Sie sind auf Seite 1von 21

Ann. N.Y. Acad. Sci.

ISSN 0077-8923

A N N A L S O F T H E N E W Y O R K A C A D E M Y O F SC I E N C E S
Special Issue: The Year in Neurology and Psychiatry
REVIEW

Recent developments in understanding the role of the gut


microbiota in brain health and disease
Eoin Sherwin,1 Timothy G. Dinan,1,2 and John F. Cryan1,3
1
APC Microbiome Institute, University College Cork, Cork, Ireland. 2 Department of Psychiatry and Neurobehavioural Sciences,
University College Cork, Cork, Ireland. 3 Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland

Address for correspondence: John F. Cryan, Department of Anatomy and Neuroscience, Office 386, Western Gateway
Building, University College Cork, Cork, Ireland. J.Cryan@ucc.ie

There is a growing appreciation of the role of the gut microbiota in all aspects of health and disease, including
brain health. Indeed, roles for the bacterial commensals in various psychiatric and neurological conditions, such
as depression, autism, stroke, Parkinson’s disease, and Alzheimer’s disease, are emerging. Microbiota dysregulation
has been documented in all of these conditions or in animal models thereof. Moreover, depletion or modulation
of the gut microbiota can affect the severity of the central pathology or behavioral deficits observed in a variety of
brain disorders. However, the mechanisms underlying such effects are only slowly being unraveled. Additionally,
recent preclinical and clinical evidence suggest that targeting the microbiota through prebiotic, probiotic, or dietary
interventions may be an effective “psychobiotic” strategy for treating symptoms in mood, neurodevelopmental
disorders, and neurodegenerative diseases.

Keywords: microbiota; brain; aging; neurodegeneration; behavior; diet

Introduction Over the past decade, it has become clear that


the bidirectional communication pathway between
Advances in sequencing and metabolomics tech-
gut bacteria and the central nervous system
nologies have enabled scientists to explore the
(CNS), the microbiota–gut–brain axis, exerts a
full extent of how microbes affect every aspect
profound influence on key brain processes, such
of the world we live in. Indeed, almost all areas
as neuroinflammation, activation of the stress axes,
of medicine have been shown to be affected by
neurotransmission, and neurogenesis, in addition
the trillions of bacteria that reside within the gut,
to modulating complex behaviors, such as socia-
including neurology and psychiatry. However, it is
bility and anxiety.4–13 Gut bacteria influence these
easily forgotten that the concept of microorganisms
central processes through their ability to synthesize
conferring host health benefits has existed for some
neurotransmitters (i.e., ␥ -amino butyric acid
time. It is now over 100 years since the Nobel laure-
(GABA), noradrenaline, and dopamine) and mod-
ate Elie Metchnikoff of the Pasteur Institute in Paris
ulate activation of the immune system, along with
proposed that lactic acid bacteria may play a key role
their ability to produce metabolites, such as short-
in the programming of the aging process.1 Hubert
chain fatty acids (SCFAs), that possess neuroactive
J. Norman, working at the Camberwell House
properties.4 Moreover, the gut microbiota and the
asylum, and George Porter Phillips, working in the
brain are linked through additional pathways, such
Bethlem Royal Hospital, both in London, tested
as the vagus pathway, and through the modulation
the concept that lactic acid bacteria may amelio-
of key dietary amino acids, such as tryptophan14–17
rate the symptoms of depression.2,3 Since then,
(Fig. 1). Given the close association between the gut
considerable advances have been made in under-
microbiota and the brain, it is not all that surprising
standing how the bacterial commensals within our
that gut bacteria have roles to play in neurological
gastrointestinal system can influence host health.
and psychiatric diseases. Evidence now suggests that

doi: 10.1111/nyas.13416
Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 
C 2017 New York Academy of Sciences. 5
The gut microbiota in brain health and disease Sherwin et al.

Figure 1. Key mechanisms underlying communication between the gut microbiota and the brain. Gut bacteria can signal to
the brain through a variety of mechanisms. These include the production of SCFA metabolites, modulation of immune signaling
to the brain, and transmission via the vagus nerve. These mechanisms serve to influence central homeostatic processes, such
as neurotransmission, neurogenesis, and neuroinflammation, all of which are involved in several neurological and psychiatric
conditions. SCFAs, short-chain fatty acids; BBB, blood–brain barrier; HPA, hypothalamic–pituitary–adrenal axis; CNS, central
nervous system; GABA, ␥-amino butyric acid.

the bacterial commensals of the gut can influence Aging


conditions such as depression, schizophrenia, Advances in modern medicine have led to an
and autism.5,15,18–20 While great strides have been increase in human longevity. While humans are liv-
taken in elucidating how bacterial commensals ing longer, the onus is now on medical and scientific
may be involved in these conditions, more insight research to mitigate the severity of age-related dis-
is required before microbiota-based therapies eases (i.e., cognitive decline, stroke, gastrointestinal
can be rationally employed as viable treatment disturbances, cancer, etc.). Although there is a
options. Perhaps what is surprising, however, growing body of evidence as to how bacterial com-
is the extent to which gut bacteria appear to mensals are affected with age,21–23 there is limited
be involved in other facets of brain health and information to date about how such changes may
behavior. Emerging research now suggests that affect the microbiota–gut–brain axis. However, this
the microbiota–gut–brain axis is also involved in deficit is beginning to be addressed.24 For instance,
neurodegenerative diseases, such as Alzheimer’s we have recently characterized the microbiota com-
disease and Parkinson’s disease, and psychiatric position of aged (20–21 months old) versus young
conditions, such as addiction. (2–3 months old) mice.25 Aged animals displayed an
altered microbiota, with changes observed in phyla
Microbiota and neurology: aging that have previously been associated with inflamma-
and neurodegeneration tion, which is consistent with previous reports.25,26
With an increase in the age that humans are living Aged animals display increased basal intestinal
there is a need to better understand the biologi- permeability, which is exacerbated in response to
cal mechanisms underpinning the aging process. In stress. Such increases in intestinal permeability are
light of this, it is important to elucidate how aging likely to increase the risk for the translocation of
affects the microbiota–gut–brain axis, as it may pro- bacteria or bacterial components (i.e., lipopolysac-
vide us with a greater insight into age-related dis- charide (LPS)) from the gut lumen into the
eases, such as stroke and neurodegenerative diseases circulation, where they can elicit an inflammatory
(i.e., Parkinson’s disease and Alzheimer’s disease). response. Indeed, aged mice display an increase

6 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

in several circulating inflammatory cytokines (i.e., While these are promising preclinical studies, such
interleukin (IL)-1␤, IL-4, IL-2, and tumor necrosis modulators of the microbiota will need to be
factor (TNF)-␣) compared with young mice.25 assessed in humans in order to determine whether
Other studies assessing the effect of age on they display efficacy in mitigating the deleterious
the murine microbiota have reported conflicting effects of aging in humans. Nonetheless, these are
results. For instance, Conley et al. reported no dif- promising findings that suggest that targeting the
ferences in the relative ratio of Firmicutes to Bac- microbiota may help to prolong human longevity
teroidetes in aged mice (26 months old), with an and attenuate age-related inflammation.
increase in the Muscipirillum genus their most sig-
nificant outcome.27 However, a separate study by Parkinson’s disease
Kim et al. identified an increase in the level of Parkinson’s disease is characterized pathologically
Firmicutes and Actinobacteria with a correspond- by degeneration of the pars compacta of the substan-
ing decrease in Bacteroidetes in aged animals (18 tia nigra of the mid brain and widespread neuroin-
months).28 However, these differences may be due flammation. Neurologically, parkinsonian patients
to the fact that aged female C57 mice were used in exhibit a range of motor-related impairments,
the study by Conley et al., while Kim et al. used such as bradykinesia, resting tremors, muscular
aged male C57 mice.27,28 Although these studies rigidity, and impairments in posture. Before the
report different alterations to the microbiota with development of the motor symptoms, parkinsonian
respect to age, both argue that the altered microbiota patients often report prodromal nonmotor-related
may be driving the observed elevated inflammatory symptoms, such as depression, sleep disturbances,
response in aged animals.27,28 The elevated circulat- and constipation, suggestive of gastrointestinal
ing inflammatory profile of aged animals may help dysfunction.31,32 Moreover, gastrointestinal func-
to explain the cognitive behavioral deficits reported tion is further exacerbated following progression
in these animals.25 of the disease with constipation, impaired gastric
While the microbiota appears to be affected emptying, and difficulties with defecation all
by aging, evidence suggests that targeting the gut reported in patients.32 Furthermore, ␣-synuclein,
commensals provides an opportunity to mitigate the protein aggregate hallmark of Parkinson’s dis-
age-associated inflammation and other deleterious ease pathology in the brain, has also been identified
conditions associated with age. For instance, in the mucosal and submucosal nerve fibers and
treatment of aged mice (18 months old) with the ganglia of the enteric nervous systems (ENSs)33,34
Lactobacillus brevis OW38 strain was found to of parkinsonian patients, with some preclinical
improve the expression of intestinal tight junction evidence even suggesting that ␣-synuclein in the
proteins, along with reducing the Firmicutes to Bac- gut can transport to the brain via the vagus nerve.35
teroidetes ratio.29 Moreover, this Lactobacillus strain Given the gastrointestinal disturbances reported
attenuated circulating levels of inflammatory mark- in Parkinson’s disease, it stands to reason that
ers (IL-1␤ and TNF-␣) while also increasing the the microbiota–gut–brain axis is affected in this
expression of the neurotrophin brain-derived neu- neurodegenerative disease. Indeed, Scheperjans
rotrophic factor in the hippocampus.29 Polyphenols et al. identified a reduced abundance of the
have also demonstrated efficacy in attenuating Prevotella species in fecal samples from parkin-
age-associated inflammation and dysregulation to sonian patients.36 A more recent clinical study
the gut microbiota of mice. Treatment of middle- also identified a reduction in the abundance of
aged mice (40 weeks old) with the polyphenolic Prevotella species (P. copri) in the microbiota of
lignan syringaresinol reduced circulating levels of parkinsonian patients, along with increases in
the proinflammatory marker LPS binding protein Akkermansia muciniphilia.37 Given that Prevotella
(LBP), while also increasing the humoral response species produce mucin, which serves to enhance
to vaccination with the influenza virus.30 Moreover, the integrity of the intestinal barrier, a reduced
syringaresinol modulated the gut microbiota by abundance of this bacteria strain may lead to
increasing the relative abundance of Lactobacillus increases in intestinal permeability in parkinsonian
and Bifidobacterium species, while decreasing levels patients and subsequent bacterial translocation.36
of the potentially harmful Akkermansia genus.30 In a separate study, the fecal microbiota of

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 7
The gut microbiota in brain health and disease Sherwin et al.

