Next Article in Journal / Special Issue
Understanding How Commensal Obligate Anaerobic Bacteria Regulate Immune Functions in the Large Intestine
Previous Article in Journal
Metazoan Remaining Genes for Essential Amino Acid Biosynthesis: Sequence Conservation and Evolutionary Analyses
Previous Article in Special Issue
Dysbiotic Events in Gut Microbiota: Impact on Human Health
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Diet and Lifestyle on Gut Microbiota and Human Health

CSIRO Food and Nutrition Flagship, Kintore Ave, Adelaide, SA 5000, Australia
*
Author to whom correspondence should be addressed.
Nutrients 2015, 7(1), 17-44; https://doi.org/10.3390/nu7010017
Submission received: 17 September 2014 / Accepted: 9 December 2014 / Published: 24 December 2014
(This article belongs to the Special Issue Microbiome and Human Health)

Abstract

:
There is growing recognition of the role of diet and other environmental factors in modulating the composition and metabolic activity of the human gut microbiota, which in turn can impact health. This narrative review explores the relevant contemporary scientific literature to provide a general perspective of this broad area. Molecular technologies have greatly advanced our understanding of the complexity and diversity of the gut microbial communities within and between individuals. Diet, particularly macronutrients, has a major role in shaping the composition and activity of these complex populations. Despite the body of knowledge that exists on the effects of carbohydrates there are still many unanswered questions. The impacts of dietary fats and protein on the gut microbiota are less well defined. Both short- and long-term dietary change can influence the microbial profiles, and infant nutrition may have life-long consequences through microbial modulation of the immune system. The impact of environmental factors, including aspects of lifestyle, on the microbiota is particularly poorly understood but some of these factors are described. We also discuss the use and potential benefits of prebiotics and probiotics to modify microbial populations. A description of some areas that should be addressed in future research is also presented.

1. Introduction

There are approximately 10 times as many microorganisms within the gastro-intestinal (GI) tract of humans (approximately 100 trillion) as there are somatic cells within the body. While most of the microbes are bacteria, the gut can also harbor yeasts, single-cell eukaryotes, viruses and small parasitic worms. The number, type and function of microbes vary along the length of the GI tract but the majority is found within the large bowel where they contribute to the fermentation of undigested food components, especially carbohydrates/fiber, and to fecal bulk. Some of the most commonly found or recognized genera of gut bacteria in adults are Bifidobacterium, Lactobacillus, Bacteroides, Clostridium, Escherichia, Streptococcus and Ruminococcus. Approximately 60% of the bacteria belong to the Bacteroidetes or Firmicutes phyla [1]. Microbes which produce methane have been detected in about 50% of individuals and are classified as Archaea and not bacteria [2]. Although individuals may have up to several hundred species of microbes within their gut, recent findings from The Human Microbiome Project and others [3,4] show that thousands of different microbes may inhabit the gut of human populations collectively and confirm a high degree of variation in the composition of these populations between individuals. Despite this variation in taxa the abundance of many of the microbial genes for basic or house-keeping metabolic activities are quite similar between individuals [3]. There is growing evidence that imbalances in gut microbial populations can be associated with disease, including inflammatory bowel disease (IBD) [5], and could be contributing factors. Consequently, there is increased awareness of the role of the microbiota in maintaining health and significant research and commercial investment in this area. Gut microbes produce a large number of bioactive compounds that can influence health; some like vitamins are beneficial, but some products are toxic. Host immune defenses along the intestine, including a mucus barrier, help prevent potentially harmful bacteria from causing damage to tissues. The maintenance of a diverse and thriving population of beneficial gut bacteria helps to keep harmful bacteria at bay by competing for nutrients and sites of colonization. Dietary means, particularly the use of a range of fibers, may be the best way of maintaining a healthy gut microbiota population. Strategies such as ingestion of live beneficial bacteria (probiotics) may also assist in maintaining health. In this review, we will expand upon these subjects relating to diet and lifestyle, the gut microbiota and health, and provide some indication of opportunities and knowledge gaps in this area.

2. Microbial Products that Impact Health—Beneficial and Harmful

Microbial mass is a significant contributor to fecal bulk, which in turn is an important determinant of bowel health. Consumption of dietary fibers reduces the risk of colorectal cancer (CRC) [6] at least partly as a consequence of dilution and elimination of toxins through fecal bulk, driven by increases in fermentative bacteria and the presence of water-holding fibers [7,8,9]. Aspects of this will be discussed in more detail later in the review.
Gut microbes are capable of producing a vast range of products, the generation of which can be dependent on many factors, including nutrient availability and the luminal environment, particularly pH [10]. A more in-depth review of gut microbial products can be found elsewhere [11]. Microbial products can be taken up by GI tissues, potentially reach circulation and other tissues, and be excreted in urine or breath. Fermentation of fiber and protein by large bowel bacteria results in some of the most abundant and physiologically important products, namely short chain fatty acids (SCFA) which act as key sources of energy for colorectal tissues and bacteria, and promote cellular mechanisms that maintain tissue integrity [12,13,14]. SCFA can reach the circulation and impact immune function and inflammation in tissues such as the lung [15]. However, some protein fermentation products such as ammonia, phenols and hydrogen sulphide can also be toxic. There are many other products which deserve mention for their influence on health. Bacteria such as Bifidobacterium can generate vitamins (e.g., K, B12, Biotin, Folate, Thiamine) [11]. Synthesis of secondary bile acids, important components of lipid transport and turnover in humans, is mediated via bacteria, including Lactobacillus, Bifidobacterium and Bacteroides [11]. Numerous lipids with biological activity are produced by bacteria, including lipopolysaccharide (LPS), a component of the cell wall of gram negative bacteria that can cause tissue inflammation [16]. Also, many enteropathogenic bacteria (e.g., some E. coli strains) can produce toxins or cause diahorrea under the right conditions, but under normal circumstances other non-pathogenic commensal bacteria with similar metabolic activities outcompete and eventually eliminate them [17]. Bacteria such as Bifidobacterium can also help prevent pathogenic infection through production of acetate [18].
Many enzymes produced by microbes influence digestion and health. Indeed, much of the microbial diversity in the human gut may be attributable to the spectrum of microbial enzymatic capacity needed to degrade nutrients, particularly the many forms of complex polysaccharides that are consumed by humans [19]. Some bacteria such as Bacteroides thetaiotamicron have the capacity to produce an array of enzymes needed for carbohydrate breakdown [20], but in general numerous microbes appear to be required in a step-wise breakdown and use of complex substrates. Bacterial phytases of the large intestine degrade phytic acid present in grains, releasing minerals such as calcium, magnesium and phosphate that are complexed with it [21], making these available to host tissues (e.g., bone). Enzymes which degrade mucins help bacteria meet their energy needs and assist in the normal turnover of the mucus barrier lining the gut.
Competition between bacteria for substrates has a significant influence on which products are generated. Hydrogen is used by many bacteria and there is a hydrogen economy within the gut based around production by some bacteria and its use by others, including methanogens and sulphate-reducing bacteria (SRB) [22,23]. The use of hydrogen for production of methane by methanogenic Archaea may limit acetate production by other microbes, thereby potentially limiting production of beneficial butyrate and impacting health [2,23]. The role of methanogens in health is not yet clear. Breath methane correlates with levels of constipation in irritable bowel syndrome (IBS) [24] but methanogens numbers are depleted in IBD [2].
Production of gases such as methane, hydrogen, hydrogen sulphide and carbon dioxide is associated with digestion and fermentation within the GI tract. While excess production may cause GI problems such as bloating and pain, the gases may serve useful purposes. However, there is debate over whether hydrogen sulphide is largely beneficial or detrimental [23].
There is a strong interaction between the host immune system and the microbiota, with both producing compounds that influence the other. Some bacteria such as the key butyrate-producer Faecalibacterium prausnitzii may produce anti-inflammatory compounds [25]. Microbes also produce substances that allow communication between each other.

3. Lifestage and Lifetstyle Impacts on the Microbiota and the Influence of Nutrition

3.1. Lifestage

Microbes colonise the human gut during or shortly after birth. The fact that babies delivered naturally have higher gut bacterial counts at 1 month of age than those delivered by caesarean section [26] suggests gut colonization by microbes begins during, and is enhanced by, natural birth. The growth and development of a robust gut microbiota is important for the development of the immune system [27] and continues during breast-feeding, a stage which seems to be important for the long-term health of the individual. Oligosaccharides present in breast milk promote the growth of Lactobacillus and Bifidobacterium, which dominate the infant gut, and this can strengthen or promote development of the immune system and may help prevent conditions such as eczema and asthma [28,29,30]. These bacteria are undetectable in the stool of preterm infants in their first weeks of life [31]. A significant shift in the populations of gut microbes occurs when infants switch to a more solid and varied diet, including a decline in populations of Lactobacillus and Bifidobacterium to only a small percentage of the large bowel microbiota [32]. A wide diversity of microorganisms is needed to utilize the many fibers and other nutrients present in adult diets [19,33]. Functional maturation of the human microbiota, including the capacity to produce vitamins, increases during the early years of life [34].
The complexities and variability of adult gut microbial populations have become increasingly evident in recent years. The variability may relate to the influence of numerous factors, including diet and host genetics. The composition and activity of gut bacteria can vary according to (and possibly a result of) life events, including puberty, ovarian cycle, pregnancy and menopause [11]. The diets of children being weaned may have particular influence on microbial diversity in later life. Another broad shift in gut microbe populations occurs with age. The Bacteroidetes phylum bacteria tend to dominate numerically during youth but numbers decline significantly by old age, whereas the reverse trend occurs for bacteria of the Firmicutes phylum [11]. The consequences and reason for this change are not yet clear. However, the gut microbiota profiles of the elderly may not be optimal. One study found a high prevalence of potentially toxic Clostridium perfringens and lower numbers of Bifidobacterium and Lactobacillus in those in long-term care [35]. The latter also have a reduced microbial diversity compared to the elderly living in the community and this is related to increased frailty and changes in nutrition [36].

3.2. Lifestyle

The impact of non-dietary lifestyle factors on the gut microbiota has been largely ignored. Smoking and lack of exercise can significantly impact the large bowel (and potentially the microbiota) as they are risk factors for CRC [37]. Indeed, smoking has a significant influence on gut microbiota composition, increasing Bacteroides-Prevotella in individuals with Crohn’s Disease (CD) and healthy individuals [38]. Smoking-induced changes in microbial populations could potentially contribute to increased risk of CD. Air-borne toxic particles can reach the large bowel via mucociliary clearance from the lungs, and increased environmental pollution associated with industrialization could contribute to concomitant increases in IBD cases [39].
Another lifestyle factor, stress, has an impact on colonic motor activity via the gut-brain axis which can alter gut microbiota profiles, including lower numbers of potentially beneficial Lactobacillus [40]. Stress may contribute to IBS, one of the most common functional bowel disorders, and the associated changes in microbial populations via the central nervous system (CNS). The gut-brain axis is bi-directional, involving both hormonal and neuronal pathways [41], and so changes in the gut microbiota may influence brain activity, including mood [42]. Autism, a neurodevelopmental disorder, is associated with significant shifts in gut microbiota populations [43,44,45].
Obesity is associated with excess energy intakes and sedentary lifestyles. Exercise (or rather a lack of it) may be an important influence on any shifts in microbial populations that are associated with obesity. This is highlighted by a recent study that showed an increase in the diversity of gut microbial populations in professional athletes in response to exercise and the associated diet [46]. In humans and animal models with obesity, shifts in gut microbial populations occur, with increases in the Firmicutes and decreases in the Bacteroidetes, which could potentially contribute to adiposity through greater energy harvest [47,48,49]. However, other data suggests the shifts in microbial populations are driven primarily by the high fat obesogenic diets [50,51]. Irrespective of the cause, there are associated increases in gut bacteria linked with poor health outcomes (e.g., Staphylococcus, E. coli, Enterobacteriaceae) [52,53]. Dietary saturated fats may increase numbers of pro-inflammatory gut microbes by stimulating the formation of taurine-conjugated bile acids that promotes growth of these pathogens [54].
Geography also has a strong bearing on the composition of gut microbial populations. The diversity of fecal microbes in children from rural Africa is greater than that of children of developed communities in the EU, as is the number of bacteria associated with breakdown of fiber [55], suggesting dietary differences contributes significantly to the microbial differences. In another study, the type of fecal bacteria and their functional genes differed between individuals in the USA and in rural areas of Venezuela and Malawi [34].
Other environmental factors may also influence health via gut microbes. Travel, particularly to overseas destinations, increases the risk of contracting and spreading infectious diseases, including those causing diarrhoea. Some infections may go undiagnosed but result in long-term GI problems, including IBS [56]. Poor sanitary conditions in developing countries, and poor personal hygiene, can facilitate the spread of infectious agents. Circadian disorganization, occurring because of travel, shift work or other reasons, also impacts gut health and alters gut microbial populations [57].

