Hostname: page-component-8448b6f56d-xtgtn Total loading time: 0 Render date: 2024-04-23T06:38:00.000Z Has data issue: false hasContentIssue false

Human gut microbiota: does diet matter?

Published online by Cambridge University Press:  26 August 2014

Johanna Maukonen*
Affiliation:
VTT Technical Research Centre of Finland, P.O. Box 1000, FI-02044 VTT, Finland
Maria Saarela
Affiliation:
VTT Technical Research Centre of Finland, P.O. Box 1000, FI-02044 VTT, Finland
*
*Corresponding author: J. Maukonen, fax +358 20 722 7071, email johanna.maukonen@vtt.fi
Rights & Permissions [Opens in a new window]

Abstract

The human oro-gastrointestinal (GI) tract is a complex system, consisting of oral cavity, pharynx, oesophagus, stomach, small intestine, large intestine, rectum and anus, which all together with the accessory digestive organs constitute the digestive system. The function of the digestive system is to break down dietary constituents into small molecules and then absorb these for subsequent distribution throughout the body. Besides digestion and carbohydrate metabolism, the indigenous microbiota has an important influence on host physiological, nutritional and immunological processes, and commensal bacteria are able to modulate the expression of host genes that regulate diverse and fundamental physiological functions. The main external factors that can affect the composition of the microbial community in generally healthy adults include major dietary changes and antibiotic therapy. Changes in some selected bacterial groups have been observed due to controlled changes to the normal diet e.g. high-protein diet, high-fat diet, prebiotics, probiotics and polyphenols. More specifically, changes in the type and quantity of non-digestible carbohydrates in the human diet influence both the metabolic products formed in the lower regions of the GI tract and the bacterial populations detected in faeces. The interactions between dietary factors, gut microbiota and host metabolism are increasingly demonstrated to be important for maintaining homeostasis and health. Therefore the aim of this review is to summarise the effect of diet, and especially dietary interventions, on the human gut microbiota. Furthermore, the most important confounding factors (methodologies used and intrinsic human factors) in relation to gut microbiota analyses are elucidated.

Type
Conference on ‘Diet, gut microbiology and human health’
Copyright
Copyright © The Authors 2014 

In the past 10 years, there has been a wealth of studies in which the relationship between the human gut microbiota and human health has been investigated. Moreover, recently there have been several human health-related microbiota studies with partly contradictory results regarding e.g. obesity-related microbiota and abundance of bifidobacteria in the faecal microbiota of babies. As it is likely that at least some of the differences may be explained by the methodology applied, it is of utmost importance that when reading articles related to human gut microbiota studies the most important confounding factors are known.

Human gut microbiota

In an adult human individual, resident bacteria outnumber human cells by a factor of ten; each adult harbours on average 1013 mammalian cells and 1014 microbial cells( Reference Savage 1 ). Most of the microbes, typically 1011–1012 microbes/g, can be found in faeces and from the large intestine( Reference Finegold, Sutter, Mathisen and Hentges 2 Reference Harmsen, Raangs and He 4 ), which is considered to be a complex fermentor with a metabolic potential to rival that of the liver( Reference Marchesi and Shanahan 5 ). The environmental determinants, namely temperature, pH, redox potential, atmospheric composition, water activity, salinity and light, within each region of the human oro-gastrointestinal (GI) tract are very different, and therefore each region has its own distinctive microbiota( Reference Wilson 6 ). Since digestive enzymes are not secreted by the mucosa of the large intestine, further breakdown of dietary constituents is carried out by the resident microbiota( Reference Wilson 6 ). Carbohydrates are mainly fermented in the proximal colon, whereas the fermentation of proteins takes place mainly in the distal colon. Transit time of digesta through the colon strongly influences the activities of the gut microbiota. The mean transit time of the oro-GI tract has been reported to be approximately 70 h in UK people consuming a normal daily diet( Reference Cummings and Macfarlane 7 , Reference Stevens, VanSoest and Robertson 8 ). The primary activity of the caecum and colon microbiota is the breakdown of carbohydrates not digested in the ileum to SCFA, which are then rapidly absorbed. The principal products of carbohydrate fermentation are SCFA (acetate, propionate and butyrate), hydrogen and CO2 gasses and bacterial cell mass (biomass)( Reference Cummings and Macfarlane 9 ). The amount of energy derived from SCFA accounts for up to 10 % of the total energy requirement of human subjects( Reference McNeil 10 ). From a nutritional point of view, the SCFA are important since they not only provide the body with energy but are also metabolised in different tissues( Reference Cummings and Macfarlane 9 ).

The microbiota in the colon and faeces is extremely diverse and on the basis of estimations from culture-based and molecular studies more than 1200 prevalent bacterial species altogether reside there. Each individual harbours at least 160 such species( Reference Rajilić-Stojanović, Smidt and De Vos 11 , Reference Qin, Li and Raes 12 ). Under normal circumstances, predominant intestinal microbiota of an adult individual is fairly stable. However, in studies where the long-term temporal stability of the predominant microbiota has been assessed from healthy subjects, the number of subjects has been limited( Reference Zoetendal, Akkermans and De Vos 13 Reference Seksik, Rigottier-Gois and Gramet 16 ). The human GI-tract, although harbouring a vast number of microbes, has only a limited diversity at the phylum level. Microbes from seven bacterial phyla (Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, Fusobacteria, Verromicrobia and Cyanobacteria-like) and one archael phylum (Euryarchaeota) have been detected in the human intestine( Reference Eckburg, Bik and Bernstein 17 Reference Turnbaugh, Hamady and Yatsunenko 21 ). However, the majority of the GI-tract population are representatives of three phyla: the Firmicutes (Families Lachnospiraceae and Ruminococcaceae), the Bacteroidetes (Bacteroidaceae, Prevotellaceae and Rikenellaceae) and the Actinobacteria (Bifidobacteriaceae and Coriobacteriaceae).

Faecal microbiota is dominated by bacteria from Families Lachnospiraceae (Eubacterium rectale group) and Ruminococcaceae (Clostridium leptum group) and Bacteroidaceae/Prevotellaceae( Reference Franks, Harmsen and Raangs 3 , Reference Harmsen, Raangs and He 4 , Reference Eckburg, Bik and Bernstein 17 , Reference Suau, Bonnet and Sutren 18 , Reference Sghir, Gramet and Suau 22 ), which together account for 60–80 % of the faecal bacteria of healthy adults( Reference Sghir, Gramet and Suau 22 Reference Mueller, Saunier and Hanisch 24 ). The bacteria belonging to the Family Lachnospiraceae comprise on average 10–45 % of the total faecal bacteria as detected with quantitative hybridisation-based methods( Reference Franks, Harmsen and Raangs 3 , Reference Harmsen, Raangs and He 4 , Reference Sghir, Gramet and Suau 22 Reference Marteau, Pochart and Doré 30 ) and the bacteria within Ruminococcaceae 16–27 %, thus co-dominating with the bacteria within Lachnospiraceae( Reference Sghir, Gramet and Suau 22 Reference Maukonen, Satokari and Mättö 25 , Reference Marteau, Pochart and Doré 30 Reference Saunier, Rouge and Lay 32 ). Many members of the Lachnospiraceae and Ruminococcaceae are polysaccharolytic and produce butyrate. In addition, they are obligately anaerobic. Members of the Bacteroidetes phylum comprise 12–60 % of the total faecal bacteria depending on which method has been used for the quantification of this group( Reference Sghir, Gramet and Suau 22 Reference Mueller, Saunier and Hanisch 24 , Reference Doré, Schir and Hannequart-Gramet 33 , Reference Rigottier-Gois, Rochet and Garrec 34 ). Bacteroides spp. are saccharolytic bacteria that contain a wealth of polysaccharide-degrading enzymes. They have an excellent ability to ferment simple and complex sugars and polysaccharides, producing acetate and succinate as major metabolic end products. In addition, most species are weakly proteolytic( Reference Shah, Balows, Trüper, Dworkin, Harder and Schleifer 35 ). Most of the bacteria within Prevotellaceae and Porphyromonadaceae have been isolated from the oral cavity and are commonly found in oral sites in culture-based studies, whereas with DNA-based molecular studies members of Prevotellaceae and Porphyromonadaceae have also been found in faeces.

Bifidobacterium spp., Lactobacillus-group and bacteria within the Family Coriobacteriaceae although found in most human subjects, comprise smaller populations among faecal bacteria in adults (bifidobacteria 0·5–6 %, lactobacilli <1–3 % and Coriobacteriaceae 1–5 % of the total faecal bacteria)( Reference Franks, Harmsen and Raangs 3 Reference Harmsen, Raangs and He 4 , Reference Sghir, Gramet and Suau 22 Reference Mueller, Saunier and Hanisch 24 , Reference Marteau, Pochart and Doré 30 ). Moreover, there are several bacteria/bacterial groups, which usually comprise <1–2 % of the total faecal microbiota of healthy adults, such as Akkermancia muciniphila, ‘true clostridia’ (Family Clostridiaceae), enterobacteria, streptococci, bacteria within the Families Peptostreptococcaceae, Erysipelotrichaceae, Veillonellaceae, Eubacteriaceae, in addition to sulphate-reducing bacteria and/or methanogens( Reference Franks, Harmsen and Raangs 3 , Reference Sghir, Gramet and Suau 22 Reference Lay, Rigottier-Gois and Holmstrøm 23 , Reference Rigottier-Gois, Le Bourhis and Gramet 27 , Reference Collado, Derrien and Isolauri 36 Reference Gibson, Macfarlane and Cummings 39 ). Besides bacteria, low levels of viruses, archaea (<1 %) and eukaryotes (≤2 %) are also found from the large intestine( Reference Gibson, Macfarlane and Cummings 39 Reference Doré, Gramet and Goderel 41 ).

The effect of sampling and used analysis methods on the results obtained from gastrointestinal-tract samples

Conditions during sample transportation have a major impact on sample quality( Reference Roesch, Casella and Simell 42 Reference Molbak, Sommer and Johnsen 44 ) and therefore the time between sampling and further processing and storing should be limited to the minimum. It should be kept in mind that regardless of how sophisticated the applied method for characterisation of the microbial community is, the result can be only as good as the sample that arrives in the laboratory. In addition, when molecular techniques are applied to study microbial communities of the GI tract, oral bacteria may be detected with molecular techniques from faeces as well, since the human oro-GI tract is an open system. In studies with snapshot samples, it is difficult to confer that the detected bacteria are resident and not just transient. Therefore to be able to establish resident microbiome, follow-up samples would be needed.

Most of the steps in the processing of faecal samples may affect the microbiological results obtained. The sampling and storage conditions affect the results( Reference Roesch, Casella and Simell 42 Reference Molbak, Sommer and Johnsen 44 ), although not as extensively as the following steps, especially the DNA extraction( Reference Kennedy, Walker and Berry 45 Reference Salonen, Nikkilä and Jalanka-Tuovinen 48 ). One week storage at −20 °C decreases the detected numbers of Gram-negative Bacteroides spp. by over one logarithmic unit as compared with the fresh samples, whereas the storage temperature does not have such a significant effect on the Gram-positive bacteria (e.g. Firmicutes and Actinobacteria)( Reference Maukonen, Simões and Saarela 47 ). Since the analysis of fresh faecal samples, especially in the case of large cohorts, is impractical and mostly also impossible, the effect of the storage conditions on the results obtained should be taken into account when the results are discussed.