parkinsonian patients were found to contain lower the microbiota–gut–brain axis may be involved in
levels of anti-inflammatory–associated bacteria the neurodegenerative disease. In the absence of
from the genera Blautia and Roseburia and a higher a microbiota, ␣-synuclein–overexpressing mice do
level of proinflammatory-associated bacteria from not show as great an impairment in motor and gas-
the genus Faecalibacterium when compared with trointestinal symptoms when compared with con-
healthy controls.38 Recent work by Hill-Burns et al. ventionally colonized mice of the same genotype.47
also identified alterations to the gut microbiota Microglia in the striatum of germ-free (mice com-
in parkinsonian patients; however, the changes pletely devoid of a microbiota; see Fig. 2 for a sum-
reported were different from those in previous mary of the neurobiological changes observed in
reports.39 Although these studies report different germ-free mice), ␣-synuclein–overexpressing mice
alterations to the gut microbiota of parkinsonian displayed a less activated phenotype relative to con-
patients, they collectively suggest that there is ventional colonized mice of the same genotype, sug-
dysregulation to the gut commensals that may gesting a less reactive phenotype in the absence of
facilitate inflammation and bacterial translocation. a microbiota, which is consistent with reports from
Further corroborating a dysregulated microbiota– previous studies in germ-free mice.47,48 Moreover,
gut–brain axis in Parkinson’s disease was a recent administration of SCFAs (acetate, butyrate, and pro-
observation by Unger et al., who demonstrated pionate) to germ-free, ␣-synuclein–overexpressing
that the levels of SCFAs—the microbial metabolites mice exacerbated motor deficits to a level compara-
acetate, butyrate, and propionate—were all signif- ble to conventionally colonized mice of the same
icantly lower in parkinsonian patients relative to genotype, suggesting that SCFAs are the facilita-
age-matched healthy controls.40 Given that SCFAs tors of the adverse effect that the microbiota has
are able to cross into the brain and possess neu- on motor symptoms.47 It is worth noting, however,
roactive properties, such as regulating microglial that the concentration of SCFAs administered to
activation,41 a reduction in their production in mice in this study were considerably higher than
Parkinson’s disease may affect disease pathology. what would expected to normally be found in vivo.
Other reports of microbiota dysregulation in While the data are compelling in suggesting that
Parkinson’s disease include small intestine bacterial SCFAs are the mediators of motor dysfunction in
overgrowth or high prevalence rates of colonization these experiments, more work is required to deter-
with the Gram-negative bacterium Helicobacter mine whether elevated levels of endogenous SCFAs
pylori.42,43 Parkinsonian patients are found to are driving these behavioral changes. Moreover, the
have increased urinary indoxyl sulfate levels, authors demonstrate that microbiota derived from
which is indicative of overgrowth of bacteria in parkinsonian patients exacerbated motor symptoms
the small intestine or constipation.44 H. pylori of ␣-synuclein–overexpressing mice compared with
infection may contribute to the development of those mice who received a microbiota from a healthy
parkinsonian symptoms through degenerating human, suggesting that dysregulation of the micro-
dopaminergic neurons in the brain.45 Interest- biota can worsen motor symptoms when combined
ingly, eradication of H. pylori has been shown with a genetic predisposition.47 Given these obser-
to enhance the onset time of levodopa (first-line vations, targeting the microbiota–gut–brain axis
pharmacological treatment for Parkinson’s dis- may help alleviate some of the neurological and gas-
ease symptoms) while also improving tremor, trointestinal symptoms of Parkinson’s disease.
rigidity, and walking ability.43,46 Metagenomic
analysis has revealed that genes involved in the Alzheimer’s disease
biosynthesis of LPS are upregulated in parkin- Alzheimer’s disease is the most common form
sonian patients, suggesting that a dysregulated of age-related dementia worldwide. The neu-
microbiota is potentially driving aberrant immune rodegenerative disease is characterized by the
activation in the neurodegenerative disease.38 accumulation of amyloid plaques, tau fibrils, and
Animal models of Parkinson’s disease have pro- widespread neuroinflammation that culminates in
vided considerable insight into the pathology of severe cognitive impairments, such as long-term
the neurodegenerative disease. Recent intriguing memory loss, and other symptoms, such as physical
work has identified a potential mechanism for how disability and exhaustion. Given the neurological

8 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

Figure 2. Germ-free mice display alterations to central physiological processes. In the absence of a microbiota, germ-free mice
display several unique alterations to brain physiology and chemistry. Microglia from germ-free mice display an immature phenotype
and fail to respond to bacterial-associated molecular patterns (i.e., LPS) as efficiently as specific pathogen-free or conventional
mice. These mice also display increases in the permeability of the blood–brain barrier (BBB), which corresponds to a reduction in
the expression of tight junction proteins. Reduction in the integrity of the BBB may allow for the translocation of immune cells
and bacterial components into the brain of germ-free mice. Germ-free mice also exhibit increased neurogenesis, which corresponds
to an increase in the volume of the hippocampus of these animals in comparison with specific pathogen-free or conventional
animals. Finally, germ-free mice display several unique alterations to neuronal homeostasis. For instance, these animals exhibit
hypermyelination of the frontal cortex along with increases in serotonin turnover in the hippocampus. Moreover, there is an increase
in markers of synaptic and neuronal plasticity in these animals, which may correspond to the increases in neurogenesis and brain
volume observed in these animals. For a comprehensive review on germ-free mice, see Ref. 122. LPS, lipopolysaccharide.

nature of this disease, perhaps it is surprising that brain parenchyma and blood vessels of Alzheimer’s
emerging evidence suggests that the microbiota– patients.50 Moreover, LPS was found to colocalize
gut–brain axis may have a role to play in this with A␤1–40 in amyloid plaques, suggesting that
condition. Recently, Cattaneo et al. identified that bacterial components do indeed translocate from
the Escherichia/Shigella bacterial genera, which the gut into the systemic circulation and reach the
are associated with mediating inflammation, were brain in Alzheimer’s disease.50 The microbiota may
increased in fecal samples from Alzheimer’s patients influence neurodegeneration in conditions like
relative to control subjects. Moreover, the increase Alzheimer’s disease through molecular mimicry.
in these bacterial taxa positively correlated with an Dendritic cells in the intestine may sense and
increase in the expression of the proinflammatory transport bacterial amyloid antigens to immune
cytokines IL-1␤ and CXCL2 in whole blood.49 Such cells in Peyer’s patches. For instance, the bacterial
results suggest a causal link between dysregulation extracellular amyloid protein curli is sensed by the
of the microbiota and systemic inflammation, A11 A␤ oligomer antibody, which suggests that
which may initiate or exacerbate the neurodegen- there are configurations of amyloid proteins that
eration that is occurring in the brain in Alzheimer’s have the same structure as microbial proteins.51
disease. Given that intestinal permeability increases Preclinical data have provided a greater insight
with age,25 bacteria or bacterial components (i.e., into the role that the microbiota–gut–brain axis may
LPS) may translocate from the lumen of the gut play in Alzheimer’s disease. In a recent study, both
and mediate systemic and neuroinflammation. In young and old germ-free mice overexpressing amy-
support of this, levels of LPS and the Escherichia coli loid precursor protein and presenilin 1 (APP/PS1)
K99 pilli protein have been found to be higher in the were found to display reduced levels of A␤42

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 9
The gut microbiota in brain health and disease Sherwin et al.

compared with conventional APP/PS1 mice, along vitro, suggesting that a polyphenol-rich diet may
with a reduction in microglial activation.52 Addi- attenuate amyloid accumulation in the brain.55
tionally, these germ-free APP/PS1 mice displayed The probiotic mixture VSL#3 (combination of
increased levels of A␤-degrading enzymes, insulin- Streptococcus thermophilus, Bifidobacterium breve,
degrading enzyme, and neprilysin-degrading B. longum, B. infantis, Lactobacillus acidophilus,
enzyme, suggesting that, in the absence of a micro- L. plantarum, L. paracasei, and L. delbrueckii sub-
biota, the murine brain is better equipped to tar- species Bulgaricus) attenuated age-related deficits
get the amyloid pathology.52 Moreover, in a sepa- in long-term potentiation, suggesting an improve-
rate study, chronic treatment of APP/PS1 transgenic ment in memory, in rats while also ameliorating
mice with an antibiotic cocktail reduced microglial age-related microglial activation.56 Despite these
and astrocyte accumulation surrounding amyloid interesting preliminary preclinical studies, more
plaques in the hippocampus.53 This observation is studies are required with both aged and transgenic
not all that surprising, given that the microbiota can animals to determine whether diet or probiotics may
influence glial cell activation and neuroinflamma- be employed into the clinical setting as a preventa-
tion in the brain.48 Interestingly, however, antibi- tive measure for the onset of Alzheimer’s disease.
otic treatment was found to decrease insoluble A␤
plaques while leading to a corresponding increase Multiple sclerosis
in the concentration of soluble A␤ levels, which Multiple sclerosis is an autoimmune neurodegener-
suggests that the gut microbiota has some bear- ative disease caused by the progressive loss of myelin
ing on amyloid load in the brain.53 Other ani- sheaths surrounding the axons of neurons. The
mal models of Alzheimer’s disease have also doc- degenerative disease is characterized by a variety of
umented alterations to the microbiota–gut–brain neurological symptoms, such as vision impairment,
axis. In the 5xFAD transgenic model of Alzheimer’s ataxia, muscle spasms, paralysis in severe cases, and
disease (which rapidly develops amyloid plaques), fatigue. Moreover, bladder- and gastrointestinal-
levels of human A␤ precursor protein are elevated related symptoms have also been reported in multi-
not only in the brains of these mice compared with ple sclerosis. For instance, Buscarinu et al. recently
wild type but also in the gastrointestinal system demonstrated that there is an increase in intestinal
(cecum, colon, jejunum, etc.).54 Moreover, these permeability along with a reduction in intestinal
5xFAD mice displayed an altered microbiota relative absorption in patients with the relapsing/remitting
to wild-type controls; with an increase observed in form of the condition.57 A separate clinical study
the Firmicutes/Bacteroidetes ratio. Within the Fir- identified an increase in circulating LBP in patients
micutes phylum, levels of the Clostridium leptum with relapsing/remitting multiple sclerosis, poten-
species, which are associated with inflammation, tially indicating an increase in the translocation of
were found to be transiently increased in 5xFAD Gram-negative bacteria from the intestinal lumen
mice.54 While these preclinical studies suggest that to the circulation.58 In addition to alterations in
the microbiota–gut–brain axis may be involved in gastrointestinal physiology, there are reports of dys-
Alzheimer’s disease, more work is required to deter- regulation to the composition of the gut microbiota
mine whether alterations to the bacterial commen- in patients with the condition. Relapsing/remitting
sals are facilitating the disease or whether a dysreg- multiple sclerosis patients displayed high interindi-
ulated microbiota is a consequence of the central vidual variability compared with healthy patients,
neurodegeneration. while there were no significant differences reported
While little is currently known regarding the role in terms of species richness.59 At the genus level,
of the microbiota–gut–brain axis in Alzheimer’s the gut microbiota of patients with multiple
disease, preliminary preclinical evidence indicates sclerosis displayed significant reductions in Fae-
that diet or modulators of the microbiota (i.e., calibacterium, Prevotella, and Anaerostiples.59 More
probiotics) may provide a means to ameliorate the clinical data are required to determine whether these
neurodegenerative disease. For instance, grape seed alterations to the gut microbiota and the gastroin-
polyphenol extracts 3-hydroxybenzoic acid and testinal system in multiple sclerosis are driving
3-(3-hydroxyphenyl)propionic acid were shown to the pathology or if these alterations are simply a
prevent the assembly of A␤1–42 into toxic fibrils in comorbid consequence of the disease.

