Next Article in Journal
Examining the Influence of Cultural Immersion on Willingness to Try Fruits and Vegetables among Children in Guam: The Traditions Pilot Study
Next Article in Special Issue
The Importance of Fatty Acids as Nutrients during Post-Exercise Recovery
Previous Article in Journal
Characterizing the Composition of the Pediatric Gut Microbiome: A Systematic Review
Previous Article in Special Issue
Lifestyle Interventions with a Focus on Nutritional Strategies to Increase Cardiorespiratory Fitness in Chronic Obstructive Pulmonary Disease, Heart Failure, Obesity, Sarcopenia, and Frailty
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mutual Interactions among Exercise, Sport Supplements and Microbiota

Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 (PU) Urbino, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this review.
Equally contributing senior authors.
Nutrients 2020, 12(1), 17; https://doi.org/10.3390/nu12010017
Submission received: 25 October 2019 / Revised: 10 December 2019 / Accepted: 17 December 2019 / Published: 20 December 2019
(This article belongs to the Special Issue Nutrition and Exercise Metabolism)

Abstract

:
The adult gut microbiota contains trillions of microorganisms of thousands of different species. Only one third of gut microbiota are common to most people; the rest are specific and contribute to enhancing genetic variation. Gut microorganisms significantly affect host nutrition, metabolic function, immune system, and redox levels, and may be modulated by several environmental conditions, including physical activity and exercise. Microbiota also act like an endocrine organ and is sensitive to the homeostatic and physiological changes associated with training; in turn, exercise has been demonstrated to increase microbiota diversity, consequently improving the metabolic profile and immunological responses. On the other side, adaptation to exercise might be influenced by the individual gut microbiota that regulates the energetic balance and participates to the control of inflammatory, redox, and hydration status. Intense endurance exercise causes physiological and biochemical demands, and requires adequate measures to counteract oxidative stress, intestinal permeability, electrolyte imbalance, glycogen depletion, frequent upper respiratory tract infections, systemic inflammation and immune responses. Microbiota could be an important tool to improve overall general health, performance, and energy availability while controlling inflammation and redox levels in endurance athletes. The relationship among gut microbiota, general health, training adaptation and performance, along with a focus on sport supplements which are known to exert some influence on the microbiota, will be discussed.

1. Introduction

The human adult gut microbiota contains trillions of microorganisms belonging to thousands of different species, and it is recognized as having a pivotal role in overall health and disease of the host [1]. The normal human gut microbiota comprises of two major phyla, namely Bacteroidetes and Firmicutes, and of less present phyla, such as Actinobacteria and Verrucomicrobia [2]. These microorganisms inhabit the human gut and form a complex community interacting with each other and with the host. Several environmental factors shape the gut microbiota; even if a dominant role is played by diet, physical exercise is also emerging as an important modulator [3,4]. Dietary composition influences the composition of gut microbiota: Diets with a low amount of fiber and rich in refined carbohydrates and fat cause a reduction in community diversity with alterations in structure and activity of the microorganisms [5,6,7]. Apart from diet, exercise is considered as one of the main environmental factors possibly influencing gut microbiota composition in animals and humans [3,8,9].
Microbiota acts as an endocrine organ, and the changes in its status induced by exercise have been correlated with modifications in host physiology, metabolism, immunity, and behaviour [10,11,12]. The effects of exercise on gut microbial microorganisms depends on its intensity and timing as well as on the anamnestic characteristics of the subject enrolled. The microbiota can also influence muscle mass, as reported by Ticinesi et al. [13]. Sarcopenia rat models exhibit a reduction of several taxa with anti-inflammatory and pro-anabolic effects in gut microbiota [14]. To compensate for their increased energy consumption and maximize their adaptation to physical loads, athletes must adopt adequate dietary habits, which increasingly include a broad range of sports nutritional supplements. These supplements could also exert specific effects on the microbiota composition, metabolism, and functionality, thus representing another, yet underestimated, variable.
In this review, we analyze the effects of mutual interactions between exercise, sport dietary supplements, and microbiota on the overall general health and performance of athletes.

2. Functions of Microbiota

Metabolomics, metagenomics, transcriptomics, proteomics, and glycomics have significantly contributed to clarifying the gut microbiome composition—which is now almost completely known—and its association with human health [15]. The MetaHIT and Microbiome Project (HMP) consortia worked hard on determining the baseline structure and function of microbiomes, producing a complete list of species and related genes. However, diversities at sub-species and strain levels are still emerging, and several microbial genes remain functionally uncharacterized [8].
The major phyla included in the human microbiome are Firmicutes and Bacteroidetes, but also a large number of minor phyla (i.e., Actinobacteria, Verrucomicrobia, and Proteobacteria) showing high metabolic activity are present [16,17]. Bacteroides, Bifidobacterium, Streptococcus, Enterobacteriaceae, Enterococcus, Clostridium, Lactobacillus, and Ruminococcus are the predominant luminal microorganisms, while Clostridium, Lactobacillus, Enterococcus, and Akkermansia are the predominant mucosa and mucus-associated genera.
The gut microbiota reflects the health status of the colonized organisms since it is involved in nutrient and drug metabolism, intestinal barrier function, and protection from colonization of pathogens, working collaboratively with the immune system. It participates in the signaling network and is involved in barrier functions and in the maintenance of its homeostasis [18].
Some features have been associated with healthy gut microbiota: The Firmicutes:Bacteroidetes ratio has been widely discussed. The Firmicutes:Bacteroidetes ratio undergoes an increase from birth to adulthood and is further altered with advanced age [19]. Ley et al. [20] reported a higher relative proportion of Bacteroidetes in lean people compared to obese ones, and found an increase in this phyla to be associated with weight loss, suggesting that obesity has a microbial component. Other authors failed to find weight loss diet-dependent significant changes in the proportion of Bacteroidetes in obese subjects, but evidenced a reduction in a group of butyrate-producing Firmicutes [21]. No association between BMI and Firmicutes:Bacteroidetes ratio or taxonomic composition in adults was found [22], while Sze et al. [23] detected only a weak association between microbial communities and obesity, evidencing that detection is confounded by a large interpersonal variation in microbiota composition. Recently, a positive association between Bacteroides fragilis and childhood obesity and a decrease in Bacteroidetes phylum related to high BMI were reported. For the Firmicutes phylum, a positive correlation with weight gain was found, and some Firmicutes species are associated with adipose tissue storage, even if further well-designed studies on this topic are required [24]. It has also been demonstrated that a low amount of Proteobacteria together with a high abundance of Bacteroidetes, Prevotella, and Ruminococcus is favourable for health [25].
The most important nutrient source for gut microbiota is dietary indigestible carbohydrates, whose fermentation produces acetate, propionate, and butyrate—named short-chain fatty acids (SCFAs)—the gases H2 and CO2, ammonia, amines, phenols, and energy, which the bacteria use for growth and the maintenance of cellular function [26].
Bacteria of the genus Bacteroides are known to possess very large numbers of genes that encode carbohydrate-active enzymes, such as glycosyl transferases, glycoside hydrolases, and polysaccharide lyases, performing carbohydrate fermentation [27]. The main SCFAs produced are acetic acid (C2), propionic acid (C3), and butyric acid (C4) (ratio 60:20:20), and in lower amounts, lactic acid (mainly L-lactate), 2-hydroxy propanoic acid, valeric, and caproic acids. Acetic and propionic acids are mainly absorbed from the colonic lumen, while butyric acid is the substrate of colonocytes. Inflammatory bowel disease, irritable bowel syndrome, cancer, and other pathologies are strictly related to SCFAs production [28].
Proteolysis is also affected by gut microbiota since several bacteria produce their proteinases and peptidases that work in synergy with human ones; furthermore, histamine is produced from L-histidine and γ-amino butyric acid from glutamate, and important antimicrobial peptides (bacteriocins) are generated [29,30]. Gut microbiota greatly contributes to the synthesis of vitamin K, biotin, folic acid, thiamine, glycans, amino acids, and conjugated linoleic acid (CLA) that possess several health-protective activities [31,32]. The lipid metabolism is influenced by gut microbiota, promoting the lipoprotein lipase activity in adipocytes and alteration of bile acid profile [33]. The up-regulation of the colipase by Bacteroides thetaiotaomicron augments the efficiency of lipid digestion [34]. Furthermore, some species, such as Escherichia coli and Bacteroides intestinalis and Bacteroides fragilis, transform the primary bile acids into the secondary bile acids in the human colon [35]. Baars et al. [36], reported that the sex-specific differences in lipid metabolism are modulated via the gut microbiota in mice and bile acids possibly play a role in the crosstalk between the microbiome and sex-specific regulation of lipid metabolism.
The gut microbiota contributes to the metabolization of polyphenol compounds, assumed with a diet rich in plants, fruit, and plant-derived products, such as cocoa, berries, wine and tea. An important role of gut microbiota on xenobiotic and drug metabolism, therefore in the outcome of pharmacological therapies, has also been reported.
Microbiota composition shows a certain degree of inter-individual differences that can be due to microbiome intrinsic factors (i.e., age-dependent state, compositional state) and microbiome extrinsic factors, such as host extrinsic factors (i.e., lifestyle, dietary habits, physical activity), host intrinsic factors (i.e., genetics), and environmental factors (i.e., vertical maternal transmission) [8]. With regard to age differences, the intestinal composition is greatly determined in childhood and reaches adult shape at three-years of age, influencing immune system functionality [37,38,39]. In adulthood, the human gut microorganisms remain relatively stable over time, exhibiting resilience to disruptors such as stress and acute diseases [40,41], even if some evidence shows that distinct compositional types of the gut microbiome, the enterotypes, can be associated with different responses to perturbations [42,43]. For example, Prevotella copri and Bacteroides vulgatus, that characterize different enterotypes, are involved in insulin resistance [44], and modification on glucose metabolism following a dietary intervention can be influenced by the Prevotella:Bacteroides ratio [8,45]. In the older age, species richness and diversity of the microbiome decreases and inter-individual variability increases, while the resilience to perturbations is reduced [46,47].

3. The Relationships between Microbiota and Exercise

The gut bacterial population is modulated by gender, genetics, age, and ethnicity, i.e., non-modifiable factors, and by modifiable factors such as host health, physical activity, diet, and eventual antibiotic therapies. Campbell et al. [48] showed that exercise manifested a unique effect on the microbiome independent of diet. Physical exercise is linked with the positive modulation of gut microbiota biodiversity; the potential mechanisms by which exercise might alter the gut microbiome have been investigated in animal and human studies.
The modifications of gut microbiota induced by physical exercise are due to the gut transit time [49], the modification of the bile acids profile [50], the production of SCFAs via AMPK activation [9,51], the modulation of the Toll-Like Receptors (TLRs) signaling pathway [52,53], immunoglobulin A (IgA) [54], the number of B and CD4+ T cells, and finally, to the weight loss [55].
Furthermore, regular exercise reduces the heat shock proteins’ response to heat stress, preventing the breakdown of tight junction proteins between intestine epithelial cells. Thus, the exercise represents a hormetic stressor to the gut that stimulates beneficial adaptations and improves the long-term resilience of the gut barrier [56].

3.1. Effect of Exercise on Gut Microbiota Metabolism

Several factors should be considered when analyzing the impact of exercise on microbiota metabolite production, such as age, body fat, and training status of subjects. Exercise effects are amplified and start early in life; alongside an increase in Bacteroidetes and a decrease in Firmicutes, physical activity promotes, more in the young than in adults, an increase in lean body mass through an adaptation of host metabolism mediated by bacteria composition [57]. Furthermore, microbial species promoted by exercise at this age favour optimal brain function development [58]. In a study analyzing active and sedentary women under 40 years of age after exercise training, the changes in several bacterial taxa were significantly correlated with body mass index (BMI) [59]. Even if the microbiota composition of all the participants was changed for a period following exercise, species with known anti-inflammatory properties and the ability to produce SCFAs were higher in lean subjects [59]. Athlete microbiomes have been found to contain distinct microbial compositions defined by the elevated abundance of Veillonellaceae, Bacteroidetes, Prevotella, Methanobevibacter, and Akkermansia [60]. The abundance of taxa involved in energy and carbohydrate metabolism, such as Prevotella and Methanobrevibacter smithii, were found to be significantly higher in professional than amateur cyclists, and were correlated with the frequency of training [61]. Christensen et al. [62] demonstrated in overweight adults, following a fiber and whole-grain-rich diet for six weeks, that Prevotella abundance is predictive of weight loss, suggesting that enterotype should be considered in personalized nutritional strategies to counteract obesity. SCFA production, in particular butyrate, an important marker of gut health, increases after exercise in humans [9]. In fact, women performing three hours of exercise or more per week increased the relative abundance of butyrate-producing taxa, and in particular some species such as Faecalibacterium prausnitzii, Roseburia hominis, and Akkermansia muciniphila, as well as the Coprococcus genus, compared to sedentary counterparts [59]. These species have been related to health-promoting effects. A high abundance of A. muciniphila has been previously described in the microbiota of athletes, while low levels have been linked to metabolic disorders (obesity, metabolic syndrome, and type II diabetes) in patients with inflammatory bowel disease (IBD) [63,64]. Furthermore, the exercise may positively impact the gut mucus layer, an important substrate for mucosa-associated bacteria such as A. muciniphila. Butyrate produced by both R. hominis and F. prausnitzii has a beneficial effect on health with a positive impact on intestinal function and lipid metabolism [65,66]. F. prausnitzii also produces metabolites with anti-inflammatory action [67]. The Coprococcus genus, a butyrate-producing genus, was more abundant in active women, promoting some exercise-related health effects [59]. In another study comparing lean and obese adults participating in a six-week supervised endurance exercise program under dietary control, an increase in butyrate-producing taxa was found only in lean subjects. Furthermore, lean adults had an increase in Faecalibacterium species, while obese adults had a decrease, and Bacteroides species had an opposite trend, confirming the influence of BMI [9]. Interestingly, the abundance of butyrate-producing taxa was higher in individuals with greater levels of aerobic fitness after normalisation for BMI, diet, and age [68].
Physical activity based on long-distance running led to significant increase in the Coriobacteriaceae family. Notably, this bacterial family—along with others—is involved in the conversion of scarcely absorbed dietary polyphenols to bioavailable and bioactive derivatives [69], as well as in the metabolism of bile salts and steroids, in particular the steroid aldosterone 18-glucuronide. This metabolite of aldosterone exerts many important functions, such as cell signaling, fuel and energy storage, and membrane integrity and stability [70].
During and after exercise, significant amounts of lactate are released into the blood. In particular, lactate has an important role in endurance performance, inasmuch as it is used as fuel from several organs and tissues. The more these organs and tissues “learn” to use lactate as a substrate, the more the performance improves [71].
Recently, Scheiman et al. [60] demonstrated that systemic lactate can cross the gut barrier into the lumen and then can be converted into propionate by the genus Veillonella. These authors have recently reported that Veillonella abundance increases in the gut microbiota and its Methylmalonyl-CoA pathway is overexpressed post-exercise; furthermore, they demonstrated that in mice, Veillonella inoculation improved treadmill performance, which was also improved by propionate administered via intracolonic infusion. Thus, these studies suggest that the modulation of SCFA production by gut microbiota affects energy metabolism during exercise and hence contributes to exercise-induced adaptation. These products of microbiota fermentation can also be used as sources of energy in the liver and muscle cells to improve endurance performance by maintaining glycaemia over time [72,73].
Again, it is important to highlight that human gut microbiota composition remains relatively stable over time, exhibiting resilience to perturbations [16], or returning totally or partially to the previous composition after stimulus cessation [74]. The resilience to perturbations implies that positive changes in exercise and dietary habits aimed at inducing positive effects on the microbiome need to be maintained for a long period to be effective. A summary of studies assessing the impact of physical activity or exercise intervention on the human gut microbiome is detailed in Table 1 [9,11,12,48,57,59,60,61,63,68,70,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93].

