Content uploaded by Thomas W Buford
Author content
All content in this area was uploaded by Thomas W Buford on Jan 24, 2019
Content may be subject to copyright.
Downloaded from https://journals.lww.com/acsm-essr by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3ne1tVsBOHA7NfuNkkALWNVlMWCz+c/f1KPgNpeVV7nk= on 01/24/2019
Downloadedfromhttps://journals.lww.com/acsm-essr by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3ne1tVsBOHA7NfuNkkALWNVlMWCz+c/f1KPgNpeVV7nk= on 01/24/2019
Exercise and Sport Sciences Reviews articles in the Published Ahead-of-Print section have been peer-reviewed and
accepted for publication. However, during copyediting, page composition, or proof review changes may be made
that could affect the content.
Copyright © 2019 by the American College of Sports Medicine www.acsm-essr.org
ESSR Published Ahead-of-Print
Heading: Perspectives for Progress
Accepted: 01/18/2019
Editor: Marni D. Boppart, ScD, FACSM
Exercise and the gut microbiome: a review of the evidence, potential mechanisms, and
implications for human health
Lucy J. Mailing1, Jacob M. Allen2, Thomas W. Buford3, Christopher J. Fields4, Jeffrey A.
Woods1,5
1 Division of Nutritional Sciences, University of Illinois Urbana-Champaign, IL
2 Center for Microbial Pathogenesis, Nationwide Children‘s Hospital, Columbus, OH
3 Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
4 High Performance Computing in Biology, Carver Biotechnology Center, University of Illinois
Urbana-Champaign, IL
5 Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign,
IL
ACCEPTED
Exercise and the gut microbiome: a review of the evidence, potential
mechanisms, and implications for human health
Lucy J. Mailing1, Jacob M. Allen2, Thomas W. Buford3, Christopher J. Fields4, Jeffrey A.
Woods1,5
1 Division of Nutritional Sciences, University of Illinois Urbana-Champaign, IL, United States
2 Center for Microbial Pathogenesis, Nationwide Children‘s Hospital, Columbus, OH, United
States
3 Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United
States
4 High Performance Computing in Biology, Carver Biotechnology Center, University of Illinois
Urbana-Champaign, IL, United States
5 Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign,
IL, United States
Corresponding Author:
Jeffrey A. Woods, PhD
1206 South 4th Street
1008A Khan Annex, Huff Hall
University of Illinois at Urbana-Champaign
Champaign IL 61820
Woods1@illinois.edu
Exercise and Sport Sciences Reviews, Publish Ahead of Print
DOI: 10.1249/JES.0000000000000183
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
Funding: The authors have no funding to disclose for the preparation of this manuscript. TB‘s
effort toward this manuscript partially supported by grants from the National Institutes of Health
(P2CHD086851 and P30AG050886).
Conflicts of interest: The authors have no conflicts of interest to report.
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
ABSTRACT
The gastrointestinal tract contains trillions of microbes (collectively known as the gut
microbiota) that play essential roles in host physiology and health. Studies from our group and
others have demonstrated that exercise independently alters the composition and functional
capacity of the gut microbiota. Here, we review what is known about the gut microbiota, how it
is studied, how it is influenced by exercise training, and discuss the potential mechanisms and
implications for human health and disease.
KEYWORDS: Endurance exercise; gut microbiota; short-chain fatty acids; butyrate; gut health;
inflammatory bowel disease
KEY POINTS
The trillions of microbes in the gut play essential roles in human health
Exercise training alters the composition and functional capacity of the gut microbiota,
independent of diet
Exercise-induced alterations of the gut microbiota may depend on obesity status, exercise
modality, and exercise intensity.
Exercise-induced alterations of the gut microbiota are likely to have numerous benefits
for human health
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
INTRODUCTION
Scientists have only recently begun to appreciate the human gut as a complex ecosystem of
bacteria, archaea, eukaryotes, and viruses that have co-evolved with humans over thousands of
years. Known collectively as the gut microbiota, these microbes can weigh up to two kilograms
and are imperative to host digestion, metabolic function, and resistance to infection (1). The
human gut microbiota has an enormous metabolic capacity, with over 1000 different unique
bacterial species and over 3 million unique genes (2). Collectively, the sum of the microbial
genes in the gut is called the gut microbiome.
Given the numerous roles of the gut microbiota in host physiology and pathophysiology, it‘s not
surprising that there is great interest in identifying ways to manipulate microbial communities in
health and disease (3–5). While diet is well known to modulate the composition of the gut
microbiota, recent studies suggest that exercise can alter gut microbial communities as well. This
will be the focus of the present review. Key questions include: Does exercise independently alter
the gut microbiota? If yes, by what mechanism? With what implications for the gut and other
organ systems? Can exercise beneficially modulate the gut microbiota in states of disease?
Before we attempt to answer these questions, we will first review advances in technology that
have improved the understanding of the microbiota‘s contribution to health and disease and
enabled investigations into exercise‘s impact on gut microbial communities.
BASIC MICROBIOME METHODOLOGY
Until the 1990s, scientific study of gut microbes primarily relied on culture, staining, and
microscopy (6). Growth media and conditions typically favored fast-growing, aerobic microbes,
meaning that many anaerobic microbes could not be effectively cultured or studied (7). This
changed with the advent of DNA sequencing. 16S bacterial ribosomal RNA (rRNA) gene
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
sequencing (hereafter 16S) quickly became the most popular (8). The conserved regions of this
gene are used to design broad-spectrum PCR primers that allow for the amplification of the more
rapidly-evolving hypervariable regions across a broad spectrum of microbes (Figure 1). The
resulting amplified hypervariable region sequences can then be classified taxonomically by
comparing them to a curated database of fully sequenced bacterial 16S genes (9).
16S is still the most widely used method to cost-effectively characterize bacterial communities in
a research setting (10), but it does have several limitations. First, taxonomic classifications are
limited primarily to bacteria. Second, sequence classification is normally limited to the genus
level, as multiple species may have the same sequence within the studied hypervariable region
(11). 16S is also susceptible to primer bias (12,13). Finally, 16S analysis does not provide direct
information about the function of gut microbes or the potential interactions with host physiology,
though tools such as PICRUSt (Phylogenetic Investigation of Communities by Reconstruction of
Unobserved States) can infer potential functional pathways from 16S results utilizing
information from the Human Microbiome Project (14).
TARGETED GENOMICS, META-OMICS, AND METABOLITES
Recently, interest has increased for moving beyond 16S (‗who‘s there?‘) to better understand the
role of the gut microbiota in states of health and disease (‗what are they doing?‘). One way to do
this involves using specific degenerate primers to perform quantitative PCR (qPCR) for a
specific conserved microbial gene. For example, our lab has targeted the butyryl-CoA:acetate-
CoA transferase (BCoAT) gene, which encodes the primary enzyme involved in gut bacterial
production of the short-chain fatty acid (SCFA) butyrate (15). This targeted genomics approach
is relatively quick and inexpensive but limited in overall scope.
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
In contrast, metagenomics (i.e. shotgun sequencing) involves assessing the entire gene content of
a given microbial community (16) to assess microbial functions and allow for identification of
bacteria, archaea, viruses, and fungi with greater specificity (2,17). Current limitations for
metagenomics include high cost, sequencing biases, complexity in both data processing and
analysis, and incomplete databases for taxonomy and genomic assignment (16). Nevertheless,
newer sequencing technologies as well as upgraded downstream pipelines (18,19) and reference
databases (20,21) have greatly improved taxonomic and functional genomic profiling of the gut
microbiota. Other meta-omics, such as meta-transcriptomics, -proteomics, and -metabolomics
can help elucidate which genes are actually expressed and become functional proteins capable of
carrying out diverse metabolic functions (22). These high-throughput, high-resolution techniques
are rapidly improving in speed, quality, and cost, and will soon be the norm for microbiome
research.
Metabolites can also be measured directly using gas or liquid chromatography and mass
spectrometry to provide insight into the collective metabolic function of the gut. However,
quantitation of certain volatile metabolites requires prompt collection and acidification or ethanol
treatment of samples after collection (23). Moreover, the fecal concentration of many gut
metabolites will depend on gut transit time, cross-feeding interactions between microbes, and
rate of host absorption (24), so it is not necessarily representative of luminal concentrations.
STATISTICAL INTERPRETATION OF GUT MICROBIOME DATA
Microbiome data analysis is typically performed using an open-source bioinformatics pipeline,
such as Quantitative Insights Into Microbial Ecology 2 (QIIME 2) (25) or Mothur (26) for 16S
and Metagenomics Reports (METAREP) (27), Metagenome Analyzer (MEGAN) (28), or
Metagenomic Rapid Annotations using Subsystems Technology (MG-RAST) (29) for
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
comparative metagenomics. For 16S, sequences from samples must be de-multiplexed, quality
filtered, and clustered into operational taxonomic units (OTUs) based on sequence identity (ID)
(i.e. about 95% ID for genus; 97% ID for species). These are then taxonomically classified using
common reference databases (30,31) and visualized as a phylogenetic tree or represented using
bar plots.