4. Impacts of Macronutrients on the Gut Microbiota and Relevance to Health

4.1. Substrate Supply to the Colonic Microbiota

An adult colon contains approximately 500 g of contents, most of which is bacteria [58], and about 100 g/day is voided as stool. A typical western type diet supplies the colonic microbiota with about 50 g daily of potentially fermentable substrate, predominantly dietary fiber (DF). Non-starch polysaccharides (NSP) are major components of DF and account for 20%–45% of the dry matter supplied to the colon. Simple sugars and oligosaccharides each represent a further 10% whereas starch (and starch hydrolysis products) supplies less than 8% of dry matter. Some sugar alcohols also escape small intestine (SI) absorption and are minor dietary substrates for the colonic microbiota [59]. About 5–15 g of protein and 5–10 g of lipid passes into the proximal colon daily, largely of dietary origin. Various other minor dietary constituents, including polyphenols, catechins, lignin, tannins and micronutrients also nourish colonic microbes. About 90% of the approximately 1 g/day of dietary polyphenols escapes digestion and absorption in the SI [60,61] and can have significant influence on microbial populations and activities [62,63,64].

4.2. Carbohydrates—Importance for Large Bowel Fermentation and Health

Carbohydrates are the principal carbon and energy source for colonic microbes. Collectively, they have an immense capacity to hydrolyse a vast range of these nutrients, especially complex polysaccharides [65].
DF is integral to a healthy diet and Australian adults consume ~27 g each day [66], which is greater than in other high income countries, including the USA (<20 g/day). Epidemiological and experimental studies show that DF is both preventative and therapeutic for many large bowel disorders and other conditions or diseases, including cardiovascular diseases, type II diabetes and obesity [67,68,69,70,71].
One mechanism by which fiber promotes and maintains bowel health is through increasing digesta mass. Incompletely fermented fiber (e.g., insoluble NSP such as cellulose), increases digesta mass primarily though its physical presence and ability to adsorb water. An increase in digesta mass dilutes toxins, reduces intracolonic pressure, shortens transit time and increases defecation frequency. Fibers can also increase fecal mass to a lesser degree by stimulating fermentation, which leads to bacterial proliferation and increased biomass [7].
Many of the health benefits ascribed to fiber are a consequence of their fermentation by the colonic microbiota and the metabolites that are produced. Carbohydrates are fermented to organic acids that provide energy for other bacteria, the bowel epithelium and peripheral tissues. SCFA are the major endproducts of carbohydrate fermentation. These weak acids (pKa ~4.8) help lower the pH within the colon thereby inhibiting the growth and activity of pathogenic bacteria. Other minor organic acids produced include lactate, succinate and formate. Branched-chain SCFA (e.g., isobutyrate and isovalerate) results from fermentation of branched chain amino acids [72].
There are spatial gradients in microorganisms along the length of the gut. Bacterial growth and metabolic activity (fermentation) is greatest in the proximal colon where substrate availability is at a maximum [13,73]. Accordingly, pH progressively increases as stool progresses from the proximal to distal colon (from 5.8 to 7.0–7.5), largely because of the progressive depletion of carbohydrate substrates and absorption of SCFA, and increasing efficiency of protein fermentation and production of alkaline metabolites [72]. Total SCFA concentrations are highest in the proximal colon (~100 mM) and decline progressively toward the distal colon. Acetate, propionate and butyrate are the major individual SCFA, accounting for 90% of the total, with molar ratios approximating 65:20:15 [74].
Butyrate has attracted significant attention because it serves as the principal source of metabolic energy for the colonocytes [75], is instrumental in maintaining mucosal integrity, modulates intestinal inflammation and promotes genomic stability. The capacity of butyrate to regulate colonocyte differentiation and apoptosis, promoting removal of dysfunctional cells, underscores its potential to protect against colon cancer [76].
The SCFA also have roles beyond the gut and may improve risk of metabolic and immune system diseases and disorders, such as osteoarthritis, obesity, type II diabetes and cardiovascular disease [13,76].
More than 90% of the total SCFA produced in the colon is absorbed by the epithelium, through mechanisms that are not fully elucidated. SCFA-stimulated sodium-coupled transport in the apical membrane of colonocytes is especially important as it mediates (co)absorption of water and helps recover electrolytes as well as energy [77]. The SCFA can bind to G-protein coupled receptors in colorectal tissues, particularly GPR 41 and 43, which may influence immune function and tumour suppression, but these pathways are still relatively poorly characterized [76].
Most of the absorbed acetate reaches the liver via the portal vein, whereas propionate, and butyrate to an even larger extent, is metabolized extensively by colonocytes. Acetate and propionate are used by the liver for oxidation, and for lipogenesis and gluconeogenesis, respectively. Hepatic metabolic clearance of SCFA is very high and so concentrations in the systemic bloodstream are about 100-fold lower than those in colonic digesta and feces (~50 µM versus 100 mM, respectively) [13].

4.3. Protein

Dietary proteins are an important part of a balanced diet. Humans are unable to synthesize numerous amino acids and must obtain them from proteins in food to maintain health. Some protein-rich foods such as meat, eggs and nuts are also good sources of vitamins or nutrients such as iron. There is good evidence that a diet containing moderate to high amounts of protein can also contribute to weight loss in overweight individuals, particularly if combined with exercise [78], thereby minimising the health risks associated with obesity. Dietary proteins also have a significant impact on gut health. Depending on the type of protein and the other nutrients present in the food this can be beneficial or harmful. Some epidemiological studies, particularly large studies (up to 500,000 people), indicate a slight but significant association between CRC risk and the consumption of high levels of red and processed meats [79,80,81,82]. Not all epidemiological studies show such an association and the inconsistent findings may relate to the many factors which may contribute to CRC [83,84].
The potential for protein to harm colorectal tissues is explicable using current knowledge. An increase in protein intake usually results in more of the macronutrient, and hence fermentable substrate, reaching the colon. Although protein digestibility has an important influence on how much reaches the colon, most common dietary protein sources are highly susceptible to hydrolysis by SI enzymes. Dietary protein serves as the major source of nitrogen for colonic microbial growth and is essential to their assimilation of carbohydrates and the production of beneficial products such as SCFA. Hence, a combination of protein and carbohydrates in the large bowel can contribute to bowel health. However, unlike carbohydrates, fermentation of protein sources by the microbiota produces a much greater diversity of gases and metabolites, and increasing the nitrogenous substrate for the microbiota can also increase putrefactive fermentation products [85]. As digesta passes down the bowel its carbohydrate content dwindles and protein fermentation becomes progressively more important. Putrefactive fermentation has been implicated in the development and progression of many common bowel diseases given their greater prevalence in the distal colon [86], including CRC and IBD. Many of these protein fermentation endproducts, which include ammonia, hydrogen sulphide, amines, phenols, thiols and indoles, have been shown to be cytotoxins, genotoxins and carcinogens [87], in in vitro and animal models [88]. Generally, fecal levels of protein fermentation products, such as sulphide, are positively associated with dietary protein consumption in humans and there is evidence from rat studies that higher dietary protein intake (including higher red meat intake) is associated with greater DNA damage in colonic mucosa when dietary levels of fermentable carbohydrate are low [88,89,90,91]. Recently completed studies suggest that this relationship holds true for humans [92,93,94]. However, higher protein intake does not always result in higher fecal levels of protein fermentation products [95] nor does it necessarily increase the genotoxicity of fecal water in humans [96].
Although ammonia is a well-known toxin [97] it is used as an N source by the microbiota and most is excreted via stool or absorbed in the gut and eliminated in urine. Diets promoting microbial protein synthesis (and concomitant increased utilisation of ammonia), effectively reroute systemic N excretion from the kidneys to the fecal stream, which has benefits for renal health [98]. Other components derived from dietary protein sources such as red meat may also influence the gut microbiota and health. Microbial metabolism of l-carnitine, abundant in red meat, may generate products such as trimethylamine-N-oxide that could increase risk of atherosclerosis [99].

4.4. Fat

Dietary fat also influences the composition and metabolic activity of the gut microbiota and some evidence for this has been described earlier in relation to obesity.
High fat diets induce increased circulating levels of bacteria-derived LPS in humans, possibly as a consequence of increased intestinal permeability [100]. LPS is an immune system modulator and potent inflammatory agent linked to the development of common metabolic diseases.
The influence of dietary fat on the gut microbiota may be indirectly mediated by bile acids. Hepatic production and release of bile acids from the gall bladder into the SI, and the amount that escapes enterohepatic recycling and enters the colon, is increased with fat intake. Secondary bile acids, produced by 7 α-dehydroxylation of primary bile acids by colonic microbiota, are potentially carcinogenic and have been implicated in the aetiology of CRC and other GI diseases [101,102]. Further research is required on the interactions between dietary fat, the type and amount of bile acids that reach the large bowel, and the population structure and function of the microbiota in that viscus.

5. Effects of Polyphenols on the Microbiota

Dietary polyphenols, sourced from many foods including grapes, grains, tea, cocoa and berries, generally promote health and are linked to prevention of diseases such as cancer and cardiovascular disease [103]. Although many dietary polyphenols may have biological impacts through anti-oxidant effects or anti-inflammatory pathways [103], polyphenols which reach the colon can be metabolized by the resident microbiota and result in bioactive products, but our understanding of the microbial bioconversion processes is limited [104,105,106]. Metabolic profiling of polyphenolic products in excreta and blood using tools such as NMR is enabling greater insights into effects of dietary polyphenols in humans [107] but linking the metabolic changes to health outcomes remains a challenge [108]. Individual differences in microbiota populations may result in different capacities for polyphenol bioconversion [109] with potential consequences for health. In this context, it is noteworthy that the gut microbiota population profiles of individuals with IBD are significantly different from healthy individuals, and also that the polyphenolic metabolite profiles are also different between the two groups [110].