10 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

Preclinical studies have provided a greater insight beneficial Lactobacillus strains could be used as a
into how the microbiota–gut–brain axis is affected preventative strategy for multiple sclerosis.
in multiple sclerosis and whether it contributes
to the observed pathology. For instance, recent Stroke and brain injury
work from our group has demonstrated that the To date, only a few studies have investigated how
gut microbiota influences myelination within the the gut microbiota may be involved in stroke and
prefrontal cortex of mice.60 Germ-free mice dis- ischemic brain damage. Clinical data are limited
played increased expression of myelin-related genes at the moment, but some studies have identified
(i.e., Mag, Mbp, Mog, and Mobp) in the prefrontal dysregulation to the microbiota in patients follow-
cortex relative to conventional mice, which was ing stroke.65 Alterations to the human microbiota
restored to control levels following recolonization following stroke have been observed, with specific
with microbiota.60 Antibiotic administration also decreases observed in the Bacteroides fragilis group
increased the expression of myelin-related genes in and increases in an Atopobium cluster noted.66
the prefrontal cortex of nonobese diabetic mice, Moreover, microbial metabolism was affected by
which further corroborates that the gut microbiota stroke, with decreases in fecal concentration of
influences myelination within the CNS.61 Interest- acetic acid and increases in valeric acid and isova-
ingly, germ-free mice display significantly attenu- leric acid.66 Preclinical data have provided us with a
ated experimental autoimmune encephalomyelitis greater insight into how the microbiota–gut–brain
(EAE; animal model of multiple sclerosis) scores axis may be affected in stroke and brain injury. For
relative to conventional controls.62 Kyung et al. instance, depletion of gut bacteria through antibi-
demonstrated that the reduced EAE severity in otic administration worsened the survival rate of
germ-free mice was due to a reduced ability of den- mice following the induction of ischemia.67 In the
dritic cells in these animals to activate proinflam- middle cerebral artery occlusion (MCAO) preclin-
matory T cells.62 Consequently, in the absence of ical model of stroke, cerebral ischemia is associated
a microbiota, activation of proinflammatory TH 17 with a dysregulation of the murine microbiota,
cells, which drive neurological damage in multi- with a reduction in bacterial ␣-diversity, along with
ple sclerosis, is blunted. Interestingly, when germ- reductions in intestinal motility and intestinal bar-
free mice were colonized with segmented filament rier dysfunction observed as an increase in intestinal
bacteria, EAE deficits were restored in these ani- permeability.68,69 Microbial-derived metabolites
mals, further corroborating that the microbiota may also influence stroke susceptibility through
modulates the proinflammatory status of the brain modulating platelet activation and thrombosis. For
during EAE. In a separate study, Miller et al. instance, elevated levels of the microbial metabolite
demonstrated that TNF receptor signaling plays trimethylamine N-oxide (TMAO; produced from
a role in the cross talk between the host micro- the microbial metabolism of TMA-rich foods)
biota and the immune system during multiple scle- were shown to correlate with an increased risk for
rosis. Inhibition of TNFR2 signaling exacerbated thrombosis in cardiovascular patients.70 TMAO
multiple sclerosis pathology in myelin oligoden- promoted platelet aggregation in the presence of
drocyte glycoprotein-specific 2D2 T cell receptor thrombin through enhancing intracellular calcium
(TCR) mice.63 However, antibiotic administration release.70 Interestingly, while dietary choline or
attenuated disease severity and mortality in these TMA increased TMAO production and thrombosis
mice.63 These findings are likely to have an impor- in conventional mice, this effect was not observed
tant bearing on anti-TNF therapies for multiple scle- in germ-free mice, indicating that the microbiota
rosis in patients. Interestingly, the composition of facilitates the production of TMAO through the
the gut microbiota may determine the severity of metabolism of choline-rich foods. Bacteria within
multiple sclerosis. Rats subjected to EAE were more the Allobaculum taxa (associated with choline
resistant toward developing multiple sclerosis–like metabolism) were positively correlated with throm-
symptoms if they contained a higher diversity of bosis, indicating that these bacteria may drive
Lactobacillus spp. relative to those rats that had a TMAO production.70 Wang et al. demonstrated
lower diversity of the lactic acid bacteria.64 Such that inhibiting microbial TMA lyases with the
results suggest that targeting the microbiota using choline analogue 3,3-dimethyl-1-butanol reduced

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 11
The gut microbiota in brain health and disease Sherwin et al.

in vivo levels of TMAO while also reducing of certain species of Prevotella in vitro.75 Increased
atherosclerosis, which suggests that the microbiota activation of the sympathetic nervous system fol-
may present a therapeutic target for the treatment lowing stroke or traumatic brain injury may lead
of cardiovascular diseases, such as stroke.71 to increased noradrenergic innervation of the gas-
There is considerable evidence to suggest that the trointestinal system that may help to account for
gut microbiota modulates immune signaling dur- the observed dysregulation to the microbiota in
ing stroke to influence the pathological outcome. these conditions. In support of this, Houlden et al.
Transplantation of a dysregulated poststroke micro- administered 6-hydroxy-dopamine, which results in
biota into germ-free mice exacerbated the subse- a potent increase in systemic noradrenaline release,
quent cerebral damage mediated by MCAO in these to mice and observed a decrease in the abundance
animals, which was associated with increased traf- of Prevotellaceae.74 Additionally, recent preclinical
ficking of proinflammatory T lymphocytes (TH 1 data suggest that the gut microbiota can influence
and TH 17 phenotype) of intestinal origin to the the severity of neurological damage following spinal
cerebral infarct site.68 Further evidence for a role cord injury.76
of intestinal IL-17 in mediating ischemic damage An additional complication associated with
comes from comprehensive experiments performed stroke is poststroke infection, which has a high mor-
by Benakis et al. The authors eloquently demon- tality rate. Evidence suggests that the gut microbiota
strated that dysregulation of the murine micro- can influence the severity of poststroke infection.
biota following antibiotic treatment resulted in a For instance, poststroke infection was observed only
reduction in the trafficking of the proinflamma- in specific pathogen–free mice and not germ-free
tory IL-17+ ␥ ␦ T cells, which was associated with a mice, indicating that the bacteria driving the sys-
reduction in IL-17–associated chemokine expres- temic inflammation following stoke originated from
sion in brain parenchyma, along with reduced within the host.69 Moreover, it has been demon-
neutrophil accumulation and a reduction in infarct strated that, following MCAO in mice, there is an
volume of the ischemic site.72 Thus, the gut micro- increase in intestinal permeability, which suggests
biota appears to influence the magnitude of post- that intestinal bacteria are capable of translocating
stroke neuroinflammation by modulating intestinal from the lumen of the gut into the systemic circu-
T cell trafficking to the meninges. A recent study lation whereby they can invade other organs (i.e.,
by Sadler et al. highlighted that mice of the same the lungs) and elicit an inflammatory response.69 In
strain (C57BL/6) but provided from different ven- support of this, 16S sequencing of the lung micro-
dors harbor distinct microbiotas, which influences biota revealed an increase in bacterial species in
the neuroinflammatory response following experi- the lungs that were predicted to originate from the
mental stroke.73 Notably, the absence of segmented small intestine.69 Thus, it appears that the intestinal
filamented bacteria in C57 mice from one ven- microbiota can not only influence the neuroinflam-
dor resulted in a reduced ability to produce anti- matory status through influencing T cell migration
inflammatory Treg cells following stroke.73 Such to the brain but also systemic inflammation follow-
results suggest that the use of mice from different ing stroke through bacterial translocation from the
vendors introduces a potential confound of differ- gut into the circulation, whereby they can invade
ing microbiotas, even if they are the same strain of other organs. On the basis of these results, target-
animal. ing the increased intestinal permeability in stroke
Increased activation of the sympathetic nervous patients may help to limit bacterial translocation
system may also influence dysregulation to the leading to sepsis. Moreover, developing a probiotic
microbiota following stroke or traumatic brain strain that could modulate T lymphocyte signal-
injury. MCAO or traumatic brain injury in mice ing to the brain may help to ameliorate the neu-
resulted in a dysregulation to the microbiota, roinflammatory status in patients following stroke.
observed as a decrease in the relative abundance While data are currently limited, there is some pre-
of Prevotellaceae and Peptococcaceae, which corre- clinical evidence to suggest that probiotic strains
lated with an increase in noradrenaline release and can ameliorate the neurological damage caused
noradrenergic innervation of the cecum.74 Nora- by stroke. Clostridium butyricum, for instance,
drenaline has been shown to inhibit the expression prevented neuronal apoptosis mediated by bilateral

12 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

common carotid artery occlusion (BCAO) in mice implicated in depression.80 Zheng et al. corrobo-
while also improving performance in the Morris rated these results when they demonstrated that
water maze task, suggesting an improvement in transplantation of a dysregulated microbiota from
cognition in these animals.77 This bacterial strain depressed humans to germ-free mice conferred a
also increased levels of beneficial gut bacteria, such depressive-related phenotype in these animals.81
as Bifidobacteria, Lactobacilli, and Fecalibacterium Recent work from De Palma et al. also demonstrated
prausnitzii, in BCAO mice.77 the successful transfer of behavior from humans to
mice following microbiota transplantation. Trans-
Microbiota and stress-related disorders plantation of microbiota from patients with irrita-
Accumulating evidence suggests that there is dysreg- ble bowel syndrome with anxiety to germ-free mice
ulation to the microbiota–gut–brain axis in stress- resulted in gastrointestinal symptoms and anxiety-
related disorders, such as depression and anxiety. related behaviors in these animals.82 These findings
Understanding how the gut microbiota is affected indicate that dysregulation to the gut microbiota is
in such conditions will allow for the development of capable of facilitating the behavioral and physiolog-
microbiota-based therapies, which may provide us ical symptoms of depression and anxiety.
with safer and more efficacious treatment options
for such conditions. Microbiota-based therapies for the treatment
of depression
Depression While a greater understanding is required as to how
Evidence for a role of the gut microbiota in depres- the gut microbiota can mediate the behavioral and
sion and other stress-related disorders has pre- physiological symptoms of depression, emerging
dominantly arisen from preclinical studies (for preclinical and clinical data are highlighting how
reviews on this topic, see Refs. 4, 14, 18, and 78). modulators of the gut commensals (i.e., prebiotics
For instance, it was recently shown that chronic and probiotics) may be an appropriate therapy for
stress results in dysregulation of the microbiota and depression and other stress-related disorders. For
metabolome of rats, with decreases observed in the instance, recent work from our group has shown
Firmicutes/Bacteroidetes ratio, and, more specifi- that a prebiotic combination of fructooligosaccha-
cally, decreases in the relative abundances of Lac- ride and galactooligosaccharide (GOS) improved
tobacillus and increases in Oscillibacter.79 While the depression and anxiety-related behavior in
preclinical evidence is rather compelling, only a few mice subjected to chronic psychosocial stress.83
clinical studies to date have performed microbiota Moreover, this prebiotic combination reduced
analysis in depressed patients to assess for any poten- stress-induced activation of the hypothalamic–
tial dysregulation. We have recently combined both pituitary–adrenal (HPA) axis while also amelio-
clinical and preclinical research to demonstrate how rating dysregulation of the microbiota brought
the microbiota–gut–brain axis is affected in depres- about by chronic stress.83 Other groups have also
sion. Depressed patients were found to have a dys- documented the beneficial effects of GOS and other
regulated microbiota, observed as a reduction in prebiotics on anxiety-related behavior in pre-
species richness and microbial diversity.80 Interest- clinical models.84,85 Additionally, GOS supple-
ingly, when the fecal microbiotas of these depressed mentation was found to lower the cortisol awak-
patients were transplanted into microbiota-depleted ening response in healthy volunteers while shifting
(antibiotic cocktail) rats, the depression behavioral attention from negative toward positive stimuli,
phenotype was also transferred into the animals.80 demonstrating that the prebiotic also displays
Rats that received the microbiota from depressed efficacy in humans.86
patients displayed a dysregulated microbiota them- The probiotic strain Lactobacillus rhamnosus
selves, along with an anhedonic and anxiety-related JB-1 has been shown to improve anxiety and social-
behavioral phenotype. Moreover, these animals dis- related behaviors in mice following chronic social
played an elevated kynurenine/tryptophan ratio, defeat,87 which is in line with previous work from
indicating that perhaps the depressed microbiota our group, which also demonstrated the beneficial
is facilitating the conversion of tryptophan into behavioral effects of this particular strain.88 Such
the deleterious metabolite kynurenine, which is findings support the use of microbiota modulators

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 13
The gut microbiota in brain health and disease Sherwin et al.