When DNA extraction is performed using solely chemical treatments combined with heat treatments, the results for some bacterial groups may be as much as four logarithmic units lower as compared with a DNA extraction method in which rigorous mechanical disruption is applied( Reference Maukonen, Simões and Saarela 47 ). Actinobacteria, in particular, are underestimated with many currently used techniques, most probably due to their high guanine–cytosine content( Reference Krogius-Kurikka, Kassinen and Paulin 49 ). In a study of Nakamura et al.( Reference Nakamura, Gaskins and Collier 50 ) it was shown that when the same samples were examined with quantitative PCR (enzymatic DNA extraction) and fluorescent in situ hybridisation (FISH), the difference in results was enormous. The proportions of bifidobacteria with quantitative PCR was 0·1–1·7 %, whereas the proportion with FISH was 28–84 %( Reference Nakamura, Gaskins and Collier 50 ). Moreover, the number of bacteria within Family Coriobacteriaceae has been shown to be 5–6 logarithmic units higher in a sample that was initially stored at +4 °C for 2 d and thereafter at −70 °C and when DNA extraction was performed mechanically than on the same sample that was stored at −70 °C and the DNA extraction was performed enzymatically( Reference Maukonen, Simões and Saarela 47 ). This may also explain why in some studies members of Coriobacteriaceae are not considered to be the part of the normal dominant microbiota( Reference Eckburg, Bik and Bernstein 17 ), whereas in others, especially those conducted with FISH, bacteria within Coriobacteriaceae constitute 1–8 % of the total population of the human gut microbiota( Reference Lay, Rigottier-Gois and Holmstrøm 23 , Reference Mueller, Saunier and Hanisch 24 , Reference Harmsen, Wildeboer-Veloo and Grijpstra 38 , Reference Matsuki, Watanabe and Fujimoto 51 ).

Another methodological aspect that may cause confusion is that different probes/primers target different bacterial populations, such as in the detection of Bacteroidetes and Firmicutes. In metagenomic/16S rRNA-based next-generation sequencing studies the authors usually refer to Bacteroidetes, whereas in studies conducted with FISH/PCR, the studied bacterial population is usually narrower (e.g. Bacteroides spp. or Bacteroides-Prevotella group)( Reference Hoyles and McCartney 52 ). The same controversy applies to Firmicutes. In many next-generation sequencing studies, Firmicutes are discussed as a single group, whereas in fact it contains several different bacterial classes e.g. Bacilli, Clostridia, Erysipelotrichia, which all have different metabolic properties. Furthermore, when PCR/FISH is used, each of these groups is usually studied separately. Moreover, since the phylum Bacteroidetes is more thoroughly characterised than the phylum Firmicutes, it is possible that the representation of Bacteroidetes in metagenomic studies is overestimated.

When microbial communities are examined, especially using several different techniques and/or next-generation sequencing, the quantity of data generated is enormous. Therefore the use of suitable algorithms e.g. measurement of microbial diversity and correct interpretation of the results are of utmost importance( Reference Walker, Duncan and Louis 53 Reference Achtman and Wagner 61 ). It should also be borne in mind that even if statistical significance is achieved, the observed changes may not necessarily be of biological significance.

Factors that affect the gut microbiota

There are several factors that affect the composition of the human GI microbiota, such as genetics, sex, ethnicity, age, medication, diseases/disorders and last but not least the diet.

Genetics of the host and geography

Twins, especially monozygotic (MZ) twins, have been reported to have more similar interindividual faecal microbiota than unrelated people. In addition, twins and their mothers have a more similar microbiota than unrelated individuals. These findings have led to the conclusion that the host genotype affects the development of the gut microbiota and gut bacterial composition( Reference Zoetendal, Akkermans and Akkermans-van Vliet 14 , Reference Turnbaugh, Hamady and Yatsunenko 21 , Reference Turnbaugh, Quince and Faith 62 , Reference Stewart, Chadwick and Murray 63 ). However, the aforementioned studies have mainly been conducted with MZ pairs concordant for leanness or obesity. Simoes et al. ( Reference Simoes, Maukonen and Kaprio 64 ) studied MZ twins concordant and discordant for obesity, and found that the concordant normal-weight MZ twins had more similar bacterial populations than the MZ twins discordant for obesity. These findings also address the importance of the diet in addition to the genetic drivers( Reference Simoes, Maukonen and Kaprio 64 ). Although the genetics or the shared environmental factors during upbringing result in more similar bacterial populations, viromes have been shown to be unique to individuals regardless of their degree of genetic relatedness( Reference Reyes, Haynes and Hanson 40 ).

Besides genetics, the effect of the geographic origin (which may also include genetic differences and environmental sources of variation) has also been studied in relation to the gut microbiota composition. Even in Europe some differences may be found between different countries, e.g. proportions of bifidobacteria have been found to be 2- to 3-fold higher in an Italian adult study population than in other European study populations( Reference Lay, Rigottier-Gois and Holmstrøm 23 , Reference Mueller, Saunier and Hanisch 24 ). In addition, when 6-week-old infants across Europe were studied, geography was a more prominent factor than delivery mode, breast-feeding and antibiotics. In infants, children from Northern European countries had more bifidobacteria, whereas the Southern European infants were associated with more diverse microbiota and higher numbers of Bacteroides than the Northern European children( Reference Fallani, Young and Scott 65 ). In the studies involving people from different continents, the bacterial population differences have been more distinct. In a few recent studies, there have been consistent results that the European and North-American faecal microbiota differ significantly from the Southern-American, African and Chinese faecal microbiota( Reference Yatsunenko, Rey and Manary 66 Reference De Filippo, Cavalieri and Di Paola 68 ). However, in all of these studies it is not possible to exclude the impact of other possible confounding factors, and therefore dietary habits and genetics may also contribute to the differences.

Age

Bacterial colonisation of the infant GI tract is influenced by e.g. mode of delivery, prematurity, type of feeding (breast feeding v. formula feeding), antibiotic treatment of the child or the mother, lifestyle and geographics( Reference Fallani, Young and Scott 65 , Reference Fallani, Amarri and Uusijarvi 69 Reference Bezirtzoglou, Tsiotsias and Welling 74 ). The earliest colonisers are usually facultative anaerobic bacteria such as Enterobacteriaceae, streptococci and staphylococci, whereas later colonisers tend to be strict anaerobes e.g. bifidobacteria, clostridia and Bacteroides spp. regardless of the infant's geographical origin and methods used for the detection( Reference Fallani, Young and Scott 65 , Reference Favier, De Vos and Akkermans 75 Reference Wang, Ahrne and Antonsson 80 ). Immediately after birth, the rectal microbiota of vaginally delivered babies resembles their own mother's vaginal microbiota, whereas the rectal microbiota of babies delivered by Caesarean section resembles that of the skin( Reference Dominguez-Bello, Costello and Contreras 81 ). The gut microbiota of preterm infants is less diverse than those of full-term babies( Reference Rougé, Goldenberg and Ferraris 70 , Reference Schwiertz, Gruhl and Löbnitz 73 , Reference Magne, Abély and Boyer 82 Reference Hall, Cole and Smith 84 ). There are numerous studies in which the predominance of bifidobacteria in exclusively breast-fed infants has been found( Reference Fallani, Young and Scott 65 , Reference Yatsunenko, Rey and Manary 66 , Reference Bezirtzoglou, Tsiotsias and Welling 74 , Reference Bullen, Tearle and Willis 85 Reference Magne, Hachelaf and Suau 88 ). At age 3–6 weeks, exclusively breast-fed infants harbour higher numbers of bifidobacteria, whereas formula-fed babies have more diverse microbiota, lower numbers of bifidobacteria and higher numbers of Bacteroides, Lachnospiraceae, Lactobacillus group, Clostridium difficile and Coriobacteriaceae( Reference Fallani, Young and Scott 65 , Reference Penders, Thijs and Vink 71 , Reference Bezirtzoglou, Tsiotsias and Welling 74 , Reference Harmsen, Wildeboer-Veloo and Raangs 87 , Reference Penders, Vink and Driessen 89 , Reference Stark and Lee 90 ). By the end of the first year of life, when the child has already started to eat the same foods as the adults, the gut microbiota starts to converge towards a profile characteristic of the adult microbiota( Reference Palmer, Bik and DiGiulio 76 , Reference Stark and Lee 90 ). However, the faecal bacterial diversity is still lower. By the end of the second to third year, the phylogenetic composition evolves towards the adult-like composition( Reference Yatsunenko, Rey and Manary 66 , Reference Hentges 91 ).

The GI microbiota evolves with age( Reference Gorbach, Nahas and Lerner 92 ). Dental deterioration, salivary function, digestion, slower intestinal transit time and changes in diet and physical activity may affect the GI microbiota of ageing people. Interest, as well as the number of studies, in the GI microbiota of elderly people has grown as life expectancy in the Western world has rapidly increased. The elderly have been reported to have relatively stable microbiota( Reference Rajilic-Stojanovic, Heilig and Molenaar 93 Reference Woodmansey, McMurdo and Macfarlane 95 ). However, the microbiota of the elderly has been reported to be more diverse and to contain partly different core microbiota as compared with younger adults( Reference Rajilic-Stojanovic, Heilig and Molenaar 93 , Reference Claesson, Cusack and O'Sullivan 94 , Reference Blaut, Collins and Welling 96 , Reference Maukonen, Mättö and Kajander 97 ). Moreover, inter-individual variation is greater in elderly people as compared with younger adults( Reference Finegold, Sutter, Mathisen and Hentges 2 , Reference Claesson, Cusack and O'Sullivan 94 , Reference Claesson, Jeffery and Conde 98 ).

Medication

In addition to dietary components, the other important external factor affecting the microbiota is antibiotic use. However, different types of antibiotics have different types of action mechanisms and thus different effects on the human microbiota( Reference Kohanski, Dwyer and Collins 99 , Reference Sullivan, Edlund and Nord 100 ). In addition, individual human responses may be different. Bifidobacteria are typically susceptible to the majority of clinically relevant antibiotics such as penicillins (β-lactam antibiotics), cephalosporins and macrolides( Reference Delgado, Flórez and Mayo 101 Reference Huys, D'Haene and Cnockaert 106 ), and most of the commensal gut bacteria (e.g. bifidobacteria, Bacteroides spp.) to amoxicillin (β-lactam antibiotic) and clavulanate( Reference Jousimies-Somer, Summanen and Citron 102 ).

The effect of diet on human gut microbiota

The importance of the metabolic activities of the gut microbiota from the host's perspective

From the host's perspective, there are numerous activities of the commensal gut microbiota that are of great importance to health. Carbohydrates are mainly fermented in the proximal colon, whereas the fermentation of proteins takes place mainly in the distal colon. However, the metabolic output of the microbial community depends not only on available substrates, but also on the gut environment, with the pH playing a major role. For example, at pH 6·7 Bacteroides spp. predominate, whereas at pH 5·5 bacteria related to E. rectale predominate( Reference Louis, Scott and Duncan 107 Reference Walker, Duncan and Carol McWilliam Leitch 109 ). The main saccharolytic genera in the human GI-tract are Bacteroides, Bifidobacterium, Clostridium, Eubacterium, Lactobacillus and Ruminococcus. The saccharolytic genera are able to produce SCFA, which have both local and systematic beneficial biological effects. A wide range of bacteria have proteolytic activities, such as clostridia, and species within genera Propionibacterium, Prevotella, Bifidobacterium and Bacteroides. Protein metabolism, however, is not as favourable to the host as carbohydrate metabolism; some of the end-products of amino acid metabolism may be deleterious to the host, e.g. ammonia, amines and phenol compounds. Some species of the genera Bacteroides, ruminococci and Akkermansia are able to break down mucin. Moreover, several Eubacterium spp. and Clostridium spp. are able to dehydroxylate bile acids, some Clostridium spp. transform conjugated bilirubin, Eubacterium coprostanoligenes is able to convert cholesterol to coprostanol and Bacteroides spp. inactivate tryptic activity, have dipeptidase activity and play a key role in the enterohepatic circulation of bile acids( Reference Cummings and Macfarlane 7 , Reference Shah, Balows, Trüper, Dworkin, Harder and Schleifer 35 , Reference Wade, Stackebrandt, Jones, Holzapfel, Reboli and Farrar 110 Reference Derrien, Vaughan and Plugge 114 ).