3.2. Microbiota and Exercise-Induced Stress

Alongside the positive effects of physical activity on gut microbiota documented by the studies previously described, unfavorable changes in host physiology have been described. High-intensity exercise not supported by an adequate training level, or overtraining syndrome, may be a stressor for the organism and may also have a negative effect on gut microbiota [13,94]. This is in agreement with the hormetic effects of reactive oxidative species (ROS) generated by exercise and by specific commensal gut microbiota [95,96,97,98,99], that at physiologically normal levels promote positive effects by participating in specific signaling pathways; while at higher concentrations, attained in the presence of toxic or pathological conditions, exert detrimental effects. Indeed, a recent key discovery in this direction was the demonstration that several species of human commensal gut bacteria are capable of generating low levels of ROS, which may contribute to sharpening the beneficial effects promoted by normal microbiota [99,100]. Modulation of ROS-mediated signaling may occur during quantitative or qualitative changes in the composition of gut microbiota following rapid dietary changes, antibiotic therapy, or probiotic intake [99]: These modulations may consequently modify and/or reduce the generally positive influence of microbiota-derived ROS. Changing from a “beneficial” to a “detrimental” response will depend on many variables, which also include the duration and intensity of muscle effort and the overall antioxidant status of the organism. In this scenario, it is important to consider the novel role of microbiota, which is capable of consistently modulating the diverging effects of ROS.
The basic principle in training theory is to push athletes to increase their training volume and intensity to the limits to maximize their performance. High-intensity exercise, especially if not proportional to training level, has a profound impact on oxidative stress, muscle damage, systemic inflammation, and immune responses [101]. Excessive exercise and inadequate recovery cause physical and psychological stress that are interrelated and lead to performance decline, fatigue, insomnia, anxiousness, inflammation, and immunosuppression [102].
From the gut microbiota perspective, prolonged exercise determines an increased intestinal permeability, altering the gut-barrier function and promoting bacterial translocation from the colon [103,104]. High-intensity exercise can cause changes in the immune response by reducing the gastrointestinal (GI) blood flow, thus increasing tissue hyperthermia and permeability of the GI epithelial wall. Research on exercise and barrier function suggests that the more strenuous the exercise, the greater the barrier disruption. GI illness symptoms, including abdominal pain, nausea, and diarrhoea, might affect 70% of athletes after strenuous exercises, and the frequency is higher in elite athletes than in recreational exercisers [105]. Excessive exercise induces a stress that increases intestinal permeability, which might lead bacteria and their toxic products, including microbiota-derived LPS, to enter into the bloodstream and activate systemic inflammation [106]. The translocated LPS activates TLRs, promoting NF-kB-pathway activation and inflammatory cytokine production, which can eventually result in endotoxemia [13]. These pro-inflammatory cytokines also increase the intestinal permeability through tight junction opening [107].
Exhaustive and acute endurance exercise has been demonstrated to induce apoptosis and altered permeability in a murine model [108]. Karl et al. [109] showed that an intense military training resulted in increased intestinal permeability concomitant with changes in intestinal microbiota composition (increased abundance of less dominant taxa and decreased abundance of more dominant taxa such as Bacteroides) and markers of inflammation. A half marathon race in amateur athletes changed the fecal microbiome functionality, causing a pro-inflammatory profile [70].
In a study in mice on endurance swimming time, Hsu et al. [81] demonstrated a relationship between performance and antioxidant activity of gut microbiota, suggesting that “gut microbiota status could be crucial for exercise performance and its potential action linked with the antioxidant enzyme system in athletes”.
Alongside single species activity and the effects on exercise response, performance could be affected by the ecological community as a whole. Indeed, the gut microbiota has a key role in controlling oxidative stress, inflammatory responses, metabolism, and energy expenditure during intense exercise.
Exercise alters the turnover of molecules involved in metabolic patterns and stimulates the release of neuroendocrine hormones interacting with the gut directly or indirectly through the immune system [110]. Alongside the adaptation of gut microbiota to exercise training, intestinal microbiota’s influence on exercise performance should be considered.
Upper respiratory tract infections (URTI) represent a common problem in sport [105,111]. URTI frequency is higher during intense training or competition, especially in individual sports such as triathlons and marathons, and lower in team sports [112]. Moreover, travel, stress, and low energy can increase URTI risk in athletes [113]. URTI can greatly affect performance, reducing muscular strength and coordination, aerobic capacity, and concentration. Due to the role of gut microbiota in programming the mucosal immune system, it can affect the respiratory tract susceptibility to infections [111]. Indeed, gut microorganisms are able to regulate mucosal sites distal from the intestine through the common mucosal immune system induction of immunoglobulins [114].
The notion that intestinal microbiota influence respiratory health is also highlighted by recent studies demonstrating that gut health-promoting products, such as probiotics and prebiotics, might reduce susceptibility to URTI [115,116], as detailed in the Section 5.2.
The activation of sympatho-adrenomedullary and the hypothalamus–pituitary–adrenal axis during exercise leads to catecholamine and glucocorticoid release [117]; furthermore, the autonomic nervous system increases neurotransmitters in peripheral tissues, such as the GI tract, and interacting with the enteric nervous system, regulates the passage of material through the GI tract [118]. The gut itself produces hormones such as gamma aminobutyric acid and neuropeptide Y [119], and microbiota are able to secrete molecules such as SCFAs and tryptophan [120,121] which can, in turn, modulate inhibitory and excitatory neurotransmitters and neurotransmitter-like molecules [122]. Dopamine production during exercise-induced stress is released in the GI tract depending on the levels of tyrosine, the type of stress, sex, and the presence of enteric bacteria capable of producing this non-essential amino acid [123]. The release of stress hormones can result in increased intestinal permeability.
Fatigue is an important limiting factor during strenuous exercise; it is a complex phenomenon resulting from muscle exhaustion and modification in the central nervous system linked to increased serotonin levels. The presence of elevated cerebral serotonin levels observed in rats under fatigue [124] is the basis of a well-accepted theory to account for the onset or increase of central fatigue in humans. Indeed, during sustained exercise, increased brain uptake of the serotonin precursor tryptophan has been observed in humans [125,126]. This theory has recently been bolstered by Kavanagh et al. [127], whose study based on paroxetine administration in humans demonstrated the influence of serotonin availability in increasing central fatigue under prolonged maximal contractions. It is also worth noting that dopamine is an important factor involved in the motivation of athletes [128]. The larger amount of secreted serotonin is produced by intestinal enterochromaffin cells, but a significant proportion is produced through gut microbiota synthesis [129]. As such, when dopamine levels decrease together with increased serotonin, it may trigger a state of increasing tiredness during exhaustive exercise, and be involved in fatigue insurgence during exercise impairing performance [130]. The maintenance of a healthy microbiota can also influence an athlete’s mood, motivation, inflammation, and susceptibility to infections: All of these effects improve performance and reduce fatigue onset and perception.
As above-mentioned, diet can greatly affect microbiota composition; the America Dietetic Association suggest a moderate to high intake of animal proteins to restore muscle, a high amount of simple carbohydrates to maintain glucose homeostasis, and a moderate intake of fats and fibers to reduce GI problems in athletes [131]. In addition, animal proteins can negatively affect microbiota through the production of toxic by-products, while a high simple carbohydrate diet leads to a reduction of beneficial SCFAs and immune function [132]. Furthermore, insufficient fibers and resistant starch can cause a reduction in microbiota diversity and functionality [133] (see also 5.3 for the effects of protein intake and supplementation).

4. Effects of Dietary Supplementation on Gut Microbiota Profiles: Can They Affect Performance?

The composition of the diet and food-associated microbes in athletes can influence the composition of gut microorganisms able to improve energy metabolism, oxidative stress, and systemic inflammation status, and the microbial species that produce toxic metabolites from protein degradation [134].
With this in mind, we will describe and discuss the supplements that have been reported to have some effects on microbiota composition, and discuss what the implications of such interactions may be for physical exercise and sport practice. To this end, we narrowed our search to the agents and supplements that have been recognized as useful and beneficial to athletes’ health and/or performance by Close et al. [135] (Table 2), and their effects on microbiota are also summarized in Table 3.

5. Sports Supplements with Strong Evidence of Interactions with Microbiota

5.1. Antioxidants (Polyphenols)

It is known that exhaustive exercise may lead to a strong increase in ROS, which may exceed the endogenous antioxidant capacity and cause severe oxidative damage, muscle weakness, and fatigue. Furthermore, muscle injuries—via cytokine production—recruit and activate neutrophils and macrophages which in turn can produce excessive and/or additional ROS levels [136]. The frequent incidence of the above situations in athletes represents the basis for today’s popularity of antioxidant-containing sport nutrition supplements. A key category among antioxidants are polyphenols, a wide group of natural plant-derived compounds [137], commonly included in supplements intended for sport use (i.e., berry extracts, etc.) and are also known to interact with microbiota. Polyphenols’ biological activity is not limited to their antioxidant capacity: Indeed, they exert other effects, which may be beneficial in supporting sports practice. For example, pomegranate polyphenols are renowned for their anti-inflammatory and anti-infective activity [138].
Once taken orally, polyphenols have been shown to interact with microbiota at different levels [139]. For example, flavonoids can affect and reshape the composition of intestinal flora exerting prebiotic and bactericidal effects and—although the evidence is not conclusive—their systemic anti-inflammatory effects could be at least in part associated with the modulation of microbiota [140,141]. The ‘prebiotic-like’ effects of polyphenols have been observed using both in vitro studies with human gut microbiota, and in vivo in preclinical and clinical trials where supplemental polyphenols and polyphenols-rich foods were shown to modulate gut microbiota, stimulating the growth of beneficial bacteria such as Lactobacilli and Bifidobacteria [139]. Other beneficial species whose growth is favored by polyphenols include Akkermansia, Faecalibacterium prausnitzii, and Roseburia spp. [142,143]. In consideration of the above-mentioned positive interactions between polyphenols and gut microbiota, Marchesi et al. [121] introduced the concept of the “three P’s” for gut health: The three P’s stand for probiotics, prebiotics, and polyphenols. Hence polyphenols were promoted at the same biological level as prebiotics.
Unfortunately, many natural polyphenols, such as condensed or hydrolysable tannins and glycosylated polyphenols derivatives (bound with sugars such as glucose, galactose, rhamnose, ribulose, arabinopyranose, and arabinofuranose) are characterized by low gut absorption in humans. The reduced oral bioavailability represents a severe limit to the potential beneficial effects of these compounds [144]. Interestingly, these polyphenols, which usually remain inactive in the diet, are biotransformed to active compounds after removal of the sugar moiety by gut microbiota [2]. These metabolites may retain parental compounds’ antioxidant and pleiotropic activity, whilst also exhibiting increased gut absorption and better bioavailability. Hence, flavonoids, through the biotransformation operated by microbiota, can more easily reach the bloodstream and exert their biologically-relevant effects at the systemic level [145].
With regard to these effects, the traditional point of view of the health benefits attributed to the polyphenol fraction of foods and herbal supplements should be redirected towards their absorbable gut microbiota metabolites.
On the whole, it can be concluded that the reciprocally positive interactions between microbiota and polyphenols might converge toward the promotion of beneficial conditions supporting athletes in sport practice.

5.2. Probiotics

Probiotics are live non-pathogenic microorganisms, which, when administered in adequate amounts, confer microbial balance, particularly in the GI tract [146]. They have received a renewed interest in the past years in athletes, since they can contribute to general health and, indirectly, sustain and/or improve performance. Probiotics have been demonstrated to lower the frequency and duration of diarrhea and to stimulate humoral and cellular immunity [147]; furthermore, they show significant antioxidant activity both in vivo and in vitro [148,149]. As previously discussed, immune perturbations caused by heavy training and competition, increase susceptibility to URTI and impact the GI, and hence greatly affect performance [150]. Probiotics are effective in decreasing susceptibility to URTI and in reducing their incidence and/or their duration or severity [115,116,151] depending on factors such as the type of sport, athletes’ training status, and kind and duration of supplementation [152]. These effects can be explained by modulation of serum cytokines and secretory immunoglobulin A (SIgA), and by changes in number and activity of innate and adaptive immune cells [116,153]. Regular consumption of probiotics can modify intestinal epithelial cell proliferation and reduce permeability, increasing the expression of mucin genes, and mucin and antimicrobial peptide secretion, enhancing the barrier function of the mucosa [154]. Lamprecht et al. [155] analyzed the effects of a multi-species probiotic supplementation on markers of the intestinal barrier, oxidation and inflammation, at rest and after intense exercise in a randomized, double-blinded, placebo-controlled trial. They found that zonulin, a marker of enhanced gut permeability, decreased to normal range after supplementation for 14 weeks, and TNF-α and exercise-induced protein oxidation were also positively affected. Thus, probiotic supplementation can modulate intestinal barrier function, redox homeostasis, and low-grade inflammation after sustained exercise.
Roberts et al. [156] examined the effect of a multistrain probiotic/prebiotic/antioxidant intervention for 12 weeks preceding a long-distance triathlon on endotoxin unit levels and GI permeability in recreational athletes. They divided subjects into three groups: One group received a probiotic/prebiotic/antioxidant supplement (LAB4ANTI), one group received a pre/probiotic supplement only (LAB4), and the third group was a placebo; values were assessed at baseline, pre-race and six days post-race. The pre/probiotic supplementation (LAB4 and LAB4ANTI) significantly reduced endotoxin levels six days after the race, but only LAB4ANTI was effective pre-race. Even if faster times were registered in both supplemented groups, there was no statistically significant difference in mean race times. The authors concluded that “combined multistrain pro/prebiotic use may reduce endotoxin unit levels, with LAB⁴ANTI potentially conferring an additive effect via combined GI modulation and antioxidant protection”.
Investigations have revealed that Lactobacillus spp. has various biological effects, for instance, Lactobacillus rhamnosus CNCMI–4317 was able to regulate cellular function and maintenance, lymphoid tissue structure and development, and immune system response [157]; furthermore, Lactobacillus spp., producing lactic acid, lead lactate-utilizing bacteria to produce butyrate [158].
Recently, the possible ergogenic and anti-fatigue effects of probiotics have been investigated. L. plantarum TWK10 has been shown to promote an increase in muscle mass, performance, and resistance to fatigue in supplemented mice [159]. In a human study, Huang et al. [160] analyzed the effect of six weeks TWK10 supplementation during exhaustive exercise. The probiotics significantly improved endurance performance and glucose content, suggesting that TWK10 can be beneficial to energy harvest. Thus, in addition to the well-documented antioxidant action, supplemental TWK10 might have an ergogenic anti-fatigue function [161,162]. However, few studies have focused on lactobacilli (probiotic) and their interaction with gut microbiota, and the possible ergogenic anti-fatigue function. The previously described results of Scheiman et al. [60] on Veillonella effects on athletic results can suggest a possible use of these strains as a supplement to improve performance. In a similar direction, Soares et al. [163] recently highlighted the benefits to the host metabolism resulting from Saccaharomyces boulardii (Sb) supplementation. In this study, the authors evaluated the effect of Sb supplementation on the rate of oxygen consumption (VO2), mechanical efficiency, and aerobic performance of Wistar rats subjected to fatiguing, incremental-speed exercise, and found that supplemental Sb does not affect resting aerobic metabolism, but significantly increases VO2 max and aerobic performance.
Among the most popular probiotics, kefir (KF), a drink rich in lactic ferments obtained from the fermentation of milk, has been proposed against hypertension, GI diseases, allergies, ischemic heart disease [164,165], fatigue and fatigue-related metabolites [166,167], and exercise-induced immune suppression [168]. KF is able to lower plasma lactate, ammonia, and creatine kinase (CK) levels, increasing the exercise performance and counteracting physical fatigue in mice [169], altering gut microbiota composition.
Interestingly probiotics are known to promote an improvement of mood state and disorders [170] and, based on literature [171], it is conceivable that the probiotic-mediated improvement of mood status may indirectly and positively impact on athlete’s conditions. The mechanisms whereby microbiota can influence mood, and more generally, CNS, have been comprehensively reviewed by Bravo et al. [172]. Although there are very few studies specifically dealing with this issue, based on the prevalence of animal studies, some authors reported beneficial effects of gut microbiota health on mood disorders which impact on performance, such as anxiety and depression [173,174,175,176]. The effects of a six-month administration of a probiotic multivitamin preparation in adults suffering from stress or exhaustion were studied by Gruenwald et al. [174]. The results showed a significant improvement in the general conditions of those who participated in the study, including a 41% improvement in perceived stress. Messaoudi et al. [175] investigated the similar-anxiolytic effects of Lactobacillus helveticus and Bifidobacterium longum supplementation on healthy human volunteers. The authors reported that a 30-day supplementation regimen promoted beneficial psychological responses. Rao et al. [176] investigated the effects of probiotic intake on anxiety and depression in chronic fatigue syndrome patients: Eight-week consumption of Lactobacillus casei resulted in a significant reduction in anxiety scores as compared to a placebo group. Finally, a multispecies probiotic supplementation was reported to positively affect the cognitive reactivity to a sad mood [174]. Although the participants in this study had no diagnosis of mood disorders, those who consumed the probiotic over four weeks significantly reduced their cognitive reactivity to sad mood, i.e., a condition that may favour progression into clinical depression. As to animal studies, although more research work is still needed, similarly to humans, there is some evidence indicating that gut microbiota health and composition affect animal depressive symptoms and behavior [177,178,179]. On the whole, the results of the relatively few animal and human studies suggest that the use of probiotics might be beneficial to mood, particularly in individuals with a history of mood disorders. In a different direction, there are also studies showing a correlation between physical activity and stress levels, with physical activity generally promoting beneficial effects on mood and affective state [180,181]. From a biochemical perspective, a positive correlation between physical exercise and the levels of tryptophan was described by Gostner et al. [182]: The authors suggested that physical exercise allows for increased levels of tryptophan which—once taken-up by the brain—may boost the synthesis of serotonin, subsequently leading to an increase in mood state. On the whole, it is conceivable that combining probiotic supplementation to maintain good microbiota conditions with an appropriate level of physical exercise may have additive—if not synergistic—beneficial effects on mood.