6. Western-Style Diets

The Western lifestyle, including diet, is associated with high incidences of chronic diseases, such as cardiovascular disease, CRC and type II diabetes which individually and collectively carry a hefty socioeconomic burden [111]. Most Western populations over-consume highly refined, omnivorous diets of poor nutritional quality. Those diets are energy dense, high in animal protein, total and saturated fats, and simple sugars but low in fruits, vegetables and other plant-based foods. Consequently, they are typically low in DF, NSP in general and RS in particular. For Western civilisations, refined cereal products (e.g., white bread) are the main DF source. Overfeeding (and sedentary behaviour) is also a hallmark of these populations.
Much of what is known about the diversity and complexity of human gut microbiota comes from molecular analysis of fecal samples obtained mainly from small cohorts of Caucasian adults habitually consuming Western style diets. Considerably less is understood about how other dietary patterns (e.g., vegetarian, Mediterranean) might influence the community structure and metabolic activity of microbiota.

7. Diet and Dietary Change

In humans, the microbial gene set is 150 times larger than the gene complement of the host [112]. However, only about 50 species belonging to just five or six genera and two phyla account for 99% of biomass. Of the genera Bacteroides, Bifidobacterium and Eubacterium are numerically the most important and may account for more than 60% of culturable bacteria present in human stool. Clostridium, Enterobacteriaceae and Streptococcus are also important but less numerous. Nearly all (~90%) of the bacteria in the human gut can be mapped to just two phyla, Bacterioidetes and Firmicutes. The relative proportions of the two dominant phyla vary, and can be influenced by a range of factors, but most people have similar proportions of each [113].
Long-term, habitual diet (i.e., dietary pattern) and shorter term dietary variation influences gut microbiota composition. The population structure is responsive to acute dietary change (daily variation), as evidenced by rapid and substantial increases in populations at the genus and species level. However, dietary change does not necessarily result in a permanent (paradigm) compositional shift, at least at phylum level, although evidence for this assertion is limited [114].

8. Dietary Patterns, Macronutrients and Microbiota Taxonomic Composition

8.1. Observational Studies

Cross-sectional studies have shown some evidence that Western-style diets are associated with gut microbial populations that are typified by a Bacteroides enterotype whereas traditional diets rich in plant polysaccharides are associated with a Prevotella enterotype [114]. The Prevotella enterotype was only weakly associated with components that typify Western diets but strongly linked to carbohydrates and simple sugars. The fecal microbiota of children in the USA is dominated by Bacteroides [34,115]. Similarly, Italian children have high levels of Enterobacteriaceae (mainly Shigella, Escherichia and Salmonella). In contrast, the stool of children in rural Africa and South America consuming traditional plant-based diets was enriched in Bacteroidetes, in particular the Prevotella enterotype and species associated with fiber utilization (e.g., Xylanibacter) [55]. Prevotella and (Xylanibacter) are known to use cellulose and xylans as substrates [55,116]. Diets of North American and Italian urban children are much richer in animal protein and saturated fats whereas the diets for the other two populations are plant-based and have higher levels of fiber. The Bacteroidetes:Firmicutes ratio was lower for children in the Western countries.
As stated earlier, there is a paucity of data on the association between vegetarian dietary patterns and the gut microbiota, especially using molecular methods. A study that used PCR-denaturing gradient gel electrophoresis (DGGE) for microbial population fingerprinting found no significant differences in the fecal microbiota of vegetarians and omnivores, although the abundance of Clostridium cluster IV in the latter tended to be greater [117]. In a cohort of female college students from rural India, the fecal microbiota of those whose dietary pattern was omnivorous had a greater relative abundance of Clostridium cluster XIVa bacteria, specifically Roseburia-E. rectale (butyrate-producing bacteria), compared to the lacto-vegetarians [118]. There were no differences in the relative proportions of other major bacterial groups targeted. A gene encoding for a pivotal enzyme (butyryl-CoA CoA-transferase) involved in butyrate synthesis was also upregulated in the omnivores. The study demonstrates differences in the composition and functional capacity of the microbiota of individuals with two markedly diverse dietary patterns.
The taxonomic diversity of the fecal microbiota of individuals on habitual Western diets appears to be less than for those consuming plant-based diets. Also, individuals who are obese or have type II diabetes, inflammatory diseases (osteoarthritis) and other major health problems (prevalent in Western societies) have a sub-optimal fecal microbiota profile. Specifically, it is less diverse than that of healthy controls [119,120] and there are also major compositional differences at the phylum level. Obesity is associated with an increased fecal Bacteroidetes:Firmicutes ratio relative to lean subjects [121]. Whether a microbiota with lower compositional diversity is less resilient to environmental challenges and is less “healthier” for the host is not yet known [122].
The fecal hydrogenotrophic microbiota of native Africans, whose diet is low in animal products, compared to that of African and European Americans consuming a typical Western diet was more diverse and contained different populations of hydrogenotrophic Archaea and methanogenic Archaea as well as SRB populations [123]. The differences in bacterial community structures of native African populations were reflective of the diets of the hosts. Those on Western diets, characterized by higher intakes of dietary animal proteins (as meat, milk and eggs), may deliver greater amounts of sulphur compounds to the colonic microbiota [124], thus favouring sulfidogenic hydrogen disposal whereas in native Africans methane is the major hydrogen sink. Native African populations have lower intake of animal products and higher breath methane concentrations than westernized populations [123,125].

8.2. Dietary Interventions

Replacing a habitual Western diet with one high in fiber elicited rapid (within 24 h) and marked alterations in fecal microbiota composition, although the changes were insufficient to produce a broad switch from Bacteroides to Prevotella enterotype [114].
In an inpatient study [126], altering dietary energy load in lean and obese adults induced rapid changes in the proportional abundance of Bacteroidetes and Firmicutes. The former decreased whereas the latter increased with increasing energy intake. Further studies are required to determine if the changes in microbiota composition were the result of the increase in dietary fat or another macronutrient. High fat diets are also associated with substantial compositional changes in the colonic microbiota at the phylum and genus levels, including reductions in both Gram positive (e.g., Bifidobacterium spp.) and Gram negative bacteria (e.g., Bacteroides) [123].
Animal models are also proving useful in understanding factors that impact the gut microbiota, particularly in regards to high fat diets and obesity. A study using a murine (RELMβ) knockout model showed that dietary fat-induced changes to gut microbiome composition were independent of obesity [127]. In conventional mice, increased dietary fat intake resulted in fewer numbers of Bacteroidetes and increases in Firmicutes and Proteobacteria. A high fat diet also reduced cecal Bifidobacterium numbers and increased circulating LPS concentrations [128,129] and has also been shown to reduce the abundance of Clostridium cluster XIVa, including Roseburia spp. [130]. Diet-induced changes in mucosal integrity have been shown to promote metabolic endotoxemia and trigger systemic low grade inflammatory responses in a range of tissues [100,128,129].

9. Microbes and Mucosal Health

A layer of mucus, produced by goblet cells, lines the epithelium of the GI tract and acts as a barrier to microbial invasion of tissues and can contribute to intestinal homeostasis [131,132]. The basic component of mucus is mucin. Some bacterial products (SCFA) stimulate the production of mucus in response to dietary components such as NSP [133]. Over-utilization of the mucus by bacteria or reduced production can lead to thinning of the barrier under certain dietary conditions [88]. In the colon, “mucin-depleted foci” may develop as one of the features associated with tumorigenesis in rodents and humans in response to carcinogens [134]. However, the role of mucin depletion in oncogenesis is not clear as a recent study in rats showed that inflammation associated with mucin-depleted foci was not due to infiltration of bacteria, whereas colonic tumors did appear to be colonized by bacteria [135]. Many bacteria can adhere to and degrade the outer layer of colonic mucus but the inner layer is generally bacteria free [136]. Although break-down of mucus by bacteria is a normal part of mucus barrier turnover, an overabundance of mucus-degrading bacteria, such as Akkermansia muciniphila in the adherent mucus layer of individuals with IBD [137,138], could contribute to tissue inflammation by weakening the barrier.
Tight junctions between cells also helps prevent translocation of bacteria and molecules (including toxins) across gut epithelial tissues. A loss of this integrity (a so-called “leaky gut”) may have serious consequence for health. In the first few years of life, interactions between the gut microbiota and the mucosal barrier appear important and perturbations in the relationship that lead to excessive gut permeability and immune changes may result in susceptibility to a range of diseases in later life [139]. A significant proportion of the activities of the immune system occur within the gut. Gut-associated lymphatics contribute substantially to this defense but other cells lining the gut also produce a range of molecules which can neutralise pathogenic microbes. Dendritic cells sample the gut luminal environment for harmful bacteria and can induce a suite of responses including the activation of macrophages, B cells and T cells within mucosal tissues and the release of broad specificity ant-microbial agents such as Immunoglobulin A and α-defensins into the luminal environment [140].
A loss of gut barrier function may contribute to numerous diseases. An example is Parkinsons disease (PD), a multi-system disease in which there is dysfunction of the GI tract, including changes in the enteric nervous system which appear before obvious degeneration of the CNS [141,142]. Individuals with PD have increased intestinal permeability, greater intestinal infiltration of E. coli and greater endotoxin (LPS) exposure, and these changes correlate with the enteric neuronal damage [143], leading to suggestions that a pathogen may be responsible for PD [144] and a breakdown in mucosal barrier function may play a central role. An impaired gut barrier may also contribute to symptoms or complications of autism, kidney disease, type 2 diabetes, cardiovascular disease, metabolic syndrome, obesity, and liver diseases [45,100,145,146,147,148,149].

10. Inter-Individual Variation in Gut Microbiota and Responses to Diet

Each individual has a distinct combination of gut microbial species. This has become increasingly evident from molecular analyses of recent decades, including The Human Microbiome Project. One metagenomic analysis also suggested that the gut microbiota of each human is typified by one of three enterotypes, with each enterotype characterised by distinct dominant groups of microbes [150], namely Bacteroides, Prevotella and Ruminococcus. However, subsequent studies, including those of The Human Microbiome Project, have been unable to provide clear support for the concept as initially proposed [4,114]. More recent findings and analysis of the evidence roughly support typing with Prevotella or Bacteroides dominance of the microbiota but the numerous factors, especially dietary, that impact gut microbial populations means there is considerable variation in numbers of these genera, making it difficult to classify populations as a particular “type” [113].
Inter-individual differences in populations of the gut microbiota may lead to different capacities to utilize dietary components and to different levels of disease risk. For example, some individuals have consistently low stool levels of the microbial fermentation product butyrate, levels which generally remain lower relative to others despite concentrations increasing in response to a diet high in RS [151]. Butyrate production is important for the maintenance of colorectal tissue integrity and may protect against colorectal diseases [13,76]. Individual differences in numbers and functions of bacteria such as Ruminococcus bromii, important for the generation of SCFA in response to RS in humans [152,153], could potentially influence colorectal health.