In addition to the individual activities, cross-feeding between the gut microbes and the metabolic networks thus created are also of great importance. For example, it has been shown in vitro that lactate produced by Bifidobacterium adolescentis as a fermentation product from fructo-oligosaccharide (FOS) and starch was further utilised by butyrate producers, which were not able to grow solely on FOS and starch( Reference Belenguer, Duncan and Calder 115 ). In addition, Roseburia intestinalis and Anaerostipes caccae were able to grow with Bifidobacterium longum using FOS; R. intestinalis was able to grow on the FOS-supplemented medium when acetate, a major fermentation product of B. longum, was added to the medium, whereas A. caccae was able to utilise fructose that was released during the bifidobacterial fermentation of FOS( Reference Falony, Vlachou and Verbrugghe 116 ). Besides other survival mechanisms, gene transfer within and from outside the gut microbiota has also been shown to occur. In Japan, where consumption of marine algae is high, a Japanese gut bacterium (Bacteroides plebeius) has acquired genes coding for porphyromonases, agarases and associated proteins and thus the ability to utilise marine algae. These algae are not readily fermentable by Western gut microbiota( Reference Hehemann, Correc and Barbeyron 117 ).

Dietary interventions v. habitual diet

The main external factors that can affect the composition of the microbial community in generally healthy adults include major dietary changes and antibiotic therapy. Changes in some selected bacterial groups have been observed due to controlled changes to the normal diet e.g. high-protein diet( Reference Hentges, Maier and Burton 118 , Reference Russell, Gratz and Duncan 119 ), prebiotics( Reference Maukonen, Mättö and Kajander 97 , Reference Tuohy, Kolida and Lustenberger 120 Reference Langlands, Hopkins and Coleman 122 ), probiotics( Reference Rinne, Kalliomaki and Salminen 123 Reference Lyra, Krogius-Kurikka and Nikkila 125 ), weight-loss diet( Reference Ley, Turnbaugh and Klein 20 , Reference Duncan, Lobley and Holtrop 126 , Reference Nadal, Santacruz and Marcos 127 ) and berries( Reference Puupponen-Pimiä, Seppänen-Laakso and Nohynek 128 ). More specifically, changes in the type and quantity of non-digestible carbohydrates in the human diet influence both the metabolic products formed in the lower regions of the GI tract and levels of bacterial populations detected in faeces( Reference Duncan, Belenguer and Holtrop 129 ). The interactions between dietary factors, gut microbiota and host metabolism are important for maintaining homeostasis and health( Reference Flint, Duncan and Scott 130 ).

The impact of habitual diet on faecal microbiota has been studied for decades. In older culture-based studies, it was found that numbers of bacteroides were lower and numbers of enterococci and Escherichia coli higher in Ugandan, Indian and Japanese people on a high-carbohydrate diet as compared with people on a Western diet( Reference Drasar, Crowther and Goddard 131 , Reference Finegold, Attebery and Sutter 132 ). However, when English people on a strictly vegetarian diet were studied, their microbiota resembled those of people on a Western diet more than the microbiota of other vegetarian people from different continents( Reference Drasar, Crowther and Goddard 131 ). Similarly, numbers of bacteroides and clostridia were lower in the Nigerian Maguzawa tribal people (predominantly cereal diet) than in the other dietary groups( Reference Drasar, Montgomery and Tomkins 133 ). In more recent molecular studies, in which gut microbiota from different parts of the world has been compared, a significant correlation between habitual diet and faecal microbiota has also been found. African children consuming a diet low in fat and animal protein and rich in starch, fibre and plant polysaccharides (predominantly vegetarian) had significantly more Bacteroidetes and Actinobacteria and less Firmicutes and Proteobacteria than European children, who had a diet high in animal protein, fat, sugar and starch, but low in fibre. Moreover, members of the genera Prevotella and Xylanibacter were found exclusively from the African children( Reference De Filippo, Cavalieri and Di Paola 68 ). Partly similarly to these findings, Prevotella enterotype was previously found to be associated with high consumption of carbohydrates and with both vegetarian and vegan habitual diets, whereas Bacteroides enterotype was associated with high consumption of animal protein, amino acids and saturated fats( Reference Wu, Chen and Hoffmann 134 ). Strict vegetarians have higher percentages and/or numbers of Lachnospiraceae and Clostridium ramosum group (Clostridial cluster XVIII) bacteria than omnivorous people( Reference Hayashi, Sakamoto and Benno 135 , Reference Hippe, Zwielehner and Liszt 136 ), whereas vegans have lower faecal numbers of Bacteroides spp., bifidobacteria and Enterobacteriaceae( Reference Zimmer, Lange and Frick 137 ). In older culture-based studies, no fusobacteria were found in faecal samples of vegetarian people. In addition, the total count of anaerobic streptococci, Peptostreptococcus spp., Actinomyces spp. and Lactobacillus spp. were higher in strict vegetarians as compared with people on a traditional Western diet( Reference Finegold, Sutter, Mathisen and Hentges 2 , Reference Finegold, Attebery and Sutter 132 , Reference Finegold, Flora and Attebery 138 , Reference Finegold, Sutter and Sugihara 139 ).

Hospitalisation of Scottish elderly people has resulted in a decrease of faecal Bacteroides–Prevotella group and Ruminococcus albus prevalence, while increasing the Enterococcus faecalis prevalence, as compared with healthy elderly people. Moreover, when the hospitalisation was combined with antibiotic treatment, reductions in other bacterial groups were also observed( Reference Bartosch, Fite and Macfarlane 140 ). Long-term residential care of Irish elderly people has been associated with a higher proportion of bacteria belonging to the phylum Bacteroidetes and several genera from other phyla, whereas community-dwelling elderly people had higher proportions of bacteria belonging to the phylum Firmicutes. In addition, the microbiota of elderly people in long-term care was significantly less diverse. However, the results also correlated with the diet; if the elderly people were grouped according to their diet, the clustering was similar to that based on the residence location. After 1 month residential care, the diet was converged to ‘long-term diet’, but it took 1 year for the microbiota to clearly cluster within the long-term residential type. Collectively this indicates that the composition of the faecal microbiota is determined rather by the composition and diversity of the diet than by the location of residence( Reference Claesson, Jeffery and Conde 98 ).

Since it has not always been clear which part of the habitual diet induces the changes observed in the GI-tract microbiota, numerous dietary interventions have been conducted to elucidate the changes induced by specific nutritional substances, e.g. different types of fibres, proteins, fats and polyphenols. In addition, the effects of probotics and prebiotics on the GI-tract microbiota have been widely studied. Therefore, the effects of the aforementioned nutritional substances on GI-tract microbiota are addressed in more detail.

Fermentable dietary carbohydrates

Dietary components that escape digestion by endogenous enzymes in the upper GI tract become available substrates in the large intestine( Reference Cummings and Macfarlane 7 ). Dietary fibre (DF) is a normal constituent of most foods derived from plants( Reference Blaut 141 ). These ‘non-digestible’ dietary carbohydrate substrates include resistant starch, plant cell-wall material (non-starch polysaccharides) and oligosaccharides( Reference Cummings and Macfarlane 7 ). In the human colon, DF is metabolised by the microbiota to SCFA, comprising mainly acetic, propionic and butyric acids. SCFA have been implicated to have both local and systemic beneficial biological effects in the human body; acetate is readily absorbed and transported to the liver; propionate is a substrate for hepatic gluconeogenesis; butyrate is the preferred fuel of the colonocytes and also plays a major role in the regulation of cell proliferation and differentiation( Reference Cummings and Macfarlane 7 ). Several excellent review-papers already exist( Reference Flint, Duncan and Scott 130 , Reference Flint, Scott and Duncan 142 Reference Flint 147 ), and therefore we will not go into detail with the dietary interventions with fibre.

In vitro and in vivo evidence indicate that a bacterial group related to Faecalibacterium prausnitzii, Roseburia and E. rectale plays a major role in mediating the butyrogenic effect of fermentable dietary carbohydrates( Reference Louis, Scott and Duncan 107 , Reference Aminov, Walker and Duncan 148 Reference Pryde, Duncan and Hold 150 ). In addition, it has been shown in numerous studies that as dietary carbohydrate content is reduced in the diet the count of F. prausnitzii declines, respectively( Reference Duncan, Belenguer and Holtrop 129 , Reference Benus, van der Werf and Welling 151 ). However, some lactate-utilising bacteria within Lachnospiraceae produce less butyrate in the presence of lactate-utilising sulphate-reducing bacteria. Moreover, in the presence of higher abundance of lactate, the formation of butyrate was reduced even more and the formation of hydrogen sulphide was promoted( Reference Marquet, Duncan and Chassard 152 ).

Cereal grains are a good source of DF. The main DF components of cereal grains are arabinoxylan, cellulose, β-glucan, fructan, resistant starch and lignin( Reference Karppinen, Liukkonen and Aura 153 ). The gut microbiota stimulating activities of arabinoxylan (stimulates Bacteroides spp. and Roseburia spp.), resistant starch (stimulates bifidobacteria, Bacteroides spp., Ruminococcus bromii, E. rectale and Roseburia spp.), β-glucan (stimulates bifidobacteria) and fructan (stimulates bifidobacteria, Bacteroides spp., lactobacilli and butyrate-producers) are well recognised( Reference Flint, Bayer and Rincon 144 , Reference Hughes, Shewry and Gibson 154 Reference Abell, Cooke and Bennett 161 ). In addition, arabinoxylan-oligosaccharides, which are enzymatic hydrolysis products of arabinoxylan, have been shown to stimulate the growth of bifidobacteria in some studies( Reference Broekaert, Courtin and Verbeke 156 ). The effects of cellulose and lignin on gut microbiota are less well known( Reference Ehle, Robertson and Van Soest 162 , Reference Robert and Bernalier-Donadille 163 ). However, it should be noted that e.g. the size of the grain flakes may lead to different bacterial responses: i.e. smaller-sized whole-oat grain flakes (0·53–0·63 mm) resulted in a significant increase in the numbers of BacteroidesPrevotella group bacteria, whereas in a fermentation with larger oat flakes (0·85–1·00 mm) bifidobacterial numbers increased( Reference Connolly, Lovegrove and Tuohy 164 ). It has also been shown in in vitro model studies using different substrates that the majority of the bacteria attached to wheat bran belonged to Lachnospiraceae and some bacteria were Bacteroides spp., whereas R. bromii, B. adolescentis, Bifidobacterium breve and E. rectale were found attached to starch. When mucin was used as a substrate, the most commonly found bacteria were Bifidobacterium bifidum and an uncultured relative of Ruminococcus lactaris ( Reference Leitch, Walker and Duncan 165 ).

