6,533
Views
15
CrossRef citations to date
0
Altmetric
Review

Microbial metabolites as modulators of the infant gut microbiome and host-microbial interactions in early life

ORCID Icon, ORCID Icon & ORCID Icon
Article: 2192151 | Received 03 Oct 2022, Accepted 06 Mar 2023, Published online: 21 Mar 2023

ABSTRACT

The development of infant gut microbiome is a pivotal process affecting the ecology and function of the microbiome, as well as host health. While the establishment of the infant microbiome has been of interest for decades, the focus on gut microbial metabolism and the resulting small molecules (metabolites) has been rather limited. However, technological and computational advances are now enabling researchers to profile the plethora of metabolites in the infant gut, allowing for improved understanding of how gut microbial-derived metabolites drive microbiome community structuring and host-microbial interactions. Here, we review the current knowledge on development of the infant gut microbiota and metabolism within the first year of life, and discuss how these microbial metabolites are key for enhancing our basic understanding of interactions during the early life developmental window.

Introduction

A relatively simple microbial community at birth develops into a diverse and complex microbial ecosystem across the early life windowCitation1, which plays a vital role in postnatal maturation and development of the endocrine, nervous and immune systemsCitation2,Citation3. The gut microbiome thus has a profound influence on both early and long-term health and developmentCitation4,Citation5. In addition to influencing development and maturation of the host immune system, the gut microbiota has a number of important functions in the host, including breakdown of ingested complex carbohydrates, protection against pathogenic bacteria and metabolic functions including production of vitamins, short chain fatty acids (SCFAs) and bile acid biotransformationCitation6. The importance of neonatal bacterial colonization for the growth of a healthy individual is clear and exhibits the power that the microbiome has on its host from an early stage in development.

The gut microbiota composition is initially known to be in a state of flux and consequently stabilizes, so that by age 2.5–4 years, it begins to resemble the composition and characteristics of an adult gutCitation7,Citation8. In this early phase, the gut microbiota is more vulnerable to modulation than in the adult stateCitation9, suggesting that the first 1000 days of life is a critical window of opportunity for optimal bacterial colonization. Disruption to microbial development during this period is believed to have long-lasting consequences, impacting on normal immune and neurocognitive development and has been associated with increased risk of developing diseases such as asthma and metabolic syndrome later in lifeCitation10,Citation11.

The gut microbiota may also be modulated at this critical phase in a beneficial manner. Strategies to optimize early microbial acquisition during this key developmental window of opportunity may help to prevent and treat disease. Interventions to modulate the early microbiome include the use of probiotic and prebiotic interventions or combinations of both, as either synbioticsCitation12 or postbioticsCitation13. The most promising results for probiotics have been seen in the prevention or treatment of diarrhea in children and in necrotizing enterocolitis in preterm infantsCitation14,Citation15. Pharmabiotics including microbially produced bacteriocins, bacteriophages and SCFAs have gained interest for potential application for microbiota modulationCitation16–19. Furthermore, there has been a shift in recent years to expand microbiome research beyond profiling taxonomic composition using 16S rRNA gene sequencing and to focus on gut microbial functionality and activity using whole metagenome sequencing (WMS) and metabolomics. WMS while more costly than 16S rRNA sequencing provides insight on the functions encoded by the genes present in the microbiome. WMS also facilitates community structure to be examined at a far greater and reliable taxonomic resolution beyond genus level to examine species and subspeciesCitation20. In addition, metabolomics is a useful complementary approach to measure metabolites in the gut derived from the diet, host or the microbiota (collectively denoted the gut metabolome)Citation21. Advances in informatics such as machine learning have also provided superior insight and facilitated meaningful interpretations of functional microbiome researchCitation22,Citation23. This review briefly discusses the factors influencing the gut microbiota in early life, summarizes current literature available on the diet-dependent development of the infant gut metabolome, and discusses how microbiota-derived metabolites may be key for early life microbiome and host development.

Factors influencing the gut microbiota in early life

Non-nutritional factors

Delivery mode exerts a significant influence of gut microbiota composition, exposing the infant to various microbes associated with the maternal gut and vaginal tractCitation24. Passage of the infant through the birth canal exposes it to the mother’s vaginal and fecal bacteria thereby contributing to microbial and metabolic diversity in the infant gutCitation25. Cesarean section impairs the transfer of certain maternal strains resulting in an infant gut microbiota with increased abundance of potentially pathogenic microbes, and a lack of beneficial taxa such as BifidobacteriumCitation26. Other factors reported to influence the development of the infant gut microbiome include gestational age, host genetics, feeding regime, geographical location and perinatal antibiotic usageCitation27,Citation28. Gestational age is an important factor in gut microbiota development with preterm birth associated with reduced diversity and delayed colonization with Bifidobacterium compared with full-term infantsCitation29. Exposure to antibiotics in early life has been reported to induce an increased abundance of members of the Enterobacteriaceae family and decrease in abundance of Bifidobacterium Citation30,Citation31. Antibiotic exposure has also been linked with the development of metabolic and inflammatory diseases later in lifeCitation32. In recent years, evidence also supports the existence of a breast milk microbiome, which is a source of beneficial microbes to the infantCitation27,Citation33,Citation34. Infants also acquire microbes that colonize their gut from the environment including siblings, pets and the hospitalCitation5,Citation28,Citation35.

Breastmilk

Human breast milk can meet all nutritional requirements essential for infant growth, health and developmentCitation36. Breast milk contains essential nutrients such as caseins, whey, fatty acids, and lactose as well as micronutrients including Immunoglobulin A, Immunoglobulin G, Immunoglobulin M, calcium and vitamin ACitation28,Citation37. Furthermore, breast milk is a source of an array of bioactive components such as human milk oligosaccharides (HMOs). HMOs consist of > 150 known complex glycans with the most common being 2′-fucosyllactose (2′FL), accounting for 20–30% of all HMOsCitation37–39. HMOs are not digested in the upper gastrointestinal (GI) tract and are degraded in the colon by specific HMO-degrading bacteria, mostly Bifidobacterium, into metabolites such as acetate and lactate with potential beneficial host health effectsCitation28,Citation38. The ability of some species of Bifidobacterium to efficiently degrade HMOs makes them dominant members of the microbiota of breastfed infants. Host secretor status, based on active or inactive copy of the fucosyltransferase 2 (FUT2) gene, determines a mother’s ability to produce HMOsCitation40–43. Non-secretors have been associated with reduced Bifidobacterium and Bacteroides abundances and lower microbial diversity in their infant’s gut microbiotaCitation44–47. The absence/decrease of 2′FL in non-secretor women has potential implications for infant short and long term health and developmentCitation48–50.

Formula milk

Compositional analysis of the infant gut microbiota has shown altered community structures in formula-fed infants compared to breastfed infantsCitation51,Citation52. Higher abundance of potentially pathogenic bacteria such as Enterobacteriaceae and Clostridium, as well as antimicrobial resistance genes, is observed in formula-fed infants compared to exclusively breastfed infantsCitation8,Citation51,Citation53,Citation54. Conversely, breastmilk feeding is associated with low microbial diversity, dominated by HMO-utilizing Bifidobacterium species and saccharolytic fermentation compared to formula-feedingCitation27,Citation55. Due to formula milk’s higher protein content, formula fed infants may also exhibit a higher degree of proteolytic metabolism in the gutCitation56,Citation57. Proteolytic microbiota-derived metabolites have been linked to detrimental cardiovascular and metabolic health effects in adultsCitation58–61, as discussed in greater details below. Yet, whether changes in colonic fermentation observed in formula fed infants influence the risk of developing diseases later remains unknown. Nonetheless, many infant formulas have in recent years been supplemented with prebiotics in an attempt to mimic health benefits of HMOs in breast milk. Prebiotic supplementation has predominantly consisted of short-chain galacto-oligosaccharides (scGOS), long-chain fructo-oligosaccharides (lcFOS), and HMOs such as 2-fucosyllactose and Lacto-N-neotetraoseCitation62–64.

Weaning and beyond

During the transition period to solid foods, typically between 4–6 months of age, changes appear in the richness and diversity of the infant gut microbiota. Milk adapted bacterial species such as B. longum subsp. infantis typically decrease in this periodCitation65. Due to the increased consumption of protein and dietary fibers during this period, microbes capable of digesting more complex nutrients increase in abundance such as Lachnospiraceae, Bacteroidaceae and RuminococcaceaeCitation66–68. Microbiota maturation occurs continuously even beyond five years of age where the children have acquired a diverse, stable and adult-like configuration, but with a lower community richness compared to the adult microbiotaCitation69.

Malnutrition

Food is a major factor shaping the gut microbiota in early life. This is also evident in children who do not receive adequate nutrition during infancy and early childhood. Malnourished infants and children develop an immature gut microbiomeCitation70, which may contribute to impairment of child growthCitation71,Citation72. The disturbance in microbial succession in malnourished children could contribute to lifelong deficits in growth and development as reviewed elsewhereCitation73. Intriguingly, a recent 3-month-long intervention study showed that a microbiota-directed complementary food prototype in comparison with an existing ready-to-use supplementary food promoted weight gain in children with moderate acute malnutrition between the ages of 12 months and 18 months of ageCitation74. The study suggested that the improved weight gain could be mediated by a more complete repair of the gut microbiotaCitation74. Supporting this hypothesis, a placebo-controlled trial in 2- to 6-month-old undernourished Bangladeshi infants with severe acute malnutrition found that supplementation with a Bifidobacterium longum subspecies infantis strain promoted weight gain that was associated with reduced levels of intestinal inflammation markersCitation75. Globally in 2020, 149 million children under the age of five years were estimated by be stuntedCitation76, emphasizing the need for also understanding the role of the immature gut microbiome in malnourished children.

Colonic fermentation and microbial metabolites in early life

The transitioning from a sole source of milk to a diverse range of foodsCitation77 not only shapes gut microbiome compositionCitation1,Citation8,Citation78, but also shapes gut microbial metabolism in early life resulting in the generation of a plethora of small molecules (metabolites) ().

Figure 1. Gut microbiota-derived metabolites in early life. Gut microbial metabolism changes in early life with the progression in early nutrition from breastfeeding to solid foods concurrent with a progression in microbiome and metabolome diversity. During breastfeeding, the dominance of the infant gut by human milk oligosaccharide (HMO) degrading Bifidobacterium species results in high levels of lactate and acetate, as well as in aromatic lactic acids (i.e. phenyllactic acid, 4-hydroxyphenyllactic acid and indolelactic acid). With the progression in diet, the dietary complexity increases and more indigestible proteins and fiber end up in the colon of the child. Consequently, colonic fermentation changes resulting in the formation of short-chain fatty acids (SCFA, i.e. acetate, propionate and butyrate) and gases (i.e. hydrogen and methane). Furthermore, proteins are degraded into amino acids, which are fermented by the resident gut microbes into branched SCFAs (i.e. isobutyrate, isovalerate and 2-methylbutyrate), amines (i.e. histamine, dopamine, tyramine, γ-aminobutyric acid (GABA), tryptamine), as well as aromatic acetic and propionic acids (e.g. indoleacetic acid and indolepropionic acid).

Figure 1. Gut microbiota-derived metabolites in early life. Gut microbial metabolism changes in early life with the progression in early nutrition from breastfeeding to solid foods concurrent with a progression in microbiome and metabolome diversity. During breastfeeding, the dominance of the infant gut by human milk oligosaccharide (HMO) degrading Bifidobacterium species results in high levels of lactate and acetate, as well as in aromatic lactic acids (i.e. phenyllactic acid, 4-hydroxyphenyllactic acid and indolelactic acid). With the progression in diet, the dietary complexity increases and more indigestible proteins and fiber end up in the colon of the child. Consequently, colonic fermentation changes resulting in the formation of short-chain fatty acids (SCFA, i.e. acetate, propionate and butyrate) and gases (i.e. hydrogen and methane). Furthermore, proteins are degraded into amino acids, which are fermented by the resident gut microbes into branched SCFAs (i.e. isobutyrate, isovalerate and 2-methylbutyrate), amines (i.e. histamine, dopamine, tyramine, γ-aminobutyric acid (GABA), tryptamine), as well as aromatic acetic and propionic acids (e.g. indoleacetic acid and indolepropionic acid).

