Hostname: page-component-7c8c6479df-27gpq Total loading time: 0 Render date: 2024-03-27T19:54:55.254Z Has data issue: false hasContentIssue false

Intestinal microbiota during early life – impact on health and disease

Published online by Cambridge University Press:  05 June 2014

Lotta Nylund*
Affiliation:
Functional Foods Forum, University of Turku, Turku, Finland Department of Veterinary Biosciences, Division of Microbiology and Epidemiology, University of Helsinki, Helsinki, Finland
Reetta Satokari
Affiliation:
Functional Foods Forum, University of Turku, Turku, Finland Department of Veterinary Biosciences, Division of Microbiology and Epidemiology, University of Helsinki, Helsinki, Finland
Seppo Salminen
Affiliation:
Functional Foods Forum, University of Turku, Turku, Finland
Willem M. de Vos
Affiliation:
Department of Veterinary Biosciences, Division of Microbiology and Epidemiology, University of Helsinki, Helsinki, Finland Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands Immunobiology Research Program, Department of Bacteriology & Immunology, University of Helsinki, Helsinki, Finland
*
*Corresponding author: L. Nylund, email lotta.nylund@utu.fi
Rights & Permissions [Opens in a new window]

Abstract

In the first years after birth, the intestinal microbiota develops rapidly both in diversity and complexity while being relatively stable in healthy adults. Different life-style-related factors as well as medical practices have an influence on the early-life intestinal colonisation. We address the impact of some of these factors on the consecutive microbiota development and later health. An overview is presented of the microbial colonisation steps and the role of the host in that process. Moreover, new early biomarkers are discussed with examples that include the association of microbiota and atopic diseases, the correlation of colic and early development and the impact of the use of antibiotics in early life. Our understanding of the development and function of the intestinal microbiota is constantly improving but the long-term influence of early-life microbiota on later life health deserves careful clinical studies.

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

Major colonisation of the intestinal microbiota starts at delivery when an infant comes into contact with microbes from the extrauterine environment. The microbial community co-evolves with its human host and after reaching the full complexity, outnumbers human cells by one or more orders of magnitude( Reference Zoetendal, Rajilić-Stojanović and de Vos 1 , Reference Scholtens, Oozeer and Martin 2 ). The collective genome of these microbes (also called microbiome) contributes significantly to our genetic coding capacity as they are found to contain over 3 million genes( Reference Qin 3 ). Unlike our own genome, the intestinal microbiome is not strictly vertically inherited. Moreover, this personalised ‘organ’ may be modified by a variety of foods, food components and pharmaceutical treatments targeting microbiota composition, stability and activity. While developing rapidly in diversity and complexity in the first years following birth, the intestinal microbiota becomes relatively stable in adulthood. A recent longitudinal study showed that the subject-specificity of the adult intestinal microbiota was maintained for over 10 years( Reference Rajilić-Stojanović, Heilig and Tims 4 ). This long-term stability provides further support for the cross-talk between the human host and microbiota( Reference Rajilić-Stojanović 5 ).

A series of changes in life style, such as improved hygienic conditions, reduced contact with companion or farm animals and a higher frequency of caesarean deliveries as well as expanded broad spectrum antibiotic use and dietary habits of both the infant and mother affect the intestinal colonisation process( Reference Rook, Raison and Lowry 6 ) (Fig. 1). Many of the factors described earlier are typical for an urban life style and are likely to lead to decreased exposure to microbes from the natural environment. Such exposure is thought to be essential both for the development of balanced microbiota diversity and composition and the optimal maturation of immune system( Reference Rook, Raison and Lowry 6 ), thus having lasting impact on later life health.

Fig. 1. (colour online) Modern life style factors associated with the development of intestinal microbiota and later life health.

Prenatal exposure to microbes and microbial compounds

Traditionally the fetus has been thought to be microbiologically sterile before birth. The presence of microbes in the amniotic fluid and placenta has mainly been associated with preterm deliveries due to maternal intrauterine infections and other pathological conditions( Reference Pararas, Skevaki and Kafetzis 7 Reference DiGiulio, Romero and Amogan 9 ) and the presence of bacterial DNA in amniotic fluid has been associated with lower gestational age and with low mean birth weight( Reference DiGiulio, Romero and Amogan 9 , Reference DiGiulio, Romero and Kusanovic 10 ). However, recent studies utilising molecular methods have shown that DNA of non-pathogenic bacteria can be detected in placenta and amniotic fluid samples in normal conditions( Reference Satokari, Grönroos and Laitinen 11 , Reference Rautava, Collado and Salminen 12 ). Hence, ingestion of amniotic fluid during pregnancy continuously exposes the fetus to bacteria and/or microbe-associated molecular patterns (MAMP). The exact mechanism(s) of bacterial entry into the intrauterine environment remains elusive. However, ascension from the vagina, by retrograde spread from abdominal cavity, haematogeneously through placenta and contamination during medical procedures (such as amniocentesis) have been suggested as potential routes( Reference Goldenberg, Culhane and Iams 13 ). Also MAMP can induce the immunostimulatory effects, for example via the stimulation of Toll-like receptors (TLR), without the need for microbial cells to enter the amniotic cavity. This is supported by the study by Rautava et al. where the presence of bacterial DNA, indicative for the presence of other MAMP too, in placenta and amniotic fluid was associated with the induction of expression profiles of TLR, especially TLR2 and TLR5 in fetal intestine( Reference Rautava, Collado and Salminen 12 ).

Furthermore, the expression of different TLR, including TLR9, has been shown to change during the maturation of gut epithelial cells( Reference Nanthakumar, Meng and Goldstein 14 , Reference Gribar, Sodhi and Richardson 15 ). TLR9 recognises unmethylated CpG motifs in bacterial DNA and its signalling maintains the gut epithelial homeostasis by improving the barrier functions and by inducing tolerance towards other MAMP( Reference Kant, de Vos and Palva 16 ). In utero the intestinal expression of TLR9 of mouse embryos decreases from days 14 to 18 and then increases again during the postnatal period( Reference Gribar, Sodhi and Richardson 15 ). Thus, it appears that a full-term newborn is programmed to receive TLR9 stimulation, which will improve the tolerance towards commensal bacteria. Consistently with this, necrotising enterocolitis in preterm infants has been associated with decreased TLR9 and increased TLR4 expression of the intestinal epithelium( Reference Gribar, Sodhi and Richardson 15 ). Remarkably, TLR9 activation via CpG-DNA supplementation significantly reduced necrotising enterocolitis severity( Reference Gribar, Sodhi and Richardson 15 ) suggesting that microbiota rich in CpG motifs but poor in TLR4 ligands, such as lipopolysaccharide-carrying Gram-negative bacteria could be optimal for the prevention or alleviation of necrotising enterocolitis. In this regard human breast milk, which supports the growth of Bifidobacteria, organisms with high-guanine–cytosine content genomes that are especially rich in CpG-motifs, appears to be a ‘superfood’ for the newborns. A number of strains of commercially produced lactobacilli have also been found to be rich in CpG-DNA( Reference Kant, de Vos and Palva 16 ) and probiotic interventions have shown some promising results in the prevention and alleviation of necrotising enterocolitis ( Reference Downard, Renaud and St Peter 17 ). In a mouse model, TLR9 signalling was indeed observed to be an essential mediator of anti-inflammatory effects of probiotics( Reference Rachmilewitz, Katakura and Karmeli 18 ). Furthermore, DNA of Bifidobacterium and Lactobacillus spp., both rich in CpG motifs, have been found in human placenta( Reference Satokari, Grönroos and Laitinen 11 ). Thus, it seems that prenatal exposure to MAMP is an important step in programming the development of gut epithelium and immune system already in utero.

Early-life microbiota

Meconium is the very first faecal specimen produced by the infant after birth. It consists mainly of amniotic fluid but includes also mucus, intestinal epithelial cells and concentrate of metabolites such as bile acids and pancreatic secretions( Reference Kumagai, Kimura and Takei 19 ). Several reports have described meconium microbiota composition providing further evidence for the suggestion that microbiological colonisation may begin already in utero ( Reference Moles, Gomez and Heilig 20 Reference Gosalbes, Llop and Valles 23 ).

Bacteria belonging to four major bacterial phyla in the intestine, Actinobacteria, Bacteroidetes, Firmicutes and Proteobacteria, are already detectable in the meconium. The predominant cultured bacteria seem to be bacilli within the Firmicutes phylum such as enterococci and staphylococci, or certain Proteobacteria such as Esherichia coli, Klebsiella and Enterobacter spp.( Reference Moles, Gomez and Heilig 20 Reference Hu, Nomura and Bashir 24 ). This is in agreement with the reports that these facultative anaerobes are present in faeces of healthy newborn infants( Reference Adlerberth and Wold 25 , Reference Palmer, Bik and Digiulio 26 ). In addition, Enterococcus spp. are commonly present, ~40 and 50 % of infants colonised at day 3, respectively( Reference Adlerberth and Wold 25 ). Following the colonisation of facultative bacteria, anaerobic bacteria appear in the infant faeces within the first weeks of life, decreasing the abundance of facultative anaerobes and thus introducing a shift in microbiota community structure( Reference Avershina, Storro and Oien 27 ). It should be noted, however, that this shift may represent an outgrowth of specific groups of bacteria and does not preclude the fact that their colonisation might have already occurred at low level. Especially the abundance of Bifidobacteria increases rapidly from ~3·5–10 % in meconium( Reference Moles, Gomez and Heilig 20 , Reference Jimenez, Marin and Martin 22 , Reference de Weerth, Fuentes and Puylaert 28 ) to 50–70 % and even up to 90 % in the faeces of breast-fed infants at ages 1 month and 3 months, respectively( Reference Fallani, Young and Scott 29 Reference Roger and McCartney 32 ). However, large inter-individual variations are characteristic for infant microbiota and the abundance of Bifidobacteria varies from 5 to 100 % in breast-fed infants( Reference Roger and McCartney 32 ). Considering formula-fed infants, Bifidobacteria (determined by fluorescent in situ hybridisation) may form a minor part of the microbiota, constituting ~25 % of the total microbiota( Reference Roger and McCartney 32 ). In addition to the individual variation, the Bifidobacterial abundance seems to vary greatly according to the geographic origin; infants from (northern) European countries harbour in general high numbers of Bifidobacteria( Reference Turroni, Peano and Pass 30 , Reference Roger and McCartney 32 , Reference Fallani, Amarri and Uusijärvi 33 ), whereas these bacteria are less predominant in Asian and American infants( Reference Palmer, Bik and Digiulio 26 , Reference Fan, Huo and Li 34 , Reference Koenig, Spor and Scalfone 35 ). This observation can be mainly explained by demographic differences and by differences in the rate and duration of breast-feeding between the countries and potentially also the differences in use of antibiotics.