Prebiotics

Prebiotics are non-digestible (by the host) food ingredients that have a beneficial effect through their selective metabolism in the intestinal tract. The prebiotics that currently fulfil the prebiotic criteria are inulin, FOS, galacto-oligosaccharides and lactulose( Reference Gibson, Probert and Van Loo 166 ). The best sources of naturally occurring prebiotics may be found in vegetables such as artichokes, onions, chicory, garlic and leeks( Reference Macfarlane and Cummings 167 ). There are numerous studies in which the bifidogenic properties of prebiotics are shown( Reference Walton, van den Heuvel and Kosters 121 , Reference Langlands, Hopkins and Coleman 122 , Reference Gibson, Probert and Van Loo 166 , Reference Tannock, Munro and Bibiloni 168 Reference Roberfroid, Gibson and Hoyles 171 ). In addition, increase in abundance of lactobacilli( Reference Walton, van den Heuvel and Kosters 121 , Reference Langlands, Hopkins and Coleman 122 ) and F. prausnitzii ( Reference Ramirez-Farias, Slezak and Fuller 170 , Reference Louis, Young and Holtrop 172 ) has been shown. Moreover, in infant formulas, galacto-oligosaccharides + FOS supplementation of cow's milk-based formula has led to a bifidobacterial population which resembled more that of breast-fed infants than purely formula-fed infants( Reference Rinne, Gueimonde and Kalliomaki 173 ). FOS have also positive effects on the intestinal barrier function( Reference Cani 174 ).

Protein

Endogenous protein sources make up approximately one-third of the exogenous dietary protein pool ( Reference Alberts, Johnson and Lewis 175 , Reference Silk 176 ). Bacterial amino acid catabolism in the human gut occurs via a number of mechanisms involving either deamination or decarboxylation reactions. The types of SCFA produced from amino acids are dependent on the chemical compositions of the substrates. In addition to SCFA, branched chain-fatty acids and aromatic compounds, namely phenol, indole and a range of phenolic and indolic substituted fatty acids derived from phenylalanine, tyrosine and tryptophan may be formed. Moreover, branched-chain amino acids are slowly fermented by colonic bacteria, with the main acidic products being branched chain-fatty acids one carbon atom shorter than the parent amino acid( Reference Smith and Macfarlane 177 ). Many metabolites produced by amino acid fermentation are harmful to the host. Phenolic and indolic compounds are also thought to act as co-carcinogens, while amines serve as precursors of nitrosamine production( Reference Smith and Macfarlane 177 ).

Culture-based studies have shown( Reference Hentges, Maier and Burton 118 ) that the counts of Bacteroides spp. and clostridia increased significantly, whereas counts of B. adolescentis decreased significantly during a high-beef diet as compared with a meatless diet (fat and fibre contents were essentially the same in both diets). In addition, sulphide concentrations were high on a high-beef diet( Reference Magee, Richardson and Hughes 178 ). Since hydrogen sulphide is toxic to the colonic epithelium and sulphide inhibits butyrate oxidation, dietary sulphide may selectively stimulate the growth of a single group of bacteria, namely sulphate-reducing bacteria, with potentially harmful effects on the epithelium( Reference Cummings and Macfarlane 9 , Reference Pitcher and Cummings 179 ). In addition, an intervention diet with a high protein and low carbohydrate content reduced the numbers of Roseburia/E. rectale group, while increasing proportions of branched-chain fatty acids and concentrations of phenylacetic acid and N-nitroso compounds( Reference Russell, Gratz and Duncan 119 ). Moreover, it should be noted that the World Cancer Research Fund released in May 2011 a report based on 1012 clinical trials, in which red and processed meat were convincingly associated with increased risk, whereas foods containing DF, in particular cereal fibre and whole grains, were associated with decreased risk of colorectal cancer( 180 ).

Fat

Fats are composed of fatty acids that are divided into SFA and unsaturated fatty acids. Dietary SFA are mainly obtained from animal products, such as meats and dairy foods, but may also be obtained from some plant sources, such as coconut, cottonseed and palm kernel oils. The major dietary MUFA is oleic acid. Oleic acid is the primary component of olive oil, but may also be found in hazelnut, rapeseed and peanut oils. A carbon chain that contains two or more cis double bonds characterises the families of n-3 or n-6 PUFA. These families cannot be synthesised by the human body( Reference Ortega, Varela and Bermudez 181 ). Linoleic (n-6 PUFA) and α-linolenic (n-3 PUFA) acids form the majority of PUFA in most Western diets. The long-chain n-3 PUFA EPA and DHA are found in seafood, especially oily fish( Reference Calder, Ahluwalia and Brouns 182 ).

High intake of dietary fat may increase the quantities of bile acids and fat that reach the colon. It has been suggested that the gut microbiota may metabolise dietary fats (producing diacylglycerols from polyunsaturated fats), convert primary bile acids into secondary bile acids and impact on the enterohepatic circulation of bile acids and fat absorption from the small intestine( Reference Fava, Gitau and Griffin 183 ). However, there are only a few human studies in which the effect of high-fat diet on the human intestinal microbiota has been investigated, and especially those in which the correlations between the different types of dietary fat and intestinal microbiota have been investigated. In a study of Brinkworth et al. ( Reference Brinkworth, Noakes and Clifton 184 ), it was shown that a very low-carbohydrate, high-fat diet resulted in a significant reductions in bifidobacterial numbers, concentrations of butyrate and total SCFA, defecation frequency and faecal excretion as compared with isoenergetic high-carbohydrate, high-fibre and low-fat diet.

High MUFA-containing dietary intervention that lasted 4 weeks reduced the total bacterial numbers but did not affect the specific bacterial groups( Reference Fava, Gitau and Griffin 183 ). Conversely, high habitual intake of MUFA has been associated with lower numbers of bifidobacteria and slightly higher numbers of Bacteroides spp.( Reference Simoes, Maukonen and Kaprio 64 ). In a recent metagenomic study in healthy volunteers, the Bacteroides enterotype was found to be highly associated with the consumption of MUFA and SFA( Reference Wu, Chen and Hoffmann 134 ). These observations suggest that the consumption of fat and animal-derived products, typically present in the Western diet, are associated with increased Bacteroides spp. prevalence in the human gut microbiota.

Habitual n-3 PUFA intake has been shown to have a significant positive association with Lactobacillus group abundance( Reference Simoes, Maukonen and Kaprio 64 ). The increase in Lactobacillus group bacterial numbers in stool after n-3 PUFA intake has also been reported in a mouse study( Reference Pachikian, Neyrinck and Portois 185 ). In addition, in a human study by Santacruz et al. ( Reference Santacruz, Marcos and Wärnberg 186 ) the numbers of lactobacilli remained at the same level, even though the ingested amount of total PUFA was greatly reduced. The increase in n-3 PUFA is effective in supporting epithelial barrier integrity by improving transepithelial resistance and by reducing IL-4-mediated permeability( Reference Willemsen, Koetsier and Balvers 187 ), and several lactobacilli enhance the function of the intestinal barrier( Reference Anderson, Cookson and McNabb 188 , Reference Donato, Gareau and Wang 189 ). Maternal salmon (marine n-3 PUFA) consumption before delivery has also lowered the number of Coriobacteriaceae in bottle-fed infants( Reference Urwin, Miles and Noakes 190 ).

Higher habitual n-6 PUFA intake has been associated with decreased numbers of bifidobacteria( Reference Simoes, Maukonen and Kaprio 64 ). It has also been reported that high n-6 PUFA intakes decrease certain immune functions, such as antigen presentation, adhesion molecule expression, proinflammatory cytokines and T-helper 1 and T-helper 2 responses( Reference Harbige 191 ). Furthermore, genomic DNA of some bifidobacterial strains is able to stimulate the production of T-helper 1 and proinflammatory cytokines, interferon-γ and TNF-α( Reference Medina, Izquierdo and Ennahar 192 ). Overall, these results indicate an association between dietary fat types and their distinct effect on the faecal microbiota. As a consequence, it seems that balanced diet with regard to fat consumption is critical not only for the host's health, but also for the gut microbiota.

Polyphenols

Plant foods contain significant amounts of phenolic compounds( Reference Aura 193 ). Plant polyphenols are a class of chemically diverse secondary metabolites that possess many different biological activities both within the plant and in the human subjects eating these plants. Plant polyphenols have the potential to affect certain risk factors of CVD, as well as being antioxidants, have antimicrobial properties and possessing inherent free radical scavenging abilities( Reference Tuohy, Conterno and Gasperotti 194 ). The main dietary sources of polyphenols are berries, fruits, beverages (e.g. coffee, tea and wine), chocolate, whole-grain cereals, vegetables and legume seeds( Reference Aura 193 ).

The human gut microbiota has extensive hydrolytic activities and breaks down many complex polyphenols into smaller phenolic acids, which can be absorbed across the intestinal mucosa( Reference Tuohy, Conterno and Gasperotti 194 ). Daily consumption of red wine polyphenols for 4 weeks significantly increased numbers of bacteria within genera Enterococcus, Prevotella, Bacteroides, Bifidobacterium, Eggerthella and Family Lachnospiraceae( Reference Queipo-Ortuno, Boto-Ordonez and Murri 195 ), whereas consumption of high cocoa flavanol drink for 4 weeks significantly increased the bifidobacterial and lactobacilli numbers but significantly decreased clostridial counts( Reference Tzounis, Rodriguez-Mateos and Vulevic 196 ). Human dietary intervention with ellagitannins, which are polyphenols abundant in strawberries, raspberries and cloudberries, induced changes in the composition of Lachnospiraceae and Ruminococcaceae( Reference Puupponen-Pimiä, Seppänen-Laakso and Nohynek 128 ). Tea phenolics (e.g. epicatechin, catechin, gallic acid and caffeic acid) significantly repressed certain bacteria such as Clostridium perfringens and C. difficile and members of the Bacteroides spp., whereas bifidobacteria, Lactobacillus spp. and nonpathogenic Clostridium spp. were less severely affected( Reference Lee, Jenner and Low 197 ). Moreover, many phenolic compounds have in vitro antimicrobial activities towards pathogenic bacteria, such as Salmonella spp., C. perfringens, C. difficile, E. coli and Staphylococcus aureus ( Reference Lee, Jenner and Low 197 Reference Requena, Monagas and Pozo-Bayón 199 ).

Probiotics

Probiotics are live micro-organisms which when administered in adequate amounts confer a health benefit on the host, according to the widely accepted definition by Food and Agriculture Organisation WHO( Reference Araya, Morelli and Reid 200 ). Most of the currently used probiotics belong to the genera Bifidobacterium and Lactobacillus. However, probiotic preparations containing species of the genera Enterococcus, Pediococcus, Streptococcus, Lactococcus, Propionibacterium, Bacillus and Saccharomyces are also used( Reference Champagne, Ross and Saarela 201 ). The past two decades have seen a marked increase in the inclusion of probiotic bacteria in various types of food products, especially in fermented milks( Reference Daly and Davis 202 ). During recent years probiotics have also been increasingly incorporated into non-dairy foods such as fruit and berry juices and e.g. cereals( Reference Champagne, Ross and Saarela 201 ). In good quality products, the daily dose should be approximately 109 colony-forming units/d( Reference Donnet-Hughes, Rochat and Serrant 203 ). Probiotics do not usually colonise the GI tract, and therefore the products should be consumed daily for the health benefits( Reference Macfarlane and Cummings 167 ). In most of the studies, probiotics have not caused any significant changes in the predominant faecal microbiota of healthy adults. However, there are very few studies in which e.g. Lactobacillus rhamnosus GG has modulated the faecal microbiota and increased overall bacterial diversity in infants( Reference Rinne, Kalliomaki and Salminen 123 , Reference Cox, Huang and Fujimura 204 ). In addition, Bifidobacterium animalis subsp. lactis Bb12 has reduced the numbers of Enterobacteriaceae and Clostridium spp. in preterm infants( Reference Mohan, Koebnick and Schildt 124 ).