Colonic fermentation of carbohydrates

The most studied microbiota-derived metabolites are the SCFAs. SCFAs are products of colonic fermentation of HMOs, dietary fiber and proteins, and known to affect human metabolism in numerous ways via activation of several host receptors in different organsCitation79. SCFAs also serve as energy substrates both for the resident gut microbes (as discussed further below), as well as human cells with butyrate being a primary energy substrate for colonocytesCitation80, and propionate being a substrate for gluconeogenesis in the intestineCitation81 and liverCitation82,Citation83. In adults, the major SCFAs acetate, propionate and butyrate are typically found in a 3:1:1 ratio, whereas succinate and lactate appear as intermediates and therefore do not appear in consistent levels. However, in early life, SCFA profiles change concordant with the child’s progression in dietary and microbiome diversityCitation84,Citation85. During breastfeeding (early phase), the SCFA profile is characterized by low acetate and high succinate, during complementary feeding (middle phase) by high lactate, pyruvate and formate, and after cessation of breastfeeding (late phase) by high propionate and butyrateCitation84. The high abundances of lactate, pyruvate and formate measured in the infant gut during the early phase reflect a less-developed microbiome since these metabolites, in the context of a developed gut microbiome, typically would have been converted into other metabolitesCitation79,Citation86. Compared to non-breast infants, exclusive breastfed children have been reported to have lower absolute concentrations of total SCFAs including acetate, butyrate, propionate, valerate, isobutyrate, and isovalerate, yet higher concentrations of lactate at four months of ageCitation87. Bifidobacterium are highly abundant in breastfed infantsCitation88 due to their ability to metabolize HMOsCitation89, which results in the production of acetate and lactateCitation90. Yet, with the cessation of breastfeeding and development of the gut microbiomeCitation1, butyrate production increases in the infant gutCitation84. Despite the large dynamics in SCFAs observed in early life and their potential importance as signaling molecules, the relationships between SCFAs, the gut microbiome and given carbohydrate substrates are rather limited. Furthermore, with over 200 HMO varieties identified in human milk and with large variations in HMO profiles between mothers and throughout lactationCitation91,Citation92, it also remains to be elucidated how differences in breastmilk HMO profiles affect the infant’s gut microbial metabolism.

Colonic fermentation of proteins

Studies on gut microbial metabolites in the context of early life have mainly been limited to the study of SCFAsCitation90,Citation93–95. However, given that intake of dietary fiber and proteins increases with the complementary dietCitation77,Citation96, which is associated with increased gut microbial diversityCitation68 and capacity to transport amino acids during infancyCitation8,Citation78, a multitude of microbial-derived metabolites of dietary fiber and proteinsCitation58,Citation59,Citation97–104 are likely to change with this dietary transition. For example, it was recently reported that HMO-degrading Bifidobacterium species convert aromatic amino acids (i.e. tryptophan, phenylalanine and tyrosine) into aromatic lactic acids (i.e. indolelactic acid, phenyllactic acid and 4-hydroxyphenyllactic acid) in the infant gutCitation88. In agreement with breastfed infants being dominated by Bifidobacterium Citation68, aromatic lactic acids have been found in higher levels in feces of breastfed infants compared to formula-fed infantsCitation85,Citation105 and weaned infantsCitation88, emphasizing the strong interplay between early life nutrition and specific gut microbes affecting the levels of microbial aromatic amino acid catabolites in early life.

Other examples of microbiota-derived metabolites changing in early life are the branched-chain fatty acids (BCFA, i.e. isobutyrate, isovalerate, 2-methylbutyrate). The BCFA reflect increased proteolytic fermentation as they originate from conversion of the branched-chain amino acids valine, leucine and isoleucineCitation21, which are implicated in insulin resistanceCitation106. The levels of proteins and amino acids could likely play a role in dictating the levels of BCFA, since a study found that breastfed infants had significantly lower levels of fecal BCFAs compared to infants fed two infant formulas with extensively hydrolyzed proteins and free amino acids, respectively, at one and two months of ageCitation107. Infant formula typically contains more protein and lower free amino acids than breast milkCitation108,Citation109, suggesting that more proteins are being fermented in the colon of formula-fed infants. In line herewith, a study in newborn rhesus monkeys found that by reducing the protein content in formula, urinary levels of microbial-derived proteolytic compounds (i.e. 3-indoxylsulfate, 4-hydroxyphenylacetate, and 2-hydroxyisobutyrate) could be reducedCitation110. Fecal BCFA levels have also been associated with variations in breastmilk lipid and HMO compositionsCitation111. Furthermore, BCFA have been reported to increase in the infant gut over time independent of early feeding (breastfeeding versus formula feeding)Citation57,Citation84,Citation93, suggesting that the lack of HMO following a progression toward solid food promotes a shift toward amino acid fermentation in the infant gut. Also biogenic amines (i.e. cadaverine and tyramine) have been reported in higher levels in feces of formula-fed infants compared to breastfed infantsCitation112,Citation113. These differences could be due to differences in the gut microbiome composition, as infant formula-associated genera include Streptococcus and EnterococcusCitation114, which are producers of putrescine, tyramine and cadaverineCitation115,Citation116. Another prevalent commensal in early life as children progress toward a solid diet is Ruminococcus gnavusCitation117, which is also able to produce tryptamine in vitroCitation118. However, it remains to be investigated whether R. gnavus indeed produces tryptamine in the infant gut. In fact, many microbial proteolytic-derived metabolites associated with detrimental effects on cardiovascular and metabolic health in adults, such as trimethylamine-N-oxide (TMAO, derivative of carnitine, betaine and choline)Citation58, imidazole propionate (derivative of histidine)Citation59, phenylacetylglutamine (derivative of phenylalanine)Citation60, p-cresol sulfate and indoxyl sulfate (derivatives of tyrosine and tryptophan, respectively)Citation61, remain understudied in the context of early life. These metabolites would be expected to arise when children transition toward adult-like solid foods. Yet, very few studies have looked into the impact of the complementary diet on the infant gut microbiomeCitation68.

Colonic fermentation of lipids and bile acids

Dietary fat is also metabolized by microbes in the gastrointestinal tract. Infants are, through breast milk and to some extent formula milk, provided with the essential fatty acids linoleic acid (LA) and alpha-linoleic acid (ALA), as well as the long chain polyunsaturated fatty acids (LC-PUFA): eicosapentaenoic acid (EPA), arachidonic acid (AA) and docosahexaenoic acid (DHA, omega-3 fatty acid)Citation77. Besides having direct effects on the gut microbiome composition and activityCitation119, the polyunsaturated fatty acids can be metabolized by colonic microbes into hydroxy fatty acids, oxo fatty acids, conjugated fatty acids, and partially saturated trans-fatty acids as intermediatesCitation120–122. Interestingly, specific Bifidobacterium strains have been found to be able to convert LA to conjugated linoleic acid (CLA)Citation122,Citation123, which has been associated with a variety of systemic health promoting effectsCitation124. Although the production and importance of CLA in the infant gut are currently unknown, another LA-derivate measured in feces, 12,13-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) has been suggested to impede immune tolerance in early lifeCitation125,Citation126. This metabolite was recently reported to be produced by common early-life gut microbiome members, namely B. bifidum and E. faecalis strainsCitation125. Furthermore, a recent randomized, controlled, clinical trial reported that infants receiving formula milk containing HMOs (i.e. 2-fucosyllactose and LNnT) had reduced fecal levels of two dihydroxy fatty acids (12,13-DiHOME and 9,10-dihydroxy-12Z-octadecenoic acid (9,10-diHOME)) compared to infants receiving control formula milk without HMOs, suggesting that HMO-induced alterations in the microbiome may affect microbial conversion of fatty acidsCitation127. Therefore, more studies are warranted to investigate intestinal fatty acid metabolism in the infant gut, which is likely to depend on the amount of fatty acids ingested, the efficacy of absorption in the small intestine, as well as the individual gut microbiome composition.

Upon intake of fat, bile is released into the small intestine to aid digestion of lipids. Bile acids constitute the primary component of bile and is conjugated by glycine (dominant in humans) and taurine, respectivelyCitation128. While approximately 95% of the bile acids are reabsorbed in the ileum and returned to the liver via the portal vein, unabsorbed bile acids end up in the colon where they undergo chemical transformations by the microbiota resulting in secondary bile acidsCitation129. Microbial deconjugation of bile acids reduces their reuptake in the small intestine, which affects the pool of bile acids entering the colonCitation129. Furthermore, microbial modification of bile acids affects their chemical structure leading to altered signaling via host receptors including the nuclear farnesoid X receptor (FXR) and G protein coupled bile acid receptor 1 (TGR5)Citation128, which may have major impact on host metabolism as discussed elsewhereCitation130,Citation131. Concordant with the succession of the gut microbial community in early life, the intestinal bile acid profiles develop according to both the microbiota-stage and nutritional stage. A longitudinal study with 10 healthy Japanese subjects found that during the first six months of life, infants dominated by Bifidobacterium species had high fecal levels of unconjugated primary bile acids whereas infants dominated by Enterobacteriaceae had high fecal levels of conjugated primary bile acidsCitation132. Indeed, bile salt hydrolase activity, catalyzing deconjugation of conjugated bile acids, is common in BifidobacteriumCitation133, suggesting that Bifidobacterium is a key player deconjugating bile acids during breastfeeding. Yet, as infants progress toward a solid diet, dietary energy intake goes upCitation77, dietary complexity and gut microbial diversity increaseCitation68. Consequently, both the total excretion of bile acids in the feces as well as the fecal levels of secondary bile acids increaseCitation85,Citation132. Furthermore, decreasing level of primary bile acids and an increase of secondary bile acids in feces have consistently been observed after weaningCitation132,Citation134,Citation135. Differences in fecal bile acid profiles have also been observed when comparing breastfed and formula-fed infants with breastfed infants showing higher levels of sulfated bile acids and formula fed infants showing higher levels of secondary bile acidsCitation85. Finally, a study in mice has shown that oral administration of bile acids to newborn mice accelerate postnatal microbiome maturationCitation136, suggesting that the progression in intestinal bile acid profiles during infancy also drives maturation of the postnatal gut microbiome.

Microbial metabolites as modulators of the microbiome community structuring

Gut microbial conversion of dietary substrates into metabolites represents a potential regulatory mechanism by which specific gut microbes impact the wider gut ecosystem (). In particular HMO-derived metabolites produced by primary HMO degraders drive cross-feeding networks that facilities growth of other early life taxa and crucial interspecies interactionsCitation137,Citation138. HMOs are poorly digested by the infant but are favored growth substrates for certain Bifidobacterium species and strains that can enzymatically degrade these complex dietary components that include LN(n)T, fucosylated and sialylated structuresCitation139. Along with Bifidobacterium, Bacteroides, Ruminococcus and more recently Akkermansia have also been shown to degrade these complex carbohydrates, although in most cases this relates to enzymatic cleavage of sugar side chains and correlates with associated mucin degradation pathwaysCitation140–142. Certain Bifidobacterium species seem to have limited impact on wider ecosystem structuring due to their ‘selfish’ digestion of HMOs. This is particularly true for B. longum subsp. infantis strains where they actively transport (via ABC transporters) and internalize whole (intact) HMOs to perform intracellular breakdown, utilizing the monosaccharide metabolites for their own cellular processes, rather than sharing among other early life microbesCitation143. Other species such as B. bifidum, Bifidobacterium pseudocatenulatum, B. longum subsp. longum and B. breve also carry out extracellular hydrolysis of complex HMOs, and then import and metabolize the resulting mono- and di-saccharides. These strains therefore provide an important keystone function during early life by providing simpler metabolites for cross-feedingCitation144,Citation145. Metabolites that can be used by other secondary degraders include the SCFAs acetate and lactate, formate, succinate, pyruvate, fucose, and 1,2-propanediol (1,2-PD)Citation84,Citation146. Previous work has indicated that within ‘mini’ Bifidobacterium ecosystems (from individual infants), there is often a keystone member that actively breakdown different HMOs, which provides key metabolites such as fucose, galactose, acetate and N-acetylglucosamine then utilized by other non-HMO degrading strainsCitation138. Other work supports this concept, showing that during growth on HMOs, cross-feeding interactions between multiple bifidobacterial strains increase overall cell numbers, which was linked to production of formate, acetate, 1,2-propanediol, and lactate during co-cultivationCitation147. Most recently, a study has indicated how ‘timing’ of sugars in the infant gut shape the wider Bifidobacterium ecosystem, with B. bifidum and B. longum subsp. infantis dominating through inhibitory priority effects based on their ability to digest HMOs, whilst B. breve benefits from facilitative priority effects and dominates by utilizing fucose (aligning with profiles observed in metagenomic datasets)Citation148. These studies highlight the importance of cross-feeding term ‘syntrophy’ for Bifidobacterium, which has been defined as ‘obligately mutualistic metabolism’ whereby without a complete mini ecosystem all or combinations of strains cannot survive without the other. This collective approach to HMO utilization may indicate additional processes by which multiple Bifidobacterium species and strains dominate the breastfed infant gut, and successfully outcompete other bacterial taxa. Indeed, acetate and lactate production by Bifidobacterium (in response to HMO-metabolism) is associated with lowering intestinal pH, which also correlates with changes in microbial composition, such as reducing levels of potential pathogens like Enterobacteriaceae, thus enhancing colonization resistance in the early life gutCitation149,Citation150. In agreement, in vitro experiments have found that SCFAs directly inhibit growth of enteric pathogens belonging to the Enterobacteriaceae family including Escherichia coli, Klebsiella and Salmonella in a pH-dependent mannerCitation151–153.