The alpha ()-diversity metrics Chao1, Shannon index, and Simpson index are a measure of the
diversity within a sample and take into account both the number of unique OTUs in a sample
(richness) and the relative abundance of these OTUs (evenness). In contrast, beta ()-diversity
metrics like Bray-Curtis and UniFrac are measures of the diversity between samples. When more
than two samples are used, -diversity is calculated for every pair of samples to create a distance
(dissimilarity) matrix. The data present in the -diversity distance matrix can be visualized using
a 2-D or 3-D Principle Coordinates Analysis (PCoA) plot, where each axis explains a certain
percentage of variation present in the dataset. Each sample is represented by a single point, and
the distance between points reflects how compositionally different the samples are from one
another (32). Statistical significance of community-level differences can be assessed using a
PERMANOVA test (33).
FACTORS INFLUENCING THE GUT MICROBIOME
Using these methodologies, several factors have been identified that influence gut microbial
composition and metabolic capacity beginning at birth. The fetal gut contains few if any
microbes as the womb is largely sterile (34). Microbial colonization begins at birth and is
significantly influenced by mode of delivery (vaginal or Cesarean section) and infant diet
(breastmilk or formula) (35). Other factors, including increased sanitation, reduced exposure to
infection through vaccination, elimination of entero-pathogens, and exposure to antibiotics and
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
non-antibiotic drugs can also alter the commensal, or native, microbiota (36). Dietary intake also
has a significant impact on microbial composition throughout life (37).
EXERCISE AND THE GUT MICROBIOME: EVIDENCE IN ANIMAL STUDIES
Emerging research from our group and others suggests that exercise also influences the gut
microbiota. Over a dozen controlled animal studies have shown that exercise training
independently alters the composition and functional capacity of the gut microbiota (38–51).
Matsumoto et al. (2008) was the first to find that five weeks of exercise training resulted in an
increase in the bacterial metabolite butyrate. Several other studies have recapitulated this finding
and shown that exercise training increases the relative abundance of butyrate-producing taxa
(38,41). Butyrate is a short-chain fatty acid (SCFA) produced from the bacterial fermentation of
dietary fiber. As the primary fuel for colonocytes, butyrate has been shown to increase colonic
epithelial cell proliferation, promote gut barrier integrity, and regulate the host immune system
and gene expression (52,53).
Drawing other broad conclusions as to how and to what degree exercise alters the rodent gut
microbiota has proved difficult due to incongruities in diet, species/strain, animal age, and
exercise modality used. For instance, several studies suggest that exercise increases the ratio of
Firmicutes to Bacteroidetes phyla (38–40), while some studies suggest that exercise reduces this
ratio (41–44). Still others have found no change at the phylum level (45,46). Several factors may
influence the disparate results observed in these studies. For instance, we recently reported that
voluntary wheel running (VWR) and forced treadmill running (FTR) – the two most common
modes of endurance exercise used with rodents – differentially altered the gut microbiota (47).
Mika et al. (2015) found that microbial genera were more robustly altered by VWR in juvenile,
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
as compared to the adult rats (44), while Evans et al. (2014) found that VWR increased microbial
diversity, but only in mice fed a high-fat diet (41).
EXERCISE AND THE GUT MICROBIOME: HUMAN CROSS-SECTIONAL
EVIDENCE
Evidence for a role of exercise in shaping the human gut microbiota first emerged from cross-
sectional studies (Table 1). Clarke et al. (2014) found that the gut microbiota of professional
rugby players had greater alpha diversity and a higher relative abundance of 40 different
bacterial taxa than the gut microbiota of lean sedentary controls. The athletes also had lower
abundance of Bacteroides and Lactobacillus species (spp.) than their lean sedentary counterparts
(54). More recently, Bressa et al. (2017) compared active women to sedentary controls (55) and
observed that women who performed at least three hours of exercise per week had increased
levels of Faecalibacterium prausnitzii, Roseburia hominis, and Akkermansia muciniphila. F.
prausnitzii and R. hominis are known butyrate producers (56), while A. muciniphila has been
associated with a lean BMI and improved metabolic health (57).
Several studies have also attempted to correlate the composition and metabolic capacity of the
microbiota with cardiorespiratory fitness. Durk et al. (2018) showed that a higher ratio of
Firmicutes to Bacteroidetes, the two predominant phyla in the human gut microbiota, was
significantly correlated with maximal oxygen uptake (VO2 max) (58). Estaki et al. (2016) found
that in younger adults, microbial diversity and abundance of butyrate-producing bacterial taxa
were positively correlated with cardiorespiratory fitness (59), while Barton et al. (2017) showed,
using metagenomic analyses, that athletes have altered gut microbial pathways for amino acid
biosynthesis and carbohydrate metabolism and greater fecal SCFA concentrations (60).
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
Nevertheless, all of these studies were limited by their cross-sectional design and their inability
to control for the effects of diet (and perhaps other factors) on the gut microbiota. There is
considerable inter-individual variability in the composition of the microbiota, and active
individuals tend to eat differently from sedentary individuals. For instance, Clarke et al. (2014)
found that increased protein intake by elite rugby players accounted for many of the observed
differences in the gut microbiota (54). These limitations suggested the need for longitudinal
studies to determine whether exercise independently alters the gut microbiota in humans.
EXERCISE AND THE GUT MICROBIOME: HUMAN LONGITUDINAL STUDIES
Recently, members of our group published findings from the first controlled longitudinal study to
assess the effects of exercise on the gut microbiome (61). In total, 32 sedentary adults (lean [BMI
<25] or obese [BMI>30]) participated in a six-week supervised endurance exercise program (30-
60 minutes duration, 3x/week) with stringent dietary controls. Several taxa were differentially
altered by exercise depending on BMI status. For instance, exercise increased Faecalibacterium
spp. in lean subjects, but reduced its abundance in obese subjects; Bacteroides spp. decreased in
the lean subjects and increased in the obese subjects. Six weeks of exercise also increased the
abundance of butyrate-producing taxa and fecal acetate and butyrate concentrations, but only in
lean subjects. Interestingly, most bacterial taxa and SCFAs that increased with exercise
subsequently decreased during the six-week sedentary washout period that followed, indicating
that the effects of exercise on the microbiota were both transient and reversible.
Similarly, Cronin et al. (2018) sought to determine whether a short-term exercise regime, with or
without whey protein supplementation, could alter gut microbial composition and function in
predominantly overweight or obese male and female adults (n=90) (62). Those randomized to the
exercise groups were required to perform moderate intensity aerobic training (18-32 minutes
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
duration) and a progressive resistance training program three times per week for eight weeks.
Post-intervention assessment did not reveal any significant changes in taxonomic composition or
metabolic pathways in either exercise group compared to baseline. However, a trend was seen
for an increase in bacterial diversity in the exercise and exercise + whey protein groups,
compared to the group that received whey protein alone. Metagenomic and metabolomic
analyses revealed only modest alterations of microbial metabolism. While the study had a fairly
large sample size, the authors note that self-reported maintenance of usual dietary intake and a
wide BMI range may have prevented detection of more significant changes.
Munukka et al. (2018) performed a similar study to determine whether endurance exercise could
affect the gut metagenome in previously sedentary overweight women (n=17) (63). Six-weeks of
light to moderate intensity cycling resulted in an increased relative abundance of Akkermansia
muciniphila and a decrease in Proteobacteria. Most interestingly, only about half of the subjects‘
microbiomes responded considerably to exercise. Metagenomic analysis revealed that exercise
training decreased the abundance of several genes related to fructose and amino acid metabolism.
Together, these findings suggest that exercise has independent effects on the gut microbiota, but
longer duration or higher intensity aerobic training may be required to induce significant
taxonomic and metagenomic changes. Furthermore, the microbiota of lean individuals may be
more responsive to an exercise intervention than that of overweight or obese individuals.
POTENTIAL MECHANISMS
There are several potential mechanisms by which exercise might alter the gut microbiota (Figure
2). The gut-associated lymphoid tissue, or GALT, lies through the small and large intestine and
contains about 70 percent of the body‘s immune cells. Several animal studies performed by
Hoffman-Goetz et al. have found that exercise alters the gene expression of intraepithelial
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
lymphocytes, downregulating pro-inflammatory cytokines and upregulating anti-inflammatory
cytokines and antioxidant enzymes (64–66). These immune cells reside in close proximity to
microbial communities and produce antimicrobial factors that are essential for mediating host-
microbial homeostasis (67). Similarly, exercise may impact the integrity of the gut mucus layer,
which plays important role in keeping microbes from adhering to the gut epithelium and serves
as an important substrate for certain mucosa-associated bacteria, such as Akkermansia
muciniphila.
Exercise raises core temperature and results in heat stress, particularly when performed for long
durations or in a hot environment (68). Exercise can also reduce intestinal blood flow by more
than 50 percent, with significant gut ischemia occurring within 10 minutes of high-intensity
exercise (69). Upon rest, the splanchnic bed undergoes rapid reperfusion. While the intestine is
an anaerobic environment, gut epithelial cells primarily utilize oxidative metabolism, and high-
intensity exercise is known to transiently impair gut barrier function (69,70). Thus, exercise-
induced heat stress and ischemia may briefly result in more direct contact between the gut
mucosal immune system and the microbes that reside in the gut lumen and mucosa, with
potential consequences for gut microbial communities.
