Download PDF
Review  |  Open Access  |  23 Sep 2024

A microbial symphony: a literature review of the factors that orchestrate the colonization dynamics of the human colonic microbiome during infancy and implications for future health

Views: 209 |  Downloads: 58 |  Cited:  1
Microbiome Res Rep 2024;3:50.
10.20517/mrr.2024.32 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

Since the advent of new sequencing and bioinformatic technologies, our understanding of the human microbiome has expanded rapidly over recent years. Numerous studies have indicated causal links between alterations to the microbiome and a range of pathological conditions. Furthermore, a large body of epidemiological data is starting to suggest that exposure, or lack thereof, to specific microbial species during the first five years of life has key implications for long-term health outcomes. These include chronic inflammatory and metabolic conditions such as diabetes, asthma, inflammatory bowel disease (IBD), and obesity, with the effects lasting into adulthood. Human microbial colonisation during these first five years of life is a highly dynamic process, with multiple environmental exposures recently being characterised to have influence before the microbiome stabilises and resembles that of an adult at 3-5 years. This short period of time, known as the window of opportunity, appears to “prime” immunoregulation for later life. Understanding and appreciating this aspect of human physiology is therefore crucial for clinicians, scientists, and public health officials. This review outlines the most recent evidence for the pre- and post-natal environments that order the development of the microbiome, how these influences metabolic and immunoregulatory pathways, and their associated health outcomes. It also discusses the limitations of the current knowledge base, and describes the potential microbiome-mediated interventions and public health measures that may have therapeutic potential in the future.

Keywords

Microbiome, paediatric, infant, inflammation, IBD, asthma, diabetes, newborn, microbiology, birth

INTRODUCTION

The term “microbiome” was first coined by Professor John Whipps in 1988 to describe the microorganisms living in soil[1]. Colloquially, as well as in the scientific and medical lexicon, the term has anthropomorphically shifted, with the assumption of reference being to the human microbial ecosystem rather than the rhizosphere. The human microbiome is defined as the totality of microbial species found living in and on a human - that is, all the species of bacteria, archaea, fungi, algae, small protists, bacteriophages, and viruses, as well as extracellular DNA[2,3]. It also encompasses their “theatre of activity” - the secondary metabolites produced by these communities [e.g., toxins, lipopeptides, polysaccharides, signalling molecules, and other (in)organic matter] that often have important metabolic and immunogenic effects. The human microbiome is diverse, with populations differing greatly depending on anatomical location. As an example, the species found in the alar crease are significantly different from those in the inguinal crease, despite both being on the skin surface[4].

The human-microbiome symbiosis should be viewed as one of co-evolution, with the microbiome aiding in the metabolism of food into short-chain fatty acids (SCFAs) (e.g., fermentation of the otherwise poorly digested plant polysaccharides and unhydrolyzed starches), the production of vitamins (e.g., thiamine, folate, riboflavin, pantothenic acid, biotin and vitamin K), and the control and competitive exclusion of pathogenic bacteria[5]. In response, the host provides a stable and rich environment with sophisticated immunological mechanisms to sense and control specific species[6]. Archaeological evidence from coprolites, dental plaque and tissue stored in permafrost have provided insight into how our microbiome has shifted away from that of the great apes to one of lower alpha diversity, with particularly decreased Methanobrevibacter and Fibrobacter, and significantly more Bacteroides species[7]. The recent (in evolutionary timescales) agricultural revolution, adoption of using heat to cook food, and Westernised diet (one of high meat and ultra-processed food consumption) have induced a rapid change in the makeup of our microbiome, which appears to have introduced mal-adaptions that are involved in diseases of modernity[8,9].

The contemporary literature reports some ~3.8 × 1013 bacteria dwelling in the 70 kg “reference” male; however, it is the colon that is the true microbial powerhouse, making up 92% of the total microbiome[10], and unless otherwise stated, hereon microbiome will be in reference to the human colonic microbiome. To regulate this, the gastrointestinal tract (GIT) maintains a fine balance of immune regulation and, accordingly, is the largest immune organ in the body[11]. This complex system involves the sensing of symbiotic species without stimulating the immune system, while detecting and mounting immune responses to specific pathogens[12]. If the interplay between this masterly inactivity and overactive defence mechanisms goes astray, there are numerous negative health-related consequences, ranging from inflammatory bowel disease (IBD), diabetes, cancer, obesity, cardiovascular disease, hypertension, depression, and anxiety (to name but a few)[13]. The “priming”, or education, of what the GIT immune system should consider a pathogen is, therefore, vital for future health[12]. The evidence suggests that this process is largely determined during the first five years of life; thus, it is of the utmost importance to fully understand and characterise the colonisation of a healthy microbiome during infancy.

Accordingly, this paper will review the latest literature on colonisation dynamics during the early years of life and what variables are at play. Specifically, we will seek out the mechanistic evidence for how bacterial species are transferred from one environment to the colonic microbiome of an individual, and discuss how this can be potentially utilised for public health. We will discuss any gaps and limitations of the scientific knowledge base and where the future of this new discipline is going.

MICROBIOME VARIATION WITH AGE

The first months and years of life are a very dynamic period for the microbiome, with wide-reaching consequences for future health. Several large international longitudinal studies have identified three distinct periods of rapid and dynamic microbiomic progression[14-17]. These can be summarised as a developmental phase (the first 12 months of life) that is dominated by the effects of breastfeeding, a transitional phase (the second year of life, or the cessation of breast milk feeding) where the microbiome is highly plastic and adjusts to solid food, and culminating in a stable phase at approximately 36-60 months that closely resembles the microbiome of an adult[14,18].

Starting with a blank slate?

There has been a long-held belief that the human foetus develops in a sterile uterine environment[19]. The origin of this goes back to research that was unable to detect microbial life in the meconium, amniotic fluid, or placenta using traditional culture-based methods and microscopy[20]. Indeed, chorioamnionitis, an intra-amniotic infection typically secondary to E. coli or group B streptococcus, is associated with miscarriage, preterm birth, and increased neonatal mortality, ergo the presence of bacteria was deemed to be an abnormal phenomenon.

New techniques, however, such as next-generation sequencing (NGS), have transformed the detection and identification of microbiota. A growing body of evidence has raised questions over the “sterile uterus hypothesis”. Aagaard et al. were the first group to systematically describe a microbiome of placental samples from both healthy pregnancies and preterm births. They found a low-abundance but metabolically rich microbiome, which interestingly reflected the mother’s dental microbiome, raising the possibility of haematogenous spread from the oral cavity to the placenta[21]. Several other studies have since published evidence for distinct microbiomes relating to the placenta, amniotic fluid, and meconium in healthy neonates, resembling the faecal, oral, and vaginal microbiome of the mother[22-24]. Furthermore, animal models have shown a similar effect. A Spanish group inoculated pregnant BALB/c mice with an oral solution of a tagged mutant strain of Enterococcus faecium. This strain was then detected from samples of amniotic fluid taken at the time of caesarean section (CS), an effect not seen in the control group[25]. In our view, this is perhaps the most compelling evidence against the sterile uterus hypothesis.

This antithesis to the canon has caused controversy, however. Several papers have subsequently highlighted methodological errors pertaining to contamination from both the collection method (e.g., epidermal contamination from the puncture site for amniocentesis, or contamination during veterinary CS), and the typical 48 h delay in collecting meconium that might allow ex-utero colonisation to occur[26-28]. Accordingly, an international group repeated these experiments, with more rigorous controls for contamination, and failed to find evidence of intra-uterine colonization of the foetal GIT[29].

An international consensus statement has made clear that the principal viewpoint of the academic community is still that of the sterile womb, citing, amongst several reasons, the high rates of false-positive results that exquisitely sensitive NGS can produce[28]. It should be noted that even in the event of true-positive results, bacterial DNA does not equate to living and metabolically active bacterial species that have colonised the foetal microbiome. Furthermore, there have been over seven decades of germ-free animal experimentation[30], where mammals are born via CS at term. A valid assumption follows that if in-utero colonisation had occurred, this would have been detected in the numerous control groups during this time[27].

Perhaps a nuanced slant to this, with increasing experimental data behind it, would be that immunogenic microbial products (bacterial DNA fragments, and metabolites) from the mother’s commensal microbiota are able to cross the placental barrier, giving the opportunity to educate the foetal immune system and promote immune tolerance before birth[31-33]. With the rapid progress in ex-utero embryogenesis technology[34], it is conceivable that in the near future, an animal model will be able to categorically prove if these immunogenic microbial products have any demonstrable relevance to foetal immunogenic and microbiomic development. For now, the evidence is insufficient to move away from the orthodoxy of the sterile womb hypothesis.

Birth

The very first moment of life has direct implications for the newborn’s microbiome. Delivery by CS or vaginal delivery (VD) has been shown to result in divergent neonatal microbiomes. This is believed to be due to the direct contact of the neonate with the maternal vaginal and perineal microbiome, allowing direct vertical transmission to occur. Although very few mechanistic studies have been performed, transmission to the neonatal colon is believed to be via the oral route, a process phrased as the “bacterial baptism of birth”[35,36]. An interesting physiological adaption arises during the third trimester that confers a survival benefit to the child. In response to increasing oestrogen and progesterone, the mother’s vaginal microbiome is altered, a decrease in alpha diversity occurs, and the abundance of Lactobacillus spp. increases[37,38]. These species produce lactic acid, decreasing vaginal pH and consequentially reducing acid-sensitive pathobionts such as Group B Streptococcus[38], which carry a high rate of neonatal sepsis and mortality.

This “bacterial baptism” during VD is inherently absent in CS; instead, other microbial environments appear to play a role and lead to divergent microbiomes in the newborn. Multiple large studies have demonstrated that VD is associated with increased abundances of Bacteroidetes (particularly Bacteroides fragilis), Bacillota (chiefly Lactobacillus spp.), and Actinomycetota (almost exclusively from the Bifidobacterium spp.), all of which are typically found in the vagina or recto-anal microbiomes. In contrast, CS has been associated with species typically found on maternal skin and microbiomes found in the hospital, with neonates having an increased abundance of Bacillota (notably Staphylococcus spp.), Firmicutes, Enterococcus, and Klebsiella[14,39-46].

