Dalton (2019) Exercise & Gut-Brain Axis
Dalton (2019) Exercise & Gut-Brain Axis
Dalton (2019) Exercise & Gut-Brain Axis
REVIEW
CONTACT Micah Zuhl [email protected] Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM, USA
© 2019 Taylor & Francis Group, LLC
556 A. DALTON ET AL.
The gut has long been overlooked when it comes exercise may have the potential to reverse the con-
to human health and disease prevention. Emerging ditions associated with obesity, metabolic diseases,
research has proved that an imbalanced diet of poor diet, along with neural and behavioral
highly saturated fats, high sugar, and low fiber disorders.42-45 To date, the impact of exercise on
intake has a large influence on the composition of the relationship between the gut and the brain is
the microbiome.8 The alterations induced by poor unknown. Therefore, the purposes of this review
dietary habits contribute to gastrointestinal are to briefly introduce the complex interaction
(GI) dysfunction that may further lead to the devel- between the microbiome, gut, and brain and to
opment of inflammatory diseases.25 Interestingly, further explain how exercise impacts these rela-
a prebiotic intervention that promoted changes in tionships. For this review, we have chosen to
the microbiota of diabetic rats lowered inflamma- state this relationship as the microbiome–gut–
tion while improving glucose intolerance.26,27 brain axis where the composition of the gut micro-
Moreover, individuals diagnosed with inflamma- biota influences both the GI and central nervous
tory conditions such as IBS and obesity have been systems (CNSs).34,46 More specifically, alterations
found to have a comorbidity of lower cognitive to the microbiome may impact (both positively
function28,29 and higher instances of stress-related and negatively) GI (e.g., secretion, motility,
psychiatric symptoms, such as anxiety.30-32 In and integrity) and higher brain function (e.g.,
accordance, interventions that specifically treat neurotransmission, neurogenesis, and behavior),
neural disorders such as selective serotonin uptake and these influences may be bidirectional.34
inhibitors (SSRIs) have been shown to improve GI
function.33 These data have sparked curiosity
among investigators, which has led to a growth of Microbiome–gut–brain axis pathways of
communication
research into understanding the connection
between the GI tract and the brain.7,9,11,30,34 The relationship between the gut and the brain
Further interest has grown in discovering the role begins in utero as the CNS and the enteric nervous
of the microbiome in mediating the connection system (ENS) are derived from the same tissues
between the gut and brain. Altering the gut micro- during fetal development.7 The communication
biome through probiotic supplementation has between the gut and the brain is a bidirectional path-
improved symptoms of both psychological disor- way that is mediated through the autonomic ner-
ders (e.g., depression and anxiety) and cognition vous system (ANS) efferent and afferent signals via
and positively supports GI function.35-39 This the vagus nerve; neuroendocrine signaling through
demonstrates the complexity of the relationship the hypothalamic–pituitary–adrenal axis (HPA
between the gut microbiome and the brain. axis), and serotonin (5-HT) regulation.47-50 It has
Regular aerobic exercise has been shown to now been established that alterations of gut micro-
prevent age-related global brain atrophy and biome enact some influence on the communication
increases brain volume in the frontal lobes and between the gut and brain through these pathways.
left superior temporal lobe, which are important The vagus nerve serves a critical role in commu-
for cognition and control of attention and nication between the gut microbes and the brain, as it
memory.40 Moderate-intensity aerobic exercise connects the CNS to the ENS.51,52 The vagus nerve is
training has also promoted improvement (in able to communicate with gut microbiota in the ENS
older adults aged 60–79 years) in functional acti- and transfer vital information to the CNS where it is
vation in the brain which allows for increased deciphered, to then generate a response based on the
efficiency when completing tasks, as well as regu- information received. For example, if the information
lating behavior and mood.41 More recently, aero- received via the vagus nerve from the gut indicates an
bic exercise has been shown to impact the gut by imbalance in gut microbiome bacteria, the CNS will
increasing microbiome diversity and functional then decipher if an inflammatory response is neces-
metabolism in both humans and mice. Altering sary. Decreased vagus nerve activity has been asso-
the bacterial profiles and influencing the by- ciated with conditions such as IBD,53 IBS,54 and
products produced from gut bacteria through depression55-57 and is attributed to dysfunction of
GUT MICROBES 557
gastric motility and gastric emptying.58 Specifically the gut microbiome is important in the adequate
targeted probiotic supplements that alter the micro- development of both the HPA axis and the CNS
biome have also been shown to improve brain func- during early life.64,69 While extremely complex,
tion, and this positive neural benefit is abolished when when a diverse and healthy microbiome communi-
a vagotomy procedure is performed among rodents.59 cates with the CNS, tighter HPA control occurs,
More specifically, mice induced with colitis demon- which may further promote both neural and GI
strated lower anxiety-like behavior after 21 days of growth through BDNF regulation.70,71
being fed a probiotic supplement containing Serotonin (5-HT) is a crucial neurotransmitter and
Bifidobacterium longum. The anxiolytic effect of hormone and is often known as one of the primary
Bifidobacterium longum was lost when vagotomized mood and cognition regulators.72 Lower platelet ser-
mice were fed the supplement. The authors concluded otonin receptor function is associated with higher
that the behavior benefits of the Bifidobacterium levels of anxiety and depression, while higher platelet
longum is vagally mediated.59 This provides evidence serotonin is associated with improved mood.73 It is
that vagal communication between the gut and brain also a major contributor to the modulation of intest-
is influenced by the release of molecules from the gut inal secretion and motility,74,75 as well as a key signal-
microbiota. ing molecule within the microbiome–gut–brain
The HPA axis regulates an organism’s response to axis.30,65 Recently, it has been demonstrated that indi-
a multitude of stressors (e.g., physical or mental).60,61 genous bacteria in the gut regulate serotonin synthesis
The HPA is influenced by the GI system through and release.76 In addition, germ-free animals have
a complex neural-immunoregulatory mechanism. demonstrated higher levels of serotonin in the hippo-
Afferent feedback from the gut via the vagus nerve campal region of the brain while showing lower
acts on the hypothalamus and hippocampus regions expression in the colon.76,77 This demonstrates that
of the brain, resulting in activation of the HPA.62 It the microbiome plays a role in the regulation of
also appears that disruption to the gut mucosal layer serotonin and may influence both brain and gut func-
via lipopolysaccharide (LPS) insult promotes the tion. It appears that various gut microbe strains
release of pro-inflammatory cytokines, which are synthesize serotonin and may be the reason why
capable of exaggerating HPA activation.47,63 HPA germ-free rodents have lower expression of 5-HT in
axis hyperresponsiveness and disturbances in the the gut. Other strains control tryptophan metabolism,
gut microbiome are found in those suffering from the precursor to 5-HT synthesis, and have been
both IBS and psychological disorders.61 This demon- shown to influence both tryptophan and serotonin
strates potential roles for the neuroendocrine system levels.78,79 For example, the Bacteroides fragilis bacter-
and the gut microbiome in the regulation of both the ium is a known consumer of tryptophan, and supple-
gut and brain. Rodents raised in a germ-free envir- mentation has been linked to improving intestinal
onment (no-microorganism exposure and no poten- wall stability, and also protection against CNS demye-
tial for gut microbiota colonization) from birth are linating disease.65,80,81 Autism spectrum disorder is
found to have an inflated release of adrenocortico- a developmental disorder that affects behavior and is
tropic hormone (ACTH) and corticosterone levels in commonly associated with GI symptoms. Autistic like
response to physical restraint stress exposure when mice demonstrated improved GI function and also
compared to free-living control rodents.6,34,48,64-66 began to resemble normal behaving animals after
The germ-free rodents also displayed lower expres- supplementation with Bacteroides fragilis.81
sion of brain-derived neurotrophic factor (BDNF) in In summary, the microbiome is capable of influen-
the hippocampal and cortex regions of the brain. cing both brain and gut function by modulating vagus
BDNF promotes neurogenesis and is vital for CNS nerve afferent feedback; influencing the hyperrespon-
growth and health.67 BDNF has also been shown to siveness of the HPA axis; and altering the regulation
be an important regulator of GI tight junction pro- of the tryptophan and serotonin synthesis pathways.