Figure 2. Microbial metabolites as modulators of the infant gut microbial ecosystem. An illustration of the ways in which microbial metabolites are thought to impact the development of the gut microbiome ecosystem during infancy. Human milk oligosaccharides (HMOs) of breast milk are likely to enter the distal gut and be degraded by HMO degraders (for example Bifidobacterium Bacteroides, Ruminococcus and Akkermansia). Among the HMO degraders, particular Bifidobacterium species are important as they are specialized in HMO degradation. Some Bifidobacterium species (mainly B. longum subsp. longum, B. bifidum and B. breve) degrade HMOs extracellular, whereas other Bifidobacterium species (mainly B. longum subsp. infantis) take up the HMOs via ABC transporters and metabolize them inside the cell. The sharing or lack of sharing of nutrients (both carbon sources and micronutrients) have consequences for cross-feeding and competition in the infant gut microbiome affecting the microbial community structure. As the child transitions from a milk-based diet to a solid food-based diet, the microbiome diversity increases and the cross-feeding networks change. Other primary degraders of dietary fibers (typically belonging to Lachnospiracea, Bacteroideacea, Clostridiacea) take over and replace the Bifidobacterium species. Generation of specific microbial metabolites may be key for colonization resistance toward pathobionts when microbiome diversity is low. The pathobionts may be excluded or kept low in abundance through low intestinal pH (due to short-chain fatty acids, SCFAs), through direct antimicrobial activity such as bacteriocins, which are antimicrobial, or simply through nutrient limitations.

Figure 2. Microbial metabolites as modulators of the infant gut microbial ecosystem. An illustration of the ways in which microbial metabolites are thought to impact the development of the gut microbiome ecosystem during infancy. Human milk oligosaccharides (HMOs) of breast milk are likely to enter the distal gut and be degraded by HMO degraders (for example Bifidobacterium Bacteroides, Ruminococcus and Akkermansia). Among the HMO degraders, particular Bifidobacterium species are important as they are specialized in HMO degradation. Some Bifidobacterium species (mainly B. longum subsp. longum, B. bifidum and B. breve) degrade HMOs extracellular, whereas other Bifidobacterium species (mainly B. longum subsp. infantis) take up the HMOs via ABC transporters and metabolize them inside the cell. The sharing or lack of sharing of nutrients (both carbon sources and micronutrients) have consequences for cross-feeding and competition in the infant gut microbiome affecting the microbial community structure. As the child transitions from a milk-based diet to a solid food-based diet, the microbiome diversity increases and the cross-feeding networks change. Other primary degraders of dietary fibers (typically belonging to Lachnospiracea, Bacteroideacea, Clostridiacea) take over and replace the Bifidobacterium species. Generation of specific microbial metabolites may be key for colonization resistance toward pathobionts when microbiome diversity is low. The pathobionts may be excluded or kept low in abundance through low intestinal pH (due to short-chain fatty acids, SCFAs), through direct antimicrobial activity such as bacteriocins, which are antimicrobial, or simply through nutrient limitations.

Looking beyond Bifidobacterium, HMO-derived metabolites also cross-feed other early life taxa. In vitro studies have shown that growth of Limosilactobacillus reuteri is enhanced through 1,2-propanediol produced by B. breve UCC2003Citation146. There has also been a number of studies exploring the cross-feeding impacts on a range of butyrate-producing bacteria, given their key health-promoting role. Specifically, butyrate-producing bacteria can utilize acetate and lactate, and HMO-derived monosaccharides as carbon sources, which serves to increase their levels in the infant gut. For example, Anaerostipes caccae has previously shown to only grow in co-culture with B. longum subsp. infantis, leading butyrate productionCitation154. Additional SCFAs production by Eubacterium hallii in response to (fucosylated) HMO breakdown by B. breve and/or B. longum subsp. infantis enables utilization of L-fucose (for production of butyrate), and 1,2-PD uptake for propionate productionCitation137, although the latter has yet to be demonstrated in the infant gut. The availability of L-fucose also supports growth of other microbes encoding Fuc catabolic pathways such as B. thetaiotaomicron and A. muciniphila, but these microbes can also cleave fucose from fucosylated HMOs, according to their fucosidase activityCitation155. Notably, some pathogenic bacteria have evolved strategies to use fucose to their advantage, including Salmonella and C. jejuni, which is the latter case appears to be enriched in breastfed infants when compared to those on formulaCitation156. Thus, the availability of different metabolites in the gut may both have beneficial and detrimental consequences depending on the ecological circumstances.

In some cases there may be competitive interactions via production of metabolites know to inhibit metabolic reactions and growth of other microbiota members e.g. hydrogen sulfide (H2S), or SCFAs (lowering the pH), or via production of metabolites with direct antimicrobial activity such as bacteriocins. Bacteriocins are ribosomally-synthesized antimicrobial peptides, which are active against other bacteria, and can be either narrow spectrum (i.e. targeting similar species) or broad spectrum (i.e. across diverse genera), with the host bacterium being resistantCitation157. A number of early life microbiota members, including Bifidobacterium, are known to produce these antimicrobials which may be linked to their ability to modulate the wider microbiota, and also inhibit overgrowth of potentially pathogenic bacteria that would otherwise be harmful to the infantCitation158. The gene clusters required for biosynthesis appear to be very strain specific; previous work has indicated that the bacteriocin bifidin (produced by Bifidobacterium bifidum NCDC 1452) has activity against a range of Gram positive and Gram negative’s including Escherichia coli and Staphylococcus aureusCitation158, which are common members of the early life gut microbiota. There are other Bifidobacterium-associated bacteriocins, however these have shown activity against specific food-borne pathogens including Salmonella spp. and Bacillus cereus, thus may not be so relevant in the context of the first 12 months of life. More recently, Bifidobacterium longum subsp. infantis LH_664 has been shown to encode and produce a narrow spectrum bacteriocin (putatively named Bifidococcin_664) that whilst not impacting growth of Lactobacillus and Enterococcus strains, does have discrete activity against a potentially important pathobiont in the (preterm) gut, Clostridium perfringensCitation159. The ability of these active peptides to target specific bacteria, may be particularly important in the context of early life perturbations (such as preterm birth, C-section and/or antibiotic treatments), when levels of pathobionts may be higher given the absence of these key metabolite/peptide modulating factors that would otherwise be produced by Bifidobacterium which may be depleted in these contexts. However at this stage it is unclear if the dietary (e.g. HMO) milieu in the gut plays a role in shaping the production of these peptides, and further work is required to disentangle these relationships.

With the introduction of additional complex dietary components (particularly plant-derived) during weaning there is a significant expansion in cross-feeding networks due to the resultant complex milieu of metabolites produced by primary degradersCitation144,Citation145. This is due to the huge variety of glycoside hydrolases encoded across different taxa (particularly in the Bacteroides genus, many of which act as primary degraders) that enables the diverse array of bonds between monosaccharide units to be ‘broken’, thus allowing these simple sugars to enter into different metabolism pathways to provide energy (ATP) for cellular responses and growthCitation160. Fermentation end products are further metabolized by cross-feeders which often results in the production of SCFAs, with increasing production of butyrate and propionate, in comparison to the acetate dominant SCFA profile observed during exclusive milk-based feedingCitation161. Additional key components that can be produced as a result of protein fermentation are amino acids, which play a central metabolism (enzyme co-factors) role for many bacteria, driving growth and changes in motility, biofilm formation and virulence factor expressionCitation162. Furthermore, there are strain level differences with respect to B vitamins biosynthesis, which is required by the host, and by other strains unable to de novo synthesize these key metabolitesCitation163. For example, several early life taxa are predicted, and have been experimentally confirmed, to produce vitamin B1 including some Bacteroides spp., Clostridium spp., Lactobacillus spp., and Bifidobacterium spp.Citation164. These strains are expected to play a key role for other species and strains that lack a vitamin B1 synthesis (e.g. Faecalibacterium spp.), highlighting the cross-feeding and also competition for this essential metabolite in the infant gutCitation165. It is clear that this time-period represents a highly dynamic period of ecosystem development, with burgeoning cross-feeding networks being established, and like other underdeveloped ecosystems, may be particularly sensitive to diet-derived perturbationsCitation166. However, clear evidence on the longer-term effects during this time period are required via both longitudinal cohorts and experimental studies. For a comprehensive review of this with a more complex (adult-like) ecosystem see a review by Oliphant and Allen-VercoCitation161.

Finally, the spatial context of these potential metabolite interactions should be considered. Clearly the levels and diffusability of key metabolites would be expected to have the most pronounced impact on ecosystem structure on those microbes in close proximity to each other. However, given the complex ‘geography’ of the gut, including the overall length, mucus layers, and micro-structure e.g crypts, it is likely that the studies undertaken so far may not completely replicate these spatial considerations. Therefore, additional model systems e.g. organoids and mice, may be required to pinpoint relative contributions of microbial metabolites as modulators of the infant gut microbiome.

Microbial metabolites as mediators of host-microbial cross talk in early life

Given the dynamics of microbial metabolites in the gut and their absorption into the circulationCitation167, these microbiota-derived molecules not only shape the gut microbiome ecosystem, but also likely play a central role in mediating host-microbial cross-talk in early life as they may communicate with host cells via host receptors in the intestine and distant organs ()Citation21,Citation168,Citation169. However, only few studies have studied the gut luminal and circulating concentrations of the metabolites in early life. This is an important gap to fill in order to figure out whether concentrations of the metabolites are within the range of the dose-response curve for the particular metabolite on the host receptor. Here we shortly discuss four areas where microbial metabolites may play a vital role in early life ().

Figure 3. Dynamics of selected signaling metabolites in the infant gut and their interaction with host receptors. The figure illustrates the change in relative abundances of known key microbiota-derived metabolites generated from saccharolyticCitation84 and proteolytic fermentationCitation84,Citation88 as well as from bile acid metabolismCitation132 in the infant gut. The metabolite profiles change as the infant goes from breastfeeding to solid foods concurrent with a maturation of the gut microbiome from a less to a more diverse microbiome. The main host receptor targets for each of the signaling metabolites is indicated to the right. As indicated by the arrows, some metabolites can bind several different host receptorsCitation168.

Figure 3. Dynamics of selected signaling metabolites in the infant gut and their interaction with host receptors. The figure illustrates the change in relative abundances of known key microbiota-derived metabolites generated from saccharolyticCitation84 and proteolytic fermentationCitation84,Citation88 as well as from bile acid metabolismCitation132 in the infant gut. The metabolite profiles change as the infant goes from breastfeeding to solid foods concurrent with a maturation of the gut microbiome from a less to a more diverse microbiome. The main host receptor targets for each of the signaling metabolites is indicated to the right. As indicated by the arrows, some metabolites can bind several different host receptorsCitation168.

Figure 4. Gut microbiota-derived metabolites as mediators of host-microbial cross-talk in early life. The microbiota-derived metabolites can impact host development in various ways as they activate different host receptors. For example, microbiota-derived metabolites can stimulate the intestinal immune system (affecting release of cytokines), enteroendocrine cells (affecting release of gut hormones), and enterochromaffin cells (affecting release of serotonin). Subsequently, the secreted molecules can affect the intestinal barrier and immune function, the intestinal nervous system with effects on intestinal motility and the gut-brain axis, and potentially the host metabolism and inflammation throughout the body upon absorption and circulation through the portal vein.