An increasing number of epidemiological studies have demonstrated a correlation between CS and an increased risk for negative long-term infant health outcomes. These findings are diverse and not always consistent, but there are reported increases in risk for type 1 diabetes mellitus (T1DM)[47-49], obesity[48,50], asthma[51,52], eczema[53], respiratory tract infections[54], coeliac disease[55], IBD[56,57], attention deficit/hyperactivity disorder[58], and autism[59]. A recent meta-analysis of the paediatric consequences of CS, with over two million subjects, demonstrated increased risks for childhood asthma [OR 1.23 (1.14, 1.33)], T1DM [OR 1.07 (0.90, 1.27)], and obesity [OR 1.35 (1.29, 1.41) ][43]. With more than 25% of births being CS in Europe and North America, the potential implications to long-term health are a significant public health concern[60,61]. However, it is still unclear what causes these phenomena. Several explanations have been proposed, from the lack of stress hormones during an elective CS to an absence of intraocular pressure being suggested to alter the inflammatory response of the newborn[62]. However, the leading hypothesis relates to the divergent microbiomes associated with these methods of delivery[63]. The first 24 h of life have been shown to be a prime opportunity for bacterial colonisation of the colon, in that the pH of the stomach is relatively neutral thanks to the ingestion of amniotic fluid in utero[43]. This has been shown to complement the vertical transmission during VD of Lactobacillus, Bifidobacterium, and Bacteroides. These have been shown to regulate the developing immune system, influence the concentration of natural killer (NK) cells, regulate the population of T-lymphocytes, increase the secretion of Immunoglobulin A (IgA) antibodies, and aid in the metabolism of human milk oligosaccharides (HMOs)[64-70]. Furthermore, Bifidobacterium has been shown in both human and animal studies to reduce the risk of necrotising enterocolitis in newborns[71].

This microbiomic theory of disease has a large and convincing body of evidence to support it. However, when looking at the literature, there are two important deficits that require attention. Firstly, the majority of studies showing metagenomic differences in VD and CS children, while replicated many times, were not designed to determine why these differences occur. Are the differences related to the method of delivery itself - i.e., is the vaginal microbiome the only variable, or are there other unrecognised environmental factors at play? Studies traditionally have used genetic operational taxonomic units (OTUs) to show significant similarity in intra-mother-child dyads vs. inter-dyads[72]; thereby, evidence of VD vertical transmission is only strongly inferred. To the best of our knowledge, there have been no mechanistic studies (e.g., using tagged bacterial species) to definitively show colonisation from the vaginal microbiome to the neonate’s GIT. Indeed, Bacteroides fragilis, the species that correlates strongly with VD, is only found in 6.6% of the vaginal microbiome of pregnant women, suggesting inoculation from an environment other than the vagina[73]. Interestingly, studies using cultured vaginal bacteria from the mother, administered orally to the neonate shortly after CS, have reported both positive and negative effects on the infant’s microbiome compared to placebo, raising questions over the extent played by VD in forming the newborn’s microbiome[74,75]. In a similar vein, there is an increasing trend of the lay press in the advocation of “vaginal seeding”, inoculating a swab or gauze with vaginal fluid to transfer the flora to a newborn. More research is urgently needed to assess not only the efficacy of this but also to assess for any safety concerns, a view reflected by the American College of Obstetricians and Gynaecologists[76]. Another variable that may influence the paradigm is the routine use of preoperative antibiotics during CS, which has significant effects on the neonatal microbiome[77]. The evidence suggests that the routine use of pre-cord clamping antibiotics significantly reduces the abundance of bacteria, predominantly from reduced Clostridium spp., an effect that can still be observed after one year of age[77]. Interest in antibiotic usage after cord clamping is increasing internationally, where the antibiotic compound does not reach the foetus. Randomised control trials of this method have shown that it successfully mitigates the effects on the neonatal microbiome[77]. Furthermore, a Swiss lead non-inferiority trial found that the technique had no negative effect on the mother (e.g., surgical site infection), and its routine adoption into clinical practice should be encouraged[78]. However, it should be noted that there is conflicting evidence on this; a Dutch group has recently reported no significant difference in microbiomic composition between infants born to CS with antibiotics given pre- vs. post-cord clamping[79]. Finally, maternal factors seem to influence microbiome dynamics, with one study showing a significant difference in children born by emergency CS compared to elective CS, suggesting that antenatal health regulates the microbiomic outcome[80]. Recent papers have shown that the differences in the microbiome of CS vs. VD are very short-lived. In one large longitudinal study using whole genome shotgun (WGS) metagenomic analysis, a more sensitive method compared to the commonly used 16S rRNA sequencing, the microbiome between CS and VD becomes indistinguishable at six weeks[81]. This provides a very limited window of opportunity for such a dynamic microbiome to determine long-term health outcomes for the child.

The second aspect to mention is the absence of prospective trials showing a pathogenic mechanism linking CS and negative long-term health outcomes. The evidence that does exist, exclusively from epidemiological studies, has been inconsistent in controlling for variables that are known to impact the health of a newborn. These include: (i) maternal and neonatal co-morbidity during pregnancy and after birth; (ii) rates of breastfeeding (known to be lower after CS); (iii) the impact of a hospital microbiome compared to a maternity unit / home birth; (iv) increased maternal age, all of which are known to be independently associated with paediatric health[82]. This list is not exhaustive and, importantly, neglects the unknown unknowns. Moreover, several of these variables, such as maternal co-morbidity, also independently increase the likelihood of having a CS, potentially compounding the effect.

Breastfeeding (the developmental phase)

During this period, even when accounting for the mode of delivery, the most significant factor associated with the structure of the microbiome is the receipt of breast milk, either exclusively or partially[14]. Contrary to previously held dogma, human breast milk is not sterile[83] and has been found to be rich in the genera Bacillus, Bifidobacterium, Enterococcus, Lactobacillus, Lactococcus, Weisella, Staphylococcus, and Streptococcus[84-87]. One paper has recorded a median bacterial load of 106 cells/mL of breast milk, suggesting that exclusively breastfed (EBF) infants consume approximately 8 billion bacteria per day[88]. The origin of the lactational mammary duct microbiome is complex and likely multifaceted. DNA sequencing of the milk has demonstrated species otherwise typically found in the oral cavity and maternal skin. It is reasonable to assume colonisation occurs during suckling of the infant as well as from direct contact with adjacent skin microbiomes. This, however, does not explain the presence of obligate anaerobes found in milk that are prototypical of an adult’s colonic microbiome - Enterococcus, Bifidobacterium, Bacteroides, Clostridium, and Faecalibacterium[89,90]. Recent evidence has suggested an entero-mammary pathway whereby bacterial cells from the colonic lumen are phagocytosed by dendritic and CD18+ cells, taken into the systemic lymphatic and cardiovascular circulation and then deposited in the mammary gland[91]. This mechanism is analogous to the established entero-mammary circulation, where B cells migrate to the mammary epithelium and release secretory (S) IgA into the milk supply[92]. If this mode of bacterial translocation is confirmed with mechanistic studies, it would provide a clear route for the initial colonisation of strict anaerobic species that are not typically found on surface environments, and the absence of this mode of transmission could have potentially profound consequences on the development of the colonic immune system. This may go some way to explain how certain bacterial species that are exquisitely vulnerable to atmospheric conditions can reach the human GIT. For example, Akkermansia mucinophilia, a species that is inversely related to IBD and obesity, is notoriously difficult to culture and survives for very short periods under atmospheric conditions[93]; thus, it has no known extra-colonic environmental niche to provide a stable supply for colonisation. A recent study has detected this species in the colostrum and milk supply from mothers[94], perhaps being a key moment for this, and similar species, to establish themselves for permanent colonisation of the newborn’s GIT. It has previously been demonstrated that maternal diet affects the milk microbiome[95], and this may be a branch of interesting future research to develop specialised maternal diets to modulate the developing neonatal microbiome with the aim of improving long-term health outcomes.

Infants fed exclusively formula milk (FM) do not receive these bacterial species. Accordingly, they have remarkably different microbiomes to EBF infants. One meta-analysis of 684 infants[96] from five countries showed an increased abundance of Bacteroides, Eubacterium, Firmicutes, and Veillonella in formula-fed babies. EBF infants had a lower alpha diversity due to the increased abundance of the genus Bifidobacterium. This increased Bifidobacterium abundance is associated with a growth in microbial metabolic pathways favouring lipid and vitamin metabolism. Several studies have taken these findings, along with the increased Bacteroides [associated with increased body mass index (BMI)], to suggest that FM may cause obesity, diabetes, and other adverse health outcomes in adulthood as a result of the altered nutrient metabolism in the colon[97,98]. Furthermore, the differences in microbiomes between infants who are EBF and FM appear to be subtly more pronounced if the mode of delivery was CS. The evidence suggests that the relative absence of anaerobes (typically found in the microbiota of CS-delivered children) is exacerbated for infants who are then subsequently on FM[99], particularly in Proteobacteria, suggesting an increased importance of EBF after CS. Another interesting finding is the microbiomic difference between infants who are EBF and those who receive human breast milk from a bottle[100]. For instance, while metagenomic studies of fresh milk and milk stored at 4 °C show no significant difference, there is a significant reduction in viable bacterial species - for instance, Lactobacillus and Streptococcus spp., possibly explaining the differences in the microbiome of these two cohorts of children (i.e., fresh human breast milk vs. refrigerated human breast milk)[101].

Human breast milk has a complex mixture of essential macronutrients that help a newborn’s normal development, as well as acting as a prebiotic. It also contains immunogenic compounds, such as SIgA (and, to a lesser extent, IgM and IgG), which provide a degree of passive immunity for the newborn to specific pathogens[102,103]. As with the mode of delivery, the difference between the microbiomes of children who are formula- vs. breast milk-fed has been proposed as a causative factor for several associated pathologies. Voluminous epidemiological studies and the cumulative power of their meta-analyses have shown clear associations between breastfeeding and beneficial health outcomes. The comprehensive list includes a reduced risk of: asthma[104]*, eczema[104], respiratory infections[105]*, IBD[106]*, infectious diarrhoeal diseases[105,107]*, leukaemia[108]*, diabetes melitus[109,110], obesity[111]*, acute otitis media[112], dental caries[113], and general intelligence[114]. Several of these papers have gone further to demonstrate a protective dose response to the aforementioned (*) pathologies, giving credence to a mechanistic link. The precise time when the benefits from EBF start to taper off is not clear, with conflicting evidence between four and six months as the cut-off times. A Cochrane review found that the balance of evidence supports EBF for six months, which is reflected in the international guidance[115-117].