for the treatment of depression and stress-related Microbiota, sociability, and


disorders. However, we must err on the side of neurodevelopmental disorders
caution with our use of animal models for assessing
There is growing evidence to support a role for
the efficacy of prebiotics and probiotics and how
the microbiota in regulating social behaviors in
this translates to the clinical setting. For instance,
mammals.4 How the gut microbiota modulates
we have recently shown that the L. rhamnosus
social behavior remains to be fully elucidated. Given
JB-1 strain, which has demonstrated efficacy
the complexity and variability of the gut microbiota
preclinically, displayed no effect in healthy male
from one species to another, identifying specific
volunteers in terms of cognition, subjective stress
microbial compositions or even specific bacterial
measures, activation of the HPA axis, or immune
species that promote social behavior is challenging.
activation.89 A lack of clinical efficacy of L. rham-
Moreover, the mechanisms through which the
nosus JB-1 may be due to the fact that mice possess
gut microbiota promote social behavior remain
a different gut microbiota than that of humans and,
unknown. However, there is growing evidence to
thus, may react differently to the probiotic strain.
suggest a role for microbiota modulation of certain
Moreover, the two studies that demonstrated bene-
neurotransmitter/peptide systems in the brain.95,96
ficial behavioral effects in mice used either an inher-
ently anxious mouse strain (BALB/c) or employed The gut microbiota influences social behaviors
chronic stress to promote an anxious phenotype, The concept that the gut microbiota may influence
which would have different reactions to stress than social behavior arose from experiments with
healthy male humans.87,88 Despite the probiotic germ-free mice. Germ-free mice were found to have
effects of L. rhamnosus JB-1 strain being somewhat deficits in social behavior in the three-chamber
lost in translation, other candidate bacterial strains interaction test.97,98 Germ-free mice exhibit social
have demonstrated efficacy as potential psychobi- avoidance behavior in this test, observed as an
otics in humans. For instance, the probiotic strain increase in the time spent in the empty chamber
B. longum 1714 demonstrated efficacy in reducing and a decrease in time spent in the chamber with
stress-induced cortisol release, along with attenuat- a novel conspecific mouse.98 This social deficit is
ing daily self-reported levels of stress and improving reversed following recolonization with microbiota,
visuospatial memory in healthy male volunteers.90 further confirming that gut bacteria modulate social
A fermented milk product containing several dif- behaviors. However, microbiota reconstitution did
ferent probiotic strains altered the activity of brain not improve social cognitive deficits in the three-
regions associated with emotionality in healthy chamber test, as these mice failed to spend more
female participants, suggesting potential psychobi- time with a novel conspecific mouse over a familiar
otic potential.91 Moreover, a recent pilot study in animal.98 Moreover, the transfer of cued food infor-
patients with irritable bowel syndrome demon- mation through social contact and communication
strated that the probiotic B. longum NCC3001 in the social transmission of food preference test was
improved scores of depression, but not anxiety, similar between conventional controls and germ-
while also improving gastrointestinal symptoms.92 free mice. Such results suggest that some facets of
While these studies are certainly promising, future social behavior are amenable through manipulation
studies assessing the therapeutic potential of of the microbiota, while others are not. Further
psychobiotics for the treatment of depression reaffirming this association between microbiota and
will require large-scale recruitment of depressed social behavior is that depletion of microbiota of
patients to determine the extent of their efficacy. conventionally colonized mice following antibiotic
In addition to prebiotics and probiotics, emerging treatment also results in social deficits.99 Interest-
evidence has demonstrated that microbiota trans- ingly, Arentsen et al. reported opposing behavioral
plantation is also capable of modifying behavior and results, demonstrating that germ-free mice display
may be an effective treatment strategy for depres- increases in social behaviors in the three-chamber
sion and other neuropsychiatric disorders.93,94 test relative to specific pathogen–free mice.100 This
However, robust clinical data are required to may be due to differences in performing the three-
support the use of microbiota transplantation as a chamber social interaction test or to differences in
potential treatment strategy for depression. animal breeding/husbandry techniques.

14 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

A growing body of evidence suggests that, in open-label study by Kang et al. demonstrated that
a variety of animal models of autism spectrum transfer of a standardized human gut microbiota
disorders, there is an altered microbiota composi- mixture was capable of improving gastrointestinal
tion. For instance, in the in utero valproate animal and behavioral symptoms in autistic children.108
model of autism, the pups display an autistic-like Moreover, these improvements to the gastrointesti-
phenotype along with intestinal inflammation and nal and behavioral symptoms through microbiota
dysregulation to the microbiota.101,102 Specifically, transfer were maintained for up to 8 weeks follow-
these animals display an increase in the Firmi- ing the cessation of the therapy, suggesting that
cutes/Bacteroidetes ratio along with increases in the intervention had long-lasting effects on the
cecal butyrate concentrations, suggesting alter- microbiota.108 While these results are preliminary
ations to microbial metabolism.101 Interestingly, and lack appropriate controls, it lends support to
there were specific increases reported in the the use of microbiota-based therapies in the future
operational taxonomic units assigned to the genera to manage the gastrointestinal and behavioral symp-
Alistipes, Mollicutes, Lactobacillales, and Enterorhab- toms of autism. However, as autism is a genetic
dus in valproate in utero–exposed males. Given that disorder,109 more research is needed to investigate
autism is more prevalent in males than in females, the relationship between host genetics and the gut
perhaps the microbiota has a bearing on the greater microbiota in shaping behaviors in autism.
risk of autism in males than in females. Alterations Emerging preclinical research suggests that can-
in gut microbiota composition have also been didate bacterial strains are capable of improv-
reported in other environmental models of autism, ing the core behavioral symptoms of autism. In
such as the maternal immune activation model, the maternal immune model of autism, Hsiao
suggesting that exposure to an in utero stress (i.e., et al. demonstrated that B. fragilis was capable of
teratogens or inflammagens) negatively affects gut improving stereotyped and anxiety-related behav-
commensals and behavior.103 Genetic animal mod- iors in this animal model while also improving
els of autism allow us to investigate the relationship intestinal permeability.103 This led Gilbert et al. to
between the host genome and the microbiota. The provocatively and, perhaps prematurely, propose
BTBR animal model of autism displays a robust the concept of probiotics for treating autism.110
autistic-like phenotype, with deficits in social More recently, Buffington et al. identified that, in
behaviors, repetitive behaviors, and anxiety-related an animal model of autism (offspring of moth-
behaviors frequently reported.104–106 Recently, it ers fed a high-fat diet), fecal levels of Lactobacil-
was demonstrated that BTBR mice display a reduc- lus reuteri were lower compared with nonautistic
tion in the Firmicutes/Bacteroidetes ratio, along controls.97 These autistic-like mice also displayed
with increases in the abundance of species such as reduced oxytocin immunoreactivity in the par-
A. mucinphilia and reductions in Bifidobacterium aventricular nucleus (PVN) of the hypothalamus.97
spp, suggestive of microbiota dysregulation.107 This Interestingly, supplementation with L. reuteri ame-
strain may prove a useful model for dissecting liorated autistic-related behaviors in these mice
the association of host genetics and microbiota in through enhancing oxytocin immunoreactivity in
facilitating an autistic-like phenotype. the PVN of the hypothalamus.97 These behavioral
and physiological effects appeared to be specific
Microbiota-based therapies for the treatment to L. reuteri, as supplementation with L. john-
of autism: hype or hope? sonii, which was also found to be reduced in these
Although more insight is required into how gut mice with autistic-like behaviors, had no effect
bacteria influence social behaviors and other behav- on these parameters.97 Given the association of
ioral aspects associated with autism, recent evidence oxytocin with social behaviors and autism, it is
suggests that modulating the microbiota through likely that the hormone plays a role in the psy-
diet, probiotics, and microbiota transfer is capa- chobiotic effects of the bacterium. It may be a
ble of modifying some aspects of behavior relevant peptide released from the bacterium, as health-
to autism. While most studies that have demon- killed L. reuteri failed to increase oxytocin
strated the beneficial effects of modulating the immunoreactivity within the PVN,97 while lysed
microbiota have been preclinical, a recent small L. reuteri was still capable of mediating this effect.111

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 15
The gut microbiota in brain health and disease Sherwin et al.

There remain several unanswered questions regard- preclinical and clinical data suggest that chronic
ing the prosocial effects of L. reuteri. For instance, alcohol consumption can negatively affect the gut
more information regarding L. reuteri’s effect on microbiota, which is perhaps not all that surprising
other brain regions related to the processing of given that ethanol is consumed via the gastrointesti-
social behaviors (i.e., the amygdala) is required. In nal system.114,115 Recently, it was shown that chronic
the study by Buffington et al., the authors noted ethanol consumption in alcoholics results in alter-
that there was no effect of the bacterial strain ations to their microbiota; decreasing the relative
on repetitive or anxiety-related behaviors. Thus, a abundance of the Bacteroidetes phylum and increas-
greater understanding of the neurocircuitry affected ing the relative abundance of Proteobacteria.116
in autism is required. Moreover, is this behavioral Intestinal permeability is also found to be increased
effect a direct consequence of consuming L. reuteri? in a subset of alcoholics, increasing the risk for
Or does this bacterial strain alter the host micro- bacterial translocation in these individuals.117
biota, which then improves social behavior? Aside Moreover, alcoholics with increased intestinal per-
from probiotics, the SCFA butyrate has recently meability also displayed a dysregulated microbiota,
been shown to improve autistic-related behaviors with increases in the Blautia and Megasphera gen-
in BTBR mice.106 Butyrate treatment improved era, and decreased levels of the anti-inflammatory
deficits in social and repetitive behaviors in BTBR Faecalibacterium prausnitzii.117 F. prausnitzii has
mice while also modulating the expression of genes been shown to inhibit the production of the proin-
related to excitatory and inhibitory neurotransmis- flammatory cytokine IL-8 in vitro, demonstrating
sion, suggesting that such microbial metabolites its anti-inflammatory potential.118 Interestingly,
may present a potential therapeutic opportunity for low levels of F. prausnitzii were correlated with
the treatment of autistic behavior.106 elevated levels of circulating IL-8 in alcoholics
with increased intestinal permeability, suggestive
Microbiota and addiction of proinflammatory immune activation.117 Addi-
While considerable advances are being made with tionally, the psychological status of alcoholics with
regard to the role of the microbiota–gut–brain axis increased intestinal permeability was worse than
in conditions, such as neurodegenerative diseases, that of controls and alcoholics with regular intesti-
and psychiatric conditions, such as depression, little nal permeability, which suggests that a dysregulated
is known with regard to the role that gut microbes microbiota–gut–brain axis is facilitating the
play in substance abuse disorders. When we consider psychological symptoms observed in alcoholics.117
drug addiction and any potential association with Recent work from our group demonstrated that
a dysregulated microbiota, it is important to con- chronic intermittent ethanol exposure in mice,
sider comorbidities, such as depression and anx- which models chronic alcohol exposure in humans,
iety. Moreover, adequate and nutritional dietary resulted in an increase in the Allistipes genera along
intake is likely to be lacking in drug addicts, which with a reduction in Clostridium cluster IV.115 Bac-
may also affect the composition of the microbiota. terial species found within Clostridium cluster IV,
Finally, many pharmacological agents,112 includ- such as F. prausnitzii, possess anti-inflammatory
ing psychotropics,113 can have direct effects on the properties.114 Thus, a loss of this genus following
microbiota, which complicates any interpretation. chronic alcohol exposure may promote a proinflam-
Nonetheless, there is growing evidence that the matory immune state in the gastrointestinal system.
microbiota can modulate behaviors and physiolog- In support of this, germ-free mice that were human-
ical changes relevant to substance abuse. ized with the microbiota of alcoholics with hepatitis
displayed a higher level of inflammation and per-
Alcohol meability of the intestine compared with controls.114
Chronic abuse of alcohol can lead to several serious Moreover, the microbiota of these humanized germ-
health complications, such as addiction, nutrient free mice lacked known beneficial bacteria, such
deficiency, liver disease, and colorectal cancer. as F. prausnitzii, and contained bacterial species
Moreover, chronic alcohol exposure can lead to that have been previously associated with intestinal
psychological and cognitive deficits, such as major inflammation, such as Bilophila wadsworthia.114,119
depression and Korsakoff ’s syndrome. Recent Thus, chronic alcohol exposure appears to lead to a