After the introduction of solid foods and weaning the relative abundance of Bifidobacteria decreases gradually being ~60 % at 4 months, 25 % at 6 months and 10 % at 2 years( Reference Avershina, Storro and Oien 27 , Reference Nylund, Satokari and Nikkilä 36 , Reference Ringel-Kulka, Cheng and Ringel 37 ). Simultaneously, the relative abundance of lactobacilli decreases, whereas bacteria predominant in adult microbiota, such as Bacteroidetes and bacteria belonging to the Clostridium clusters XIVa and IV increase( Reference Roger and McCartney 32 , Reference Koenig, Spor and Scalfone 35 Reference Ringel-Kulka, Cheng and Ringel 37 ). However, the early-life microbiota composition is characterised by high inter-individual variation( Reference Avershina, Storro and Oien 27 , Reference Nylund, Satokari and Nikkilä 36 , Reference Avershina, Storro and Oien 38 ). The bacterial abundances in healthy infant microbiota vary greatly in a subject-wise manner and fluctuate further in response to the changes in different life events such as antibiotic treatments and introduction of solid foods( Reference Palmer, Bik and Digiulio 26 , Reference Koenig, Spor and Scalfone 35 ) (Fig. 1). During and after weaning major changes occur in microbiota diversity and composition, this transitional phase being more pronounced in breast-fed than in formula-fed infants( Reference Roger and McCartney 32 ). The succession of Bacteroides spp. and bacteria belonging to the Clostridium clusters XIVa and IV proceeds rapidly while the relative proportion of Bifidobacteria decreases( Reference Roger and McCartney 32 , Reference Koenig, Spor and Scalfone 35 ).

Previously, it has been suggested that the microbiota diversity and composition stabilise and reach the level of adult microbiota within the first year or two( Reference Palmer, Bik and Digiulio 26 , Reference Mackie, Sghir and Gaskins 39 ). However, recent studies have shown that microbiota maturation will continue longer( Reference Nylund, Satokari and Nikkilä 36 , Reference Ringel-Kulka, Cheng and Ringel 37 , Reference Agans, Rigsbee and Kenche 40 ). Interestingly, the establishment of bacteria belonging to Clostridium cluster XIVa at a level similar to adults has been observed already in young children (age 1–4 years)( Reference Ringel-Kulka, Cheng and Ringel 37 ), while other bacterial groups still remain at low-level abundance. This indicates that the microbiota development is a gradual process, where some bacterial groups may reach the degree of stabilisation earlier than others. However, considering the major physiological changes taking place in the human body within childhood and adolescence it may be argued that the development of the intestinal microbiota continues throughout this time period and is not finished until the human host reaches adulthood. The first studies on adolescent microbiota also point to this direction as they reported significant differences between the microbiota composition of adolescent children (age 11–18 years) and adults, the most striking difference being the almost 2-fold higher abundance of Bifidobacteria in adolescent subjects (9 v. 5·5 % of total microbiota, respectively)( Reference Agans, Rigsbee and Kenche 40 ). However, in order to comprehensively understand the microbiota development and stabilisation, more longitudinal studies analysing time-series samples from the same individuals over a long-time period are needed.

Effect of breast milk on microbiota composition

Human milk oligosaccharides (HMO) have an essential role in the promotion of the development of normal physiology of intestine and immune system in infants. Human milk contains a complex mixture of oligosaccharides, their exact composition varying according to different extrinsic and intrinsic factors. These factors include the genetic background of the mother, maternal health status, diet, secretor status and Lewis blood group type( Reference Bode 41 Reference Albrecht, Schols and van den Heuvel 43 ). Oligosaccharide molecules participate in the maintenance of a healthy gut microbiota in three ways. (1) They block the colonisation of pathogenic bacteria by acting as receptor analogues and binding to the bacterial surface, thus preventing the pathogens from binding to their target oligosaccharides on the epithelial cell surface( Reference Zivkovic, German and Lebrilla 44 ). (2) They act as prebiotic substrates promoting the growth of beneficial bacteria, notably Bifidobacteria, concurrently preventing the adherence of potentially harmful bacteria via colonisation resistance( Reference Bode 41 ). (3) They have also been suggested to modulate intestinal epithelial cells, lymphocyte cytokine production and leucocyte rolling and adhesion (comprehensively reviewed by Bode( Reference Bode 41 )).

Human infants lack the extensive set of enzymes needed for the digestion of glycan residues of HMO. Thus, these molecules pass undigested to the lower part of the intestinal tract, where they can be consumed by the specific members of infant gut microbiota( Reference Marcobal and Sonnenburg 45 ). Since a wide repertoire of enzymes are needed for the degradation and utilisation of the intricate structures of both HMO and plant polysaccharides, such processes most likely involve several different commensal bacteria acting synergistically. The two major bacterial genera described to have the capability for milk oligosaccharide utilisation are Bifidobacterium spp. and Bacteroides spp. Bifidobacteria, such as Bifidobacterium longum subsp. infantis and Bifidobacterium bifidum, typically abundant in infant microbiota, harbour a complex set of genes specifically related to HMO utilisation( Reference Sela, Garrido and Lerno 46 ). The B. longum subsp. infantis genome harbours entire gene clusters controlling the expression of glycosidases, membrane-spanning transporters and other proteins dedicated to human milk oligosaccharide utilisation( Reference Sela, Garrido and Lerno 46 , Reference Sela, Chapman and Adeuya 47 ). In contrast, B. longum subsp. longum, which is more abundant in adult microbiota, is unable to use diverse HMO, but has the capability to utilise short-chain oligosaccharides( Reference Sela, Garrido and Lerno 46 ). However, HMO have reported to up-regulate the expression of several pathways in B. longum subsp. Longum, such as genes involved in carbohydrate degradation and cell adherence( Reference Gonzalez, Klaassens and Malinen 48 ). Possibly, B. longum subsp. longum relies on cross-feeding with other bacteria, which first degrade complex polysaccharides to shorter units and thereby can also use HMO as a nutrient source.

Bacteroides spp. genomes harbour a specific gene cluster termed polysaccharide utilisation loci, enabling a wide range of saccharolytic ability( Reference Marcobal and Sonnenburg 45 , Reference Martens, Lowe and Chiang 49 ). For example, Bacteroides thetaiotaomicron can degrade more than a dozen different types of glycans( Reference Marcobal and Sonnenburg 45 ), most likely also HMO. In addition, in vitro utilisation of HMO by Bacteroides fragilis and Bacteroides vulgatus has been reported( Reference Marcobal and Sonnenburg 45 ). Consistently, B. fragilis and B. vulgatus are the predominant Bacteroides spp. found in breast-fed infants( Reference Tannock, Lawley and Munro 50 ). The abundance of bacterial groups, which have restricted capacity to utilise different polysaccharide compounds, are likely to fluctuate more in response to the type of incoming carbohydrates, whereas bacteria with a wide glycan-degrading capability may have a competitive advantage in the gut. Bacteroides spp. are among the first groups colonising the gut( Reference Palmer, Bik and Digiulio 26 , Reference Penders, Stobberingh and Thijs 51 ), increase further after the introduction of solid food and weaning( Reference Roger and McCartney 32 , Reference Koenig, Spor and Scalfone 35 ) and are part of the common core microbiota in adults( Reference Qin 3 , Reference Rajilić-Stojanović, Heilig and Molenaar 52 , Reference Huse, Ye and Zhou 53 ). Moreover, the ability of Bacteroides spp. to switch substrate specificity in response to the changing ingestion of nutrients indicates that they are adapted to the symbiotic life with human host and are permanent colonisers of the gut.

Human milk is also a source of bacteria to the infant. The predominant bacteria observed in human milk samples are Bacilli, such as Streptococcus spp. and Staphylococcus spp.( Reference Hunt, Foster and Forney 54 , Reference Jost, Lacroix and Braegger 55 ). In addition, Bifidobacterium spp. are present and Bacteroidetes and specific Clostridia such as butyrate-producing bacteria Faecalibacterium and Roseburia spp. have been detected( Reference Hunt, Foster and Forney 54 , Reference Jost, Lacroix and Braegger 56 , Reference Martin, Jimenez and Heilig 57 ). The bacterial composition of breast milk varies depending on the genetic background, maternal dietary habits and demographic differences between the mothers. For example, European mothers commonly harbour Lactobacillus and Bifidobacterium spp. in their breast milk, whereas these bacteria were rarely detected in mothers from the USA, possibly as a result of technical differences and drawbacks in DNA extraction( Reference Jost, Lacroix and Braegger 55 , Reference Jost, Lacroix and Braegger 56 , Reference Ward, Hosid and Ioshikhes 58 ). Furthermore, the mode of delivery has been shown to affect the milk microbiota composition( Reference Cabrera-Rubio, Collado and Laitinen 59 ). Milk samples from mothers who delivered their infants vaginally contained more Leuconostocaceae and less Carnobacteriaceae than milk samples from mothers who had gone through an elective caesarean section.

Maternal health status seems to have a major effect on the milk microbiota composition. For example, milk microbiota of overweight mothers differs from that of normal weight mothers( Reference Cabrera-Rubio, Collado and Laitinen 59 , Reference Collado, Laitinen and Salminen 60 ). The bacterial composition of breast milk seems to be stable at intra-individual level over time, while representing a great inter-individual variation. This suggests that human milk microbiota is highly personalised, in a manner similar to intestinal microbiota( Reference Costello, Lauber and Hamady 61 Reference Ursell, Clemente and Rideout 63 ).