Figure 4. Gut microbiota-derived metabolites as mediators of host-microbial cross-talk in early life. The microbiota-derived metabolites can impact host development in various ways as they activate different host receptors. For example, microbiota-derived metabolites can stimulate the intestinal immune system (affecting release of cytokines), enteroendocrine cells (affecting release of gut hormones), and enterochromaffin cells (affecting release of serotonin). Subsequently, the secreted molecules can affect the intestinal barrier and immune function, the intestinal nervous system with effects on intestinal motility and the gut-brain axis, and potentially the host metabolism and inflammation throughout the body upon absorption and circulation through the portal vein.

Microbial metabolites and intestinal barrier function

Although a ‘strong’ barrier is considered essential for host health, during very early life a balance is required; allowing key microbial metabolites to cross and initiate immune programming in the underlying mucosal and at more distal and systemic sites. Butyrate is the main ‘fuel’ used by intestinal epithelial cells (IECs) for growthCitation170, however concentrations of butyrate are relatively low during milk feeding, but steadily increase across the weaning period and beyondCitation84, due to the expansion in butyrate producing bacteria given the increasingly complex dietary environmentCitation84. Acetate (and propionate) can be consumed by IECs, however this is a less efficient process than for butyrate. Nevertheless, acetate is also associated with inducing additional anti-inflammatory responses in the gut epithelium such as enhancing tight junction protein expression and anti-inflammatory cytokine productionCitation171. This appears to be mediated through Toll-like receptors (TLRs) and G protein-coupled receptors (GCPRs), including FFA2 pathway stimulation, as defined using KO mouse models. This acetate-ligand interaction can lead to NLRP3 inflammasome activationCitation172, which also links to a more efficient clearance of enteric pathogens from the gutCitation173, and enhanced production of sIgA (via Dendritic Cell-B cell activation). Of the other metabolites discussed above, such as B vitamins, it is appreciated these also directly modulate host immune responses, including the epithelial barrier (for a comprehensive overview see Yoshii et al.Citation174, and there is also work suggesting that bacteriocins may have a dual role, providing direct antimicrobial activity and also promoting immune responses, however this seems to be more with respect to innate immune cells like macrophagesCitation159. Furthermore, aromatic lactic acids could be important in early life since they may modulate immune function via both the aryl hydrocarbon receptor (AhR) and hydroxycarboxylic acid receptor 3 (HCA3)Citation88,Citation175,Citation176. During the last decade, tryptophan catabolites have emerged as potential key AhR-ligands playing a role in regulating intestinal barrier and immune functionCitation98. It was recently shown that fecal levels of indolelactic acid were positively correlated with the fecal samples’ ability to activate AhR in an AhR-reporter cell lineCitation88, suggesting that indolelactic acid is a key AhR-ligand in early life. However, it remains to be determined whether indolelactic acid and/or other microbiota-derived AhR ligands indeed play a role for immune barrier development in the context of early life. For all of these studies, it is important to note that most work in this area has concentrated on adult models. Previous work has suggested that microbes (including Bifidobacterium) drive distinct responses in the neonate vs. later life time pointsCitation177, thus there is a pressing need to determine how microbial metabolites such as aromatic lactic acids and SCFAs modulate host intestinal responses during the early life developmental window.

Microbial metabolites and intestinal motility

Stool frequency and consistency vary significantly according to early life nutrition between and within infants during the first year of lifeCitation178. Despite this, very little is known about the interplay between the diet, the developing gut microbiota, and the ever-changing bowel habits in early life. Several microbial metabolites, which have been measured in the infant gut, could potentially play a role in affecting gut motility in early life, although this has not yet been investigated. For example, tryptamine has been found to accelerate transit time in vivo through the 5-HT4 receptor (5-HT4R), a receptor uniquely expressed in the colonic epitheliumCitation118,Citation179. Other tryptophan-derived catabolites may also affect intestinal motility via AhR signaling in enteric neurons in the distal gastrointestinal tractCitation180. Furthermore, SCFAs can stimulate the release of serotonin (5-HT) in enterochromaffin cells by activating GPCRs such as FFA3 (GPR41), FFA2 (GPR43), and HCA2 (GPR109A). Serotonin has multiple paracrine and endocrine roles including promotion of peristalsis via the enteric nervous systemCitation181. Secondary bile acids and SCFAs may also through TGR5 and FFA2/FFA3, respectively, stimulate release of GLP-1 and PYY in enteroendocrine cells and thereby affect colonic motilityCitation182,Citation183. Variations in intestinal transit time have been associated with the gut microbiome composition and activity in adultsCitation184. Furthermore, given the large fluctuations and changes in SCFAs, secondary bile acids and proteolytic metabolites during the first year of life, it is possible that the gut microbiome contributes to inter- and intra-individual variations in bowel habits during infancy. Such variations could potentially be linked to common functional gastrointestinal disorders in infants such as infantile colic and functional constipationCitation185.

Microbial metabolites and whole body metabolism

SCFAs are proposed to affect whole body metabolism and inflammation upon absorption and circulation in the blood stream to distant organs through interactions with host receptorsCitation169. Furthermore, SCFAs can indirectly affect whole body metabolism by stimulating secretion of gut hormones (e.g. GLP-1 and PYY) in enteroendocrine cells affecting appetite and through stimulation of immune cells that regulate whole body metabolismCitation79. The high gut luminal levels of SCFAs, lactate, and succinate measured during infancy could possibly be important for recruitment of macrophages and neutrophils acting through FFA2, HCA1, and GPR91, respectivelyCitation168. However, in the context of early life, little is known about the role of SCFAs regulating whole body metabolism and inflammation. High fecal levels of formate during early infancy (3–4 months of age) have been associated with lower BMI z-score, whereas high fecal levels of butyrate have been associated with a higher BMI z-score at 3 years of age in Canadian infantsCitation186. Based on these observations one could speculate that the development of the infant gut metabolome from high formate (reflecting a low-diversity microbiome) to high butyrate (reflecting a more diverse microbiome) could play a role in growth trajectoriesCitation186. Yet, the timing of the gut metabolome development may be important, since another study found that Danish infants with normal weight gain at nine months of age had significantly higher fecal concentrations of butyrate compared to infants with high weight gainCitation111. In line with this, both exclusive formula feeding as well as early introduction to solid foods (vs later) in infancy has been associated with altered gut microbiota and increased risk of being overweight in early childhoodCitation52,Citation187. Prospective studies focusing on the infant gut metabolome development are warranted to understand whether the gut microbiota via production of metabolites is indeed casually involved in the programming of host metabolism, which may trigger long-lasting physiological effects that promote or prevent diseases.

Concluding remarks

Together, there is a clear need to uncover and explore microbial metabolites and their interactions with both gut microbes and host cells within early life. These microbial interactions may be pivotal for establishment of the gut microbiome and appropriate cross-talk between gut commensals and host cells with both short- and long-term physiological consequences. Currently, much of the work with regards to microbial metabolites and ecosystem structuring have been done in the context of the adult gut. Furthermore, only few aspects of cross-feeding in the infant gut such as Bifidobacterium-metabolism of HMOs and the generation of SCFAs have been studied. Therefore, prospective infant cohort studies following infants’ progression in diet, concurrent with their progression in gut microbiome composition and activity, are needed to further advance the field. By collecting and analyzing longitudinal fecal samples by untargeted metabolomics, unpreceded insights into known and novel microbial-derived metabolites, as well as their dynamics across and within infants during early life, can be providedCitation88. Combined with high-resolution microbiome profiling, enabling strain level classificationCitation20, as well as immune cell profilingCitation176, this may take the field to a whole new level. Furthermore, there is a need for relevant animal models and human organoids mimicking the context of early life to uncover the functional and mechanistic impact of metabolites on host development and specific pathways. Such insights are needed to develop/establish targeted strategies that contribute to healthy growth and development in infants, which may include manipulation of the infant gut microbiota using prebiotics, probiotics or postbiotics. However, before we can tackle these more translational aspects, we need to gain deeper insights into the range and mode-of-action of key microbial metabolites in the infant gut.

Author contributions

H.M.R, C.S., and L.H. wrote the manuscript. H.M.R. designed the figures. All authors approved the final version of the manuscript.

Disclosure statement

No potential conflict of interest was reported by the authors.

Data availability statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Additional information

Funding

This work was supported by the Independent Research Fund Denmark (MOTILITY; grant no. 0171-00006B to H.M.R.). LJH is supported by Wellcome Trust Investigator Award 220876/Z/20/Z; the Biotechnology and Biological Sciences Research Council (BBSRC), Institute Strategic Programme Gut Microbes and Health BB/R012490/1, and its constituent projects BBS/E/F/000PR10353 and BBS/E/F/000PR10356. CS is supported by Science Foundation Ireland, APC Microbiome Ireland and Vistamilk. The funders had no role in decision to publish and preparation of the manuscript.