Conclusions

To answer the question posed in the title: does diet matter in regard to human microbiota? Yes, it does. From the host's perspective, there are numerous activities of the commensal gut microbiota that are of great importance to health. Moreover, by choosing what we eat, we can decide which bacteria we feed. However, even though diet matters, the results from dietary interventions are not always straight forward. It should be remembered that the detected effect is dependent on the study subjects, study protocols, used DNA-extraction techniques, used methodologies, and in case of next-generation sequencing also the algorithms used for cleaning and analysing the data. In addition, individual variation in human intestinal microbiota is so wide that the subtle changes may not be detected if the study cohort is not large enough. Therefore, studies in which different habitual diets have been compared with each other, usually get clearer correlations with nutrients v. bacteria than those observed in dietary interventions.

In order to get as much information from future dietary interventions there would be need for big enough group sizes, long enough trials, long enough wash-out periods in cross-over designs, sufficient background information (i.e. baseline 7-d food records and clinical parameters) and last but not least more interdisciplinary research across microbiology, nutrition, immunology, genetics, epigenetics, proteomics, transcriptomics, metabolomics and human physiology.

Financial Support

This work was supported by the EU-funded projects TORNADO (grant no. FP7-KBBE-222720) and ETHERPATHS (grant no. FP7-KBBE-222639). The funders had no role in the design, analysis or writing of the present paper.

Conflicts of Interest

None.

Authorship

J. M. drafted the manuscript and undertook the literature searches. M. S. reviewed the literature on polyphenols, proteins and fats. J. M. and M. S. reviewed and revised the manuscript.