After breastfeeding (the transitional and stable phases)

At approximately 12 months, coinciding when the majority of infants have completed weaning, the microbiome changes sharply. The cessation of EBF and the introduction of solid food results in a shift towards species that are predominantly fermenters of plant-based polysaccharides and fibres, converting them into SCFAs, such as butyrate[17]. SCFAs are known to be essential for colonocyte metabolic health, as well as having anti-inflammatory and anti-carcinogenic properties[118]. This is characterised by an increase in Bacteroides, Clostridium, Enterobacteria, Enterococci, and Streptococcus[17,119]. Infants with a high-fibre diet display a considerable increase in Bacteroidetes[120]. In the distal colon, these phyla further contribute to the host through the biosynthesis of vitamins and essential amino acids[119]. It has been suggested that a westernised diet during this period reduces the populations of Bacteroidetes and Prevotella, which may lead to dysbiosis and partly explain the increased risk of IBD[119]. Interestingly, this shift in the microbiome is most apparent when breastfeeding stops, rather than when food is introduced[17]. This effect is less evident in formula-fed infants as their diet already included complex nutritional compounds that had resulted in a more diverse microbiome earlier in life[121]. After 12-24 months, this highly plastic phase starts to stabilise and resembles the microbiome of adults. It is during this period that other environmental stimuli start to have an impact on the microbiome.

ENVIRONMENTAL FACTORS

The home environment

Studies have shown that modern humans spend just under 90% of their time indoors[122]. It, therefore, logically follows that the home environment would have a significant environmental influence on the microbiome of children. The microbiome of modern homes, compared to pre-industrial home environments that were intertwined with soil, plant and animal microbial life, is low in both diversity and abundance. This is exacerbated by the scarcity of nutrients, modern surface materials, and chemicals used to keep homes clean. These selective pressures result in a relative increase in environmental exposure to Basidiomycota, Deinococcus, Enhydrobacter, Micrococcus, Paracoccus, Staphylococcus, and Streptococcus[123,124]. It has been suggested that indoor plants go some way in increasing microbial diversity and provide a protected environment for microbial life, allowing stabilisation of the home microbiome, although there have been no epidemiological studies assessing if this makes any difference to human health[125].

Therefore, it is perhaps surprising that despite the significant time spent in the home environment, several studies have found that it is only responsible for a small proportion of the variation seen in the infant’s microbiome[126,127]. One study looked at adopted children and compared their microbiome to their genetically unrelated siblings and found that the shared home environment explained 6% of the variance[128]. The number of occupants in the shared house (i.e., how many siblings or parents cohabit in the house) has also failed to provide consistent results for any effect on the microbiome, although the presence of older siblings appears to alter the rate of maturation of the infant microbiome[127,129]. To add to the complexity that researchers face in trying to elucidate environment variables that affect microbiomic dynamics, twin studies have demonstrated increased concordance among monozygotic compared to dizygotic twins, showing that there is a genetic influence on the microbiome[130]. For example, the LCT, Fut2, irak4, and Nod2 genes have been directly linked to an alteration in the abundance of Bifidobacterium, Lachnospiraceae, Roseburia, Bacteroidetes, and Firmicutes, respectively[131-134]. This, in part, explains why genetic mutations (e.g., in Nod2) causing perturbations in the microbiome might be pathogenic for IBD[132,135].

Although no consistent effect has been found on the microbiome of adults[136], household pets modulate the microbiome of children[137]. Specifically, Bifidobacterium is increased in children with household dogs[137]. Having a dog in the house during infancy reduces the risk of childhood asthma, an atopic disease that appears to have an inverse relationship with Bifidobacterium[137]. A recent meta-analysis using cohort studies of children with household dogs adds weight to this, with a risk reduction for asthma of 0.85 (95%CI = 0.73-0.97)[138]. Interestingly, there appears to be a “dose response” with this association, with one study showing that two or more dogs had a greater effect than a single dog[139]. While we believe that the prevailing evidence supports household dogs being protective, albeit with potentially a small clinical effect, there is still conflicting evidence in the literature. As an example, one meta-analysis using a mix of cohort and case-control data came to an antithetical conclusion[140].

Day care

Children attending day care facilities are exposed to a group of peers with different microbiomes during their dynamic period of colonisation. One study found that compared to homeschooled children, day care resulted in a significantly more diverse and abundant microbiome, with an increased abundance in Bacteroidetes, Firmicutes, Lachnospiraceae, and Ruminococcaceae[141]. Perhaps unsurprisingly, the same group found that the microbiomes of peers at individual day care centres become more similar to each other compared to children in separate facilities. Previous studies have noted that the mixing of children in day care facilities is inversely related to childhood diabetes, suggesting that the microbiome might influence the pathogenesis[142]. Taken together, it appears that the mixing of children from outside of the household has a larger effect on the microbiome than mixing within the household; this is presumed to be due to the relative homogeneity between parents and offspring compared to the more diverse microbiomes of multiple classmates.

Geographical proximity to the natural environment

There is significant heterogeneity between global populations[143]; for example, populations living in agrarian societies generally have a higher abundance of Bacillus, Clostriales, Prevotella, Proteobacteria, Ruminobacter, Spirochaetes, and Succinivibrionaceae, while that of modern urban populations have higher Bacteroides, Bifidobacterium, Blautia, and Firmicutes[144-146]. However, elucidating if this is due to the geography of nations (e.g., urban USA vs. rural Malawi) or their respective diet, cultural habits, and climate, among others, has proved hard to determine. One review has noted that even within individual nations, urban dwellers are more likely to be born via CS, less likely to have been EBF, and more likely to have been exposed to microbiome-altering medications (e.g., antibiotics) during childhood, making the distinction between their rural counterparts more complicated[147].

It is, however, clear that when looking at the granular level, i.e., intranational, the geography of where children live has significant impacts on the microbiome and health outcomes of children. Numerous epidemiological studies have shown that children who grow up in rural locations have a significantly lower probability of IBD, asthma, and diabetes in later life[148-150]. The mechanism of this association has only recently started to become causally linked to the microbiome. As an example, one study looked at children growing up near (< 500 m) rural environments and compared their microbiome to those in urban areas, finding that the proximity to nature increases the abundance of Proteobacteria and Verrucomicrobiales, genera known to be beneficial symbionts and from the microbial rich soil[151]. These findings have been replicated with children growing up on farms[14]. A Finnish study found that children who grow up with this rural microbiome have reduced inflammatory response to bacterial cell wall antigens, with reduced expression of the pro-inflammatory cytokines IL-1β, IL-6, and IL-12[152]. Moreover, an elegantly designed study has demonstrated a linear “dose response” to a child’s proximity to greenspaces, showing that infants growing up in urban areas near a greenspace (e.g., a park) have a degree of protection to IBD, and as the distance to urban greenspaces increases, this protective effect decreases[153].

The soil found in rural areas, particularly forest floors, appears to be a crucial reservoir for beneficial microbial species that modulate the immune system, particularly in terms of atopic disease and IBD[154,155]. Indeed, soil is the most microbially rich substrate to be described[156] and the soil-dwelling species (e.g., Bacillus) have been identified as candidates for Rook’s old friends hypothesis[157]. This theory posits that we have co-evolved with environmental microbiomes typically encountered as forest hunter-gatherer omnivores, and these microbial species drive immunoregulatory and metabolic pathways[158]. Without these microbial populations, our GIT has an increased risk of dysregulation and results in the chronic inflammatory conditions often seen in the urbanised West. This does not explain the whole pathogenesis, but the mounting evidence does suggest it at least explains part of it[159]. Furthermore, animal studies have shown that exposure to soil during early life impacts the metabolome of the microbiome, increasing beneficial SCFAs, an effect that persists into adulthood[160]. Our group has recently shown that exposure to a strain of Bacillus velezensis, a prototypical soil dweller that produces bioactive cyclic-lipopeptides, can modulate the immune system in a beneficial way that ameliorates acute ulcerative colitis[161]. One group from Finland has gone one step further and designed an interventional trial for children aged 3-5 growing up in urban day care centres. These children were exposed to a specific rural microbiome by covering their outdoor areas with soil transplanted from a forest. They found this modulated the children’s microbiome with a particular increase in Proteobacteria, and noted an increase in plasma IL-10, a potent anti-inflammatory cytokine[162]. How these soil bacteria colonise the human GIT is poorly understood. It has been hypothesised that we are exposed to them by eating unwashed food, and by direct contact with the soil (e.g., playing in a field)[163,164]. The recently characterised aerobiome may also be responsible, at least in part, for colonisation dynamics in infants. As air blows over the ground and vegetation, microbial species are picked up and transported, and studies have shown significant differences in urban and rural aerobiomes, with rural aerobiomes having a greater abundance particularly in Bacillus and Proteobacteria than urban ones[165,166]. One group has experimentally demonstrated how exposure to a soil-based aerobiome might modulate the microbiome[167]. This might explain the linear proximity effect described earlier for children living in cities but near greenspaces, although more research on this new science is needed.