11. Use of Probiotics and Prebiotics as Nutritional Strategies to Improve Health

Probiosis and prebiosis are diet-based processes/strategies for promoting the health of the host through improving the composition of the colonic microbiota. Although both prebiotics and probiotics have been shown to increase numbers of selected bacteria at the species and genus level, typically Bifidobacterium and Lactobacillus, changes in the overall composition of the gut microbiota are often relatively small, and generally persist only for as long as the period of the intervention. Also, definitive proof that the identified compositional alterations are directly responsible for an improvement in host health generally remains elusive. While the concepts have practical relevance they are simplistic given the current limited understanding of the complex and dynamic interplay between the host and their gut microbiota.
Prebiotics are dietary substrates that selectively promote proliferation and/or activity of “beneficial” bacteria indigenous to the colon. The concept, first published by Gibson and Roberfroid [154] in 1995, has been refined and redefined on several occasions. Prebiotics are defined currently as “selectively fermented ingredients that result in specific changes, in the composition and/or activity in the GI microbiota, thus conferring benefit(s) upon host health” [155].
To qualify as a prebiotic all of the following properties must be demonstrated: (i) a food ingredient that escapes assimilation in the small intestine; (ii) upon reaching the colon its fermentation by the microbiota flora selectively alters its taxonomic composition and/or activity which (iii) confers demonstrable health benefits for the consumer [156].
The validity of the prebiotic concept and evidence of a role for prebiotics in promoting health and reducing risk of bowel and systemic diseases have been recently reviewed in depth [157,158,159,160]. Data from studies in animals provides strong evidence of the potential of prebiotics to afford protection against a range of chronic diseases or conditions common in humans (e.g., CRC, IBD, type 2 diabetes, obesity) by preventing colonization by enteric pathogens [61,158,161]. Prebiotics have been shown to improve bowel and immune function, metabolic health and mineral bioavailability in humans but the evidence is strong only for bowel habit and colonic uptake of calcium and magnesium. There is mounting evidence that prebiotics both directly and indirectly modulate the immune system and reduce the risk and severity of bowel infectious and inflammatory conditions, such as IBD, as well as functional bowel disorders, notably IBS [159].
Short-chain nondigestible carbohydrates (inulin-type fructans, fructo-oligosaccharides (FOS) and galacto-oligosaccharides (GOS)) are the quintessential prebiotics and the target bacterial groups are typically Bifidobacterium and Lactobacillus. Fructan prebiotics, such as inulin and FOS, occur naturally in various foods including cereals, fruits and vegetables and so are ubiquitous in most diets. Dietary intakes have been estimated to be ~5–10 g/day [162].
The prebiotic concept as it currently stands is probably too narrowly focused. It has been proposed [163,164] that the taxonomic focus should be widened beyond the Bifidobacterium and Lactobacillus which have been historical targets. These genera may not be the most important contributors to host health. Emerging candidates include Ruminococcus bromii, Roseburia intestinalis, Eubacterium rectale, and Faecalibactrium prausnitzii, but there are many others that may be of benefit. It has been suggested that a prebiotic index might offer greater utility for evaluating the efficacy of different prebiotics [165]. The prebiotic concept also encompasses selective improvements in metabolic activity of the microbiota but this has been given little attention to date. Changes in concentration patterns of key beneficial microbial metabolites such as butyrate should be integrated into prebiotic index models.
All established prebiotics to date are carbohydrates, specifically inulin type fructans and GOS. However, other dietary carbohydrates also qualify as prebiotics, for instance resistant starch (RS) [156,166], but evidence from human studies is limited. More studies are required on the prebiotic properties of different types, doses and food sources of RS. The inter-individual variability in the microbial response to RS suggests successful dietary interventions with RS need to be personalised [167]. Dietary constituents other than carbohydrates conceivably could function as prebiotics. For instance, cocoa flavonols can increase the relative abundance of Bifidobacterium and Lactobacillus at the expense of potentially pathogenic bacteria, notably the C. histolyticum group [64].
Probiotics are defined as live microorganisms which when administered in adequate amounts confer a health benefit on the host. The most commonly consumed probiotics belong to the genus Lactobacillus and Bifidobacterium. Mechanisms by which probiotics might improve host health include immune function augmentation through reinforcing mucosal barrier function, reducing mucosal transfer of luminal organisms and metabolites to the host, increasing mucosal antibody production, strengthening epithelia integrity and direct antagonism of pathogenic microorganisms. However, the results of studies in humans are varied, due most likely to methodological differences (dose and duration of probiotic administration, sampling regimen and microbiological techniques) and differences in host cohorts (age, health status). Perhaps most importantly, it is clear from in vivo studies in humans and animal models that probiotic efficacy in promoting health is strain dependent and not species and genus specific. For a more comprehensive and detailed description of the health benefits of probiotics and their prophylactic potential for various gut diseases the reader is referred to recent narrative and systematic reviews [168,169,170,171,172,173].

12. Gaps in Understanding

There are still many gaps in our understanding of the interactions between diet, lifestyle, gut microbes and health. Here, we present some of the areas we believe should be addressed to help fill that knowledge gap.
There is a growing need for an understanding of the activities of gut microbes, particularly their physiological relevance. Current molecular methods such as sequencing technologies are allowing the identification of the many hundreds of microbial species present in human GI tracts and are beginning to identify the types of genes that they possess. The next steps will be to understand the functions of the many poorly characterised microbes, particularly their roles in the breakdown of food and how the associated by-products contribute to health and disease.
The majority of gut microbes are present within the large bowel and most GI microbiology research has focused on this area. However, the SI can, like the colon and rectum, become inflamed in cases of IBD and bacteria are implicated. The role that bacteria play in SI enteropathies and leaky gut is also yet to be clearly elucidated. The contribution of diet to maintenance of SI health is also not well understood. Understanding in this area is hampered by the general inaccessibility to these sites within human subjects, especially in healthy individuals who have no need to visit a gastroenterologist.
The integrity of the gut mucosa is of critical importance to health. Understanding which foods or dietary components strengthen or weaken that barrier may assist in tailoring diets to prevent microbes and toxins such as LPS from accessing tissues and causing inflammation. A better understanding of the interactions between the host immune system and gut bacteria, particularly in children, should also shed light on how microbes may contribute to lifelong susceptibility to some diseases, and how diet may be used to promote optimal microbial populations.
The gut-brain axis is increasingly viewed as having an important role in health with bi-directional communication of information of relevance to areas such as satiety, mood and gut motility and suggestions of roles in conditions that include IBS and autism. The gut microbiota has been implicated in some of these conditions and there is great scope for research into understanding which of the many microbial products reach the CNS and impact health, including mental health, via the brain, and consequently for understanding how dietary manipulation of the microbiota then impacts these important areas.
Since it has been shown that many microbial products can influence health, the inter-individual variation in gut microbial profiles in humans may lead to differences in disease risk. A better understanding of the origins of the variation may ultimately allow the microbial profiles to be modulated. Environmental and dietary factors appear to play some role during a child’s early development but the extent to which host genetics contribute to the variation is not known. Studies which follow the development of microbial profiles in children, and the impact that diet and environment have on these, are sorely needed. It may be possible to develop diets that lead to optimal microbial population and health outcomes.
The extent to which long-term dietary patterns can shift the composition of microbial populations is yet to be clearly determined despite some emerging knowledge and should be investigated further. Our understanding of how different sources and forms of macronutrients such as carbohydrates, proteins and lipids interact and affect the GI tract is still lacking. Knowledge of how micronutrients impact the gut and its microbes is even scarcer.
Food structure is also an important determinant of how a food impacts the body, with particle size and the associated food matrix influencing the accessibility of host and microbial enzymes to nutrients. For example, smaller starch granules may be more readily degraded due to higher surface area to volume ratios and this increases the rate at which sugars are absorbed by tissues, an important issue when considering glycaemic control. Since most gut microbes are within the large bowel, food structures which minimize SI digestion and allow food to pass into the colon will have the greatest impact on the microbiota. Cooking practices can potentially impact food structure and digestibility of foods. Strategies which optimize the structure of foods for defined benefits (i.e., glycemic control) are already being implemented but more work is needed in this area to achieve a broader range of health outcomes.
Ingestion of probiotics, prebiotics and microencapsulated nutrients, beneficial molecules or microbes [174,175] is designed to deliver a health benefit to the body by increasing numbers of beneficial microbes or their products with the gut. A greater knowledge of which microbes and functions are beneficial is needed to effectively culture, deliver and/or stimulate the growth of the appropriate microbes. Presently, only a small number of adult human gut microbes have been used for probiotics, or targeted in assessments of the impacts of diets (including prebiotics) on gut health.

13. Conclusions

We have sought to provide a broad picture of how diet, and to some extent lifestyle, can have significant and wide-ranging impacts on human health and shown that the microbes which inhabit the GI tract play an important role in mediating these effects. Although significant gains have been made recently in our understanding of the complexity of gut microbial populations, a more detailed understanding is needed of microbial functions and products that maintain (or negatively impact) the integrity of tissues, at sites within and distant from the gut. Alongside this is a need to understand which factors within the diet supply substrates to the microbes so that this knowledge can be harnessed to generate the desired shifts in microbial populations, products and health outcomes. Particularly challenging will be the task of understanding what constitutes a healthy population of gut microbes. Certain microbial population profiles may be associated with diseases and conditions. In many cases, it is not clear if environmental/lifestyle factors, diet or genetic predisposition leads to these profiles, or indeed whether the altered microbial populations contribute to the condition. While dietary intervention can induce significant change, it is possible that the level of impact may not always be sufficient to engineer the changes in microbial populations that are conducive to better health. The use of probiotics and other strategies may be required. An understanding of the ontogenesis of our gut microbial population profiles, and how this contributes to the development of our immune system, may enable early intervention or prevention of the formation of undesirable microbial profiles and the consequences. In this context, defining the factors which dictate the development of our human microbial populations in early life will be important.

Acknowledgments

All authors have read and approved the final manuscript. This review was supported in part by funds provided by Meat and Livestock Australia. The authors wish to thank David Topping for constructive input.

Author Contributions

Both authors contributed to the writing of the manuscript and approved the final version.

Conflicts of Interest

The authors declare a potential conflict of interest resulting from the financial support of Meat and Livestock Australia.