tein expression and regulation.68 Treatment with However, it is important to understand that other
Bifidobacterium ameliorated the exaggerated HPA communication channels in the gut–brain axis such
response in the germ-free rodents and restored as GABA-glutamate and catecholamines have been
BDNF levels in the brain.64 It has become clear that shown to be influenced by specific microbiota.82,83
558 A. DALTON ET AL.
Exercise effect on the microbiota–gut–brain of microbes that contribute to the biodiversity of the
axis bacteria in the gut.42-44,66 Exercise appears to be
a potential external influence on the capacity to
The bulk of previous exercise research on GI phy- alter the gut biodiversity in both quantitative and
siology has shown a negative effect. For example, in qualitative ways.6 Exercise alterations to the gut
several studies from our group and others, intense microbiome may provide a link to the exercise-
exercise leads to an increase in intestinal permeabil- related benefits on GI function, mood, and higher
ity, GI damage, and mild endotoxemia.84-86 brain centers. Figure 1 highlights the impact of
However, targeted exercise therapies have been exercise on the microbiome, and how alterations
developed to treat those suffering from IBS with may mediate improvements in both the gut and
reported improvements in symptoms of the brain.
disorder.87 Exercise also promotes cognition and Research conducted among volunteers from the
improved symptoms of both mood and psychologi- American Gut Project95 concluded that those who
cal disorders among humans. Cognitive improve- exercised at a higher frequency demonstrated
ments have been observed among stroke patients, diversity in the Firmcutes phylum (specifically,
while symptoms of depression and schizophrenia Faecalibacterium prausnitzii and species from the
have also been improved after aerobic exercise genus Oscillospira, Lachnospira, and Coprococcus).
training.88-90 Interestingly, the improvements Athletes have been shown to exhibit higher diver-
observed among IBS patients after exercise were sity of gut microorganisms compared to nonath-
linked to changes in mental health and emotion.87 letes. More specifically, in a group of professional
Evidence suggests that the bacterial diversity of the rugby players, 22 phyla were detected, while only
gut contributes to the disordered state of GI disor- 11 phyla were identified in age-matched low BMI
ders by negatively impacting GI function and the control and 9 phyla in an age-matched high BMI
psychological state.91,92 Altering the microbiome control. The distinct phyla among the athletes
through probiotic supplements improves both GI correlated with both dietary protein intake and
integrity and mood among IBS patients.92,93 creatine kinase, suggesting that diet and exercise
Probiotic supplements also reduced stress-induced facilitate diversity in the gut. The researchers iden-
GI symptoms (abdominal pain and nausea) among tified higher proportions of the Firmicutes phyla
participants with a history of elevated stress.94 and lower Bacteroidetes phyla in the athlete group
Exciting research within the last 5 years has demon- compared to the high BMI control.42 While the
strated that exercise promotes a diverse community athlete group had greater diversity than the low
Figure 1. The role of exercise in the microbiome–gut–brain axis. Exercise and the gut microbiome have been independently shown
to improve symptoms of IBS and stabilize the TJ barrier and have also been linked to decrease psychological disorders (e.g.
depression, anxiety), promote neurogenesis (through BDNF), and improve HPA axis control. It has been demonstrated that aerobic
exercise caused greater diversity in the microbiome along with increasing genera of the Firmicutes phylum, which produce short-
chain fatty acids. In addition, athletes have demonstrated higher levels of the Akkermansia genus, which has been linked to
metabolic and neural diseases. The extent to which the effect of exercise on the gut and brain is mediated through alterations in the
microbiome is unknown. The dotted line represents a proposed mechanism. Solid lines represent known mechanisms. BDNF – brain-
derived neurotropic factor, SCFA – short-chain fatty acids, ZO-1 – zona occluden-1 protein, HPA axis – hypothalamic–pituitary–
adrenal axis, TJ – tight junction.
GUT MICROBES 559
BMI control, it was difficult for the researchers to aerobic fitness (VO2max) and fecal SCFAs, indi-
identify specific bacteria that help to understand cating that more aerobically fit humans produce
the role of extreme exercise on the microbiome in higher rates of SFCAs.101 Total dietary protein
this study. Interestingly, both the athletes and low intake was also a significant contributor to the
BMI group demonstrated a higher proportion of diversity and functional changes (SFCA produc-
the Akkermansia genus (Verrucomicrobia phy- tion) in the gut, further highlighting the role of
lum), which has been negatively associated with diet. Higher SFCA production is attributed to
metabolic dysfunction in humans and rodents.42 many health benefits, including protection against
The administration of Akkermansia has been inflammation and atherosclerosis.102 SCFAs pro-
shown to reversed diet-induced metabolic disor- duced from the microbiome also appear to provide
ders in obese mice.96 The Akkermansia genus has nutrients to brain microglia to support healthy
also been linked to cognitive impairment as maturation and function.103 In summary, a link
a result of poor dietary habits.97 Mice demon- has been established between the functional by-
strated a 5.4-fold reduction in Akkermansia and products (i.e., SFCA) of the gut microbiome and
associated behavioral decline in response to the healthy behaviors (exercise and diet).99
Western diet (high fat and sugar intake).98 While The bulk of research examining the role of exercise
the link between exercise changes to the gut interventions on changes in the gut microbiome has
microbiome and resulting changes in brain func- been performed in rodent models. Kang et al.104 ana-
tion is unknown, the alterations among athletes lyzed changes in the gut bacteria among mice after
appear to be a result of exercise and dietary beha- a 16-week exercise program (forced wheel running
viors, which may provide protection against both 5 days/week) compared to a high-fat diet group.104
metabolic and cognitive decline. Exercise appeared to increase the bacteria of phylum
In a follow-up study, the same group of Firmicutes and lower Bacteroidetes, which was sur-
researchers confirmed the microbial diversity prisingly similar to the changes observed in the high-
among elite rugby athletes compared to nonactive, fat diet group. Conversely, Evans et al.105 reported
age-matched control and again demonstrated opposite findings with lower Firmicutes and higher
higher levels of Akkermansia.99 The researchers Bacteroidetes after 12 weeks of voluntary wheel run-
also performed metabolomic phenotyping of the ning. The group also demonstrated exercise preven-
microbiome to further understand the differences tion of high-fat diet-induced obesity and suggested
between athletes and controls at the functional a microbial altering mechanism of exercise on avert-
metabolic level. The athletes reported higher activ- ing high-fat diet-related obesity.105 The differential
ity of several pathways, including amino acid results between the Kang and Evans groups may be
synthesis, and carbohydrate metabolism. As attributed to the forced vs. voluntary wheel running
a result, athletes demonstrated an enriched profile protocols utilized in the studies, respectively (dis-
of SCFAs, indicating greater production and host cussed in next section). A link has been made between
absorption rates. Identified SFCAs in the athletes low gut Bacteriodetes and obesity and further sup-
were acetate, propionate, and butyrate, which are ported by increased Bacteriodetes in response to
produced by gut microbes from protein, fibers, weight loss among obese individuals.106 The richness
and nondigestible starches.100 The researchers and diversity of the gut microbiome has been corre-
attributed the differences between athletes and lated with various metabolic markers where low rich-
controls to heavy physical activity and the asso- ness corresponds with higher adiposity, insulin
ciated diet. They highlighted the impact of the resistance, and dyslipidemia.107 Exercise is an estab-
high protein and fiber diet among the athletic lished treatment for metabolic disorders, and these
group and the resulting metabolism by gut results may indicate that exercise-related changes in
microbes. In support of these findings, Estaki et - the gut microbiome may support metabolic health.
al.101 distinctly identified an association between In a short voluntary wheel running exercise proto-
cardiorespiratory fitness and microbiome function col (6 days), Queipo-Ortuno et al.108 reported an
rather than diversity of specific bacterial taxa. increase in genera Lactobacillus and Blautia coccoides-
A strong relationship was observed between Eubacterium rectale from the Firmicutes phylum,
560 A. DALTON ET AL.
along with genus Bifidobacterium from the These findings suggest that aerobic exercise affects
Actinobaceria phylum. All three genera are capable the content and diversity of the gut microbiome in
of producing SCFAs,109 which may be considered humans and may additionally raise the question
a functional change in the gut microbiome as about how gut microbes influence adaptations to
a result of exercise. Queipo-Ortuno et al.108 also exercise.
posted a positive association between Lactobacillus The identified “exercise” genera by Allen et al.114
and Bifidobacterium and serum leptin levels in the were all producers of SCFAs and more specifically
animals. Leptin is a hormone secreted from adipose butyrate. Butyrate and other SFCAs are known
tissue and helps to control appetite and eating energy substrates for colonocytes, with butyrate
behavior.110 These results demonstrate that exercise being the primary energy source.115,116 This may
alterations to the gut microbiome are related to diet- explain why exercise improves symptoms and
ary food intake and may indicate a connection reduces the risk for IBS and colon cancer.117
between exercise, the gut, and behavioral change. Further, butyrate has been shown to stimulate
Several recent exercise intervention studies have neural proliferation in the dentate gyrus brain
been conducted among human subjects. While the regions of mice and has also been used to induce
primary aim of this review is to assess the impact of neurogenesis after ischemic brain insult in adult
exercise training on the gut microbiome. Zhao et al.- rodents.118,119 These results may provide a link
111 between exercise-induced microbial SCFA produc-
analyzed fecal samples before and after a half-
marathon event among human participants. An tion and improvements in both brain and GI func-
increase in the richness of the Actinobacteria phylum tion. However, it is unknown if the positive benefits
was reported with a notable correlation between the of exercise on gut and brain are mediated through
Coriobacteriaceae family and several metabolites. adjustments to the microbiome (see Figure 1).