Antibiotic exposure

We have already discussed how antibiotic use during the antenatal period can influence the neonatal microbiome. Perhaps unsurprisingly, antibiotic exposure in early life, either administered directly, or indirectly via breast milk if the mother is taking antibiotics, also reduces the alpha diversity of the microbiome[168]. Surprisingly, however, this effect can still be found long after the child stops taking antibiotics. Unlike in adults, where studies have shown that the microbiome recovers approximately two weeks after stopping antibiotics[169], children have a much less stable microbiome during their developmental and transitional phases and can take months to years to recover[170,171]. Specific changes to the microbiome from antibiotic use in children are characterised by reduced Actinobacteria, Bacteroidetes, Firmicutes, and Verrucomicrobia populations, which are significant producers of beneficial SCFAs[172]. These perturbations in the microbiome in early life have been associated with childhood obesity, IBD, asthma, and allergies[173-176]. Furthermore, there are extensive mechanistic data from animal models supporting the causal link between the dysbiosis in the microbiome and subsequent pathology[177,178]. In contrast, certain bacterial cadres have been shown to increase in abundance after antibiotic use, such as E. coli, E. cloacae, K. pneumoniae, C. difficile, Erysipelotrichaceae spp., and Enterococcus spp., which are considered pathogenic and are associated with infective diarrhoeal disease, opportunistic infections, and IBD [177,179,180]. The use of antibiotics also alters the resistome, in that it increases the abundance of antimicrobial-resistant species, raising the risk of future clinically significant antibiotic-resistant infections[180]. Certain multi-strain probiotics may be suitable for rapid replenishment of the microbiome after antibiotic use, with one randomised control trial showing a significant reduction in antibiotic-related diarrhoea in children under the age of 18[181]. Furthermore, evidence summarised in a recent Cochrane review suggests there may be a utility for probiotic use in necrotising enterocolitis when used in combination with standard care[182]. While no powerful study has looked at the long-term outcomes of children taking probiotics or live bacterial therapeutics (LBT), there is evidence that Lactobacillus-, Bifidobacterium- and Bacillus-based LBTs can beneficially modulate the microbiome and reduce atopy, respiratory, ear and gastrointestinal infections[183-187]. While antibiotic use has a large and perhaps obvious impact on the microbiome, it should also be noted that the use of gastric acid-suppressing medications (e.g., proton pump inhibitors and histamine receptor H2 agonists antagonists) that are used in children with symptomatic reflux also influences the colonisation dynamics of infants. They have been shown to increase the abundance of Enterobacteriaceae, Clostridium, and Haemophilus, while reducing Firmicutes and Lactobacillus[188,189].

Gaps in the literature and future research potential

This literature review has described the latest evidence on numerous variables that impact the orchestration of the infant microbiome during the highly dynamic period of the first half decade of life. It has documented the evidence for health implications of these variables, and sought clarity on research for mechanistic causation.

The body of evidence on this topic is large and growing rapidly. PubMed has over 10,500 results for “paediatric microbiome” from the last decade, and more than half of these were published in the last three years alone. A significant proportion of these academic papers are based on epidemiological data. Accordingly, for many of the results presented, causality can only be inferred. To address this, several research groups have used animal models to control for environmental stimuli. These animal models require meticulous methodology, and often employ germ-free or gnotobiotic species that are greatly divorced from natural conditions. As such, the applicability to general human microbiomic dynamics is hard to fully establish. Consequently, and despite significant research, there are two important deficits in our understanding of this vital period of microbial colonisation: (i) the causal relationship from a specific environmental stimulus to establishing a colony in the GIT; and (ii) how specific microbiomes influence long-term health.

Addressing the first point, definitively showing that exposure to environment A leads directly to microbiomic outcome B, might appear pedantic given the voluminous observational data. However, without knowing specifically how species rarely found in the mostly aerobic outdoor environment colonise the infant GIT, such as Akkermansia Mucinophilia, devising future interventions to aid the developing microbiome is going to be problematic and imprecise. This can be seen contemporarily in novel vaginal seeding techniques used after CS, which have so far failed to convincingly replicate the microbiome of children born via VD. The entero-mammary pathway of translocating bacteria from the maternal colon to breast milk appears to be a fruitful avenue for future research. Furthermore, while the widespread use of 16S genomic profiling has transformed our understanding and ability to measure the microbiome in a cost-effective way, it does not provide the entire picture. For example, 16S RNA is not sufficient for fungal or viral profiling, and both methods fail to give clarity on strain-specific population changes. As an example, it has been estimated that 75% of our microbiome, at the bacterial strain level, has yet to be characterised, colloquially known as dark matter[190,191]. Furthermore, metagenomics is not specific to the metabolome of the microbiome, which provides the mainstay of bioactivity. The fine details of this are beyond the scope of this paper, but a recent review covers the topic well[192]. New technologies are showing promise to give researchers the tools to track specific strains that might produce clear evidence of transfer from one environment to the GIT, and the variation in metabolic output from different strains (e.g., WISH-tagging); however, economic viability has limited widespread use, and they have not yet been employed in this specific field[192,193]. The use of genetically tagged bacteria has been used for several decades as a cost-effective way of tracking microbiome dynamics in adults across different disciplines[194,195]. To date, and to the best of our knowledge, there has only been one study using this technique on the infant microbiome[25]. More studies using these techniques are needed to clarify with precision modes of microbial transmission. Addressing the second point, observational studies that demonstrate differences in health outcomes all suffer from well-established biases and the inability to control for unknown confounders. As already highlighted, these studies can, therefore, only infer causality[196]. Due to the nature of the subject matter (the paediatric population), it is not surprising that randomised trials have not been performed. Ethical approval for a randomised trial into CS vs. VD would be near impossible to justify, let alone finding volunteers for recruitment. Animal models have been, therefore, crucial for deepening our understanding of how differences in the microbiome affect metabolic and immunogenic health. Despite the critiques of observational data listed above, pragmatism and the highly consistent and reproducible evidence of associated long-term health benefits for the infant should, and do, guide public health organisations as well as professional bodies to advocate for VD over CS, and for exclusive breastfeeding for at least six months. It should be noted that there are further benefits to breastfeeding that have not been discussed, such as the nutrition provided by breast milk in nations that have food insecurity or unsanitary water conditions[197]; the protective effects of breastfeeding on the mother’s risk of breast cancer, etc.[198], but these are beyond the scope of this paper.

The critical window of opportunity

It is clear that the infant microbiome is influenced by numerous variables in a highly orchestrated and dynamic ecosystem that follows a similar pattern across different global populations. CS is associated with an altered microbiome compared to VD, although the mechanism of this difference is not fully established. The largest single effector of the microbiome is breastfeeding, with very clear evidence that once this ceases, the microbiome of the infant quickly starts to change to that of an adult. During this time, environmental stimuli also become more apparent (e.g., diet, living in a rural environment, going to day care, etc.) and the microbiomic differences from variables seen earlier in life, such as mode of birth or breast feeding, start to converge. At around the age of 3-5 years, the microbiome resembles that of an adult, and populations stabilise.

This raises two interesting points. Firstly, if the associated health differences seen in observational studies are causally related to the microbiomic differences, the effect occurs during a very short period of time. This has been referred to as the “critical window”. Animal studies have shown that during this window, the immune system is exposed to microbial antigens via specific pattern recognition receptors (e.g., Toll-Like Receptors), and primed to tolerate these specific species that direct the immune system in later life, with evidence suggesting that the microbiota need to follow a specific sequence of colonisation to ensure an optimal cascade of immune and metabolic development[199-204]. Furthermore, several experiments using germ-free mice that were exposed to microbiota before weaning demonstrated less inflammation, compared to those exposed later in life[70,205]. Innate-like T cells appear to be crucial. These cells develop immunological effector characteristics before thymic emergence, and recent work has shown that these cells are particularly responsive to early-life antigen exposure, thereby promoting the maintenance of tissue homeostasis[206,207]. Early exposure to commensal microbes correlates to the abundance of mucosal-associated invariant T cells (MAIT) and NK cells, and the absence of these early life antigen exposures cannot be compensated for in later life, with work showing that these T cell populations are vital for maintaining homeostasis and preventing excessive inflammation[206]. As an example, one murine study showed that Bacteroidetes (a genus associated with breastfeeding) increased colonic CD4+ cytotoxic T lymphocytes and increased FoxP3 protein expression, polarising the immune system away from Th2 type allergic response, in favour of Th1-mediated anti-inflammatory IL-10 production[208]. Evidence in humans supports this theory of a critical window of opportunity. For instance, cohort studies have shown that children who live in rural environments have a lower prevalence of IBD and asthma in later life compared to those born in urban areas; however, this protective effect is diminished if the child moves into a rural location after the critical window (i.e., is not exposed from birth), and the protective effect is absent in adults[148,209,210].

Secondly, if this critical window of opportunity during the first 3 to 5 years of life is proved to be true - and based on the available evidence, it is our opinion that this is a reasonable position to have - the highly dynamic infant microbiome offers a potential opportunity for health intervention. Building on public health advice that already exists (e.g., breastfeeding), interventions could be used to modulate the microbiome. We have discussed a Finnish group that has already shown that using soil from a forest transplanted to urban day care facilities changes the microbiome of the attendees, but large long-term studies need to be performed to see if this can influence health outcomes. Other approaches, such as using similar environmental inoculants in the home or using probiotics, have also been suggested, although the impact of this has not been assessed. Animal models are being employed in increasing numbers in an attempt to elucidate what specific microbial populations are important in orchestrating beneficial health outcomes, which in the future may be able to increase the precision of any microbiomic intervention.

CONCLUSION

There has been a remarkable increase in knowledge over a short period of time, covering the subtleties of the infant microbiome and its variation depending on specific external stimuli. It is clear that the first five years of life are a highly complex and dynamic period for the infant microbiome. We have described how this is influenced by numerous variables in a highly orchestrated and dynamic ecosystem that follows a similar pattern across different global populations. An individual’s microbiome can be seen to diverge from birth, with CS being associated with an altered microbiome compared to VD, although the mechanism of this difference is not fully established. It is apparent that the largest single effector of the microbiome is breastfeeding, with very clear evidence that once this ceases, the microbiome of the infant quickly starts to change to that of an adult. Very few mechanistic studies have been performed to study these processes in detail. As such, observational data have been employed to infer how the environment influences the microbiome. Several studies have identified key external influences on the infant’s microbiome, but consistently breastfeeding has the largest effect size. Moreover, the available evidence suggests that as a single variable, it provides a significant and long-lasting health benefit to the infant. After breastfeeding stops, other environmental cues, such as diet, geography, and exposure to soil, start to influence the microbiome to a greater degree. This period of early life, the window of opportunity, appears key to priming the immune system and avoiding chronic inflammatory states in later life. These modifiable variables show growing promise for future clinical utility for a microbiome-based public health intervention. As our methodology improves, higher resolution analysis should make it possible to dissect specific strain-level configurations as well as the non-bacterial species of the microbiome, and the ecosystems they originate from, to enable us to fully understand how the microbiome influences the normal development of the immune and metabolic systems in humans.

DECLARATIONS

Authors’ contributions

Conceptualization, writing - original draft, visualization: Horwell E

Writing - review & editing: Bearn P, Cutting SM

Availability of data and materials

Not applicable.