References

  1. Backhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-bacterial mutualism in the human intestine. Science 2005, 307, 1915–1920. [Google Scholar] [CrossRef] [PubMed]
  2. Scanlan, P.D.; Shanahan, F.; Marchesi, J.R. Human methanogen diversity and incidence in healthy and diseased colonic groups using mcrA gene analysis. BMC Microbiol. 2008, 8, 79. [Google Scholar] [CrossRef]
  3. Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T.; Creasy, H.H.; Earl, A.M.; Fitzgerald, M.G.; Fulton, R.S.; et al. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [Green Version]
  4. Huse, S.M.; Ye, Y.; Zhou, Y.; Fodor, A.A. A core human microbiome as viewed through 16S rRNA sequence clusters. PLoS One 2012, 7, e34242. [Google Scholar] [CrossRef]
  5. Manichanh, C.; Rigottier-Gois, L.; Bonnaud, E.; Gloux, K.; Pelletier, E.; Frangeul, L.; Nalin, R; Jarrin, C.; Chardon, P.; Marteau, P.; et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 2006, 55, 205–211. [Google Scholar]
  6. Bingham, S.A.; Day, N.E.; Luben, R.; Ferrari, P.; Slimani, N.; Norat, T.; Clavel-Chapelon, F.; Kesse, E.; Nieters, A.; Boeing, H.; et al. Dietary fibre in food and protection against colorectal cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC): An observational study. Lancet 2003, 361, 1496–1501. [Google Scholar]
  7. Stephen, A.M.; Cummings, J.H. Mechanism of action of dietary fiber in the human colon. Nature 1980, 284, 283–284. [Google Scholar] [CrossRef] [PubMed]
  8. Cummings, J.H.; Bingham, S.A.; Heaton, K.W.; Eastwood, M.A. Fecal weight, colon cancer risk, and dietary-intake of nonstarch polysaccharides (dietary fiber). Gastroenterology 1992, 103, 1783–1789. [Google Scholar] [PubMed]
  9. Birkett, A.M.; Jones, G.P.; de Silva, A.M.; Young, G.P.; Muir, J.G. Dietary intake and faecal excretion of carbohydrate by Australians: Importance of achieving stool weights greater than 150 g to improve faecal markers relevant to colon cancer risk. Eur. J. Clin. Nutr. 1997, 51, 625–632. [Google Scholar] [CrossRef] [PubMed]
  10. Duncan, S.H.; Louis, P.; Thomson, J.M.; Flint, H.J. The role of pH in determining the species composition of the human colonic microbiota. Environ. Microbiol. 2009, 11, 2112–2122. [Google Scholar] [CrossRef] [PubMed]
  11. Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed]
  12. Cummings, J.H.; Macfarlane, G.T. The control and consequences of bacterial fermentation in the human colon. J. Appl. Bacteriol. 1991, 70, 443–459. [Google Scholar] [CrossRef] [PubMed]
  13. Topping, D.L.; Clifton, P.M. Short-chain fatty acids and human colonic function: Roles of resistant starch and nonstarch polysaccharides. Physiol. Rev. 2001, 81, 1031–1064. [Google Scholar] [PubMed]
  14. Donohoe, D.R.; Garge, N.; Zhang, X.; Sun, W.; O’Connell, T.M.; Bunger, M.K.; Bultman, S.J. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell. Metab. 2011, 13, 517–526. [Google Scholar] [CrossRef] [PubMed]
  15. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harries, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–168. [Google Scholar]
  16. Trent, M.S.; Stead, C.M.; Tran, A.X.; Hankins, J.V. Diversity of endotoxin and its impact on pathogenesis. J. Endotoxin Res. 2006, 12, 205–223. [Google Scholar] [CrossRef] [PubMed]
  17. Kamada, N.; Chen, G.; Nunez, G. Harnessing pathogen-commensal relations. Nat. Med. 2012, 18, 1190–1191. [Google Scholar] [CrossRef] [PubMed]
  18. Fukuda, S.; Toh, H.; Hase, K.; Oshima, K.; Nakanishi, Y.; Yoshimura, K.; Tobe, T.; Clarke, J.M.; Topping, D.L.; Suzuki, T.; et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011, 469, 543–547. [Google Scholar]
  19. Cantarel, B.L.; Lombard, V.; Henrissat, B. Complex carbohydrate utilization by the healthy human microbiome. PLoS One 2012, 7, e28742. [Google Scholar] [CrossRef]
  20. Xu, J.; Bjursell, M.K.; Himrod, J.; Deng, S.; Carmichael, L.K.; Chiang, H.C.; Hooper, L.V.; Gordon, J.I. A genomic view of the human-Bacteroides thetaiotaomicron symbiosis. Science 2003, 299, 2074–2076. [Google Scholar] [CrossRef] [PubMed]
  21. Sandberg, A.S.; Andlid, T. Phytogenic and microbial phytases in human nutrition. Int. J. Food Sci. Technol. 2002, 37, 823–833. [Google Scholar] [CrossRef]
  22. Morvan, B.; Bonnemoy, F.; Fonty, G.; Gouet, P. Quantitative determination of H2-utilizing acetogenic and sulfate-reducing bacteria and methanogenic archaea from digestive tract of different mammals. Curr. Microbiol. 1996, 32, 129–133. [Google Scholar] [CrossRef] [PubMed]
  23. Carbonero, F.; Benefiel, A.C.; Alizadeh-Ghamsari, A.H.; Gaskins, H.R. Microbial pathways in colonic sulphur metabolism and links with health and disease. Front. Physiol. 2012, 3, 448. [Google Scholar] [CrossRef]
  24. Chatterjee, S.; Park, S.; Low, K.; Kong, Y.; Pimentel, M. The degree of breath methane production in IBS correlates with the severity of constipation. Am. J. Gastroenterol. 2007, 102, 837–841. [Google Scholar] [CrossRef] [PubMed]
  25. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermudez-Humaran, 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]
  26. Huurre, A.; Kalliomaki, M.; Rautava, S.; Rinne, M.; Salminen, S.; Isolauri, E. Mode of delivery—Effects on gut microbiota and humoral immunity. Neonatology 2008, 93, 236–240. [Google Scholar] [CrossRef] [PubMed]
  27. Kelly, D.; King, T.; Aminov, R. Importance of microbial colonization of the gut in early life to the development of immunity. Mutat. Res. 2007, 622, 58–69. [Google Scholar] [CrossRef] [PubMed]
  28. Harmsen, H.J.; Wildeboer-Veloo, A.C.M.; Raangs, G.C.; Wagendorp, A.A.; Klijn, N.; Bindels, J.G.; Welling, G.W. Analysis of intestinal flora development in breast-fed and formula-fed infants by using molecular identification and detection methods. J. Pediatr. Gastroenterol. Nutr. 2000, 30, 61–67. [Google Scholar] [CrossRef]
  29. Coppa, G.V.; Zampini, L.; Galeazzi, T.; Gabrielli, O. Prebiotics in human milk: A review. Dig. Liver Dis. 2006, 38, S291–S294. [Google Scholar] [CrossRef] [PubMed]
  30. Arslanoglu, S.; Moro, G.E.; Schmitt, J.; Tandoi, L.; Rizzardi, S.; Boehm, G. Early dietary intervention with a mixture of prebiotic oligosaccharides reduces the incidence of allergic manifestations and infections during the first two years of life. J. Nutr. 2008, 138, 1091–1095. [Google Scholar] [PubMed]
  31. Barrett, M.J.; Donoghue, V.; Mooney, E.E.; Slevin, M.; Persaud, T.; Twomey, E.; Ryan, S.; Laffan, E.; Twomey, A. Isolated acute non-cystic white matter injury in term infants presenting with neonatal encephalopathy. Arch. Dis. Child. Fetal Neonatal Ed. 2013, 98, F158–F160. [Google Scholar] [CrossRef] [PubMed]
  32. Sghir, A.; Gramet, G.; Suau, A.; Rochet, V.; Pochart, P.; Dore, J. Quantification of bacterial groups within human fecal flora by oligonucleotide probe hybridization. Appl. Environ. Microbiol. 2000, 66, 2263–2266. [Google Scholar] [CrossRef] [PubMed]
  33. Pokusaeva, K.; Fitzgerald, G.F.; van Sinderen, D. Carbohydrate metabolism in Bifidobacteria. Genes Nutr. 2011, 6, 285–306. [Google Scholar] [CrossRef] [PubMed]
  34. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar]
  35. Lakshminarayanan, B.; Harris, H.M.B.; Coakley, M.; O’Sullivan, O.; Stanton, C.; Pruteanu, M.; Shanahan, F.; O’Toole, P.W.; Ross, R.P.; Consortium, E.; et al. Prevalence and characterization of Clostridium perfringens from the faecal microbiota of elderly Irish subjects. J. Med. Microbiol. 2013, 62, 457–466. [Google Scholar]
  36. Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O’Connor, E.M.; Cusack, S.; Harris, H.M.B.; Coakley, M.; Lakshminarayanan, B.; O’Sulliva, O.; et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012, 488, 178–184. [Google Scholar]
  37. Huxley, R.R.; Ansary-Moghaddam, A.; Clifton, P.; Czernichow, S.; Parr, C.L.; Woodward, M. The impact of dietary and lifestyle risk factors on risk of colorectal cancer: A quantitative overview of the epidemiological evidence. Int. J. Cancer 2009, 125, 171–180. [Google Scholar] [CrossRef]
  38. Benjamin, J.L.; Hedin, C.R.H.; Koutsoumpas, A.; Ng, S.C.; McCarthy, N.E.; Prescott, N.J.; Pessoa-Lopes, P.; Mathew, C.G.; Sanderson, J.; Hart, A.L.; et al. Smokers with active Crohn’s disease have a clinically relevant dysbiosis of the gastrointestinal microbiota. Inflamm. Bowel Dis. 2012, 18, 1092–1100. [Google Scholar]
  39. Beamish, L.A.; Osornio-Vargas, A.R.; Wine, E. Air pollution: An environmental factor contributing to intestinal disease. J. Crohns Colitis 2011, 5, 279–286. [Google Scholar] [CrossRef] [PubMed]
  40. Lutgendorff, F.; Akkermans, L.M.A.; Soderholm, J.D. The role of microbiota and probiotics in stress-induced gastrointestinal damage. Curr. Mol. Med. 2008, 8, 282–298. [Google Scholar] [CrossRef] [PubMed]
  41. Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbe communication in health and disease. Front. Physiol. 2011, 2, 94. [Google Scholar] [CrossRef]
  42. Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef] [PubMed]
  43. Finegold, S.M.; Dowd, S.E.; Gontcharova, V.; Liu, C.; Henley, K.E.; Wolcott, R.D.; Youn, E.; Summanen, P.H.; Granpeesheh, D.; Dixon, D.; et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe 2010, 16, 444–453. [Google Scholar]
  44. Parracho, H.; Bingham, M.O.; Gibson, G.R.; McCartney, A.L. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J. Med. Microbiol. 2005, 54, 987–991. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, L.; Christophersen, C.T.; Sorich, M.J.; Gerber, J.P.; Angley, M.T.; Conlon, M.A. Low relative abundances of the mucolytic bacterium Akkermansia muciniphila and Bifidobacterium spp. in feces of children with autism. Appl. Environ. Microbiol. 2011, 77, 6718–6721. [Google Scholar] [CrossRef]
  46. Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucy, 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]
  47. Ley, R.E.; Backhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef] [PubMed]
  48. 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] [PubMed]