Coriobacteriaceae has been associated with higher The limited amount of research explaining how
levels of high-density lipoprotein (HDL) and meta- exercise influences the microbiome has indicated
bolic improvements, and the researchers identified that genera from the Firmicutes phylum appear to
the change in bacteria as a potential mechanism for be the most responsive to exercise-induced
exercise-induced health benefits.112,113 These results changes. Given that the Firmicutes phylum con-
indicate that an acute bout of exercise, while extreme sists of over 250 genera, it is difficult to under-
(i.e., half-marathon) has a profound impact on the stand which genera lead to systemic inflammation
gut microbiome. and which lead to a healthy gut environment. The
In the first known human exercise study, Allen work from Allen et al.114 has implied that genera
et al.114 compared microbiota changes between that produce SCFA appear to respond to exercise;
lean and obese humans before and after a 6-week however, this is the only known study among
aerobic exercise program and further evaluated humans to evaluate changes after an aerobic exer-
changes after a period of de-training. Exercise cise intervention, and then upon cessation of exer-
caused changes in gut microbiome diversity and cise. In the only other human exercise study,
microbial production of SCFAs among both the Cronin et al.120 reported minimal change in gut
lean and obese subjects. The authors aimed to bacterial diversity among healthy male and females
identify groups of bacteria that responded to exer- who participated in an 8-week combined aerobic
cise training and identified the Faecalibacterium, and resistance exercise program. The conflicting
Lachnospira, Clostridia, and Roseburia genera, results may be due to the exercise regimen where
which are all part of the Firmicutes phylum. the aerobic training program in the Allen et al.114
These genera increased in abundance in response study appeared to be more vigorous, and 100%
to exercise training and returned to baseline upon compliance was reported. Importantly, Cronin et -
cessation. A notable finding was that changes in al.120 implemented a nonexercise control to accu-
the gut were associated with changes in lean mass rately assess and compare sedentary and exercise
and reduction in fat mass, meaning that shifts in behavior. They concluded that while the gut
microbial content were more substantial among microbiome is adaptable, 8 weeks of exercise may
those subjects that demonstrate these changes. not be enough stimulus to enact change. These
GUT MICROBES 561
studies have advanced the field and future work is results in apoptosis (death of cells) in mice.
needed to fully characterize how the microbiome Decreased levels of BDNF in the hippocampus
adapts to exercise, and other various forms of have also been associated with anxiety and depres-
physical activity. sion and is often comorbid with gut inflammatory
diseases, such as IBS, and or inflammatory-bowel
disease (IBD).8,133 Oral supplementation with
Exercise influences the microbiome–gut– Bifidobacterium has been shown to increase
brain axis pathways of communication
BDNF expression in the brains of rodents, while
As mentioned previously, the vagus nerve is at the aerobic exercise in piglets has also been shown to
interface of the microbiome–gut–brain axis. increase Bifidobacterium in the gut.64,134 Antibiotic
Altered vagus nerve activation is commonly found treatments that killed gut bacteria also delayed the
in patients with both GI (IBS, IBD) and psycholo- growth of brain cells in the hippocampus regions
gical disorders (depression).121,122 Various forms of of mice.135 Both probiotics and aerobic exercise
exercise, including yoga and aerobic exercise train- individually rescued the decline in neurogenesis
ing, have been shown to improve parasympathetic and cognitive function among mice treated with
tone.123 Meta-analytical studies have identified the antibiotic.135 However, it is unknown if the
yoga as a proven mode of exercise to improve exercise effects on brain neurogenesis and cogni-
symptoms of IBS and depression,124,125 which may tion are mediated through Bifidobacterium.136
be partially mediated by autonomic influences. As Elevated HPA axis activation and gut–microbiome
previously discussed, alterations in the gut micro- disruption is seen when an exercising individual
biome affect vagal communication between the gut exceeds 60% of maximum oxygen uptake (VO2max)
and brain.126 An intact vagus nerve is critical for the or during prolonged exercise (>90 min).
benefits of probiotics supplements containing Psychological stress, such as the kind experienced by
strains of Bifidobacterium and Lactobacillus to be athletes pre-competition, has also led to an increased
effective.55 Further, any mediating effect of probio- HPA axis activation and greater gut disruption.10
tics on the HPA axis and depression-related symp- Likewise, research in mice has found that moderate
toms are abolished through the vagotomy forced treadmill running (8–12 m/min at a 5% grade
procedure.127 Therefore, it is logical to suggest for 40 min, 5 times per week) exacerbated colitis
that any alterations that exercise may have on the symptoms when compared to voluntary exercise,
diversity and expression of gut flora would poten- which attenuated symptoms.137 The researchers con-
tially be manifested through influence on vagal cluded that the forced exercise may have been per-
communication between the gut and brain. ceived as a psychological stressor by the mice, whereas
Unfortunately, empirical evidence currently does the voluntary wheel running is a conscious decision.
not exist to verify this conclusion. Hyperresponsive HPA axis appears to be influenced
Improvements in brain structure and function by the diversity of the gut microbiome where the lack
have been documented as an adaptation to repeti- of, or absence of bacteria, caused robust HPA activa-
tive aerobic exercise.40,128,129 Regular moderate- tion in response to psychological stress.64
intensity aerobic exercise prevents age-related Supplementation with Bifidobacterium species
brain volume loss and increased brain volume in reversed HPA overactivation and alleviated symp-
areas responsible for cognition and control of toms of anxiety and distress among rodents and
attention and memory.40 The positive benefits of humans, respectively.35 While previously noted, the
exercise on neural function appear to be mediated Bifidobacterium strains are influenced by aerobic
through regulation of BDNF. BDNF is central in exercise, where animals with free access to wheel
the growth and the survival of striatal neurons in running showed increased numbers of
108
the brain, and it also plays a critical role in reg- Bifidobacterium. Furthermore, force wheel running
ulating mood disorders, along with learning and resulted in reduced levels of Lactobacillus, which is
memory.34,130,131 According to Linnarsson et al.,132 important for protection against disease-causing
BDNF is known for its protective role in the adult microorganisms.138 It is important to mention that
brain, because the genetic deletion of this protein microbiome-altering effect of voluntary wheel
562 A. DALTON ET AL.
running has only been shown in male mice, whereas exercise can be a therapeutic factor and strategy for
null findings were reported in a combined cohort of both psychological and GI disorders. The main
male and female mice.138,139 However, both voluntary identified phyla that respond to exercise are
and forced exercise appear to influence the average Firmicutes and Actinobacteria, which contain the
operational taxonomic units (OTUs) of several micro- Lactobacillus and Bifidobacterium genera, respec-
bial species. For example, force running increased tively. In addition, the SCFA-producing genera
Ruminococcus, Butyrivibrio, and Oscillospira, which from the Firmicutes phylum also appear to increase
are all within Firmicutes phylum. Of note, many of in response to exercise. Future work should target
these various species are being considered as psycho- underlying mechanisms of how specifically exercise
biotics due to their role in neuropsychiatric influences the microbiome, and the mediators
disorders.140 In summary, it appears that the psycho- involved in the gut–brain axis. This may lead to
logical state of exercise influences the microbiome, the development of various treatment combina-
and these changes may further impact the HPA axis tions (exercise + probiotics) to ameliorate (or
regulation and mood disorder symptoms. improve) specific disease states.