References

1. Savage, DC (1977) Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol 131, 107133.CrossRefGoogle Scholar
2. Finegold, SM, Sutter, VL, Mathisen, GE (1983) Normal indigenous intestinal flora. In Human Intestinal Microflora in Health and Disease, pp. 331 [Hentges, DJ, editor]. New York, NY: Academic Press.Google Scholar
3. Franks, AH, Harmsen, HJM, Raangs, GC et al. (1998) Variations of bacterial populations in human feces measured by fluorescent in situ hybridization with group-specific 16S rRNA-targeted oligonucleotide probes. Appl Environ Microbiol 64, 33363345.CrossRefGoogle ScholarPubMed
4. Harmsen, HJM, Raangs, GC, He, T et al. (2002) Extensive set of 16S rRNA-based probes for detection of bacteria in human feces. Appl Environ Microbiol 68, 29822990.CrossRefGoogle ScholarPubMed
5. Marchesi, J & Shanahan, F (2007) The normal intestinal microbiota. Curr Opin Infect Dis 20, 508513.Google Scholar
6. Wilson, M (2008) Bacteriology of Humans. Oxford, UK: Blackwell Publishing.Google Scholar
7. Cummings, JH & Macfarlane, GT (1991) The control and consequences of bacterial fermentation in the human colon. J Appl Bacteriol 70, 443459.CrossRefGoogle ScholarPubMed
8. Stevens, J, VanSoest, PJ, Robertson, JB et al. (1987) Mean transit time measurement by analysis of a single stool after ingestion of multicolored plastic pellets. Am J Clin Nutr 46, 10481054.Google Scholar
9. Cummings, JH & Macfarlane, GT (1997) Colonic microflora: nutrition and health. Nutrition 13, 476478.Google Scholar
10. McNeil, NI (1984) The contribution of the large intestine to energy supplies in man. Am J Clin Nutr 39, 338342.Google Scholar
11. Rajilić-Stojanović, M, Smidt, H & De Vos, WM (2007) Diversity of the human gastrointestinal tract microbiota revisited. Environ Microbiol 9, 21252136.Google Scholar
12. Qin, J, Li, R, Raes, J et al. (2010) A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464, 5965.CrossRefGoogle ScholarPubMed
13. Zoetendal, EG, Akkermans, ADL & De Vos, WM (1998) Temperature gradient gel electrophoresis analysis of 16S rRNA from human fecal samples reveals stable and host-specific communities of active bacteria. Appl Environ Microbiol 64, 38543859.Google Scholar
14. Zoetendal, EG, Akkermans, ADL, Akkermans-van Vliet, WM et al. (2001) The host genotype affects the bacterial community in the human gastrointestinal tract. Microb Ecol Health Dis 13, 129134.Google Scholar
15. Vanhoutte, T, Huys, G, De Brandt, E et al. (2004) Temporal stability analysis of the microbiota in human feces by denaturing gradient gel electrophoresis using universal and group-specific 16S rRNA gene primers. FEMS Microbiol Ecol 48, 437446.Google Scholar
16. Seksik, P, Rigottier-Gois, L, Gramet, G et al. (2003) Alterations of the dominant faecal bacterial groups in patients with Crohn's disease of the colon. Gut 52, 237242.Google Scholar
17. Eckburg, PB, Bik, EM, Bernstein, CN et al. (2005) Microbiology: diversity of the human intestinal microbial flora. Science 308, 16351638.CrossRefGoogle ScholarPubMed
18. Suau, A, Bonnet, R, Sutren, M et al. (1999) Direct analysis of genes encoding 16S rRNA from complex communities reveals many novel molecular species within the human gut. Appl Environ Microbiol 65, 47994807.Google Scholar
19. Tap, J, Mondot, S, Levenez, F et al. (2009) Towards the human intestinal microbiota phylogenetic core. Environ Microbiol 11, 25742584.Google Scholar
20. Ley, RE, Turnbaugh, PJ, Klein, S et al. (2006) Microbial ecology: human gut microbes associated with obesity. Nature 444, 10221023.Google Scholar
21. Turnbaugh, PJ, Hamady, M, Yatsunenko, T et al. (2009) A core gut microbiome in obese and lean twins. Nature 457, 480484.CrossRefGoogle ScholarPubMed
22. Sghir, A, Gramet, G, Suau, A et al. (2000) Quantification of bacterial groups within human fecal flora by oligonucleotide probe hybridization. Appl Environ Microbiol 66, 22632266.Google Scholar
23. Lay, C, Rigottier-Gois, L, Holmstrøm, K et al. (2005) Colonic microbiota signatures across five northern European countries. Appl Environ Microbiol 71, 41534155.Google Scholar
24. Mueller, S, Saunier, K, Hanisch, C et al. (2006) Differences in fecal microbiota in different European study populations in relation to age, gender, and country: a cross-sectional study. Appl Environ Microbiol 72, 10271033.Google Scholar
25. Maukonen, J, Satokari, R, Mättö, J et al. (2006) Prevalence and temporal stability of selected clostridial groups in irritable bowel syndrome in relation to predominant faecal bacteria. J Med Microbiol 55, 625633.Google Scholar
26. Rochet, V, Rigottier-Gois, L, Rabot, S et al. (2004) Validation of fluorescent in situ hybridization combined with flow cytometry for assessing interindividual variation in the composition of human fecal microflora during long-term storage of samples. J Microbiol Methods 59, 263270.CrossRefGoogle ScholarPubMed
27. Rigottier-Gois, L, Le Bourhis, A, Gramet, G et al. (2003) Fluorescent hybridisation combined with flow cytometry and hybridisation of total RNA to analyse the composition of microbial communities in human faeces using 16S rRNA probes. FEMS Microbiol Ecol 43, 237245.CrossRefGoogle ScholarPubMed
28. Hold, GL, Schwiertz, A, Aminov, RI et al. (2003) Oligonucleotide probes that detect quantitatively significant groups of butyrate-producing bacteria in human feces. Appl Environ Microbiol 69, 43204324.Google Scholar
29. Zoetendal, EG, Ben-Amor, K, Harmsen, HJM et al. (2002) Quantification of uncultured Ruminococcus obeum-like bacteria in human fecal samples by fluorescent in situ hybridization and flow cytometry using 16S rRNA-targeted probes. Appl Environ Microbiol 68, 42254232.Google Scholar
30. Marteau, P, Pochart, P, Doré, J et al. (2001) Comparative study of bacterial groups within the human cecal and fecal microbiota. Appl Environ Microbiol 67, 49394942.Google Scholar
31. Lay, C, Sutren, M, Rochet, V et al. (2005) Design and validation of 16S rRNA probes to enumerate members of the Clostridium leptum subgroup in human faecal microbiota. Environ Microbiol 7, 933946.Google Scholar
32. Saunier, K, Rouge, C, Lay, C et al. (2005) Enumeration of bacteria from the Clostridium leptum subgroup in human faecal microbiota using Clep1156 16S rRNA probe in combination with helper and competitor oligonucleotides. Syst Appl Microbiol 28, 454464.CrossRefGoogle ScholarPubMed
33. Doré, J, Schir, A, Hannequart-Gramet, G et al. (1998) Design and evaluation of a 16S rRNA-targeted oligonucleotide probe for specific detection and quantitation of human faecal Bacteroides populations. Syst Appl Microbiol 21, 6571.Google Scholar
34. Rigottier-Gois, L, Rochet, V, Garrec, N et al. (2003) Enumeration of Bacteroides species in human faeces by fluorescent in situ hybridisation combined with flow cytometry using 16S rRNA probes. Syst Appl Microbiol 26, 110118.Google Scholar
35. Shah, HN (1992) The genus bacteroides and related taxa. In The Prokaryotes, 2nd ed., pp. 3593–607 [Balows, A, Trüper, HG, Dworkin, M, Harder, W, Schleifer, K-H, editors]. New York, NY: Springer-Verlag.CrossRefGoogle Scholar
36. Collado, MC, Derrien, M, Isolauri, E et al. (2007) Intestinal integrity and Akkermansia muciniphila, a mucin-degrading member of the intestinal microbiota present in infants, adults, and the elderly. Appl Environ Microbiol 73, 77677770.Google Scholar
37. Derrien, M, Collado, MC, Ben-Amor, K et al. (2008) The mucin degrader Akkermansia muciniphila is an abundant resident of the human intestinal tract. Appl Environ Microbiol 74, 16461648.Google Scholar
38. Harmsen, HJM, Wildeboer-Veloo, ACM, Grijpstra, J et al. (2000) Development of 16S rRNA-based probes for the Coriobacterium group and the Atopobium cluster and their application for enumeration of Coriobacteriaceae in human feces from volunteers of different age groups. Appl Environ Microbiol 66, 45234527.CrossRefGoogle ScholarPubMed
39. Gibson, GR, Macfarlane, GT & Cummings, JH (1988) Occurrence of sulphate-reducing bacteria in human faeces and the relationship of dissimilatory sulphate reduction to methanogenesis in the large gut. J Appl Bacteriol 65, 103111.Google Scholar
40. Reyes, A, Haynes, M, Hanson, N et al. (2010) Viruses in the faecal microbiota of monozygotic twins and their mothers. Nature 466, 334338.Google Scholar
41. Doré, J, Gramet, G, Goderel, I et al. (1998) Culture-independent characterisation of human faecal flora using rRNA-targeted hybridisation probes. Genet Sel Evol 30, Suppl., S287S296.Google Scholar
42. Roesch, LF, Casella, G, Simell, O et al. (2009) Influence of fecal sample storage on bacterial community diversity. Open Microbiol J 3, 4046.CrossRefGoogle ScholarPubMed
43. Ott, SJ, Musfeldt, M, Timmis, KN et al. (2004) In vitro alterations of intestinal bacterial microbiota in fecal samples during storage. Diag Microbiol Infect Dis 50, 237245.Google Scholar
44. Molbak, L, Sommer, HM, Johnsen, K et al. (2006) Freezing at −80 °C distorts the DNA composition of bacterial communities in intestinal samples. Curr Issues Intest Microbiol 7, 2934.Google Scholar
45. Kennedy, NA, Walker, AW, Berry, SH et al. (2014) The impact of different DNA extraction kits and laboratories upon the assessment of human gut microbiota composition by 16S rRNA gene sequencing. PLoS ONE 9, e88982.Google Scholar
46. Dridi, B, Henry, M, El Khéchine, A et al. (2009) High prevalence of Methanobrevibacter smithii and Methanosphaera stadtmanae detected in the human gut using an improved DNA detection protocol. PLoS ONE 4, e7063.CrossRefGoogle ScholarPubMed
47. Maukonen, J, Simões, C & Saarela, M (2012) The currently used commercial DNA extraction methods give different results of clostridial and Actinobacterial populations derived from human fecal samples. FEMS Microbiol Ecol 79, 697708.CrossRefGoogle ScholarPubMed
48. Salonen, A, Nikkilä, J, Jalanka-Tuovinen, J et al. (2010) Comparative analysis of fecal DNA extraction methods with phylogenetic microarray: effective recovery of bacterial and archaeal DNA using mechanical cell lysis. J Microbiol Methods 81, 127134.Google Scholar
49. Krogius-Kurikka, L, Kassinen, A, Paulin, L et al. (2009) Sequence analysis of percent G + C fraction libraries of human faecal bacterial DNA reveals a high number of Actinobacteria. BMC Microbiol 9, 68.Google Scholar
50. Nakamura, N, Gaskins, HR, Collier, CT et al. (2009) Molecular ecological analysis of fecal bacterial populations from term infants fed formula supplemented with selected blends of prebiotics. Appl Environ Microbiol 75, 11211128.Google Scholar
51. Matsuki, T, Watanabe, K, Fujimoto, J et al. (2004) Use of 16S rRNA gene-targeted group-specific primers for real-time PCR analysis of predominant bacteria in human feces. Appl Environ Microbiol 70, 72207228.Google Scholar
52. Hoyles, L & McCartney, AL (2009) What do we mean when we refer to Bacteroidetes populations in the human gastrointestinal microbiota? FEMS Microbiol Lett 299, 175183.Google Scholar
53. Walker, AW, Duncan, SH, Louis, P et al. (2014) Phylogeny, culturing, and metagenomics of the human gut microbiota. Trends Microbiol 22, 267274.Google Scholar
54. Kuczynski, J, Lauber, CL, Walters, WA et al. (2011) Experimental and analytical tools for studying the human microbiome. Nat Rev Genet 13, 4758.Google Scholar
55. Corry, JE, Jarvis, B, Passmore, S et al. (2007) A critical review of measurement uncertainty in the enumeration of food micro-organisms. Food Microbiol 24, 230253.Google Scholar
56. Ramette, A (2007) Multivariate analyses in microbial ecology. FEMS Microbiol Ecol 62, 142160.Google Scholar
57. Rudi, K, Zimonja, M, Trosvik, P et al. (2007) Use of multivariate statistics for 16S rRNA gene analysis of microbial communities. Int J Food Microbiol 120, 9599.Google Scholar
58. Bent, SJ & Forney, LJ (2008) The tragedy of the uncommon: understanding limitations in the analysis of microbial diversity. ISME J 2, 689695.Google Scholar
59. Lozupone, CA, Hamady, M, Kelley, ST et al. (2007) Quantitative and qualitative beta diversity measures lead to different insights into factors that structure microbial communities. Appl Environ Microbiol 73, 15761585.Google Scholar
60. Werner, JJ, Koren, O, Hugenholtz, P et al. (2012) Impact of training sets on classification of high-throughput bacterial 16s rRNA gene surveys. ISME J 6, 94103.CrossRefGoogle ScholarPubMed
61. Achtman, M & Wagner, M (2008) Microbial diversity and the genetic nature of microbial species. Nat Rev Microbiol 6, 431440.Google Scholar
62. Turnbaugh, PJ, Quince, C, Faith, JJ et al. (2010) Organismal, genetic, and transcriptional variation in the deeply sequenced gut microbiomes of identical twins. Proc Natl Acad Sci USA 107, 75037508.Google Scholar
63. Stewart, JA, Chadwick, VS & Murray, A (2005) Investigations into the influence of host genetics on the predominant eubacteria in the faecal microflora of children. J Med Microbiol 54, 12391242.Google Scholar
64. Simoes, CD, Maukonen, J, Kaprio, J et al. (2013) Habitual dietary intake is associated with the stool microbiota composition of Finnish monozygotic twins. J Nutr 143, 417423.Google Scholar
65. Fallani, M, Young, D, Scott, J et al. (2010) Intestinal microbiota of 6-week-old infants across Europe: geographic influence beyond delivery mode, breast-feeding, and antibiotics. J Pediatr Gastroenterol Nutr 51, 7784.CrossRefGoogle ScholarPubMed