  49. 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] [PubMed]
  50. Delzenne, N.M.; Cani, P.D. Interaction between obesity and the gut microbiota: Relevance in nutrition. Ann. Rev. Nutr. 2011, 31, 15–31. [Google Scholar] [CrossRef]
  51. Lin, H.V.; Frassetto, A.; Kowalik, E.J., Jr.; Nawrocki, A.R.; Lu, M.M.; Kosinski, J.R.; Hubert, J.A.; Szeto, D.; Yao, X.; Forrest, G.; et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One 2012, 7, e35240. [Google Scholar] [CrossRef]
  52. Collado, M.C.; Isolauri, E.; Laitinen, K.; Salminen, S. Distinct composition of gut microbiota during pregnancy in overweight and normal-weight women. Am. J. Clin. Nutr. 2008, 88, 894–899. [Google Scholar] [PubMed]
  53. Kalliomaki, M.; Collado, M.C.; Salminen, S.; Isolauri, E. Early differences in fecal microbiota composition in children may predict overweight. Am. J. Clin. Nutr. 2008, 87, 534–538. [Google Scholar] [PubMed]
  54. Devkota, S.; Wang, Y.; Musch, M.W.; Leone, V.; Fehlner-Peach, H.; Nadimpalli, A.; Antonopoulos, D.A.; Jabri, B.; Chang, E.B. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature 2012, 487, 104–108. [Google Scholar] [PubMed]
  55. 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]
  56. Verdu, E.F.; Riddle, M.S. Chronic gastrointestinal consequences of acute infectious diarrhea: Evolving concepts in epidemiology and pathogenesis. Am. J. Gastroenterol. 2012, 107, 981–989. [Google Scholar] [CrossRef] [PubMed]
  57. Voigt, R.M.; Forsyth, C.B.; Green, S.J.; Mutlu, E.; Engen, P.; Vitaterna, M.H.; Turek, F.W.; Keshavarzian, A. Circadian disorganization alters intestinal microbiota. PLoS One 2014, 9, e97500. [Google Scholar] [CrossRef]
  58. Hill, M.J. Bacterial fermentation of complex carbohydrate in the human colon. Eur. J. Cancer Prev. 1995, 4, 353–358. [Google Scholar] [CrossRef] [PubMed]
  59. Payne, A.N.; Chassard, C.; Lacroix, C. Gut microbial adaptation to dietary consumption of fructose, artificial sweeteners and sugar alcohols: Implications for host-microbe interactions contributing to obesity. Obes. Rev. 2012, 13, 799–809. [Google Scholar] [CrossRef] [PubMed]
  60. Touvier, M.; Druesne-Pecollo, N.; Kesse-Guyot, E.; Andreeva, V.A.; Fezeu, L.; Galan, P.; Hercberg, S.; Latino-Martel, P. Dual association between polyphenol intake and breast cancer risk according to alcohol consumption level: A prospective cohort study. Breast Cancer Res. Treat. 2013, 137, 225–236. [Google Scholar] [CrossRef] [PubMed]
  61. Tuohy, K.M.; Conterno, L.; Gasperotti, M.; Viola, R. Up-regulating the human intestinal microbiome using whole plant foods, polyphenols, and/or fiber. J. Agric. Food Chem. 2012, 60, 8776–8782. [Google Scholar] [CrossRef] [PubMed]
  62. Lee, H.C.; Jenner, A.M.; Low, C.S.; Lee, Y.K. Effect of tea phenolics and their aromatic fecal bacterial metabolites on intestinal microbiota. Res. Microbiol. 2006, 157, 876–884. [Google Scholar] [CrossRef] [PubMed]
  63. Tzounis, X.; Rodriguez-Mateos, A.; Vulevic, J.; Gibson, G.R.; Kwik-Uribe, C.; Spencer, J.P.E. Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study. Am. J. Clin. Nutr. 2011, 93, 62–72. [Google Scholar] [CrossRef] [PubMed]
  64. Martin, F.-P.J.; Montoliu, I.; Nagy, K.; Moco, S.; Collino, S.; Guy, P.; Redeuil, K.; Scherer, M.; Rezzi, S.; Kochhar, S.; et al. Specific dietary preferences are linked to differing gut microbial metabolic activity in response to dark chocolate intake. J. Proteome Res. 2012, 11, 6252–6263. [Google Scholar]
  65. Gill, S.R.; Pop, M.; DeBoy, R.T.; Eckburg, P.B.; Turnbaugh, P.J.; Samuel, B.S.; Gordon, J.I.; Relman, D.A.; Fraser-Liggett, C.M.; Nelson, K.E.; et al. Metagenomic analysis of the human distal gut microbiome. Science 2006, 312, 1355–1359. [Google Scholar]
  66. Baghurst, P.A.; Baghurst, K.I.; Record, S.J. Dietary fibre, non-starch polysaccharides and resistant starch—A review. Food Aust. 1996, 48, S3–S35. [Google Scholar]
  67. Murphy, N.; Norat, T.; Ferrari, P.; Jenab, M.; Bueno-de-Mesquita, B.; Skeie, G.; Dahm, C.C.; Overvad, K.; Olsen, A.; Tjønneland, A.; et al. Dietary fibre intake and risks of cancers of the colon and rectum in the European Prospective Investigation into Cancer and Nutrition (EPIC). PLoS One 2012, 7, e39361. [Google Scholar] [CrossRef]
  68. Aune, D.; Chan, D.S.M.; Lau, R.; Vieira, R.; Greenwood, D.C.; Kampman, E.; Norat, T. Dietary fibre, whole grains, and risk of colorectal cancer: Systematic review and dose-response meta-analysis of prospective studies. BMJ 2011, 343, d6617. [Google Scholar] [CrossRef]
  69. Bodinham, C.L.; Smith, L.; Wright, J.; Frost, G.S.; Robertson, M.D. Dietary fibre improves first-phase insulin secretion in overweight individuals. PloS One 2012, 7, e40834. [Google Scholar] [CrossRef]
  70. Hauner, H.; Bechthold, A.; Boeing, H.; Broenstrup, A.; Buyken, A.; Leschik-Bonnet, E.; Linseisen, J.; Schulze, M.; Strohm, D.; Wolfram, G.; et al. Evidence-based guideline of the German Nutrition Society: Carbohydrate Intake and prevention of nutrition-related diseases. Ann. Nutr. MeTab. 2012, 60, 1–58. [Google Scholar]
  71. Sleeth, M.; Psichas, A.; Frost, G. Weight gain and insulin sensitivity: A role for the glycaemic index and dietary fibre? Br. J. Nutr. 2013, 109, 1539–1541. [Google Scholar] [CrossRef] [PubMed]
  72. Windey, K.; de Preter, V.; Verbeke, K. Relevance of protein fermentation to gut health. Mol. Nutr. Food Res. 2012, 56, 184–196. [Google Scholar] [CrossRef] [PubMed]
  73. Mitchell, B.L.; Lawson, M.J.; Davies, M.; Grant, A.K.; Roediger, W.E.W.; Illman, R.J.; Topping, D.L. Volatile fatty-acids in the human intestine—Studies in surgical patients. Nutr. Res. 1985, 5, 1089–1092. [Google Scholar] [CrossRef]
  74. Spiller, G.A.; Chernoff, M.C.; Hill, R.A.; Gates, J.E.; Nassar, J.J.; Shipley, E.A. Effect of purified cellulose, pectin, and a low-residue diet on fecal volatile fatty-acids, transit-time, and fecal weight in humans. Am. J. Clin. Nutr. 1980, 33, 754–759. [Google Scholar] [PubMed]
  75. Roediger, W.E.W. Role of anaerobic-bacteria in the metabolic welfare of the colonic mucosa in man. Gut 1980, 21, 793–798. [Google Scholar] [CrossRef] [PubMed]
  76. Fung, K.Y.C.; Cosgrove, L.; Lockett, T.; Head, R.; Topping, D.L. A review of the potential mechanisms for the lowering of colorectal oncogenesis by butyrate. Br. J. Nutr. 2012, 108, 820–831. [Google Scholar] [CrossRef] [PubMed]
  77. Binder, H.J. Role of colonic short-chain fatty acid transport in diarrhea. Ann. Rev. Physiol. 2010, 72, 297–313. [Google Scholar] [CrossRef]
  78. Wycherley, T.P.; Noakes, M.; Clifton, P.M.; Cleanthous, X.; Keogh, J.B.; Brinkworth, G.D. A high-protein diet with resistance exercise training improves weight loss and body composition in overweight and obese patients with type 2 diabetes. Diabetes Care 2010, 33, 969–976. [Google Scholar] [CrossRef] [PubMed]
  79. Chao, A.; Thun, M.J.; Connell, C.J.; McCullough, M.L.; Jacobs, E.J.; Flanders, W.D.; Rodriguez, C.; Sinha, R.; Calle, E.E. Meat consumption and risk of colorectal cancer. JAMA 2005, 97, 906–916. [Google Scholar]
  80. Norat, T.; Bingham, S.; Ferrari, P.; Slimani, N.; Jenab, M.; Mazuir, M.; Overvad, K.; Olsen, A.; Tjonneland, A.; Clavel, F.; et al. Meat, fish, and colorectal cancer risk: The European Prospective Investigation into Cancer and Nutrition. J. Natl. Cancer Inst. 2005, 97, 906–916. [Google Scholar]
  81. World Cancer Research Fund. Food, Nutrition, Physical Activity, and the Prevention of Colon Cancer: A Global Perspective; American Institute for Cancer Research: Washington, DC, USA, 2007. [Google Scholar]
  82. World Cancer Research Fund. Food, Nutrition, Physical Activity, and the Prevention of Colorectal Cancer; Continuous Update Project Report; American Institute for Cancer Research: Washington, DC, USA, 2011. [Google Scholar]
  83. Alexander, D.D.; Cushing, C.A. Red meat and colorectal cancer: A critical summary of prospective epidemiological studies. Obes. Rev. 2011, 12, e472–e493. [Google Scholar] [CrossRef] [PubMed]
  84. Oostindjer, M.; Alexander, J.; Vang, G.; Andersen, G.; Bryan, N.S.; Chen, D.; Corpet, D.E.; de Smet, S.; Dragsted, L.O.; Haug, A.; et al. The role of red and processed meat in colorectal cancer development: A perspective. Meat Sci. 2014, 97, 583–596. [Google Scholar]
  85. Silvester, K.R.; Cummings, J.H. Does digestibility of meat protein help explain large-bowel cancer risk. Nutr. Cancer 1995, 24, 279–288. [Google Scholar] [CrossRef] [PubMed]
  86. Macfarlane, G.T.; Macfarlane, S. Bacteria, colonic fermentation, and gastrointestinal health. J. AOAC Int. 2012, 95, 50–60. [Google Scholar] [CrossRef] [PubMed]
  87. Hughes, R.; Magee, E.A.; Bingham, S. Protein degradation in the large intestine: Relevance to colorectal cancer. Curr. Issues Intest. Microbiol. 2000, 1, 51–58. [Google Scholar] [PubMed]
  88. Toden, S.; Bird, A.R.; Topping, D.L.; Conlon, M.A. Resistant starch attenuates colonic DNA damage induced by higher dietary protein in rats. Nutr. Cancer 2005, 51, 45–51. [Google Scholar] [CrossRef] [PubMed]
  89. Toden, S.; Bird, A.R.; Topping, D.L.; Conlon, M.A. Differential effects of dietary whey, casein and soya on colonic DNA damage and large bowel SCFA in rats fed diets low and high in resistant starch. Br. J. Nutr. 2007, 97, 535–543. [Google Scholar] [CrossRef] [PubMed]
  90. Toden, S.; Bird, A.R.; Topping, D.L.; Conlon, M.A. Dose-dependent reduction of dietary protein-induced colonocyte DNA damage by resistant starch in rats correlates more highly with caecal butyrate than with other short chain fatty acids. Cancer Biol. Ther. 2007, 6, 253–258. [Google Scholar] [CrossRef] [PubMed]
  91. Toden, S.; Bird, A.R.; Topping, D.L.; Conlon, M.A. High red meat diets induce greater numbers of colonic DNA double-strand breaks than white meat in rats: Attenuation by high-amylose maize starch. Carcinogenesis 2007, 28, 2355–2362. [Google Scholar] [CrossRef] [PubMed]