Recently, the existence of a ‘core’ milk microbiota has been suggested( Reference Hunt, Foster and Forney 54 ). The milk core microbiota consisted of nine operational taxonomic units, corresponding to Staphylococcus, Streptococcus (Firmicutes), Corynebacterium, Propionibacterium (Actinobacteria) and Serratia, Pseudomonas, Ralstonia, Sphingomonas and Bradyrhizobiaceae (Proteobacteria), constituting approximately half the total bacterial community. It is noteworthy that many of the core microbiota genera are typically found from the skin and it seems likely that some part of the breast milk microbiota originates from the skin. Another origin of bacteria in human milk may be the intestinal tract of the mother. It has been suggested that intestinal bacteria could transfer within the phagocytosing cells from the gut to human milk via entero-mammary circulation of immune cells( Reference Grönlund, Gueimonde and Laitinen 64 ). Interestingly, Bifidobacterium breve is one of the most commonly detected Bifidobacterial species in human milk samples( Reference Martin, Jimenez and Heilig 57 , Reference Boesten, Schuren and Ben Amor 65 Reference Solis, de Los Reyes-Gavilan and Fernandez 69 ) and it produces exopolysaccharide, which masks other surface antigens and presents an ability to remain immunologically ‘silent’( Reference Fanning, Hall and Cronin 70 ). The production of exopolysaccharide seems to be important for the persistence of B. breve in the gut( Reference Fanning, Hall and Cronin 70 ). Speculatively, exopolysaccharide may also play a role in the survival of this bacterium within immune cells, enabling its transfer via the enteromammary circulation route. Moreover, B. breve and other Bifidobacteria are known to produce specific pili that are assumed to play a role in colonisation( Reference O'Connell Motherway, Zomer and Leahy 71 ). While the origin of bacteria in human milk remains an open question, human milk bacteria should be considered as an important source of bacteria in the establishment of intestinal microbiota during early life (Fig. 1).

Perturbations in the development of microbiota diversity and composition

Antibiotic treatments

The administration of antibiotics has been considered the most remarkable extrinsic factor affecting the microbiota composition and development during early infancy and childhood (Fig. 1). The majority of antibiotics used to treat early-life infections have a rather broad-spectrum antimicrobial activity, inhibiting the growth of both pathogenic bacteria and the beneficial members of commensal microbiota. Especially in children, the major effect is the reduction of bacteria considered to have health-promoting properties such as Bifidobacterium spp. and Lactobacillus spp., which may have long-term effects on the infants’ health later in life.

After the antibiotic treatment, the overall microbiota diversity is generally decreased and the microbiota composition is often characterised by a dominance of a few bacterial groups( Reference Fouhy, Guinane and Hussey 72 , Reference Dethlefsen and Relman 73 ). In a recent study, the effect of early-life antibiotic treatment (ampicillin and gentamicin) on microbiota composition of newborn infants was analysed using a high-throughput sequencing( Reference Fouhy, Guinane and Hussey 72 ). The authors reported higher proportions of Proteobacteria and reduced abundances of genera Bifidobacterium and Lactobacillus in antibiotic-treated infants when compared with untreated controls 1 month after the cessation of treatment( Reference Fouhy, Guinane and Hussey 72 ). These findings are in line with previous observations considering the microbiota deviations after the administration of different types of antibiotics( Reference Fallani, Young and Scott 29 , Reference Rea, Dobson and O'Sullivan 74 ). Moreover, a declined prevalence of Bacteroidetes and higher abundances of enterobacteria and enterococci have been reported in antibiotic-treated infants when compared with healthy controls( Reference Fallani, Young and Scott 29 , Reference Fouhy, Guinane and Hussey 72 , Reference Tanaka, Kobayashi and Songjinda 75 ). In addition, a decreased abundance of Bifidobacteria have been reported in patients who have received antibiotics( Reference Hussey, Wall and Gruffman 76 , Reference Mangin, Leveque and Magne 77 ). However, the sensitivity for antibiotics seems to be a strain-specific feature and thus different Bifidobacterium species may be distinctly affected. In a study by Mangin et al. ( Reference Mangin, Suau and Gotteland 78 ), the total number of Bifidobacteria was not decreased after amoxicillin treatment for 7 d. Instead, the diversity of Bifidobacterium spp. population and a shift in species composition was observed. Specifically, a complete loss of Bifidobacterium adolescentis group and a decreased amount of B. bifidum were detected, whereas B. longum and B. catenulatum group bacteria were not affected( Reference Mangin, Suau and Gotteland 78 ). Thus, high diversity of Bifidobacterial species may protect the infant from more extensive effects of specific antibiotics.

The microbiota recovery begins shortly after the cessation of antibiotic administration but it seems to be rather slow and gradual process and the recovery remains often incomplete( Reference Dethlefsen and Relman 73 , Reference Jernberg, Lofmark and Edlund 79 , Reference Jakobsson, Jernberg and Andersson 80 ). In a recent study, the elevated amounts of Proteobacteria could still be detected 2 months after the termination of antibiotic medication, whereas Bifidobacteria and Lactobacilli abundances were more or less recovered( Reference Fouhy, Guinane and Hussey 72 ). The overgrowth of Proteobacteria, especially Enterobacteriaceae after the antibiotic treatment(s) has been widely reported. This effect can be explained by the competitive advantage obtained by the production of β-lactamases. These enzymes degrade the β-lactam antibiotic structure, thus providing resistance against several antibiotics such as amoxicillin, ampicillin and gentamicin.

Interestingly, early-life antibiotic treatment(s) have been associated with the increased risk for health problems later in life such as the risk for coeliac disease (CD) development( Reference Mårild, Ye and Lebwohl 81 ), allergic diseases( Reference Foliaki, Pearce and Björksten 82 ) and the increased risk of obesity at school age( Reference Ajslev, Andersen and Gamborg 83 ). Furthermore, prenatal exposure to antibiotics may have long-term effects on the health later in life, since maternal antibiotic use during pregnancy has been associated with an increased risk of cow's milk allergy, asthma, eczema and hay fever in their infants( Reference McKeever, Lewis and Smith 84 , Reference Metsälä, Lundqvist and Virta 85 ). However, only a limited number of studies utilising high-throughput analysis methods on the evaluation of microbiota diversity and composition after antibiotic therapies have been published. Further studies are required to assess the impact of antibiotics on host-microbe cross-talk and interactions. Furthermore, the long-term effects of antibiotics on both microbiota and on later health status of the paediatric patients need to be urgently assessed.

Colic

Colic crying is one of the most common problems in early life confronting ~10–25 % of otherwise healthy infants within the first months of life( Reference Pärtty, Luoto and Kalliomäki 86 , Reference Milidou, Sondergaard and Jensen 87 ). Colic cry is characterised by inexplicable, excessive crying >3 h/d for 3 d or more in 1 week, whereas it does not respond to any interventions such as feeding, diaper change or other solicitude procedures( Reference Wessel, Cobb and Jackson 88 ). Usually colic crying starts from 2-week-old to 3-month-old infants and declines after a few months. Although its aetiology and pathogenesis remain obscure, an association between colic cry and immaturity of intestinal function and/or neurodevelopmental maturity as well as excessive colonic gas production has been suggested( Reference Savino, Cordisco and Tarasco 89 , Reference Rhoads, Fatheree and Norori 90 ). In addition, an aberrant microbiota composition has been suggested to promote colicky symptoms. Microbiota diversity and stability have been observed to be lower in infants suffering from colic than in healthy control subjects( Reference de Weerth, Fuentes and Puylaert 28 , Reference Rhoads, Fatheree and Norori 90 ).

The most consistent finding is the decreased amounts of Bifidobacterium spp. and Lactobacillus spp. in infants with colic or extensive crying( Reference de Weerth, Fuentes and Puylaert 28 , Reference Pärtty, Kalliomäki and Endo 91 ). Conversely, elevated numbers of these bacteria have been linked to decreased colicky symptoms( Reference Roos, Dicksved and Tarasco 92 ). A recent study reported an association between delayed colonisation by Bifidobacterium infantis and increased risk of irritability in preterm infants( Reference Pärtty, Luoto and Kalliomäki 86 ). Previously, the colonisation of B. infantis has been associated with normal development of immune tolerance and the species has been shown to be capable of normalising the permeability of intestinal mucosa( Reference Chichlowski, De Lartigue and German 93 ). This effect is most likely mediated by bioactive factors secreted by B. infantis, which have been shown to induce the expression of tight junction proteins, thus tightening the connections between enterocytes( Reference Ewaschuk, Diaz and Meddings 94 ). Moreover, Bifidobacteria have been associated with reduced abdominal pain and discomfort in adults( Reference Jalanka-Tuovinen, Salonen and Nikkilä 95 ).

Bacteria that are increased in infants with excessive crying and colic symptoms include anaerobic Gram-negative and coliform bacteria( Reference de Weerth, Fuentes and Puylaert 28 , Reference Savino, Cordisco and Tarasco 89 , Reference Rhoads, Fatheree and Norori 90 , Reference Savino, Cordisco and Tarasco 96 ). It has been speculated that coliform bacteria such as E. coli may overtake the beneficial bacteria in colicky infants resulting in reduced induction of regulatory T cells by beneficial commensals and increased production of cytokines by antigen-presenting cells, thus leading to immune dysregulation and increased permeability of intestinal epithelium( Reference Savino, Cordisco and Tarasco 89 ). A recent study utilising high-throughput microarray analysis reported a negative association between crying and butyrate-producing bacteria such as Butyrivibrio crossotus, Eubacterium rectale and Eubacterium hallii, which were found to be 1·5-fold more abundant in healthy infants without colic symptoms( Reference de Weerth, Fuentes and Puylaert 28 ). Butyrate-producing bacteria have been shown to reduce the pain sensation( Reference Vanhoutvin, Troost and Kilkens 97 ) and proposed to reinforce gut defense barrier by increasing the production of mucins( Reference Burger-van Paassen, Vincent and Puiman 98 ). Butyrate also up-regulates the expression of tight junction proteins, thus leading to decreased intestinal permeability( Reference Ma, Fan and Li 99 , Reference Wang, Wang and Wang 100 ).

The role of aberrant microbiota composition in colic is supported by the observation that colicky symptoms could be alleviated by probiotic supplementation of Lactobacillus reuteri DSM 17938( Reference Savino, Cordisco and Tarasco 101 ). The authors suggested the improvement of gut motility and function and the reduction of visceral pain as possible mechanisms of probiotic action. Interestingly, the probiotic strain L. reuteri DSM 17938 has also been shown to reduce gastric distension and accelerate gastric emptying rate, which could potentially alleviate colic symptoms( Reference Indrio, Riezzo and Raimondi 102 ). Furthermore, two L. reuteri strains have been shown to inhibit the growth of colic-associated coliforms in vitro ( Reference Savino, Cordisco and Tarasco 96 ). This inhibition was mediated by bacteriocins and other inhibitory molecules produced by L. reuteri. Moreover, the potential of Lactobacillus rhamnosus GG in alleviation of colic symptoms have been reported( Reference Pärtty, Luoto and Kalliomäki 86 , Reference Pärtty and Isolauri 103 ). In contrast, a recent study has shown that L. reuteri strain DSM 17938 was not effective in protecting newborns from colic( Reference Sung, Hiscock and Tang 104 ). Thus, these studies warrant further assessment and well-planned intervention studies to characterise the potential effects of other probiotic strains in addition to L. reuteri and L. rhamnosus GG.