While intestinal permeability occurs briefly during acute exercise, contact between microbes and
the immune system may be reduced at rest with regular physical activity. Trained athletes have
lower levels of circulating bacterial endotoxin lipopolysaccharide (LPS) at rest than sedentary
individuals (71), and a greater heat shock protein response to heat stress (72). Increased heat
shock proteins in the gut have been shown to prevent breakdown of tight junction proteins
between epithelial cells (73). Thus, it‘s plausible that exercise represents a hormetic stressor to
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
the gut that stimulates beneficial adaptations and improves the long-term resilience of the gut
barrier.
Altered gut motility and/or activity of the enteric nervous system is another mechanism by which
exercise may influence the gut microbiome. Exercise reduces transit time in the large intestine
and has been shown to accelerate the movement of gas through the gastrointestinal (GI) tract
(74,75). Exercise is also well known to impact the autonomic nervous system increasing vagal
and overall sympathetic tone (76), but it‘s impact on the complex mesh-like network of neurons
that innervate the gut has not been well elucidated. Nonetheless, regional or global changes in GI
transit are likely to have profound effects on intestinal pH, mucus secretion, biofilm formation,
and availability of nutrients to microbes. Mechanical forces are also increased in the abdomen
during most forms of aerobic exercise, which could potentially influence gut motility or increase
the mixing of intestinal contents.
Exercise training may also alter the entero-hepatic circulation of bile acids. Meissner et al.
(2011) found that hypercholesterolemic mice that were given access to a running wheel for 12
weeks displayed increased bile acid secretion and increased fecal bile acid outputs compared to
hypercholesterolemic mice that remained sedentary (77). Bile acids are potent regulators of gut
microbiota community structure, and an absence of these molecules is associated with significant
alterations in gut microbial communities (i.e. gut dysbiosis) (78). Thus, changes in the bile acid
pool could significantly shift the gut microbiome with exercise.
Lastly, exercise significantly alters metabolic flux (the rate of turnover of molecules through
metabolic pathways) and requires contraction of skeletal muscle, which stimulates the release of
myokines, metabolites, and neuroendocrine hormones that may interact with the gut directly or
indirectly through a common interface with the immune system (79). Significant amounts of
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
lactate are released into the blood during exercise, which could alter intestinal pH if any of this
lactate is secreted into the gut lumen. Overall, more research is needed to determine which of
these mechanisms are responsible for the adaptation of the gut microbiota to exercise training.
IMPLICATIONS FOR THE GUT AND BEYOND
Exercise-induced alterations of the gut microbiota likely have implications for gut and whole-
body health. Physical activity has been shown to be protective against many chronic diseases and
offers an attractive and cost-effective way to improve quality of life (80). Though under-
recognized to date, many of these benefits may be derived via interactions with the gut
microbiota (Figure 3). Here, we discuss potential example conditions for which the gut
microbiota may play a pivotal role, though they almost certainly do not represent the full
spectrum of potential benefits. It should also be noted that in most cases the potential attribution
of beneficial effects to the gut microbiota remains speculative due to the lack of definitive data in
this area.
Colorectal cancer
Observational studies indicate that physically active individuals have a 24% reduced risk for
colorectal cancer compared to sedentary individuals (23). Beginning an exercise program after
the onset of colorectal cancer may also improve quality of life and reduce overall mortality (82).
In preclinical animal studies, VWR has been shown to reduce colon tumor incidence (83). One
mechanism for this may be increased butyrate production from exercise. Colorectal cancer
patients have been shown to have an altered gut microbiota characterized by a reduced
abundance of butyrate-producing taxa, including Roseburia and Lachnospiraceae (84).
In vitro studies have shown that butyrate differentially regulates gene expression in healthy and
cancerous cells (85). In healthy epithelial cells, butyrate is rapidly metabolized via the
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
mitochondrial tricarboxylic acid (TCA) cycle. This results in a buildup of cytosolic citrate and
acetyl CoA and increases the acetylation of histones by histone acetyltransferases (HATs). This
epigenetic modification increases expression of genes involved in cell proliferation and cell
turnover, effectively strengthening the intestinal barrier (85).
In colorectal cancer cells, however, mitochondrial dysfunction results in an accumulation of
butyrate in the cytosol. Free butyrate inhibits histone deacetylases (HDACs), which results in the
epigenetic suppression of proliferation and promotion of cell death pathways (85). This may
ultimately lead to a reduction in tumor size and reduces the chance of metastasis. Indeed,
Basterfield & Mathers et al. (2010) found that Min mice, which are genetically predisposed to
intestinal adenomas, had a reduced number of large tumors in the colon and a trend towards
reduced tumor multiplicity with exercise training. There was a weak correlation between fecal
butyrate concentrations and tumor number (86).
Inflammatory bowel disease
Inflammatory bowel disease (IBD) includes both Crohn‘s disease and ulcerative colitis (UC) and
is characterized by inappropriate gut immune responses and an altered microbiota. IBD patients
have an increased relative abundance of Enterobacteriaceae and reduced abundance of
Roseburia, a genus known to produce butyrate and induce regulatory T cell formation (87).
Regulatory T cells are important for modulating the immune system, promoting tolerance to self-
antigens, preventing autoimmune disease, and dampening inflammation. Metagenomic analysis
also revealed reduced carbohydrate metabolism and amino acid biosynthesis in the fecal
microbiome of IBD patients compared to healthy controls (87) – two pathways that exercise has
been reported to increase. Indeed, higher self-reported physical activity levels are associated with
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
a 22 percent reduced risk of active UC (88), and a ten week intervention that included moderate
exercise improved quality of life in patients with moderately active UC (89).
Our group and others have performed several preclinical animal studies on the effects of exercise
on colitis. Szalai et al. (2014) found that six weeks of VWR increased expression of heme
oxygenase and nitric oxide synthase, increased anti-inflammatory cytokines, and reduced
inflammatory markers and the severity of mucosal damage in TNBS-induced colitis (90), while
Liu et al. (2015) found that one month of VWR suppresses pro-inflammatory cytokine
production in response to dextran sodium sulfate (DSS)-induced colitis by up-regulating
glucocorticoid-mediated peroxisome proliferator-activated receptor gamma (PPAR‐γ)
expression in the colon (91). PPAR‐γ regulates fatty acid storage and glucose metabolism. In
2013, members of our group confirmed that VWR conferred protection against DSS-induced
colitis and reduced disease-related symptoms and mortality, but additionally observed that FTR
exacerbated symptoms and led to higher mortality (92). Further study revealed that VWR and
FTR resulted in distinct changes in the gut microbiota (47).
To determine if exercise-induced alterations in the gut microbiota were directly responsible for
the protective effects of VWR, members of our group transferred cecal contents from exercised
or sedentary mice into naïve, sedentary germ-free mice in the first-ever ―exercise‖ fecal
microbiota transplant (FMT). When recipient mice were later subjected to an acute colitis insult
with DSS, those that had received a microbiota from exercised mice lost significantly less body
weight and had fewer clinical symptoms than those that received a microbiota from sedentary
mice. Mice receiving the microbiota from exercised mice also had a more regenerative cytokine
profile, with significantly higher levels of transforming growth factor beta (TGF-β), forkhead
box P3 (FoxP3), and interleukin (IL-22) gene expression in the distal colon (93). More studies
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
are needed to determine whether exercise can beneficially modulate the gut microbiota in
humans with IBD, and whether compositional alterations parallel improvements in
symptomology.
Obesity and metabolic disease
Several studies have shown that the gut microbiota is closely associated with obesity and
metabolic syndrome. A seminal paper by Turnbaugh et al. (2006) showed that transplanting fecal
material from an obese mouse into a germ-free mouse resulted in rapid weight gain (94). The
obese microbiota has a significantly higher capacity for energy harvest from the diet and may
also promote intestinal permeability, allowing the influx of endotoxin into the bloodstream.
Endotoxemia itself has been shown to result in weight gain and insulin resistance (95).
Evidence from animal studies suggests that exercise may attenuate the gut dysbiosis and altered
intestinal villi morphology induced from high-fat diet feeding (45). Queipo-Ortuño et al. (2013)
found that just six days of VWR increased the relative abundance of Lactobacillus and
Bifidobacterium spp. in male rats, which were positively correlated with serum leptin levels (42),
while Lambert et al. (2015) found significant interactions between exercise and diabetic state on
the gut microbiota in an animal model of type 2 diabetes (40).
Lai et al. (2018) showed that high-fat diet fed obese mice receiving FMT from exercised,
normal-fat diet fed donor mice showed improvements in metabolic parameters, including weight
loss, reduced fasting blood glucose, and lower hepatic expression of pro-inflammatory cytokines
(96). Notably, two of the taxa that were highly associated with FMT from exercised donors,
Odoribacter and AF12 of the family Rikenellaceae, are known butyrate-producers. In animal
models of obesity, butyrate has been shown to increase energy expenditure, improve insulin
sensitivity, and reduce adiposity (97). Butyrate and other SCFAs also stimulate the production of
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
satiety hormones, which help regulate food intake, and may help delay or attenuate the
development of diabetes by improving gut barrier function (98).