16 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

dysregulation in the composition of the microbiota; in such countries.125,126 It is important to consider


promoting a proinflammatory state within the gut, the factors that have driven the evolution of the
which could affect the microbiota–gut–brain axis Westernized microbiota from that of more rural
and behavior in these individuals. microbiotas, such as that of the Hadza community.
Sonnenburg et al. demonstrated that a diet con-
Cocaine and cannabis sisting of low microbiota–accessible carbohydrates
Data regarding the effects of other drugs of abuse resulted in a reduction in the diversity of gut bac-
upon the microbiota are currently limited. How- teria of mice (that harbored a human microbiota),
ever, there is some preliminary evidence to suggest which was largely reversible following changing the
that certain drugs, such as cannabis and cocaine, diet to a higher level of carbohydrates that bacteria
affect the microbiota–gut–brain axis. Depletion could consume. Interestingly, however, gut bacte-
of the murine gut microbiota with an antibi- rial diversity worsened across generations in mice
otic cocktail served to enhance unbiased condi- on a low microbiota–accessible diet, which became
tional place preference for cocaine, which was less amenable to improvement following restora-
reversed following treatment of antibiotic-treated tion with a high-carbohydrate diet.123 The results of
mice with SCFAs.120 Such data indicate that the this study highlight how the human gut microbiota
gut microbiota can influence drug-seeking behavior. may have potentially evolved to be less diverse and
9 -Tetrahydrocannibinol (THC), the active psy- amenable to modulation following the movement
chotropic chemical in cannabis, was shown to alter of humans from more indigenous, rural communi-
the microbiota of mice, observed as an increase ties to Westernized lifestyles. Moreover, these results
in Clostridium leptum and decreases in Roseburia may help to explain why diseases and psychiatric dis-
species.121 Moreover, in diet-induced obese mice, orders are more prominent in Western countries.
THC lowered the Firmicutes/Bacteroidetes ratio Despite this, modifying one’s diet has been shown
while also increasing A. muciniphilia, a bacterial to improve many facets of behavior.
strain that has been previously associated with The consumption of a Mediterranean-style diet
weight loss, reducing insulin resistance, and improv- (i.e., fruits, vegetables, unsalted nuts, fish, lean
ing intestinal barrier function.122 Whether THC red meat, etc.) is linked to a reduced likelihood
affects gut–brain signaling to improve weight loss for the development of depression in comparison
(i.e., modulation of satiety peptides) is unknown. to the Western-style diet, which is linked to a
However, such preliminary data warrant a greater greater risk for developing the mood disorder.126
investigation into how THC can influence gut Preclinical evidence has identified the beneficial
microbiota composition and weight loss. While effects of polyunsaturated fatty acids. For instance,
more work is required into understanding how perinatal omega-3 fatty acid supplementation
drugs of abuse, such as cannabis and cocaine, affect lowered stress-induced activation of the HPA axis
the microbiota and, subsequently, the gut–brain in adolescent mice while also enhancing cognition
axis, these studies provide an insight into how this in adulthood, demonstrating the beneficial effects
axis may be affected in addicts. of dietary supplements on stress and behavior.127
Moreover, the omega-3 fatty acids docosahexaenoic
The diet–microbiota–gut–brain axis:
acid and eicosapentaenoic acid have been shown to
toward nutritional psychiatry
improve anxiety and depression-related behaviors
Diet has been shown to have an important bear- in female rats.128 Given that stress-related psychi-
ing on the composition of the microbiota.123 For atric conditions are more prevalent in females, these
instance, the fecal microbiota of the rural Hadza preclinical data suggest that diet may be an effective
community, who mostly forage in the wild for their strategy for treating stress-related disorders in
diet, was found to be unique from that of a West- women. Until recently, most evidence regarding the
ernized population.124 Moreover, the diets of West- effect of diet on mood had been limited to animal
ernized countries are found to be suboptimal with studies and observational studies in humans.126
respect to the consumption of nutrients and high However, in a recent randomized controlled trial
in saturated fats, which may help to explain the conducted by Jacka et al., the authors demonstrated
prevalence of diseases and psychiatric conditions that a modified version of the Mediterranean diet

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 17
The gut microbiota in brain health and disease Sherwin et al.

(ModiMedDiet) was a beneficial adjunctive therapy brain, such as through the vagus nerve, immune
for the treatment of clinical depression.129 Individ- system, production of microbial metabolites, and
uals with mild depression reported improvement modulation of circulating tryptophan.8,10,17,135
in depression symptomology as assessed by the Understanding how the gut commensals modulate
Montgomery-Åsberg Depression rating scale behavior remains one of the biggest unanswered
(MADS) following the 12-week dietary trial. questions in microbiota–gut–brain axis research.
While these are preliminary results, they are rather However, emerging studies are providing some
promising, and they suggest that modifying one’s insight into the mechanisms underlying how gut
diet to incorporate a more Mediterranean-style of bacteria influence social behaviors. In a recent study
food intake is a beneficial strategy for the treatment from the Karolinska Institute, the authors identify
of depression. Indeed, dietary changes may be a a role for bacterial-sensing molecules in regulating
less-costly and safer strategy for the treatment of social development.136 Genetic knockdown of the
mild depression for patients rather than conven- peptidoglycan-sensing molecule PGLYRP2 was
tional antidepressant medication. However, much found to increase social behaviors in both male
more research is needed to give an evidence base to and female mice in the three-chamber test while
such hypotheses. also modulating the expression of genes related to
Additionally, emerging preclinical data sug- synaptic plasticity.136 The authors argue that bacte-
gest that the diet is also capable of improving rial components, such as peptidoglycan, cross the
autistic-related behavior. For instance, a high-fat, blood–brain barrier during postnatal development
low-carbohydrate ketogenic diet has also been and bind to molecules, such as PGLYRP2, to influ-
shown to improve deficits in social behaviors ence neuronal circuits linked to social behaviors.136
and repetitive behaviors in the BTBR mice as Other biological mechanisms, such as the
well as in the environmental models of autism, immune system and vagal signaling, are also likely
maternal immune activation, and valproate in utero to mediate the gut microbiota’s influence on social
exposure.130–132 Moreover, a modified amino acid behavior. D’Mello et al. demonstrated that periph-
diet (containing high concentrations of histidine, eral inflammation, mediated by bile duct ligation,
lysine, and threonine, with low concentrations resulted in social withdrawal behavior in mice.137
of leucine, isoleucine, and valine) was shown to This behavioral deficit was ameliorated following
improve repetitive self-grooming behavior in BTBR treatment of mice with the probiotic mixture VSL#3,
mice while also attenuating mammalian target of indicating that gut bacteria can influence how the
rapamycin (mTOR) signaling in the prefrontal immune system signals to the brain to modulate
cortex.133 Currently, there is little information behaviors, such as sociability and anxiety.137 In a
available as to whether the ketogenic diet or other separate study, vagotomy blocked the anxiogenic
diets may improve behavioral symptoms in autistic effects of intestinal colitis in mice, further corrobo-
individuals. However, recently, it was demonstrated rating that the vagal pathway is an important medi-
that a 6-week gluten-free diet intervention was ben- ator for the gut microbiota to influence behavior.138
eficial in reducing gastrointestinal symptoms as well Gut bacteria may also signal via the vagus nerve to
as modestly improving the behavioral symptoms of influence social behaviors. Oral administration of
autism in children.134 While these studies highlight the probiotic strain L. reuteri has been shown to
the beneficial effects that diet may have upon increase the central expression and secretion of the
behavior, a greater understanding of the mecha- prosocial hormone oxytocin in mice.97,139 However,
nisms underlying how diet or dietary components this effect of L. reuteri was lost following vagotomy,
improves social behavior and mood is required. indicating that gut commensals signal via the vagus
nerve to increase the synthesis and secretion of oxy-
Moving toward mechanisms
tocin from the brain. The production of neuroactive
In the last number of years, there has been a clear metabolites by gut bacteria may also influence social
focus on understanding the mechanisms underlying behaviors. Levels of the microbial metabolite cresol,
how microbiota can influence gut–brain function were found to be elevated in the ceca of socially
(Fig. 3). It is clear that there are multiple mecha- withdrawn mice.61 Interestingly, in vitro experi-
nisms through which gut bacteria can signal to the ments demonstrated that cresol was capable of

18 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

Figure 3. Overview of the gut–microbiota–brain axis in neurology and psychiatry. Evidence now suggests that the microbiota–gut–
brain axis is involved in a variety of neurological and psychiatric conditions, such as depression, addiction, stroke, and Parkinson’s
disease. Moreover, modulating the microbiota in these conditions using probiotics, prebiotics, or through diet has displayed efficacy
in preclinical studies, with some clinical studies also demonstrating efficacy. However, a greater understanding of how the intestinal
commensals are affected in these various conditions will allow for the rational development of microbiota-based therapies in these
various disorders.

inhibiting the expression of myelin genes and the The gut microbiota is also capable of influencing
differentiation of oligodendrocytes.61 Thus, ele- anorexigenic signaling to the brain, thereby mod-
vated levels of cresol may be driving social avoid- ulating feelings of satiety. Within the lumen of the
ance behaviors through modulating the expres- gastrointestinal system, the gut microbiota is in close
sion of myelin-related genes. In support of this, we proximity to enteroendocrine cells, and evidence
have recently demonstrated that the gut microbiota suggests that gut microbes are capable of modu-
influences myelination in the prefrontal cortex of lating the secretion of satiety peptides from these
mice.60 In addition to producing various bioactive cells. Breton et al. demonstrated that the E. coli
metabolites, the gut microbiota also synthesize var- chaperone protein caseinolytic protease (Clp) B is
ious neurotransmitters, such as noradrenaline and capable of increasing plasma levels of the satiety
GABA.140,141 Moreover, certain spore-forming bac- peptides glucagon-like peptide 1 (GLP-1) and pep-
teria have been shown to influence the secretion of tide YY (PYY) while also suppressing food intake
serotonin from enterochromaffin cells, which subse- and increasing neuronal activation within hypotha-
quently influences local gut physiology.142 Whether lamic nuclei that are associated with satiety.144
microbially derived neurotransmitters are capable ClpB likely mediates this increase in GLP-1 and
of reaching the brain and influencing their specified PYY through activating melanocortin receptor 4
circuitry and behavior is unknown. However, the (MCR4) expressed on the cell surface of enteroen-
microbiota has been shown to influence the level docrine cells. In support of this, ClpB was found to
of some of these neurotransmitters in the brain. be an antigen mimetic of ␣-melanocyte–stimulating
For instance, germ-free mice display increased hip- hormone (␣-MSH), which binds to and activates
pocampal serotonin levels relative to conventionally MCR4.145,146 In addition to indirectly activating
colonized animals.17,143 This is most likely due to the satiety-related nuclei in the brain through increas-
observed increased availability of circulating tryp- ing the secretion of GLP-1 and PYY, Breton et al.
tophan in these animals owing to the absence of gut also demonstrated that ClpB was capable of acti-
bacteria that utilize the amino acid.17 vating proopiomelanocortin neurons when applied

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 19
The gut microbiota in brain health and disease Sherwin et al.