Atopic diseases

Atopic diseases are chronic and relapsing disorders usually starting in early childhood. Atopy has been characterised as a genetic disposition to develop an allergic reaction and produce elevated levels of IgE upon exposure to an environmental antigen( Reference Bieber 105 ). Atopic diseases include eczema (atopic dermatitis), allergic rhinitis (hay fever), allergic conjunctivitis and allergic asthma. In early life, the most common form of atopic disease is eczema, its prevalence being ~15–30 % depending on the country studied( Reference Deckers, McLean and Linssen 106 ). The incidences of eczema and other allergic diseases are more common in industrialised countries, the highest prevalence typically found in Northern Europe( Reference Bieber 105 ). During the past decades, associations between the composition of intestinal microbiota and atopic diseases have been studied intensively.

Some of the studies evaluating the associations between microbiota composition and atopy have also addressed the microbiota composition preceding the development of disease. Reduced diversity at early life (i.e. at ages 1 week, 1 month or 4 months) has been associated with an increased risk of developing atopy or allergic disease( Reference Abrahamsson, Jakobsson and Andersson 107 Reference Kalliomäki, Kirjavainen and Eerola 112 ). However, after age 1 year the total microbiota diversity in children either developing or having eczema is comparable or even higher than that of healthy children( Reference Nylund, Satokari and Nikkilä 36 , Reference Abrahamsson, Jakobsson and Andersson 107 ). In addition, the pathogenesis of atopic diseases is associated with an impaired gut barrier function and increased intestinal permeability and gastrointestinal symptoms are common among the patients( Reference Rosenfeldt, Benfeldt and Valerius 113 ). Thus, it seems that sufficient diversity of microbiota in early infancy is essential for modulation of the expression of genes involved in the normal pattern of intestinal development such as postnatal intestinal maturation and maintenance of mucosal barrier( Reference Sjögren, Tomicic and Lundberg 114 , Reference Hooper and Macpherson 115 ). However, microbiota development and diversification should not happen too expeditiously, since prematurely occurring changes towards an adult-type microbiota may predispose infants, e.g. with eczema( Reference Nylund, Satokari and Nikkilä 36 ). It is possible that an infant-type microbiota supports an adequate gut barrier function and tolerance against allergens in an immature gut and affects the maturation of the gut epithelium and immune functions in a way that results in reinforcement of the normal mucosal barrier function( Reference Rosenfeldt, Benfeldt and Valerius 113 , Reference Maynard, Elson and Hatton 116 ).

The results on specific bacteria either increasing or decreasing the risk of developing atopic diseases or associated with their onset are still conflicting( Reference Nylund, Satokari and Nikkilä 36 , Reference Penders, Stobberingh and Thijs 51 , Reference Sepp, Julge and Mikelsaar 117 Reference Mah, Chin and Wong 121 ). Aberrations in Bifidobacterial community have been associated with children with atopic diseases, most often characterised by either reduced total abundance or shifts in species community( Reference Maynard, Elson and Hatton 116 , Reference Sepp, Julge and Mikelsaar 117 , Reference Johansson, Sjögren and Persson 119 , Reference Gore, Munro and Lay 120 ). Furthermore, decreased amounts of Bacteroides spp. and increased amounts of specific Firmicutes such as Staphylococcus aureus and different clostridial groups, have been associated with the development and onset of allergic diseases( Reference Nylund, Satokari and Nikkilä 36 , Reference Abrahamsson, Jakobsson and Andersson 107 , Reference Björksten, Sepp and Julge 122 Reference Thompson-Chagoyan, Fallani and Maldonado 125 ). Interestingly, both Bifidobacterium spp. and Bacteroides spp. have been reported to have anti-inflammatory properties via their ability to direct the cellular and physical maturation of the developing immune system( Reference Hooper, Wong and Thelin 126 , Reference Pagnini, Saeed and Bamias 127 ). For example, polysaccharide A from B. fragilis is able to direct the development of CD4+ T cells, thus inducing the differentiation of T helper (Th) 1 lineage and correction of the Th1/Th2 imbalance( Reference Mazmanian, Liu and Tzianabos 128 ). Furthermore, this polysaccharide has been shown to promote immunologic tolerance through induction of regulatory T cells, resulting in suppression of IL-17 responses( Reference Round, Lee and Li 129 ). Moreover, both Bifidobacterium and Bacteroides spp. have high frequency of immunostimulatory CpG motifs in their genomes, thus being rich in TLR9 ligands( Reference Kant, de Vos and Palva 16 ). TLR9 stimulation is known to both enhance epithelial integrity and direct immune responses towards Th1 type (reviewed in Kant et al.( Reference Kant, de Vos and Palva 16 )). These effects may be diminished in allergic subjects, who have reduced numbers of Bifidobacterium and Bacteroides spp.

In recent studies, increased levels of IL-17 have been associated with asthma( Reference Alyasin, Karimi and Amin 130 , Reference Ramirez-Velazquez, Castillo and Guido-Bayardo 131 ). Furthermore, one of the most important defence mechanisms in the epithelial barrier is IgA, which is present at high concentrations in the intestinal mucus layer( Reference Brandtzaeg 132 ). Low levels of IgA predisposes infants to increased binding of antigens to mucosal membrane, to increased mucosal leakiness and an increased uptake of dietary antigens( Reference Johansen, Pekna and Norderhaug 133 ). Low levels of IgA have also been associated with increased risk for the development of IgE-mediated allergic diseases in children( Reference Kukkonen, Kuitunen and Haahtela 134 ). Furthermore, it has been suggested that high numbers of Clostridium spp. may be associated with degradation of antigen-specific IgA, which could debilitate the immature gut barrier( Reference Pärtty, Luoto and Kalliomäki 86 ). The protective role of specific bacteria and their compounds against atopy and allergic diseases is further supported by several clinical studies reporting the effects of probiotic strains on the alleviation of allergic symptoms even when the probiotics failed to modify the microbiota composition or diversity( Reference Nermes, Kantele and Atosuo 135 , Reference Elazab, Mendy and Gasana 136 ). These effects can be related to the probiotic effects on the hosts’ immunological functions such as improvement of the barrier function and increasing allergen-specific IgA levels, which are essential for the development of tolerance and can be considered as a marker for immune maturation( Reference Rosenfeldt, Benfeldt and Valerius 113 , Reference Kukkonen, Kuitunen and Haahtela 134 , Reference Nermes, Kantele and Atosuo 135 , Reference Mantis, Rol and Corthesy 137 Reference Di Mauro, Neu and Riezzo 139 ). Furthermore, probiotics have been suggested to have immunomodulatory impacts that affect the Th1/Th2 balance such as stimulation of Th1-type immune responses, induction of apoptosis of Th2 cells and induction of regulatory T and dendritic cells( Reference Kant, de Vos and Palva 16 , Reference Rautava, Arvilommi and Isolauri 138 , Reference Kwon, Lee and So 140 Reference West, Hammarstrom and Hernell 144 ).

Coeliac disease

CD is an autoimmune disorder of the small intestine that occurs in genetically predisposed individuals. It is caused by a reaction to dietary gluten and related prolamines, which are proteins found in maize such as wheat, barley and rye. Upon exposure to gluten, inflammatory cascade is induced in the small intestinal epithelium leading to a villous atrophy and crypt hyperplasia( Reference Green and Jabri 145 ). Typical symptoms include different gastrointestinal symptoms such as diarrhoea, abdominal pain and distension( Reference Murch, Jenkins and Auth 146 ). Untreated CD may lead to weight loss, malabsorption and growth disturbances in paediatric patients( Reference Murch, Jenkins and Auth 146 ). The CD is a multifactorial disorder and its pathogenesis involves both genetic and environmental factors. For example, a high frequency of infectious episodes early in life( Reference Stene, Honeyman and Hoffenberg 147 , Reference Myleus, Hernell and Gothefors 148 ), antibiotic treatments( Reference Mårild, Ye and Lebwohl 81 ) as well as the timing of gluten introduction into the diet( Reference Sellitto, Bai and Serena 149 , Reference Ivarsson, Persson and Nystrom 150 ) have been associated with the onset of CD in genetically susceptible infants (Fig. 1).

A specific role for the intestinal microbiota in CD development has been suggested( Reference Sellitto, Bai and Serena 149 , Reference Sanz, De Pama and Laparra 151 , Reference De Palma, Nadal and Medina 152 ). Indeed, deviations in faecal and duodenal microbiota associated with CD have been reported( Reference Sellitto, Bai and Serena 149 , Reference Sanz, De Pama and Laparra 151 Reference Schippa, Iebba and Barbato 153 ), although recent studies utilising high-throughput methods have reported comparable microbiota compositions in patients and healthy controls( Reference de Meij, Budding and Grasman 154 Reference Cheng, Kalliomäki and Heilig 157 ). A recent study utilising a high-throughput microarray method in analysing duodenal biopsies of paediatric CD patients in Finland found that while the overall microbiota composition was comparable between CD and healthy subjects, a profile of eight bacterial groups was observed to distinguish patients from healthy controls( Reference Cheng, Kalliomäki and Heilig 157 ). This profile was characterised by higher abundances of bacteria related to Prevotella melaninogenica, Haemophilus and Serratia spp., whereas those related to P. oralis, P. cinnamivorans, Ruminococcus bromiii, Proteus and Clostridium stercorarium were decreased in CD patients( Reference Cheng, Kalliomäki and Heilig 157 ). Also the total abundance of Prevotella spp. was found to be slightly increased (did not reach a statistical significance)( Reference Cheng, Kalliomäki and Heilig 157 ), which supports the previous findings by a Swedish research group, who found an association of elevated total abundance of Prevotella spp. and CD( Reference Forsberg, Fahlgren and Horstedt 158 , Reference Ou, Hedberg and Horstedt 159 ). Microbiota dysbiosis of CD patients may be characterised by an increased microbiota diversity( Reference De Palma, Nadal and Medina 152 , Reference Schippa, Iebba and Barbato 153 , Reference Nadal, Donat and Ribes-Koninckx 160 , Reference Sanchez, Donat and Ribes-Koninckx 161 ), but these findings have been contradicted recently( Reference Cheng, Kalliomäki and Heilig 157 ). Furthermore, patients with active CD seem to have an increased inter-individual similarity when compared with patients in remission state or healthy controls( Reference Schippa, Iebba and Barbato 153 , Reference Forsberg, Fahlgren and Horstedt 158 ). It has been suggested that altered glycosylation patterns observed in mucosa of CD patients( Reference Forsberg, Fahlgren and Horstedt 158 ) may create a more selective pressure leading to a more homogenous microbial colonisation( Reference Schippa, Iebba and Barbato 153 ).