  92. Russell, W.R.; Gratz, S.W.; Duncan, S.H.; Holtrop, G.; Ince, J.; Scobbie, L.; Duncan, G.; Johnstone, A.M.; Lobley, G.E.; Wallace, R.J.; et al. High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health. Am. J. Clin. Nutr. 2011, 93, 1062–1072. [Google Scholar]
  93. Shaughnessy, D.T.; Gangarosa, L.M.; Schliebe, B.; Umbach, D.M.; Xu, Z.; MacIntosh, B.; Knize, M.G.; Matthews, P.P.; Swank, A.E.; Sandler, R.S.; et al. Inhibition of fried meat-induced colorectal DNA damage and altered systemic genotoxicity in humans by crucifera, chlorophyllin, and yogurt. PLoS One 2011, 6, e18707. [Google Scholar] [CrossRef]
  94. Humphreys, K.J.; Conlon, M.A.; Young, G.P.; Topping, D.L.; Hu, Y.; Winter, J.M.; Bird, A.R.; Cobiac, L.; Kennedy, N.A.; Michael, M.A.; et al. Dietary manipulation of oncogenic microRNA expression in human rectal mucosa: A randomized trial. Cancer Prev. Res. 2014, 7, 786–795. [Google Scholar]
  95. Brinkworth, G.D.; Noakes, M.; Clifton, P.M.; Bird, A.R. Comparative effects of very low-carbohydrate, high-fat and high-carbohydrate, low-fat weight-loss diets on bowel habit and faecal short-chain fatty acids and bacterial populations. Br. J. Nutr. 2009, 101, 1493–1502. [Google Scholar] [CrossRef] [PubMed]
  96. Windey, K.; de Preter, V.; Iouat, T.; Schuit, F.; Herman, J.; Vansant, G.; Verbeke, K. Modulation of protein fermentation does not affect fecal water toxicity: A randomized cross-over study in healthy subjects. PLoS One 2012, 7, e52387. [Google Scholar] [CrossRef]
  97. Lin, H.C.; Visek, W.J. Colon mucosal cell damage by ammonia in rats. J. Nutr. 1991, 121, 887–893. [Google Scholar] [PubMed]
  98. Kramer, H. Dietary patterns, calories, and kidney disease. Adv. Chronic Kidney Dis. 2013, 20, 135–140. [Google Scholar] [CrossRef] [PubMed]
  99. Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar]
  100. Moreira, A.P.B.; Texeira, T.F.S.; Ferreira, A.B.; Peluzio Mdo, C.; Alfenas Rde, C. Influence of a high-fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. Br. J. Nutr. 2012, 108, 801–809. [Google Scholar] [CrossRef] [PubMed]
  101. Ou, J.; de Lany, J.P.; Zhang, M.; Sharma, S.; O’Keefe, S.J.D. Association between low colonic short-chain fatty acids and high bile acids in high colon cancer risk populations. Nutr. Cancer 2012, 64, 34–40. [Google Scholar] [CrossRef] [PubMed]
  102. Ridlon, J.M.; Kang, D.-J.; Hylemon, P.B. Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 2006, 47, 241–259. [Google Scholar] [CrossRef] [PubMed]
  103. Soto-Vaca, A.; Gutierrez, A.; Losso, J.N.; Xu, Z.; Finley, J.W. Evolution of phenolic compounds from color and flavor problems to health benefits. J. Agric. Food Chem. 2012, 60, 6658–6677. [Google Scholar] [CrossRef] [PubMed]
  104. 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]
  105. Selma, M.V.; Espin, J.C.; Tomas-Barberan, F.A. Interaction between phenolics and gut microbiota: Role in human health. J. Agric. Food Chem. 2009, 57, 6485–6501. [Google Scholar] [CrossRef] [PubMed]
  106. Forester, S.C.; Waterhouse, A.L. Metabolites are key to understanding health effects of wine polyphenolics. J. Nutr. 2009, 139, 1824S–1831S. [Google Scholar] [CrossRef] [PubMed]
  107. Grün, C.H.; van Dorsten, F.A.; Jacobs, D.M.; le Belleguic, M.; van Velzen, E.J.J.; Bingham, M.O.; Janssen, H.-G.; van Duynhoven, J.P.M. GC-MS methods for metabolic profiling of microbial fermentation products of dietary polyphenols in human and in vitro intervention studies. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2008, 871, 212–219. [Google Scholar] [CrossRef] [PubMed]
  108. Lee, C.Y. Challenges in providing credible scientific evidence of health benefits of dietary polyphenols. J. Funct. Foods 2013, 5, 524–526. [Google Scholar] [CrossRef]
  109. Gross, G.; Jacobs, D.M.; Peters, S.; Possemiers, S.; van Duynhoven, J.; Vaughan, E.E.; van de Wiele, T. In vitro bioconversion of polyphenols from black tea and red wine/grape juice by human intestinal microbiota displays strong interindividual variability. J. Agric. Food Chem. 2010, 58, 10236–10246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Van Nuenen, M.; Venema, K.; van der Woude, J.C.J.; Kuipers, E.J. The metabolic activity of fecal microbiota from healthy individuals and patients with inflammatory bowel disease. Dig. Dis. Sci. 2004, 49, 485–491. [Google Scholar] [CrossRef] [PubMed]
  111. Cordain, L.; Eaton, S.B.; Sebastian, A.; Mann, N.; Lindeberg, S.; Watkins, B.A.; O’Keefe, J.H.; Brand-Miller, J. Origins and evolution of the Western diet: Health implications for the 21st century. Am. J. Clin. Nutr. 2005, 81, 341–354. [Google Scholar] [PubMed]
  112. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [Green Version]
  113. Jeffery, I.B.; Claesson, M.J.; O’Toole, P.W.; Shanahan, F. Categorization of the gut microbiota: Enterotypes or gradients? Nat. Rev. Microbiol. 2012, 10, 591–592. [Google Scholar] [CrossRef] [PubMed]
  114. 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]
  115. Lin, A.; Bik, E.M.; Costello, E.K.; Dethlefsen, L.; Haque, R.; Relman, D.A.; Singh, U. Distinct distal gut microbiome diversity and composition in healthy children from Bangladesh and the United States. PLoS One 2013, 8, e53838. [Google Scholar] [CrossRef]
  116. Purushe, J.; Fouts, D.E.; Morrison, M.; White, B.A.; Mackie, R.I.; North American Consortium for Rumen Bacteria; Coutinho, P.M.; Henrissat, B.; Nelson, K.E. Comparative genome analysis of Prevotella ruminicola and Prevotella bryantii: Insights into their environmental niche. Microb. Ecol. 2010, 60, 721–729. [Google Scholar] [CrossRef] [PubMed]
  117. Liszt, K.; Zwielehner, J.; Handschur, M.; Hippe, B.; Thaler, R.; Haslberger, A.G. Characterization of bacteria, clostridia and Bacteroides in faeces of vegetarians using qPCR and PCR-DGGE fingerprinting. Ann. Nutr. Metab. 2009, 54, 253–257. [Google Scholar] [CrossRef] [PubMed]
  118. Kabeerdoss, J.; Devi, R.S.; Mary, R.R.; Ramakrishna, B.S. Faecal microbiota composition in vegetarians: Comparison with omnivores in a cohort of young women in southern India. Br. J. Nutr. 2012, 108, 953–957. [Google Scholar] [CrossRef] [PubMed]
  119. Frank, D.N.; Amand, A.L.S.; Feldman, R.A.; Boedeker, E.C.; Harpaz, N.; Pace, N.R. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. USA 2007, 104, 13780–13785. [Google Scholar] [CrossRef] [PubMed]
  120. Gore, C.; Munro, K.; Lay, C.; Bibiloni, R.; Morris, J.; Woodcock, A.; Custovic, A.; Tannock, G.W. Bifidobacterium pseudocatenulatum is associated with atopic eczema: A nested case-control study investigating the fecal microbiota of infants. J. Allergy Clin. Immunol. 2008, 121, 135–140. [Google Scholar] [CrossRef] [PubMed]
  121. Schwiertz, A.; Taras, D.; Schaefer, K.; Beijer, S.; Bos, N.A.; Donus, C.; Hardt, P.D. Microbiota and SCFA in lean and overweight healthy subjects. Obesity 2010, 18, 190–195. [Google Scholar] [CrossRef] [PubMed]
  122. 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] [PubMed]
  123. Nava, G.M.; Carbonero, F.; Ou, J.; Benefiel, A.C.; O’Keefe, S.J.; Gaskins, H.R. Hydrogenotrophic microbiota distinguish native Africans from African and European Americans. Environ. Microbiol. Rep. 2012, 4, 307–315. [Google Scholar] [CrossRef] [PubMed]
  124. Magee, E.A.; Richardson, C.J.; Hughes, R.; Cummings, J.H. Contribution of dietary protein to sulfide production in the large intestine: An in vitro and a controlled feeding study in humans. Am. J. Clin. Nutr. 2000, 72, 1488–1494. [Google Scholar] [PubMed]
  125. O’Keefe, S.J.D.; Kidd, M.; Espitalier-Noel, G.; Owira, P. Rarity of colon cancer in Africans is associated with low animal product consumption, not fiber. Am. J. Gastroenterol. 1999, 94, 1373–1380. [Google Scholar] [CrossRef] [PubMed]
  126. Jumpertz, R.; Duc Son, L.; Turnbaugh, P.J.; Trinidad, C.; Bogardus, C.; Gordon, J.I.; Krakoff, J. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am. J. Clin. Nutr. 2011, 94, 58–65. [Google Scholar] [CrossRef] [PubMed]
  127. Hildebrandt, M.A.; Hoffmann, C.; Sherrill-Mix, S.A.; Keilbaugh, S.A.; Hamady, M.; Chen, Y.-Y.; Knight, R.; Ahima, R.S.; Bushman, F.; Wu, G.D.; et al. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology 2009, 137, 1716–1724. [Google Scholar]
  128. Cani, P.D.; Neyrinck, A.M.; Fava, F.; Knauf, C.; Burcelin, R.G.; Tuohy, K.M.; Gibson, G.R.; Delzenne, N.M. Selective increases of Bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 2007, 50, 2374–2383. [Google Scholar] [CrossRef] [PubMed]
  129. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar]
  130. Neyrinck, A.M.; Possemiers, S.; Verstraete, W.; de Backer, F.; Cani, P.D.; Delzenne, N.M. Dietary modulation of clostridial cluster XIVa gut bacteria (Roseburia spp.) by chitin-glucan fiber improves host metabolic alterations induced by high-fat diet in mice. J. Nutr. Biochem. 2012, 23, 51–59. [Google Scholar]
  131. Deplancke, B.; Gaskins, H.R. Microbial modulation of innate defense: Goblet cells and the intestinal mucus layer. Am. J. Clin. Nutr. 2001, 73, 1131S–1141S. [Google Scholar] [PubMed]
  132. Kim, Y.S.; Ho, S.B. Intestinal goblet cells and mucins in health and disease: Recent insights and progress. Curr. Gastroenterol. Rep. 2010, 12, 319–330. [Google Scholar] [CrossRef] [PubMed]
  133. Hedemann, M.S.; Theil, P.K.; Knudsen, K.E.B. The thickness of the intestinal mucous layer in the colon of rats fed various sources of non-digestible carbohydrates is positively correlated with the pool of SCFA but negatively correlated with the proportion of butyric acid in digesta. Br. J. Nutr. 2009, 102, 117–125. [Google Scholar] [CrossRef] [PubMed]
  134. Femia, A.P.; Giannini, A.; Fazi, M.; Tarquini, E.; Salvadori, M.; Roncucci, L.; Tonelli, F.; Dolara, P.; Caderni, G. Identification of mucin depleted foci in the human colon. Cancer Prev. Res. 2008, 1, 562–567. [Google Scholar] [CrossRef]
  135. Femia, A.P.; Swidsinski, A.; Dolara, P.; Salvadori, M.; Amedei, A.; Caderni, G. Mucin depleted foci, colonic preneoplastic lesions lacking Muc2, show up-regulation of Tlr2 but not bacterial infiltration. PLoS One 2012, 7, e29918. [Google Scholar] [CrossRef]