Mental and cognitive health
The gut microbiota has also been implicated in mental health and cognition, and the existence of
a gut-brain axis is well established (99,100). Gut microbiota-derived metabolites have been
shown to activate receptors on vagal afferents of the enteric nervous system, and certain
microbes are also capable of producing neurotransmitters; for example, Lactobacillus spp. can
produce both serotonin and gamma-aminobutyric acid (GABA) (101). Serotonin is thought to
play a role in emotion and cognitive functions, and low levels have been linked to depression.
GABA is the chief inhibitory neurotransmitter in the central nervous system and typically has
anti-anxiety and relaxant effects. Thus, it‘s no surprise that germ-free mice that lack a
commensal microbiota exhibit altered brain function, abnormal behaviors, and an exaggerated
hypothalamic-pituitary-adrenal response to stress (102).
Gut dysbiosis may also contribute to impaired mental health. Human patients with major
depressive disorder have an altered gut microbiota, characterized by changes in the relative
abundance of Firmicutes, Bacteroidetes, and Actinobacteria (103). Notably, transferring fecal
material from these patients into germ-free mice confers depression-like behaviors in the
recipient mice (103). Stevens et al. (2017) found that patients with a depressive or anxiety
disorder had a unique predicted gut metagenomic profile and increased levels of plasma markers
of intestinal permeability (104).
Exercise is well-known to have benefits for mental and neurological health (105), and it is
plausible that some of the beneficial effects of exercise on the brain are mediated by the gut
microbiota. For instance, Kang et al. (2014) found that an hour of daily wheel running increased
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
the relative abundance of Lachnospiraceae, a family of known butyrate-producing microbes,
which was negatively correlated with anxiety-like behavior in adult C57Bl/6J mice (38).
Butyrate itself has been shown to upregulate brain-derived neurotrophic factor (BDNF)
expression in the hippocampus and frontal cortex of mice, which helps to support the survival of
existing neurons and encourage the formation of new neurons and synapses. Butyrate has also
been shown to regulate the activation of microglial cells, a specialized population of immune
cells in the brain (106,107). Like exercise, butyrate also appears to increase neuroplasticity and
has anti-depressant activity, boosting brain serotonin levels (108).
FUTURE PERSPECTIVES
Overall, increasing evidence suggests that regular aerobic exercise confers benefits to the gut
microbiota, which may be partially responsible for the widespread benefits of regular physical
activity on human health. This area of research will no doubt have many exciting developments
in the coming decade, and there are many questions that are yet be answered (Figure 2). In
addition to elucidating the mechanisms involved, the effects of different forms of exercise
necessitates further study. Open questions include: ―What frequency, mode, or intensity of
exercise is best? How does exercise impact the gut microbiome in children or the elderly? In
healthy or diseased states? How does exercise interact with diet in shaping the gut microbiome?
Do probiotics or prebiotics influence gut responses to an exercise intervention? What about
resistance exercise?‖
Future research should also employ methodologies to elucidate the effects of exercise on the
microbiome in various regions of the GI tract, including microbes associated with the gut mucus
layer. While this will likely involve more invasive endoscopic procedures for human studies, it is
critical to understand the true dynamics of the gut environment. A recent study by Zmora et al.
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
(2018) suggests that fecal samples often under- or over-represent the relative abundance of
various bacterial genera and species in the human gut (109).
While we have learned a great deal about how exercise influences bacterial communities, future
research should also seek to understand how exercise influences archaea, fungi, and viruses in
the human gut, and how exercise influences gut competition and ecological patterns. The
increasing feasibility of metagenomic studies will also help to elucidate which bioactive
metabolites produced by the gut microbiota might be most affected by exercise training.
Gnotobiotic, or germ-free, animal studies will also help to determine how exercise-induced
alterations in the gut microbiota are causally linked to alterations in disease risk. Ultimately, we
can imagine a future of personalized microbiome-based lifestyle medicine, where baseline gut
microbiota, diet, and other host factors might help predict which exercise program might be most
effective for a given individual.
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
ACKNOWLEDGEMENTS
TB‘s effort toward this manuscript partially supported by grants from the National Institutes of
Health (P2CHD086851 and P30AG050886).
Funding: The authors have no funding to disclose for the preparation of this manuscript.
Conflicts of interest: The authors have no conflicts of interest to report.
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
REFERENCES
1. Brestoff JR, Artis D. Commensal bacteria at the interface of host metabolism and the
immune system. Nature Immunology [Internet]. 2013 Jul [cited 2018 Aug 5];14(7):676–84.
Available from: https://www.nature.com/articles/ni.2640
2. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut
microbial gene catalogue established by metagenomic sequencing. Nature [Internet]. 2010
Mar [cited 2018 Aug 5];464(7285):59–65. Available from:
https://www.nature.com/articles/nature08821
3. Marchesi JR, Adams DH, Fava F, Hermes GDA, Hirschfield GM, Hold G, et al. The gut
microbiota and host health: a new clinical frontier. Gut [Internet]. 2015 Sep 2 [cited 2018
Sep 17];gutjnl-2015-309990. Available from:
https://gut.bmj.com/content/early/2015/09/02/gutjnl-2015-309990
4. Scott KP, Jean-Michel A, Midtvedt T, Hemert S van. Manipulating the gut microbiota to
maintain health and treat disease. Microbial Ecology in Health and Disease [Internet]. 2015
Dec 1 [cited 2018 Sep 17];26(1):25877. Available from:
https://www.tandfonline.com/doi/abs/10.3402/mehd.v26.25877
5. Kootte RS, Vrieze A, Holleman F, Dallinga‐Thie GM, Zoetendal EG, Vos WM de, et al.
The therapeutic potential of manipulating gut microbiota in obesity and type 2 diabetes
mellitus. Diabetes, Obesity and Metabolism [Internet]. 2012 Feb 1 [cited 2018 Sep
17];14(2):112–20. Available from: https://onlinelibrary.wiley.com/doi/abs/10.1111/j.1463-
1326.2011.01483.x
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
6. Hiergeist A, Gläsner J, Reischl U, Gessner A. Analyses of Intestinal Microbiota: Culture
versus Sequencing. ILAR J [Internet]. 2015 Aug 31 [cited 2018 Sep 17];56(2):228–40.
Available from: https://academic.oup.com/ilarjournal/article/56/2/228/650795
7. Rappé MS, Giovannoni SJ. The uncultured microbial majority. Annu Rev Microbiol.
2003;57:369–94.
8. Matsuki T, Watanabe K, Fujimoto J, Miyamoto Y, Takada T, Matsumoto K, et al.
Development of 16S rRNA-Gene-Targeted Group-Specific Primers for the Detection and
Identification of Predominant Bacteria in Human Feces. Appl Environ Microbiol [Internet].
2002 Nov 1 [cited 2018 Sep 17];68(11):5445–51. Available from:
https://aem.asm.org/content/68/11/5445
9. Janda JM, Abbott SL. 16S rRNA Gene Sequencing for Bacterial Identification in the
Diagnostic Laboratory: Pluses, Perils, and Pitfalls. J Clin Microbiol [Internet]. 2007 Sep 1
[cited 2018 Aug 5];45(9):2761–4. Available from: http://jcm.asm.org/content/45/9/2761
10. Pollock J, Glendinning L, Wisedchanwet T, Watson M. The madness of microbiome:
Attempting to find consensus ―best practice‖ for 16S microbiome studies. Appl Environ
Microbiol [Internet]. 2018 Feb 2 [cited 2018 Sep 17];AEM.02627-17. Available from:
https://aem.asm.org/content/early/2018/01/29/AEM.02627-17
11. Srinivasan R, Karaoz U, Volegova M, MacKichan J, Kato-Maeda M, Miller S, et al. Use of
16S rRNA Gene for Identification of a Broad Range of Clinically Relevant Bacterial
Pathogens. PLoS One [Internet]. 2015 Feb 6 [cited 2018 Sep 17];10(2). Available from:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4319838/
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
12. Tremblay J, Singh K, Fern A, Kirton ES, He S, Woyke T, et al. Primer and platform effects
on 16S rRNA tag sequencing. Front Microbiol [Internet]. 2015 Aug 4 [cited 2018 Sep
17];6. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4523815/
13. Suzuki MT, Giovannoni SJ. Bias caused by template annealing in the amplification of
mixtures of 16S rRNA genes by PCR. Appl Environ Microbiol [Internet]. 1996 Feb 1 [cited
2018 Sep 17];62(2):625–30. Available from: https://aem.asm.org/content/62/2/625
14. Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, et al.
Predictive functional profiling of microbial communities using 16S rRNA marker gene
sequences. Nat Biotechnol. 2013 Sep;31(9):814–21.