5.3. Proteins

Athletes are subjected to multi-stress conditions of regular, intensive, and/or prolonged exercise routines; to promote protein synthesis and the consequent adaptations to training, they need an adequate protein intake. Proteins should be consumed before and after the performance and regularly during the day to guarantee an efficient supply of essential amino acids [183], especially when increasing intensity and duration of the training session and competitions.
To clarify the impact of proteins on gut microbiota composition and functionality, protein quantity, quality, processing history (which impacts on protein digestion, presentation, and overall function), and source must be taken into consideration [184]. Protein sources with a different amino acid composition can impact differently on gut microbiota. Some studies suggest that plant-derived proteins are more useful than animal-derived proteins for microbiota, and consequently, for host metabolism. A high animal protein consumption has been linked to the Bacteroides enterotype [7].
Beaumont et al. [185] studied the effect of a three-weeks isocaloric supplementation with casein, soy-protein, or a maltodextrin compound in overweight humans. They reported that the high-protein diets had no effect on the bacterial composition and diversity of fecal microbiota. However, interestingly, both high-protein diets caused a shift of fecal and urinary metabolite (amino acid-derived isobutyrate, microbiota-host cometabolites indoxyl sulphate, and phenylacetylglutamine) concentration produced by gut microbiota. A decrease in fecal butyrate in both high-protein groups was found, suggesting a possible negative effect of a high-protein intake on colonic health. The two high-protein diets also led to an increase in the concentration of amino acid-derived bacterial metabolites, due to higher protein degradation by the gut microbiota.
Moreno-Perez et al. [186] evaluated the effect of 10-week protein supplementation (10 g of whey isolate plus 10 g of beef hydrolysate protein per day) on gut microbiota in recreational endurance athletes. Although there was no effect of SCFAs, ammonia, or faecal pH, they reported a decrease in health-promoting bacteria (Roseburia, Blautia, and Bifidobacterium longum); based on these data, the authors concluded that long-term protein supplementation might harm the gut microbiota of athletes. The effects of long-term high-protein supplement consumption on intestinal microbiota and amino acid fermentation have not been further investigated in recreational sports people and lifestyle users of protein supplements.
Microbial diversity has been found to positively correlate with protein intake and exercise in elite athletes [63], and thus, exercise training is suggested to be essential for healthy and balanced gut microbiota, and muscle mass and function. According to these observations, well-balanced protein intake should support not only athletic performance but also the well-being of the gut [187]. Although the physiological dietary protein requirements are elevated for athletes, high-protein diets may affect the gut microbiota and protein fermentation levels [94], with consequences for health [187]. Effects of protein supplementation for microbiota in athletes should be taken into consideration since nutritional recommendations regarding protein consumption in this population are higher than for the general population. The long-term consequences of the decrease in these bacterial taxa (Blautia, Roseburia, and Bifidobacterium longum) for gut health are unknown [186].

6. Sports Supplements with Moderate/Emerging Evidence of Interactions with Microbiota

6.1. Branched Chain Amino Acids (BCAAs)

Studies regarding the influence of BCAA supplementation on gut microbiota are still limited to animal studies, where several beneficial effects have been documented in pigs and mice [188,189,190,191,192]. Recently, it has been reported that dietary supplementation with BCAAs promotes intestinal development, enhances enterocyte proliferation, increases intestinal absorption of amino acids (AA) and glucose, and improves the immune defenses of piglets [193]. Yang et al. [194] reported an increased abundance of Akkermansia and Bifidobacterium and a decreased level of Enterobacteriaceae, together with changes in lipid and sugar metabolism in BALB/C mice supplemented with a BCAA-enriched mixture (1.5 mg/g body weight per day, beginning at the 11th to the 15th month of age). The Akkermansia genus has been reported to reduce metabolic disorders and insulin resistance by reversing the effects of high fat diets, and by enhancing intestinal levels of endocannabinoids controlling inflammation, as well as enhancing the intestinal barrier and intestinal peptide secretion [195]. However, since the above data have been obtained in animals, specific studies are needed to understand the effects of BCAA supplementation on gut microbiota in humans.

6.2. Glutamine

Glutamine is the most abundant amino acid in human blood, skeletal muscle, and the free amino acid pool [196]. At the intestinal level, it promotes enterocyte proliferation, regulates tight junction proteins [197], improves intestinal barrier function and reduces intestinal inflammatory response [198]. Glutamine is widely used in sports nutrition; in particular, its supplementation seems to increase muscle glycogen synthesis, reduce ammonia accumulation induced by exercise, and attenuate markers of muscle damage [199]. Analyzing the intervention of oral intake of 30 g per day of L-glutamine for a short period (14 days) in overweight and obese adults, de Souza et al. [200] noticed some alteration in the composition of gut microbiota. First, the authors reported a reduced Firmicutes:Bacteroidetes ratio compared to the control group; moreover, the number of bacteria from the Veillonella genus decreased, whilst that of Prevotella genus increased. To this regard, it has been reported that an increased amount of Veillonella is associated with higher levels of gut inflammation and the development of colitis and Crohn’s disease [201,202]; on the contrary, an increased the abundance of the Prevotella genus has been described as a shield against inflammation and non-infectious diseases of the colon [132]. Furthermore, a recent study by Abboud et al. [203] showed that 14 days of 30 g of glutamine supplementation induced a significant diminution in the waist circumference and circulating LPS levels in overweight subjects, and reduced the insulin levels in obese subjects. These findings suggest that glutamine supplementation in humans may promote a healthier microbiota composition with anti-inflammatory implications; the latter probably due to the decrease of Veillonella and increase of Prevotella genera. However, although these beneficial effects are well-documented in studies carried out on animals [204], the literature in humans is still scarce, making it necessary to develop more studies.

6.3. Sodium Bicarbonate

Sodium bicarbonate is a cheap and widely available household product. The consumption of bicarbonate-rich mineral water is used as a sport supplement to improve performance [205]. This supplement, taken before training or competition, is reputed to enhance intracellular and/or extracellular buffering capacity by managing the progressive increase in intracellular acidosis during intense exercise [206]. Microbiome analysis demonstrated that the consumption (500 mL per day, for 7 days) of bicarbonate-rich mineral water (ca. 2.5 g/l of bicarbonate ions) might promote a reshape of microbiota, increasing lean-inducible bacteria (Christensenellaecae and Dehalobacteriaceae) and decreasing Bifidobacteriaceae [207]. Interestingly, higher Christensenellaceae abundance has been previously reported in lean subjects (BMI < 25) as compared to obese ones (BMI > 30) [208].

6.4. Vitamin D

Vitamin D is known to significantly contribute to skeletal mineralization regulating calcium and phosphorus-homeostasis, and promoting endocrine effects on bone, intestine, parathyroid glands and kidney [209]. There is also growing evidence that Vitamin D regulates many other cell functions, and its potential impact on skeletal muscle mass and strength is receiving more considerable attention. The biological actions of vitamin D on muscle cell differentiation, metabolism, and function may be multiple, acting through direct and indirect, genomic and non-genomic pathways [210]. Furthermore, athletes may exhibit vitamin D deficiency, particularly during the winter in northern countries [211]. For these reasons, vitamin D supplementation is one of the most popular in sports nutrition. An additional and emerging rationale for its supplementation might be that vitamin D deficiency negatively affects the intestinal microbiome, hampering its capacity to produce B vitamins in the intestine, whose reduced availability may negatively impact the immune system [212].
An experimental study conducted on healthy volunteers [213] showed that high-dose (≥34,300 IU/week for eight weeks) intakes of oral vitamin D caused, in the upper gastrointestinal tract, a decline in Proteobacteria and an increase in Bacteroidetes, and in general, in bacterial richness. In parallel, a decrease of opportunistic pathogens like Pseudomonas and Escherichia/Shigella was observed. More recently, a comprehensive review [214] found evidence in the literature indicating that vitamin D influences the composition of the gastrointestinal microbiome, supporting that vitamin D supplementation could be useful in healthy subjects and hence, also in athletes. However, given the paucity and heterogeneity of the studies conducted so far, and the emerging importance in health and disease of the relationships between vitamin D and the microbiome, further and specifically designed studies and trials are warranted.

6.5. Omega-3 and Polyunsaturated Fatty Acids (PUFAs)

Polyunsaturated fatty acids (PUFAs) are known to maintain the fluidity of cell membranes and regulate cell signaling, gene expression, and cellular function, also serving as substrates for the synthesis of lipid mediators. The effects of PUFAs on gut microbiota are not well defined. However, experimental studies suggested that omega-3 PUFAs work with host immune cells to ensure intestinal wall integrity; furthermore, they can work with microbiota in regulating inflammation and the immune system [215,216]. Omega-3 PUFAs can exert anti-inflammatory effects post-exercise, interacting with prostaglandin synthesis [217], and can affect gut integrity by improving epithelial barrier function in animal models [218]. They have also been shown to decrease the Firmicutes:Bacteroidetes ratio and the levels of Coprococcus and Faecalibacterium; increasing the abundance of Bifidobacterium, Lachnospira, Roseburia, and Lactobacillus and other butyrate-producing bacterial genera [219,220,221]. Furthermore, omega-3 PUFA plasma levels correlate with the Lachnospiraceae family, a SCFA-producing bacteria [219]. Neuropsychiatric disorders and dysbiosis induced by social instability stress during adolescence were prevented by supplementation with omega-3 and the protective effect was long-term maintained through adulthood [222,223]. Since omega-3 PUFAs are known to modulate the gut microbiota composition, they have been recently proposed as prebiotics [224].

6.6. Carbohydrate-Electrolyte Sport Drinks

Carbohydrate sports drinks provide worthwhile benefits to endurance performance compared with noncaloric control beverages [225]. Carbohydrates in different formulations (liquid, gels, or solid) are frequently associated with electrolytes. Moreover, some authors reported that these supplements could increase gut discomfort depending on the intestinal absorption, formulation, and or/the co-presence of specific components [226]. However, there are no studies aimed at highlighting changes in the abundance of the different bacterial strains of intestinal flora compared to the different formulations of carbohydrate (CHO)-sport drinks, therefore specific studies are needed to clarify their interactions with microbiota.

6.7. L-Carnitine

L-carnitine-containing supplements are popular and widely used by athletes to increase physical performance [227]—due to its role in mitochondrial fatty-acid oxidation—and to facilitate recovery from exercise [228]. Carnitine may be absorbed both in the small and in the large intestine. The gut microbial population may influence the absorption rate of carnitine by the large intestine: Indeed, a high intestinal bacterial population can inhibit carnitine absorption. Carnitine plays a role in maintaining the ability of the colonic microbiota of fiber fermentation; furthermore, low dietary fiber intake can affect the metabolic carnitine pathways that have been linked to an increased risk of atherosclerosis and cardiovascular diseases [229]. Indeed, L-carnitine might be converted into atherosclerosis- and thrombosis-promoting metabolites via gut microbiota-dependent transformations [230,231,232]; the extent of this transformation is dependent on the diet habit, i.e., omnivorous > vegan/vegetarian diets. However, these results should be taken with caution due to the low sample size in these studies. On the other hand, carnitine may instead have a beneficial effect on metabolic health and cardiovascular function via stimulation of glucose oxidation, improved glucose tolerance, and insulin sensitivity [230].

6.8. Caffeine

Caffeine consumption is very popular among sports practitioners due to its supposed beneficial effects on performance. Indeed, caffeine, acting as a competitor for adenosine in CNS, is reputed to hamper the negative effects that adenosine causes on neurotransmission, excitation, and pain perception [233]. However, although caffeine supplementation is widely utilized by athletes and there is vast literature on its biological and pharmacological effects, its influence on gut microbiota is not well known. Caffeine can be taken in various and very normal ways as it is contained in coffee, multiple drinks, extracts, and foods. To date, there are few studies on the possible effects of caffeine intake on the composition of gut microbiota. For instance, Jaquet et al. [234] found that a daily dose of three cups of coffee (over three weeks in 16 healthy adults) promoted an increase in the number of the reportedly beneficial Bifidobacteria strains. However, as declared by the authors themselves, this study was affected by some limitations, such as the absence of control and placebo. More recently, Janssens et al. [235] found that Green Team (GT) supplementation (corresponding to a daily intake of 0.27/0.45 g caffeine) for 12 weeks did not have a significant effect on the composition of the gut microbiota; no significant differences in the fecal bacterial diversity and community structure were observed between treatment (green tea or placebo) and time (baseline and week 12) for the measured variables. Therefore, they concluded that catechin- and caffeine-rich GT supplementation has no long-term effects on the composition of the gut microbiota in healthy normal-weight and overweight subjects.
However, the main limitation of both studies, which gave apparently conflicting results, is that they investigated the effects of products (coffee or green tea dry extract) containing not only caffeine, but also a complex mixture of other bioactive compounds such as polyphenols. Hence, to date, although caffeine is a prominent ingredient in many popular sports supplements, it is not yet possible to establish if it may produce any significant effect on microbiota composition.

7. Conclusions

In summary, intensity, timing, and type of exercise can influence the composition of gut microbiota, as it also relates to gender, age, health, and training status of subjects. It has been demonstrated that physical activity performed at low levels, but continuously, can increase microbiota diversity, improving the metabolic profile and immunological responses of the subjects, while acute strenuous exercise may cause deleterious effects on the athletes’ microbiota and his general health.
Gut microbiota composition can influence training adaptation and athletic performance: Indeed, it is involved in the metabolism and delivery of nutrients, hormones, and vitamins important to sustain exercise. Furthermore, specific gut bacterial species can be an aid for athletes by producing beneficial metabolites, such as the antioxidant bioactive molecules, SCFAs, which may help to improve metabolic, immune and barrier function, and anti-inflammatory and antioxidant systemic effects. On the whole, good gut microbiota conditions positively affect athletes’ health with beneficial consequences on their training adaptation and performances (Figure 1).
Some supplements intended for sport use contain probiotics, proteins, polyphenols, and other compounds which are known to interact with microbiota at multiple levels, for example in promoting the selection and growth of healthy specific microorganisms, such as Bifidobacteria and Lactobacillus, and at the same time, limiting the bacterial species that produce toxic metabolites.
To this end, the design of methods, including adequate and rational supplementation, capable of implementing the richness and metabolic capacity of the microbiota could be a useful strategy to improve athletes’ health, training response, adaptations, and ultimately, performance. Disappointingly, the interactions and interplay between exercise, gut microbiota, and sport nutritional supplements are still largely unknown, and should be a research focus in the near future.