  136. Johansson, M.E.V.; Phillipson, M.; Petersson, J.; Velcich, A.; Holm, L.; Hansson, G.C. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc. Natl. Acad. Sci. USA 2008, 105, 15064–15069. [Google Scholar] [CrossRef] [PubMed]
  137. Png, C.W.; Linden, S.K.; Gilshenan, K.S.; Zoetendal, E.G.; McSweeney, C.S.; Sly, L.I.; McGuckin, M.A.; Florin, T.H.J. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am. J. Gastroenterol. 2010, 105, 2420–2428. [Google Scholar] [CrossRef] [PubMed]
  138. Collado, M.C.; Derrien, M.; Isolauri, E.; de Vos, W.M.; Salminen, S. Intestinal integrity and Akkermansia muciniphila, a mucin-degrading member of the intestinal microbiota present in infants, adults, and the elderly. Appl. Environ. Microbiol. 2007, 73, 7767–7770. [Google Scholar] [CrossRef] [PubMed]
  139. Kerr, C.A.; Grice, D.M.; Tran, C.D.; Bauer, D.C.; Li, D.; Hendry, P.; Hannan, G.N. Early life events influence whole-of-life metabolic health via gut microflora and gut permeability. Crit. Rev. Microbiol. 2014, in press. [Google Scholar]
  140. Hooper, L.V.; Littman, D.R.; Macpherson, A.J. Interactions between the microbiota and the immune system. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef] [PubMed]
  141. Lebouvier, T.; Chaumette, T.; Paillusson, S.; Duyckaerts, C.; des Varannes, S.B.; Neunlist, M.; Derkinderen, P. The second brain and Parkinson’s disease. Eur. J. Neurosci. 2009, 30, 735–741. [Google Scholar] [CrossRef] [PubMed]
  142. Awad, R.A. Neurogenic bowel dysfunction in patients with spinal cord injury, myelomeningocele, multiple sclerosis and Parkinson’s disease. World J. Gastroenterol. 2011, 17, 5035–5048. [Google Scholar] [CrossRef] [PubMed]
  143. Forsyth, C.B.; Shannon, K.M.; Kordower, J.H.; Voigt, R.M.; Shaikh, M.; Jaglin, J.A.; Estes, J.D.; Dodiya, H.B.; Keshavarzian, A. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s Disease. PLoS One 2011, 6, e28032. [Google Scholar] [CrossRef]
  144. Braak, H.; Rub, U.; Gai, W.P.; del Tredici, K. Idiopathic Parkinson’s disease: Possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J. Neural Transm. 2003, 110, 517–536. [Google Scholar] [CrossRef] [PubMed]
  145. Esteve, E.; Ricart, W.; Fernandez-Real, J.-M. Gut microbiota interactions with obesity, insulin resistance and type 2 diabetes: Did gut microbiote co-evolve with insulin resistance? Curr. Opin. Clin. Nutr. Metab. Care 2011, 14, 483–490. [Google Scholar] [CrossRef] [PubMed]
  146. Frazier, T.H.; DiBaise, J.K.; McClain, C.J. Gut microbiota, intestinal permeability, obesity-induced inflammation, and liver injury. J. Parenter. Enter. Nutr. 2011, 35, 14S–20S. [Google Scholar] [CrossRef]
  147. Cani, P.D.; Osto, M.; Geurts, L.; Everard, A. Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes 2012, 3, 279–288. [Google Scholar] [CrossRef] [PubMed]
  148. Anders, H.-J.; Andersen, K.; Stecher, B. The intestinal microbiota, a leaky gut, and abnormal immunity in kidney disease. Kidney Int. 2013, 83, 1010–1016. [Google Scholar] [CrossRef] [PubMed]
  149. Piya, M.K.; Harte, A.L.; McTernan, P.G. Metabolic endotoxaemia: Is it more than just a gut feeling? Curr. Opin. Lipidol. 2013, 24, 78–85. [Google Scholar] [CrossRef] [PubMed]
  150. 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]
  151. McOrist, A.L.; Miller, R.B.; Bird, A.R.; Keogh, J.B.; Noakes, M.; Topping, D.L.; Conlon, M.A. Fecal butyrate levels vary widely among individuals but are usually increased by a diet high in resistant starch. J. Nutr. 2011, 141, 883–889. [Google Scholar] [CrossRef] [PubMed]
  152. Abell, G.C.J.; Cooke, C.M.; Bennett, C.N.; Conlon, M.A.; McOrist, A.L. Phylotypes related to Ruminococcus bromii are abundant in the large bowel of humans and increase in response to a diet high in resistant starch. FEMS Microbiol. Ecol. 2008, 66, 505–515. [Google Scholar] [CrossRef] [PubMed]
  153. Walker, A.W.; Ince, J.; Duncan, S.H.; Webster, L.M.; Holtrop, G.; Ze, X.; Brown, D.; Stares, M.D.; Scott, P.; Bergerat, A.; et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2011, 5, 220–230. [Google Scholar]
  154. Gibson, G.R.; Roberfroid, M.B. Dietary modulation of the human colonic microbiota—Introducing the concept of prebiotics. J. Nutr. 1995, 125, 1401–1412. [Google Scholar] [PubMed]
  155. Gibson, G.R.; Scott, K.P.; Rastall, R.A.; Tuohy, K.M.; Hotchkiss, A.; Dubert-Ferrandon, A.; Gareau, M.; Murphy, E.F.; Saulnier, D.; Loh, G.; et al. Dietary prebiotics: Current status and new definition. Food Sci. Technol. Bull. Funct. Foods 2010, 7, 1–19. [Google Scholar] [CrossRef]
  156. Bird, A.R.; Topping, D.L. Resistant starch as a prebiotic. In Therapeutic Microbiology: Probiotics and Related Strategies; Versalovic, J., Wilson, M., Eds.; ASM Press: Washington, DC, USA, 2008; pp. 159–173. [Google Scholar]
  157. Clark, M.J.; Robien, K.; Slavin, J.L. Effect of prebiotics on biomarkers of colorectal cancer in humans: A systematic review. Nutr. Rev. 2012, 70, 436–443. [Google Scholar] [CrossRef] [PubMed]
  158. Roberfroid, M.; Gibson, G.R.; Hoyles, L.; McCartney, A.L.; Rastall, R.; Rowland, I.; Wolvers, D.; Watzl, B.; Szajewska, H.; Stahl, B.; et al. Prebiotic effects: Metabolic and health benefits. Br. J. Nutr. 2010, 104, S1–S63. [Google Scholar]
  159. Brownawell, A.M.; Caers, W.; Gibson, G.R.; Kendall, C.W.C.; Lewis, K.D.; Ringel, Y.; Slavin, J.L. Prebiotics and the health benefits of fiber: Current regulatory status, future research, and goals. J. Nutr. 2012, 142, 962–974. [Google Scholar] [CrossRef] [PubMed]
  160. Saad, N.; Delattre, C.; Urdaci, M.; Schmitter, J.M.; Bressollier, P. An overview of the last advances in probiotic and prebiotic field. LWT Food Sci. Technol. 2013, 50, 1–16. [Google Scholar] [CrossRef]
  161. Delzenne, N.M.; Neyrinck, A.M.; Backhed, F.; Cani, P.D. Targeting gut microbiota in obesity: Effects of prebiotics and probiotics. Nat. Rev. Endocrinol. 2011, 7, 639–646. [Google Scholar] [CrossRef] [PubMed]
  162. Van Loo, J.; Coussement, P.; de Leenheer, L.; Hoebregs, H.; Smits, G. On the presence of inulin and oligofructose as natural ingredients in the western diet. Crit. Rev. Food Sci. Nutr. 1995, 35, 525–552. [Google Scholar] [CrossRef] [PubMed]
  163. Bird, A.R.; Conlon, M.A.; Christophersen, C.T.; Topping, D.L. Resistant starch, large bowel fermentation and a broader perspective of prebiotics and probiotics. Benef. Microbes 2010, 1, 423–431. [Google Scholar] [CrossRef] [PubMed]
  164. Conlon, M.A.; Bird, A.R.; Regina, A.; Morell, M.K.; Lockett, T.; Kang, S.; Molloy, P.; Kerr, C.A.; Shaw, J.; McSweeney, C.; et al. Resistant starches protect against colonic DNA damage and alter microbiota and gene expression in rats fed a western diet. J. Nutr. 2012, 142, 832–840. [Google Scholar]
  165. Roberfroid, M. Prebiotics: The concept revisited. J. Nutr. 2007, 137, 830S–837S. [Google Scholar] [PubMed]
  166. Bird, A.R.; Lopez-Rubio, A.; Shrestha, A.K.; Gidley, M.J. Resistant starch in vitro and in vivo: Factors determining yield, structure, and physiological relevance. In Modern Biopolymer Science: Bridging the Divide between Fundamental Treatise and Industrial Application; Kasapsis, S., Norton, I.T., Ubbink, J.B., Eds.; Academic Press: Burlington, MA, USA, 2009; pp. 449–510. [Google Scholar]
  167. Tremaroli, V.; Backhed, F. Functional interactions between the gut microbiota and host metabolism. Nature 2012, 489, 242–249. [Google Scholar] [CrossRef] [PubMed]
  168. Crittenden, R.; Bird, A.R.; Gopal, P.; Henriksson, A.; Lee, Y.K.; Payne, M.J. Probiotic research in Australia, New Zealand and the Asia-Pacific region. Curr. Pharm. Des. 2005, 11, 37–53. [Google Scholar] [CrossRef] [PubMed]
  169. Floch, M.H.; Walker, W.A.; Madsen, K.; Sanders, M.E.; Macfarlane, G.T.; Flint, H.J.; Dieleman, L.A.; Ringel, Y.; Guandalini, S.; Kelley, C.P.; et al. Recommendations for probiotic use—2011 update. J. Clin. Gastroenterol. 2011, 45, S168–S171. [Google Scholar]
  170. Khani, S.; Hosseini, H.M.; Taheri, M.; Nourani, M.R.; Imani Fooladi, A.A. Probiotics as an alternative strategy for prevention and treatment of human diseases: A review. Inflamm. Allergy Drug Targets 2012, 11, 79–89. [Google Scholar] [CrossRef] [PubMed]
  171. Mugambi, M.N.; Musekiwa, A.; Lombard, M.; Young, T.; Blaauw, R. Probiotics, prebiotics infant formula use in preterm or low birth weight infants: A systematic review. Nutr. J. 2012, 11, 58. [Google Scholar] [CrossRef]
  172. Johnston, B.C.; Ma, S.S.Y.; Goldenberg, J.Z.; Thorlund, K.; Vandvik, P.O.; Loeb, M.; Guyatt, G.H. Probiotics for the prevention of Clostridium difficile-associated diarrhea: A systematic review and meta-analysis. Ann. Intern. Med. 2012, 157, 878–888. [Google Scholar] [CrossRef] [PubMed]
  173. Hosseini, A.; Nikfar, S.; Abdollahi, M. Probiotics use to treat irritable bowel syndrome. Expert Opin. Biol. Ther. 2012, 12, 1323–1334. [Google Scholar] [CrossRef] [PubMed]
  174. Augustin, M.A.; Sanguansri, L.; Lockett, T. Nano- and micro-encapsulated systems for enhancing the delivery of resveratrol. Ann. N. Y. Acad. Sci. 2013, 1290, 107–112. [Google Scholar] [CrossRef] [PubMed]
  175. Cook, M.T.; Tzortzis, G.; Charalampopoulos, D.; Khutoryanskiy, V.V. Microencapsulation of probiotics for gastrointestinal delivery. J. Control. Release 2012, 162, 56–67. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Conlon, M.A.; Bird, A.R. The Impact of Diet and Lifestyle on Gut Microbiota and Human Health. Nutrients 2015, 7, 17-44. https://doi.org/10.3390/nu7010017

AMA Style

Conlon MA, Bird AR. The Impact of Diet and Lifestyle on Gut Microbiota and Human Health. Nutrients. 2015; 7(1):17-44. https://doi.org/10.3390/nu7010017

Chicago/Turabian Style

Conlon, Michael A., and Anthony R. Bird. 2015. "The Impact of Diet and Lifestyle on Gut Microbiota and Human Health" Nutrients 7, no. 1: 17-44. https://doi.org/10.3390/nu7010017

Article Metrics

Back to TopTop