66. Yatsunenko, T, Rey, FE, Manary, MJ et al. (2012) Human gut microbiome viewed across age and geography. Nature 486, 222227.Google Scholar
67. Li, M, Wang, B, Zhang, M et al. (2008) Symbiotic gut microbes modulate human metabolic phenotypes. Proc Natl Acad Sci USA 105, 21172122.Google Scholar
68. De Filippo, C, Cavalieri, D, Di Paola, M et al. (2010) Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci USA 107, 1469114696.CrossRefGoogle ScholarPubMed
69. Fallani, M, Amarri, S, Uusijarvi, A et al. (2011) Determinants of the human infant intestinal microbiota after the introduction of first complementary foods in infant samples from five European centres. Microbiology 157, 13851392.Google Scholar
70. Rougé, C, Goldenberg, O, Ferraris, L et al. (2010) Investigation of the intestinal microbiota in preterm infants using different methods. Anaerobe 16, 362370.Google Scholar
71. Penders, J, Thijs, C, Vink, C et al. (2006) Factors influencing the composition of the intestinal microbiota in early infancy. Pediatrics 118, 511521.Google Scholar
72. Alm, JS, Swartz, J, Björkstén, B et al. (2002) An anthroposophic lifestyle and intestinal microflora in infancy. Pediatr Allergy Immunol 13, 402411.Google Scholar
73. Schwiertz, A, Gruhl, B, Löbnitz, M et al. (2003) Development of the intestinal bacterial composition in hospitalized preterm infants in comparison with breast-fed, full-term infants. Pediatr Res 54, 393399.CrossRefGoogle ScholarPubMed
74. Bezirtzoglou, E, Tsiotsias, A & Welling, GW (2011) Microbiota profile in feces of breast- and formula-fed newborns by using fluorescence in situ hybridization (FISH). Anaerobe 17, 478482.Google Scholar
75. Favier, CF, De Vos, WM & Akkermans, ADL (2003) Development of bacterial and bifidobacterial communities in feces of newborn babies. Anaerobe 9, 219229.Google Scholar
76. Palmer, C, Bik, EM, DiGiulio, DB et al. (2007) Development of the human infant intestinal microbiota. PLoS Biol 5, 15561573.Google Scholar
77. Favier, CF, Vaughan, EE, de Vos, WM et al. (2002) Molecular monitoring of succession of bacterial communities in human neonates. Appl Environ Microbiol 68, 219226.Google Scholar
78. Rotimi, VO & Duerden, BI (1981) The development of the bacterial flora in normal neonates. J Med Microbiol 14, 5162.Google Scholar
79. Songjinda, P, Nakayama, J, Kuroki, Y et al. (2005) Molecular monitoring of the developmental bacterial community in the gastrointestinal tract of Japanese infants. Biosci Biotechnol Biochem 69, 638641.CrossRefGoogle ScholarPubMed
80. Wang, M, Ahrne, S, Antonsson, M et al. (2004) T-RFLP combined with principal component analysis and 16S rRNA gene sequencing: an effective strategy for comparison of fecal microbiota in infants of different ages. J Microbiol Methods 59, 5369.CrossRefGoogle ScholarPubMed
81. Dominguez-Bello, MG, Costello, EK, Contreras, M et al. (2010) Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA 107, 1197111975.Google Scholar
82. Magne, F, Abély, M, Boyer, F et al. (2006) Low species diversity and high interindividual variability in faeces of preterm infants as revealed by sequences of 16S rRNA genes and PCR-temporal temperature gradient gel electrophoresis profiles. FEMS Microbiol Ecol 57, 128138.Google Scholar
83. Arboleya, S, Binetti, A, Salazar, N et al. (2012) Establishment and development of intestinal microbiota in preterm neonates. FEMS Microbiol Ecol 79, 763772.Google Scholar
84. Hall, MA, Cole, CB, Smith, SL et al. (1990) Factors influencing the presence of faecal lactobacilli in early infancy. Arch Dis Child 65, 185188.Google Scholar
85. Bullen, CL, Tearle, PV & Willis, AT (1976) Bifidobacteria in the intestinal tract of infants: an in vivo study. J Med Microbiol 9, 325333.Google Scholar
86. Mariat, D, Firmesse, O, Levenez, F et al. (2009) The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol 9, 123.Google Scholar
87. Harmsen, HJM, Wildeboer-Veloo, ACM, Raangs, GC et al. (2000) Analysis of intestinal flora development in breast-fed and formula-fed infants by using molecular identification and detection methods. J Pediatr Gastroenterol Nutr 30, 6167.Google Scholar
88. Magne, F, Hachelaf, W, Suau, A et al. (2006) A longitudinal study of infant faecal microbiota during weaning. FEMS Microbiol Ecol 58, 563571.Google Scholar
89. Penders, J, Vink, C, Driessen, C et al. (2005) Quantification of Bifidobacterium spp., Escherichia coli and Clostridium difficile in faecal samples of breast-fed and formula-fed infants by real-time PCR. FEMS Microbiol Lett 243, 141147.Google Scholar
90. Stark, PL & Lee, A (1982) The microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of life. J Med Microbiol 15, 189203.Google Scholar
91. Hentges, DJ (1993) The anaerobic microflora of the human body. Clin Infect Dis 16, Suppl. 4, S175S180.Google Scholar
92. Gorbach, SL, Nahas, L, Lerner, PI et al. (1967) Studies of intestinal microflora. I. Effects of diet, age, and periodic sampling on numbers of fecal microorganisms in man. Gastroenterology 53, 845855.CrossRefGoogle ScholarPubMed
93. Rajilic-Stojanovic, M, Heilig, HG, Molenaar, D et al. (2009) Development and application of the human intestinal tract chip, a phylogenetic microarray: analysis of universally conserved phylotypes in the abundant microbiota of young and elderly adults. Environ Microbiol 11, 17361751.Google Scholar
94. Claesson, MJ, Cusack, S, O'Sullivan, O et al. (2011) Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci USA 108, Suppl. 1, 45864591.Google Scholar
95. Woodmansey, EJ, McMurdo, ME, Macfarlane, GT et al. (2004) Comparison of compositions and metabolic activities of fecal microbiotas in young adults and in antibiotic-treated and non-antibiotic-treated elderly subjects. Appl Environ Microbiol 70, 61136122.Google Scholar
96. Blaut, M, Collins, MD, Welling, GW et al. (2002) Molecular biological methods for studying the gut microbiota: the EU human gut flora project. Br J Nutr 87, Suppl. 2, S203S211.Google Scholar
97. Maukonen, J, Mättö, J, Kajander, K et al. (2008) Diversity and temporal stability of fecal bacterial populations in elderly subjects consuming galacto-oligosaccharide containing probiotic yoghurt. Int Dairy J 18, 386395.Google Scholar
98. Claesson, MJ, Jeffery, IB, Conde, S et al. (2012) Gut microbiota composition correlates with diet and health in the elderly. Nature 488, 178184.Google Scholar
99. Kohanski, MA, Dwyer, DJ & Collins, JJ (2010) How antibiotics kill bacteria: from targets to networks. Nat Rev Microbiol 8, 423435.Google Scholar
100. Sullivan, A, Edlund, C & Nord, CE (2001) Effect of antimicrobial agents on the ecological balance of human microflora. Lancet Infect Dis 1, 101114.CrossRefGoogle ScholarPubMed
101. Delgado, S, Flórez, AB & Mayo, B (2005) Antibiotic susceptibility of Lactobacillus and Bifidobacterium species from the human gastrointestinal tract. Curr Microbiol 50, 202207.Google Scholar
102. Jousimies-Somer, HR, Summanen, P, Citron, DM et al. (2002) Wadsworth – KTL Anaerobic Bacteriology Manual , 6th ed. Belmont, CA: Star Publishing Company.Google Scholar
103. Mättö, J, van Hoek, AHAM, Domig, KJ et al. (2007) Susceptibility of human and probiotic Bifidobacterium spp. to selected antibiotics as determined by the Etest method. Int Dairy J 17, 11231131.Google Scholar
104. Moubareck, C, Gavini, F, Vaugien, L et al. (2005) Antimicrobial susceptibility of bifidobacteria. J Antimicrob Chemother 55, 3844.Google Scholar
105. Saarela, M, Mayrhofer, S, Domig, KJ et al. (2008) Susceptibility of bifidobacteria originating from different sources to tetracycline, erythromycin, streptomycin and vancomycin. Int J Prob Preb 3, 269270.Google Scholar
106. Huys, G, D'Haene, K, Cnockaert, M et al. (2010) Intra- and interlaboratory performances of two commercial antimicrobial susceptibility testing methods for bifidobacteria and nonenterococcal lactic acid bacteria. Antimicrob Agents Chemother 54, 25672574.Google Scholar
107. Louis, P, Scott, KP, Duncan, SH et al. (2007) Understanding the effects of diet on bacterial metabolism in the large intestine. J Appl Microbiol 102, 11971208.Google Scholar
108. Duncan, SH, Louis, P, Thomson, JM et al. (2009) The role of pH in determining the species composition of the human colonic microbiota. Environ Microbiol 11, 21122122.Google Scholar
109. Walker, AW, Duncan, SH, Carol McWilliam Leitch, E et al. (2005) pH and peptide supply can radically alter bacterial populations and short-chain fatty acid ratios within microbial communities from the human colon. Appl Environ Microbiol 71, 36923700.Google Scholar
110. Wade, WG (2006) The genus Eubacterium and related genera. In Prokaryotes, pp. 823835 [Stackebrandt, E, Jones, D, Holzapfel, W, Reboli, A, Farrar, W, editors]. New York, NY: Springer-Verlag.Google Scholar
111. Wexler, HM (2007) Bacteroides: the good, the bad, and the nitty-gritty. Clin Microbiol Rev 20, 593621.Google Scholar
112. Shah, HN & Gharbia, SE (1993) Ecophysiology and taxonomy of Bacteroides and related taxa. Clin Infect Dis 16, Suppl. 4, S160S167.Google Scholar
113. Karlsson, FH, Ussery, DW, Nielsen, J et al. (2011) A closer look at Bacteroides: phylogenetic relationship and genomic implications of a life in the human gut. Microb Ecol 61, 473485.CrossRefGoogle Scholar
114. Derrien, M, Vaughan, EE, Plugge, CM et al. (2004) Akkermansia municiphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol 54, 14691476.Google Scholar
115. Belenguer, A, Duncan, SH, Calder, AG et al. (2006) Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut. Appl Environ Microbiol 72, 35933599.CrossRefGoogle ScholarPubMed
116. Falony, G, Vlachou, A, Verbrugghe, K et al. (2006) Cross-feeding between Bifidobacterium longum BB536 and acetate-converting, butyrate-producing colon bacteria during growth on oligofructose. Appl Environ Microbiol 72, 78357841.Google Scholar
117. Hehemann, JH, Correc, G, Barbeyron, T et al. (2010) Transfer of carbohydrate-active enzymes from marine bacteria to Japanese gut microbiota. Nature 464, 908912.Google Scholar
118. Hentges, DJ, Maier, BR, Burton, GC et al. (1977) Effect of a high-beef diet on the fecal bacterial flora of humans. Cancer Res 37, 568571.Google Scholar
119. Russell, WR, Gratz, SW, Duncan, SH et al. (2011) High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health. Am J Clin Nutr 93, 10621072.Google Scholar
120. Tuohy, KM, Kolida, S, Lustenberger, AM et al. (2001) The prebiotic effects of biscuits containing partially hydrolysed guar gum and fructo-oligosaccharides-a human volunteer study. Br J Nutr 86, 341348.Google Scholar
121. Walton, GE, van den Heuvel, EG, Kosters, MH et al. (2012) A randomised crossover study investigating the effects of galacto-oligosaccharides on the faecal microbiota in men and women over 50 years of age. Br J Nutr 107, 14661475.CrossRefGoogle ScholarPubMed
122. Langlands, SJ, Hopkins, MJ, Coleman, N et al. (2004) Prebiotic carbohydrates modify the mucosa associated microflora of the human large bowel. Gut 53, 16101616.Google Scholar
123. Rinne, M, Kalliomaki, M, Salminen, S et al. (2006) Probiotic intervention in the first months of life: short-term effects on gastrointestinal symptoms and long-term effects on gut microbiota. J Pediatr Gastroenterol Nutr 43, 200205.Google Scholar
124. Mohan, R, Koebnick, C, Schildt, J et al. (2006) Effects of Bifidobacterium lactis Bb12 supplementation on intestinal microbiota of preterm infants: a double-blind, placebo-controlled, randomized study. J Clin Microbiol 44, 40254031.Google Scholar
125. Lyra, A, Krogius-Kurikka, L, Nikkila, J et al. (2010) Effect of a multispecies probiotic supplement on quantity of irritable bowel syndrome-related intestinal microbial phylotypes. BMC Gastroenterol 10, 110.Google Scholar
126. Duncan, SH, Lobley, GE, Holtrop, G et al. (2008) Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes 32, 17201724.Google Scholar
127. Nadal, I, Santacruz, A, Marcos, A et al. (2009) Shifts in clostridia, bacteroides and immunoglobulin-coating fecal bacteria associated with weight loss in obese adolescents. Int J Obes 33, 758767.Google Scholar
128. Puupponen-Pimiä, R, Seppänen-Laakso, T, Nohynek, L et al. (2013) Effects of ellagitannin rich berries on blood lipid profiles, gut microbiota and metabolism of phenolic compounds in metabolic syndrome. Mol. Nutr Food Res 57, 22582263.Google Scholar
129. Duncan, SH, Belenguer, A, Holtrop, G et al. (2007) Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Appl Environ Microbiol 73, 10731078.CrossRefGoogle ScholarPubMed
130. Flint, HJ, Duncan, SH, Scott, KP et al. (2007) Interactions and competition within the microbial community of the human colon: links between diet and health: minireview. Environ Microbiol 9, 11011111.Google Scholar
131. Drasar, BS, Crowther, JS, Goddard, P et al. (1973) The relation between diet and the gut microflora in man. Proc Nutr Soc 32, 4952.Google Scholar
132. Finegold, SM, Attebery, HR & Sutter, VL (1974) Effect of diet on human fecal flora: comparison of Japanese and American diets. Am J Clin Nutr 27, 14561469.Google Scholar
133. Drasar, BS, Montgomery, F & Tomkins, AM (1986) Diet and faecal flora in three dietary groups in rural northern Nigeria. J Hyg (Lond) 96, 5965.Google Scholar