Such microbiota deviations are only partly restored after long-term treatment with gluten-free diet. A higher diversity and a complete rearrangement in Eubacterium species community as well as changed metabolomic profiles were observed in CD patients who had followed gluten-free diet for 2 years when compared to healthy controls( Reference Di Cagno, De Angelis and De Pasquale 162 ). In contrast, the proportions of E. coli and Staphylococcus were observed to normalise after treatment with gluten-free diet( Reference Collado, Donat and Ribes-Koninckx 163 ).

In the active phase of CD, the reduction of Gram-positive bacteria population, especially the numbers or proportion of Bifidobacterium spp. has been reported( Reference De Palma, Nadal and Medina 152 , Reference Collado, Donat and Ribes-Koninckx 163 ). Such findings may be of interest, since Bifidobacteria have been suggested to alleviate gastrointestinal symptoms of adult coeliac patients( Reference Smecuol, Hwang and Sugai 164 ) and have been associated with reduced abdominal pain and discomfort in healthy adults( Reference Jalanka-Tuovinen, Salonen and Nikkilä 95 ). In contrast to declined proportions of Gram-positives, Gram-negative bacteria such as Clostridium groups( Reference Nadal, Donat and Ribes-Koninckx 160 , Reference Collado, Donat and Ribes-Koninckx 163 ), Prevotella spp.( Reference Cheng, Kalliomäki and Heilig 157 , Reference Ou, Hedberg and Horstedt 159 ) and E. coli ( Reference Schippa, Iebba and Barbato 153 , Reference Collado, Donat and Ribes-Koninckx 163 ) seem to be increased in paediatric CD patients. The most constant finding is the higher abundance of Bacteroides spp. in faeces and duodenal biopsies of CD patients( Reference De Palma, Nadal and Medina 152 , Reference Schippa, Iebba and Barbato 153 , Reference Collado, Donat and Ribes-Koninckx 163 ), although a complete lack of the members of phylum Bacteroidetes was observed in CD predisposed infants in a prospective study( Reference Sellitto, Bai and Serena 149 ).

Furthermore, another study reported a reduction in IgA-coated bacteria, especially IgA-coated Bacteroides in faeces of untreated and treated CD patients when compared to healthy controls( Reference Sanz, De Pama and Laparra 151 ). The authors stated that host defences against this bacterial group might be reduced in coeliac disease, thus allowing its increased colonisation. Moreover, shifts in Bacteroides spp. composition in early-life microbiota have been reported in infants with high genetic risk for CD development compared to infants with low genetic risk( Reference Collado, Donat and Ribes-Koninckx 163 ). In detail, Bacteroides uniformis, B. ovatus and B. plebeius were associated with a low genetic risk, whereas B. vulgatus seems to be more prevalent both in high-risk infants( Reference Sanchez, De Palma and Capilla 165 ) and in infants with active CD( Reference Schippa, Iebba and Barbato 153 ). Furthermore, B. fragilis has been associated with an increased risk for CD development in genetically predisposed infants who were formula-fed( Reference Palma, Capilla and Nova 166 ). Interestingly, polysaccharide A produced by B. fragilis has been shown to induce the differentiation of Th1-type immune cells( Reference Mazmanian, Liu and Tzianabos 128 ). In addition, a decreased duodenal expression of TLR2 and increased expression of TLR9 and IL-8 have been observed in infants with CD( Reference Kalliomäki, Satokari and Lähteenoja 155 ). It has been suggested that increased TLR9 signalling in the duodenum may contribute to the Th1 response found in the small intestinal mucosa of CD subjects( Reference Kalliomäki, Satokari and Lähteenoja 155 , Reference Cheng, Kalliomäki and Heilig 157 ). Furthermore, the expression of tight junction protein coding ZO-1 is significantly down-regulated in untreated CD patients when compared to patients with treated CD( Reference Cheng, Kalliomäki and Heilig 157 ). Thus, a synergistic effect of Bacteroides spp. and increased TLR9 signalling may lead to an excessive induction of Th1-type immune response, which may contribute to the onset and/or remission of the coeliac disease. Collectively, it seems that both altered microbiota composition and dysregulated host–microbe interaction may have a role in CD.

Conclusions

The microbiota development is a gradual process, which begins during early phases of pregnancy. During the succession of microbes some bacterial groups reach the degree of maturation earlier than others. The development of the intestinal microbiota is likely to continue throughout childhood and adolescence and may not be completed until the human host reaches adulthood. The course of development is affected by both life-style factors and medical practices that direct the intestinal colonisation and have an impact on health later in life. Our understanding of both the compositional development and the diversity and function of the intestinal microbiota and its effects on health and disease is constantly improving but further studies are still needed to address the long-term influence of early-life gut microbiota on intestinal, systemic immunity and other organ systems.

Acknowledgements

None.

Financial Support

This work was supported by The Finnish Graduate School on Applied Bioscience: Bioengineering, Food and Nutrition, Environment (for L. N.), The Academy of Finland (Grant no. 258438 for R. S. and Grant numbers 137389 and 141140 for W. de V.) and the ERC Advanced Grant no. 250172 (Microbes Inside) of the European Research Council.

Conflicts of Interest

None.