Author Contributions

Conceptualization, S.D.Z. and D.A.; Methodology, S.A., D.S.; Resources, S.D.Z., D.A., M.G., G.A., S.A., F.F., G.P. and E.B.; Writing—Original Draft Preparation, S.D.Z. and D.A.; Writing—Review & Editing, S.D.Z., D.A., G.A., S.A. and P.S.; Supervision, P.S. and V.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partly granted by “Progetti di Valorizzazione DISB 2017”.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mitchell, C.M.; Davy, B.M.; Hulver, M.W.; Neilson, A.P.; Bennett, B.J.; Davy, K.P. Does Exercise Alter Gut Microbial Composition? A Systematic Review. Med. Sci. Sports Exerc. 2019, 51, 160–167. [Google Scholar] [CrossRef] [PubMed]
  2. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef] [PubMed]
  3. Monda, V.; Villano, I.; Messina, A.; Valenzano, A.; Esposito, T.; Moscatelli, F.; Viggiano, A.; Cibelli, G.; Chieffi, S.; Monda, M.; et al. Exercise Modifies the Gut Microbiota with Positive Health Effects. Oxid. Med. Cell. Longev. 2017, 2017, 3831972. [Google Scholar] [CrossRef] [PubMed]
  4. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [Green Version]
  5. Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef]
  6. Sonnenburg, E.D.; Smits, S.A.; Tikhonov, M.; Higginbottom, S.K.; Wingreen, N.S.; Sonnenburg, J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016, 529, 212–215. [Google Scholar] [CrossRef] [Green Version]
  7. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef] [Green Version]
  8. Schmidt, T.S.B.; Raes, J.; Bork, P. The Human Gut Microbiome: From Association to Modulation. Cell 2018, 172, 1198–1215. [Google Scholar] [CrossRef]
  9. Allen, J.M.; Mailing, L.J.; Niemiro, G.M.; Moore, R.; Cook, M.D.; White, B.A.; Holscher, H.D.; Woods, J.A. Exercise Alters Gut Microbiota Composition and Function in Lean and Obese Humans. Med. Sci. Sports Exerc. 2018, 50, 747–757. [Google Scholar] [CrossRef]
  10. Choi, J.J.; Eum, S.Y.; Rampersaud, E.; Daunert, S.; Abreu, M.T.; Toborek, M. Exercise attenuates PCB-induced changes in the mouse gut microbiome. Environ. Health Perspect. 2013, 121, 725–730. [Google Scholar] [CrossRef]
  11. Kang, S.S.; Jeraldo, P.R.; Kurti, A.; Miller, M.E.; Cook, M.D.; Whitlock, K.; Goldenfeld, N.; Woods, J.A.; White, B.A.; Chia, N.; et al. Diet and exercise orthogonally alter the gut microbiome and reveal independent associations with anxiety and cognition. Mol. Neurodegener. 2014, 9, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Evans, C.C.; LePard, K.J.; Kwak, J.W.; Stancukas, M.C.; Laskowski, S.; Dougherty, J.; Moulton, L.; Glawe, A.; Wang, Y.; Leone, V.; 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, e92193. [Google Scholar] [CrossRef] [PubMed]
  13. Ticinesi, A.; Lauretani, F.; Tana, C.; Nouvenne, A.; Ridolo, E.; Meschi, T. Exercise and immune system as modulators of intestinal microbiome: Implications for the gut-muscle axis hypothesis. Exerc. Immunol. Rev. 2019, 25, 84–95. [Google Scholar] [PubMed]
  14. Siddharth, J.; Chakrabarti, A.; Pannérec, A.; Karaz, S.; Morin-Rivron, D.; Masoodi, M.; Feige, J.N.; Parkinson, S.J. Aging and sarcopenia associate with specific interactions between gut microbes, serum biomarkers and host physiology in rats. Aging 2017, 9, 1698–1720. [Google Scholar] [CrossRef] [Green Version]
  15. Donovan, S.M. Introduction to the special focus issue on the impact of diet on gut microbiota composition and function and future opportunities for nutritional modulation of the gut microbiome to improve human health. Gut Microbes 2017, 8, 75–81. [Google Scholar] [CrossRef]
  16. Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef] [Green Version]
  17. Greenhalgh, K.; Meyer, K.M.; Aagaard, K.M.; Wilmes, P. The human gut microbiome in health: Establishment and resilience of microbiota over a lifetime. Environ. Microbiol. 2016, 18, 2103–2116. [Google Scholar] [CrossRef]
  18. Zhang, Z.; Tang, H.; Chen, P.; Xie, H.; Tao, Y. Demystifying the manipulation of host immunity, metabolism, and extraintestinal tumors by the gut microbiome. Signal Transduct. Target. Ther. 2019, 4, 41. [Google Scholar] [CrossRef]
  19. Mariat, D.; Firmesse, O.; Levenez, F.; Guimaraes, V.; Sokol, H.; Dore, J.; Corthier, G.; Furet, J.P. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009, 9, 123. [Google Scholar] [CrossRef]
  20. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef]
  21. Duncan, S.H.; Lobley, G.E.; Holtrop, G.; Ince, J.; Johnstone, A.M.; Louis, P.; Flint, H.J. Human colonic microbiota associated with diet, obesity and weight loss. Int. J. Obes. 2008, 32, 1720–1724. [Google Scholar] [CrossRef] [Green Version]
  22. Finucane, M.M.; Sharpton, T.J.; Laurent, T.J.; Pollard, K.S. A taxonomic signature of obesity in the microbiome? Getting to the guts of the matter. PLoS ONE 2014, 9, e84689. [Google Scholar] [CrossRef] [Green Version]
  23. Sze, M.A.; Schloss, P.D. Looking for a Signal in the Noise: Revisiting Obesity and the Microbiome. MBio 2016, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Indiani, C.; Rizzardi, K.F.; Castelo, P.M.; Ferraz, L.F.C.; Darrieux, M.; Parisotto, T.M. Childhood Obesity and Firmicutes/Bacteroidetes Ratio in the Gut Microbiota: A Systematic Review. Child. Obes. 2018, 14, 501–509. [Google Scholar] [CrossRef] [PubMed]
  25. Hollister, E.B.; Gao, C.; Versalovic, J. Compositional and functional features of the gastrointestinal microbiome and their effects on human health. Gastroenterology 2014, 146, 1449–1458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2014, 7, 17–44. [Google Scholar] [CrossRef]
  27. Cantarel, B.L.; Lombard, V.; Henrissat, B. Complex carbohydrate utilization by the healthy human microbiome. PLoS ONE 2012, 7, e28742. [Google Scholar] [CrossRef] [Green Version]
  28. Primec, M.; Mičetić-Turk, D.; Langerholc, T. Analysis of short-chain fatty acids in human feces: A scoping review. Anal. Biochem. 2017, 526, 9–21. [Google Scholar] [CrossRef]
  29. De Biase, D.; Pennacchietti, E. Glutamate decarboxylase-dependent acid resistance in orally acquired bacteria: Function, distribution and biomedical implications of the gadBC operon. Mol. Microbiol. 2012, 86, 770–786. [Google Scholar] [CrossRef]
  30. Garcia-Gutierrez, E.; Mayer, M.J.; Cotter, P.D.; Narbad, A. Gut microbiota as a source of novel antimicrobials. Gut Microbes 2019, 10, 1–21. [Google Scholar] [CrossRef] [Green Version]
  31. Baddini Feitoza, A.; Fernandes Pereira, A.; da Costa, N.F.; Goncalves Ribeiro, B. Conjugated linoleic acid (CLA): Effect modulation of body composition and lipid profile. Nutr. Hosp. 2009, 24, 422–428. [Google Scholar] [PubMed]
  32. Devillard, E.; McIntosh, F.M.; Paillard, D.; Thomas, N.A.; Shingfield, K.J.; Wallace, R.J. Differences between human subjects in the composition of the faecal bacterial community and faecal metabolism of linoleic acid. Microbiology 2009, 155, 513–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Boursier, J.; Diehl, A.M. Nonalcoholic Fatty Liver Disease and the Gut Microbiome. Clin. Liver Dis. 2016, 20, 263–275. [Google Scholar] [CrossRef] [PubMed]
  34. Hooper, L.V.; Wong, M.H.; Thelin, A.; Hansson, L.; Falk, P.G.; Gordon, J.I. Molecular analysis of commensal host-microbial relationships in the intestine. Science 2001, 291, 881–884. [Google Scholar] [CrossRef] [Green Version]
  35. Velagapudi, V.R.; Hezaveh, R.; Reigstad, C.S.; Gopalacharyulu, P.; Yetukuri, L.; Islam, S.; Felin, J.; Perkins, R.; Borén, J.; Oresic, M.; et al. The gut microbiota modulates host energy and lipid metabolism in mice. J. Lipid Res. 2010, 51, 1101–1112. [Google Scholar] [CrossRef] [Green Version]
  36. Baars, A.; Oosting, A.; Lohuis, M.; Koehorst, M.; El Aidy, S.; Hugenholtz, F.; Smidt, H.; Mischke, M.; Boekschoten, M.V.; Verkade, H.J.; et al. Sex differences in lipid metabolism are affected by presence of the gut microbiota. Sci. Rep. 2018, 8, 13426. [Google Scholar] [CrossRef] [Green Version]
  37. Gensollen, T.; Iyer, S.S.; Kasper, D.L.; Blumberg, R.S. How colonization by microbiota in early life shapes the immune system. Science 2016, 352, 539–544. [Google Scholar] [CrossRef] [Green Version]
  38. Kabat, A.M.; Srinivasan, N.; Maloy, K.J. Modulation of immune development and function by intestinal microbiota. Trends Immunol. 2014, 35, 507–517. [Google Scholar] [CrossRef]
  39. Thaiss, C.A.; Levy, M.; Suez, J.; Elinav, E. The interplay between the innate immune system and the microbiota. Curr. Opin. Immunol. 2014, 26, 41–48. [Google Scholar] [CrossRef]
  40. McCoy, K.D.; Ignacio, A.; Geuking, M.B. Microbiota and Type 2 immune responses. Curr. Opin. Immunol. 2018, 54, 20–27. [Google Scholar] [CrossRef]
  41. McLean, M.H.; Dieguez, D.; Miller, L.M.; Young, H.A. Does the microbiota play a role in the pathogenesis of autoimmune diseases? Gut 2015, 64, 332–341. [Google Scholar] [CrossRef] [PubMed]
  42. Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef] [PubMed]
  43. Costea, P.I.; Hildebrand, F.; Arumugam, M.; Bäckhed, F.; Blaser, M.J.; Bushman, F.D.; de Vos, W.M.; Ehrlich, S.D.; Fraser, C.M.; Hattori, M.; et al. Enterotypes in the landscape of gut microbial community composition. Nat. Microbiol. 2018, 3, 8–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Pedersen, H.K.; Gudmundsdottir, V.; Nielsen, H.B.; Hyotylainen, T.; Nielsen, T.; Jensen, B.A.; Forslund, K.; Hildebrand, F.; Prifti, E.; Falony, G.; et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 2016, 535, 376–381. [Google Scholar] [CrossRef] [PubMed]
  45. Kovatcheva-Datchary, P.; Nilsson, A.; Akrami, R.; Lee, Y.S.; De Vadder, F.; Arora, T.; Hallen, A.; Martens, E.; Björck, I.; Bäckhed, F. Dietary Fiber-Induced Improvement in Glucose Metabolism Is Associated with Increased Abundance of Prevotella. Cell Metab. 2015, 22, 971–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O’Connor, E.M.; Cusack, S.; Harris, H.M.; Coakley, M.; Lakshminarayanan, B.; O’Sullivan, O.; et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012, 488, 178–184. [Google Scholar] [CrossRef]
  47. O’Toole, P.W.; Jeffery, I.B. Gut microbiota and aging. Science 2015, 350, 1214–1215. [Google Scholar] [CrossRef]
  48. Campbell, S.C.; Wisniewski, P.J.; Noji, M.; McGuinness, L.R.; Häggblom, M.M.; Lightfoot, S.A.; Joseph, L.B.; Kerkhof, L.J. The Effect of Diet and Exercise on Intestinal Integrity and Microbial Diversity in Mice. PLoS ONE 2016, 11, e0150502. [Google Scholar] [CrossRef] [Green Version]
  49. Oettlé, G.J. Effect of moderate exercise on bowel habit. Gut 1991, 32, 941–944. [Google Scholar] [CrossRef] [Green Version]
  50. Hagio, M.; Matsumoto, M.; Yajima, T.; Hara, H.; Ishizuka, S. Voluntary wheel running exercise and dietary lactose concomitantly reduce proportion of secondary bile acids in rat feces. J. Appl. Physiol. 2010, 109, 663–668. [Google Scholar] [CrossRef] [Green Version]
  51. Cerda, B.; Perez, M.; Perez-Santiago, J.D.; Tornero-Aguilera, J.F.; Gonzalez-Soltero, R.; Larrosa, M. Gut Microbiota Modification: Another Piece in the Puzzle of the Benefits of Physical Exercise in Health? Front. Physiol. 2016, 7, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Francaux, M. Toll-like receptor signalling induced by endurance exercise. Appl. Physiol. Nutr. Metab. 2009, 34, 454–458. [Google Scholar] [CrossRef] [PubMed]
  53. Frosali, S.; Pagliari, D.; Gambassi, G.; Landolfi, R.; Pandolfi, F.; Cianci, R. How the Intricate Interaction among Toll-Like Receptors, Microbiota, and Intestinal Immunity Can Influence Gastrointestinal Pathology. J. Immunol. Res. 2015, 2015, 489821. [Google Scholar] [CrossRef] [PubMed]
  54. Viloria, M.; Lara-Padilla, E.; Campos-Rodríguez, R.; Jarillo-Luna, A.; Reyna-Garfias, H.; López-Sánchez, P.; Rivera-Aguilar, V.; Salas-Casas, A.; Berral de la Rosa, F.J.; García-Latorre, E. Effect of moderate exercise on IgA levels and lymphocyte count in mouse intestine. Immunol. Investig. 2011, 40, 640–656. [Google Scholar] [CrossRef] [Green Version]
  55. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
  56. Mailing, L.J.; Allen, J.M.; Buford, T.W.; Fields, C.J.; Woods, J.A. Exercise and the Gut Microbiome: A Review of the Evidence, Potential Mechanisms, and Implications for Human Health. Exerc. Sport Sci. Rev. 2019, 47, 75–85. [Google Scholar] [CrossRef]
  57. Mika, A.; Van Treuren, W.; González, A.; Herrera, J.J.; 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 2015, 10, e0125889. [Google Scholar] [CrossRef]
  58. Stilling, R.M.; Ryan, F.J.; Hoban, A.E.; Shanahan, F.; Clarke, G.; Claesson, M.J.; Dinan, T.G.; Cryan, J.F. Microbes & neurodevelopment—Absence of microbiota during early life increases activity-related transcriptional pathways in the amygdala. Brain Behav. Immun. 2015, 50, 209–220. [Google Scholar] [CrossRef]
  59. Bressa, C.; Bailén-Andrino, M.; Pérez-Santiago, J.; González-Soltero, R.; Pérez, M.; Montalvo-Lominchar, M.G.; Maté-Muñoz, J.L.; Domínguez, R.; Moreno, D.; Larrosa, M. Differences in gut microbiota profile between women with active lifestyle and sedentary women. PLoS ONE 2017, 12, e0171352. [Google Scholar] [CrossRef] [Green Version]
  60. Scheiman, J.; Luber, J.M.; Chavkin, T.A.; MacDonald, T.; Tung, A.; Pham, L.D.; Wibowo, M.C.; Wurth, R.C.; Punthambaker, S.; Tierney, B.T.; et al. Meta-omics analysis of elite athletes identifies a performance-enhancing microbe that functions via lactate metabolism. Nat. Med. 2019, 25, 1104–1109. [Google Scholar] [CrossRef]
  61. Petersen, L.M.; Bautista, E.J.; Nguyen, H.; Hanson, B.M.; Chen, L.; Lek, S.H.; Sodergren, E.; Weinstock, G.M. Community characteristics of the gut microbiomes of competitive cyclists. Microbiome 2017, 5, 98. [Google Scholar] [CrossRef] [PubMed]
  62. Christensen, L.; Vuholm, S.; Roager, H.M.; Nielsen, D.S.; Krych, L.; Kristensen, M.; Astrup, A.; Hjorth, M.F. Prevotella Abundance Predicts Weight Loss Success in Healthy, Overweight Adults Consuming a Whole-Grain Diet Ad Libitum: A Post Hoc Analysis of a 6-Wk Randomized Controlled Trial. J. Nutr. 2019. [Google Scholar] [CrossRef] [PubMed]
  63. Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014, 63, 1913–1920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Derrien, M.; Belzer, C.; de Vos, W.M. Akkermansia muciniphila and its role in regulating host functions. Microb. Pathog. 2017, 106, 171–181. [Google Scholar] [CrossRef] [PubMed]