References

  • Stewart CJ, Ajami NJ, O’brien JL, Hutchinson DS, Smith DP, Wong MC, Ross MC, Lloyd RE, Doddapaneni H, Metcalf GA, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018;562(7728):583–22. doi:10.1038/s41586-018-0617-x.
  • Tamburini S, Shen N, Wu HC, Clemente JC. The microbiome in early life: implications for health outcomes. Nat Med. 2016;22(7):713–722. doi:10.1038/nm.4142.
  • Dominguez-Bello MG, Godoy-Vitorino F, Knight R, Blaser MJ. Role of the microbiome in human development. Gut. 2019;68:1108–1114. doi:10.1136/gutjnl-2018-317503.
  • Ratsika A, Codagnone MC, O’mahony S, Stanton C, Cryan JF. Priming for life: early life nutrition and the microbiota-gut-brain axis. Nutrients. 2021;13:1–33. doi:10.3390/nu13020423.
  • Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, Belzer C, Delgado Palacio S, Arboleya Montes S, Mancabelli L, et al. The first microbial colonizers of the human gut: composition, activities, and health implications of the infant gut microbiota. Microbiol Mol Biol Rev. 2017;81(4). doi:10.1128/MMBR.00036-17.
  • Turroni F, Milani C, Duranti S, Lugli GA, Bernasconi S, Margolles A, Di Pierro F, van Sinderen D, Ventura M. The infant gut microbiome as a microbial organ influencing host well-being. Ital J Pediatr. 2020;46(1). doi:10.1186/s13052-020-0781-0.
  • Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227. doi:10.1038/nature11053.
  • Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, Li Y, Xia Y, Xie H, Zhong H, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17(5):690–703. doi:10.1016/j.chom.2015.04.004.
  • Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16(1):35–56. doi:10.1038/s41575-018-0061-2.
  • Bisgaard H, Li N, Bonnelykke K, Chawes BLK, Skov T, Paludan-Müller G, Stokholm J, Smith B, Krogfelt KA. Reduced diversity of the intestinal microbiota during infancy is associated with increased risk of allergic disease at school age. J Allergy Clin Immunol. 2011;128(3):646–652.e5. doi:10.1016/j.jaci.2011.04.060.
  • Cowan CSM, Dinan TG, Cryan JF. Annual Research Review: critical windows – the microbiota–gut–brain axis in neurocognitive development. J Child Psychol Psychiatry. 2020;61(3):353–371. doi:10.1111/jcpp.13156.
  • Swanson KS, Gibson GR, Hutkins R, Reimer RA, Reid G, Verbeke K, Scott KP, Holscher HD, Azad MB, Delzenne NM, et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat Rev Gastroenterol Hepatol. 2020;17(11):687–701. doi:10.1038/s41575-020-0344-2.
  • Żółkiewicz J, Marzec A, Ruszczyński M, Feleszko W. Postbiotics—a step beyond pre- and probiotics. Nutrients. 2020;12(8):1–17. doi:10.3390/nu12082189.
  • Murphy K, Ross RP, Ryan CA, Dempsey EM, Stanton CP. Prebiotics, and synbiotics for the prevention of necrotizing enterocolitis. Front Nutr. 2021;8. doi:10.3389/fnut.2021.667188.
  • Florez ID, Veroniki A-A, Al Khalifah R, Yepes-Nuñez JJ, Sierra JM, Vernooij RWM, Acosta-Reyes J, Granados CM, Pérez-Gaxiola G, Cuello-Garcia C, et al. Comparative effectiveness and safety of interventions for acute diarrhea and gastroenteritis in children: a systematic review and network meta-analysis. PLoS One. 2018;13(12):e0207701. doi:10.1371/journal.pone.0207701.
  • Quigley EMM, Gajula P. Recent advances in modulating the microbiome. F1000res. 2020;9:46. doi:10.12688/f1000research.20204.1.
  • Rehman S, Ali Z, Khan M, Bostan N, Naseem S. The dawn of phage therapy. Rev Med Virol. 2019;29:e2041. doi:10.1002/rmv.2041.
  • Dalile B, van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat Rev Gastroenterol Hepatol. 2019;16(8):461–478. doi:10.1038/s41575-019-0157-3.
  • Hegarty JW, Guinane CM, Ross RP, Hill C, Cotter PD. Bacteriocin production: a relatively unharnessed probiotic trait? F1000res. 2016;5:2587. doi:10.12688/f1000research.9615.1.
  • Enav H, Bäckhed F, Ley RE. The developing infant gut microbiome: a strain-level view. Cell Host Microbe. 2022;30(5):627–638. doi:10.1016/j.chom.2022.04.009.
  • Roager HM, Dragsted LO. Diet‐derived microbial metabolites in health and disease. Nutr Bull. 2019;44(3):216–227. doi:10.1111/nbu.12396.
  • Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A, Ben-Yacov O, Lador D, Avnit-Sagi T, Lotan-Pompan M, et al. Personalized nutrition by prediction of glycemic responses. Cell. 2015;163(5):1079–1094. doi:10.1016/j.cell.2015.11.001.
  • Berry SE, Am V, DA D, Asnicar F, Mazidi M, Wolf J, Capdevila J, Hadjigeorgiou G, Davies R, Al KH, et al. Human postprandial responses to food and potential for precision nutrition. Nat Med 2020. 2020;26:1–10.
  • Munyaka PM, Khafipour E, Ghia JE. External influence of early childhood establishment of gut microbiota and subsequent health implications. Front Pediatr. 2014;2. doi:10.3389/fped.2014.00109.
  • Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, Knight R. 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(26):11971–11975. doi:10.1073/pnas.1002601107.
  • Shao Y, Forster SC, Tsaliki E, Vervier K, Strang A, Simpson N, Kumar N, Stares MD, Rodger A, Brocklehurst P, et al. Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth. Nature. 2019;574(7776):117–121. doi:10.1038/s41586-019-1560-1.
  • Laursen MF, Bahl MI, Licht TR. Settlers of our inner surface – factors shaping the gut microbiota from birth to toddlerhood. FEMS Microbiol Rev. 2021;45(4). doi:10.1093/femsre/fuab001.
  • Laursen MF. Gut microbiota development: influence of diet from infancy to toddlerhood. Ann Nutr Metab. 2021;77(Suppl. 3):21–34. doi:10.1159/000517912.
  • Hill CJ, Lynch DB, Murphy K, Ulaszewska M, Jeffery IB, O’shea CA, Watkins C, Dempsey E, Mattivi F, Tuohy K, et al. Erratum to: evolution of gut microbiota composition from birth to 24 weeks in the infantmet Cohort. Microbiome. 2017;5(1):21. doi:10.1186/s40168-017-0240-3.
  • Fouhy F, Guinane CM, Hussey S, Wall R, Ryan CA, Dempsey EM, Murphy B, Ross RP, Fitzgerald GF, Stanton C, et al. High-throughput sequencing reveals the incomplete, short-term recovery of infant gut microbiota following parenteral antibiotic treatment with ampicillin and gentamicin. Antimicrob Agents Chemother. 2012;56(11):5811–5820. doi:10.1128/AAC.00789-12.
  • Reyman M, van Houten MA, Watson RL, Chu MLJN, Arp K, de Waal WJ, Schiering I, Plötz FB, Willems RJL, van Schaik W, et al. Effects of early-life antibiotics on the developing infant gut microbiome and resistome: a randomized trial. Nat Commun. 2022;13(1). doi:10.1038/s41467-022-28525-z.
  • Hernández E, Bargiela R, Diez MS, Friedrichs A, Pérez-Cobas AE, Gosalbes MJ, Knecht H, Martínez-Martínez M, Seifert J, von Bergen M, et al. Functional consequences of microbial shifts in the human gastrointestinal tract linked to antibiotic treatment and obesity. Gut Microbes. 2013;4(4):306–315. doi:10.4161/gmic.25321.
  • Murphy K, Curley D, O’callaghan TF, O’shea C-A, Dempsey EM, O’toole PW, Ross RP, Ryan CA, Stanton C. The composition of human milk and infant faecal microbiota over the first three months of life: a pilot study. Sci Rep. 2017;7(1). doi:10.1038/srep40597.
  • Jost T, Lacroix C, Braegger C, Chassard C. Assessment of bacterial diversity in breast milk using culture-dependent and culture-independent approaches. Br J Nutr. 2013;110(7):1253–1262. doi:10.1017/S0007114513000597.
  • Laursen MF, Zachariassen G, Bahl MI, Bergström A, Høst A, Michaelsen KF, Licht TR. Having older siblings is associated with gut microbiota development during early childhood. BMC Microbiol. 2015;15(1):154. doi:10.1186/s12866-015-0477-6.
  • Victora CG, Bahl R, Barros AJD, França GVA, Horton S, Krasevec J, Murch S, Sankar MJ, Walker N, Rollins NC. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet. 2016;387(10017):475–490. doi:10.1016/S0140-6736(15)01024-7.
  • Ahern GJ, Hennessy AA, Anthony Ryan C, Paul Ross R, Stanton C. Advances in infant formula science. Annu Rev Food Sci Technol. 2019;10(1):75–102. doi:10.1146/annurev-food-081318-104308.
  • Davis JCC, Totten SM, Huang JO, Nagshbandi S, Kirmiz N, Garrido DA, Lewis ZT, Wu LD, Smilowitz JT, German JB, et al. Identification of oligosaccharides in feces of breast-fed infants and their correlation with the gut microbial community. Mol Cell Proteomics. 2016;15(9):2987–3002. doi:10.1074/mcp.M116.060665.
  • Bode L. The functional biology of human milk oligosaccharides. Early Hum Dev. 2015;91(11):619–622. doi:10.1016/j.earlhumdev.2015.09.001.
  • Totten SM, Zivkovic AM, Wu S, Ngyuen U, Freeman SL, Ruhaak LR, Darboe MK, German JB, Prentice AM, Lebrilla CB. Comprehensive profiles of human milk oligosaccharides yield highly sensitive and specific markers for determining secretor status in lactating mothers. J Proteome Res. 2012;11(12):6124–6133. doi:10.1021/pr300769g.
  • Chung S, Bode L, Hall DA. Point-of-care human milk testing for maternal secretor status. Anal Bioanal Chem. 2022;414(10):3187–3196. doi:10.1007/s00216-021-03697-7.
  • Korpela K, Salonen A, Hickman B, Kunz C, Sprenger N, Kukkonen K, Savilahti E, Kuitunen M, de Vos WM. Fucosylated oligosaccharides in mother’s milk alleviate the effects of caesarean birth on infant gut microbiota. Sci Rep. 2018;8(1). doi:10.1038/s41598-018-32037-6.
  • Kunz C, Meyer C, Collado MC, Geiger L, García-Mantrana I, Bertua-Ríos B, Martínez-Costa C, Borsch C, Rudloff S. Influence of gestational age, secretor, and lewis blood group status on the oligosaccharide content of human milk. J Pediatr Gastroenterol Nutr. 2017;64(5):789–798. doi:10.1097/MPG.0000000000001402.
  • Cabrera-Rubio R, Kunz C, Rudloff S, García-Mantrana I, Crehuá-Gaudiza E, Martínez-Costa C, Collado MC. Association of maternal secretor status and human milk oligosaccharides with milk microbiota: an observational pilot study. J Pediatr Gastroenterol Nutr. 2019;68(2):256–263. doi:10.1097/MPG.0000000000002216.
  • Kumar H, Wacklin P, Nakphaichit M, Loyttyniemi E, Chowdhury S, Shouche Y, Mättö J, Isolauri E, Salminen S. Secretor status is strongly associated with microbial alterations observed during pregnancy. PLoS One. 2015;10(7):e0134623. doi:10.1371/journal.pone.0134623.
  • Wacklin P, Tuimala J, Nikkilä J, Tims S, Mäkivuokko H, Alakulppi N, Laine P, Rajilic-Stojanovic M, Paulin L, de Vos WM, et al. Faecal microbiota composition in adults is associated with the FUT2 gene determining the secretor status. PLoS One. 2014;9(4):e94863. doi:10.1371/journal.pone.0094863.
  • Wacklin P, Mäkivuokko H, Alakulppi N, Nikkilä J, Tenkanen H, Räbinä J, Partanen J, Aranko K, Mättö J. Secretor genotype (FUT2 gene) is strongly associated with the composition of Bifidobacteria in the human intestine. PLoS One. 2011;6(5):e20113. doi:10.1371/journal.pone.0020113.
  • Lodge CJ, Lowe AJ, Milanzi E, Bowatte G, Abramson MJ, Tsimiklis H, Axelrad C, Robertson B, Darling AE, Svanes C, et al. Human milk oligosaccharide profiles and allergic disease up to 18 years. J Allergy Clin Immunol. 2021;147(3):1041–1048. doi:10.1016/j.jaci.2020.06.027.
  • Seppo AE, Autran CA, Bode L, Järvinen KM. Human milk oligosaccharides and development of cow’s milk allergy in infants. J Allergy Clin Immunol. 2017;139(2):708–711.e5. doi:10.1016/j.jaci.2016.08.031.