Aerobic exercise has been shown to also influence
serotonin regulation. In rats, 5-HT synthesis and
metabolism increased in both the brain stem and the Author contributions
hippocampus in response to exercise,141 and this AD, CM, and MZ all participated in manuscript develop-
increase in 5-HT resulted in the reduction of depres- ment, design, and drafting.
sive and anxiety symptoms.142 Wipfili et. al.142
described the effects of exercise as similar to the
those of SSRIs used to treat depression and anxiety. Disclosure of Potential Conflicts of Interest
The following bacteria in the gut microbiome have
No potential conflicts of interest were disclosed.
been found to produce serotonin: Lactococcus lactis,
Lactobacillus plantarum, Streptococcus thermophile,143
Morganella morganii, and Klebsiella pneumonia.144 Funding
Unfortunately, there are no previous studies that thor-
This research did not receive any specific grant from funding
oughly examined if exercise has any influence on agencies in the public, commercial, or not-for-profit sectors.
these specific strains. Because some of these seroto-
nin-producing strains stem from the lactobacilis gen-
era (which is a component of Firmicutes phyla), References
exercise may support the increased presence of these
strains through the natural diversification of gut 1. Sender R, Fuchs S, Milo R. Are we really vastly out-
microbiota that occurs as a result of exercise. numbered? Revisiting the ratio of bacterial to host cells
in humans. Cell. 2016;164(3):337–340. doi:10.1016/j.
Therefore, the production of serotonin through an cell.2016.01.013.
exercise-adapted gut microbiome might possibly be 2. Chen J, He X, Huang J. Diet effects in gut microbiome
an explanation as to why subjects exhibit lower stress- and obesity. J Food Sci. 2014;79:4. doi:10.1111/1750-
related symptoms, such as anxiety and depression, 3841.12397.
after participating in physical activity. These state- 3. Peters H, De Vries W, Vanberge-Henegouwen G,
ments, however, are purely assumptions and more Akkermans L. Potential benefits and hazards of physi-
cal activity and exercise on the gastrointestinal tract.
research is needed to confirm this suggestion. Gut. 2001;48:435–439.
4. Mach N, Fuster-Botella D. Endurance exercise and gut
microbiota: A review. J Sport Health Sci. 2017;6
Conclusion (2):179–197. doi:10.1016/j.jshs.2016.05.001.
Although the exact causal relationship is unknown, 5. Cronin O, Molloy MG, Shanahan F. Exercise, fitness,
and the gut. Curr Opin Gastroenterol. 2016;32
current evidence allows for the assumption that
(2):67–73. doi:10.1097/MOG.0000000000000240.
exercise may mediate a bidirectional relationship 6. Bermon S, Petriz B, Kajeniene A, Prestes J, Castell L,
between the gut and brain through alterations in Franco OL. The microbiota: an exercise immunology
the microbiome. This relationship may explain why perspective. Exerc Immunol Rev. 2015;21:9.
GUT MICROBES 563
7. Sharon G, Sampson TR, Geschwind DH, microbiota changes with age. BMC Microbiol. 2009;9
Mazmanian SK. The central nervous system and the (1):123. doi:10.1186/1471-2180-9-123.
gut microbiome. Cell. 2016;167(4):915–932. 19. Conlan S, Kong HH, Segre JA. Species-level analysis of
doi:10.1016/j.cell.2016.10.027. DNA sequence data from the NIH human microbiome
8. Proctor C, Thiennimitr P, Chattipakorn N, project. PLoS One. 2012;7(10):e47075. doi:10.1371/
Chattipakorn SC. Diet, gut microbiota and cognition. journal.pone.0047075.
Metab Brain Dis. 2017;32(1):1–17. doi:10.1007/s11011- 20. Group NHW, Peterson J, Garges S, Giovanni M,
016-9917-8. McInnes P, Wang L, Schloss JA, Bonazzi V,
9. Foster JA, Rinaman L, Cryan JF. Stress & the gut-brain McEwen JE, Wetterstrand KA, et al. The NIH human
axis: regulation by the microbiome. Neurobiol Stress. microbiome project. Genome Res. 2009;19
2017;7:124–136. doi:10.1016/j.ynstr.2017.03.001. (12):2317–2323. doi:10.1101/gr.096651.109.
10. Clark A, Mach N. Exercise-induced stress behavior, 21. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK,
gut-microbiota-brain axis and diet: a systematic review Knight R. Diversity, stability and resilience of the
for athletes. J Int Soc Sports Nutr. 2016;13:43. human gut microbiota. Nature. 2012;489(7415):220.
doi:10.1186/s12970-016-0155-6. doi:10.1038/nature11468.
11. Burokas A, Arboleya S, Moloney RD, Peterson VL, 22. Hartstra AV, Bouter KE, Bäckhed F, Nieuwdorp M.
Murphy K, Clarke G, Stanton C, Dinan TG, Cryan Insights into the role of the microbiome in obesity and
JF. Targeting the microbiota-gut-brain axis: prebiotics type 2 diabetes. Diabetes Care. 2015;38(1):159–165.
have anxiolytic and antidepressant-like effects and doi:10.2337/dc14-0769.
reverse the impact of chronic stress in mice. Biol 23. Li D, Kirsop J, Tang WW. Listening to our gut: con-
Psychiatry. 2017;82(7):472–487. doi:10.1016/j. tribution of gut microbiota and cardiovascular risk in
biopsych.2016.12.031. diabetes pathogenesis. Curr Diab Rep. 2015;15(9):63.
12. Braniste V, Al-Asmakh M, Kowal C, Anuar F, doi:10.1007/s11892-015-0634-1.
Abbaspour A, Tóth M, Korecka A, Bakocevic N, 24. Dinan TG, Cryan JF. Melancholic microbes: a link
Ng LG, Guan NL, et al. The gut microbiota influences between gut microbiota and depression?
blood-brain barrier permeability in mice. Sci Transl Neurogastroenterol Motil. 2013;25(9):713–719.
Med. 2014;6(263):263ra158–263ra158. doi:10.1126/ doi:10.1111/nmo.12198.
scitranslmed.3009759. 25. Cani PD, Delzenne NM. Involvement of the gut micro-
13. Wong JM, de Souza R, Kendall CW, Emam A, biota in the development of low grade inflammation
Jenkins DJ. Colonic health: fermentation and short associated with obesity: focus on this neglected partner.
chain fatty acids. J Clin Gastroenterol. 2006;40:235–243. Acta Gastroenterol Belg. 2010;73:267–269.
14. Lin HV, Frassetto A, Kowalik EJ Jr, Nawrocki AR, 26. Cani PD, Possemiers S, Van de Wiele T, Guiot Y,
Lu MM, Kosinski JR, Hubert JA, Szeto D, Yao X, Everard A, Rottier O, Geurts L, Naslain D,
Forrest G, et al. Butyrate and propionate protect Neyrinck A, Lambert DM, et al. Changes in gut micro-
against diet-induced obesity and regulate gut hormones biota control inflammation in obese mice through
via free fatty acid receptor 3-independent mechanisms. a mechanism involving GLP-2-driven improvement of
PLoS One. 2012;7(4):e35240. doi:10.1371/journal. gut permeability. Gut. 2009;58(8):1091–1103.
pone.0035240. doi:10.1136/gut.2008.165886.
15. Tang WW, Wang Z, Levison BS, Koeth RA, Britt EB, 27. Everard A, Lazarevic V, Derrien M, Girard M,
Fu X, Wu Y, Hazen SL. Intestinal microbial metabo- Muccioli GG, Neyrinck AM, Neyrinck AM,
lism of phosphatidylcholine and cardiovascular risk. Possemiers S, Van Holle A, François P, et al.
N Engl J Med. 2013;368(17):1575–1584. doi:10.1056/ Responses of gut microbiota and glucose and lipid
NEJMoa1109400. metabolism to prebiotics in genetic obese and
16. Kelly D, Campbell JI, King TP, Grant G, Jansson EA, diet-induced leptin-resistant mice. Diabetes. 2011;60
Coutts AG, Pettersson S, Conway S. Commensal anae- (11):2775–2786. doi:10.2337/db11-0227.
robic gut bacteria attenuate inflammation by regulating 28. Mayer EA. Gut feelings: the emerging biology of gut–
nuclear-cytoplasmic shuttling of PPAR-γ and RelA. brain communication. Nat Rev Neurosci. 2011;12
Nat Immunol. 2004;5(1):104. doi:10.1038/ni1018. (8):453. doi:10.1038/nrn3071.
17. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, 29. Kennedy P, Clarke G, O‘Neill A, Groeger J, Quigley E,
Blekhman R, Beaumont M, Van Treuren W, Knight R, Shanahan F, Cryan JF, Dinan TG. Cognitive perfor-
Bell JT, et al. Human genetics shape the gut mance in irritable bowel syndrome: evidence of a
microbiome. Cell. 2014;159(4):789–799. doi:10.1016/j. stress-related impairment in visuospatial memory.
cell.2014.09.053. Psychol Med. 2014;44(7):1553–1566. doi:10.1017/
18. Mariat D, Firmesse O, Levenez F, Guimarăes V, S0033291713002171.
Sokol H, Doré J, Corthier G, Furet J-P. The 30. Kennedy PJ, Cryan JF, Dinan TG, Clarke G. Irritable
Firmicutes/Bacteroidetes ratio of the human bowel syndrome: a microbiome-gut-brain axis
564 A. DALTON ET AL.
disorder? World J Gastroenterol. 2014;20(39):14105. extremes impact on gut microbial diversity. Gut. 2014;63
doi:10.3748/wjg.v20.i39.14105. (12):1913–1920. doi:10.1136/gutjnl2013–306541.
31. Castanon N, Lasselin J, Capuron L. Neuropsychiatric 43. Mika A, Van Treuren W, González A, Herrera JJ,
comorbidity in obesity: role of inflammatory processes. Knight R, Fleshner M. Exercise is more effective at
Front Endocrinol (Lausanne). 2014;5:74. doi:10.3389/ altering gut microbial composition and producing
fendo.2014.00074. stable changes in lean mass in juvenile versus adult
32. Lackner JM, Ma CX, Keefer L, Brenner DM, male F344 rats. PLoS One. 2015;10(5):e0125889.
Gudleski GD, Satchidanand N, Firth R, Sitrin MD, doi:10.1371/journal.pone.0125889.
Katz L, Krasner SS, et al. Type, rather than number, 44. Welly RJ, Liu T-W, Zidon TM, Rowles JL III, Park Y-M,
of mental and physical comorbidities increases the Smith TN, Swanson KS, Padilla J, Vieira-Potter VJ.
severity of symptoms in patients with irritable bowel Comparison of diet vs. exercise on metabolic function &
syndrome. Clin Gastroenterol Hepatol. 2013;11 gut microbiota in obese rats. Med Sci Sports Exerc. 2016;48
(9):1147–1157. doi:10.1016/j.cgh.2013.03.011. (9):1688. doi:10.1249/MSS.0000000000000964.
33. Thorkelson G, Bielefeldt K, Szigethy E. Empirically 45. Monda V, Villano I, Messina A, Valenzano A,
supported use of psychiatric medications in adolescents Esposito T, Moscatelli F, Viggiano A, Cibelli G,
and adults with IBD. Inflamm Bowel Dis. 2016;22 Chieffi S, Monda M, et al. Exercise modifies the gut
(6):1509–1522. doi:10.1097/MIB.0000000000000734. microbiota with positive health effects. Oxid Med Cell
34. Cryan JF, O’mahony S. The microbiome-gut-brain Longev. 2017;2017. doi:10.1155/2017/3831972.
axis: from bowel to behavior. Neurogastroenterol 46. Clarke G, Dinan T, Cryan J. Microbiome–gut–brain
Motil. 2011;23(3):187–192. doi:10.1111/j.1365- Axis. In: Highlander SK, Rodriguez-Valera F,
2982.2010.01664.x. White BA, editors. Encyclopedia of metagenomics:
35. Messaoudi M, Lalonde R, Violle N, Javelot H, environmental metagenomics. Boston (MA): Springer
Desor D, Nejdi A, Bisson J-F, Rougeot C, US; 2015. p. 425–437.
Pichelin M, Cazaubiel M, et al. Assessment of 47. Grenham S, Clarke G, Cryan JF, Dinan TG. Brain–gut–
psychotropic-like properties of a probiotic formula- microbe communication in health and disease. Front
tion (Lactobacillus helveticus R0052 and Physiol. 2011;2:94. doi:10.3389/fphys.2011.00094.
Bifidobacterium longum R0175) in rats and human 48. Cryan JF, Dinan TG. Mind-altering microorganisms:
subjects. Br J Nutr. 2011;105(5):755–764. doi:10.1017/ the impact of the gut microbiota on brain and
S0007114510004319. behaviour. Nat Rev Neurosci. 2012;13(10):701.
36. Benton D, Williams C, Brown A. Impact of consuming doi:10.1038/nrn3346.
a milk drink containing a probiotic on mood and 49. Forsythe P, Kunze W, Bienenstock J. Moody microbes
cognition. Eur J Clin Nutr. 2007;61(3):355. or fecal phrenology: what do we know about the
doi:10.1038/sj.ejcn.1602546. microbiota-gut-brain axis? BMC Med. 2016;14(1):58.
37. Desbonnet L, Garrett L, Clarke G, Bienenstock J, doi:10.1186/s12916-016-0604-8.
Dinan TG. The probiotic Bifidobacteria infantis: an 50. Brookes SJ, Spencer NJ, Costa M, Zagorodnyuk VP.
assessment of potential antidepressant properties in Extrinsic primary afferent signalling in the gut. Nat Rev
the rat. J Psychiatr Res. 2008;43(2):164–174. Gastroenterol Hepatol. 2013;10(5):286. doi:10.1038/
doi:10.1016/j.jpsychires.2008.03.009. nrgastro.2013.29.
38. Hoveyda N, Heneghan C, Mahtani KR, Perera R, 51. Forsythe P, Kunze WA, Bienenstock J. On communi-
Roberts N, Glasziou P. A systematic review and cation between gut microbes and the brain. Curr Opin
meta-analysis: probiotics in the treatment of irritable Gastroenterol. 2012;28(6):557–562. doi:10.1097/
bowel syndrome. BMC Gastroenterol. 2009;9(1):15. MOG.0b013e3283572ffa.
doi:10.1186/1471-230X-9-15. 52. Forsythe P, Bienenstock J, Kunze WA. Vagal pathways for
39. Ringel Y, Quigley EM, Lin HC. Using probiotics in microbiome-brain-gut axis communication. In: Microbial
gastrointestinal disorders. Am J Gastroenterol Suppl. endocrinology: the microbiota-gut-brain axis in health and
2012;1(1):34. doi:10.1038/ajgsup.2012.7. disease. Adv Exp Med Biol. 2014;817:115–133. doi:10.1007/
40. Colcombe SJ, Erickson KI, Scalf PE, Kim JS, Prakash R, 978-1-4939-0897-4.
McAuley E, Elavsky S, Marquez DX, Hu L, Kramer AF. 53. Pellissier S, Dantzer C, Mondillon L, Trocme C,
Aerobic exercise training increases brain volume in Gauchez A-S, Ducros V, Mathieu N, Toussaint B,
aging humans. J Gerontol A Biol Sci Med Sci. 2006;61 Fournier A, Canini F, et al. Relationship between vagal
(11):1166–1170. doi:10.1093/gerona/61.11.1166. tone, cortisol, TNF-alpha, epinephrine and negative affects
41. Gomez-Pinilla F, Hillman C. The influence of exercise in Crohn’s disease and irritable bowel syndrome. PLoS One.
on cognitive abilities. Compr Physiol. 2013;3(1):403– 2014;9(9):e105328. doi:10.1371/journal.pone.0105328.
428. doi:10.1002/cphy/c110063. 54. Liu Q, Wang EM, Yan XJ, Chen SL. Autonomic func-
42. Clarke SF, Murphy EF, O’sullivan O, Lucey AJ, tioning in irritable bowel syndrome measured by heart
Humphreys M, Hogan A. Hayes P, O'Reilly M, Jeffery rate variability: A meta-analysis. J Dig Dis. 2013;14
IB, Wood-Martin R, et al. Exercise and associated dietary (12):638–646. doi:10.1111/1751-2980.12092.
GUT MICROBES 565
55. Malick M, Gilbert K, Daniel J, Arseneault-Breard J, between women with active lifestyle and sedentary
Tompkins T, Godbout R, Rousseau G. Vagotomy pre- women. PLoS One. 2017;12(2):e0171352. doi:10.1371/
vents the effect of probiotics on caspase activity in journal.pone.0171352.
a model of postmyocardial infarction depression. 67. Schecterson LC, Bothwell M. Novel roles for neurotrophins
Neurogastroenterol Motil. 2015;27(5):663–671. are suggested by BDNF and NT-3 mRNA expression in
doi:10.1111/nmo.12540. developing neurons. Neuron. 1992;9:449–463.