Financial support and sponsorship

This work was supported by funding from the UK Medical Research Council (MRC) under grant number (MR/R026262/1 to Cutting SM). The doctoral fellowship of Dr Edward Horwell was paid for by Ashford and St Peter’s NHS Foundation Trust (UK).

Conflicts of interest

Cutting SM is the CEO of SporeGen Ltd and has no financial gain in this publication. There are no other conflicts of interest to declare.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024.

REFERENCES

1. Whipps JM, Lewis K, Cooke RC. Mycoparasitism and plant disease control. In: Burge MN, editors. Fungi in biological control systems. Manchester University Press; 1988.p.161-187. Available from: https://books.google.com/books?hl=zh-CN&lr=&id=qoK7AAAAIAAJ&oi=fnd&pg=PR7&dq=Fungi+in+biological+control+systems&ots=ZGZywqBQQc&sig=SF3yncFR8kM6WBVld-jY1Ymmxec#v=onepage&q=Fungi%20in%20biological%20control%20systems&f=false. [Last accessed on 13 Sep 2024].

2. Berg G, Rybakova D, Fischer D, et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome 2020;8:103.

3. Ursell LK, Metcalf JL, Parfrey LW, Knight R. Defining the human microbiome. Nutr Rev 2012;70 Suppl 1:S38-44.

4. Grice EA, Segre JA. The skin microbiome. Nat Rev Microbiol 2011;9:244-53.

5. Morowitz MJ, Carlisle EM, Alverdy JC. Contributions of intestinal bacteria to nutrition and metabolism in the critically ill. Surg Clin North Am 2011;91:771-85.

6. Chow J, Lee SM, Shen Y, Khosravi A, Mazmanian SK. Host–bacterial symbiosis in health and disease. Adv Immunol 2010;107:243-74.

7. Davenport ER, Sanders JG, Song SJ, Amato KR, Clark AG, Knight R. The human microbiome in evolution. BMC Biol 2017;15:127.

8. Dominguez-Bello MG, Godoy-Vitorino F, Knight R, Blaser MJ. Role of the microbiome in human development. Gut 2019;68:1108-14.

9. Bragazzi NL, Del Rio D, Mayer EA, Mena P. We are what, when, and how we eat: the evolutionary impact of dietary shifts on physical and cognitive development, health, and disease. Adv Nutr 2024;15:100280.

10. Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol 2016;14:e1002533.

11. Takiishi T, Fenero CIM, Câmara NOS. Intestinal barrier and gut microbiota: shaping our immune responses throughout life. Tissue Barriers 2017;5:e1373208.

12. Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res 2020;30:492-506.

13. Afzaal M, Saeed F, Shah YA, et al. Human gut microbiota in health and disease: unveiling the relationship. Front Microbiol 2022;13:999001.

14. Stewart CJ, Ajami NJ, O'Brien JL, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature 2018;562:583-8.

15. Bäckhed F, Roswall J, Peng Y, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe 2015;17:690-703.

16. Vatanen T, Franzosa EA, Schwager R, et al. The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature 2018;562:589-94.

17. Bergström A, Skov TH, Bahl MI, et al. Establishment of intestinal microbiota during early life: a longitudinal, explorative study of a large cohort of danish infants. Appl Environ Microbiol 2014;80:2889-900.

18. Sarkar A, Yoo JY, Valeria Ozorio Dutra S, Morgan KH, Groer M. The association between early-life gut microbiota and long-term health and diseases. J Clin Med 2021;10:459.

19. Wassenaar TM, Panigrahi P. Is a foetus developing in a sterile environment? Lett Appl Microbiol 2014;59:572-9.

20. Escherich T. The intestinal bacteria of the neonate and breast-fed infant. Rev Infect Dis 1989;11:352-6.

21. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta harbors a unique microbiome. Sci Transl Med 2014;6:237ra65.

22. Morais J, Marques C, Teixeira D, et al. Extremely preterm neonates have more lactobacillus in meconium than very preterm neonates - the in utero microbial colonization hypothesis. Gut Microbes 2020;12:1785804.

23. Seferovic MD, Pace RM, Carroll M, et al. Visualization of microbes by 16S in situ hybridization in term and preterm placentas without intraamniotic infection. Am J Obstet Gynecol 2019;221:146.e1-e23.

24. Stout MJ, Conlon B, Landeau M, et al. Identification of intracellular bacteria in the basal plate of the human placenta in term and preterm gestations. Am J Obstet Gynecol 2013;208:226.e1-7.

25. Jiménez E, Fernández L, Marín ML, et al. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr Microbiol 2005;51:270-4.

26. Gschwind R, Fournier T, Kennedy S, et al. Evidence for contamination as the origin for bacteria found in human placenta rather than a microbiota. PLoS One 2020;15:e0237232.

27. Perez-Muñoz ME, Arrieta MC, Ramer-Tait AE, Walter J. A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: implications for research on the pioneer infant microbiome. Microbiome 2017;5:48.

28. Kennedy KM, de Goffau MC, Perez-Muñoz ME, et al. Questioning the fetal microbiome illustrates pitfalls of low-biomass microbial studies. Nature 2023;613:639-49.

29. Kennedy KM, Gerlach MJ, Adam T, et al. Fetal meconium does not have a detectable microbiota before birth. Nat Microbiol 2021;6:865-73.

30. Gustafsson B. Germ-free rearing of rats. Acta Anat (Basel) 1946;2:376-91.

31. Stinson LF, Boyce MC, Payne MS, Keelan JA. The not-so-sterile womb: evidence that the human fetus is exposed to bacteria prior to birth. Front Microbiol 2019;10:1124.

32. Kaisanlahti A, Turunen J, Byts N, et al. Maternal microbiota communicates with the fetus through microbiota-derived extracellular vesicles. Microbiome 2023;11:249.

33. Li Y, Toothaker JM, Ben-Simon S, et al. In utero human intestine harbors unique metabolome, including bacterial metabolites. JCI Insight 2020;5:138751.

34. Aguilera-Castrejon A, Oldak B, Shani T, et al. Ex utero mouse embryogenesis from pre-gastrulation to late organogenesis. Nature 2021;593:119-24.

35. Singh A, Mittal M. Neonatal microbiome - a brief review. J Matern Fetal Neonatal Med 2020;33:3841-8.

36. Mueller NT, Hourigan SK, Hoffmann DE, et al. Bacterial baptism: scientific, medical, and regulatory issues raised by vaginal seeding of c-section-born babies. J Law Med Ethics 2019;47:568-78.

37. Prince AL, Chu DM, Seferovic MD, Antony KM, Ma J, Aagaard KM. The perinatal microbiome and pregnancy: moving beyond the vaginal microbiome. Cold Spring Harb Perspect Med 2015;5:a023051.

38. Starc M, Lučovnik M, Eržen Vrlič P, Jeverica S. Protective effect of lactobacillus crispatus against vaginal colonization with group b streptococci in the third trimester of pregnancy. Pathogens 2022;11:980.

39. Aagaard K, Stewart CJ, Chu D. Una destinatio, viae diversae: does exposure to the vaginal microbiota confer health benefits to the infant, and does lack of exposure confer disease risk? EMBO Rep 2016;17:1679-84.

40. Reyman M, van Houten MA, van Baarle D, et al. Impact of delivery mode-associated gut microbiota dynamics on health in the first year of life. Nat Commun 2019;10:4997.

41. Wernroth ML, Peura S, Hedman AM, et al. Development of gut microbiota during the first 2 years of life. Sci Rep 2022;12:9080.

42. Dominguez-Bello MG, Costello EK, Contreras M, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A 2010;107:11971-5.

43. Słabuszewska-Jóźwiak A, Szymański JK, Ciebiera M, Sarecka-Hujar B, Jakiel G. Pediatrics consequences of caesarean section-a systematic review and meta-analysis. Int J Environ Res Public Health 2020;17:8031.

44. Stokholm J, Thorsen J, Chawes BL, et al. Cesarean section changes neonatal gut colonization. J Allergy Clin Immunol 2016;138:881-889.e2.

45. Wampach L, Heintz-Buschart A, Fritz JV, et al. Birth mode is associated with earliest strain-conferred gut microbiome functions and immunostimulatory potential. Nat Commun 2018;9:5091.

46. Xie J, Tang C, Hong S, et al. Maternal vaginal fluids play a major role in the colonization of the neonatal intestinal microbiota. Front Cell Infect Microbiol 2023;13:1065884.

47. Clausen TD, Bergholt T, Eriksson F, Rasmussen S, Keiding N, Løkkegaard EC. Prelabor cesarean section and risk of childhood type 1 diabetes: a nationwide register-based cohort study. Epidemiology 2016;27:547-55.

48. Hyde MJ, Modi N. The long-term effects of birth by caesarean section: the case for a randomised controlled trial. Early Hum Dev 2012;88:943-9.

49. Cardwell CR, Stene LC, Joner G, Cinek O, Svensson J, Goldacre MJ, et al. Caesarean section is associated with an increased risk of childhood-onset type 1 diabetes mellitus: a meta-analysis of observational studies. Diabetologia 2008;51:726-35.

50. Rutayisire E, Wu X, Huang K, Tao S, Chen Y, Tao F. Cesarean section may increase the risk of both overweight and obesity in preschool children. BMC Pregnancy Childbirth 2016;16:338.

51. Tollånes MC, Moster D, Daltveit AK, Irgens LM. Cesarean section and risk of severe childhood asthma: a population-based cohort study. J Pediatr 2008;153:112-6.

52. Thavagnanam S, Fleming J, Bromley A, Shields MD, Cardwell CR. A meta-analysis of the association between caesarean section and childhood asthma. Clin Exp Allergy 2008;38:629-33.

53. Dahlen HG, Downe S, Wright ML, Kennedy HP, Taylor JY. Childbirth and consequent atopic disease: emerging evidence on epigenetic effects based on the hygiene and EPIIC hypotheses. BMC Pregnancy Childbirth 2016;16:4.

54. Tefera M, Assefa N, Mengistie B, Abrham A, Teji K, Worku T. Elective cesarean section on term pregnancies has a high risk for neonatal respiratory morbidity in developed countries: a systematic review and meta-analysis. Front Pediatr 2020;8:286.