Authorship

L. N. wrote the paper; R. S. designed Fig. 1; all authors corrected and approved the manuscript.

References

1. Zoetendal, EG, Rajilić-Stojanović, M & de Vos, WM (2008) High-throughput diversity and functionality analysis of the gastrointestinal tract microbiota. Gut 57, 16051615.CrossRefGoogle ScholarPubMed
2. Scholtens, PA, Oozeer, R, Martin, R et al. (2012) The early settlers: intestinal microbiology in early life. Annu Rev Food Sci Technol 3, 425447.Google Scholar
3. Qin, J & Members of MetaHIT Consortium (2010) A human gut microbial gene catalog established by deep metagenomic sequencing. Nature 464, 5965.CrossRefGoogle Scholar
4. Rajilić-Stojanović, M, Heilig, HG, Tims, S et al. (2012) Long-term monitoring of the human intestinal microbiota composition. Environ Microbiol 15, 11461159.CrossRefGoogle Scholar
5. Rajilić-Stojanović, M (2013) Function of the microbiota. Best Pract Res Clin Gastroenterol 27, 516.Google Scholar
6. Rook, GA, Raison, CL & Lowry, CA (2014) Microbial “Old Friends”, immunoregulation and socio-economic status. Clin Exp Immunol (In the Press).CrossRefGoogle Scholar
7. Pararas, MV, Skevaki, CL & Kafetzis, DA (2006) Preterm birth due to maternal infection: causative pathogens and modes of prevention. Eur J Clin Microbiol Infect Dis 25, 562569.Google Scholar
8. Wang, X, Buhimschi, CS, Temoin, S et al. (2013) Comparative microbial analysis of paired amniotic fluid and cord blood from pregnancies complicated by preterm birth and early-onset neonatal sepsis. PLoS ONE 8, e56131.Google Scholar
9. DiGiulio, DB, Romero, R, Amogan, HP et al. (2008) Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PLoS ONE 3, e3056.Google Scholar
10. DiGiulio, DB, Romero, R, Kusanovic, JP et al. (2010) Prevalence and diversity of microbes in the amniotic fluid, the fetal inflammatory response, and pregnancy outcome in women with preterm pre-labor rupture of membranes. Am J Reprod Immunol 64, 3857.CrossRefGoogle ScholarPubMed
11. Satokari, R, Grönroos, T, Laitinen, K et al. (2009) Bifidobacterium and Lactobacillus DNA in the human placenta. Lett Appl Microbiol 48, 812.CrossRefGoogle ScholarPubMed
12. Rautava, S, Collado, MC, Salminen, S et al. (2012) Probiotics modulate host-microbe interaction in the placenta and fetal gut: a randomized, double-blind, placebo-controlled trial. Neonatology 102, 178184.CrossRefGoogle Scholar
13. Goldenberg, RL, Culhane, JF, Iams, JD et al. (2008) Epidemiology and causes of preterm birth. Lancet 371, 7584.CrossRefGoogle ScholarPubMed
14. Nanthakumar, N, Meng, D, Goldstein, AM et al. (2011) The mechanism of excessive intestinal inflammation in necrotizing enterocolitis: an immature innate immune response. PLoS ONE 6, e17776.CrossRefGoogle ScholarPubMed
15. Gribar, SC, Sodhi, CP, Richardson, WM et al. (2009) Reciprocal expression and signaling of TLR4 and TLR9 in the pathogenesis and treatment of necrotizing enterocolitis. J Immunol 182, 636646.CrossRefGoogle ScholarPubMed
16. Kant, R, de Vos, WM, Palva, A et al. (2013) Immunostimulatory CpG motifs in the genomes of gut bacteria and their role in human health and disease. J Med Microbiol 63, 293308.CrossRefGoogle ScholarPubMed
17. Downard, CD, Renaud, E, St Peter, SD et al. (2012) Treatment of necrotizing enterocolitis: an American Pediatric Surgical Association Outcomes and Clinical Trials Committee systematic review. J Pediatr Surg 47, 21112122.CrossRefGoogle ScholarPubMed
18. Rachmilewitz, D, Katakura, K, Karmeli, F et al. (2004) Toll-like receptor 9 signaling mediates the anti-inflammatory effects of probiotics in murine experimental colitis. Gastroenterology 126, 520528.CrossRefGoogle ScholarPubMed
19. Kumagai, M, Kimura, A, Takei, H et al. (2007) Perinatal bile acid metabolism: bile acid analysis of meconium of preterm and full-term infants. J Gastroenterol 42, 904910.CrossRefGoogle ScholarPubMed
20. Moles, L, Gomez, M, Heilig, H et al. (2013) Bacterial diversity in meconium of preterm neonates and evolution of their fecal microbiota during the first month of life. PLoS ONE 8, e66986.CrossRefGoogle ScholarPubMed
21. 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
22. Jimenez, E, Marin, ML, Martin, R et al. (2008) Is meconium from healthy newborns actually sterile? Res Microbiol 159, 187193.CrossRefGoogle ScholarPubMed
23. Gosalbes, MJ, Llop, S, Valles, Y et al. (2013) Meconium microbiota types dominated by lactic acid or enteric bacteria are differentially associated with maternal eczema and respiratory problems in infants. Clin Exp Allergy 43, 198211.CrossRefGoogle ScholarPubMed
24. Hu, J, Nomura, Y, Bashir, A et al. (2013) Diversified microbiota of meconium is affected by maternal diabetes status. PLoS ONE 8, e78257.CrossRefGoogle ScholarPubMed
25. Adlerberth, I & Wold, AE (2009) Establishment of the gut microbiota in Western infants. Acta Paediatr 98, 229238.CrossRefGoogle ScholarPubMed
26. Palmer, C, Bik, EM, Digiulio, DB et al. (2007) Development of the human infant intestinal microbiota. PLoS ONE 5, e177.CrossRefGoogle ScholarPubMed
27. Avershina, E, Storro, O, Oien, T et al. (2013) Major faecal microbiota shifts in composition and diversity with age in a geographically restricted cohort of mothers and their children. FEMS Microbiol Ecol 87, 280290.CrossRefGoogle Scholar
28. de Weerth, C, Fuentes, S, Puylaert, P et al. (2013) Intestinal microbiota of infants with colic: development and specific signatures. Pediatrics 131, e550e558.Google Scholar
29. 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.Google Scholar
30. Turroni, F, Peano, C, Pass, DA et al. (2012) Diversity of bifidobacteria within the infant gut microbiota. PLoS ONE 7, e36957.CrossRefGoogle ScholarPubMed
31. Bezirtzoglou, E & Stavropoulou, E (2011) Immunology and probiotic impact of the newborn and young children intestinal microflora. Anaerobe 17, 369374.CrossRefGoogle Scholar
32. Roger, LC & McCartney, AL (2010) Longitudinal investigation of the faecal microbiota of healthy full-term infants using fluorescence in situ hybridization and denaturing gradient gel electrophoresis. Microbiology 156, 33173328.CrossRefGoogle ScholarPubMed
33. Fallani, M, Amarri, S, Uusijärvi, 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
34. Fan, W, Huo, G, Li, X et al. (2013) Impact of diet in shaping gut microbiota revealed by a comparative study in infants during the six months of life. J Microbiol Biotechnol 24, 133143.Google Scholar
35. Koenig, JE, Spor, A, Scalfone, N et al. (2011) Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci USA 108, S4578S4585.Google Scholar
36. Nylund, L, Satokari, R, Nikkilä, J et al. (2013) Microarray analysis reveals marked intestinal microbiota aberrancy in infants having eczema compared to healthy children in at-risk for atopic disease. BMC Microbiol 13, 12.Google Scholar
37. Ringel-Kulka, T, Cheng, J, Ringel, Y et al. (2013) Intestinal microbiota in healthy U.S. young children and adults – a high throughput microarray analysis. PLoS ONE 8, e64315.CrossRefGoogle ScholarPubMed
38. Avershina, E, Storro, O, Oien, T et al. (2013) Bifidobacterial succession and correlation networks in a large unselected cohort of mothers and their children. Appl Environ Microbiol 79, 497507.Google Scholar
39. Mackie, RI, Sghir, A & Gaskins, HR (1999) Developmental microbial ecology of the neonatal gastrointestinal tract. Am J Clin Nutr 69, 1035S1045S.CrossRefGoogle ScholarPubMed
40. Agans, R, Rigsbee, L, Kenche, H et al. (2011) Distal gut microbiota of adolescent children is different from that of adults. FEMS Microbiol Ecol 77, 404412.CrossRefGoogle ScholarPubMed
41. Bode, L (2012) Human milk oligosaccharides: every baby needs a sugar mama. Glycobiology 22, 11471162.Google Scholar
42. Thurl, S, Munzert, M, Henker, J et al. (2010) Variation of human milk oligosaccharides in relation to milk groups and lactational periods. Br J Nutr 104, 12611271.Google Scholar
43. Albrecht, S, Schols, HA, van den Heuvel, EG et al. (2011) Occurrence of oligosaccharides in feces of breast-fed babies in their first six months of life and the corresponding breast milk. Carbohydr Res 346, 25402550.CrossRefGoogle ScholarPubMed
44. Zivkovic, AM, German, JB, Lebrilla, CB et al. (2011) Human milk glycobiome and its impact on the infant gastrointestinal microbiota. Proc Natl Acad Sci USA 108, S4653S4658.CrossRefGoogle ScholarPubMed
45. Marcobal, A & Sonnenburg, JL (2012) Human milk oligosaccharide consumption by intestinal microbiota. Clin Microbiol Infect 18, Suppl. 4, 1215.CrossRefGoogle ScholarPubMed
46. Sela, DA, Garrido, D, Lerno, L et al. (2012) Bifidobacterium longum subsp. infantis ATCC 15697 alpha-fucosidases are active on fucosylated human milk oligosaccharides. Appl Environ Microbiol 78, 795803.CrossRefGoogle ScholarPubMed
47. Sela, DA, Chapman, J, Adeuya, A et al. (2008) The genome sequence of Bifidobacterium longum subsp. infantis reveals adaptations for milk utilization within the infant microbiome. Proc Natl Acad Sci USA 105, 1896418969.Google Scholar
48. Gonzalez, R, Klaassens, ES, Malinen, E et al. (2008) Differential transcriptional response of Bifidobacterium longum to human milk, formula milk, and galactooligosaccharide. Appl Environ Microbiol 74, 46864694.Google Scholar
49. Martens, EC, Lowe, EC, Chiang, H et al. (2011) Recognition and degradation of plant cell wall polysaccharides by two human gut symbionts. PLoS Biol 9, e1001221.Google Scholar
50. Tannock, GW, Lawley, B, Munro, K et al. (2013) Comparison of the compositions of the stool microbiotas of infants fed goat milk formula, cow milk-based formula, or breast milk. Appl Environ Microbiol 79, 30403048.Google Scholar
51. Penders, J, Stobberingh, E, Thijs, C et al. (2006) Molecular fingerprinting of the intestinal microbiota of infants in whom atopic eczema was or was not developing. Clin Exp Allergy 36, 16021608.Google Scholar
52. Rajilić-Stojanović, M, Heilig, H, Molenaar, D et al. (2009) Development and application of The Human Intestinal Tract Chip (HITChip), a phylogenetic microarray: absence of universally conserved phylotypes in the abundant microbiota of young and elderly adults. Environ Microbiol 11, 17361743.Google Scholar
53. Huse, SM, Ye, Y, Zhou, Y et al. (2012) A core human microbiome as viewed through 16S rRNA sequence clusters. PLoS ONE 7, e34242.CrossRefGoogle ScholarPubMed
54. Hunt, KM, Foster, JA, Forney, LJ et al. (2011) Characterization of the diversity and temporal stability of bacterial communities in human milk. PLoS ONE 6, e21313.Google Scholar
55. Jost, T, Lacroix, C, Braegger, CP et al. (2013) Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding. Environ Microbiol (In the Press).Google Scholar
56. Jost, T, Lacroix, C, Braegger, C et al. (2013) Assessment of bacterial diversity in breast milk using culture-dependent and culture-independent approaches. Br J Nutr 110, 12531262.CrossRefGoogle ScholarPubMed
57. Martin, R, Jimenez, E, Heilig, H et al. (2009) Isolation of bifidobacteria from breast milk and assessment of the bifidobacterial population by PCR-denaturing gradient gel electrophoresis and quantitative real-time PCR. Appl Environ Microbiol 75, 965969.Google Scholar
58. Ward, TL, Hosid, S, Ioshikhes, I et al. (2013) Human milk metagenome: a functional capacity analysis. BMC Microbiol 13, 116.Google Scholar
59. Cabrera-Rubio, R, Collado, MC, Laitinen, K et al. (2012) The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery. Am J Clin Nutr 96, 544551.Google Scholar
60. Collado, MC, Laitinen, K, Salminen, S et al. (2012) Maternal weight and excessive weight gain during pregnancy modify the immunomodulatory potential of breast milk. Pediatr Res 72, 7785.Google Scholar
61. Costello, EK, Lauber, CL, Hamady, M et al. (2009) Bacterial community variation in human body habitats across space and time. Science 326, 16941697.Google Scholar
62. Human Microbiome Project Consortium (2012) Structure, function and diversity of the healthy human microbiome. Nature 486, 207214.CrossRefGoogle Scholar
63. Ursell, LK, Clemente, JC, Rideout, JR et al. (2012) The interpersonal and intrapersonal diversity of human-associated microbiota in key body sites. J Allergy Clin Immunol 129, 12041208.CrossRefGoogle ScholarPubMed
64. Grönlund, MM, Gueimonde, M, Laitinen, K et al. (2007) Maternal breast-milk and intestinal bifidobacteria guide the compositional development of the Bifidobacterium microbiota in infants at risk of allergic disease. Clin Exp Allergy 37, 17641772.CrossRefGoogle ScholarPubMed
65. Boesten, R, Schuren, F, Ben Amor, K et al. (2011) Bifidobacterium population analysis in the infant gut by direct mapping of genomic hybridization patterns: potential for monitoring temporal development and effects of dietary regimens. Microb Biotechnol 4, 417427.Google Scholar
66. Alp, G, Aslim, B, Suludere, Z et al. (2010) The role of hemagglutination and effect of exopolysaccharide production on bifidobacteria adhesion to Caco-2 cells in vitro . Microbiol Immunol 54, 658665.Google Scholar
67. Turroni, F, Foroni, E, Serafini, F et al. (2011) Ability of Bifidobacterium breve to grow on different types of milk: exploring the metabolism of milk through genome analysis. Appl Environ Microbiol 77, 74087417.CrossRefGoogle Scholar
68. Roger, LC, Costabile, A, Holland, DT et al. (2010) Examination of faecal Bifidobacterium populations in breast- and formula-fed infants during the first 18 months of life. Microbiology 156, 33293341.CrossRefGoogle ScholarPubMed
69. Solis, G, de Los Reyes-Gavilan, CG, Fernandez, N et al. (2010) Establishment and development of lactic acid bacteria and bifidobacteria microbiota in breast-milk and the infant gut. Anaerobe 16, 307310.CrossRefGoogle ScholarPubMed
70. Fanning, S, Hall, LJ, Cronin, M et al. (2012) Bifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection. Proc Natl Acad Sci USA 109, 21082113.Google Scholar
71. O'Connell Motherway, M, Zomer, A, Leahy, SC et al. (2011) Functional genome analysis of Bifidobacterium breve UCC2003 reveals type IVb tight adherence (Tad) pili as an essential and conserved host-colonization factor. Proc Natl Acad Sci USA 108, 1121711222.Google Scholar
72. Fouhy, F, Guinane, CM, Hussey, S et al. (2012) High-throughput sequencing reveals the incomplete, short-term recovery of infant gut microbiota following parenteral antibiotic treatment with ampicillin and gentamicin. Antimicrob Agents Chemother 56, 58115820.Google Scholar
73. Dethlefsen, L & Relman, DA (2011) Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci USA 108, S4554S4561.Google Scholar
74. Rea, MC, Dobson, A, O'Sullivan, O et al. (2011) Effect of broad- and narrow-spectrum antimicrobials on Clostridium difficile and microbial diversity in a model of the distal colon. Proc Natl Acad Sci USA 108, S4639S4644.Google Scholar
75. Tanaka, S, Kobayashi, T, Songjinda, P et al. (2009) Influence of antibiotic exposure in the early postnatal period on the development of intestinal microbiota. FEMS Immunol Med Microbiol 56, 8087.CrossRefGoogle ScholarPubMed
76. Hussey, S, Wall, R, Gruffman, E et al. (2011) Parenteral antibiotics reduce bifidobacteria colonization and diversity in neonates. Int J Microbiol doi: 10.1155/2011/130574.Google Scholar
77. Mangin, I, Leveque, C, Magne, F et al. (2012) Long-term changes in human colonic Bifidobacterium populations induced by a 5-day oral amoxicillin-clavulanic acid treatment. PLoS ONE 7, e50257.Google Scholar
78. Mangin, I, Suau, A, Gotteland, M et al. (2010) Amoxicillin treatment modifies the composition of Bifidobacterium species in infant intestinal microbiota. Anaerobe 16, 433438.Google Scholar
79. Jernberg, C, Lofmark, S, Edlund, C et al. (2010) Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology 156, 32163223.Google Scholar
80. Jakobsson, HE, Jernberg, C, Andersson, AF et al. (2010) Short-term antibiotic treatment has differing long-term impacts on the human throat and gut microbiome. PLoS ONE 5, e9836.CrossRefGoogle ScholarPubMed
81. Mårild, K, Ye, W, Lebwohl, B et al. (2013) Antibiotic exposure and the development of coeliac disease: a nationwide case-control study. BMC Gastroenterol 13, 109.CrossRefGoogle ScholarPubMed
82. Foliaki, S, Pearce, N, Björksten, B et al. (2009) Antibiotic use in infancy and symptoms of asthma, rhinoconjunctivitis, and eczema in children 6 and 7 years old: International Study of Asthma and Allergies in Childhood Phase III. J Allergy Clin Immunol 124, 982989.CrossRefGoogle Scholar
83. Ajslev, TA, Andersen, CS, Gamborg, M et al. (2011) Childhood overweight after establishment of the gut microbiota: the role of delivery mode, pre-pregnancy weight and early administration of antibiotics. Int J Obes (Lond) 35, 522529.Google Scholar
84. McKeever, TM, Lewis, SA, Smith, C et al. (2002) The importance of prenatal exposures on the development of allergic disease: a birth cohort study using the West Midlands General Practice Database. Am J Respir Crit Care Med 166, 827832.Google Scholar
85. Metsälä, J, Lundqvist, A, Virta, LJ et al. (2013) Mother's and offspring's use of antibiotics and infant allergy to cow's milk. Epidemiology 24, 303309.Google Scholar
86. Pärtty, A, Luoto, R, Kalliomäki, M et al. (2013) Effects of early prebiotic and probiotic supplementation on development of gut microbiota and fussing and crying in preterm infants: a randomized, double-blind, placebo-controlled trial. J Pediatr 163, 12721277. e1–2.Google Scholar
87. Milidou, I, Sondergaard, C, Jensen, MS et al. (2013) Gestational age, small for gestational age, and infantile colic. Paediatr Perinat Epidemiol 28, 138145.Google Scholar
88. Wessel, MA, Cobb, JC, Jackson, EB et al. (1954) Paroxysmal fussing in infancy, sometimes called colic. Pediatrics 14, 421435.Google Scholar
89. Savino, F, Cordisco, L, Tarasco, V et al. (2009) Molecular identification of coliform bacteria from colicky breastfed infants. Acta Paediatr 98, 15821588.CrossRefGoogle ScholarPubMed
90. Rhoads, JM, Fatheree, NY, Norori, J et al. (2009) Altered fecal microflora and increased fecal calprotectin in infants with colic. J Pediatr 155, 823828.Google Scholar
91. Pärtty, A, Kalliomäki, M, Endo, A et al. (2012) Compositional development of Bifidobacterium and Lactobacillus microbiota is linked with crying and fussing in early infancy. PLoS ONE 7, e32495.Google Scholar
92. Roos, S, Dicksved, J, Tarasco, V et al. (2013) 454 pyrosequencing analysis on faecal samples from a randomized DBPC trial of colicky infants treated with Lactobacillus reuteri DSM 17938. PLoS ONE 8, e56710.Google Scholar
93. Chichlowski, M, De Lartigue, G, German, JB et al. (2012) Bifidobacteria isolated from infants and cultured on human milk oligosaccharides affect intestinal epithelial function. J Pediatr Gastroenterol Nutr 55, 321327.Google Scholar
94. Ewaschuk, JB, Diaz, H, Meddings, L et al. (2008) Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function. Am J Physiol Gastrointest Liver Physiol 295, G1025G1034.Google Scholar
95. Jalanka-Tuovinen, J, Salonen, A, Nikkilä, J et al. (2011) Intestinal microbiota in healthy adults: temporal analysis reveals individual and common core and relation to intestinal symptoms. PLoS ONE 6, e23035.Google Scholar
96. Savino, F, Cordisco, L, Tarasco, V et al. (2011) Antagonistic effect of Lactobacillus strains against gas-producing coliforms isolated from colicky infants. BMC Microbiol 11, 157.Google Scholar
97. Vanhoutvin, SA, Troost, FJ, Kilkens, TO et al. (2009) The effects of butyrate enemas on visceral perception in healthy volunteers. Neurogastroenterol Motil 21, 952–e76.Google Scholar
98. Burger-van Paassen, N, Vincent, A, Puiman, PJ et al. (2009) The regulation of intestinal mucin MUC2 expression by short-chain fatty acids: implications for epithelial protection. Biochem J 420, 211219.Google Scholar
99. Ma, X, Fan, PX, Li, LS et al. (2012) Butyrate promotes the recovering of intestinal wound healing through its positive effect on the tight junctions. J Anim Sci 90, S266S268.Google Scholar
100. Wang, HB, Wang, PY, Wang, X et al. (2012) Butyrate enhances intestinal epithelial barrier function via up-regulation of tight junction protein Claudin-1 transcription. Dig Dis Sci 57, 31263135.Google Scholar
101. Savino, F, Cordisco, L, Tarasco, V et al. (2010) Lactobacillus reuteri DSM 17938 in infantile colic: a randomized, double-blind, placebo-controlled trial. Pediatrics 126, e526e533.Google Scholar
102. Indrio, F, Riezzo, G, Raimondi, F et al. (2011) Lactobacillus reuteri accelerates gastric emptying and improves regurgitation in infants. Eur J Clin Invest 41, 417422.Google Scholar
103. Pärtty, A & Isolauri, E (2012) Gut microbiota and infant distress – the association between compositional development of the gut microbiota and fussing and crying in early infancy. Microb Ecol Health Dis 23 (online publication; doi:10.3402/mehd.v23i0.18577).Google Scholar
104. Sung, V, Hiscock, H, Tang, ML et al. (2014) Treating infant colic with the probiotic Lactobacillus reuteri: double blind, placebo controlled randomised trial. Br Med J 348, g2107.Google Scholar
105. Bieber, T (2010) Atopic dermatitis. Ann Dermatol 22, 125137.Google Scholar
106. Deckers, IA, McLean, S, Linssen, S et al. (2012) Investigating international time trends in the incidence and prevalence of atopic eczema 1990–2010: a systematic review of epidemiological studies. PLoS ONE 7, e39803.Google Scholar
107. Abrahamsson, TR, Jakobsson, HE, Andersson, AF et al. (2012) Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin Immunol 129, 434440. e2.Google Scholar
108. Bisgaard, H, Li, N, Bonnelykke, K et al. (2011) Reduced diversity of the intestinal microbiota during infancy is associated with increased risk of allergic disease at school age. J Allergy Clin Immunol 128, 646652. e1–5.Google Scholar
109. Forno, E, Onderdonk, AB, McCracken, J et al. (2008) Diversity of the gut microbiota and eczema in early life. Clin Mol Allergy 6, 11.Google Scholar
110. Wang, M, Karlsson, C, Olsson, C et al. (2008) Reduced diversity in the early fecal microbiota of infants with atopic eczema. J Allergy Clin Immunol. 121, 129134.Google Scholar