56. Li S, Zhai X, Rong P, McCabe MF, Wang X, Zhao J, 68. Li C, Cai -Y-Y, Yan Z-X. Brain-derived neurotrophic
Ben H, Wang S, Slattery DA. Therapeutic effect of factor preserves intestinal mucosal barrier function and
vagus nerve stimulation on depressive-like behavior, alters gut microbiota in mice. Kaohsiung J Med Sci.
hyperglycemia and insulin receptor expression in 2018;34(3):134–141. doi:10.1016/j.kjms.2017.11.002.
Zucker fatty rats. PLoS One. 2014;9(11):e112066. 69. Bravo JA, Julio-Pieper M, Forsythe P, Kunze W,
doi:10.1371/journal.pone.0112066. Dinan TG, Bienenstock J, Cryan JF. Communication
57. Grimonprez A, Raedt R, Baeken C, Boon P, Vonck K. between gastrointestinal bacteria and the nervous
The antidepressant mechanism of action of vagus nerve system. Curr Opin Pharmacol. 2012;12(6):667–672.
stimulation: evidence from preclinical studies. doi:10.1016/j.coph.2012.09.010.
Neurosci Biobehav Rev. 2015;56:26–34. doi:10.1016/j. 70. von Boyen GB, Reinshagen M, Steinkamp M, Adler G,
neubiorev.2015.06.019. Kirsch J. Enteric nervous plasticity and development:
58. Kheder SH, Heller J, Bär J, Wutzler A, Menge B, dependence on neurotrophic factors. J Gastroenterol.
Juckel G. Autonomic dysfunction of gastric motility 2002;37(8):583–588. doi:10.1007/s005350200093.
in major depression. J Affect Disord. 71. Heijtz RD, Wang S, Anuar F, Qian Y, Björkholm B,
2018;226:196–202. doi:10.1016/j.jad.2017.09.049. Samuelsson A, Hibberd ML, Forssberg H, Pettersson S.
59. Bercik P, Park A, Sinclair D, Khoshdel A, Lu J, Huang X, Normal gut microbiota modulates brain development
Deng Y, Blennerhassett PA, Fahnestock M, Moine D, and behavior. Proc Natl Acad Sci. 2011;108
et al. The anxiolytic effect of Bifidobacterium longum (7):3047–3052. doi:10.1073/pnas.1010529108.
NCC3001 involves vagal pathways for gut–brain commu- 72. Jenkins TA, Nguyen JC, Polglaze KE, Bertrand PP.
nication. Neurogastroenterol Motil. 2011;23 Influence of tryptophan and serotonin on mood and
(12):1132–1139. doi:10.1111/j.1365-2982.2011.01796.x. cognition with a possible role of the gut-brain axis.
60. Tsigos C, Chrousos GP. Hypothalamic–pituitary–adre- Nutrients. 2016;8(1):56. doi:10.3390/nu8010056.
nal axis, neuroendocrine factors and stress. 73. Young SN. How to increase serotonin in the human
J Psychosom Res. 2002;53:865–871. brain without drugs. J Psychol Psychiatr Neuroscil.
61. Farzi A, Fröhlich EE, Holzer P. Gut microbiota and the 2007;32:394.
neuroendocrine system. Neurotherapeutics. 2018;15 74. Gershon MD, Tack J. The serotonin signaling system:
(1):1–18. doi:10.1007/s13311-017-0600-5. from basic understanding to drug development for
62. Hosoi T, Okuma Y, Matsuda T, Nomura Y. Novel functional GI disorders. Gastroenterology. 2007;132
pathway for LPS-induced afferent vagus nerve activa- (1):397–414. doi:10.1053/j.gastro.2006.11.002.
tion: possible role of nodose ganglion. Auton Neurosci: 75. Mawe GM, Hoffman JM. Serotonin signalling in the
Basic Clin. 2005;120(1):104–107. doi:10.1016/j. gut—functions, dysfunctions and therapeutic targets.
autneu.2004.11.012. Nat Rev Gastroenterol Hepatol. 2013;10(8):473.
63. Dinan TG, Quigley EM, Ahmed SM, Scully P, doi:10.1038/nrgastro.2013.105.
O’Brien S, O’Mahony L, O’Mahony S, Shanahan F, 76. Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P,
Keeling PWN. Hypothalamic-pituitary-gut axis dysre- Ma L, Nagler CR, Ismagilov RF, Mazmanian SK,
gulation in irritable bowel syndrome: plasma cytokines Hsiao EY. Indigenous bacteria from the gut microbiota
as a potential biomarker? Gastroenterology. 2006;130 regulate host serotonin biosynthesis. Cell. 2015;161
(2):304–311. doi:10.1053/j.gastro.2005.11.033. (2):264–276. doi:10.1016/j.cell.2015.02.047.
64. Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, 77. Clarke G, Grenham S, Scully P, Fitzgerald P,
Kubo C, Koga Y. Postnatal microbial colonization pro- Moloney R, Shanahan F, Dinan TG, Cryan JF. The
grams the hypothalamic–pituitary–adrenal system for microbiome-gut-brain axis during early life regulates
stress response in mice. J Physiol. 2004;558(1):263–275. the hippocampal serotonergic system in a
doi:10.1113/jphysiol.2004.063388. sex-dependent manner. Mol Psychiatry. 2013;18
65. O’Mahony S, Clarke G, Borre Y, Dinan T, Cryan J. (6):666. doi:10.1038/mp.2012.77.
Serotonin, tryptophan metabolism and the brain-gut- 78. Yanofsky C. RNA-based regulation of genes of trypto-
microbiome axis. Behav Brain Res. 2015;277:32–48. phan synthesis and degradation, in bacteria. Rna.
doi:10.1016/j.bbr.2014.07.027. 2007;13(8):1141–1154. doi:10.1261/rna.620507.
66. Bressa C, Bailén-Andrino M, Pérez-Santiago J, 79. Strasser B, Gostner JM, Fuchs D. Mood, food, and
González-Soltero R, Pérez M, Montalvo-Lominchar cognition: role of tryptophan and serotonin. Curr
MG, Maté-Muñoz JL, Domínguez R, Moreno D, Opin Clin Nutr Metab Care. 2016;19(1):55–61.
Larrosa M, et al. Differences in gut microbiota profile doi:10.1097/MCO.0000000000000237.
566 A. DALTON ET AL.
80. Ochoa-Reparaz J, Mielcarz D, Wang Y, Begum- 91. Jeffery IB, O’toole PW, Öhman L, Claesson MJ,
Haque S, Dasgupta S, Kasper D, Kasper L. Deane J, Quigley EM, Simrén M. An irritable bowel
A polysaccharide from the human commensal bacter- syndrome subtype defined by species-specific altera-
oides fragilis protects against CNS demyelinating tions in faecal microbiota. Gut. 2012;61(7):997–1006.
disease. Mucosal Immunol. 2010;3(5):487. doi:10.1136/gutjnl-2011-301501.
doi:10.1038/mi.2009.138. 92. Kajander K, Myllyluoma E, Rajilić-Stojanović M,
81. Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, Kyrönpalo S, Rasmussen M, Järvenpää S,
McCue T, Codelli JA, Chow J, Reisman SE, Zoetendal EG, de Vos WM, Vapaatalo H, Korpela R.
Petrosino JF, et al. Microbiota modulate behavioral Clinical trial: multispecies probiotic supplementation
and physiological abnormalities associated with neuro- alleviates the symptoms of irritable bowel syndrome
developmental disorders. Cell. 2013;155(7):1451–1463. and stabilizes intestinal microbiota. Aliment
doi:10.1016/j.cell.2013.11.024. Pharmacol Ther. 2008;27(1):48–57. doi:10.1111/
82. Yunes R, Poluektova E, Dyachkova M, Klimina K, j.1365-2036.2007.03542.x.
Kovtun A, Averina O, Orlova VS, Danilenko VN. 93. Choi CH, Jo SY, Park HJ, Chang SK, Byeon J-S,
GABA production and structure of gadB/gadC genes Myung S-J. A randomized, double-blind,
in lactobacillus and bifidobacterium strains from placebo-controlled multicenter trial of Saccharomyces
human microbiota. Anaerobe. 2016;42:197–204. boulardii in irritable bowel syndrome: effect on quality
doi:10.1016/j.anaerobe.2016.10.011. of life. J Clin Gastroenterol. 2011;45(8):679–683.