55. Mårild K, Stephansson O, Montgomery S, Murray JA, Ludvigsson JF. Pregnancy outcome and risk of celiac disease in offspring: a nationwide case-control study. Gastroenterology 2012;142:39-45.e3.

56. Bager P, Simonsen J, Nielsen NM, Frisch M. Cesarean section and offspring’s risk of inflammatory bowel disease: a national cohort study. Inflamm Bowel Dis 2012;18:857-62.

57. Sevelsted A, Stokholm J, Bønnelykke K, Bisgaard H. Cesarean section and chronic immune disorders. Obstetrical & Gynecological Survey 2015;70:303-5.

58. Axelsson PB, Clausen TD, Petersen AH, et al. Investigating the effects of cesarean delivery and antibiotic use in early childhood on risk of later attention deficit hyperactivity disorder. J Child Psychol Psychiatry 2019;60:151-9.

59. Curran EA, Dalman C, Kearney PM, et al. Association between obstetric mode of delivery and autism spectrum disorder: a population-based sibling design study. JAMA Psychiatry 2015;72:935-42.

60. Boerma T, Ronsmans C, Melesse DY, et al. Global epidemiology of use of and disparities in caesarean sections. Lancet 2018;392:1341-8.

61. Betrán AP, Ye J, Moller AB, Zhang J, Gülmezoglu AM, Torloni MR. The increasing trend in caesarean section rates: global, regional and national estimates: 1990-2014. PLoS One 2016;11:e0148343.

62. Kiriakopoulos N, Grigoriadis S, Maziotis E, et al. Investigating stress response during vaginal delivery and elective cesarean section through assessment of levels of cortisol, interleukin 6 (IL-6), growth hormone (GH) and insulin-like growth factor 1 (IGF-1). J Clin Med 2019;8:1112.

63. Blaser MJ. The theory of disappearing microbiota and the epidemics of chronic diseases. Nat Rev Immunol 2017;17:461-3.

64. Cho S, Samuel TM, Li T, et al. Interactions between bifidobacterium and bacteroides and human milk oligosaccharides and their associations with infant cognition. Front Nutr 2023;10:1216327.

65. Thongaram T, Hoeflinger JL, Chow J, Miller MJ. Human milk oligosaccharide consumption by probiotic and human-associated bifidobacteria and lactobacilli. J Dairy Sci 2017;100:7825-33.

66. Shao Y, Forster SC, Tsaliki E, et al. Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth. Nature 2019;574:117-21.

67. Sanidad KZ, Zeng MY. Neonatal gut microbiome and immunity. Curr Opin Microbiol 2020;56:30-7.

68. Fujimura KE, Slusher NA, Cabana MD, Lynch SV. Role of the gut microbiota in defining human health. Expert Rev Anti Infect Ther 2010;8:435-54.

69. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005;122:107-18.

70. Olszak T, An D, Zeissig S, et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 2012;336:489-93.

71. Lu CY, Ni YH. Gut microbiota and the development of pediatric diseases. J Gastroenterol 2015;50:720-6.

72. Asnicar F, Manara S, Zolfo M, et al. Studying vertical microbiome transmission from mothers to infants by strain-level metagenomic profiling. mSystems 2017;2:e00164-16.

73. Leszczynski P, van Belkum A, Pituch H, Verbrugh H, Meisel-Mikolajczyk F. Vaginal carriage of enterotoxigenic bacteroides fragilis in pregnant women. J Clin Microbiol 1997;35:2899-903.

74. Wilson BC, Butler ÉM, Grigg CP, et al. Oral administration of maternal vaginal microbes at birth to restore gut microbiome development in infants born by caesarean section: a pilot randomised placebo-controlled trial. EBioMedicine 2021;69:103443.

75. Zhou L, Qiu W, Wang J, et al. Effects of vaginal microbiota transfer on the neurodevelopment and microbiome of cesarean-born infants: a blinded randomized controlled trial. Cell Host Microbe 2023;31:1232-47.e5.

76. Wharton KR, Birsner ML. Committee opinion no. 725: vaginal seeding. Obstet Gynecol 2017;130:e274-8.

77. Bossung V, Lupatsii M, Dashdorj L, et al. Timing of antimicrobial prophylaxis for cesarean section is critical for gut microbiome development in term born infants. Gut Microbes 2022;14:2038855.

78. Sommerstein R, Marschall J, Atkinson A, et al. Swissnoso. Antimicrobial prophylaxis administration after umbilical cord clamping in cesarean section and the risk of surgical site infection: a cohort study with 55,901 patients. Antimicrob Resist Infect Control 2020;9:201.

79. Dierikx T, Berkhout D, Eck A, et al. Influence of timing of maternal antibiotic administration during caesarean section on infant microbial colonisation: a randomised controlled trial. Gut 2022;71:1803-11.

80. Azad MB, Konya T, Maughan H, et al. CHILD Study Investigators. Gut microbiota of healthy canadian infants: profiles by mode of delivery and infant diet at 4 months. CMAJ 2013;185:385-94.

81. Chu DM, Ma J, Prince AL, Antony KM, Seferovic MD, Aagaard KM. Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat Med 2017;23:314-26.

82. Erbaydar N, Erbaydar T. Relationship between caesarean section and breastfeeding: evidence from the 2013 turkey demographic and health survey. BMC Pregnancy Childbirth 2020;20:55.

83. McGuire MK, McGuire MA. Got bacteria? Curr Opin Biotechnol 2017;44:63-8.

84. Urbaniak C, Angelini M, Gloor GB, Reid G. Human milk microbiota profiles in relation to birthing method, gestation and infant gender. Microbiome 2016;4:1.

85. Khodayar-Pardo P, Mira-Pascual L, Collado MC, Martínez-Costa C. Impact of lactation stage, gestational age and mode of delivery on breast milk microbiota. J Perinatol 2014;34:599-605.

86. Soto A, Martín V, Jiménez E, Mader I, Rodríguez JM, Fernández L. Lactobacilli and bifidobacteria in human breast milk: influence of antibiotherapy and other host and clinical factors. J Pediatr Gastroenterol Nutr 2014;59:78-88.

87. Lyons KE, Shea CO', Grimaud G, et al. The human milk microbiome aligns with lactation stage and not birth mode. Sci Rep 2022;12:5598.

88. Boix-Amorós A, Collado MC, Mira A. Relationship between milk microbiota, bacterial load, macronutrients, and human cells during lactation. Front Microbiol 2016;7:492.

89. Jost T, Lacroix C, Braegger CP, Rochat F, Chassard C. Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding. Environ Microbiol 2014;16:2891-904.

90. Perez PF, Doré J, Leclerc M, et al. Bacterial imprinting of the neonatal immune system: lessons from maternal cells? Pediatrics 2007;119:e724-32.

91. Rodríguez JM. The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation? Adv Nutr 2014;5:779-84.

92. Newburg DS. Innate immunity and human milk. J Nutr 2005;135:1308-12.

93. Ouwerkerk JP, van der Ark KCH, Davids M, et al. Adaptation of akkermansia muciniphila to the oxic-anoxic interface of the mucus layer. Appl Environ Microbiol 2016;82:6983-93.

94. Luna E, Parkar SG, Kirmiz N, et al. Utilization efficiency of human milk oligosaccharides by human-associated akkermansia is strain dependent. Appl Environ Microbiol 2022;88:e0148721.

95. Cortes-Macías E, Selma-Royo M, García-Mantrana I, et al. Maternal diet shapes the breast milk microbiota composition and diversity: impact of mode of delivery and antibiotic exposure. J Nutr 2021;151:330-40.

96. Ho NT, Li F, Lee-Sarwar KA, et al. Meta-analysis of effects of exclusive breastfeeding on infant gut microbiota across populations. Nat Commun 2018;9:4169.

97. Yan J, Liu L, Zhu Y, Huang G, Wang PP. The association between breastfeeding and childhood obesity: a meta-analysis. BMC Public Health 2014;14:1267.

98. Cardwell CR, Stene LC, Ludvigsson J, et al. Breast-feeding and childhood-onset type 1 diabetes: a pooled analysis of individual participant data from 43 observational studies. Diabetes Care 2012;35:2215-25.

99. Chua MC, Ben-Amor K, Lay C, et al. Effect of synbiotic on the gut microbiota of cesarean delivered infants: a randomized, double-blind, multicenter study. J Pediatr Gastroenterol Nutr 2017;65:102-6.

100. Fehr K, Moossavi S, Sbihi H, et al. Breastmilk feeding practices are associated with the co-occurrence of bacteria in mothers’ milk and the infant gut: the CHILD cohort study. Cell Host Microbe 2020;28:285-97.e4.

101. Stinson LF, Trevenen ML, Geddes DT. Effect of cold storage on the viable and total bacterial populations in human milk. Nutrients 2022;14:1875.

102. Parigi SM, Eldh M, Larssen P, Gabrielsson S, Villablanca EJ. Breast milk and solid food shaping intestinal immunity. Front Immunol 2015;6:415.

103. Atyeo C, Alter G. The multifaceted roles of breast milk antibodies. Cell 2021;184:1486-99.

104. Lodge CJ, Tan DJ, Lau MX, et al. Breastfeeding and asthma and allergies: a systematic review and meta-analysis. Acta Paediatr 2015;104:38-53.

105. Duijts L, Jaddoe VW, Hofman A, Moll HA. Prolonged and exclusive breastfeeding reduces the risk of infectious diseases in infancy. Pediatrics 2010;126:e18-25.

106. Xu L, Lochhead P, Ko Y, Claggett B, Leong RW, Ananthakrishnan AN. Systematic review with meta-analysis: breastfeeding and the risk of Crohn’s disease and ulcerative colitis. Aliment Pharmacol Ther 2017;46:780-9.

107. Ogbo FA, Agho K, Ogeleka P, Woolfenden S, Page A, Eastwood J. Global Child Health Research Interest Group. Infant feeding practices and diarrhoea in sub-saharan african countries with high diarrhoea mortality. PLoS One 2017;12:e0171792.

108. Su Q, Sun X, Zhu L, et al. Breastfeeding and the risk of childhood cancer: a systematic review and dose-response meta-analysis. BMC Med 2021;19:90.

109. Horta BL, de Lima NP. Breastfeeding and type 2 diabetes: systematic review and meta-analysis. Curr Diab Rep 2019;19:1.