  65. Canani, R.B.; Costanzo, M.D.; Leone, L.; Pedata, M.; Meli, R.; Calignano, A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J. Gastroenterol. 2011, 17, 1519–1528. [Google Scholar] [CrossRef] [PubMed]
  66. Machiels, K.; Joossens, M.; Sabino, J.; De Preter, V.; Arijs, I.; Eeckhaut, V.; Ballet, V.; Claes, K.; Van Immerseel, F.; Verbeke, K.; et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 2014, 63, 1275–1283. [Google Scholar] [CrossRef] [PubMed]
  67. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humarán, L.G.; Gratadoux, J.J.; Blugeon, S.; Bridonneau, C.; Furet, J.P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [Green Version]
  68. Estaki, M.; Pither, J.; Baumeister, P.; Little, J.P.; Gill, S.K.; Ghosh, S.; Ahmadi-Vand, Z.; Marsden, K.R.; Gibson, D.L. Cardiorespiratory fitness as a predictor of intestinal microbial diversity and distinct metagenomic functions. Microbiome 2016, 4, 42. [Google Scholar] [CrossRef] [Green Version]
  69. Braune, A.; Blaut, M. Bacterial species involved in the conversion of dietary flavonoids in the human gut. Gut Microbes 2016, 7, 216–234. [Google Scholar] [CrossRef] [Green Version]
  70. Zhao, X.; Zhang, Z.; Hu, B.; Huang, W.; Yuan, C.; Zou, L. Response of Gut Microbiota to Metabolite Changes Induced by Endurance Exercise. Front. Microbiol. 2018, 9, 765. [Google Scholar] [CrossRef]
  71. van Hall, G. Lactate kinetics in human tissues at rest and during exercise. Acta Physiol. 2010, 199, 499–508. [Google Scholar] [CrossRef] [PubMed]
  72. Tung, Y.T.; Chen, Y.J.; Chuang, H.L.; Huang, W.C.; Lo, C.T.; Liao, C.C.; Huang, C.C. Characterization of the serum and liver proteomes in gut-microbiota-lacking mice. Int. J. Med. Sci. 2017, 14, 257–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Trovato, F.M.; Martines, G.F.; Brischetto, D.; Catalano, D.; Musumeci, G.; Trovato, G.M. Fatty liver disease and lifestyle in youngsters: Diet, food intake frequency, exercise, sleep shortage and fashion. Liver Int. 2016, 36, 427–433. [Google Scholar] [CrossRef] [PubMed]
  74. Relman, D.A. The human microbiome: Ecosystem resilience and health. Nutr. Rev. 2012, 70 (Suppl. 1), S2–S9. [Google Scholar] [CrossRef] [PubMed]
  75. Matsumoto, M.; Inoue, R.; Tsukahara, T.; Ushida, K.; Chiji, H.; Matsubara, N.; Hara, H. Voluntary running exercise alters microbiota composition and increases n-butyrate concentration in the rat cecum. BioSci. Biotechnol. Biochem. 2008, 72, 572–576. [Google Scholar] [CrossRef]
  76. Queipo-Ortuno, M.I.; Seoane, L.M.; Murri, M.; Pardo, M.; Gomez-Zumaquero, J.M.; Cardona, F.; Casanueva, F.; Tinahones, F.J. 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 2013, 8, e65465. [Google Scholar] [CrossRef]
  77. Allen, J.M.; Berg Miller, M.E.; Pence, B.D.; Whitlock, K.; Nehra, V.; Gaskins, H.R.; White, B.A.; Fryer, J.D.; Woods, J.A. Voluntary and forced exercise differentially alters the gut microbiome in C57BL/6J mice. J. Appl. Physiol. 2015, 118, 1059–1066. [Google Scholar] [CrossRef] [Green Version]
  78. McFadzean, R. Exercise can Help Modulate Human Gut Microbiota; University of Colorado Boulder: Boulder, CO, USA, 2014. [Google Scholar]
  79. Petriz, B.A.; Castro, A.P.; Almeida, J.A.; Gomes, C.P.; Fernandes, G.R.; Kruger, R.H.; Pereira, R.W.; Franco, O.L. Exercise induction of gut microbiota modifications in obese, non-obese and hypertensive rats. BMC Genom. 2014, 15, 511. [Google Scholar] [CrossRef] [Green Version]
  80. Denou, E.; Marcinko, K.; Surette, M.G.; Steinberg, G.R.; Schertzer, J.D. 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, 310, E982–E993. [Google Scholar] [CrossRef]
  81. Hsu, Y.J.; Chiu, C.C.; Li, Y.P.; Huang, W.C.; Huang, Y.T.; Huang, C.C.; Chuang, H.L. Effect of intestinal microbiota on exercise performance in mice. J. Strength Cond. Res. 2015, 29, 552–558. [Google Scholar] [CrossRef]
  82. Lambert, J.E.; Myslicki, J.P.; Bomhof, M.R.; Belke, D.D.; Shearer, J.; Reimer, R.A. Exercise training modifies gut microbiota in normal and diabetic mice. Appl. Physiol. Nutr. Metab. 2015, 40, 749–752. [Google Scholar] [CrossRef] [PubMed]
  83. Liu, T.W.; Park, Y.M.; Holscher, H.D.; Padilla, J.; Scroggins, R.J.; Welly, R.; Britton, S.L.; Koch, L.G.; Vieira-Potter, V.J.; Swanson, K.S. Physical Activity Differentially Affects the Cecal Microbiota of Ovariectomized Female Rats Selectively Bred for High and Low Aerobic Capacity. PLoS ONE 2015, 10, e0136150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Barton, W.; Penney, N.C.; Cronin, O.; Garcia-Perez, I.; Molloy, M.G.; Holmes, E.; Shanahan, F.; Cotter, P.D.; O’Sullivan, O. The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut 2018, 67, 625–633. [Google Scholar] [CrossRef] [PubMed]
  85. Cook, M.D.; Allen, J.M.; Pence, B.D.; Wallig, M.A.; Gaskins, H.R.; White, B.A.; Woods, J.A. Exercise and gut immune function: Evidence of alterations in colon immune cell homeostasis and microbiome characteristics with exercise training. Immunol. Cell Biol. 2016, 94, 158–163. [Google Scholar] [CrossRef] [PubMed]
  86. Lamoureux, E.V.; Grandy, S.A.; Langille, M.G.I. Moderate Exercise Has Limited but Distinguishable Effects on the Mouse Microbiome. MSystems 2017, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Liu, Z.; Liu, H.Y.; Zhou, H.; Zhan, Q.; Lai, W.; Zeng, Q.; Ren, H.; Xu, D. Moderate-Intensity Exercise Affects Gut Microbiome Composition and Influences Cardiac Function in Myocardial Infarction Mice. Front. Microbiol. 2017, 8, 1687. [Google Scholar] [CrossRef]
  88. Paulsen, J.A.; Ptacek, T.S.; Carter, S.J.; Liu, N.; Kumar, R.; Hyndman, L.; Lefkowitz, E.J.; Morrow, C.D.; Rogers, L.Q. Gut microbiota composition associated with alterations in cardiorespiratory fitness and psychosocial outcomes among breast cancer survivors. Support. Care Cancer 2017, 25, 1563–1570. [Google Scholar] [CrossRef]
  89. Stewart, C.J.; Nelson, A.; Campbell, M.D.; Walker, M.; Stevenson, E.J.; Shaw, J.A.; Cummings, S.P.; West, D.J. Gut microbiota of Type 1 diabetes patients with good glycaemic control and high physical fitness is similar to people without diabetes: An observational study. Diabet. Med. 2017, 34, 127–134. [Google Scholar] [CrossRef]
  90. Cronin, O.; Barton, W.; Skuse, P.; Penney, N.C.; Garcia-Perez, I.; Murphy, E.F.; Woods, T.; Nugent, H.; Fanning, A.; Melgar, S.; 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 2018, 3. [Google Scholar] [CrossRef] [Green Version]
  91. Durk, R.P.; Castillo, E.; Márquez-Magaña, L.; Grosicki, G.J.; Bolter, N.D.; Lee, C.M.; Bagley, J.R. Gut Microbiota Composition Is Related to Cardiorespiratory Fitness in Healthy Young Adults. Int. J. Sport Nutr. Exerc. Metab. 2019, 29, 249–253. [Google Scholar] [CrossRef]
  92. Munukka, E.; Ahtiainen, J.P.; Puigbo, P.; Jalkanen, S.; Pahkala, K.; Keskitalo, A.; Kujala, U.M.; Pietila, S.; Hollmen, M.; Elo, L.; et al. Six-Week Endurance Exercise Alters Gut Metagenome That Is not Reflected in Systemic Metabolism in Over-weight Women. Front. Microbiol. 2018, 9, 2323. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, Y.; Shi, Y.; Wiklund, P.; Tan, X.; Wu, N.; Zhang, X.; Tikkanen, O.; Zhang, C.; Munukka, E.; Cheng, S. The Association between Cardiorespiratory Fitness and Gut Microbiota Composition in Premenopausal Women. Nutrients 2017, 9, 792. [Google Scholar] [CrossRef] [PubMed]
  94. Clark, A.; Mach, N. Exercise-induced stress behavior, gut-microbiota-brain axis and diet: A systematic review for athletes. J. Int. Soc. Sports Nutr. 2016, 13, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Reid, M.B.; Khawli, F.A.; Moody, M.R. Reactive oxygen in skeletal muscle. III. Contractility of unfatigued muscle. J. Appl. Physiol. 1993, 75, 1081–1087. [Google Scholar] [CrossRef]
  96. Powers, S.K.; Jackson, M.J. Exercise-induced oxidative stress: Cellular mechanisms and impact on muscle force production. Physiol. Rev. 2008, 88, 1243–1276. [Google Scholar] [CrossRef] [Green Version]
  97. Barbieri, E.; Sestili, P. Reactive oxygen species in skeletal muscle signaling. J. Signal. Transduct. 2012, 2012, 982794. [Google Scholar] [CrossRef] [Green Version]
  98. He, F.; Li, J.; Liu, Z.; Chuang, C.C.; Yang, W.; Zuo, L. Redox Mechanism of Reactive Oxygen Species in Exercise. Front. Physiol. 2016, 7, 486. [Google Scholar] [CrossRef] [Green Version]
  99. Jones, R.M.; Mercante, J.W.; Neish, A.S. Reactive oxygen production induced by the gut microbiota: Pharmacotherapeutic implications. Curr. Med. Chem. 2012, 19, 1519–1529. [Google Scholar] [CrossRef]
  100. Kumar, A.; Wu, H.; Collier-Hyams, L.S.; Hansen, J.M.; Li, T.; Yamoah, K.; Pan, Z.Q.; Jones, D.P.; Neish, A.S. Commensal bacteria modulate cullin-dependent signaling via generation of reactive oxygen species. EMBO J. 2007, 26, 4457–4466. [Google Scholar] [CrossRef] [Green Version]
  101. Davies, K.J.; Packer, L.; Brooks, G.A. Biochemical adaptation of mitochondria, muscle, and whole-animal respiration to endurance training. Arch. Biochem. Biophys. 1981, 209, 539–554. [Google Scholar] [CrossRef]
  102. Purvis, D.; Gonsalves, S.; Deuster, P.A. Physiological and psychological fatigue in extreme conditions: Overtraining and elite athletes. PM R 2010, 2, 442–450. [Google Scholar] [CrossRef] [PubMed]
  103. Peters, H.P.; De Vries, W.R.; Vanberge-Henegouwen, G.P.; Akkermans, L.M. Potential benefits and hazards of physical activity and exercise on the gastrointestinal tract. Gut 2001, 48, 435–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Gisolfi, C.V. Is the GI System Built For Exercise? News Physiol. Sci. 2000, 15, 114–119. [Google Scholar] [CrossRef] [PubMed]
  105. de Oliveira, E.P.; Burini, R.C. The impact of physical exercise on the gastrointestinal tract. Curr. Opin. Clin. Nutr. Metab. Care 2009, 12, 533–538. [Google Scholar] [CrossRef]
  106. Karl, J.P.; Margolis, L.M.; Madslien, E.H.; Murphy, N.E.; Castellani, J.W.; Gundersen, Y.; Hoke, A.V.; Levangie, M.W.; Kumar, R.; Chakraborty, N.; et al. Changes in intestinal microbiota composition and metabolism coincide with increased intestinal permeability in young adults under prolonged physiological stress. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 312, G559–G571. [Google Scholar] [CrossRef] [Green Version]
  107. Zuhl, M.; Schneider, S.; Lanphere, K.; Conn, C.; Dokladny, K.; Moseley, P. Exercise regulation of intestinal tight junction proteins. Br. J. Sports Med. 2014, 48, 980–986. [Google Scholar] [CrossRef]
  108. Gutekunst, K.; Kruger, K.; August, C.; Diener, M.; Mooren, F.C. Acute exercises induce disorders of the gastrointestinal integrity in a murine model. Eur. J. Appl. Physiol. 2014, 114, 609–617. [Google Scholar] [CrossRef]
  109. Karl, J.P.; Margolis, L.M.; Murphy, N.E.; Carrigan, C.T.; Castellani, J.W.; Madslien, E.H.; Teien, H.K.; Martini, S.; Montain, S.J.; Pasiakos, S.M. Military training elicits marked increases in plasma metabolomic signatures of energy metabolism, lipolysis, fatty acid oxidation, and ketogenesis. Physiol. Rep. 2017, 5. [Google Scholar] [CrossRef]
  110. Egan, B.; Zierath, J.R. Exercise metabolism and the molecular regulation of skeletal muscle adaptation. Cell Metab. 2013, 17, 162–184. [Google Scholar] [CrossRef] [Green Version]
  111. Colbey, C.; Cox, A.J.; Pyne, D.B.; Zhang, P.; Cripps, A.W.; West, N.P. Upper Respiratory Symptoms, Gut Health and Mucosal Immunity in Athletes. Sports Med. 2018, 48, 65–77. [Google Scholar] [CrossRef] [Green Version]
  112. Moreira, A.; Delgado, L.; Moreira, P.; Haahtela, T. Does exercise increase the risk of upper respiratory tract infections? Br. Med. Bull. 2009, 90, 111–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Nieman, D.C.; Nehlsen-Cannarella, S.L.; Fagoaga, O.R.; Henson, D.A.; Shannon, M.; Hjertman, J.M.; Schmitt, R.L.; Bolton, M.R.; Austin, M.D.; Schilling, B.K.; et al. Immune function in female elite rowers and non-athletes. Br. J. Sports Med. 2000, 34, 181–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kang, W.; Kudsk, K.A. Is there evidence that the gut contributes to mucosal immunity in humans? JPEN J. Parenter Enteral Nutr. 2007, 31, 246–258. [Google Scholar] [CrossRef] [PubMed]
  115. Kekkonen, R.A.; Vasankari, T.J.; Vuorimaa, T.; Haahtela, T.; Julkunen, I.; Korpela, R. The effect of probiotics on respiratory infections and gastrointestinal symptoms during training in marathon runners. Int. J. Sport Nutr. Exerc. Metab. 2007, 17, 352–363. [Google Scholar] [CrossRef]
  116. Gleeson, M.; Bishop, N.C.; Oliveira, M.; McCauley, T.; Tauler, P.; Lawrence, C. Effects of a Lactobacillus salivarius probiotic intervention on infection, cold symptom duration and severity, and mucosal immunity in endurance athletes. Int. J. Sport Nutr. Exerc. Metab. 2012, 22, 235–242. [Google Scholar] [CrossRef]
  117. Ulrich-Lai, Y.M.; Herman, J.P. Neural regulation of endocrine and autonomic stress responses. Nat. Rev. Neurosci. 2009, 10, 397–409. [Google Scholar] [CrossRef] [Green Version]
  118. Eisenstein, M. Microbiome: Bacterial broadband. Nature 2016, 533, S104–S106. [Google Scholar] [CrossRef]
  119. Rhee, S.H.; Pothoulakis, C.; Mayer, E.A. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 306–314. [Google Scholar] [CrossRef] [Green Version]
  120. Lyte, M.; Vulchanova, L.; Brown, D.R. Stress at the intestinal surface: Catecholamines and mucosa-bacteria interactions. Cell Tissue Res. 2011, 343, 23–32. [Google Scholar] [CrossRef]
  121. Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.; Hirschfield, G.M.; Hold, G.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The gut microbiota and host health: A new clinical frontier. Gut 2016, 65, 330–339. [Google Scholar] [CrossRef] [Green Version]
  122. Moloney, R.D.; Desbonnet, L.; Clarke, G.; Dinan, T.G.; Cryan, J.F. The microbiome: Stress, health and disease. Mamm. Genome 2014, 25, 49–74. [Google Scholar] [CrossRef] [PubMed]
  123. Eisenhofer, G.; Aneman, A.; Friberg, P.; Hooper, D.; Fåndriks, L.; Lonroth, H.; Hunyady, B.; Mezey, E. Substantial production of dopamine in the human gastrointestinal tract. J. Clin. Endocrinol. Metab. 1997, 82, 3864–3871. [Google Scholar] [CrossRef] [PubMed]