83. Asano Y, Hiramoto T, Nishino R, Aiba Y, Kimura T, doi:10.1097/MCG.0b013e318204593e.
Yoshihara K, Koga Y, Sudo N. Critical role of gut 94. Diop L, Guillou S, Durand H. Probiotic food supple-
microbiota in the production of biologically active, ment reduces stress-induced gastrointestinal symptoms
free catecholamines in the gut lumen of mice. Am in volunteers: a double-blind, placebo-controlled, ran-
J Physiol Gastrointest Liver Physiol. 2012;303(11): domized trial. Nutr Res. 2008;28(1):1–5. doi:10.1016/j.
G1288–G1295. doi:10.1152/ajpgi.00341.2012. nutres.2007.10.001.
84. Zuhl M, Dokladny K, Mermier C, Schneider S, Salgado R, 95. McFadzean R. Exercise can help modulate human gut
Moseley P. The effects of acute oral glutamine supplemen- microbiota. CU Scholar. 2014. Undergraduate honors
tation on exercise-induced gastrointestinal permeability and thesis 155.
heat shock protein expression in peripheral blood mono- 96. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C,
nuclear cells. Cell Stress Chaperones. 2015;20(1):85–93. Bindels LB, Guiot Y, Derrien M, Muccioli GG,
doi:10.1007/s12192-014-0528-1. Delzenne NM, et al. Cross-talk between Akkermansia
85. Lambert GP. Intestinal barrier dysfunction, endotoxe- muciniphila and intestinal epithelium controls
mia, and gastrointestinal symptoms: the ‘canary in the diet-induced obesity. Proc Natl Acad Sci USA. 2013;110
coal mine’during exercise-heat stress?. In: (22):9066–9071. doi:10.1073/pnas.1219451110.
Thermoregulation and human performance. Med 97. Noble EE, Hsu TM, Kanoski SE. Gut to brain dysbiosis:
Sport Sci. 2008;53:61–73. doi:10.1159/000151550. mechanisms linking western diet consumption, the
86. Dokladny K, Zuhl MN, Moseley PL. Intestinal epithe- microbiome, and cognitive impairment. Front Behav
lial barrier function and tight junction proteins with Neurosci. 2017;11:9. doi:10.3389/fnbeh.2017.00246.
heat and exercise. J Appl Physiol. 2015;120(6):692–701. 98. Bruce-Keller AJ, Salbaum JM, Luo M, Blanchard E IV,
doi:10.1152/japplphysiol.00536.2015. Taylor CM, Welsh DA, Berthoud H-R. Obese-type gut
87. Johannesson E, Simrén M, Strid H, Bajor A, Sadik R. microbiota induce neurobehavioral changes in the
Physical activity improves symptoms in irritable bowel absence of obesity. Biol Psychiatry. 2015;77
syndrome: a randomized controlled trial. Am (7):607–615. doi:10.1016/j.biopsych.2014.07.012.
J Gastroenterol. 2011;106(5):915. doi:10.1038/ajg.2010.480. 99. Barton W, Penney NC, Cronin O, Garcia-Perez I,
88. Wu MH, Lee CP, Hsu SC, Chang CM, Chen CY. Molloy MG, Holmes E, Shanahan F, Cotter PD,
Effectiveness of high-intensity interval training on the O’Sullivan O. The microbiome of professional athletes
mental and physical health of people with chronic differs from that of more sedentary subjects in composi-
schizophrenia. Neuropsychiatr Dis Treat. tion and particularly at the functional metabolic level. Gut.
2015;11:1255. doi:10.2147/NDT.S81482. 2018;67(4):625–633. doi:10.1136/gutjnl-2016-313627.
89. McNeil JK, LeBlanc EM, Joyner M. The effect of exer- 100. Macfarlane GT, Macfarlane S. Bacteria, colonic fer-
cise on depressive symptoms in the moderately mentation, and gastrointestinal health. J AOAC Int.
depressed elderly. Psychol Aging. 1991;6(3):487. 2012;95:50–60.
doi:10.1037/0882-7974.6.3.487. 101. Estaki M, Pither J, Baumeister P, Little JP, Gill SK,
90. Quaney BM, Boyd LA, McDowd JM, Zahner LH, He J, Ghosh S, Ahmadi-Vand Z, Marsden KR, Gibson DL.
Mayo MS, Macko RF. Aerobic exercise improves cog- Cardiorespiratory fitness as a predictor of intestinal
nition and motor function poststroke. Neurorehab microbial diversity and distinct metagenomic
Neural Repar. 2009;23(9):879–885. doi:10.1177/ functions. Microbiome. 2016;4(1):42. doi:10.1186/
1545968309338193. s40168-016-0189-7.
GUT MICROBES 567
102. Ohira H, Tsutsui W, Fujioka Y. Are short chain fatty the beneficial effects of Roux-en-Y gastric bypass on
acids in gut microbiota defensive players for inflamma- type 2 diabetes. Surg Obes Relat Dis. 2018;14
tion and atherosclerosis? J Atheroscler Thromb. (5):584–593. doi:10.1016/j.soard.2018.01.012.
2017;24(7):660–672. doi:10.5551/jat.RV17006. 114. Allen JM, Mailing LJ, Niemiro GM, Moore R,
103. Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Cook MD, White BA, Holscher HD, Woods JA.
Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Exercise alters gut microbiota composition and func-
Jakobshagen K, Buch T, et al. Host microbiota con- tion in lean and obese humans. Med Sci Sports Exerc.
stantly control maturation and function of microglia in 2018;50:747–757. doi:10.1249/MSS.0000000000001495.
the CNS. Nat Neurosci. 2015;18(7):965–977. 115. Donohoe DR, Garge N, Zhang X, Sun W,
doi:10.1038/nn.4030. O’Connell TM, Bunger MK, Bultman SJ. The micro-
104. Kang SS, Jeraldo PR, Kurti A, Miller MEB, Cook MD, biome and butyrate regulate energy metabolism and
Whitlock K, Goldenfeld N, Woods JA, White BA, autophagy in the mammalian colon. Cell Metab.
Chia N, et al. Diet and exercise orthogonally alter the 2011;13(5):517–526. doi:10.1016/j.cmet.2011.02.018.
gut microbiome and reveal independent associations 116. M Astbury S, Corfe BM. Uptake and metabolism of the
with anxiety and cognition. Mol Neurodegener. short-chain fatty acid butyrate, a critical review of the
2014;9(1):36. doi:10.1186/1750-1326-9-36. literature. Curr Drug Metab. 2012;13:815–821.
105. Evans CC, LePard KJ, Kwak JW, Stancukas MC, 117. Wolin KY, Yan Y, Colditz GA, Lee I. Physical activity
Laskowski S, Dougherty J, Moulton L, Glawe A, and colon cancer prevention: a meta-analysis. Br
Wang Y, Leone V, et al. Exercise prevents weight J Cancer. 2009;100(4):611. doi:10.1038/sj.bjc.6604917.
gain and alters the gut microbiota in a mouse model 118. Yoo DY, Kim W, Nam SM, Kim DW, Chung JY, Choi SY,
of high fat diet-induced obesity. PLoS One. 2014;9(3): Yoon YS, Won MH, Hwang IK. Synergistic effects of
e92193. doi:10.1371/journal.pone.0092193. sodium butyrate, a histone deacetylase inhibitor, on increase
106. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial of neurogenesis induced by pyridoxine and increase of
ecology: human gut microbes associated with obesity. neural proliferation in the mouse dentate gyrus.
Nature. 2006;444(7122):1022–1023. doi:10.1038/ Neurochem Res. 2011;36(10):1850. doi:10.1007/s11064-
4441022a. 011-0501-7.
107. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, 119. Kim HJ, Leeds P, Chuang DM. The HDAC inhibitor,
Falony G, Almeida M, Arumugam M, Batto J-M, sodium butyrate, stimulates neurogenesis in the
Kennedy S, et al. Richness of human gut microbiome ischemic brain. J Neurochem. 2009;110(4):1226–1240.
correlates with metabolic markers. Nature. 2013;500 doi:10.1111/j.1471-4159.2009.06212.x.