  • Sprenger N, Odenwald H, Kukkonen AK, Kuitunen M, Savilahti E, Kunz C. FUT2-dependent breast milk oligosaccharides and allergy at 2 and 5 years of age in infants with high hereditary allergy risk. Eur J Nutr. 2017;56(3):1293–1301. doi:10.1007/s00394-016-1180-6.
  • Pärnänen KMM, Hultman J, Markkanen M, Satokari R, Rautava S, Lamendella R, Wright J, McLimans CJ, Kelleher SL, Virta MP. Early-life formula feeding is associated with infant gut microbiota alterations and an increased antibiotic resistance load. Am J Clin Nutr. 2022;115(2):407–421. doi:10.1093/ajcn/nqab353.
  • Forbes JD, Azad MB, Vehling L, Tun HM, Konya TB, Guttman DS, Field CJ, Lefebvre D, Sears MR, Becker AB, et al. Association of exposure to formula in the hospital and subsequent infant feeding practices with gut microbiota and risk of overweight in the first year of life. JAMA Pediatr. 2018;172(7):e181161. doi:10.1001/jamapediatrics.2018.1161.
  • Penders J, Thijs C, van den Brandt PA, Kummeling I, Snijders B, Stelma F, Adams H, van Ree R, Stobberingh EE. Gut microbiota composition and development of atopic manifestations in infancy: the koala birth cohort study. Gut. 2007;56(5):661–667. doi:10.1136/gut.2006.100164.
  • Rahman SF, Olm MR, Morowitz MJ, Banfield JF, Segata N. Machine learning leveraging genomes from metagenomes identifies influential antibiotic resistance genes in the infant gut microbiome. mSystems. 2018;3(1). doi:10.1128/mSystems.00123-17.
  • Sakanaka M, Gotoh A, Yoshida K, Odamaki T, Koguchi H, Xiao J-Z, Kitaoka M, Katayama T. Varied pathways of infant gut-associated bifidobacterium to assimilate human milk oligosaccharides: prevalence of the gene set and its correlation with bifidobacteria-rich microbiota formation. Nutrients. 2019;12(1):71. doi:10.3390/nu12010071.
  • Chow J, Panasevich MR, Alexander D, Vester Boler BM, Rossoni Serao MC, Faber TA, Bauer LL, Fahey GC. Fecal metabolomics of healthy breast-fed versus formula-fed infants before and during in vitro batch culture fermentation. J Proteome Res. 2014;13(5):2534–2542. doi:10.1021/pr500011w.
  • He X, Parenti M, Grip T, Domellöf M, Lönnerdal B, Hernell O, Timby N, Slupsky CM. Fecal microbiome and metabolome of infants fed bovine MFGM supplemented formula or standard formula with breast-fed infants as reference: a randomized controlled trial. Sci Rep. 2019;9(1). doi:10.1038/s41598-019-48858-y.
  • Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, DuGar B, Feldstein AE, Britt EB, Fu X, Chung Y-M, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57–63. doi:10.1038/nature09922.
  • Koh A, Molinaro A, Ståhlman M, Khan MT, Schmidt C, Mannerås-Holm L, Wu H, Carreras A, Jeong H, Olofsson LE, et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell. 2018;175(4):947–961.e17. doi:10.1016/j.cell.2018.09.055.
  • Nemet I, Saha PP, Gupta N, Zhu W, Romano KA, Skye SM, Cajka T, Mohan ML, Li L, Wu Y, et al. A cardiovascular disease-linked gut microbial metabolite acts via adrenergic receptors. Cell. 2020;180(5):862–877.e22. doi:10.1016/j.cell.2020.02.016.
  • Wu I-W, Hsu K-H, Lee C-C, Sun C-Y, Hsu H-J, Tsai C-J, Tzen C-Y, Wang Y-C, Lin C-Y, Wu M-S. P-Cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease. Nephrol Dial Transplant. 2011;26(3):938–947. doi:10.1093/ndt/gfq580.
  • Bosheva M, Tokodi I, Krasnow A, Pedersen HK, Lukjancenko O, Eklund AC, Grathwohl D, Sprenger N, Berger B, Cercamondi CI. Infant formula with a specific blend of five human milk oligosaccharides drives the gut microbiota development and improves gut maturation markers: a randomized controlled trial. Front Nutr. 2022;9. doi:10.3389/fnut.2022.920362.
  • Ma C, Wasti S, Huang S, Zhang Z, Mishra R, Jiang S, You Z, Wu Y, Chang H, Wang Y, et al. The gut microbiome stability is altered by probiotic ingestion and improved by the continuous supplementation of galactooligosaccharide. Gut Microbes. 2020;12(1):1–13. doi:10.1080/19490976.2020.1785252.
  • Vandenplas Y, Berger B, Carnielli V, Ksiazyk J, Lagström H, Sanchez Luna M, Migacheva N, Mosselmans J-M, Picaud J-C, Possner M, et al. Human Milk Oligosaccharides: 2′-Fucosyllactose (2′-FL) and Lacto-N-Neotetraose (LNnT) in Infant Formula. Nutrients. 2018;10(9):1161. doi:10.3390/nu10091161.
  • Laursen MF, Bahl MI, Michaelsen KF, Licht TR. First foods and gut microbes. Front Microbiol. 2017;8:356. doi:10.3389/fmicb.2017.00356.
  • Leong C, Haszard JJ, Lawley B, Otal A, Taylor RW, Szymlek-Gay EA, Fleming EA, Daniels L, Fangupo LJ, Tannock GW, et al. Mediation analysis as a means of identifying dietary components that differentially affect the fecal microbiota of infants weaned by modified baby-led and traditional approaches. Appl Environ Microbiol. 2018;84(18). doi:10.1128/AEM.00914-18.
  • Thompson AL, Monteagudo-Mera A, Cadenas MB, Lampl ML, Azcarate-Peril MA. Milk- and solid-feeding practices and daycare attendance are associated with differences in bacterial diversity, predominant communities, and metabolic and immune function of the infant gut microbiome. Front Cell Infect Microbiol. 2015;5. doi:10.3389/fcimb.2015.00003.
  • Laursen MF, Andersen LBB, Michaelsen KF, Mølgaard C, Trolle E, Bahl MI, Licht TR. Infant gut microbiota development is driven by transition to family foods independent of maternal obesity. mSphere. 2016;1(1):1–16. doi:10.1128/mSphere.00069-15.
  • Roswall J, Olsson LM, Kovatcheva-Datchary P, Nilsson S, Tremaroli V, Simon M-C, Kiilerich P, Akrami R, Krämer M, Uhlén M, et al. Developmental trajectory of the healthy human gut microbiota during the first 5 years of life. Cell Host Microbe. 2021;29(5):765–776.e3. doi:10.1016/j.chom.2021.02.021.
  • Subramanian S, Huq S, Yatsunenko T, Haque R, Mahfuz M, Alam MA, Benezra A, DeStefano J, Meier MF, Muegge BD, et al. Persistent gut microbiota immaturity in malnourished Bangladeshi children. Nature. 2014;510(7505):417–421. doi:10.1038/nature13421.
  • Blanton LV, Charbonneau MR, Salih T, Barratt MJ, Venkatesh S, Ilkaveya O, Subramanian S, Manary MJ, Trehan I, Jorgensen JM, et al. Gut bacteria that prevent growth impairments transmitted by microbiota from malnourished children. Science. 2016;351(6275). doi:10.1126/science.aad3311.
  • Smith MI, Yatsunenko T, Manary MJ, Trehan I, Mkakosya R, Cheng J, Kau AL, Rich SS, Concannon P, Mychaleckyj JC, et al. Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science. 2013;339(6119):548–554. doi:10.1126/science.1229000.
  • Robertson RC, Manges AR, Finlay BB, Prendergast AJ. The human microbiome and child growth – first 1000 days and beyond. Trends Microbiol. 2019;27:131–147. Preprint at. doi:10.1016/j.tim.2018.09.008.
  • Chen RY, Mostafa I, Hibberd MC, Das S, Mahfuz M, Naila NN, Islam MM, Huq S, Alam MA, Zaman MU, et al. A microbiota-directed food intervention for undernourished children. N Engl J Med. 2021;384(16):1517–1528. doi:10.1056/NEJMoa2023294.
  • Barratt MJ, Nuzhat S, Ahsan K, Frese SA, Arzamasov AA, Sarker SA, Islam MM, Palit P, Islam MR, Hibberd MC, et al. Bifidobacterium infantis treatment promotes weight gain in Bangladeshi infants with severe acute malnutrition. Sci Transl Med. 2022;14(640). doi:10.1126/scitranslmed.abk1107.
  • United Nations Children’s Fund (UNICEF). World Health Organization & International Bank for Reconstruction and Development/The World Bank. Levels and Trends in Child Malnutrition: Key Findings of the 2021 Edition of the Joint Child Malnutrition Estimates. 2021.
  • Grote V, Verduci E, Scaglioni S, Vecchi F, Contarini G, Giovannini M, Koletzko B, Agostoni C. Breast milk composition and infant nutrient intakes during the first 12 months of life. Eur J Clin Nutr. 2016;70(2):250–256. doi:10.1038/ejcn.2015.162.
  • Baumann-Dudenhoeffer AM, D’souza AW, Tarr PI, Warner BB, Dantas G. Infant diet and maternal gestational weight gain predict early metabolic maturation of gut microbiomes. Nat Med. 2018;24(12):1822–1829. doi:10.1038/s41591-018-0216-2.
  • Koh A, de Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165:1332–1345. doi:10.1016/j.cell.2016.05.041.
  • McNeil NI. The contribution of the large intestine to energy supplies in man. Am J Clin Nutr. 1984;39(2):338–342. doi:10.1093/ajcn/39.2.338.
  • de Vadder F, Kovatcheva-Datchary P, Goncalves D, Vinera J, Zitoun C, Duchampt A, Bäckhed F, Mithieux G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell. 2014;156(1–2):84–96. doi:10.1016/j.cell.2013.12.016.
  • Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. 1987;28:1221–1227. doi:10.1136/gut.28.10.1221.
  • den Besten G, Lange K, Havinga R, van Dijk TH, Gerding A, van Eunen K, Müller M, Groen AK, Hooiveld GJ, Bakker BM, et al. Gut-derived short-chain fatty acids are vividly assimilated into host carbohydrates and lipids. Am J Physiol Gastrointest Liver Physiol. 2013;305(12):G900–910. doi:10.1152/ajpgi.00265.2013.
  • Tsukuda N, Yahagi K, Hara T, Watanabe Y, Matsumoto H, Mori H, Higashi K, Tsuji H, Matsumoto S, Kurokawa K, et al. Key bacterial taxa and metabolic pathways affecting gut short-chain fatty acid profiles in early life. Isme J. 2021;15(9):2574–2590. 2021 15. doi:10.1038/s41396-021-00937-7.
  • Sillner N, Walker A, Lucio M, Maier TV, Bazanella M, Rychlik M, Haller D, Schmitt-Kopplin P. Longitudinal profiles of dietary and microbial metabolites in formula- and breastfed infants. Front Mol Biosci. 2021;8. doi:10.3389/fmolb.2021.660456.
  • Duncan SH, Louis P, Flint HJ. Lactate-utilizing bacteria, isolated from human feces, that produce butyrate as a major fermentation product. Appl Environ Microbiol. 2004;70(10):5810–5817. doi:10.1128/AEM.70.10.5810-5817.2004.
  • Bridgman SL, Azad MB, Field CJ, Haqq AM, Becker AB, Mandhane PJ, Subbarao P, Turvey SE, Sears MR, Scott JA, et al. Fecal short-chain fatty acid variations by breastfeeding status in infants at 4 months: differences in relative versus absolute concentrations. Front Nutr. 2017;4. doi:10.3389/fnut.2017.00011.
  • Laursen MF, Sakanaka M, von Burg N, Mörbe U, Andersen D, Jm M, Ct P, Rivollier A, Kf M, Mølgaard C, et al. Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut. Nat Microbiol. 2021;6(11):1367–1382. 2021 6. doi:10.1038/s41564-021-00970-4.
  • Thomson P, Medina DA, Garrido D. Human milk oligosaccharides and infant gut bifidobacteria: molecular strategies for their utilization. Food Microbiol. 2018;75:37–46. doi:10.1016/j.fm.2017.09.001.
  • Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM, Topping DL, Suzuki T, et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature. 2011;469(7331):543–547. doi:10.1038/nature09646.
  • Bode L. Human milk oligosaccharides: every baby needs a sugar mama. Glycobiology. 2012;22:1147–1162. doi:10.1093/glycob/cws074.
  • Soyyilmaz B, Mikš MH, Röhrig CH, Matwiejuk M, Meszaros-Matwiejuk A, Vigsnæs LK. The mean of milk: a review of human milk oligosaccharide concentrations throughout lactation. Nutrients. 2021;13(8):2737. doi:10.3390/nu13082737.
