model of Alzheimer’s in humans. Gut Microbes 5, 404–410.
disease. Pharmacol. Biochem. Behav. 110, 46–57. Tillisch, K., Labus, J., Kilpatrick, L., Jiang, Z., Stains, J., Ebrat, B.,
Psichas, A., Sleeth, M.L., Murphy, K.G., Brooks, L., Bewick, G.A., Guyonnet, D., Legrain-Raspaud, S., Trotin, B., Naliboff, B., et al.
Hanyaloglu, A.C., Ghatei, M.A., Bloom, S.R., and Frost, G. 2015. 2013. Consumption of fermented milk product with probiotic
The short chain fatty acid propionate stimulates GLP-1 and PYY modulates brain activity. Gastroenterology 144, 1394–1401.e4.
secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. Tremaroli, V. and Backhed, F. 2012. Functional interactions between
(Lond) 39, 424–429. the gut microbiota and host metabolism. Nature 489, 242–249.
182 Kim et al.

Vetulani, J. 2013. Early maternal separation: a rodent model of de- spp. and Ruminococcus torques in feces of children with autism
pression and a prevailing human condition. Pharmacol. Rep. 65, spectrum disorder. Mol. Autism 4, 42.
1451–1461. Wang, Y. and Kasper, L.H. 2014. The role of microbiome in central
Vogt, N.M., Kerby, R.L., Dill-McFarland, K.A., Harding, S.J., Mer- nervous system disorders. Brain. Behav. Immun. 38, 1–12.
luzzi, A.P., Johnson, S.C., Carlsson, C.M., Asthana, S., Zetter- Westfall, S., Lomis, N., Kahouli, I., Dia, S.Y., Singh, S.P., and Prakash,
berg, H., and Blennow, K. 2017. Gut microbiome alterations in S. 2017. Microbiome, probiotics and neurodegenerative diseases:
Alzheimer’s disease. Sci. Rep. 7, 13537. deciphering the gut brain axis. Cell. Mol. Life Sci. 74, 3769–3787.
Wallner, S., Peters, S., Pitzer, C., Resch, H., Bogdahn, U., and Sch- Williams, B.B., Van Benschoten, A.H., Cimermancic, P., Donia,
neider, A. 2015. The granulocyte-colony stimulating factor has M.S., Zimmermann, M., Taketani, M., Ishihara, A., Kashyap,
a dual role in neuronal and vascular plasticity. Front. Cell Dev. P.C., Fraser, J.S., and Fischbach, M.A. 2014. Discovery and char-
Biol. 3, 48. acterization of gut microbiota decarboxylases that can produce
Wang, L., Christophersen, C.T., Sorich, M.J., Gerber, J.P., Angley, the neurotransmitter tryptamine. Cell Host Microbe 16, 495–503.
M.T., and Conlon, M.A. 2011. Low relative abundances of the Yang, N.J. and Chiu, I.M. 2017. Bacterial signaling to the nervous
mucolytic bacterium Akkermansia muciniphila and Bifidobac- system through toxins and metabolites. J. Mol. Biol. 429, 587–605.
terium spp. in feces of children with autism. Appl. Environ. Mic- Yano, J.M., Yu, K., Donaldson, G.P., Shastri, G.G., Ann, P., Ma,
robiol. 77, 6718–6721. L., Nagler, C.R., Ismagilov, R.F., Mazmanian, S.K., and Hsiao,
Wang, L., Christophersen, C.T., Sorich, M.J., Gerber, J.P., Angley, E.Y. 2015. Indigenous bacteria from the gut microbiota regu-
M.T., and Conlon, M.A. 2013. Increased abundance of Sutterella late host serotonin biosynthesis. Cell 161, 264–276.

You might also like