  124. Blomstrand, E.; Perrett, D.; Parry-Billings, M.; Newsholme, E.A. Effect of sustained exercise on plasma amino acid concentrations and on 5-hydroxytryptamine metabolism in six different brain regions in the rat. Acta Physiol. Scand. 1989, 136, 473–481. [Google Scholar] [CrossRef] [PubMed]
  125. Blomstrand, E.; Moller, K.; Secher, N.H.; Nybo, L. Effect of carbohydrate ingestion on brain exchange of amino acids during sustained exercise in human subjects. Acta Physiol. Scand. 2005, 185, 203–209. [Google Scholar] [CrossRef] [PubMed]
  126. Nybo, L.; Dalsgaard, M.K.; Steensberg, A.; Moller, K.; Secher, N.H. Cerebral ammonia uptake and accumulation during prolonged exercise in humans. J. Physiol. 2005, 563, 285–290. [Google Scholar] [CrossRef] [PubMed]
  127. Kavanagh, J.J.; McFarland, A.J.; Taylor, J.L. Enhanced availability of serotonin increases activation of unfatigued muscle but exacerbates central fatigue during prolonged sustained contractions. J. Physiol. 2019, 597, 319–332. [Google Scholar] [CrossRef]
  128. Foley, T.E.; Fleshner, M. Neuroplasticity of dopamine circuits after exercise: Implications for central fatigue. Neuro. Med. 2008, 10, 67–80. [Google Scholar] [CrossRef]
  129. El Aidy, S.; Dinan, T.G.; Cryan, J.F. Gut Microbiota: The Conductor in the Orchestra of Immune-Neuroendocrine Communication. Clin. Ther. 2015, 37, 954–967. [Google Scholar] [CrossRef]
  130. Bailey, S.P.; Davis, J.M.; Ahlborn, E.N. Serotonergic agonists and antagonists affect endurance performance in the rat. Int. J. Sports Med. 1993, 14, 330–333. [Google Scholar] [CrossRef]
  131. Rodriguez, N.R.; Di Marco, N.M.; Langley, S.; Association, A.D.; Canada, D.O.; Medicine, A.C.O.S. American College of Sports Medicine position stand. Nutrition and athletic performance. Med. Sci. Sports Exerc. 2009, 41, 709–731. [Google Scholar] [CrossRef]
  132. De Filippo, C.; Cavalieri, D.; Di Paola, M.; Ramazzotti, M.; Poullet, J.B.; Massart, S.; Collini, S.; Pieraccini, G.; Lionetti, P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. USA 2010, 107, 14691–14696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Hoffman, C.J.; Coleman, E. An eating plan and update on recommended dietary practices for the endurance athlete. J. Am. Diet. Assoc. 1991, 91, 325–330. [Google Scholar] [PubMed]
  135. Close, G.L.; Hamilton, D.L.; Philp, A.; Burke, L.M.; Morton, J.P. New strategies in sport nutrition to increase exercise performance. Free Radic. Biol. Med. 2016, 98, 144–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Nocella, C.; Cammisotto, V.; Pigozzi, F.; Borrione, P.; Fossati, C.; D’Amico, A.; Cangemi, R.; Peruzzi, M.; Gobbi, G.; Ettorre, E.; et al. Impairment between Oxidant and Antioxidant Systems: Short- and Long-term Implications for Athletes’ Health. Nutrients 2019, 11, 1353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Manach, C.; Scalbert, A.; Morand, C.; Remesy, C.; Jimenez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Ismail, T.; Sestili, P.; Akhtar, S. Pomegranate peel and fruit extracts: A review of potential anti-inflammatory and anti-infective effects. J. Ethnopharmacol. 2012, 143, 397–405. [Google Scholar] [CrossRef]
  139. Espin, J.C.; Gonzalez-Sarrias, A.; Tomas-Barberan, F.A. The gut microbiota: A key factor in the therapeutic effects of (poly)phenols. Biochem. Pharmacol. 2017, 139, 82–93. [Google Scholar] [CrossRef]
  140. During, A.; Larondelle, Y. The O-methylation of chrysin markedly improves its intestinal anti-inflammatory properties: Structure-activity relationships of flavones. Biochem. Pharmacol. 2013, 86, 1739–1746. [Google Scholar] [CrossRef]
  141. Etxeberria, U.; Arias, N.; Boque, N.; Macarulla, M.T.; Portillo, M.P.; Martinez, J.A.; Milagro, F.I. Reshaping faecal gut microbiota composition by the intake of trans-resveratrol and quercetin in high-fat sucrose diet-fed rats. J. Nutr. Biochem. 2015, 26, 651–660. [Google Scholar] [CrossRef]
  142. Anhe, F.F.; Roy, D.; Pilon, G.; Dudonne, S.; Matamoros, S.; Varin, T.V.; Garofalo, C.; Moine, Q.; Desjardins, Y.; Levy, E.; et al. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut 2015, 64, 872–883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Li, Z.; Henning, S.M.; Lee, R.P.; Lu, Q.Y.; Summanen, P.H.; Thames, G.; Corbett, K.; Downes, J.; Tseng, C.H.; Finegold, S.M.; et al. Pomegranate extract induces ellagitannin metabolite formation and changes stool microbiota in healthy volunteers. Food Funct. 2015, 6, 2487–2495. [Google Scholar] [CrossRef] [PubMed]
  144. Manach, C.; Williamson, G.; Morand, C.; Scalbert, A.; Remesy, C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am. J. Clin. Nutr. 2005, 81, 230S–242S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. González-Sarrías, A.; Espin, J.C.; Tomás-Barberán, F.A. Non-extractable polyphenols produce gut microbiota metabolites thatpersist in circulation and show anti-inflammatory and free radical-scavenging effects. Trends Food Sci. Technol. 2017, 69, 281–288. [Google Scholar] [CrossRef]
  146. Williams, N.T. Probiotics. Am. J. Health Syst. Pharm. 2010, 67, 449–458. [Google Scholar] [CrossRef] [PubMed]
  147. Mishra, V.; Shah, C.; Mokashe, N.; Chavan, R.; Yadav, H.; Prajapati, J. Probiotics as potential antioxidants: A systematic review. J. Agric. Food Chem. 2015, 63, 3615–3626. [Google Scholar] [CrossRef]
  148. Lin, M.Y.; Yen, C.L. Antioxidative ability of lactic acid bacteria. J. Agric. Food Chem. 1999, 47, 1460–1466. [Google Scholar] [CrossRef]
  149. Persichetti, E.; De Michele, A.; Codini, M.; Traina, G. Antioxidative capacity of Lactobacillus fermentum LF31 evaluated in vitro by oxygen radical absorbance capacity assay. Nutrition 2014, 30, 936–938. [Google Scholar] [CrossRef]
  150. West, N.P.; Pyne, D.B.; Cripps, A.W.; Hopkins, W.G.; Eskesen, D.C.; Jairath, A.; Christophersen, C.T.; Conlon, M.A.; Fricker, P.A. Lactobacillus fermentum (PCC(R)) supplementation and gastrointestinal and respiratory-tract illness symptoms: A randomised control trial in athletes. Nutr. J. 2011, 10, 30. [Google Scholar] [CrossRef] [Green Version]
  151. Bertuccioli, A.; Rocchi, M.; Morganti, I.; Vici, G.; Gervasi, M.; Amatori, S.; Sisti, D. Streptococcus salivarius K12 in pharyngotonsillitis and acute otitis media—A meta-analysis. Nutrafoods 2019, 2, 80–88. [Google Scholar] [CrossRef]
  152. West, N.P.; Pyne, D.B.; Peake, J.M.; Cripps, A.W. Probiotics, immunity and exercise: A review. Exerc. Immunol. Rev. 2009, 15, 107–126. [Google Scholar] [PubMed]
  153. Gleeson, M.; Bishop, N.C.; Oliveira, M.; Tauler, P. Daily probiotic’s (Lactobacillus casei Shirota) reduction of infection incidence in athletes. Int. J. Sport Nutr. Exerc. Metab. 2011, 21, 55–64. [Google Scholar] [CrossRef] [PubMed]
  154. Mack, D.R.; Ahrne, S.; Hyde, L.; Wei, S.; Hollingsworth, M.A. Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro. Gut 2003, 52, 827–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Lamprecht, M.; Bogner, S.; Schippinger, G.; Steinbauer, K.; Fankhauser, F.; Hallstroem, S.; Schuetz, B.; Greilberger, J.F. Probiotic supplementation affects markers of intestinal barrier, oxidation, and inflammation in trained men; a randomized, double-blinded, placebo-controlled trial. J. Int. Soc. Sports Nutr. 2012, 9, 45. [Google Scholar] [CrossRef] [Green Version]
  156. Roberts, J.D.; Suckling, C.A.; Peedle, G.Y.; Murphy, J.A.; Dawkins, T.G.; Roberts, M.G. An Exploratory Investigation of Endotoxin Levels in Novice Long Distance Triathletes, and the Effects of a Multi-Strain Probiotic/Prebiotic, Antioxidant Intervention. Nutrients 2016, 8, 733. [Google Scholar] [CrossRef] [Green Version]
  157. Jacouton, E.; Mach, N.; Cadiou, J.; Lapaque, N.; Clement, K.; Dore, J.; van Hylckama Vlieg, J.E.; Smokvina, T.; Blottiere, H.M. Lactobacillus rhamnosus CNCMI-4317 Modulates Fiaf/Angptl4 in Intestinal Epithelial Cells and Circulating Level in Mice. PLoS ONE 2015, 10, e0138880. [Google Scholar] [CrossRef] [Green Version]
  158. Duncan, S.H.; Louis, P.; Flint, H.J. Lactate-utilizing bacteria, isolated from human feces, that produce butyrate as a major fermentation product. Appl. Environ. Microbiol. 2004, 70, 5810–5817. [Google Scholar] [CrossRef] [Green Version]
  159. Chen, Y.M.; Wei, L.; Chiu, Y.S.; Hsu, Y.J.; Tsai, T.Y.; Wang, M.F.; Huang, C.C. Lactobacillus plantarum TWK10 Supplementation Improves Exercise Performance and Increases Muscle Mass in Mice. Nutrients 2016, 8, 205. [Google Scholar] [CrossRef]
  160. Huang, W.C.; Hsu, Y.J.; Li, H.; Kan, N.W.; Chen, Y.M.; Lin, J.S.; Hsu, T.K.; Tsai, T.Y.; Chiu, Y.S.; Huang, C.C. Effect of Lactobacillus Plantarum TWK10 on Improving Endurance Performance in Humans. Chin. J. Physiol. 2018, 61, 163–170. [Google Scholar] [CrossRef]
  161. Li, S.; Zhao, Y.; Zhang, L.; Zhang, X.; Huang, L.; Li, D.; Niu, C.; Yang, Z.; Wang, Q. Antioxidant activity of Lactobacillus plantarum strains isolated from traditional Chinese fermented foods. Food Chem. 2012, 135, 1914–1919. [Google Scholar] [CrossRef]
  162. Kaushik, J.K.; Kumar, A.; Duary, R.K.; Mohanty, A.K.; Grover, S.; Batish, V.K. Functional and probiotic attributes of an indigenous isolate of Lactobacillus plantarum. PLoS ONE 2009, 4, e8099. [Google Scholar] [CrossRef] [Green Version]
  163. Soares, A.D.N.; Wanner, S.P.; Morais, E.S.S.; Hudson, A.S.R.; Martins, F.S.; Cardoso, V.N. Supplementation with Saccharomyces boulardii Increases the Maximal Oxygen Consumption and Maximal Aerobic Speed Attained by Rats Subjected to an Incremental-Speed Exercise. Nutrients 2019, 11, 2352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. St-Onge, M.P.; Farnworth, E.R.; Savard, T.; Chabot, D.; Mafu, A.; Jones, P.J. Kefir consumption does not alter plasma lipid levels or cholesterol fractional synthesis rates relative to milk in hyperlipidemic men: A randomized controlled trial [ISRCTN10820810]. BMC Complement. Altern. Med. 2002, 2, 1. [Google Scholar] [CrossRef] [PubMed]
  165. Anderson, L. Handbook of Fermented Functional Foods; CRC Press: Boca Raton, FL, USA, 2003; Volume 5, pp. 101–102. [Google Scholar]
  166. Huang, C.C.; Hsu, M.C.; Huang, W.C.; Yang, H.R.; Hou, C.C. Triterpenoid-Rich Extract from Antrodia camphorata Improves Physical Fatigue and Exercise Performance in Mice. Evid. Based Complement. Altern. Med. 2012, 2012, 364741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. O’Brien, K.V.; Stewart, L.K.; Forney, L.A.; Aryana, K.J.; Prinyawiwatkul, W.; Boeneke, C.A. The effects of postexercise consumption of a kefir beverage on performance and recovery during intensive endurance training. J. Dairy Sci. 2015, 98, 7446–7449. [Google Scholar] [CrossRef]
  168. Lollo, P.C.B.; Morato, P.N.; de Moura, C.S.; de Oliveira, M.M.; Cruz, A.G.; José de Assis, F.F.; Amaya-Farfan, J.; Cristianini, M. Ultra-high temperature plus dynamic highpressure processing: An effective combinationfor potential probiotic fermented milkprocessing which attenuate exercise-inducedimmune suppression in Wistar rats. J. Funct. Foods 2015, 14, 541–548. [Google Scholar] [CrossRef]
  169. Hsu, Y.J.; Huang, W.C.; Lin, J.S.; Chen, Y.M.; Ho, S.T.; Huang, C.C.; Tung, Y.T. Kefir Supplementation Modifies Gut Microbiota Composition, Reduces Physical Fatigue, and Improves Exercise Performance in Mice. Nutrients 2018, 10, 862. [Google Scholar] [CrossRef] [Green Version]
  170. Grant, M.C.; Baker, J.S. An overview of the effect of probiotics and exercise on mood and associated health conditions. Crit. Rev. Food Sci. Nutr. 2017, 57, 3887–3893. [Google Scholar] [CrossRef] [Green Version]
  171. Bar, K.J.; Markser, V.Z. Sport specificity of mental disorders: The issue of sport psychiatry. Eur. Arch. Psychiatry Clin. Neurosci. 2013, 263 (Suppl. 2), S205–S210. [Google Scholar] [CrossRef]
  172. Bravo, J.A.; Julio-Pieper, M.; Forsythe, P.; Kunze, W.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Communication between gastrointestinal bacteria and the nervous system. Curr. Opin. Pharmacol. 2012, 12, 667–672. [Google Scholar] [CrossRef]
  173. Benton, D.; Williams, C.; Brown, A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 2007, 61, 355–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Gruenwald, J.; Graubaum, H.J.; Harde, A. Effect of a probiotic multivitamin compound on stress and exhaustion. Adv. Ther. 2002, 19, 141–150. [Google Scholar] [CrossRef] [PubMed]
  175. Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Rao, A.V.; Bested, A.C.; Beaulne, T.M.; Katzman, M.A.; Iorio, C.; Berardi, J.M.; Logan, A.C. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog. 2009, 1, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Desbonnet, L.; Garrett, L.; Clarke, G.; Bienenstock, J.; Dinan, T.G. The probiotic Bifidobacteria infantis: An assessment of potential antidepressant properties in the rat. J. Psychiatr. Res. 2008, 43, 164–174. [Google Scholar] [CrossRef] [PubMed]
  178. Lyte, M.; Li, W.; Opitz, N.; Gaykema, R.P.; Goehler, L.E. Induction of anxiety-like behavior in mice during the initial stages of infection with the agent of murine colonic hyperplasia Citrobacter rodentium. Physiol. Behav. 2006, 89, 350–357. [Google Scholar] [CrossRef] [PubMed]
  179. Goehler, L.E.; Park, S.M.; Opitz, N.; Lyte, M.; Gaykema, R.P. Campylobacter jejuni infection increases anxiety-like behavior in the holeboard: Possible anatomical substrates for viscerosensory modulation of exploratory behavior. Brain Behav. Immun. 2008, 22, 354–366. [Google Scholar] [CrossRef] [Green Version]
  180. Schultchen, D.; Reichenberger, J.; Mittl, T.; Weh, T.R.M.; Smyth, J.M.; Blechert, J.; Pollatos, O. Bidirectional relationship of stress and affect with physical activity and healthy eating. Br. J. Health Psychol. 2019, 24, 315–333. [Google Scholar] [CrossRef] [Green Version]
  181. Kanning, M.K.; Ebner-Priemer, U.W.; Schlicht, W.M. How to Investigate Within-Subject Associations between Physical Activity and Momentary Affective States in Everyday Life: A Position Statement Based on a Literature Overview. Front. Psychol. 2013, 4, 187. [Google Scholar] [CrossRef] [Green Version]