  • Bazanella M, Maier TV, Clavel T, Lagkouvardos I, Lucio M, Maldonado-Gòmez MX, Autran C, Walter J, Bode L, Schmitt-Kopplin P, et al. Randomized controlled trial on the impact of early-life intervention with bifidobacteria on the healthy infant fecal microbiota and metabolome. Am J Clin Nutr. 2017;106(5):1274–1286. doi:10.3945/ajcn.117.157529.
  • Wopereis H, Sim K, Shaw A, Warner JO, Knol J, Kroll JS. Intestinal microbiota in infants at high risk for allergy: effects of prebiotics and role in eczema development. J Allergy Clin Immunol. 2018;141(4):1334–1342.e5. doi:10.1016/j.jaci.2017.05.054.
  • Differding MK, Benjamin-Neelon SE, Hoyo C, Østbye T, Mueller NT. Timing of complementary feeding is associated with gut microbiota diversity and composition and short chain fatty acid concentrations over the first year of life. BMC Microbiol. 2020;20(1):56. doi:10.1186/s12866-020-01723-9.
  • Michaelsen KF, Grummer-Strawn L, Bégin F. Emerging issues in complementary feeding: global aspects. Matern Child Nutr. 2017;13:e12444. doi:10.1111/mcn.12444.
  • Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, Le A, Cowan TM, Nolan GP, Fischbach MA, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551(7682):648–652. doi:10.1038/nature24661.
  • Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun. 2018;9(1):3294. doi:10.1038/s41467-018-05470-4.
  • Guo C-J, Allen BM, Hiam KJ, Dodd D, Van Treuren W, Higginbottom S, Nagashima K, Fischer CR, Sonnenburg JL, Spitzer MH, et al. Depletion of microbiome-derived molecules in the host using Clostridium genetics. Sci (1979). 2019;366(6471):eaav1282. doi:10.1126/science.aav1282.
  • Singh R, Chandrashekharappa S, Bodduluri SR, Baby BV, Hegde B, Kotla NG, Hiwale AA, Saiyed T, Patel P, Vijay-Kumar M, et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat Commun. 2019;10(1):89. doi:10.1038/s41467-018-07859-7.
  • Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, Jensen BAH, Forslund K, Hildebrand F, Prifti E, Falony G, et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature. 2016;535(7612):376–381. doi:10.1038/nature18646.
  • Chen H, Pk N, Yang Y, Ce R, Aa B, Kuchroo M, Gw C, Ac K, Am R, Jm C, et al. A forward chemical genetic screen reveals gut microbiota metabolites that modulate host physiology. Cell. 2019;177(5):1217–1231.
  • Mazzoli R, Pessione E. The Neuro-endocrinological Role of Microbial Glutamate and GABA Signaling. Front Microbiol. 2016;7:1934. doi:10.3389/fmicb.2016.01934.
  • Fernández-Reina A, Urdiales JL, Sánchez-Jiménez F. What we know and what we need to know about aromatic and cationic biogenic amines in the gastrointestinal tract. Foods. 2018;7(9):145. doi:10.3390/foods7090145.
  • Brink LR, Mercer KE, Piccolo BD, Chintapalli SV, Elolimy A, Bowlin AK, Matazel KS, Pack L, Adams SH, Shankar K, et al. Neonatal diet alters fecal microbiota and metabolome profiles at different ages in infants fed breast milk or formula. Am J Clin Nutr. 2020;111(6):1190–1202. doi:10.1093/ajcn/nqaa076.
  • Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, Haqq AM, Shah SH, Arlotto M, Slentz CA, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9(4):311–326. doi:10.1016/j.cmet.2009.02.002.
  • Kok CR, Brabec B, Chichlowski M, Harris CL, Moore N, Wampler JL, Vanderhoof J, Rose D, Hutkins R. Stool microbiome, pH and short/branched chain fatty acids in infants receiving extensively hydrolyzed formula, amino acid formula, or human milk through two months of age. BMC Microbiol. 2020;20(1). doi:10.1186/s12866-020-01991-5.
  • Agostoni C, Boniglia E, Carratù B, Riva C, Sanzini E. Free amino acid content in standard infant formulas: comparison with human milk. J Am Coll Nutr. 2000;19(4):434–438. doi:10.1080/07315724.2000.10718943.
  • Heinig MJ, Nommsen LA, Peerson JM, Lonnerdal B, Dewey KG. Energy and protein intakes of breast-fed and formula-fed infants during the first year of life and their association with growth velocity: the DARLING Study. Am J Clin Nutr. 1993;58(2):152–161. doi:10.1093/ajcn/58.2.152.
  • He X, Sotelo-Orozco J, Rudolph C, Lönnerdal B, Slupsky CM. The role of protein and free amino acids on intake, metabolism, and gut microbiome: a comparison between breast-fed and formula-fed rhesus monkey infants. Front Pediatr. 2020;7. doi:10.3389/fped.2019.00563.
  • Pekmez CT, Larsson MW, Lind MV, Vazquez Manjarrez N, Yonemitsu C, Larnkjær A, Bode L, Mølgaard C, Michaelsen KF, Dragsted LO. Breastmilk lipids and oligosaccharides influence branched short-chain fatty acid concentrations in infants with excessive weight gain. Mol Nutr Food Res. 2020;64(3):1900977. doi:10.1002/mnfr.201900977.
  • Suárez L, Moreno-Luque M, Martínez-Ardines I, González N, Campo P, Huerta-Cima P, Sánchez M. Amine variations in faecal content in the first weeks of life of newborns in relation to breast-feeding or infant formulas. Br J Nutr. 2019;122(10):1130–1141. doi:10.1017/S0007114519001879.
  • Murray KE, Adams RF, Earl JW, Shaw KJ. Studies of the free faecal amines of infants with gastroenteritis and of healthy infants. Gut. 1986;27:1173–1180. doi:10.1136/gut.27.10.1173.
  • Ma J, Li Z, Zhang W, Zhang C, Zhang Y, Mei H, Zhuo N, Wang H, Wang L, Wu D. Comparison of gut microbiota in exclusively breast-fed and formula-fed babies: a study of 91 term infants. Sci Rep. 2020;10(1). doi:10.1038/s41598-020-72635-x.
  • Pugin B, Barcik W, Westermann P, Heider A, Wawrzyniak M, Hellings P, Akdis CA, O’mahony L. A wide diversity of bacteria from the human gut produces and degrades biogenic amines. Microb Ecol Health Dis. 2017;28(1):1353881. doi:10.1080/16512235.2017.1353881.
  • Ladero V, Fernández M, Calles-Enríquez M, Sánchez-Llana E, Cañedo E, Martín MC, Alvarez MA. Is the production of the biogenic amines tyramine and putrescine a species-level trait in enterococci? Food Microbiol. 2012;30(1):132–138. doi:10.1016/j.fm.2011.12.016.
  • Sagheddu V, Patrone V, Miragoli F, Puglisi E, Morelli L. Infant early gut colonization by lachnospiraceae: high frequency of ruminococcus gnavus. Front Pediatr. 2016;4. doi:10.3389/fped.2016.00057.
  • Williams BB, Van Benschoten A, Cimermancic P, Donia M, Zimmermann M, Taketani M, Ishihara A, Kashyap P, Fraser J, Fischbach M. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell Host Microbe. 2014;16(4):495–503. doi:10.1016/j.chom.2014.09.001.
  • Nejrup RG, Licht TR, Hellgren LI. Fatty acid composition and phospholipid types used in infant formulas modifies the establishment of human gut bacteria in germ-free mice. Sci Rep. 2017;7(1):1–11. 2017 7. doi:10.1038/s41598-017-04298-0.
  • Devillard E, McIntosh FM, Duncan SH, Wallace RJ. Metabolism of linoleic acid by human gut bacteria: different routes for biosynthesis of conjugated linoleic acid. J Bacteriol. 2007;189(6):2566. doi:10.1128/JB.01359-06.
  • Kishino S, Takeuchi M, Sb P, Hirata A, Kitamura N, Kunisawa J, Kiyono H, Iwamoto R, Isobe Y, Arita M, et al. Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition. Proc Natl Acad Sci U S A. 2013;110(44):17808–17813. doi:10.1073/pnas.1312937110.
  • Hennessy AA, Barrett E, Paul Ross R, Fitzgerald GF, Devery R, Stanton C. The production of conjugated α-linolenic, γ-linolenic and stearidonic acids by strains of bifidobacteria and propionibacteria. Lipids. 2012;47(3):313–327. doi:10.1007/s11745-011-3636-z.
  • Mei Y, Chen H, Yang B, Zhao J, Zhang H, Chen W. Research progress on conjugated linoleic acid bio-conversion in Bifidobacterium. Int J Food Microbiol. 2022;369:109593. doi:10.1016/j.ijfoodmicro.2022.109593.
  • Kim JH, Kim Y, Kim YJ, Park Y. Conjugated linoleic acid: potential health benefits as a functional food ingredient. Annu Rev Food Sci Technol. 2016;7(1):221–244. doi:10.1146/annurev-food-041715-033028.
  • Levan SR, Stamnes KA, Lin DL, Panzer AR, Fukui E, McCauley K, Fujimura KE, McKean M, Ownby DR, Zoratti EM, et al. Elevated faecal 12,13-diHOME concentration in neonates at high risk for asthma is produced by gut bacteria and impedes immune tolerance. Nat Microbiol. 2019;4(11):1851–1861. doi:10.1038/s41564-019-0498-2.
  • Fujimura KE, Sitarik AR, Havstad S, Lin DL, Levan S, Fadrosh D, Panzer AR, LaMere B, Rackaityte E, Lukacs NW, et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat Med. 2016;22(10):1187–1191. doi:10.1038/nm.4176.
  • Martin F-P, Tytgat HLP, Krogh Pedersen H, Moine D, Eklund AC, Berger B, Sprenger N. Host–microbial co-metabolites modulated by human milk oligosaccharides relate to reduced risk of respiratory tract infections. Front Nutr. 2022;9:1742. doi:10.3389/fnut.2022.935711.
  • Wahlström A, Sayin SI, Marschall HU, Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016;24(1):41–50. doi:10.1016/j.cmet.2016.05.005.
  • Ridlon JM, Kang D-J-J, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lipid Res. 2006;47(2):241–259. doi:10.1194/jlr.R500013-JLR200.
  • Thomas C, Pellicciari R, Pruzanski M, Auwerx J, Schoonjans K. Targeting bile-acid signalling for metabolic diseases. Nat Rev Drug Discov. 2008;7(8):678–693. doi:10.1038/nrd2619.
  • Hylemon PB, Zhou H, Pandak WM, Ren S, Gil G, Dent P. Bile acids as regulatory molecules. J Lipid Res. 2009;50(8):1509–1520. doi:10.1194/jlr.R900007-JLR200.
  • Tanaka M, Sanefuji M, Morokuma S, Yoden M, Momoda R, Sonomoto K, Ogawa M, Kato K, Nakayama J. The association between gut microbiota development and maturation of intestinal bile acid metabolism in the first 3 y of healthy Japanese infants. Gut Microbes. 2020;11(2):205–216. doi:10.1080/19490976.2019.1650997.
  • Tanaka H, Doesburg K, Iwasaki T, Mierau I. Screening of lactic acid bacteria for bile salt hydrolase activity. J Dairy Sci. 1999;82(12):2530–2535. doi:10.3168/jds.S0022-0302(99)75506-2.
  • Khine WWT, Rahayu ES, See TY, Kuah S, Salminen S, Nakayama J, Lee Y-K. Indonesian children fecal microbiome from birth until weaning was different from microbiomes of their mothers. Gut Microbes. 2020;12(1):1761240. doi:10.1080/19490976.2020.1761240.
  • Xiong JJ, Hu H-W, Xu C-Z, Yin J-W, Liu M, Zhang L-Z, Duan Y, Huang Y-K. Developmental patterns of fecal bile acids in healthy neonates and children. Med Sci Monit. 2021;27. doi:10.12659/MSM.928214.
  • van Best N, Rolle-Kampczyk U, Schaap FG, Basic M, Olde Damink SWM, Bleich A, Savelkoul PHM, von Bergen M, Penders J, Hornef MW. Bile acids drive the newborn’s gut microbiota maturation. Nat Commun. 2020;11(1). doi:10.1038/s41467-020-17183-8.
  • Schwab C, Ruscheweyh H-J, Bunesova V, Pham VT, Beerenwinkel N, Lacroix C. Trophic interactions of infant bifidobacteria and eubacterium hallii during L-fucose and fucosyllactose degradation. Front Microbiol. 2017;8:95. doi:10.3389/fmicb.2017.00095.
  • Lawson MAE, O’neill IJ, Kujawska M, Gowrinadh Javvadi S, Wijeyesekera A, Flegg Z, Chalklen L, Hall LJ. Breast milk-derived human milk oligosaccharides promote Bifidobacterium interactions within a single ecosystem. Isme J. 2020;14(2):635–648. doi:10.1038/s41396-019-0553-2.
  • Masi AC, Stewart CJ. Untangling human milk oligosaccharides and infant gut microbiome. iScience. 2022;25:103542. doi:10.1016/j.isci.2021.103542.