15. Louis P, Flint HJ. Development of a Semiquantitative Degenerate Real-Time PCR-Based
Assay for Estimation of Numbers of Butyryl-Coenzyme A (CoA) CoA Transferase Genes
in Complex Bacterial Samples. Appl Environ Microbiol [Internet]. 2007 Mar 15 [cited 2018
Aug 6];73(6):2009–12. Available from: http://aem.asm.org/content/73/6/2009
16. Wang W-L, Xu S-Y, Ren Z-G, Tao L, Jiang J-W, Zheng S-S. Application of metagenomics
in the human gut microbiome. World J Gastroenterol [Internet]. 2015 Jan 21 [cited 2018
Aug 5];21(3):803–14. Available from:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4299332/
17. Human Microbiome Project Consortium. Structure, function and diversity of the healthy
human microbiome. Nature. 2012 Jun 13;486(7402):207–14.
18. Truong DT, Franzosa EA, Tickle TL, Scholz M, Weingart G, Pasolli E, et al. MetaPhlAn2
for enhanced metagenomic taxonomic profiling. Nature Methods [Internet]. 2015 Oct [cited
2018 Aug 6];12(10):902–3. Available from: https://www.nature.com/articles/nmeth.3589
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
19. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, et al. Metabolic
reconstruction for metagenomic data and its application to the human microbiome. PLoS
Comput Biol. 2012;8(6):e1002358.
20. Kanehisa M, Goto S, Sato Y, Kawashima M, Furumichi M, Tanabe M. Data, information,
knowledge and principle: back to metabolism in KEGG. Nucleic Acids Res. 2014
Jan;42(Database issue):D199-205.
21. Activities at the Universal Protein Resource (UniProt). Nucleic Acids Res [Internet]. 2014
Jan 1 [cited 2018 Aug 6];42(Database issue):D191–8. Available from:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3965022/
22. Morgan XC, Huttenhower C. Meta‘omic analytic techniques for studying the intestinal
microbiome. Gastroenterology. 2014 May;146(6):1437-1448.e1.
23. Torii T, Kanemitsu K, Wada T, Itoh S, Kinugawa K, Hagiwara A. Measurement of short-
chain fatty acids in human faeces using high-performance liquid chromatography: specimen
stability. Ann Clin Biochem. 2010 Sep;47(Pt 5):447–52.
24. Donia MS, Fischbach MA. HUMAN MICROBIOTA. Small molecules from the human
microbiota. Science. 2015 Jul 24;349(6246):1254766.
25. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al.
QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010
May;7(5):335–6.
26. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, et al. Introducing
mothur: Open-Source, Platform-Independent, Community-Supported Software for
Describing and Comparing Microbial Communities. Appl Environ Microbiol [Internet].
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
2009 Dec [cited 2018 Aug 5];75(23):7537–41. Available from:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2786419/
27. Goll J, Rusch DB, Tanenbaum DM, Thiagarajan M, Li K, Methé BA, et al. METAREP:
JCVI metagenomics reports—an open source tool for high-performance comparative
metagenomics. Bioinformatics [Internet]. 2010 Oct 15 [cited 2018 Aug 5];26(20):2631–2.
Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2951084/
28. Huson DH, Auch AF, Qi J, Schuster SC. MEGAN analysis of metagenomic data. Genome
Res [Internet]. 2007 Mar [cited 2018 Nov 12];17(3):377–86. Available from:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1800929/
29. Meyer F, Paarmann D, D‘Souza M, Olson R, Glass E, Kubal M, et al. The metagenomics
RAST server – a public resource for the automatic phylogenetic and functional analysis of
metagenomes. BMC Bioinformatics [Internet]. 2008 Sep 19 [cited 2018 Nov 12];9(1):386.
Available from: https://doi.org/10.1186/1471-2105-9-386
30. McDonald D, Price MN, Goodrich J, Nawrocki EP, DeSantis TZ, Probst A, et al. An
improved Greengenes taxonomy with explicit ranks for ecological and evolutionary
analyses of bacteria and archaea. The ISME Journal [Internet]. 2012 Mar [cited 2018 Sep
17];6(3):610–8. Available from: https://www.nature.com/articles/ismej2011139
31. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naïve Bayesian Classifier for Rapid Assignment
of rRNA Sequences into the New Bacterial Taxonomy. Appl Environ Microbiol [Internet].
2007 Aug 15 [cited 2018 Sep 17];73(16):5261–7. Available from:
https://aem.asm.org/content/73/16/5261
32. Anderson MJ, Willis TJ. Canonical Analysis of Principal Coordinates: A Useful Method of
Constrained Ordination for Ecology. Ecology [Internet]. 2003 Feb 1 [cited 2018 Sep
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
18];84(2):511–25. Available from:
https://esajournals.onlinelibrary.wiley.com/doi/abs/10.1890/0012-
9658%282003%29084%5B0511%3ACAOPCA%5D2.0.CO%3B2
33. McArdle B, Anderson M. Fitting multivariate models to community data: a comment on
distance-based redundancy analysis. Ecology [Internet]. [cited 2018 Nov 12];82. Available
from: https://esajournals.onlinelibrary.wiley.com/doi/abs/10.1890/0012-
9658(2001)082%5B0290:FMMTCD%5D2.0.CO;2
34. Perez-Muñoz ME, Arrieta M-C, Ramer-Tait AE, Walter J. A critical assessment of the
―sterile womb‖ and ―in utero colonization‖ hypotheses: implications for research on the
pioneer infant microbiome. Microbiome [Internet]. 2017 Apr 28 [cited 2018 Oct
30];5(1):48. Available from: https://doi.org/10.1186/s40168-017-0268-4
35. Penders J, Thijs C, Vink C, Stelma FF, Snijders B, Kummeling I, et al. Factors Influencing
the Composition of the Intestinal Microbiota in Early Infancy. Pediatrics [Internet]. 2006
Aug 1 [cited 2018 Aug 5];118(2):511–21. Available from:
http://pediatrics.aappublications.org/content/118/2/511
36. Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, Anderson EE, et al. Extensive impact
of non-antibiotic drugs on human gut bacteria. Nature. 2018 Mar 29;555(7698):623–8.
37. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet
rapidly and reproducibly alters the human gut microbiome. Nature. 2014 Jan
23;505(7484):559–63.
38. Kang SS, Jeraldo PR, Kurti A, Miller MEB, Cook MD, Whitlock K, et al. Diet and exercise
orthogonally alter the gut microbiome and reveal independent associations with anxiety and
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
cognition. Mol Neurodegener [Internet]. 2014 Sep 13;9:36. Available from:
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4168696/
39. Petriz BA, Castro AP, Almeida JA, Gomes CP, Fernandes GR, Kruger RH, et al. Exercise
induction of gut microbiota modifications in obese, non-obese and hypertensive rats. BMC
Genomics [Internet]. 2014 Jun 21 [cited 2018 Feb 12];15:511. Available from:
https://doi.org/10.1186/1471-2164-15-511
40. Lambert JE, Myslicki JP, Bomhof MR, Belke DD, Shearer J, Reimer RA. Exercise training
modifies gut microbiota in normal and diabetic mice. Appl Physiol Nutr Metab [Internet].
2015 Feb 18 [cited 2018 Feb 12];40(7):749–52. Available from:
http://www.nrcresearchpress.com/doi/abs/10.1139/apnm-2014-0452
41. Evans CC, LePard KJ, Kwak JW, Stancukas MC, Laskowski S, Dougherty J, et al. Exercise
prevents weight gain and alters the gut microbiota in a mouse model of high fat diet-
induced obesity. PLoS ONE. 2014;9(3):e92193.
42. Queipo-Ortuño MI, Seoane LM, Murri M, Pardo M, Gomez-Zumaquero JM, Cardona F, et
al. Gut Microbiota Composition in Male Rat Models under Different Nutritional Status and
Physical Activity and Its Association with Serum Leptin and Ghrelin Levels. PLOS ONE
[Internet]. 2013 May 28 [cited 2017 Jun 19];8(5):e65465. Available from:
http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0065465
43. Denou E, Marcinko K, Surette MG, Steinberg GR, Schertzer JD. High-intensity exercise
training increases the diversity and metabolic capacity of the mouse distal gut microbiota
during diet-induced obesity. Am J Physiol Endocrinol Metab. 2016 Jun 1;310(11):E982-
993.
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
44. Mika A, Van Treuren W, González A, Herrera JJ, Knight R, Fleshner M. Exercise Is More
Effective at Altering Gut Microbial Composition and Producing Stable Changes in Lean
Mass in Juvenile versus Adult Male F344 Rats. PLoS One [Internet]. 2015 May 27;10(5).
Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4446322/
45. Campbell SC, Wisniewski PJ, Noji M, McGuinness LR, Häggblom MM, Lightfoot SA, et
al. The Effect of Diet and Exercise on Intestinal Integrity and Microbial Diversity in Mice.
PLoS ONE. 2016;11(3):e0150502.
46. Matsumoto M, Inoue R, Tsukahara T, Ushida K, Chiji H, Matsubara N, et al. Voluntary
running exercise alters microbiota composition and increases n-butyrate concentration in
the rat cecum. Biosci Biotechnol Biochem. 2008 Feb;72(2):572–6.
47. Allen JM, Miller MEB, Pence BD, Whitlock K, Nehra V, Gaskins HR, et al. Voluntary and
forced exercise differentially alters the gut microbiome in C57BL/6J mice. Journal of
Applied Physiology [Internet]. 2015 Apr 15 [cited 2017 Jun 19];118(8):1059–66. Available
from: http://jap.physiology.org/content/118/8/1059
48. Liu T-W, Park Y-M, Holscher HD, Padilla J, Scroggins RJ, Welly R, et al. Physical
Activity Differentially Affects the Cecal Microbiota of Ovariectomized Female Rats
Selectively Bred for High and Low Aerobic Capacity. PLOS ONE [Internet]. 2015 Aug 24
[cited 2018 Feb 12];10(8):e0136150. Available from:
http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0136150
49. Lamoureux EV, Grandy SA, Langille MGI. Moderate Exercise Has Limited but
Distinguishable Effects on the Mouse Microbiome. mSystems [Internet]. 2017 Aug 29
[cited 2018 Feb 12];2(4):e00006-17. Available from:
http://msystems.asm.org/content/2/4/e00006-17
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
50. Welly RJ, Liu T-W, Zidon TM, Rowles JL, Park Y-M, Smith TN, et al. Comparison of Diet
versus Exercise on Metabolic Function and Gut Microbiota in Obese Rats. Med Sci Sports
Exerc. 2016 Sep;48(9):1688–98.
51. Batacan R b., Fenning A s., Dalbo V j., Scanlan A t., Duncan M j., Moore R j., et al. A gut
reaction: the combined influence of exercise and diet on gastrointestinal microbiota in rats.
J Appl Microbiol [Internet]. 2017 Jun 1 [cited 2018 Feb 12];122(6):1627–38. Available
from: http://onlinelibrary.wiley.com/doi/10.1111/jam.13442/abstract
52. Säemann MD, Böhmig GA, Osterreicher CH, Burtscher H, Parolini O, Diakos C, et al.
Anti-inflammatory effects of sodium butyrate on human monocytes: potent inhibition of IL-
12 and up-regulation of IL-10 production. FASEB J. 2000 Dec;14(15):2380–2.
53. Peng L, Li Z-R, Green RS, Holzman IR, Lin J. Butyrate Enhances the Intestinal Barrier by
Facilitating Tight Junction Assembly via Activation of AMP-Activated Protein Kinase in
Caco-2 Cell Monolayers. J Nutr [Internet]. 2009 Sep 1 [cited 2017 Jul 18];139(9):1619–25.
Available from: http://jn.nutrition.org/content/139/9/1619
54. Clarke SF, Murphy EF, O‘Sullivan O, Lucey AJ, Humphreys M, Hogan A, et al. Exercise
and associated dietary extremes impact on gut microbial diversity. Gut. 2014
Dec;63(12):1913–20.
55. Bressa C, Bailén-Andrino M, Pérez-Santiago J, González-Soltero R, Pérez M, Montalvo-
Lominchar MG, et al. Differences in gut microbiota profile between women with active
lifestyle and sedentary women. PLOS ONE [Internet]. 2017 Feb 10 [cited 2018 Feb
12];12(2):e0171352. Available from:
http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0171352
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
56. Louis P, Flint HJ. Diversity, metabolism and microbial ecology of butyrate-producing
bacteria from the human large intestine. FEMS Microbiol Lett [Internet]. 2009 May 1 [cited
2018 Aug 5];294(1):1–8. Available from:
https://academic.oup.com/femsle/article/294/1/1/468786
57. Dao MC, Everard A, Aron-Wisnewsky J, Sokolovska N, Prifti E, Verger EO, et al.
Akkermansia muciniphila and improved metabolic health during a dietary intervention in
obesity: relationship with gut microbiome richness and ecology. Gut [Internet]. 2016 Mar 1
[cited 2018 Aug 5];65(3):426–36. Available from: https://gut.bmj.com/content/65/3/426
58. Durk RP, Castillo E, Márquez-Magaña L, Grosicki GJ, Bolter ND, Lee CM, et al. Gut
Microbiota Composition is Related to Cardiorespiratory Fitness in Healthy Young Adults.
International Journal of Sport Nutrition and Exercise Metabolism [Internet]. 2018 Jul 10
[cited 2018 Jul 13];1–15. Available from:
https://journals.humankinetics.com/doi/10.1123/ijsnem.2018-0024
59. Estaki M, Pither J, Baumeister P, Little JP, Gill SK, Ghosh S, et al. Cardiorespiratory
fitness as a predictor of intestinal microbial diversity and distinct metagenomic functions.
Microbiome [Internet]. 2016 Aug 8 [cited 2018 Feb 12];4:42. Available from:
https://doi.org/10.1186/s40168-016-0189-7
60. Barton W, Penney NC, Cronin O, Garcia-Perez I, Molloy MG, Holmes E, et al. The
microbiome of professional athletes differs from that of more sedentary subjects in
composition and particularly at the functional metabolic level. Gut [Internet]. 2017 Mar 29
[cited 2018 Feb 12];gutjnl-2016-313627. Available from:
http://gut.bmj.com/content/early/2017/03/29/gutjnl-2016-313627
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
61. Allen JM, Mailing LJ, Niemiro GM, Moore R, Cook MD, White BA, et al. Exercise Alters
Gut Microbiota Composition and Function in Lean and Obese Humans. Med Sci Sports
Exerc. 2017 Nov 20;
62. Cronin O, Barton W, Skuse P, Penney NC, Garcia-Perez I, Murphy EF, et al. A Prospective
Metagenomic and Metabolomic Analysis of the Impact of Exercise and/or Whey Protein
Supplementation on the Gut Microbiome of Sedentary Adults. mSystems [Internet]. 2018
Jun 26 [cited 2018 Jul 26];3(3):e00044-18. Available from:
https://msystems.asm.org/content/3/3/e00044-18
63. Munukka E, Ahtiainen JP, Puigbó P, Jalkanen S, Pahkala K, Keskitalo A, et al. Six-Week
Endurance Exercise Alters Gut Metagenome That Is not Reflected in Systemic Metabolism
in Over-weight Women. Front Microbiol [Internet]. 2018 Oct 3 [cited 2018 Oct 30];9.
Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6178902/
64. Packer N, Hoffman-Goetz L. Exercise training reduces inflammatory mediators in the
intestinal tract of healthy older adult mice. Can J Aging. 2012 Jun;31(2):161–71.
65. Hoffman-Goetz L, Pervaiz N, Guan J. Voluntary exercise training in mice increases the
expression of antioxidant enzymes and decreases the expression of TNF-alpha in intestinal
lymphocytes. Brain Behav Immun. 2009 May;23(4):498–506.
66. Hoffman-Goetz L. Freewheel training decreases pro- and increases anti-inflammatory
cytokine expression in mouse intestinal lymphocytes. Brain, Behavior, and Immunity
[Internet]. 2010 [cited 2017 Jul 12];(24):1105–15. Available from:
https://www.researchgate.net/publication/44637863_Freewheel_training_decreases_pro-
_and_increases_anti-inflammatory_cytokine_expression_in_mouse_intestinal_lymphocytes
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
67. Ismail AS, Severson KM, Vaishnava S, Behrendt CL, Yu X, Benjamin JL, et al. γδ
intraepithelial lymphocytes are essential mediators of host–microbial homeostasis at the
intestinal mucosal surface. PNAS [Internet]. 2011 May 24 [cited 2018 Nov
13];108(21):8743–8. Available from: http://www.pnas.org/content/108/21/8743
68. Rowell LB, Brengelmann GL, Blackmon JR, Twiss RD, Kusumi F. Splanchnic blood flow
and metabolism in heat-stressed man. Journal of Applied Physiology [Internet]. 1968 Apr 1
[cited 2018 Mar 13];24(4):475–84. Available from:
https://www.physiology.org/doi/abs/10.1152/jappl.1968.24.4.475
69. van Wijck K, Lenaerts K, van Loon LJC, Peters WHM, Buurman WA, Dejong CHC.
Exercise-induced splanchnic hypoperfusion results in gut dysfunction in healthy men. PLoS
ONE. 2011;6(7):e22366.
70. Otte JA, Oostveen E, Geelkerken RH, Groeneveld ABJ, Kolkman JJ. Exercise induces
gastric ischemia in healthy volunteers: a tonometry study. Journal of Applied Physiology
[Internet]. 2001 Aug 1 [cited 2018 Feb 27];91(2):866–71. Available from:
http://www.physiology.org/doi/full/10.1152/jappl.2001.91.2.866
71. Lira FS, Rosa JC, Pimentel GD, Souza HA, Caperuto EC, Carnevali LC, et al. Endotoxin
levels correlate positively with a sedentary lifestyle and negatively with highly trained
subjects. Lipids Health Dis. 2010 Aug 4;9:82.
72. Fehrenbach E, Niess AM, Schlotz E, Passek F, Dickhuth H-H, Northoff H. Transcriptional
and translational regulation of heat shock proteins in leukocytes of endurance runners.
Journal of Applied Physiology [Internet]. 2000 Aug 1 [cited 2018 Mar 12];89(2):704–10.
Available from: https://www.physiology.org/doi/full/10.1152/jappl.2000.89.2.704
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
73. Dokladny K, Moseley PL, Ma TY. Physiologically relevant increase in temperature causes
an increase in intestinal epithelial tight junction permeability. Am J Physiol Gastrointest
Liver Physiol. 2006 Feb;290(2):G204-212.
74. Song BK, Cho KO, Jo Y, Oh JW, Kim YS. Colon transit time according to physical activity
level in adults. J Neurogastroenterol Motil. 2012 Jan;18(1):64–9.
75. Dainese R, Serra J, Azpiroz F, Malagelada J-R. Effects of physical activity on intestinal gas
transit and evacuation in healthy subjects. Am J Med. 2004 Apr 15;116(8):536–9.
76. Freeman JV, Dewey FE, Hadley DM, Myers J, Froelicher VF. Autonomic Nervous System
Interaction With the Cardiovascular System During Exercise. Progress in Cardiovascular
Diseases [Internet]. 2006 Mar 1 [cited 2018 Oct 30];48(5):342–62. Available from:
http://www.sciencedirect.com/science/article/pii/S0033062005001386
77. Meissner M, Lombardo E, Havinga R, Tietge UJF, Kuipers F, Groen AK. Voluntary wheel
running increases bile acid as well as cholesterol excretion and decreases atherosclerosis in
hypercholesterolemic mice. Atherosclerosis [Internet]. 2011 Oct 1 [cited 2018 Nov
14];218(2):323–9. Available from:
http://www.sciencedirect.com/science/article/pii/S0021915011005624
78. Kakiyama G, Pandak WM, Gillevet PM, Hylemon PB, Heuman DM, Daita K, et al.
Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol. 2013
May;58(5):949–55.
79. Egan B, Zierath JR. Exercise Metabolism and the Molecular Regulation of Skeletal Muscle
Adaptation. Cell Metabolism [Internet]. 2013 Feb 5 [cited 2018 Oct 30];17(2):162–84.
Available from: http://www.sciencedirect.com/science/article/pii/S1550413112005037
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
80. Warburton DER, Nicol CW, Bredin SSD. Health benefits of physical activity: the evidence.
CMAJ [Internet]. 2006 Mar 14 [cited 2018 Oct 30];174(6):801–9. Available from:
http://www.cmaj.ca/content/174/6/801
81. Wolin KY, Yan Y, Colditz GA, Lee I-M. Physical activity and colon cancer prevention: a
meta-analysis. British Journal of Cancer [Internet]. 2009 Feb [cited 2018 Mar
27];100(4):611–6. Available from: https://www.nature.com/articles/6604917
82. Courneya KS, Friedenreich CM, Quinney HA, Fields ALA, Jones LW, Fairey AS. A
randomized trial of exercise and quality of life in colorectal cancer survivors. Eur J Cancer
Care (Engl). 2003 Dec;12(4):347–57.
83. Andrianopoulos G, Nelson RL, Bombeck CT, Souza G. The influence of physical activity
in 1,2 dimethylhydrazine induced colon carcinogenesis in the rat. Anticancer Res [Internet].
1987 [cited 2018 Mar 27];7(4B):849–52. Available from:
http://europepmc.org/abstract/med/3674772
84. Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, et al. Structural segregation of gut
microbiota between colorectal cancer patients and healthy volunteers. ISME J [Internet].
2012 Feb [cited 2018 Nov 6];6(2):320–9. Available from:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3260502/
85. Donohoe DR, Collins LB, Wali A, Bigler R, Sun W, Bultman SJ. The Warburg effect
dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol
Cell. 2012 Nov 30;48(4):612–26.
86. Basterfield L, Mathers JC. Intestinal tumours, colonic butyrate and sleep in exercised Min
mice. British Journal of Nutrition [Internet]. 2010 Aug [cited 2018 Nov 13];104(3):355–63.
Available from: https://www.cambridge.org/core/journals/british-journal-of-
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
nutrition/article/intestinal-tumours-colonic-butyrate-and-sleep-in-exercised-min-
mice/1D2C3CAA3C431BAADEC834D205DED89A
87. Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, et al. Dysfunction of
the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biology
[Internet]. 2012 Sep 26 [cited 2018 Nov 13];13(9):R79. Available from:
https://doi.org/10.1186/gb-2012-13-9-r79
88. Jones PD, Kappelman MD, Martin CF, Chen W, Sandler RS, Long MD. Exercise
Decreases Risk of Future Active Disease in Inflammatory Bowel Disease Patients in
Remission. Inflamm Bowel Dis [Internet]. 2015 May [cited 2018 Mar 26];21(5):1063–71.
Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4402239/
89. Elsenbruch S, Langhorst J, Popkirowa K, Müller T, Luedtke R, Franken U, et al. Effects of
mind-body therapy on quality of life and neuroendocrine and cellular immune functions in
patients with ulcerative colitis. Psychother Psychosom. 2005;74(5):277–87.
90. Szalai Z, Szász A, Nagy I, Puskás LG, Kupai K, Király A, et al. Anti-Inflammatory Effect
of Recreational Exercise in TNBS-Induced Colitis in Rats: Role of NOS/HO/MPO System
[Internet]. Oxidative Medicine and Cellular Longevity. 2014 [cited 2018 Nov 13].
Available from: https://www.hindawi.com/journals/omcl/2014/925981/abs/
91. Liu W-X, Zhou F, Wang Y, Wang T, Xing J-W, Zhang S, et al. Voluntary exercise protects
against ulcerative colitis by up-regulating glucocorticoid-mediated PPAR-γ activity in the
colon in mice. Acta Physiologica [Internet]. 2015 Sep 1 [cited 2018 Nov 13];215(1):24–36.
Available from: https://onlinelibrary.wiley.com/doi/abs/10.1111/apha.12534
92. Cook MD, Martin SA, Williams C, Whitlock K, Wallig MA, Pence BD, et al. Forced
treadmill exercise training exacerbates inflammation and causes mortality while voluntary
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
wheel training is protective in a mouse model of colitis. Brain Behav Immun. 2013
Oct;33:46–56.
93. Allen JM, Mailing LJ, Cohrs J, Salmonson C, Fryer JD, Nehra V, et al. Exercise training-
induced modification of the gut microbiota persists after microbiota colonization and
attenuates the response to chemically-induced colitis in gnotobiotic mice. Gut Microbes
[Internet]. 2017 Sep 1 [cited 2018 Feb 20];0(0):1–16. Available from:
https://doi.org/10.1080/19490976.2017.1372077
94. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-
associated gut microbiome with increased capacity for energy harvest. Nature [Internet].
2006 Dec 21 [cited 2017 Aug 3];444(7122):1027–131. Available from:
https://www.nature.com/nature/journal/v444/n7122/abs/nature05414.html
95. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic
Endotoxemia Initiates Obesity and Insulin Resistance. Diabetes [Internet]. 2007 Jul 1 [cited
2018 Jul 26];56(7):1761–72. Available from:
http://diabetes.diabetesjournals.org/content/56/7/1761
96. Lai Z-L, Tseng C-H, Ho HJ, Cheung CKY, Lin J-Y, Chen Y-J, et al. Fecal microbiota
transplantation confers beneficial metabolic effects of diet and exercise on diet-induced
obese mice. Scientific Reports [Internet]. 2018 Oct 23 [cited 2018 Nov 13];8(1):15625.
Available from: https://www.nature.com/articles/s41598-018-33893-y
97. Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, Lefevre M, et al. Butyrate improves insulin
sensitivity and increases energy expenditure in mice. Diabetes. 2009 Jul;58(7):1509–17.
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
98. Li N, Hatch M, Wasserfall CH, Douglas-Escobar M, Atkinson MA, Schatz DA, et al.
Butyrate and type 1 diabetes mellitus: can we fix the intestinal leak? J Pediatr Gastroenterol
Nutr. 2010 Oct;51(4):414–7.
99. Cryan JF, O‘Mahony SM. The microbiome-gut-brain axis: from bowel to behavior.
Neurogastroenterology & Motility [Internet]. 2011 Mar 1 [cited 2018 Oct 30];23(3):187–
92. Available from: https://onlinelibrary.wiley.com/doi/abs/10.1111/j.1365-
2982.2010.01664.x
100. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between
enteric microbiota, central and enteric nervous systems. Ann Gastroenterol [Internet]. 2015
[cited 2018 Oct 30];28(2):203–9. Available from:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4367209/
101. Forsythe P, Bienenstock J, Kunze WA. Vagal pathways for microbiome-brain-gut axis
communication. Adv Exp Med Biol. 2014;817:115–33.