directly to hypothalamic sections.144 Thus, alter- regulation of the microbiota–gut–brain axis is now
ations to the level of E. coli in the gut microbiota emerging for a broad range of neurological and
are likely to have some bearing on satiety-related psychiatric conditions from Parkinson’s disease to
behaviors. In support of this, plasma levels of ClpB depression. Preclinical studies have been beneficial
were found to be higher in female patients with eat- in elucidating how microbiota dysregulation is
ing disorders (anorexia nervosa, bulimia nervosa, involved in such conditions. Moreover, preclinical
and binge-eating disorder),147 which suggests that studies have demonstrated the efficacy of modula-
dysregulation to the microbiota, observed as an tors of the microbiota (i.e., probiotics, prebiotics,
increased production of ClpB, may be facilitating and diet), supporting their use as potential thera-
such deleterious behaviors. pies for conditions such as depression and autism.
While a great deal of attention has been given to However, caution must be taken with extrapolating
understanding how the gut microbiota influences these preclinical results to the clinical setting, given
processes in the CNS, it is also important to con- reports of probiotic strains displaying efficacy
sider the effect that gut commensals have on the in mice but not in humans. Going forward, a
ENS. Proper functioning of the ENS is required for greater emphasis should be placed on assessing
physiological processes, such as gut motility, but also the therapeutic efficacy of diet or probiotics in
to ensure gut–brain communication. In the absence the clinical setting through conducting sufficiently
of a microbiota, germ-free mice display reduced powered, rigorous clinical trials.
excitability of after-hyperpolarization cells in the
Acknowledgments
myenteric plexus.148,149 A reduction in the neuronal
firing rates of these cells within the ENS of germ-free Timothy Dinan and John Cryan are supported by
mice is likely to affect proper signaling between the the Science Foundation Ireland (SFI) (Grant Num-
gut and the brain that may help to explain some of bers 07/CE/BI368 and 12/RC/2273); the Irish Health
the behavioral deficits observed in these animals.150 Research Board; the Department of Agriculture,
Moreover, bacterial strains that have demonstrated Food and the Marine; and Enterprise Ireland.
positive effects on behavior, such as L. reuteri and
L. rhamnosus JB-1, have also been shown to mod- Competing interests
ulate excitability of the ENS, which is likely to have Timothy Dinan and John Cryan are in receipt of
some bearing on their psychobiotic effects.151,152 research funding from 4D-Pharma, Mead Johnson,
The bacterial strain B. fragilis, which has demon- Suntory Wellness, Nutricia, and Cremo. Timothy
strated efficacy in improving autistic-related behav- Dinan has been an invited speaker at meetings
iors and ameliorating gastrointestinal permeability organized by Servier, Lundbeck, Janssen, and
in the MIA model of autism,103 has also been shown AstraZeneca. John Cryan has been an invited
to increase excitability of intestinal primary afferent speaker at meetings organized by Mead Johnsen,
neurons.153 This effect of B. fragilis was found to Alkermes, and Janssen. Eoin Sherwin declares no
be dependent on its expression of the exopolysac- competing interests.
charide polysaccharide A.153 The beneficial effects
of probiotics on behavior may depend on their abil-
ity to modulate neurotransmission within the ENS, References
and future work should consider the role of the ENS 1. Mackowiak, P.A. 2013. Recycling Metchnikoff: probiotics,
in this regard. the intestinal microbiome and the quest for long life. Front.
Public Health 1: 52.
Conclusions and future perspectives 2. Phillips, J.G.P. 1910. The treatment of melancholia by the
lactic acid Bacillus. Br. J. Psychiatry 56: 422–431.
Since the gut microbiota was first proposed to influ- 3. Bested, A.C., A.C. Logan & E.M. Selhub. 2013. Intestinal
ence human health over 100 years ago, our under- microbiota, probiotics and mental health: from Metch-
standing of such a role has increased immensely. nikoff to modern advances: part II—contemporary con-
Evidence now suggests that the microbiota, through textual research. Gut Pathog. 5: 3.
4. Sherwin, E., K.V. Sandhu, T.G. Dinan & J.F. Cryan. 2016.
the bidirectional communication axis known as the May the force be with you: the light and dark sides of the
microbiota–gut–brain axis, is involved in physio- microbiota–gut–brain axis in neuropsychiatry. CNS Drugs
logical processes, such as mood and aging. Dys- 30: 1019–1041.

20 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

5. Kelly, J.R., P.J. Kennedy, J.F. Cryan, T.G. Dinan, et al. 2015. 26. Giannelli, V., V. Di Gregorio, V. Iebba, et al. 2014. Micro-
Breaking down the barriers: the gut microbiome, intesti- biota and the gut–liver axis: bacterial translocation, inflam-
nal permeability and stress-related psychiatric. Front. Cell. mation and infection in cirrhosis. WorldJ. Gastroenterol. 20:
Neurosci. 9: 392. 16795–16810.
6. Forsythe, P. & W.A. Kunze. 2013. Voices from within: gut 27. Conley, M.N., C.P. Wong, K.M. Duyck, et al. 2016. Aging
microbes and the CNS. Cell. Mol. Life Sci. 70: 55–69. and serum MCP-1 are associated with gut microbiome
7. Sampson, T.R. & S.K. Mazmanian. 2015. Control of brain composition in a murine model. Peer J. 4: e1854.
development, function, and behavior by the microbiome. 28. Kim, K.-A., J.-J. Jeong, S.-Y. Yoo & D.-H. Kim. 2016. Gut
Cell Host Microbe 17: 565–576. microbiota lipopolysaccharide accelerates inflamm-aging
8. Sarkar, A., S.M. Lehto, S. Harty, et al. 2016. Psychobiotics in mice. BMC Microbiol. 16: 9.
and the manipulation of bacteria–gut–brain signals. Trends 29. Jeong, J.-J., K.A. Kim, Y.-J. Hwang, et al. 2016. Anti-
Neurosci. 39: 763–781 inflammaging effects of Lactobacillus brevis OW38 in aged
9. Bienenstock, J., W. Kunze & P. Forsythe. 2015. Microbiota mice. Benef. Microbes 7: 1–12.
and the gut–brain axis. Nutr. Rev. 73: 28–31. 30. Cho, S.-Y., J. Kim, J.H. Lee, et al. 2016. Modulation of gut
10. Foster, J.A., M. Lyte, E. Meyer & J.F. Cryan. 2016. Gut micro- microbiota and delayed immunosenescence as a result of
biota and brain function: an evolving field in neuroscience. syringaresinol consumption in middle-aged mice. Sci. Rep.
Int. J. Neuropsychopharmacol. 19: 1–7. 6: 39026.
11. Mayer, X.E.A., R. Knight, S.K. Mazmanian, et al. 2014. Gut 31. Poirier, A.-A., B. Aubé, M. Côté, et al. 2016. Gastroin-
microbes and the brain: paradigm shift in neuroscience. J. testinal dysfunctions in Parkinson’s disease: symptoms and
Neurosci. 34: 15490–15496. treatments. Parkinsons Dis. 2016: 6762528.
12. Mayer, E.A., K. Tillisch & A. Gupta. 2015. Gut/brain axis 32. Felice, V.D., E.M. Quigley, A.M. Sullivan, et al. 2016.
and the microbiota. J. Clin. Invest. 125: 49–53. Microbiota–gut–brain signalling in Parkinson’s disease:
13. Labus, J.S., E.B. Hollister, J. Jacobs, et al. 2017. Differences implications for non-motor symptoms. Parkinsonism Relat.
in gut microbial composition correlate with regional brain Disord. 27: 1–8.
volumes in irritable bowel syndrome. Microbiome 5: 1–17. 33. Forsyth, C.B., K.M. Shannon, J.H. Kordower, et al. 2011.
14. Sherwin, E., K. Rea, T.G. Dinan & J.F. Cryan. 2016. A gut Increased intestinal permeability correlates with sigmoid
(microbiome) feeling about the brain. Curr. Opin. Gas- mucosa alpha-synuclein staining and endotoxin expo-
troenterol. 32: 96–102. sure markers in early Parkinson’s disease. PLoS One 6:
15. Dinan, T.G. & J.F. Cryan. 2015. The impact of gut micro- e28032.
biota on brain and behaviour. Curr. Opin. Clin. Nutr. 34. Hilton, D., M. Stephens, L. Kirk, et al. 2014. Accumulation
Metab. Care 18: 552–558. of ␣-synuclein in the bowel of patients in the pre-clinical
16. Dinan, T.G., R.M. Stilling, C. Stanton & J.F. Cryan. 2015. phase of Parkinson’s disease. Acta Neuropathol. 127: 235–
Collective unconscious: how gut microbes shape human 241.
behavior. J. Psychiatr. Res. 63: 1–9. 35. Holmqvist, S., O. Chutna, L. Bousset, et al. 2014. Direct
17. O’Mahony, S.M., G. Clarke, Y.E. Borre, et al. 2015. evidence of Parkinson pathology spread from the gastroin-
Serotonin, tryptophan metabolism and the brain–gut– testinal tract to the brain in rats. Acta Neuropathol. 128:
microbiome axis. Behav. Brain Res. 277: 32–48. 805–820.
18. Burnet, P.W.J. & P.J. Cowen. 2013. Psychobiotics highlight 36. Scheperjans, F., V. Aho, P.A.B. Pereira, et al. 2015. Gut
the pathways to happiness. Biol. Psychiatry 74: 708–709. microbiota are related to Parkinson’s disease and clinical
19. Moos, W.H., D.V. Faller, D.N. Harpp, et al. 2016. Micro- phenotype. Mov. Disord. 30: 350–358.
biota and neurological disorders: a gut feeling. Biores. Open 37. Bedarf, J.R., F. Hildebrand, L.P. Coelho, et al. 2017. Func-
Access 5: 137–145. tional implications of microbial and viral gut metagenome
20. Severance, E.G., E. Prandovszky, J. Castiglione & R.H. changes in early stage l-DOPA-naı̈ve Parkinson’s disease
Yolken. 2015. Gastroenterology issues in schizophrenia: patients. Genome Med. 9: 1–13.
why the gut matters. Curr. Psychiatry Rep. 17: 27. 38. Keshavarzian, A., S.J. Green, P.A. Engen, et al. 2015. Colonic
21. O’Toole, P.W. & I.B. Jeffery. 2015. Gut microbiota and bacterial composition in Parkinson’s disease. Mov. Disord.
aging. Science 350: 1214–1216. 30: 1351–1360.
22. Kong, F., Y. Hua, B. Zeng, et al. 2016. Gut microbiota sig- 39. Hill-Burns, E.M., J.W. Debelius, J.T. Morton, et al. 2017.
natures of longevity. Curr. Biol. 26: R832–R833. Parkinson’s disease and Parkinson’s disease medications
23. Vaiserman, A.M., A.K. Koliada & F. Marotta. 2017. Gut have distinct signatures of the gut microbiome. Mov. Dis-
microbiota: a player in aging and a target for anti-aging ord. 32: 739–749.
intervention. Ageing Res. Rev. 35: 36–45. 40. Unger, M.M., J. Spiegel, K.-U. Dillmann, et al. 2016. Short
24. Prenderville, J.A., P.J. Kennedy, T.G. Dinan & J.F. Cryan. chain fatty acids and gut microbiota differ between patients
2015. Adding fuel to the fire: the impact of stress on the with Parkinson’s disease and age-matched controls. Parkin-
ageing brain. Trends Neurosci. 38: 13–25. sonism Relat. Disord. 32: 66–72.
25. Scott, K.A., M. Ida, V.L. Peterson, et al. 2017. Revis- 41. Huuskonen, J., T. Suuronen, T. Nuutinen, et al. 2004. Reg-
iting Metchnikoff: age-related alterations in microbiota- ulation of microglial inflammatory response by sodium
gut–brain axis in the mouse. Brain Behav. Immun. butyrate and short-chain fatty acids. Br. J. Pharmacol. 141:
https://doi.org/10.1016/j.bbi.2017.02.004. 874–880.

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 21
The gut microbiota in brain health and disease Sherwin et al.