110. Binns C, Lee M, Low WY. The long-term public health benefits of breastfeeding. Asia Pac J Public Health 2016;28:7-14.

111. Qiao J, Dai LJ, Zhang Q, Ouyang YQ. A meta-analysis of the association between breastfeeding and early childhood obesity. J Pediatr Nurs 2020;53:57-66.

112. Bowatte G, Tham R, Allen KJ, et al. Breastfeeding and childhood acute otitis media: a systematic review and meta-analysis. Acta Paediatr 2015;104:85-95.

113. Tham R, Bowatte G, Dharmage SC, et al. Breastfeeding and the risk of dental caries: a systematic review and meta-analysis. Acta Paediatr 2015;104:62-84.

114. McGowan C, Bland R. The benefits of breastfeeding on child intelligence, behavior, and executive function: a review of recent evidence. Breastfeed Med 2023;18:172-87.

115. Pérez-Escamilla R, Buccini GS, Segura-Pérez S, Piwoz E. Perspective: should exclusive breastfeeding still be recommended for 6 months? Adv Nutr 2019;10:931-43.

116. Kramer MS, Kakuma R. Optimal duration of exclusive breastfeeding. Cochrane Database Syst Rev 2012;2012:CD003517.

117. Kim JH, Lee SW, Lee JE, Ha EK, Han MY, Lee E. Breastmilk feeding during the first 4 to 6 months of age and childhood disease burden until 10 years of age. Nutrients 2021;13:2825.

118. der Beek CM, Dejong CHC, Troost FJ, Masclee AAM, Lenaerts K. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr Rev 2017;75:286-305.

119. Kumbhare SV, Patangia DV, Patil RH, Shouche YS, Patil NP. Factors influencing the gut microbiome in children: from infancy to childhood. J Biosci 2019;44:9860.

120. De Filippo C, Cavalieri D, Di Paola M, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A 2010;107:14691-6.

121. Stark PL, Lee A. The microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of life. J Med Microbiol 1982;15:189-203.

122. Klepeis NE, Nelson WC, Ott WR, et al. The National Human Activity Pattern Survey (NHAPS): a resource for assessing exposure to environmental pollutants. J Expo Anal Environ Epidemiol 2001;11:231-52.

123. Shin SK, Kim J, Ha SM, et al. Metagenomic insights into the bioaerosols in the indoor and outdoor environments of childcare facilities. PLoS One 2015;10:e0126960.

124. Kembel SW, Meadow JF, O'Connor TK, et al. Architectural design drives the biogeography of indoor bacterial communities. PLoS One 2014;9:e87093.

125. Rai S, Singh DK, Kumar A. Microbial, environmental and anthropogenic factors influencing the indoor microbiome of the built environment. J Basic Microbiol 2021;61:267-92.

126. Brito IL, Gurry T, Zhao S, et al. Transmission of human-associated microbiota along family and social networks. Nat Microbiol 2019;4:964-71.

127. Lane AA, McGuire MK, McGuire MA, et al. Household composition and the infant fecal microbiome: the INSPIRE study. Am J Phys Anthropol 2019;169:526-39.

128. Tavalire HF, Christie DM, Leve LD, Ting N, Cresko WA, Bohannan BJM. Shared environment and genetics shape the gut microbiome after infant adoption. mBio 2021;12:e00548-21.

129. Stokholm J, Blaser MJ, Thorsen J, et al. Maturation of the gut microbiome and risk of asthma in childhood. Nat Commun 2018;9:141.

130. Xie H, Guo R, Zhong H, et al. Shotgun metagenomics of 250 adult twins reveals genetic and environmental impacts on the gut microbiome. Cell Syst 2016;3:572-84.e3.

131. Org E, Parks BW, Joo JW, et al. Genetic and environmental control of host-gut microbiota interactions. Genome Res 2015;25:1558-69.

132. Rausch P, Rehman A, Künzel S, et al. Colonic mucosa-associated microbiota is influenced by an interaction of Crohn disease and FUT2 (secretor) genotype. Proc Natl Acad Sci U S A 2011;108:19030-5.

133. Rehman A, Sina C, Gavrilova O, et al. Nod2 is essential for temporal development of intestinal microbial communities. Gut 2011;60:1354-62.

134. Cahana I, Iraqi FA. Impact of host genetics on gut microbiome: take-home lessons from human and mouse studies. Animal Model Exp Med 2020;3:229-36.

135. Liu Z, Zhang Y, Jin T, Yi C, Ocansey DKW, Mao F. The role of NOD2 in intestinal immune response and microbiota modulation: a therapeutic target in inflammatory bowel disease. Int Immunopharmacol 2022;113:109466.

136. Kates AE, Jarrett O, Skarlupka JH, et al. Household pet ownership and the microbial diversity of the human gut microbiota. Front Cell Infect Microbiol 2020;10:73.

137. Nermes M, Endo A, Aarnio J, Salminen S, Isolauri E. Furry pets modulate gut microbiota composition in infants at risk for allergic disease. J Allergy Clin Immunol 2015;136:1688-90.e1.

138. Gao X, Yin M, Yang P, et al. Effect of exposure to cats and dogs on the risk of asthma and allergic rhinitis: a systematic review and meta-analysis. Am J Rhinol Allergy 2020;34:703-14.

139. Fall T, Ekberg S, Lundholm C, Fang F, Almqvist C. Dog characteristics and future risk of asthma in children growing up with dogs. Sci Rep 2018;8:16899.

140. Ji X, Yao Y, Zheng P, Hao C. The relationship of domestic pet ownership with the risk of childhood asthma: a systematic review and meta-analysis. Front Pediatr 2022;10:953330.

141. Amir A, Erez-Granat O, Braun T, et al. Gut microbiome development in early childhood is affected by day care attendance. NPJ Biofilms Microbiomes 2022;8:2.

142. Parslow RC, McKinney PA, Law GR, Bodansky HJ. Population mixing and childhood diabetes. Int J Epidemiol 2001;30:533-8.

143. Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature 2012;486:222-7.

144. Widyarman AS, Theodorea CF, Udawatte NS, et al. Diversity of oral microbiome of women from urban and rural areas of indonesia: a pilot study. Front Oral Health 2021;2:738306.

145. Brewster R, Tamburini FB, Asiimwe E, Oduaran O, Hazelhurst S, Bhatt AS. Surveying gut microbiome research in africans: toward improved diversity and representation. Trends Microbiol 2019;27:824-35.

146. Gupta VK, Paul S, Dutta C. Geography, ethnicity or subsistence-specific variations in human microbiome composition and diversity. Front Microbiol 2017;8:1162.

147. Brushett S, Sinha T, Reijneveld SA, de Kroon MLA, Zhernakova A. The effects of urbanization on the infant gut microbiota and health outcomes. Front Pediatr 2020;8:408.

148. Benchimol EI, Kaplan GG, Otley AR, et al. Rural and urban residence during early life is associated with risk of inflammatory bowel disease: a population-based inception and birth cohort study. Am J Gastroenterol 2017;112:1412-22.

149. Wong GW, Chow CM. Childhood asthma epidemiology: insights from comparative studies of rural and urban populations. Pediatr Pulmonol 2008;43:107-16.

150. Dagenais GR, Gerstein HC, Zhang X, et al. Variations in diabetes prevalence in low-, middle-, and high-income countries: results from the prospective urban and rural epidemiological study. Diabetes Care 2016;39:780-7.

151. Nielsen CC, Gascon M, Osornio-Vargas AR, et al. Natural environments in the urban context and gut microbiota in infants. Environ Int 2020;142:105881.

152. Kirjavainen PV, Karvonen AM, Adams RI, et al. Farm-like indoor microbiota in non-farm homes protects children from asthma development. Nat Med 2019;25:1089-95.

153. Elten M, Benchimol EI, Fell DB, et al. Residential greenspace in childhood reduces risk of pediatric inflammatory bowel disease: a population-based Cohort study. Am J Gastroenterol 2021;116:347-53.

154. Hanski I, von Hertzen L, Fyhrquist N, et al. Environmental biodiversity, human microbiota, and allergy are interrelated. Proc Natl Acad Sci U S A 2012;109:8334-9.

155. Hertzen L, Hanski I, Haahtela T. Natural immunity. Biodiversity loss and inflammatory diseases are two global megatrends that might be related. EMBO Rep 2011;12:1089-93.

156. Anthony MA, Bender SF, van der Heijden MGA. Enumerating soil biodiversity. Proc Natl Acad Sci U S A 2023;120:e2304663120.

157. Rook GA. 99th Dahlem conference on infection, inflammation and chronic inflammatory disorders: darwinian medicine and the ‘hygiene’ or ‘old friends’ hypothesis. Clin Exp Immunol 2010;160:70-9.

158. Rook GA. Regulation of the immune system by biodiversity from the natural environment: an ecosystem service essential to health. Proc Natl Acad Sci U S A 2013;110:18360-7.

159. Rook GAW. The old friends hypothesis: evolution, immunoregulation and essential microbial inputs. Front Allergy 2023;4:1220481.

160. Tasnim N, Quin C, Gill S, Dai C, Hart M, Gibson DL. Early life environmental exposures have a minor impact on the gut ecosystem following a natural birth. Gut Microbes 2021;13:1-15.

161. Horwell E, Vittoria M, Hong HA, Bearn P, Cutting SM. A family of cyclic lipopeptides found in human isolates of bacillus ameliorates acute colitis via direct agonism of toll-like receptor 2 in a murine model of inflammatory bowel disease. Dig Dis Sci 2024;Online ahead of print.

162. Roslund MI, Puhakka R, Grönroos M, et al. ADELE research group. Biodiversity intervention enhances immune regulation and health-associated commensal microbiota among daycare children. Sci Adv 2020;6:eaba2578.

163. Blum WEH, Zechmeister-Boltenstern S, Keiblinger KM. Does soil contribute to the human gut microbiome? Microorganisms 2019;7:287.

164. Hirt H. Healthy soils for healthy plants for healthy humans: how beneficial microbes in the soil, food and gut are interconnected and how agriculture can contribute to human health. EMBO Rep 2020;21:e51069.

165. Mhuireach GÁ, Betancourt-román CM, Green JL, Johnson BR. Spatiotemporal controls on the urban aerobiome. Front Ecol Evol 2019;7:43.