111. Ismail, IH, Oppedisano, F, Joseph, SJ et al. (2012) Reduced gut microbial diversity in early life is associated with later development of eczema but not atopy in high-risk infants. Pediatr Allergy Immunol 23, 674681.Google Scholar
112. Kalliomäki, M, Kirjavainen, P, Eerola, E et al. (2001) Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy clin Immunol 107, 129134.Google Scholar
113. Rosenfeldt, V, Benfeldt, E, Valerius, NH et al. (2004) Effect of probiotics on gastrointestinal symptoms and small intestinal permeability in children with atopic dermatitis. J Pediatr 145, 612616.Google Scholar
114. Sjögren, YM, Tomicic, S, Lundberg, A et al. (2009) Influence of early gut microbiota on the maturation of childhood mucosal and systemic immune responses. Clin Exp Allergy 39, 18421851.Google Scholar
115. Hooper, LV & Macpherson, AJ (2010) Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat Rev Immunol 10, 159169.Google Scholar
116. Maynard, CL, Elson, CO, Hatton, RD et al. (2012) Reciprocal interactions of the intestinal microbiota and immune system. Nature 489, 231241.CrossRefGoogle ScholarPubMed
117. Sepp, E, Julge, K, Mikelsaar, M et al. (2005) Intestinal microbiota and immunoglobulin E responses in 5-year-old Estonian children. Clin Exp Allergy 35, 11411146.Google Scholar
118. Stsepetova, J, Sepp, E, Julge, K et al. (2007) Molecularly assessed shifts of Bifidobacterium ssp. and less diverse microbial communities are characteristic of 5-year-old allergic children. FEMS Immunol Med Microbiol 51, 260269.Google Scholar
119. Johansson, MA, Sjögren, YM, Persson, JO et al. (2011) Early colonization with a group of Lactobacilli decreases the risk for allergy at five years of age despite allergic heredity. PLoS ONE 6, e23031.Google Scholar
120. Gore, C, Munro, K, Lay, C et al. (2008) Bifidobacterium pseudocatenulatum is associated with atopic eczema: a nested case-control study investigating the fecal microbiota of infants. J Allergy Clin Immunol 121, 135140.Google Scholar
121. Mah, K, Chin, V, Wong, W et al. (2007) Effect of a milk formula containing probiotics on the fecal microbiota of asian infants at risk of atopic diseases. Pediatr Res 62, 674679.Google Scholar
122. Björksten, B, Sepp, E, Julge, K et al. (2001) Allergy development and the intestinal microflora during the first year of life. J Allergy Clin Immunol 108, 516520.Google Scholar
123. Sjögren, YM, Jenmalm, MC, Böttcher, MF et al. (2009) Altered early infant gut microbiota in children developing allergy up to 5 years of age. Clin Exp Allergy 39, 518526.Google Scholar
124. Storro, O, Oien, T, Langsrud, O et al. (2011) Temporal variations in early gut microbial colonization are associated with allergen-specific immunoglobulin E but not atopic eczema at 2 years of age. Clin Exp Allergy 41, 15451554.Google Scholar
125. Thompson-Chagoyan, OC, Fallani, M, Maldonado, J et al. (2011) Faecal microbiota and short-chain fatty acid levels in faeces from infants with cow's milk protein allergy. Int Arch Allergy Immunol 156, 325332.Google Scholar
126. Hooper, LV, Wong, MH, Thelin, A et al. (2001) Molecular analysis of commensal host-microbial relationships in the intestine. Science 291, 881884.Google Scholar
127. Pagnini, C, Saeed, R, Bamias, G et al. (2010) Probiotics promote gut health through stimulation of epithelial innate immunity. Proc Natl Acad Sci USA 107, 454459.Google Scholar
128. Mazmanian, SK, Liu, CH, Tzianabos, AO et al. (2005) An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122, 107118.Google Scholar
129. Round, JL, Lee, SM, Li, J et al. (2011) The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 332, 974977.Google Scholar
130. Alyasin, S, Karimi, MH, Amin, R et al. (2013) Interleukin-17 gene expression and serum levels in children with severe asthma. Iran J Immunol 10, 177185.Google Scholar
131. Ramirez-Velazquez, C, Castillo, EC, Guido-Bayardo, L et al. (2013) IL-17-producing peripheral blood CD177+ neutrophils increase in allergic asthmatic subjects. Allergy Asthma Clin Immunol 9, 23.Google Scholar
132. Brandtzaeg, P (2009) Mucosal immunity: induction, dissemination, and effector functions. Scand J Immunol 70, 505515.Google Scholar
133. Johansen, FE, Pekna, M, Norderhaug, IN et al. (1999) Absence of epithelial immunoglobulin A transport, with increased mucosal leakiness, in polymeric immunoglobulin receptor/secretory component-deficient mice. J Exp Med 190, 915922.CrossRefGoogle ScholarPubMed
134. Kukkonen, K, Kuitunen, M, Haahtela, T et al. (2010) High intestinal IgA associates with reduced risk of IgE-associated allergic diseases. Pediatr Allergy Immunol 21, 6773.Google Scholar
135. Nermes, M, Kantele, JM, Atosuo, TJ et al. (2011) Interaction of orally administered Lactobacillus rhamnosus GG with skin and gut microbiota and humoral immunity in infants with atopic dermatitis. Clin Exp Allergy 41, 370377.Google Scholar
136. Elazab, N, Mendy, A, Gasana, J et al. (2013) Probiotic administration in early life, atopy, and asthma: a meta-analysis of clinical trials. Pediatrics 132, e666e676.Google Scholar
137. Mantis, NJ, Rol, N & Corthesy, B (2011) Secretory IgA's complex roles in immunity and mucosal homeostasis in the gut. Mucosal Immunol 4, 603611.Google Scholar
138. Rautava, S, Arvilommi, H & Isolauri, E (2006) Specific probiotics in enhancing maturation of IgA responses in formula-fed infants. Pediatr Res 60, 221224.Google Scholar
139. Di Mauro, A, Neu, J, Riezzo, G et al. (2013) Gastrointestinal function development and microbiota. Ital J Pediatr 39, 15.Google Scholar
140. Kwon, HK, Lee, CG, So, JS et al. (2010) Generation of regulatory dendritic cells and CD4+Foxp3+ T cells by probiotics administration suppresses immune disorders. Proc Natl Acad Sci USA 107, 21592164.Google Scholar
141. Lyons, A, O'Mahony, D, O'Brien, F et al. (2010) Bacterial strain-specific induction of Foxp3+ T regulatory cells is protective in murine allergy models. Clin Exp Allergy 40, 811819.Google Scholar
142. Marschan, E, Kuitunen, M, Kukkonen, K et al. (2008) Probiotics in infancy induce protective immune profiles that are characteristic for chronic low-grade inflammation. Clin Exp Allergy 38, 611618.Google Scholar
143. Torii, S, Torii, A, Itoh, K et al. (2011) Effects of oral administration of Lactobacillus acidophilus L-92 on the symptoms and serum markers of atopic dermatitis in children. Int Arch Allergy Immunol 154, 236245.Google Scholar
144. West, CE, Hammarstrom, ML & Hernell, O (2009) Probiotics during weaning reduce the incidence of eczema. Pediatr Allergy Immunol 20, 430437.Google Scholar
145. Green, PH & Jabri, B (2006) Celiac disease. Annu Rev Med 57, 207221.Google Scholar
146. Murch, S, Jenkins, H, Auth, M et al. (2013) Joint BSPGHAN and Coeliac UK guidelines for the diagnosis and management of coeliac disease in children. Arch Dis Child 98, 806811.Google Scholar
147. Stene, LC, Honeyman, MC, Hoffenberg, EJ et al. (2006) Rotavirus infection frequency and risk of celiac disease autoimmunity in early childhood: a longitudinal study. Am J Gastroenterol 101, 23332340.Google Scholar
148. Myleus, A, Hernell, O, Gothefors, L et al. (2012) Early infections are associated with increased risk for celiac disease: an incident case-referent study. BMC Pediatr 12, 194.Google Scholar
149. Sellitto, M, Bai, G, Serena, G et al. (2012) Proof of concept of microbiome-metabolome analysis and delayed gluten exposure on celiac disease autoimmunity in genetically at-risk infants. PLoS ONE 7, e33387.Google Scholar
150. Ivarsson, A, Persson, LA, Nystrom, L et al. (2000) Epidemic of coeliac disease in Swedish children. Acta Paediatr 89, 165171.Google Scholar
151. Sanz, Y, De Pama, G & Laparra, M (2011) Unraveling the ties between celiac disease and intestinal microbiota. Int Rev Immunol 30, 207218.Google Scholar
152. De Palma, G, Nadal, I, Medina, M et al. (2010) Intestinal dysbiosis and reduced immunoglobulin-coated bacteria associated with coeliac disease in children. BMC Microbiol 10, 63.Google Scholar
153. Schippa, S, Iebba, V, Barbato, M et al. (2010) A distinctive ‘microbial signature’ in celiac pediatric patients. BMC Microbiol 10, 175.Google Scholar
154. de Meij, TG, Budding, AE, Grasman, ME et al. (2013) Composition and diversity of the duodenal mucosa-associated microbiome in children with untreated coeliac disease. Scand J Gastroenterol 48, 530536.Google Scholar
155. Kalliomäki, M, Satokari, R, Lähteenoja, H et al. (2012) Expression of microbiota, Toll-like receptors, and their regulators in the small intestinal mucosa in celiac disease. J Pediatr Gastroenterol Nutr 54, 727732.Google Scholar
156. Nistal, E, Caminero, A, Herran, AR et al. (2012) Differences of small intestinal bacteria populations in adults and children with/without celiac disease: effect of age, gluten diet, and disease. Inflamm Bowel Dis 18, 649656.Google Scholar
157. Cheng, J, Kalliomäki, M, Heilig, HG et al. (2013) Duodenal microbiota composition and mucosal homeostasis in pediatric celiac disease. BMC Gastroenterol 13, 113.Google Scholar
158. Forsberg, G, Fahlgren, A, Horstedt, P et al. (2004) Presence of bacteria and innate immunity of intestinal epithelium in childhood celiac disease. Am J Gastroenterol 99, 894904.Google Scholar
159. Ou, G, Hedberg, M, Horstedt, P et al. (2009) Proximal small intestinal microbiota and identification of rod-shaped bacteria associated with childhood celiac disease. Am J Gastroenterol 104, 30583067.Google Scholar
160. Nadal, I, Donat, E, Ribes-Koninckx, C et al. (2007) Imbalance in the composition of the duodenal microbiota of children with coeliac disease. J Med Microbiol 56, 16691674.Google Scholar
161. Sanchez, E, Donat, E, Ribes-Koninckx, C et al. (2013) Duodenal-mucosal bacteria associated with celiac disease in children. Appl Environ Microb 79, 54725479.Google Scholar
162. Di Cagno, R, De Angelis, M, De Pasquale, I et al. (2011) Duodenal and faecal microbiota of celiac children: molecular, phenotype and metabolome characterization. BMC Microbiol 11, 219.Google Scholar
163. Collado, MC, Donat, E, Ribes-Koninckx, C et al. (2009) Specific duodenal and faecal bacterial groups associated with paediatric coeliac disease. J Clin Pathol 62, 264269.Google Scholar
164. Smecuol, E, Hwang, HJ, Sugai, E et al. (2013) Exploratory, randomized, double-blind, placebo-controlled study on the effects of Bifidobacterium infantis natren life start strain super strain in active celiac disease. J Clin Gastroenterol 47, 139147.Google Scholar
165. Sanchez, E, De Palma, G, Capilla, A et al. (2011) Influence of environmental and genetic factors linked to celiac disease risk on infant gut colonization by Bacteroides species. Appl Environ Microbiol 77, 53165323.Google Scholar
166. Palma, GD, Capilla, A, Nova, E et al. (2012) Influence of milk-feeding type and genetic risk of developing coeliac disease on intestinal microbiota of infants: the PROFICEL study. PLoS ONE 7, e30791.Google Scholar
Figure 0

Fig. 1. (colour online) Modern life style factors associated with the development of intestinal microbiota and later life health.