42. Fasano, A., F. Bove, M. Gabrielli, et al. 2013. The role of 58. Escribano, B.M., F.J. Medina-fernández, M. Aguilar-luque
small intestinal bacterial overgrowth in Parkinson’s disease. & E. Agüera. 2016. Lipopolysaccharide binding protein and
Mov. Disord. 28: 1241–1249. oxidative stress in a multiple sclerosis model. Neurothera-
43. Çamcı, G. & S. Oğuz. 2016. Association between Parkin- peutics 14: 199–211.
son’s disease and Helicobacter pylori. J. Clin. Neurol. 12: 59. Miyake, S., S. Kim, W. Suda & K. Oshima. 2015. Dysbiosis
147–150. in the gut microbiota of patients with multiple sclerosis,
44. Cassani, E., M. Barichella, R. Cancello, et al. 2015. Increased with a striking depletion of species belonging to Clostridia
urinary indoxyl sulfate (indican): new insights into gut XIVa and IV clusters. PLoS One 10: e0137429.
dysbiosis in Parkinson’s disease. Parkinsonism Relat. Disord. 60. Hoban, A.E., R.M. Stilling, F.J. Ryan, et al. 2016. Regulation
21: 389–393. of prefrontal cortex myelination by the microbiota. Transl.
45. Dobbs, R.J., S.M. Dobbs, C. Weller, et al. 2008. Helicobac- Psychiatry 6: e774.
ter hypothesis for idiopathic parkinsonism: before and 61. Gacias, M., S. Gaspari, P.M.G. Santos, et al. 2016.
beyond. Helicobacter 13: 309–322. Microbiota-driven transcriptional changes in prefrontal
46. Hashim, H., S. Azmin, H. Razlan, et al. 2014. Eradica- cortex override genetic differences in social behavior. Elife
tion of Helicobacter pylori infection improves levodopa 5: 1–27.
action, clinical symptoms and quality of life in patients 62. Kyung, Y., J.S. Menezes, Y. Umesaki & S.K. Mazmanian.
with Parkinson’s disease. PLoS One 9: e112330. 2011. Proinflammatory T-cell responses to gut micro-
47. Sampson, T.R., J.W. Debelius, T. Thron, et al. 2016. Gut biota promote experimental autoimmune encephalomyeli-
microbiota regulate motor deficits and neuroinflammation tis. Proc. Natl. Acad. Sci. U.S.A. 108: 4615–4622.
in a model of Parkinson’s disease. Cell 167: 1469–1480. 63. Miller, P.G., M.B. Bonn, C.L. Franklin, et al. 2017. TNFR2
e12. deficiency acts in concert with gut microbiota to precipitate
48. Erny, D., A.L. Hrabě de Angelis, D. Jaitin, et al. 2015. spontaneous sex-biased central nervous system demyeli-
Host microbiota constantly control maturation and func- nating autoimmune disease. J. Immunol. 195: 4668–
tion of microglia in the CNS. Nat. Neurosci. 18: 965– 4684.
977. 64. Stanisavljevic, S., J. Lukic, S. Sokivic, et al. 2016. Corre-
49. Cattaneo, A., N. Cattane, S. Galluzzi, et al. 2017. Association lation of gut microbiota composition with resistance to
of brain amyloidosis with pro-inflammatory gut bacterial experimental autoimmune encephalomyelitis in rats. Front.
taxa and peripheral inflammation markers in cognitively Microbiol. 7: 1–12.
impaired elderly. Neurobiol. Aging 49: 60–68. 65. Yin, J., S.-X. Liao, Y. He, S. Wang, et al. 2015. Dys-
50. Zhan, X., B. Stamova, L.W. Jin, et al . 2016. Gram- biosis of gut microbiota with reduced trimethylamine-
negative bacterial molecules associate with Alzheimer dis- n-oxide level in patients with large-artery atherosclerotic
ease pathology. Neurology 87: 2324–2332. stroke or transient ischemic attack. J. Am. Heart Assoc. 4:
51. Friedland, R.P. 2015. Mechanisms of molecular mimicry 1–13.
involving the microbiota in neurodegeneration. J. 66. Yamashiro, K., R. Tanaka, T. Urabe, et al. 2017. Gut dysbiosis
Alzheimers Dis. 45: 349–362. is associated with metabolism and systemic inflammation
52. Harach, T., N. Marungruang, N. Duthilleul, et al. 2017. in patients with ischemic stroke. PLoS One 12: e0171521.
Reduction of Abeta amyloid pathology in APPPS1 trans- 67. Winek, K., O. Engel, P. Koduah, et al. 2016. Depletion of
genic mice in the absence of gut microbiota. Sci. Rep. 7: cultivatable gut microbiota by broad-spectrum antibiotic
41802. pretreatment worsens outcome after murine stroke. Stroke
53. Minter, M.R., C. Zhang, V. Leone, et al. 2016. Antibiotic- 47: 1354–1363.
induced perturbations in gut microbial diversity influences 68. Singh, V., S. Roth, G. Llovera, et al. 2016. Microbiota dysbio-
neuro-inflammation and amyloidosis in a murine model sis controls the neuroinflammatory response after stroke.
of Alzheimer’s disease. Sci. Rep. 6: 30028. J. Neurosci. 36: 7428–7440.
54. Brandscheid, C., F. Schuck, S. Reinhardt, et al. 2017. Altered 69. Stanley, D., L.J. Mason, K.E. Mackin, et al. 2016. Translo-
gut microbiome composition and tryptic activity of the cation and dissemination of commensal bacteria in post-
5xFAD Alzheimer’s mouse model. J. Alzheimers Dis. 56: stroke infection. Nat. Med. 22: 1277–1284.
775–788. 70. Risk, T., W. Zhu, J.C. Gregory, et al. 2016. Gut microbial
55. Wang, D., L. Ho, J. Faith, et al. 2015. Role of intesti- metabolite TMAO enhances platelet article gut microbial
nal microbiota in the generation of polyphenol-derived metabolite TMAO enhances platelet hyperreactivity and
phenolic acid mediated attenuation of Alzheimer’s dis- thrombosis risk. Cell 165: 111–124.
ease beta-amyloid oligomerization. Mol. Nutr. Food Res. 71. Wang, Z., A.B. Roberts, J.A. Buffa, et al. 2015. Non-lethal
59: 1025–1040. inhibition of gut microbial trimethylamine production
56. Distrutti, E., J.-A. O’Reilly, C. McDonald, et al. 2014. Mod- for the treatment of article non-lethal inhibition of gut
ulation of intestinal microbiota by the probiotic VSL#3 microbial trimethylamine production for the treatment of
resets brain gene expression and ameliorates the age-related atherosclerosis. Cell 163: 1585–1595.
deficit in LTP. PLoS One 9: e106503. 72. Benakis, C., D. Brea, S. Caballero, et al. 2016. Commensal
57. Buscarinu, M.C., B. Cerasoli, V. Annibali, et al. 2017. microbiota affects ischemic stroke outcome by regulating
Altered intestinal permeability in patients with relapsing– intestinal ␥ ␦ T cells. Nat. Med. 22: 516–523.
remitting multiple sclerosis: a pilot study. Mult. Scler. 23: 73. Sadler, R., V. Singh, C. Benakis, et al. 2017. Micro-
442–446. biota differences between commercial breeders impacts

22 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

the post-stroke immune response. Brain Behav. Immun. central GABA receptor expression in a mouse via the vagus
https://doi.org/10.1016/j.bbi.2017.03.011. nerve. Proc. Natl. Acad. Sci. U.S.A. 108: 16050–16055.
74. Houlden, A., M. Goldrick, D. Brough, et al. 2016. Brain 89. Kelly, J.R., A.P. Allen, A. Temko, et al. 2017. Lost in trans-
injury induces specific changes in the caecal microbiota lation? The potential psychobiotic Lactobacillus rhamnosus
of mice via altered autonomic activity and mucoprotein (JB-1) fails to modulate stress or cognitive performance in
production. Brain Behav. Immun. 57: 10–20. healthy male subjects. Brain Behav. Immun. 61: 50–59.
75. Jentsch, H.F.R., D. März & M. Krüger. 2013. The effects of 90. Allen, A.P., W. Hutch, Y.E. Borre, et al. 2016. Bifidobac-
stress hormones on growth of selected periodontitis related terium longum 1714 as a translational psychobiotic: mod-
bacteria. Anaerobe 24: 49–54. ulation of stress, electrophysiology and neurocognition in
76. Kigerl, K.A., J.C.E. Hall, L. Wang, et al. 2016. Gut dysbiosis healthy volunteers. Transl. Psychiatry 6: e939.
impairs recovery after spinal cord injury. J. Exp. Med. 213: 91. Tillisch, K., J. Labus, L. Kilpatrick, et al. 2013. Consumption
2603–2620. of fermented milk product with probiotic modulates brain
77. Sun, J., F. Wang, Z. Ling, et al. 2016. Clostridium butyricum activity. Gastroenterology 144: 1394–1401, 1401.e1-4.
attenuates cerebral ischemia/reperfusion injury in diabetic 92. Pinto-sanchez, M.I., G.B. Hall, K. Ghajar, et al. 2017. Pro-
mice via modulation of gut microbiota. Brain Res. 1642: biotic Bifidobacterium longum NCC3001 reduces depres-
180–188. sion scores and alters brain activity: a pilot study in
78. Zhou, L. & J.A. Foster. 2015. Psychobiotics and the gut– patients with irritable bowel syndrome. Gastroenterology
brain axis: in the pursuit of happiness. Neuropsychiatr. Dis. https://doi.org/10.1053/j.gastro.2017.05.003.
Treat. 11: 715–723. 93. Xu, M., H. Cao, W. Wang, et al. 2015. Fecal microbiota
79. Yu, M., H. Jia, C. Zhou, et al. 2017. Variations in gut transplantation broadening its application beyond intesti-
microbiota and fecal metabolic phenotype associated with nal disorders. World J. Gastroenterol. 21: 102–111.
depression by 16S rRNA gene sequencing and LC/MS- 94. Evrensel, A. & M.E. Ceylan. 2016. Fecal microbiota trans-
based metabolomics. J. Pharm. Biomed. Anal. 138: 231– plantation and its usage in neuropsychiatric disorders. Clin.
239. Psychopharmacol. Neurosci. 14: 231–237.
80. Kelly, J.R., Y. Borre, C. O’ Brien, et al. 2016. Transferring the 95. Holzer, P. & A. Farzi. 2014. Neuropeptides and the
blues: depression-associated gut microbiota induces neu- microbiota–gut–brain axis. Adv. Exp. Med. Biol. 817: 39–71.
robehavioural changes in the rat. J. Psychiatr Res. 82: 109– 96. Holzer, P. 2016. Neuropeptides, microbiota, and behavior.
118. Int. Rev. Neurobiol. 131: 67–89.
81. Zheng, P., B. Zeng, C. Zhou, et al. 2016. Gut microbiome 97. Buffington, S.A., G.V. Di Prisco, T.A. Auchtung, et al. 2016.
remodeling induces depressive-like behaviors through a Microbial reconstitution reverses maternal diet-induced
pathway mediated by the host’s metabolism. Mol. Psychia- social and synaptic deficits in offspring. Cell 165: 1762–
try 21:786–796. 1775.
82. De Palma, G., M.D.J. Lynch, J. Lu, et al. 2017. Transplanta- 98. Desbonnet, L., G. Clarke, F. Shanahan, et al. 2014. Micro-
tion of fecal microbiota from patients with irritable bowel biota is essential for social development in the mouse. Mol.
syndrome alters gut function and behavior in recipient Psychiatry 19: 146–148.
mice. Sci. Transl. Med. 6397: 1–15. 99. Desbonnet, L., G. Clarke, A. Traplin, et al. 2015. Gut micro-
83. Burokas, A., S. Arboleya, D. Rachel, et al. 2017. Target- biota depletion from early adolescence in mice: implica-
ing the microbiota–gut–brain axis: prebiotics have anx- tions for brain and behaviour. Brain Behav. Immun. 48:
iolytic and antidepressant-like effects and reverse the 165–173.
impact of chronic stress in mice. Biol. Psychiatry. https:// 100. Arentsen, T., H. Raith, Y. Qian, et al. 2015. Host micro-
doi.org/10.1016/j.biopsych.2016.12.031. biota modulates development of social preference in mice.
84. Savignac, H.M., Y. Couch, M. Stratford, et al. 2016. Prebi- Microb. Ecol. Health Dis. 26: 29719.
otic administration normalizes lipopolysaccharide (LPS)- 101. de Theije, C.G.M., H. Wopereis, M. Ramadan, et al. 2014.
induced anxiety and cortical 5-HT2A receptor and IL1 lev- Altered gut microbiota and activity in a murine model of
els in male mice. Brain Behav. Immun. 52: 120–131. autism spectrum disorders. Brain Behav. Immun. 37: 197–
85. Tarr, A.J., J.D. Galley, S.E. Fisher, et al. 2015. The prebi- 206.
otics 3 sialyllactose and 6 sialyllactose diminish stressor- 102. de Theije, C.G., P.J. Koelink, G.A. Korte-Bouws, et al. 2014.
induced anxiety-like behavior and colonic microbiota Intestinal inflammation in a murine model of autism spec-
alterations: evidence for effects on the gut–brain axis. Brain trum disorders. Brain Behav. Immun. 37: 240–247.
Behav. Immun. 50: 166–177. 103. Hsiao, E.Y., S.W. McBride, S. Hsien, et al. 2013. Microbiota
86. Schmidt, K., P.J. Cowen, C.J. Harmer, et al. 2015. Prebi- modulate behavioral and physiological abnormalities asso-
otic intake reduces the waking cortisol response and alters ciated with neurodevelopmental disorders. Cell 155: 1451–
emotional bias in healthy volunteers. Psychopharmacology 1463.
(Berl.) 232: 1793–1801. 104. McFarlane, H.G., G.K. Kusek, M. Yang, et al. 2008. Autism-
87. Bharwani, A., M.F. Mian, M.G. Surette, et al. 2017. like behavioral phenotypes in BTBR T+tf/J mice. Genes
Oral treatment with Lactobacillus rhamnosus attenuates Brain Behav. 7: 152–163.
behavioural deficits and immune changes in chronic social 105. Kazdoba, T.M., R.J. Hagerman, D. Zolkowska, et al. 2015.
stress. BMC Med. 15: 7. Evaluation of the neuroactive steroid ganaxolone on social
88. Bravo, J.A., P. Forsythe, M.V. Chew, et al. 2011. Ingestion and repetitive behaviors in the BTBR mouse model of
of Lactobacillus strain regulates emotional behavior and autism. Psychopharmacology (Berl.) 233: 309–323.