166. Després VR, Huffman JA, Burrows SM, et al. Primary biological aerosol particles in the atmosphere: a review. Tellus B: Chemical and Physical Meteorology 2022;64:15598.

167. Liddicoat C, Sydnor H, Cando-Dumancela C, et al. Naturally-diverse airborne environmental microbial exposures modulate the gut microbiome and may provide anxiolytic benefits in mice. Sci Total Environ 2020;701:134684.

168. Reyman M, van Houten MA, Watson RL, et al. Effects of early-life antibiotics on the developing infant gut microbiome and resistome: a randomized trial. Nat Commun 2022;13:893.

169. Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A 2011;108 Suppl 1:4554-61.

170. Korpela K, Salonen A, Virta LJ, et al. Intestinal microbiome is related to lifetime antibiotic use in finnish pre-school children. Nat Commun 2016;7:10410.

171. Gasparrini AJ, Wang B, Sun X, et al. Persistent metagenomic signatures of early-life hospitalization and antibiotic treatment in the infant gut microbiota and resistome. Nat Microbiol 2019;4:2285-97.

172. Parker EPK, Praharaj I, John J, et al. Changes in the intestinal microbiota following the administration of azithromycin in a randomised placebo-controlled trial among infants in south India. Sci Rep 2017;7:9168.

173. Celind F, Wennergren G, Vasileiadou S, Alm B, Goksör E. Antibiotics in the first week of life were associated with atopic asthma at 12 years of age. Acta Paediatr 2018;107:1798-804.

174. Hviid A, Svanström H, Frisch M. Antibiotic use and inflammatory bowel diseases in childhood. Gut 2011;60:49-54.

175. Kelly D, Kelly A, O'Dowd T, Hayes CB. Antibiotic use in early childhood and risk of obesity: longitudinal analysis of a national cohort. World J Pediatr 2019;15:390-7.

176. Cox LM, Yamanishi S, Sohn J, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 2014;158:705-21.

177. Anthony WE, Wang B, Sukhum KV, et al. Acute and persistent effects of commonly used antibiotics on the gut microbiome and resistome in healthy adults. Cell Rep 2022;39:110649.

178. Ozkul C, Ruiz VE, Battaglia T, et al. A single early-in-life antibiotic course increases susceptibility to DSS-induced colitis. Genome Med 2020;12:65.

179. Buelow E, Bello González TDJ, Fuentes S, et al. Comparative gut microbiota and resistome profiling of intensive care patients receiving selective digestive tract decontamination and healthy subjects. Microbiome 2017;5:88.

180. Fjalstad JW, Esaiassen E, Juvet LK, van den Anker JN, Klingenberg C. Antibiotic therapy in neonates and impact on gut microbiota and antibiotic resistance development: a systematic review. J Antimicrob Chemother 2018;73:569-80.

181. Lukasik J, Dierikx T, Besseling-van der Vaart I, de Meij T, Szajewska H. Multispecies Probiotic in AAD Study Group. Multispecies probiotic for the prevention of antibiotic-associated diarrhea in children: a randomized clinical trial. JAMA Pediatr 2022;176:860-6.

182. Sharif S, Meader N, Oddie SJ, Rojas-Reyes MX, McGuire W. Probiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Database Syst Rev 2020;10:CD005496.

183. Kukkonen K, Savilahti E, Haahtela T, et al. Long-term safety and impact on infection rates of postnatal probiotic and prebiotic (synbiotic) treatment: randomized, double-blind, placebo-controlled trial. Pediatrics 2008;122:8-12.

184. Hatakka K, Savilahti E, Pönkä A, et al. Effect of long term consumption of probiotic milk on infections in children attending day care centres: double blind, randomised trial. BMJ 2001;322:1327.

185. Lundelin K, Poussa T, Salminen S, Isolauri E. Long-term safety and efficacy of perinatal probiotic intervention: evidence from a follow-up study of four randomized, double-blind, placebo-controlled trials. Pediatr Allergy Immunol 2017;28:170-5.

186. Rutten NB, Gorissen DM, Eck A, et al. Long term development of gut microbiota composition in atopic children: impact of probiotics. PLoS One 2015;10:e0137681.

187. Dhongade AR, Joshi SS, Kulkarni SV, et al. The efficacy and safety of a bacillus probiotic combination for the treatment of acute diarrhea in children: a double-blind, randomized, placebo-controlled multicentric study. Pediatric Infectious Disease 2022;4:86-91.

188. Turco R, Martinelli M, Miele E, et al. Proton pump inhibitors as a risk factor for paediatric Clostridium difficile infection. Aliment Pharmacol Ther 2010;31:754-9.

189. Huang H, Jiang J, Wang X, Jiang K, Cao H. Exposure to prescribed medication in early life and impacts on gut microbiota and disease development. EClinicalMedicine 2024;68:102428.

190. Joice R, Yasuda K, Shafquat A, Morgan XC, Huttenhower C. Determining microbial products and identifying molecular targets in the human microbiome. Cell Metab 2014;20:731-41.

191. Marcy Y, Ouverney C, Bik EM, et al. Dissecting biological “dark matter” with single-cell genetic analysis of rare and uncultivated TM7 microbes from the human mouth. Proc Natl Acad Sci U S A 2007;104:11889-94.

192. Yan Y, Nguyen LH, Franzosa EA, Huttenhower C. Strain-level epidemiology of microbial communities and the human microbiome. Genome Med 2020;12:71.

193. Daniel BBJ, Steiger Y, Sintsova A, et al. Assessing microbiome population dynamics using wild-type isogenic standardized hybrid (WISH)-tags. Nat Microbiol 2024;9:1103-16.

194. Jansson JK. Tracking genetically engineered microorganisms in nature. Curr Opin Biotechnol 1995;6:275-83.

195. Geva-Zatorsky N, Alvarez D, Hudak JE, et al. In vivo imaging and tracking of host-microbiota interactions via metabolic labeling of gut anaerobic bacteria. Nat Med 2015;21:1091-100.

196. Rivas-Ruiz F, Pérez-Vicente S, González-Ramírez AR. Bias in clinical epidemiological study designs. Allergol Immunopathol (Madr) 2013;41:54-9.

197. Turin CG, Ochoa TJ. The role of maternal breast milk in preventing infantile diarrhea in the developing world. Curr Trop Med Rep 2014;1:97-105.

198. Ciampo LA, Del Ciampo IRL. Breastfeeding and the benefits of lactation for women’s health. Rev Bras Ginecol Obstet 2018;40:354-9.

199. Hornef MW, Torow N. ‘Layered immunity’ and the ‘neonatal window of opportunity’-timed succession of non-redundant phases to establish mucosal host-microbial homeostasis after birth. Immunology 2020;159:15-25.

200. Gensollen T, Iyer SS, Kasper DL, Blumberg RS. How colonization by microbiota in early life shapes the immune system. Science 2016;352:539-44.

201. Torow N, Yu K, Hassani K, et al. Active suppression of intestinal CD4+TCRαβ+ T-lymphocyte maturation during the postnatal period. Nat Commun 2015;6:7725.

202. Sjögren YM, Tomicic S, Lundberg A, et al. Influence of early gut microbiota on the maturation of childhood mucosal and systemic immune responses. Clin Exp Allergy 2009;39:1842-51.

203. Lu L, Yu Y, Guo Y, Wang Y, Chang EB, Claud EC. Transcriptional modulation of intestinal innate defense/inflammation genes by preterm infant microbiota in a humanized gnotobiotic mouse model. PLoS One 2015;10:e0124504.

204. Reynolds HM, Bettini ML. Early-life microbiota-immune homeostasis. Front Immunol 2023;14:1266876.

205. Ronan V, Yeasin R, Claud EC. Childhood development and the microbiome-the intestinal microbiota in maintenance of health and development of disease during childhood development. Gastroenterology 2021;160:495-506.

206. Constantinides MG, Belkaid Y. Early-life imprinting of unconventional T cells and tissue homeostasis. Science 2021;374:eabf0095.

207. Constantinides MG, Link VM, Tamoutounour S, et al. MAIT cells are imprinted by the microbiota in early life and promote tissue repair. Science 2019;366:eaax6624.

208. Ottman N, Ruokolainen L, Suomalainen A, et al. Soil exposure modifies the gut microbiota and supports immune tolerance in a mouse model. J Allergy Clin Immunol 2019;143:1198-206.e12.

209. Timm S, Svanes C, Janson C, et al. Place of upbringing in early childhood as related to inflammatory bowel diseases in adulthood: a population-based cohort study in Northern Europe. Eur J Epidemiol 2014;29:429-37.

210. Feng M, Yang Z, Pan L, et al. Associations of early life exposures and environmental factors with asthma among children in rural and urban areas of Guangdong, China. Chest 2016;149:1030-41.

Cite This Article

Review
Open Access
A microbial symphony: a literature review of the factors that orchestrate the colonization dynamics of the human colonic microbiome during infancy and implications for future health
Edward HorwellEdward Horwell, ... Simon M. Cutting

How to Cite

Horwell, E.; Bearn P.; Cutting S. M. A microbial symphony: a literature review of the factors that orchestrate the colonization dynamics of the human colonic microbiome during infancy and implications for future health. Microbiome. Res. Rep. 2024, 3, 50. http://dx.doi.org/10.20517/mrr.2024.32

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click on download.

Export Citation File:

Type of Import

Tips on Downloading Citation

This feature enables you to download the bibliographic information (also called citation data, header data, or metadata) for the articles on our site.

Citation Manager File Format

Use the radio buttons to choose how to format the bibliographic data you're harvesting. Several citation manager formats are available, including EndNote and BibTex.

Type of Import

If you have citation management software installed on your computer your Web browser should be able to import metadata directly into your reference database.

Direct Import: When the Direct Import option is selected (the default state), a dialogue box will give you the option to Save or Open the downloaded citation data. Choosing Open will either launch your citation manager or give you a choice of applications with which to use the metadata. The Save option saves the file locally for later use.

Indirect Import: When the Indirect Import option is selected, the metadata is displayed and may be copied and pasted as needed.

About This Article

Special Issue

© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
209
Downloads
58
Citations
1
Comments
0
0

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Microbiome Research Reports
ISSN 2771-5965 (Online)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/