  • Kijner S, Cher A, Yassour M. The infant gut commensal bacteroides dorei presents a generalized transcriptional response to various human milk oligosaccharides. Front Cell Infect Microbiol. 2022;12:285. doi:10.3389/fcimb.2022.854122.
  • Crost EH, Tailford LE, Le Gall G, Fons M, Henrissat B, Juge N. Utilisation of mucin glycans by the human gut symbiont ruminococcus gnavus is strain-dependent. PLoS One. 2013;8(10):e76341. doi:10.1371/journal.pone.0076341.
  • Kostopoulos I, Elzinga J, Ottman N, Jt K, Blijenberg B, Aalvink S, Boeren S, Mank M, Knol J, Wm DV, et al. Akkermansia muciniphila uses human milk oligosaccharides to thrive in the early life conditions in vitro. Sci Rep. 2020;10(1):1–17. 2020 10. doi:10.1038/s41598-020-71113-8.
  • Sela DA, Mills DA. Nursing our microbiota: molecular linkages between bifidobacteria and milk oligosaccharides. Trends Microbiol. 2010;18(7):298–307. doi:10.1016/j.tim.2010.03.008.
  • Saa P, Urrutia A, Silva-Andrade C, Martín AJ, Garrido D. Modeling approaches for probing cross-feeding interactions in the human gut microbiome. Comput Struct Biotechnol J. 2021;20:79–89. doi:10.1016/j.csbj.2021.12.006.
  • Fernandez-Julia P, Commane DM, van Sinderen D, Munoz-Munoz J. Cross-feeding interactions between human gut commensals belonging to the Bacteroides and Bifidobacterium genera when grown on dietary glycans. Microbiome Res Rep. 2022;1:12. doi:10.20517/mrr.2021.05.
  • Cheng CC, Duar RM, Lin X, Perez-Munoz ME, Tollenaar S, Oh J-H, van Pijkeren J-P, Li F, van Sinderen D, Gänzle MG, et al. Ecological importance of cross-feeding of the intermediate metabolite 1,2-Propanediol between bacterial gut symbionts. Appl Environ Microbiol. 2020;86(11). doi:10.1128/AEM.00190-20.
  • Walsh C, Lane JA, van Sinderen D, Hickey RM. Human milk oligosaccharide-sharing by a consortium of infant derived Bifidobacterium species. Sci Rep. 2022;12(1):1–14. 2022 12. doi:10.1038/s41598-022-07904-y.
  • Ojima MN, Jiang L, Aa A, Yoshida K, Odamaki T, Xiao J, Nakajima A, Kitaoka M, Hirose J, Urashima T, et al. Priority effects shape the structure of infant-type Bifidobacterium communities on human milk oligosaccharides. Isme J. 2022;1–15. doi:10.1038/s41396-022-01270-3.
  • Alcon-Giner C, Dalby MJ, Caim S, Ketskemety J, Shaw A, Sim K, Lawson MAE, Kiu R, Leclaire C, Chalklen L, et al. Microbiota supplementation with bifidobacterium and lactobacillus modifies the preterm infant gut microbiota and metabolome: an observational study. Cell Rep Med. 2020;1(5):100077. doi:10.1016/j.xcrm.2020.100077.
  • Henrick BM, Hutton AA, Palumbo MC, Casaburi G, Mitchell RD, Underwood MA, Smilowitz JT, Frese SA. Elevated fecal pH indicates a profound change in the breastfed infant gut microbiome due to reduction of bifidobacterium over the past century. mSphere. 2018;3(2):e00041–18. doi:10.1128/mSphere.00041-18.
  • Sorbara MT, Dubin K, Littmann ER, Moody TU, Fontana E, Seok R, Leiner IM, Taur Y, Peled JU, van den Brink MRM, et al. Inhibiting antibiotic-resistant Enterobacteriaceae by microbiota-mediated intracellular acidification. J Exp Med. 2019;216(1):84–98. doi:10.1084/jem.20181639.
  • Jacobson A, Lam L, Rajendram M, Tamburini F, Honeycutt J, Pham T, Van Treuren W, Pruss K, Stabler SR, Lugo K, et al. A gut commensal-produced metabolite mediates colonization resistance to salmonella infection. Cell Host Microbe. 2018;24(2):296–307.e7. doi:10.1016/j.chom.2018.07.002.
  • Zhang S, Dogan B, Guo C, Herlekar D, Stewart K, Scherl EJ, Simpson KW. Short chain fatty acids modulate the growth and virulence of pathosymbiont escherichia coli and host response. Antibiot (Basel). 2020;9(8):1–20. doi:10.3390/antibiotics9080462.
  • Chia LW, Mank M, Blijenberg B, Bongers RS, van Limpt K, Wopereis H, Tims S, Stahl B, Belzer C, Knol J. Cross-feeding between Bifidobacterium infantis and Anaerostipes caccae on lactose and human milk oligosaccharides. Benef Microbes. 2021;12(1):69–83. doi:10.3920/BM2020.0005.
  • Glover JS, Ticer TD, Engevik MA. Characterizing the mucin-degrading capacity of the human gut microbiota. Sci Rep. 2022;12(1). doi:10.1038/s41598-022-11819-z.
  • Garber JM, Hennet T, Szymanski CM. Significance of fucose in intestinal health and disease. Mol Microbiol. 2021;115(6):1086–1093. doi:10.1111/mmi.14681.
  • Heilbronner S, Krismer B, Brötz-Oesterhelt H, Peschel A. The microbiome-shaping roles of bacteriocins. Nat Rev Microbiol. 2021;19(11):726–739. doi:10.1038/s41579-021-00569-w.
  • Martinez FAC, Balciunas EM, Converti A, Cotter PD, de Souza Oliveira RP. Bacteriocin production by Bifidobacterium spp. A review. Biotechnol Adv. 2013;31(4):482–488. doi:10.1016/j.biotechadv.2013.01.010.
  • Javvadi SG, Kujawska M, Papp D, Am G, Jordan A, Ma L, Ij O, Alcon-Giner C, Kiu R, Clarke P, et al. A novel bacteriocin produced by Bifidobacterium longum subsp. infantis has dual antimicrobial and immunomodulatory activity. bioRxiv. 2022;2022. doi:10.1101/2022.01.27.477972.
  • Zafar H, Saier MH. Gut Bacteroides species in health and disease. Gut Microbes. 2021;13(1):1–20. doi:10.1080/19490976.2020.1848158.
  • Oliphant K, Allen-Vercoe E. Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health. Microbiome. 2019;7(17):1–15. 2019. doi:10.1186/s40168-019-0704-8.
  • Lin R, Liu W, Piao M, Zhu H. A review of the relationship between the gut microbiota and amino acid metabolism. Amino Acids. 2017;49:2083–2090. doi:10.1007/s00726-017-2493-3.
  • Degnan PH, Taga ME, Goodman AL. Vitamin B12 as a modulator of gut microbial ecology. Cell Metab. 2014;20(5):769–778. doi:10.1016/j.cmet.2014.10.002.
  • Wan Z, Zheng J, Zhu Z, Sang L, Zhu J, Luo S, Zhao Y, Wang R, Zhang Y, Hao K, et al. Intermediate role of gut microbiota in vitamin B nutrition and its influences on human health. Front Nutr. 2022;9. doi:10.3389/fnut.2022.1031502.
  • Uebanso T, Shimohata T, Mawatari K, Takahashi A. Functional Roles of B-Vitamins in the Gut and Gut Microbiome. Mol Nutr Food Res. 2020;64(18):2000426. doi:10.1002/mnfr.202000426.
  • Valles-Colomer M, Blanco-Míguez A, Manghi P, Asnicar F, Dubois L, Golzato D, Armanini F, Cumbo F, Huang KD, Manara S, et al. The person-to-person transmission landscape of the gut and oral microbiomes. Nature. 2023;614(7946):125–135. doi:10.1038/S41586-022-05620-1.
  • Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A. 2009;106(10):3698–3703. doi:10.1073/pnas.0812874106.
  • Husted AS, Trauelsen M, Rudenko O, Hjorth SA, Schwartz TW. GPCR-Mediated signaling of metabolites. Cell Metab. 2017;25(4):777–796. doi:10.1016/j.cmet.2017.03.008.
  • Schroeder BO, Bäckhed F. Signals from the gut microbiota to distant organs in physiology and disease. Nat Med. 2016;22(10):1079–1089. doi:10.1038/nm.4185.
  • Salvi PS, Cowles RA. Butyrate and the intestinal epithelium: modulation of proliferation and inflammation in homeostasis and disease. Cells. 2021;10(7):1775. doi:10.3390/cells10071775.
  • Ma J, Piao X, Mahfuz S, Long S, Wang J. The interaction among gut microbes, the intestinal barrier and short chain fatty acids. Animal Nutr. 2022;9:159–174. doi:10.1016/j.aninu.2021.09.012.
  • Macia L, Tan J, Vieira AT, Leach K, Stanley D, Luong S, Maruya M, Ian McKenzie C, Hijikata A, Wong C, et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat Commun. 2015;6(1):6734. doi:10.1038/ncomms7734.
  • Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology. 2013;145(2):396–406.e10. doi:10.1053/j.gastro.2013.04.056.
  • Yoshii K, Hosomi K, Sawane K, Kunisawa J. Metabolism of dietary and microbial vitamin b family in the regulation of host immunity. Front Nutr. 2019;6:48. doi:10.3389/fnut.2019.00048.
  • Ehrlich AM, Pacheco AR, Henrick BM, Taft D, Xu G, Huda MN, Mishchuk D, Goodson ML, Slupsky C, Barile D, et al. Indole-3-lactic acid associated with Bifidobacterium-dominated microbiota significantly decreases inflammation in intestinal epithelial cells. BMC Microbiol. 2020;20(1). doi:10.1186/s12866-020-02023-y.
  • Henrick BM, Rodriguez L, Lakshmikanth T, Pou C, Henckel E, Arzoomand A, Olin A, Wang J, Mikes J, Tan Z, et al. Bifidobacteria-mediated immune system imprinting early in life. Cell. 2021;184(15):3884–3898.e11. doi:10.1016/j.cell.2021.05.030.
  • Kiu R, Treveil A, Harnisch LC, Caim S, Leclaire C, van Sinderen D, Korcsmaros T, Hall LJ. Bifidobacterium breve UCC2003 induces a distinct global transcriptomic program in neonatal murine intestinal epithelial cells. iScience. 2020;23(7):101336. doi:10.1016/j.isci.2020.101336.
  • Moretti E, Rakza T, Mestdagh B, Labreuche J, Turck D. The bowel movement characteristics of exclusively breastfed and exclusively formula fed infants differ during the first three months of life. Acta Paediatrica, Int J Paediatrics. 2019;108:877–881. doi:10.1111/apa.14620.
  • Bhattarai Y, Williams BB, Battaglioli EJ, Whitaker WR, Till L, Grover M, Linden DR, Akiba Y, Kandimalla KK, Zachos NC, et al. Gut microbiota-produced tryptamine activates an epithelial G-Protein-coupled receptor to increase colonic secretion. Cell Host Microbe. 2018;23(6):775–785.e5. doi:10.1016/j.chom.2018.05.004.
  • Obata Y, Castaño Á, Boeing S, Bon-Frauches AC, Fung C, Fallesen T, de Agüero MG, Yilmaz B, Lopes R, Huseynova A, et al. Neuronal programming by microbiota regulates intestinal physiology. Nature. 2020;578(7794):284–289. doi:10.1038/s41586-020-1975-8.
  • Soret R, Chevalier J, De Coppet P, Poupeau G, Derkinderen P, Segain JP, Neunlist M. Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterol. 2010;138(5):1772–1782. doi:10.1053/j.gastro.2010.01.053.
  • Ticho AL, Malhotra P, Dudeja PK, Gill RK, Alrefai WA. Bile acid receptors and gastrointestinal functions. Liver Res. 2019;3(1):31. doi:10.1016/j.livres.2019.01.001.
  • Wichmann A, Allahyar A, Greiner T, Plovier H, Lundén G, Larsson T, Drucker D, Delzenne N, Cani P, Bäckhed F. Microbial modulation of energy availability in the colon regulates intestinal transit. Cell Host Microbe. 2013;14(5):582–590. doi:10.1016/j.chom.2013.09.012.
  • Procházková N, Falony G, Lo D, Tr L, Raes J, Hm R. Advancing human gut microbiota research by considering gut transit time. Gut. 2022;72(1):180–191. gutjnl-2022-328166.
  • Vandenplas Y, Hauser B, Salvatore S. Functional gastrointestinal disorders in infancy: impact on the health of the infant and family. Pediatr Gastroenterol Hepatol Nutr. 2019;22(3):207–216. doi:10.5223/pghn.2019.22.3.207.
  • Bridgman SL, Malmuthuge N, Mandal R, Field CJ, Haqq AM, Mandhane PJ, Moraes TJ, Turvey SE, Simons E, Subbarao P, et al. Childhood body mass index and associations with infant gut metabolites and secretory IgA: findings from a prospective cohort study. Int J Obes (Lond). 2022;46(9):1712–1719. doi:10.1038/S41366-022-01183-3.
  • Differding MK, Doyon M, Bouchard L, Perron P, Guérin R, Asselin C, Massé E, Hivert M-F, Mueller NT. Potential interaction between timing of infant complementary feeding and breastfeeding duration in determination of early childhood gut microbiota composition and BMI. Pediatr Obes. 2020;15(8). doi:10.1111/ijpo.12642.