134. Wu, GD, Chen, J, Hoffmann, C et al. (2011) Linking long-term dietary patterns with gut microbial enterotypes. Science 334, 105108.Google Scholar
135. Hayashi, H, Sakamoto, M & Benno, Y (2002) Fecal microbial diversity in a strict vegetarian as determined by molecular analysis and cultivation. Microbiol Immunol 46, 819831.Google Scholar
136. Hippe, B, Zwielehner, J, Liszt, K et al. (2011) Quantification of butyryl CoA:acetate CoA-transferase genes reveals different butyrate production capacity in individuals according to diet and age. FEMS Microbiol Lett 316, 130135.CrossRefGoogle ScholarPubMed
137. Zimmer, J, Lange, B, Frick, JS et al. (2012) A vegan or vegetarian diet substantially alters the human colonic faecal microbiota. Eur J Clin Nutr 66, 5360.Google Scholar
138. Finegold, SM, Flora, DJ, Attebery, HR et al. (1975) Fecal bacteriology of colonic polyp patients and control patients. Cancer Res 35, 34073417.Google Scholar
139. Finegold, SM, Sutter, VL & Sugihara, PT (1977) Fecal microbial flora in Seventh Day Adventist populations and control subjects. Am J Clin Nutr 30, 17811792.Google Scholar
140. Bartosch, S, Fite, A, Macfarlane, GT et al. (2004) Characterization of bacterial communities in feces from healthy elderly volunteers and hospitalized elderly patients by using real-time PCR and effects of antibiotic treatment on the fecal microbiota. Appl Environ Microbiol 70, 35753581.CrossRefGoogle ScholarPubMed
141. Blaut, M (2002) Relationship of prebiotics and food to intestinal microflora. Eur J Nutr 41, Suppl. 1, 1116.Google Scholar
142. Flint, H, Scott, K, Duncan, S et al. (2012) Microbial degradation of complex carbohydrates in the gut. Gut Microbes 3, 289306.Google Scholar
143. Ze, X, Duncan, SH, Louis, P et al. (2012) Ruminococcus bromii is a keystone species for the degradation of resistant starch in the human colon. ISME J 6, 15351543.Google Scholar
144. Flint, HJ, Bayer, EA, Rincon, MT et al. (2008) Polysaccharide utilization by gut bacteria: potential for new insights from genomic analysis. Nat Rev Microbiol 6, 121131.Google Scholar
145. Russell, WR, Hoyles, L, Flint, HJ et al. (2013) Colonic bacterial metabolites and human health. Curr Opin Microbiol 16, 246254.Google Scholar
146. Scott, KP, Gratz, SW, Sheridan, PO et al. (2013) The influence of diet on the gut microbiota. Pharmacol Res 69, 5260.Google Scholar
147. Flint, HJ (2012) The impact of nutrition on the human microbiome. Nutr Rev 70, Suppl. 1, S10S13.Google Scholar
148. Aminov, RI, Walker, AW, Duncan, SH et al. (2006) Molecular diversity, cultivation, and improved detection by fluorescent in situ hybridization of a dominant group of human gut bacteria related to Roseburia spp. or Eubacterium rectale . Appl Environ Microbiol 72, 63716376.Google Scholar
149. Barcenilla, A, Pryde, SE, Martin, JC et al. (2000) Phylogenetic relationships of butyrate-producing bacteria from the human gut. Appl Environ Microbiol 66, 16541661.Google Scholar
150. Pryde, SE, Duncan, SH, Hold, GL et al. (2002) The microbiology of butyrate formation in the human colon. FEMS Microbiol Lett 217, 133139.Google Scholar
151. Benus, RF, van der Werf, TS, Welling, GW et al. (2010) Association between Faecalibacterium prausnitzii and dietary fibre in colonic fermentation in healthy human subjects. Br J Nutr 104, 693700.Google Scholar
152. Marquet, P, Duncan, SH, Chassard, C et al. (2009) Lactate has the potential to promote hydrogen sulphide formation in the human colon. FEMS Microbiol Lett 299, 128134.Google Scholar
153. Karppinen, S, Liukkonen, K, Aura, A-M et al. (2000) In vitro fermentation of polysaccharides of rye, wheat and oat brans and inulin by human faecal bacteria. J Sci Food Agric 80, 14691476.Google Scholar
154. Hughes, SA, Shewry, PR, Gibson, GR et al. (2008) In vitro fermentation of oat and barley derived β-glucans by human faecal microbiota. FEMS Microbiol Ecol 64, 482493.Google Scholar
155. Hopkins, MJ, Englyst, HN, Macfarlane, S et al. (2003) Degradation of cross-linked and non-cross-linked arabinoxylans by the intestinal microbiota in children. Appl Environ Microbiol 69, 63546360.Google Scholar
156. Broekaert, WF, Courtin, CM, Verbeke, K et al. (2011) Prebiotic and other health-related effects of cereal-derived arabinoxylans, arabinoxylan-oligosaccharides, and xylooligosaccharides. Crit Rev Food Sci Nutr 51, 178194.Google Scholar
157. Zhao, J & Cheung, PC (2011) Fermentation of beta-glucans derived from different sources by bifidobacteria: evaluation of their bifidogenic effect. J Agric Food Chem 59, 59865992.Google Scholar
158. De Vuyst, L & Leroy, F (2011) Cross-feeding between bifidobacteria and butyrate-producing colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas production. Int J Food Microbiol 149, 7380.Google Scholar
159. Martinez, I, Kim, J, Duffy, PR et al. (2010) Resistant starches types 2 and 4 have differential effects on the composition of the fecal microbiota in human subjects. PLoS ONE 5, e15046.Google Scholar
160. Walker, AW, Ince, J, Duncan, SH et al. (2011) Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J 5, 220230.Google Scholar
161. Abell, GCJ, Cooke, CM, Bennett, CN et al. (2008) 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 66, 505515.Google Scholar
162. Ehle, FR, Robertson, JB & Van Soest, PJ (1982) Influence of dietary fibers on fermentation in the human large intestine. J Nutr 112, 158166.Google Scholar
163. Robert, C & Bernalier-Donadille, A (2003) The cellulolytic microflora of the human colon: evidence of microcrystalline cellulose-degrading bacteria in methane-excreting subjects. FEMS Microbiol Ecol 46, 8189.Google Scholar
164. Connolly, ML, Lovegrove, JA & Tuohy, KM (2010) In vitro evaluation of the microbiota modulation abilities of different sized whole oat grain flakes. Anaerobe 16, 483488.Google Scholar
165. Leitch, EC, Walker, AW, Duncan, SH et al. (2007) Selective colonization of insoluble substrates by human faecal bacteria. Environ Microbiol 9, 667679.Google Scholar
166. Gibson, GR, Probert, HM, Van Loo, J et al. (2004) Dietary modulation of the human colonic microbiota: updating the concept of prebiotics. Nutr Res Rev 17, 259275.Google Scholar
167. Macfarlane, GT & Cummings, JH (1993) Probiotics and prebiotics: can regulating the activities of intestinal bacteria benefit health? Br Med J 318, 9991003.Google Scholar
168. Tannock, GW, Munro, K, Bibiloni, R et al. (2004) Impact of consumption of oligosaccharide-containing biscuits on the fecal microbiota of humans. Appl Environ Microbiol 70, 21292136.Google Scholar
169. Davis, LM, Martinez, I, Walter, J et al. (2011) Barcoded pyrosequencing reveals that consumption of galactooligosaccharides results in a highly specific bifidogenic response in humans. PLoS ONE 6, e25200.Google Scholar
170. Ramirez-Farias, C, Slezak, K, Fuller, Z et al. (2009) Effect of inulin on the human gut microbiota: stimulation of Bifidobacterium adolescentis and Faecalibacterium prausnitzii . Br J Nutr 101, 541550.Google Scholar
171. Roberfroid, M, Gibson, GR, Hoyles, L et al. (2010) Prebiotic effects: metabolic and health benefits. Br J Nutr 104, Suppl. 2, S1S63.Google Scholar
172. Louis, P, Young, P, Holtrop, G et al. (2010) Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene. Environ Microbiol 12, 304314.Google Scholar
173. Rinne, MM, Gueimonde, M, Kalliomaki, M et al. (2005) Similar bifidogenic effects of prebiotic-supplemented partially hydrolyzed infant formula and breastfeeding on infant gut microbiota. FEMS Immunol Med Microbiol 43, 5965.Google Scholar
174. Cani, PD (2012) Crosstalk between the gut microbiota and the endocannabinoid system: impact on the gut barrier function and the adipose tissue. Clin Microbiol Infect 18, Suppl. 4, 5053.Google Scholar
175. Alberts, B, Johnson, A, Lewis, J et al. (2002) Molecular Biology of the Cell, 4th ed. New York: Garland Science.Google Scholar
176. Silk, DB (1980) Digestion and absorption of dietary protein in man. Proc Nutr Soc 39, 6170.Google Scholar
177. Smith, EA & Macfarlane, GT (1997) Dissimilatory amino Acid metabolism in human colonic bacteria. Anaerobe 3, 327337.Google Scholar
178. Magee, EA, Richardson, CJ, Hughes, R et al. (2000) 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 72, 14881494.Google Scholar
179. Pitcher, MC & Cummings, JH (1996) Hydrogen sulphide: a bacterial toxin in ulcerative colitis? Gut 39, 14.Google Scholar
180. World Cancer Research Fund & American Institute for Cancer Research (2011). Continuous Update. Project Report. Food, Nutrition, Physical Activity, and the Prevention of Colorectal Cancer.Google Scholar
181. Ortega, A, Varela, LM, Bermudez, B et al. (2012) Dietary fatty acids linking postprandial metabolic response and chronic diseases. Food Funct 3, 2227.Google Scholar
182. Calder, PC, Ahluwalia, N, Brouns, F et al. (2011) Dietary factors and low-grade inflammation in relation to overweight and obesity. Br J Nutr 106, Suppl. 3, S5S78.Google Scholar
183. Fava, F, Gitau, R, Griffin, BA et al. (2013) The type and quantity of dietary fat and carbohydrate alter faecal microbiome and short-chain fatty acid excretion in a metabolic syndrome ‘at-risk’ population. Int J Obes 37, 216223.Google Scholar
184. Brinkworth, GD, Noakes, M, Clifton, PM et al. (2009) 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 101, 14931502.Google Scholar
185. Pachikian, BD, Neyrinck, AM, Portois, L et al. (2011) Involvement of gut microbial fermentation in the metabolic alterations occurring in n-3 polyunsaturated fatty acids-depleted mice. Nutr Metab (Lond) 8, 44.Google Scholar
186. Santacruz, A, Marcos, A, Wärnberg, J et al. (2009) Interplay between weight loss and gut microbiota composition in overweight adolescents. Obesity 17, 19061915.Google Scholar
187. Willemsen, LE, Koetsier, MA, Balvers, M et al. (2008) Polyunsaturated fatty acids support epithelial barrier integrity and reduce IL-4 mediated permeability in vitro . Eur J Nutr 47, 183191.Google Scholar
188. Anderson, RC, Cookson, AL, McNabb, WC et al. (2010) Lactobacillus plantarum MB452 enhances the function of the intestinal barrier by increasing the expression levels of genes involved in tight junction formation. BMC Microbiol 10, 316.Google Scholar
189. Donato, KA, Gareau, MG, Wang, YJ et al. (2010) Lactobacillus rhamnosus GG attenuates interferon-{gamma} and tumour necrosis factor-alpha-induced barrier dysfunction and pro-inflammatory signalling. Microbiology 156, 32883297.Google Scholar
190. Urwin, HJ, Miles, EA, Noakes, PS et al. (2014) Effect of salmon consumption during pregnancy on maternal and infant faecal microbiota, secretory IgA and calprotectin. Br J Nutr 111, 773784.Google Scholar
191. Harbige, LS (2003) Fatty acids, the immune response, and autoimmunity: a question of n-6 essentiality and the balance between n-6 and n-3. Lipids 38, 323341.Google Scholar
192. Medina, M, Izquierdo, E, Ennahar, S et al. (2007) Differential immunomodulatory properties of Bifidobacterium logum strains: relevance to probiotic selection and clinical applications. Clin Exp Immunol 150, 531538.Google Scholar
193. Aura, A-M (2008) Microbial metabolism of dietary phenolic compounds in the colon. Phytochem Rev 7, 407429.Google Scholar
194. Tuohy, KM, Conterno, L, Gasperotti, M et al. (2012) Up-regulating the human intestinal microbiome using whole plant foods, polyphenols, and/or fiber. J Agric Food Chem 60, 87768782.Google Scholar
195. Queipo-Ortuno, MI, Boto-Ordonez, M, Murri, M et al. (2012) Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am J Clin Nutr 95, 13231334.Google Scholar
196. Tzounis, X, Rodriguez-Mateos, A, Vulevic, J et al. (2011) Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study. Am J Clin Nutr 93, 6272.Google Scholar
197. Lee, HC, Jenner, AM, Low, CS et al. (2006) Effect of tea phenolics and their aromatic fecal bacterial metabolites on intestinal microbiota. Res Microbiol 157, 876884.Google Scholar
198. Alakomi, HL, Puupponen-Pimia, R, Aura, A-M et al. (2007) Weakening of salmonella with selected microbial metabolites of berry-derived phenolic compounds and organic acids. J Agric Food Chem 55, 39053912.Google Scholar
199. Requena, T, Monagas, M, Pozo-Bayón, MA et al. (2010) Perspectives of the potential implications of wine polyphenols on human oral and gut microbiota. Trends Food Sci Technol 21, 332344.Google Scholar
200. Araya, M, Morelli, L, Reid, G et al. (2002) Guidelines for the evaluation of probiotics in food. Joint FAO/WHO Working Group Report on Drafting Guidelines for the Evaluation of Probiotics in Food, London (ON, Canada) April 30 and May 1.Google Scholar
201. Champagne, CP, Ross, RP, Saarela, M et al. (2011) Recommendations for the viability assessment of probiotics as concentrated cultures and in food matrices. Int J Food Microbiol 149, 185193.Google Scholar
202. Daly, C & Davis, R (1998) The biotechnology of lactic acid bacteria with emphasis on applications in food safety and human health. Agric Food Sci Finl 7, 251265.Google Scholar
203. Donnet-Hughes, A, Rochat, F, Serrant, P et al. (1999) Modulation of nonspecific mechanisms of defense by lactic acid bacteria: effective dose. J Dairy Sci 82, 863869.Google Scholar
204. Cox, MJ, Huang, YJ, Fujimura, KE et al. (2010) Lactobacillus casei abundance is associated with profound shifts in the infant gut microbiome. PLoS ONE 5, e8745.Google Scholar