  182. Gostner, J.M.; Becker, K.; Sperner-Unterweger, B.; Überall, F.; Fuchs, D.; Strasser, B. Role of Tryptophan Metabolism in Mood, Behavior, and Cognition. In Targeting the Broadly Pathogenic Kynurenine Pathway; Springer: Cham, Switzerland, 2015; pp. 75–89. [Google Scholar]
  183. Thomas, D.T.; Erdman, K.A.; Burke, L.M. Position of the Academy of Nutrition and Dietetics, Dietitians of Canada, and the American College of Sports Medicine: Nutrition and Athletic Performance. J. Acad. Nutr. Diet. 2016, 116, 501–528. [Google Scholar] [CrossRef]
  184. Mills, S.; Stanton, C.; Lane, J.A.; Smith, G.J.; Ross, R.P. Precision Nutrition and the Microbiome, Part I: Current State of the Science. Nutrients 2019, 11, 923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Beaumont, M.; Portune, K.J.; Steuer, N.; Lan, A.; Cerrudo, V.; Audebert, M.; Dumont, F.; Mancano, G.; Khodorova, N.; Andriamihaja, M.; et al. Quantity and source of dietary protein influence metabolite production by gut microbiota and rectal mucosa gene expression: A randomized, parallel, double-blind trial in overweight humans. Am. J. Clin. Nutr. 2017, 106, 1005–1019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Moreno-Perez, D.; Bressa, C.; Bailen, M.; Hamed-Bousdar, S.; Naclerio, F.; Carmona, M.; Perez, M.; Gonzalez-Soltero, R.; Montalvo-Lominchar, M.G.; Carabana, C.; et al. Effect of a Protein Supplement on the Gut Microbiota of Endurance Athletes: A Randomized, Controlled, Double-Blind Pilot Study. Nutrients 2018, 10, 337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Karlund, A.; Gomez-Gallego, C.; Turpeinen, A.M.; Palo-Oja, O.M.; El-Nezami, H.; Kolehmainen, M. Protein Supplements and Their Relation with Nutrition, Microbiota Composition and Health: Is More Protein Always Better for Sportspeople? Nutrients 2019, 11, 829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Lynch, C.J.; Gern, B.; Lloyd, C.; Hutson, S.M.; Eicher, R.; Vary, T.C. Leucine in food mediates some of the postprandial rise in plasma leptin concentrations. Am. J. Physiol. Endocrinol. Metab. 2006, 291, E621–E630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Zhang, S.; Qiao, S.; Ren, M.; Zeng, X.; Ma, X.; Wu, Z.; Thacker, P.; Wu, G. Supplementation with branched-chain amino acids to a low-protein diet regulates intestinal expression of amino acid and peptide transporters in weanling pigs. Amino Acids 2013, 45, 1191–1205. [Google Scholar] [CrossRef] [PubMed]
  190. Zhang, Y.; Guo, K.; LeBlanc, R.E.; Loh, D.; Schwartz, G.J.; Yu, Y.H. Increasing dietary leucine intake reduces diet-induced obesity and improves glucose and cholesterol metabolism in mice via multimechanisms. Diabetes 2007, 56, 1647–1654. [Google Scholar] [CrossRef] [Green Version]
  191. D’Antona, G.; Ragni, M.; Cardile, A.; Tedesco, L.; Dossena, M.; Bruttini, F.; Caliaro, F.; Corsetti, G.; Bottinelli, R.; Carruba, M.O.; et al. Branched-chain amino acid supplementation promotes survival and supports cardiac and skeletal muscle mitochondrial biogenesis in middle-aged mice. Cell Metab. 2010, 12, 362–372. [Google Scholar] [CrossRef] [Green Version]
  192. Ren, M.; Zhang, S.H.; Zeng, X.F.; Liu, H.; Qiao, S.Y. Branched-chain Amino Acids are Beneficial to Maintain Growth Performance and Intestinal Immune-related Function in Weaned Piglets Fed Protein Restricted Diet. Asian-Australas. J. Anim. Sci. 2015, 28, 1742–1750. [Google Scholar] [CrossRef] [Green Version]
  193. Zhou, H.; Yu, B.; Gao, J.; Htoo, J.K.; Chen, D. Regulation of intestinal health by branched-chain amino acids. Anim. Sci. J. 2018, 89, 3–11. [Google Scholar] [CrossRef]
  194. Yang, Z.; Huang, S.; Zou, D.; Dong, D.; He, X.; Liu, N.; Liu, W.; Huang, L. Metabolic shifts and structural changes in the gut microbiota upon branched-chain amino acid supplementation in middle-aged mice. Amino Acids 2016, 48, 2731–2745. [Google Scholar] [CrossRef] [PubMed]
  195. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Calder, P.C. Glutamine and the immune system. Clin. Nutr. 1994, 13, 2–8. [Google Scholar] [CrossRef]
  197. Kim, M.H.; Kim, H. The Roles of Glutamine in the Intestine and Its Implication in Intestinal Diseases. Int. J. Mol. Sci. 2017, 18, 1051. [Google Scholar] [CrossRef] [Green Version]
  198. L’Huillier, C.; Jarbeau, M.; Achamrah, N.; Belmonte, L.; Amamou, A.; Nobis, S.; Goichon, A.; Salameh, E.; Bahlouli, W.; do Rego, J.L.; et al. Glutamine, but not Branched-Chain Amino Acids, Restores Intestinal Barrier Function during Activity-Based Anorexia. Nutrients 2019, 11, 1348. [Google Scholar] [CrossRef] [Green Version]
  199. Coqueiro, A.Y.; Rogero, M.M.; Tirapegui, J. Glutamine as an Anti-Fatigue Amino Acid in Sports Nutrition. Nutrients 2019, 11, 863. [Google Scholar] [CrossRef] [Green Version]
  200. de Souza, A.Z.; Zambom, A.Z.; Abboud, K.Y.; Reis, S.K.; Tannihao, F.; Guadagnini, D.; Saad, M.J.; Prada, P.O. Oral supplementation with L-glutamine alters gut microbiota of obese and overweight adults: A pilot study. Nutrition 2015, 31, 884–889. [Google Scholar] [CrossRef]
  201. Swidsinski, A.; Weber, J.; Loening-Baucke, V.; Hale, L.P.; Lochs, H. Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J. Clin. Microbiol. 2005, 43, 3380–3389. [Google Scholar] [CrossRef] [Green Version]
  202. Keighley, M.R.; Arabi, Y.; Dimock, F.; Burdon, D.W.; Allan, R.N.; Alexander-Williams, J. Influence of inflammatory bowel disease on intestinal microflora. Gut 1978, 19, 1099–1104. [Google Scholar] [CrossRef] [Green Version]
  203. Abboud, K.Y.; Reis, S.K.; Martelli, M.E.; Zordao, O.P.; Tannihao, F.; de Souza, A.Z.Z.; Assalin, H.B.; Guadagnini, D.; Rocha, G.Z.; Saad, M.J.A.; et al. Oral Glutamine Supplementation Reduces Obesity, Pro-Inflammatory Markers, and Improves Insulin Sensitivity in DIO Wistar Rats and Reduces Waist Circumference in Overweight and Obese Humans. Nutrients 2019, 11, 536. [Google Scholar] [CrossRef] [Green Version]
  204. Perna, S.; Alalwan, T.A.; Alaali, Z.; Alnashaba, T.; Gasparri, C.; Infantino, V.; Hammad, L.; Riva, A.; Petrangolini, G.; Allegrini, P.; et al. The Role of Glutamine in the Complex Interaction between Gut Microbiota and Health: A Narrative Review. Int. J. Mol. Sci. 2019, 20, 5232. [Google Scholar] [CrossRef] [Green Version]
  205. Carr, A.J.; Hopkins, W.G.; Gore, C.J. Effects of acute alkalosis and acidosis on performance: A meta-analysis. Sports Med. 2011, 41, 801–814. [Google Scholar] [CrossRef] [PubMed]
  206. Burke, L.M. Practical considerations for bicarbonate loading and sports performance. Nestle Nutr. Inst. Workshop Ser. 2013, 75, 15–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Murakami, S.; Goto, Y.; Ito, K.; Hayasaka, S.; Kurihara, S.; Soga, T.; Tomita, M.; Fukuda, S. The Consumption of Bicarbonate-Rich Mineral Water Improves Glycemic Control. Evid. Based Complement. Altern. Med. 2015, 2015, 824395. [Google Scholar] [CrossRef]
  208. Goodrich, J.K.; Waters, J.L.; Poole, A.C.; Sutter, J.L.; Koren, O.; Blekhman, R.; Beaumont, M.; Van Treuren, W.; Knight, R.; Bell, J.T.; et al. Human genetics shape the gut microbiome. Cell 2014, 159, 789–799. [Google Scholar] [CrossRef] [Green Version]
  209. Veldurthy, V.; Wei, R.; Oz, L.; Dhawan, P.; Jeon, Y.H.; Christakos, S. Vitamin D, calcium homeostasis and aging. Bone Res. 2016, 4, 16041. [Google Scholar] [CrossRef] [Green Version]
  210. Agostini, D.; Zeppa Donati, S.; Lucertini, F.; Annibalini, G.; Gervasi, M.; Ferri Marini, C.; Piccoli, G.; Stocchi, V.; Barbieri, E.; Sestili, P. Muscle and Bone Health in Postmenopausal Women: Role of Protein and Vitamin D Supplementation Combined with Exercise Training. Nutrients 2018, 10, 1103. [Google Scholar] [CrossRef] [Green Version]
  211. Close, G.L.; Russell, J.; Cobley, J.N.; Owens, D.J.; Wilson, G.; Gregson, W.; Fraser, W.D.; Morton, J.P. Assessment of vitamin D concentration in non-supplemented professional athletes and healthy adults during the winter months in the UK: Implications for skeletal muscle function. J. Sports Sci. 2013, 31, 344–353. [Google Scholar] [CrossRef]
  212. Gominak, S.C. Vitamin D deficiency changes the intestinal microbiome reducing B vitamin production in the gut. The resulting lack of pantothenic acid adversely affects the immune system, producing a “pro-inflammatory” state associated with atherosclerosis and autoimmunity. Med. Hypotheses 2016, 94, 103–107. [Google Scholar] [CrossRef]
  213. Bashir, M.; Prietl, B.; Tauschmann, M.; Mautner, S.I.; Kump, P.K.; Treiber, G.; Wurm, P.; Gorkiewicz, G.; Hogenauer, C.; Pieber, T.R. Effects of high doses of vitamin D3 on mucosa-associated gut microbiome vary between regions of the human gastrointestinal tract. Eur. J. Nutr. 2016, 55, 1479–1489. [Google Scholar] [CrossRef] [Green Version]
  214. Waterhouse, M.; Hope, B.; Krause, L.; Morrison, M.; Protani, M.M.; Zakrzewski, M.; Neale, R.E. Vitamin D and the gut microbiome: A systematic review of in vivo studies. Eur. J. Nutr. 2019, 58, 2895–2910. [Google Scholar] [CrossRef] [PubMed]
  215. Back, M.; Hansson, G.K. Omega-3 fatty acids, cardiovascular risk, and the resolution of inflammation. FASEB J. 2019, 33, 1536–1539. [Google Scholar] [CrossRef] [PubMed]
  216. Kim, M.; Qie, Y.; Park, J.; Kim, C.H. Gut Microbial Metabolites Fuel Host Antibody Responses. Cell Host Microbe 2016, 20, 202–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Maughan, R.J.; Burke, L.M.; Dvorak, J.; Larson-Meyer, D.E.; Peeling, P.; Phillips, S.M.; Rawson, E.S.; Walsh, N.P.; Garthe, I.; Geyer, H.; et al. IOC Consensus Statement: Dietary Supplements and the High-Performance Athlete. Int. J. Sport Nutr. Exerc. Metab. 2018, 28, 104–125. [Google Scholar] [CrossRef] [Green Version]
  218. Whiting, C.V.; Bland, P.W.; Tarlton, J.F. Dietary n-3 polyunsaturated fatty acids reduce disease and colonic proinflammatory cytokines in a mouse model of colitis. Inflamm. Bowel Dis. 2005, 11, 340–349. [Google Scholar] [CrossRef]
  219. Andersen, A.D.; Molbak, L.; Michaelsen, K.F.; Lauritzen, L. Molecular fingerprints of the human fecal microbiota from 9 to 18 months old and the effect of fish oil supplementation. J. Pediatr. Gastroenterol. Nutr. 2011, 53, 303–309. [Google Scholar] [CrossRef]
  220. Balfego, M.; Canivell, S.; Hanzu, F.A.; Sala-Vila, A.; Martinez-Medina, M.; Murillo, S.; Mur, T.; Ruano, E.G.; Linares, F.; Porras, N.; et al. Effects of sardine-enriched diet on metabolic control, inflammation and gut microbiota in drug-naive patients with type 2 diabetes: A pilot randomized trial. Lipids Health Dis. 2016, 15, 78. [Google Scholar] [CrossRef] [Green Version]
  221. Watson, H.; Mitra, S.; Croden, F.C.; Taylor, M.; Wood, H.M.; Perry, S.L.; Spencer, J.A.; Quirke, P.; Toogood, G.J.; Lawton, C.L.; et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut 2018, 67, 1974–1983. [Google Scholar] [CrossRef]
  222. Robertson, R.C.; Seira Oriach, C.; Murphy, K.; Moloney, G.M.; Cryan, J.F.; Dinan, T.G.; Paul Ross, R.; Stanton, C. Omega-3 polyunsaturated fatty acids critically regulate behaviour and gut microbiota development in adolescence and adulthood. Brain Behav. Immun. 2017, 59, 21–37. [Google Scholar] [CrossRef]
  223. Provensi, G.; Schmidt, S.D.; Boehme, M.; Bastiaanssen, T.F.S.; Rani, B.; Costa, A.; Busca, K.; Fouhy, F.; Strain, C.; Stanton, C.; et al. Preventing adolescent stress-induced cognitive and microbiome changes by diet. Proc. Natl. Acad. Sci. USA 2019, 116, 9644–9651. [Google Scholar] [CrossRef]
  224. Costantini, L.; Molinari, R.; Farinon, B.; Merendino, N. Impact of Omega-3 Fatty Acids on the Gut Microbiota. Int. J. Mol. Sci. 2017, 18, 2645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. O’Brien, W.J.; Rowlands, D.S. Fructose-maltodextrin ratio in a carbohydrate-electrolyte solution differentially affects exogenous carbohydrate oxidation rate, gut comfort, and performance. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G181–G189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Guillochon, M.; Rowlands, D.S. Solid, Gel, and Liquid Carbohydrate Format Effects on Gut Comfort and Performance. Int. J. Sport Nutr. Exerc. Metab. 2017, 27, 247–254. [Google Scholar] [CrossRef] [PubMed]
  227. Brass, E.P. Supplemental carnitine and exercise. Am. J. Clin. Nutr. 2000, 72, 618S–623S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Fielding, R.; Riede, L.; Lugo, J.P.; Bellamine, A. l-Carnitine Supplementation in Recovery after Exercise. Nutrients 2018, 10, 349. [Google Scholar] [CrossRef] [Green Version]
  229. Ghonimy, A.; Zhang, D.M.; Farouk, M.H.; Wang, Q. The Impact of Carnitine on Dietary Fiber and Gut Bacteria Metabolism and Their Mutual Interaction in Monogastrics. Int. J. Mol. Sci. 2018, 19, 1008. [Google Scholar] [CrossRef] [Green Version]
  230. Ussher, J.R.; Lopaschuk, G.D.; Arduini, A. Gut microbiota metabolism of L-carnitine and cardiovascular risk. Atherosclerosis 2013, 231, 456–461. [Google Scholar] [CrossRef]
  231. Johri, A.M.; Heyland, D.K.; Hetu, M.F.; Crawford, B.; Spence, J.D. Carnitine therapy for the treatment of metabolic syndrome and cardiovascular disease: Evidence and controversies. Nutr. Metab. Cardiovasc. Dis. 2014, 24, 808–814. [Google Scholar] [CrossRef]
  232. Koeth, R.A.; Lam-Galvez, B.R.; Kirsop, J.; Wang, Z.; Levison, B.S.; Gu, X.; Copeland, M.F.; Bartlett, D.; Cody, D.B.; Dai, H.J.; et al. l-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J. Clin. Investig. 2019, 129, 373–387. [Google Scholar] [CrossRef]
  233. Mielgo-Ayuso, J.; Marques-Jimenez, D.; Refoyo, I.; Del Coso, J.; Leon-Guereno, P.; Calleja-Gonzalez, J. Effect of Caffeine Supplementation on Sports Performance Based on Differences Between Sexes: A Systematic Review. Nutrients 2019, 11, 2313. [Google Scholar] [CrossRef] [Green Version]
  234. Jaquet, M.; Rochat, I.; Moulin, J.; Cavin, C.; Bibiloni, R. Impact of coffee consumption on the gut microbiota: A human volunteer study. Int. J. Food Microbiol. 2009, 130, 117–121. [Google Scholar] [CrossRef] [PubMed]
  235. Janssens, P.L.; Penders, J.; Hursel, R.; Budding, A.E.; Savelkoul, P.H.; Westerterp-Plantenga, M.S. Long-Term Green Tea Supplementation Does Not Change the Human Gut Microbiota. PLoS ONE 2016, 11, e0153134. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Beneficial effects of exercise, supplements, and microbiota interactions on athletes’ health and performance.