102. Luczynski P, McVey Neufeld K-A, Oriach CS, Clarke G, Dinan TG, Cryan JF. Growing up
in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on
Brain and Behavior. Int J Neuropsychopharmacol [Internet]. 2016 Aug 1 [cited 2018 Jul
26];19(8). Available from: https://academic.oup.com/ijnp/article/19/8/pyw020/2910071
103. Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, et al. Gut microbiome remodeling induces
depressive-like behaviors through a pathway mediated by the host‘s metabolism. Molecular
Psychiatry [Internet]. 2016 Jun [cited 2018 Nov 13];21(6):786–96. Available from:
https://www.nature.com/articles/mp201644
104. Stevens BR, Goel R, Seungbum K, Richards EM, Holbert RC, Pepine CJ, et al. Increased
human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
with plasma LPS and altered gut microbiome in anxiety or depression. Gut [Internet]. 2018
Aug [cited 2018 Nov 13];67(8):1555–7. Available from:
https://europepmc.orghttp://europepmc.org/articles/PMC5851874/
105. Huang T, Larsen KT, Ried-Larsen M, Møller NC, Andersen LB. The effects of physical
activity and exercise on brain-derived neurotrophic factor in healthy humans: A review.
Scand J Med Sci Sports. 2014 Feb;24(1):1–10.
106. Varela RB, Valvassori SS, Lopes-Borges J, Mariot E, Dal-Pont GC, Amboni RT, et al.
Sodium butyrate and mood stabilizers block ouabain-induced hyperlocomotion and increase
BDNF, NGF and GDNF levels in brain of Wistar rats. J Psychiatr Res. 2015 Feb;61:114–
21.
107. Matt SM, Allen JM, Lawson MA, Mailing LJ, Woods JA, Johnson RW. Butyrate and
Dietary Soluble Fiber Improve Neuroinflammation Associated With Aging in Mice. Front
Immunol [Internet]. 2018 Aug 14 [cited 2018 Nov 13];9. Available from:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6102557/
108. Intlekofer KA, Berchtold NC, Malvaez M, Carlos AJ, McQuown SC, Cunningham MJ, et
al. Exercise and sodium butyrate transform a subthreshold learning event into long-term
memory via a brain-derived neurotrophic factor-dependent mechanism.
Neuropsychopharmacology. 2013 Sep;38(10):2027–34.
109. Zmora N, Zilberman-Schapira G, Suez J, Mor U, Dori-Bachash M, Bashiardes S, et al.
Personalized Gut Mucosal Colonization Resistance to Empiric Probiotics Is Associated
with Unique Host and Microbiome Features. Cell [Internet]. 2018 Sep 6 [cited 2018 Sep
8];174(6):1388-1405.e21. Available from: https://www.cell.com/cell/abstract/S0092-
8674(18)31102-4
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
FIGURE LEGENDS
Figure 1. Methodologies commonly used to study the gut microbiome. Fecal or GI content
samples can be used for metabolite analysis or undergo chemical and mechanical digestion to
extract nucleic acids. Extracted DNA can be used for targeted genomics, shotgun sequencing, or
16S rRNA gene amplification.
Figure 2. Current unknowns and future areas of research related to exercise and the gut
microbiome. While several studies have now shown that exercise alters gut microbiota
composition, functional capacity, and metabolites, the effects of different exercise frequencies,
modes, and intensities is unknown. Assessing the effects of exercise on the gut microbiota in
different populations and its synergy with different diets also represents a key area of future
research. Mechanistic studies, such as those that utilize mice, will help determine the potential
mechanisms involved, and whether exercise-induced changes in the gut environment are
potentially disease modifying.
Figure 3. Proposed model for how exercise alters the gut microbiota and gut epithelium
with potential implications for human health. Exercise has been shown to increase butyrate-
producing taxa and fecal butyrate concentrations and reduce pro-inflammatory cytokines and
oxidative stress in the gut. Exercise is also known to have benefits on whole-body physiology,
and is protective against colon cancer, inflammatory bowel disease, depression, anxiety, and
obesity. Whether this disease protection is mediated by exercise-induced changes in the gut
microbiome and gut epithelium remains to be determined.
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
Figure 1
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
Figure 2
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
Figure 3
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
Table. Summary of cross-sectional and longitudinal studies assessing the impact of physical activity status or an exercise intervention
on the human gut microbiome.
STUDY
DESIGN
SUBJECTS
EXERCISE
TRAINING
CHANGE OR
CONTROL OF
DIET
IMPACT ON GUT MICROBIAL
COMMUNITIES
Clarke et al,
2014
Cross-
sectional
Elite rugby players
(n=40), low BMI
controls (n=23), and
high BMI controls
(n=23)
n/a
Elite athletes
consumed significantly
more protein and total
energy. Increased
protein intake
accounted for many
observed differences in
gut microbial
composition.
Greater alpha diversity in elite
athletes compared to lean sedentary
controls. ↑Akkermansia in athletes
and low BMI controls;
↑Erysipelotrichaceae, S24-7,
Prevotella and Succinivibrio and
↓Lactobacillaceae, Bacteroides, and
Lactobacillus spp. in athletes
compared to lean controls
Estaki et al.
2016
Cross-
sectional
Healthy adults with
varying
cardiorespiratory
fitness levels (n=39)
n/a
Protein intake was
highly associated with
overall microbial
community
composition.
VO2 peak accounted for more than
20 percent of the variation in species
richness. Individuals with higher
fitness had increased relative
abundance of butyrate-producing
taxa and increased fecal butyrate
concentrations.
Stewart et
al. 2016
Cross-
sectional
Adult males Type 1
diabetics with good
glycemic control and
high levels of physical
n/a
Not assessed.
Gut microbial composition of
patients with Type 1 diabetes in
good glycemic control and with high
physical fitness levels is comparable
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
fitness (n=10) and
matched healthy adult
male controls (n=10)
to those of matche people without
diabetes
Bressa et al.
2017
Cross-
sectional
Premenopausal
women, active (>3
hours of physical
exercise/wk, n=19) or
sedentary (<30
minutes 3 days/wk,
n=21)
n/a
Greater consumption
of fruits and
vegetables by active
group; sedentary group
ingested more
processed meats.
Increased relative abundance of
Faecalibacterium prausnitzii,
Roseburia hominis, and Akkermansia
muciniphila in active women;
reduced relative abundance of
Barnesiellaceae, Odoribacteraceae
Yang et al.
2017
Cross-
sectional
Premenopausal
women, all activity
levels, primarily
overweight or obese
(n=71)
n/a
No significant
differences between
groups in
macronutrient
composition, fiber, or
total energy intake.
Lower VO2 max was associated with
lower relative abundance of
Bacteroides spp. and higher rel.
abundance of Eubacterium rectale-
Clostridium coccoides group
Barton et al.
2018
Cross-
sectional
Professional rugby
players (n=40) and
sedentary controls
with low BMI (n=22)
or high BMI (n=24)
n/a
Rugby players
consumed significantly
more protein and total
energy.
Rugby players had increased amino
acid and antibiotic biosynthesis,
carbohydrate metabolism, and
increased fecal SCFAs compared to
controls
Durk et al.
2018
Cross-
sectional
Healthy young adults
(n=20 males, n=17
females) with varying
n/a
No association
between macronutrient
intake and Firmicutes
Higher ratio of Firmicutes to
Bacteroidetes was significantly
correlated with VO2 max. VO2 max
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
cardiorespiratory
fitness level
to Bacteroidetes ratio.
accounted for 22 percent of the
variance in gut microbiota
composition.
Paulsen et
al. 2017
Longitudinal
Post-primary
treatment breast
cancer survivors
(n=12)
Received written
materials regarding
benefits of physical
activity. Fitness
measured at baseline
and 3 months
No significant changes
in self-reported
carbohydrate or fiber
intake. Other
macronutrients not
reported or controlled
for.
Significant association between
cardiorespiratory fitness and beta
diversity at 3 month timepoint.
Allen et al.
2018
Longitudinal
Previously sedentary
lean or obese adults
(n=32)
6 week progressive
aerobic exercise
intervention
(moderate-high
intensity) + 6 week
sedentary washout
period
Diet stability
confirmed using 7-day
diet diaries and a 3-
day control diet prior
to each fecal
collection.
Several taxa were differentially
altered depending on BMI status:
Faecalibacterium increased in lean
subjects but decreased in obese;
Bacteroides decreased in lean
subjects but increased in obese.
Increased butyrate-producing taxa,
fecal acetate and butyrate
concentrations. Effects were
reversed upon return to sedentary
lifestyle.
Cronin et al.
2018
Longitudinal
Predominantly
overweight or obese
adults randomized to
exercise-only (E),
8 week mixed
progressive moderate
aerobic exercise (18-
32 min) and
Self-reported
maintenance of dietary
intake
No significant changes in taxonomic
composition; trend for increase in
bacterial diversity in E and EP
groups. Only modest alterations of
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED
exercise + whey
protein (EP) or whey
protein only (P)
groups (n=30 each
group)
resistance training
(3x/week)
microbial metabolic potential.
Munukka et
al. 2018
Longitudinal
Previously sedentary,
overweight women
(n=17)
6 week cycling
exercise (low-
moderate intensity)
Diet stability
confirmed in 14
subjects using 3-day
food record; only
slight increase in
energy derived from
starch.
Increased relative abundance of
Akkermansia and decreased relative
abundance of Proteobacteria. Only
half of the subjects' microbiomes
responded to exercise. Exercise
training decreased abundance of
fructose and amino acid metabolism
related genes
Copyright © 2019 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
ACCEPTED