(7464):541–546. doi:10.1038/nature12506. 120. Cronin O, Barton W, Skuse P, Penney NC, Garcia-Perez I,
108. Queipo-Ortuño MI, Seoane LM, Murri M, Pardo M, Murphy EF, Woods T, Nugent H, Fanning A, Melgar S,
Gomez-Zumaquero JM, Cardona F, Casanueva F, et al. A prospective metagenomic and metabolomic analysis
Tinahones FJ, Sanz Y. Gut microbiota composition in of the impact of exercise and/or whey protein supplementa-
male rat models under different nutritional status and tion on the gut microbiome of sedentary adults. mSystems.
physical activity and its association with serum leptin 2018;3(3):e00044–18. doi:10.1128/mSystems.00044-18.
and ghrelin levels. PLoS One. 2013;8(5):e65465. 121. Rottenberg J. Cardiac vagal control in depression:
doi:10.1371/journal.pone.0065465. a critical analysis. Biol Psychol. 2007;74(2):200–211.
109. Barcenilla A, Pryde SE, Martin JC, Duncan SH, doi:10.1016/j.biopsycho.2005.08.010.
Stewart CS, Henderson C, Flint HJ. Phylogenetic rela- 122. Gershon MD. Nerves, reflexes, and the enteric nervous
tionships of butyrate-producing bacteria from the system: pathogenesis of the irritable bowel syndrome.
human gut. Appl Environ Microbiol. J Clin Gastroenterol. 2005;39:S184–S193.
2000;66:1654–1661. 123. Vempati R, Telles S. Yoga-based guided relaxation
110. Boakes RA. Self-starvation in the rat: running versus reduces sympathetic activity judged from baseline
eating. Span J Psychol. 2007;10:251–257. levels. Psychol Rep. 2002;90(2):487–494. doi:10.2466/
111. Zhao X, Zhang Z, Hu B, Huang W, Yuan C, Zou L. pr0.2002.90.2.487.
Response of gut microbiota to metabolite changes 124. Schumann D, Anheyer D, Lauche R, Dobos G,
induced by endurance exercise. Front Microbiol. Langhorst J, Cramer H. Effect of yoga in the therapy
2018;9:765. doi:10.3389/fmicb.2018.00765. of irritable bowel syndrome: a systematic review. Clin
112. Martinez I, Wallace G, Zhang C, Legge R, Benson AK, Gastroenterol Hepatol. 2016;14(12):1720–1731.
Carr TP, Moriyama EN, Walter J. Diet-induced meta- doi:10.1016/j.cgh.2016.04.026.
bolic improvements in a hamster model of hypercho- 125. Cramer H, Lauche R, Langhorst J, Dobos G. Yoga for
lesterolemia are strongly linked to alterations of the gut depression: A systematic review and meta-analysis.
microbiota. Appl Environ Microbiol. 2009;75 Depress Anxiety. 2013;30(11):1068–1083. doi:10.1002/
(12):4175–4184. doi:10.1128/AEM.00380-09. da.22166.
113. Liu H, Zhang H, Wang X, Yu X, Hu C, Zhang X. The 126. Perez-Burgos A, Wang B, Mao Y-K, Mistry B,
family Coriobacteriaceae is a potential contributor to Neufeld K-AM, Bienenstock J, Kunze W.
568 A. DALTON ET AL.
Psychoactive bacteria Lactobacillus rhamnosus (JB-1) hippocampal neurogenesis. Cell Rep. 2016;15
elicits rapid frequency facilitation in vagal afferents. (9):1945–1956. doi:10.1016/j.celrep.2016.04.074.
Am J Physiol Gastrointest Liver Physiol. 2012;304(2): 136. Cerdá B, Pérez M, Pérez-Santiago JD, Tornero-
G211–G220. doi:10.1152/ajpgi.00128.2012. Aguilera JF, González-Soltero R, Larrosa M. Gut
127. Bravo JA, Forsythe P, Chew MV, Escaravage E, microbiota modification: another piece in the puzzle
Savignac HM, Dinan TG, Bienenstock J, Cryan JF. of the benefits of physical exercise in health? Front
Ingestion of Lactobacillus strain regulates emotional Physiol. 2016;7:51. doi:10.3389/fphys.2016.00051.
behavior and central GABA receptor expression in 137. Cook MD, Martin SA, Williams C, Whitlock K,
a mouse via the vagus nerve. Proc Natl Acad Sci. Wallig MA, Pence BD, Woods JA. Forced treadmill
2011;108(38):16050–16055. doi:10.1073/ exercise training exacerbates inflammation and causes
pnas.1102999108. mortality while voluntary wheel training is protective
128. Colcombe S, Kramer AF. Fitness effects on the cogni- in a mouse model of colitis. Brain Behav Immun.
tive function of older adults: a meta-analytic study. 2013;33:46–56. doi:10.1016/j.bbi.2013.05.005.
Psychol Sci. 2003;14(2):125–130. doi:10.1111/1467- 138. Lamoureux EV, Grandy SA, Langille MGI. Moderate
9280.t01-1-01430. exercise has limited but distinguishable effects on the
129. Kramer AF, Colcombe SJ, McAuley E, Scalf PE, mouse microbiome. mSystems. 2017;2:4. doi:10.1128/
Erickson KI. Fitness, aging and neurocognitive mSystems.00006-17.
function. Neurobiol Aging. 2005;26(1):124–127. 139. Allen JM, Berg Miller ME, Pence BD, Whitlock K,
doi:10.1016/j.neurobiolaging.2005.09.009. Nehra V, Gaskins HR, White BA, Fryer JD,
130. Rasmussen P, Brassard P, Adser H, Pedersen MV, Woods JA. Voluntary and forced exercise differentially
Leick L, Hart E, Secher NH, Pedersen BK, alters the gut microbiome in C57BL/6J mice. J Appl
Pilegaard H. Evidence for a release of brain-derived Physiol. 2015;118(8):1059–1066. doi:10.1152/
neurotrophic factor from the brain during exercise. japplphysiol.01077.2014.
Exp Physiol. 2009;94(10):1062–1069. doi:10.1113/ 140. Lou De Santis G, Kavvadia M, Abd Almajeed
expphysiol.2009.048512. Abbaas Alwardat N, Bigioni G, Zeppieri C, Cascapera S,
131. Molteni R, Wu A, Vaynman S, Ying Z, Barnard R, De Lorenzo A. Psychobiotics as integrative therapy for
Gomez-Pinilla F. Exercise reverses the harmful effects of neuropsychiatric disorders with special emphasis on the
consumption of a high-fat diet on synaptic and beha- microbiota-gut-brain axis. Biomed Prev. 2017;2:111.
vioral plasticity associated to the action of brain-derived 141. Dey S, Singh R, Dey P. Exercise training: significance of
neurotrophic factor. Neuroscience. 2004;123:429–440. regional alterations in serotonin metabolism of rat
132. Linnarsson S, Björklund A, Ernfors P. Learning deficit brain in relation to antidepressant effect of exercise.
in BDNF mutant mice. Eur J Neurosci. Physiol Behav. 1992;52:1095–1099.
1997;9:2581–2587. 142. Wipfli B, Landers D, Nagoshi C, Ringenbach S. An
133. Sarkar A, Lehto SM, Harty S, Dinan TG, Cryan JF, examination of serotonin and psychological variables
Burnet PW. Psychobiotics and the manipulation of in the relationship between exercise and mental health.
bacteria–gut–brain signals. Trends Neurosci. 2016;39 Scand J Med Sci Sports. 2011;21(3):474–481.
(11):763–781. doi:10.1016/j.tins.2016.09.002. doi:10.1111/j.1600-0838.2009.01049.x.
134. Smith F, Clark JE, Overman BL, Tozel CC, Huang JH, 143. Özoğul F, Kuley E, Özoğul Y, Özoğul İ. The function of
Rivier JE, Blikslager AT, Moeser AJ. Early weaning lactic acid bacteria on biogenic amines production by
stress impairs development of mucosal barrier function food-borne pathogens in arginine decarboxylase broth.
in the porcine intestine. Am J Physiol Gastrointest Food Sci Technol Res. 2012;18(6):795–804.
Liver Physiol. 2009;298(3):G352–G363. doi:10.1152/ doi:10.3136/fstr.18.795.
ajpgi.00081.2009. 144. Özoğul F. Production of biogenic amines by
135. Möhle L, Mattei D, Heimesaat MM, Bereswill S, Fischer A, Morganella morganii, Klebsiella pneumoniae and
Alutis M, French T, Hambardzumyan D, Matzinger P, Hafnia alvei using a rapid HPLC method. Eur Food
Dunay IR, et al. Ly6Chi monocytes provide a link between Res Technol. 2004;219(5):465–469. doi:10.1007/s00217-
antibiotic-induced changes in gut microbiota and adult 004-0988-0.