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 23
The gut microbiota in brain health and disease Sherwin et al.

106. Kratsman, N., D. Getselter & E. Elliott. 2016. Sodium and gut microbiota in mice treated chronically with ␦9-
butyrate attenuates social behavior deficits and modifies the tetrahydrocannabinol. PLoS One 10: e0144270.
transcription of inhibitory/excitatory genes in the frontal 122. Plovier, H., A. Everard, C. Druart, et al. 2017. A purified
cortex of an autism model. Neuropharmacology 102: 136– membrane protein from Akkermansia muciniphila or the
145. pasteurized bacterium improves metabolism in obese and
107. Newell, C., M.R. Bomhof, R.A. Reimer, et al. 2016. Keto- diabetic mice. Nat. Med. 23: 107–113.
genic diet modifies the gut microbiota in a murine model 123. Sonnenburg, E.D., S.A. Smits, M. Tikhonov, et al. 2016.
of autism spectrum disorder. Mol. Autism 7: 37. Diet-induced extinctions in the gut microbiota compound
108. Kang, D.-W., J.B. Adams, A.C. Gregory, et al. 2017. Micro- over generations. Nature 529: 212–215.
biota transfer therapy alters gut ecosystem and improves 124. Turroni, S., J. Fiori, S. Rampelli, et al. 2016. Fecal
gastrointestinal and autism symptoms: an open-label metabolome of the Hadza hunter-gatherers: a host–
study. Microbiome 5: 10. microbiome integrative view. Sci. Rep. 6: 32826.
109. De La Torre-Ubieta, L., H. Won, J.L. Stein & D.H. 125. Power, S.E., I.B. Jeffery, R.P. Ross, et al. 2014. Food and
Geschwind. 2016. Advancing the understanding of autism nutrient intake of Irish community-dwelling elderly sub-
disease mechanisms through genetics. Nat. Med. 22: 345– jects: who is at nutritional risk? J. Nutr. Health Aging 18:
361. 561–572.
110. Gilbert, J.A., R. Krajmalnik-Brown, D.L. Porazinska, et al. 126. Sandhu K.V., E. Sherwin, H. Schellekens, et al. 2016. Feed-
2013. Toward effective probiotics for autism and other neu- ing the microbiota–gut–brain axis: diet, microbiome and
rodevelopmental disorders. Cell 155: 1446–1448. neuropsychiatry. Transl. Res. 179: 223–244.
111. Varian, B.J., T. Poutahidis, B.T. DiBenedictis, et al. 2016. 127. Robertson, R.C., C. Seira Oriach, K. Murphy, et al. 2016.
Microbial lysate upregulates host oxytocin. Brain Behav. Omega-3 polyunsaturated fatty acids critically regulate
Immun. 61: 36–49. behaviour and gut microbiota development in adolescence
112. Spanogiannopoulos, P., E.N. Bess, R.N. Carmody & P.J. and adulthood. Brain Behav. Immun. 59: 21–37
Turnbaugh. 2016. The microbial pharmacists within us: 128. Pusceddu, M.M., P. Kelly, N. Ariffin, et al. 2015. n-3 PUFAs
a metagenomic view of xenobiotic metabolism. Nat. Rev. have beneficial effects on anxiety and cognition in female
Microbiol. 14: 273–287. rats: effects of early life stress. Psychoneuroendocrinology 58:
113. Davey, K.J., P.D. Cotter, O. O’Sullivan, et al. 2013. Antipsy- 79–90.
chotics and the gut microbiome: olanzapine-induced 129. Jacka, F.N., A. O’Neil, C. Itsiopoulos, et al. 2016. A ran-
metabolic dysfunction is attenuated by antibiotic adminis- domised, controlled trial of a dietary intervention for adults
tration in the rat. Transl. Psychiatry 3: e309. with major depression (the “SMILES” trial): results. In Pro-
114. Llopis, M., A.M. Cassard, L. Wrzosek, et al. 2016. Intesti- ceedings of the 18th Annual Conference of the International
nal microbiota contributes to individual susceptibility to Society for Bipolar Disorders held jointly with the 8th Biennial
alcoholic liver disease. Gut 65: 830–839. Conference of the International Society For Affective Disor-
115. Peterson, V.L., N.J. Jury, R. Cabrera-Rubio, et al. 2017. ders. 1–13. Amsterdam.
Drunk bugs: chronic vapour alcohol exposure induces 130. Ruskin, D.N., J. Svedova, J.L. Cote, et al. 2013. Ketogenic
marked changes in the gut microbiome in mice. Behav. diet improves core symptoms of autism in BTBR mice.
Brain Res. 323: 172–176. PLoS One 8: 4–9.
116. Tsuruya, A., A. Kuwahara, Y. Saito, et al. 2016. Eco- 131. Ruskin, D.N., M.I. Murphy, S.L. Slade & S.A. Masino. 2017.
physiological consequences of alcoholism on human gut Ketogenic diet improves behaviors in a maternal immune
microbiota: implications for ethanol-related pathogenesis activation model of autism spectrum disorder. PLos One
of colon cancer. Sci. Rep. 6: 27923. 12: e0171643.
117. Leclercq, S., S. Matamoros, P.D. Cani, et al. 2014. Intesti- 132. Castro, K., D. Baronio, I.S. Perry, et al. 2016. The effect of
nal permeability, gut–bacterial dysbiosis, and behavioral ketogenic diet in an animal model of autism induced by
markers of alcohol-dependence severity. Proc. Natl. Acad. prenatal exposure to valproic acid. Nutr. Neurosci. 9: 1–8.
Sci. U.S.A. 111: E4485–E4493. 133. Wu, J., C.G.M. de Theije, S.L. da Silva, et al. 2016. Dietary
118. Sokol, H., B. Pigneur, L. Watterlot, et al. 2008. Faecal- interventions that reduce mTOR activity rescue autistic-
ibacterium prausnitzii is an anti-inflammatory commensal like behavioral deficits in mice. Brain Behav. Immun. 59:
bacterium identified by gut microbiota analysis of Crohn 273–287.
disease patients. Proc. Natl. Acad. Sci. U.S.A. 105: 16731– 134. Ghalichi, F., J. Ghaemmaghami, A. Malek & A.
16736. Ostadrahimi. 2016. Effect of gluten free diet on gastroin-
119. Devkota, S., Y. Wang, M.W. Musch, et al. 2012. Dietary- testinal and behavioral indices for children with autism
fat-induced taurocholic acid promotes pathobiont expan- spectrum disorders: a randomized clinical trial. World J.
sion and colitis in Il10–/– mice. Nature 487: 104– Pediatr. 12: 436–442.
108. 135. Scott, K.P., J.-M. Antoine, T. Midtvedt & S. van Hemert.
120. Kiraly, D.D., D.M. Walker, E.S. Calipari, et al. 2016. Alter- 2015. Manipulating the gut microbiota to maintain health
ations of the host microbiome affect behavioral responses and treat disease. Microb. Ecol. Health Dis. 26: 25877.
to cocaine. Sci. Rep. 6: 35455. 136. Arentsen, T., Y. Qian, S. Gkotzis, et al. 2017. The bacterial
121. Cluny, N.L., C.M. Keenan, R.A. Reimer, et al. 2015. Pre- peptidoglycan-sensing molecule Pglyrp2 modulates brain
vention of diet-induced obesity effects on body weight development and behavior. Mol. Psychiatry 22: 257–266.

24 Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences.
Sherwin et al. The gut microbiota in brain health and disease

137. D’Mello, C., N. Ronaghan, R. Zaheer, et al. 2015. 146. Panaro, B.L., I.R. Tough, M.S. Engelstoft, et al. 2014. The
Probiotics improve inflammation-associated sickness melanocortin-4 receptor is expressed in enteroendocrine l
behavior by altering communication between the periph- cells and regulates the release of peptide YY and glucagon-
eral immune system and the brain. J. Neurosci. 35: 10821– like peptide 1 in vivo. Cell Metab. 20: 1018–1029.
10830. 147. Breton, J., R. Legrand, K. Akkermann, et al. 2016. Elevated
138. Bercik, P., A.J. Park, D. Sinclair, et al. 2011. The anxiolytic plasma concentrations of bacterial ClpB protein in patients
effect of Bifidobacterium longum NCC3001 involves vagal with eating disorders. Int. J. Eat Disord. 49: 805–808.
pathways for gut–brain communication. Neurogastroen- 148. Mcvey Neufeld, K.A., Y.K. Mao, J. Bienenstock, et al. 2013.
terol. Motil. 23: 1132–1139. The microbiome is essential for normal gut intrinsic pri-
139. Poutahidis, T., S.M. Kearney, T. Levkovich, et al. 2013. mary afferent neuron excitability in the mouse. Neurogas-
Microbial symbionts accelerate wound healing via the neu- troenterol. Motil. 25: 183–190.
ropeptide hormone oxytocin. PLoS One 8: e78898. 149. Mcvey Neufeld, K.A., A. Perez-Burgos, Y.K. Mao, et al.
140. Lyte, M. & J.F. Cryan, Eds. 2014. Microbial Endocrinol- 2015. The gut microbiome restores intrinsic and extrinsic
ogy: The Microbiota–Gut–Brain Axis in Health and Disease. nerve function in germ-free mice accompanied by changes
Springer. in calbindin. Neurogastroenterol. Motil. 27: 627–636.
141. Lyte, M. 2016. Microbial endocrinology in the pathogenesis 150. Luczynski, P., K.A.M.V. Neufeld, C.S. Oriach, et al. 2016.
of infectious disease. Microbiol. Spectr. 4. Growing up in a bubble: using germ-free animals to assess
142. Yano, J.M., K. Yu, G.P. Donaldson, et al. 2015. Indigenous the influence of the gut microbiota on brain and behavior.
bacteria from the gut microbiota regulate host serotonin Int. J. Neuropsychopharmacol. 19: 1–17.
biosynthesis. Cell 161: 264–276. 151. Wang, B., Y.K. Mao, C. Diorio, et al. 2009. Lactobacillus
143. Clarke, G., S. Grenham, P. Scully, et al. 2013. The reuteri ingestion and IKCa channel blockade have similar
microbiome–gut–brain axis during early life regulates the effects on rat colon motility and myenteric neurones. Neu-
hippocampal serotonergic system in a sex-dependent man- rogastroenterol. Motil. 22: 98–e33.
ner. Mol. Psychiatry 18: 666–673. 152. Wu, R.Y., M. Pasyk, B. Wang, et al. 2013. Spatiotempo-
144. Breton, J., N. Tennoune, N. Lucas, et al. 2016. Gut commen- ral maps reveal regional differences in the effects on gut
sal E. coli proteins activate host satiety pathways following motility for Lactobacillus reuteri and rhamnosus strains.
nutrient-induced bacterial growth. Cell Metab. 23: 1–11. Neurogastroenterol. Motil. 25: e205–1.
145. Tennoune, N., P. Chan, J. Breton, et al. 2014. Bacterial ClpB 153. Mao, Y.-K., D.L. Kasper, B. Wang, et al. 2013. Bacteroides
heat-shock protein, an antigen-mimetic of the anorexigenic fragilis polysaccharide A is necessary and sufficient for acute
peptide ␣-MSH, at the origin of eating disorders. Transl. activation of intestinal sensory neurons. Nat. Commun. 4:
Psychiatry 4: e458. 1465.

Ann. N.Y. Acad. Sci. 1420 (2018) 5–25 


C 2017 New York Academy of Sciences. 25

Das könnte Ihnen auch gefallen