Figure 1. Beneficial effects of exercise, supplements, and microbiota interactions on athletes’ health and performance.
Nutrients 12 00017 g001
Table 1. Exercise and microbiota.
Table 1. Exercise and microbiota.
ReferenceModelsSample SizeType of ExerciseDuration of the InterventionEffects on Microbiota and Metabolic-Functional Responses
[75]Wistar rats14 (n = 7/group)Voluntary wheel running vs. control5 weeks↑ Butyrate-producing bacteria; ↑ butyrate
[76]Sprague-Dawley rats ABA group; control ABA group40 (n = 10/group)Voluntary wheel running.
(i): rats submitted to the same food restriction schedule as ABA with no wheel access exercise, (iii) exercise group: rats feed ad libitum with free access to the activity wheel, and (iv) ad libitum group: rats feed ad libitum but without access to the activity wheel
6 daysBacteroidetes; ↓ Firmicutes; ↑ diversity; ↓ Bacteroides; ↑ Lactobacillus; ↑ organic acid lactate, butyrate
[63]Humans86
(n = 20–23/group)
Male rugby players vs. healthy male controls1 sampling point↑ Diversity in athletes vs controls; ↓ Firmicutes,Ruminococcaceae,Succinivibrionaceae, and ↓ Succinivibrio groups; ↑ Akkermansiaceae and ↑ Akkermansia in low BMI athletes and controls; ↑ protein intake; ↑ CPK in athletes
[12]Wild-type mice48 (n = 12/group)Voluntary wheel running.
(i) LF/Sed, (ii) LF/Ex, (iii) HF/Sed, and (iv) HF/Ex
12 weeksFirmicutes:Bacteroidetes ratio; ↑ Bacteroidetes phylum; ↓ Firmicutes in a manner that was proportional to the distance run in mice fed with HFD
Actinobacteria; exercise prevented DIO
[11]Wild-type mice, DIO mice40 (n = 10/group)Motorized wheel.16 weeksBacteroidetes; ↑ Firmicutes;
↑ Cognitive abilities despite HFD
[78]Humans1493Voluntary exercise (i) never, (ii) rarely, (iii) occasionally, (iv) regularly, and (v) daily1 sampling pointFaecalibacterium prausnitzii; ↑ α-proteobacteria diversity
[79]Obese (Zucker), hypertensive (SHR) and Wistar rats9 (n =  3/group)Forced treadmill running (i) obese fa/fa (homozygous); obese (Obese rats), (ii) hypertensive, and (iii) Wistar-Kyoto rats with high blood pressure4 weeksFirmicutes;Proteobacteria;Lactobacillus;Allobaculum (Hypertensive rats); ↑ Pseudomonas and Lactobacillus (Obese rats); ↑ Clostridiaceae and Bacteroidaceae families and Oscillospira and Ruminococcus genera ↔ blood lactate accumulation
[77]C57BL/6J mice30
(n = 9–10/group)
Voluntary wheel running vs. forced treadmill running or control6 weeks↓ Bacterial richness; ↔ Bacteroidetes,Firmicutes in both groups. ↓ Turicibacter in Voluntary group
[80]Wild-type mice24 (n = 8/group)HIIT6 weeksFirmicutes:Bacteroidetes ratio
[81]GF-, SPF-, BF- gnotobiotic mice24 (n = 8/group)SwimmingTo-exhaustion test↓ Glutathione, ↓ catalase, ↓ SCFAs, in GF and BF vs SPF
[82]Mice38
(n = 9–10/group)
Low-intensity treadmill running type 2 diabetic db/db vs db/+ (heterozygote); or control6 weeks↓ Enterobacteriaceae and ↑ Bifidobacterium spp Bifidobacterium spp. in exercised non-diabetic mice
Bacteroides/Prevotella spp. and Methanobrevibacter spp; ↑ Lactobacillus spp. and Clostridium leptum in both trained and untrained groups
[83]Ovariectomized rats15 (n = 7–8/group)Voluntary wheel running vs. control11 weeksBacteroidetes; ↔ Firmicutes:Bacteroidetes ratio; ↓ Firmicutes in low-capacity running rats; ↑ Firmicutes in high-capacity running rats
[57]F344 rats40 (n = 20/group)Voluntary wheel running; juvenile vs. adults6 weeksBacteroidetes;Firmicutes; ↑ bacterial genera in juvenile rats vs adults
[48]Wild-type, DIO mice36 (n = 18/group)Voluntary wheel running vs. control12 weeksFaecalibacterium prausnitzii in trained mice; ↓ Cox-2 in DIO trained mice
[85]Wild-type mice14 (n = 4/5/group)Voluntary wheel running vs. forced treadmill running or sedentary6 weeks↑ Diarrhoea; ↑ IL-6; ↑ IL-1β; IL-17 colon gene expression; ↑ mortality in treadmill group; alleviated colitis symptoms; ↓ inflammatory gene in wheel group
[68]Humans39 (n = 14-12-13/group)Observing levels of cardiorespiratory fitness1 sampling point↑ Diversity; ↑ butyrate-producing taxa in subjects with higher fitness
[84]Humans86
(n = 40–46/group)
Professional rugby players vs. control1 sampling point↑ SCFAs: acetate, propionate and butyrate, ↑ muscle turnover (fitness) in athletes vs. control group
[59]Humans, healthy women40
(n = 19–21/group)
Active women performing World Health Organization-recommended low dose of exercise vs. sedentary1 sampling pointFaecalibacterium prausnitzii; ↑ Akkermansia muciniphila; Gut microbiota composition ∝ body fat/muscular mass
[86]C57BL/6J mice42 (n = 10-11-21/group)Voluntary wheel running vs. forced treadmill running or control8 weeks↔ Abundance in gut microbes, ↑ Rikenellaceae and Lachnospiraceae; ↔ host inflammatory response in both trained and control
[87]C57BL/6J mice16 (n = 7–9/group)Forced treadmill running vs. control4 weeksButyricimonas and Akkermansia; improved cardiac function
[88]Humans, BCS12Observing levels of cardiorespiratory fitness, anxiety, fatigue3 monthsGut microbiota composition ∝ changes in cardiorespiratory fitness level and anxiety in BCS
[61]Humans33Cyclists who reported either 6–10, 11–15, 16–20, or 20+ hours of exercise per week1 sampling pointBacteroides;Prevotella; ↑ Eubacterium; ↑ Ruminococcus, and ↑ Akkermansia; ↑ Methanobrevibacter smithii archaeon had upregulation of genes involved in the production of methane
[89]Humans, T1D, and healthy controls20 (n = 10/group)Male T1D vs healthy male controls, observing levels of physical fitness, glycemic control1 sampling pointFaecalibacterium sp., Roseburia sp. and Bacteroides sp. were typically the most abundant members of the community in both patients with T1D and controls. Gut microbiota comparable between T1D-subjects in good glycaemic control + high physical fitness vs healthy control
[93]Humans, premenopausal women71
(n = 23–24/group)
Observing levels of cardiorespiratory fitness1 sampling pointBacteroides; ↓ Eubacterium rectale-Clostridium coccoides. Gut microbiota composition ∝ cardiorespiratory fitness level. The association between VO2max and EreC, however, appears to be mediated by body fatness
[70]Humans20 (16 males and 4 females)Amateur runners2 sampling points (one week before and one week after the half-marathon race)Running did not affect the α-diversity, ↑ Coriobacteriaceae and Succinivibrionaceae Coprococcus, Actinobacillus, and Ruminococcus bicirculans;Ezakiella and Romboutsia
[9]Mice29
(n = 9–10/group)
Voluntary wheel running vs. forced treadmill running or control6 weeksVoluntary wheel running vs Forced treadmill running altered many individual bacterial taxa.
Turicibacter spp. in VWR group
[90]Humans, Overweight or obese90 (n = 30/group)Overweight or obese adults randomized to exercise-only, exercise + whey protein, or whey protein only groups8 weeksNo significant changes in microbial species composition; ↑ bacterial diversity in to exercise-only, exercise + whey protein. Modest alterations of microbial metabolic potential
[91]Humans37 (n = 20 males, n = 17 females)Observing levels of cardiorespiratory fitness1 sampling pointFirmicutes:Bacteroidetes ∝ VO2max
[92]Humans17Sedentary, overweight women cycling exercise (low–moderate intensity)6 weeksAkkermansia; ↓ Proteobacteria;
↓ Fructose and amino acid metabolism-related genes
[60]Humans38
(n = 26–12/group)
Marathon athletes vs. control2 sampling points (one week before and one week after the race)Veillonella atypica converts lactate to propionate;
↑ Methylmalonyl-CoA pathway is overexpressed
[60]Male C57BL/5 64 (n = 32/group)Treated with V. atypica or L. bulgaricus3 days. 2 sampling points, (before exercise and after exercise).Veillonella inoculation improved treadmill performance; performance is improved in mice administered propionate via intracolonic infusion
Abbreviations: ↑ significant increase; ↓ significant decrease; ↔ = unchanged; ∝ = correlates; ABA = activity based anorexia; BF = Bacteroides fragilis; BMI = body mass index; BCS = breast cancer survivors; CPK = creatine-phosphokinase; DIO = diet-induced obese; Ex = exercise; GF = germ free; HIIT = high-intensity interval training; HFD = high fat diet; HF/Ex = high fat exercise; HF/Sed = high fat sedentary; LF/Ex = low fat exercise; LF/Sed = low fat sedentary; ND = normal diet; PBS = phosphate buffer saline; SCFAs = short chain fatty acids; Sed = sedentary; SPF = specific pathogen-free; SHR = strain hypertensive rats; T1D = Type 1 diabetes; VO2max = maximum rate of oxygen consumption.
Table 2. List of the most useful sport supplements able to provide benefits in terms of health and performance according to Close et al. [135].
Table 2. List of the most useful sport supplements able to provide benefits in terms of health and performance according to Close et al. [135].
Strong EvidenceModerate or Emerging EvidenceLack of Evidence
Antioxidants (Polyphenols)
Probiotics
Proteins
BCAA 1
L-Glutamine
Sodium-Bicarbonate
Vitamin D
Omega-3 PUFAs 2
CHO 3-Electrolytes Sport Drink
L-Carnitine
Caffeine
Creatine
Taurine
Beta-Alanine
Beetroot Juice
Collagen
Glucosamine
Vitamin C
1 BCAA: Branched chain amino acids; 2 PUFAs: Polyunsaturated fatty acids; 3 CHO: Carbohydrates. Supplements are grouped based on their interaction with the gut microbiota: Strong (green), moderate/emerging (yellow), and lack of evidence (orange).
Table 3. Supplements and microbiota.
Table 3. Supplements and microbiota.
Supplement CategorySupplementReferenceModelsSample SizeAdministration (Dose/Day and Duration) of SupplementationEffects on Microbiome Taxonomy
AntioxidantFlavonols (Quercetin)[141]Rats23 Wistar rats
(n = 5–6/group)
30 mg/kg BW/day for 6 weeksFirmicutes:Bacteroidetes ratio; ↓ Erysipelotrichaceae,
Bacillus, ↓ Eubacterium cylindroides
Anthocyanidins[142]Mice36 mice
(n = 12/group)
Cranberry extract (200 mg/kg) for 8 weeksAkkermansia
Pomegranate Extract (Ellagitannin)[143]Humans20 healthy subjects1000 mg for 4 weeksActinobacteria; ↓ Firmicutes
ProbioticsLAB; LAB-ANTI[156]Humans30 triathletes subject
(n = 10/group)
Two capsules in the morning for 12-week pre-race period and the six-day post-race periodBifidobacteria
Firmicutes
Bacteroides
Lactobacillus rhamnosus CNCMI-4317[157]Mice18 mice
(n = 5-6-7/group)
11 days↑ Lactobacilli
lactobacillus plantarum TWK10[159]Mice24 mice
(n = 8/group)
LP10-1X:1 capsule 2.05 × 108 CFU/kg per day and LP10-5X: 1.03 × 109 CFU/kg for 6 weeks↑ Lactobacilli
lactobacillus plantarum TWK10[160]Humans16 male adults
(n = 8/group)
1 capsule per day for 6 weeks↑ Lactobacilli
Saccaharomyces boulardii[163]Wistar Rats26 males (11 weeks old)
(n = 13/group)
108 CFU·kg−1·day−1 for 10 daysSaccharomyces
Bacteroides
FirmicutesProteobacteria
Tenericutes
Kefir[169]Mice32 mice (n = 8/group)1 -vehicle group
2 -KF-1X, (2.15 g/kg/day)
3 -KF-2X, (4.31 g/kg/day)
4 -KF-5X, (10.76 g/kg/day)
for 28 days
KF-1X: ↑ Ruminococcaceae
KF-2X: ↑ BacteroidesBacteroidia
FirmicutesClostridiales
Clostridia
KF-5X: ↑ Bacteroides
Firmicutes
Rikenellaceae, ↑ Bacteroidales, ↑ Bacteroidia
Clostridia
Probiotic formulation (PF): consisting of Lactobacillus helveticus R0052 + Bifidobacterium longum R0175[175]Humans55 subjects
(n = 26–29/group)
1·5 g/d of PF, 1 time per day, for 30 daysLactobacillus;Bifidobacteria
Lactobacillus Casei[176]Humans39 Chronic Fatigue Syndrome patient
(n = 16–19/group)
1 sachet, three time per day, 8 weeksLactobacillus;Bifidobacteria
ProteinIsolated Soy Protein (SOY); Isolated Milk Protein (CAS); Control[185]Humans38 overweight subjects
(n = 13/group)
15% of participants’ habitual energy intake for 3 weeksNo effects on microbiota composition
Whey Isolate + Beef Hydrolysate[186]Humans24 endurance recreational athletes
(n = 12/group)
10 g of whey + 10 g of beef protein for 10 weeksBacteroidetes phylum; ↓ Firmicutes phylum; ↑ Bacteroides genus; ↓ Citrobacter genera; ↑ Klebsiella genera; ↓ B. Longum
BCAABCAA-enriched mixture[194]Mice18 BALB/c male mice
(n = 9/group)
1.5 mg/g BW for 4 monthsAkkermansia; ↑ Bifidobacterium; ↓ Enterobacteriaceae
GlutamineL-Glutamine; L-Alanine[200]Humans33 overweight/obese adults
(n = 12–21/group)
30 g for 14 daysFirmicutes:Bacteroidetes ratio; ↓ Veillonella genus; ↑ Prevotella genus
Sodium BicarbonateBicarbonate-enriched water[207]Humans19 healthy subjects2.5 g/L in 500 mL for 7 daysChristensennellaceae
Dehalobacteriaceae
Bifidobacteriaceae
Vitamin DVitamin D3[213]Humans16 healthy subjects980 IU/kg BW for 4 weeks, 490 IU/kg BW for 4 weeksUpper GI tract: ↓ Gammaproteobacteria (↓ Pseudomonas spp.; ↓ Escherichia/Shigella spp.); ↑ Bacterial richness.
No major changes in the terminal GI tract.
Omega-3Mixed eicosapentaenoic acid/docosahexaenoic acid[221]Humans22 healthy adults4 g for 8 weeksBifidobacterium, ↑ Roseburia, ↑ Lactobacillus
Fish oil; Sunflower oil[219]Humans132 infants
(n = 60–72/group)
9 monthsBacteroidetes
CaffeineInstant coffee powder[234]Humans16 healthy adults3.4 g of coffee in 150–200 mL water, 3 cups/day for 3 weeksBifidobacterium spp.
Green tea extract[235]Humans58 healthy adults
(n = 28–30/group)
0.27–0.45 g/day of caffeine for 12 weeksNo effects on microbiota composition
Abbreviations: ↑ significant increase; ↓ significant decrease; BW = body weight; KF = kefir; BCAA = branched-chain amino acids; GI = gastrointestinal.

Share and Cite

MDPI and ACS Style

Donati Zeppa, S.; Agostini, D.; Gervasi, M.; Annibalini, G.; Amatori, S.; Ferrini, F.; Sisti, D.; Piccoli, G.; Barbieri, E.; Sestili, P.; et al. Mutual Interactions among Exercise, Sport Supplements and Microbiota. Nutrients 2020, 12, 17. https://doi.org/10.3390/nu12010017

AMA Style

Donati Zeppa S, Agostini D, Gervasi M, Annibalini G, Amatori S, Ferrini F, Sisti D, Piccoli G, Barbieri E, Sestili P, et al. Mutual Interactions among Exercise, Sport Supplements and Microbiota. Nutrients. 2020; 12(1):17. https://doi.org/10.3390/nu12010017

Chicago/Turabian Style

Donati Zeppa, Sabrina, Deborah Agostini, Marco Gervasi, Giosuè Annibalini, Stefano Amatori, Fabio Ferrini, Davide Sisti, Giovanni Piccoli, Elena Barbieri, Piero Sestili, and et al. 2020. "Mutual Interactions among Exercise, Sport Supplements and Microbiota" Nutrients 12, no. 1: 17. https://doi.org/10.3390/nu12010017

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop