BBC Russian
Svoboda | Graniru | BBC Russia | Golosameriki | Facebook

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Establishment of the gut microbiome during early life is a complex process with lasting implications for an individual's health. Several factors influence microbial assembly; however, breast-feeding is recognized as one of the most influential drivers of gut microbiome composition during infancy, with potential implications for function. Differences in gut microbial communities between breast-fed and formula-fed infants have been consistently observed and are hypothesized to partially mediate the relationships between breast-feeding and decreased risk for numerous communicable and noncommunicable diseases in early life. Despite decades of research on the gut microbiome of breast-fed infants, there are large scientific gaps in understanding how human milk has evolved to support microbial and immune development. This review will summarize the evidence on how breast-feeding broadly affects the composition and function of the early-life gut microbiome and discuss mechanisms by which specific human milk components shape intestinal bacterial colonization, succession, and function.

Free full text 


Logo of nihpaLink to Publisher's site
J Allergy Clin Immunol. Author manuscript; available in PMC 2023 Sep 1.
Published in final edited form as:
PMCID: PMC9463492
NIHMSID: NIHMS1828112
PMID: 36075638

Gut Microbiome and Breast-feeding: Implications for Early Immune Regulation

Abstract

Establishment of the gut microbiome during early life is a complex process with lasting implications on an individual’s health. Several factors influence microbial assembly; however, breastfeeding is recognized as one of the most influential drivers of gut microbiome composition during infancy, with potential implications for function. Differences in gut microbial communities between breastfed and formula-fed infants have been consistently observed and are hypothesized to partially mediate the relationships between breastfeeding and decreased risk for numerous communicable and non-communicable diseases in early life. Despite decades of research on the gut microbiome of breastfed infants, there are large scientific gaps in understanding how human milk has evolved to support microbial and immune development. This review will summarize the evidence on how breastfeeding broadly impacts early life gut microbiome composition and function and discuss mechanisms by which specific human milk components shape intestinal bacterial colonization, succession, and function.

Keywords: Human milk, breastfeeding, microbiome, infant immunity, IgA, human milk oligosaccharides

Introduction

Infancy is a critical period for establishment of the gut microbiome, a complex process that is known to have a long-term impact on health and risk for disease. Microbial colonization of the gastrointestinal (GI) tract is fundamentally linked to metabolic programming, immunologic maturation, and proper gastrointestinal development.1,2 Perturbations in colonization in infancy have been associated with an increased risk for multiple conditions, including asthma, atopic dermatitis, food allergy, diabetes, inflammatory bowel disease, and obesity, highlighting early life as a critical window to shape the gut microbiome.3 Multiple perinatal factors, such as gestational age, mode of delivery, and antibiotic usage, impact microbial assembly in the GI tract, but infant diet is recognized as one of the most significant factors associated with early microbial structure.2,4,5 Human milk has evolved over time to not only provide nutrients necessary for growth but also a myriad of bioactive molecules that profoundly influence microbial colonization and succession.6,7 Breastfeeding has been shown to be protective against diarrheal disease and respiratory infections and associated with decreased risk for non-communicable, inflammatory diseases8, benefits that are hypothesized to be at least partially conferred through the gut microbiome.9 This review will explore how breastfeeding impacts the early life gut microbiome with an emphasis on mechanisms by which different human milk components shape bacterial colonization and fitness.

Gut Microbiome Composition in the Breastfed Infant

Much of the understanding about the impact of breastfeeding on the infant gut microbiome has been generated by decades of research highlighting differences in fecal bacterial composition between breastfed (BF) and formula-fed (FF) infants, specifics of which have been reviewed extensively by us and others.5,911 In a simplified, albeit generalizable model of succession, the GI tract is initially colonized by facultative anaerobes, such as Staphylococcus, Streptococcus, Enterobacteriaceae, and Lactobacillus. These pioneering taxa create an environment suitable for succession of obligate anaerobes, such as Bifidobacterium, Clostridium, and Bacteroides.5,10 Broadly speaking, formula feeding is associated with a more diverse fecal microbiota as well as a more mature microbiome as assessed by microbiota age and microbiota-for-age Z-score4,9, metrics originally developed by Subramanian and colleagues to compare assembly of the microbiome in malnourished relative to healthy infants of a similar chronological age.4,9,12 These features are often a result of high abundances of certain Bifidobacterium species that predominate during infancy in breastfed infants, in part, due to their unique capacity to metabolize human milk oligosaccharides (HMOs).2,4,9 Consumption of human milk has also been associated with higher abundances of Lactobacillus and lower abundances of Proteobacteria, Veillonella, Clostridium, and Bacteroides, though results vary among studies.2,4,5,9,13 Breastfeeding exclusivity (i.e. exclusive vs partial)13 and duration14 also impact the infant fecal microbiome, and dose-dependent effects of breastfeeding on gut microbiota composition have been demonstrated.13 A meta-analysis by Ho and colleagues also identified higher abundances of Eubacterium, Veillonella, and Bacteroides in the fecal microbiome of partially breastfed compared to exclusively BF infants.9 Cessation of breastfeeding, even more so than solid food introduction, dramatically shifts the infant microbiota towards a more mature and diverse, adult-like configuration marked by increased levels of Firmicutes.2,4,15,16

Though assessed in fewer studies, the demonstrated impact of human milk is not limited to the taxonomic profile (species abundance) but also extends to both metabolic capacity (functional genes) of resident microbiota and their output (metabolites). Studies employing shotgun metagenomic sequencing, which sequences all genetic material in a microbial community, have shown breastfeeding to be associated with enrichment of microbial genes related to carbohydrate and lipid metabolism, including fatty acid biosynthesis,4 as well as oxidative phosphorylation and vitamin B synthesis.2 On the other hand, the microbiome of infants not receiving any human milk has been characterized by greater abundance of genes related to bile acid synthesis and methanogenesis2 as well as nucleotide and amino acid metabolism4, processes that are characteristic of a more mature microbiome seen in adults.

Metabolomic studies have also demonstrated that the fecal metabolite pool discriminates BF and FF infants.17,18 BF infants have been shown to exhibit lower levels of conjugated and secondary bile acids but higher levels of sulfated bile acids18, the latter of which may represent an attempt at detoxification by decreasing buildup of bile acids.19 Furthermore, breastfeeding is associated with higher levels of several aromatic amino acid metabolites, including anti-inflammatory, indole-3-lactic acid (ILA)17,18, which will be discussed in more detail later in this review. Findings on the effects of breastfeeding on fecal organic acid and short chain fatty acid (SCFA) concentrations are mixed. BF infants from 3-5 months of age have been shown to exhibit increased levels of lactate and relative proportion of acetate compared to FF infants.20 However, in another study, breastfeeding was associated with increased fecal concentrations of butyrate and isovalerate at both 3 and 6 months relative to infants receiving cow’s milk or soy formula.17 SCFA are rapidly absorbed by colonocytes21 and SCFA and organic acids can be utilized by other resident microbiota, thus fecal concentrations may not accurately represent production, which is a consistent limitation in fecal metabolomic studies. Still, how these diet-related differences in functional capacity of and metabolic output by the microbiome influence infant health and development has not been fully resolved.

The Role of Specific Human Milk Components in Shaping the Infant Gut Microbiome

Human milk has several factors that may impact the composition and function of the infant gut microbiome. We discuss some of the most prominent factors below.

Human milk is a rich source of oligosaccharides, which select for bifidobacteria that mitigate intestinal inflammation and promote intestinal immune development

Human milk has evolved to supply essential nutrients as well as other non-essential components to support infant health and survival.7 The third largest fraction of molecules secreted in human milk are glycans referred to as human milk oligosaccharides (HMOs), composed of up to five monosaccharide building blocks varying in length and composition.22 HMO biosynthesis takes place in the mammary gland and is achieved by successive action of specific glycosyltransferases.23 Approximately 200 HMO structures have been identified to date24, where fucose and sialic acid moieties contribute to diversity and complexity relative to non-human mammalian milk.25 HMO concentrations vary among individuals and across lactational stages26, but median concentrations are approximately 8 g/L.27 These molecules are non-nutritive to the infant and arrive intact to the infant colon similar to soluble dietary fibers.2729 Thus, HMOs are available exclusively for utilization by microbiota capable of doing so.30

Several taxa within the genus Bifidobacterium have evolved mechanisms to bind, import, and catabolize HMOs intra- and extra-cellularly.31,32 Bifidobacteria ferment HMOs, and as a result, decrease the pH of the gut lumen through the secretion of organic acids, i.e. acetate, lactate, and formate (Fig 1). These organic acids promote the production of additional short-chain fatty acids (SCFA) by heterologous microbiota, collectively decreasing the pH of the gut lumen. SCFA are absorbed in the gut and are used as a source of energy by colon and liver cells, as well as peripheral tissues.33,34 However, the capacity of various bifidobacteria to ferment HMOs differs according to their makeup of HMO utilization genes within their genome. Bifidobacterium longum subsp. infantis (B. infantis) possesses all five of the HMO utilization gene clusters and is therefore a “superuser” of HMOs. Clusters H1 and H4 are rather unique to B. infantis, and B. breve has some genes of H2 and H5 and even H3. Species that harbor these gene clusters have a significant competitive advantage over other bacteria, allowing them to predominate the infant gut microbiome. In addition to SCFA related to energy metabolism, bifidobacteria produce indole-3-lactic acid (ILA) as a catabolite of tryptophan.35 ILA is hypothesized to be partly responsible for the anti-inflammatory effects associated with high abundances of B. infantis colonization in infancy as it interferes with transcription of the inflammatory cytokine IL-8 in enterocytes.36 In addition, tryptophan metabolites were found to regulate the expression of the ligand-binding subunit of the IL-10 receptor, implicating their role in the maintenance and repair of the intestinal epithelial barrier.37,38 HMOs may also shape the infant gut microbiome by acting as a decoy receptor for pathogens, preventing attachment to the intestinal epithelium and subsequent infection.39

An external file that holds a picture, illustration, etc.
Object name is nihms-1828112-f0001.jpg
Human Milk Oligosaccharides (HMO) drive Bifidobacterium colonization and shape the infant metabolome.

HMOs select for and drive colonization of specific Bifidobacterium species within the infant gut microbiome. Bifidobacterium produce acetate, formate, lactate, which can be further metabolized into SCFA by other resident microorganisms. Certain species of Bifidobacterium, such as B. infantis, can metabolize tryptophan into indole-3-lactic acid (ILA), among others. SCFA and ILA can be reabsorbed into the gut epithelium and redistributed to peripheral tissues, influencing the infant immune system through a variety of proposed mechanisms. Figure created with BioRender.com

HMOs select members of the genus Bifidobacterium, which often represent 50-70% of the infant gut microbiome early in life.40,41 However, it is likely that HMOs alone do not determine the presence or abundance of bifidobacteria. Several pre- and postnatal exposures such as cesarean sections, presence of older siblings, antibiotic use, short duration or non-exclusive breastfeeding, and urban lifestyles have been demonstrated to impact the succession of bifidobacteria early in life.4245 The effect of Bifidobacterium on development of infant immunity was examined recently by Henrick et al.46 Infants with expanded bifidobacteria had contracted populations of neutrophils, basophils, plasmablasts, and memory CD8+ cells as well as invariant T cells (MAIT) indicating effects on both the innate and adaptive immune system. They also had higher frequency of non-classical monocytes, which have been considered anti-inflammatory, and antigen-experienced CD39R+ regulatory T cells (Tregs) as well as decreased levels of inflammatory cytokines TNFα, IL-17A, and IL-1α as well as Th2 cytokine IL-13. Furthermore, infants harboring high abundances of bifidobacteria also had elevated levels of Treg-associated cytokines IL-27 and IL-10 and IL-1 inhibitor, IL1RA, but also unexpectedly elevated IL-6. To our knowledge, this is the only information available on the impact of Bifidobacterium succession on early immune phenotypes, warranting further investigation and replication in future cohort studies.

The prevalence of bifidobacteria, especially B. infiantis, in western populations is reduced compared to global populations, in particular relative to developing countries where breastfeeding rates and durations are historically higher.47 It is notable that infant formulas enriched with soluble fructo- and galacto-oligosaccharides enrich the infant gut with infant-type bifidobacterial taxa, although they may do so by non-specifically promoting other bacterial groups.48

B. infantis is a model taxon to elucidate mechanisms of attenuating intestinal inflammation and dysregulation. B. infantis decreases flagellin- and pathogen-induced IL-8 secretion in epithelial cells.49 High relative abundance (>50%) of B. infantis in early infancy is associated with improved CD4 T-cell responses and vaccine memory50, and high bifidobacterial taxonomic diversity is positively correlated to salivary secretory IgA levels, a marker for protection against development of allergic symptoms.51 The absence of HMO-utilization genes from the microbial metagenome, rather than the absence of specific taxa, is associated with dysregulated immunity.46 This finding was supported through a probiotic intervention with B. infantis that fully utilizes HMOs, preventing intestinal T helper 2 and T helper 17 cytokine response in favor of T helper 1 and Tregs. This is hypothesized to be due to upregulation of galectin-1 in T cells by B. infantis-derived ILA. Additionally, supplementation with B. infantis has been shown to reduce necrotizing enterocolitis in pre-clinical and clinical trials.52,53 However, a randomized controlled trial deploying B. breve54 did not demonstrate similar effects, highlighting further both the potential utility of B. infantis as well as important functional differences among species or strains of Bifidobacterium.

SIgA drives establishment and maintenance of the infant gut microbiome through specific and non-specific mechanisms

Human colostrum and mature milk contain high amounts of immunoglobulin A (IgA) antibodies and relatively low levels of IgG, which contrasts with plasma where IgG outnumbers IgA antibodies. This difference in the relative proportions of IgA to IgG in mucosal secretions is the result of active transport of IgA across the mammary epithelium, resulting in the formation of secretory IgA (SIgA).5557 SIgA consists of two or more IgA monomers linked together by joining (J) chain and secretory component (SC). Milk SIgA is produced from mucosal-homing α4β7, CCR10+ plasma cells (PCs)58 that are induced within gut-associated lymphoid tissues in response to food antigens, enteric pathogens and commensal biota59, which then home to the mammary gland through an entero-mammary pathway.60 Gut- and other mucosa-derived PCs preferentially secrete IgA dimeric and polymeric forms of IgA (not monomeric) into the mammary gland parenchyma.61 Dimeric and polymeric IgA is recognized by the pIgR on the basolateral membrane of mammary epithelial cells and is then transcytosed through the cell and secreted apically after proteolytic cleavage of the pIgR secretory component (SC).62 SC, which is acquired during IgA transport across the mammary gland epithelium, renders SIgA resistant to intestinal proteases and plays a role in antibody localization within mucus63 (Fig 2). SIgA is highly glycosylated with over 16 N-linked glycan sites located on the Ig, SC, and J chain.64

An external file that holds a picture, illustration, etc.
Object name is nihms-1828112-f0002.jpg
The biological activities of SIgA.

SIgA antibodies exert unique effector functions depending on the species being interacted with. SIgA mediates immune exclusion and enchained growth, restricts motility, and inhibits expression of toxins and virulence factors to defend against pathogenic organisms. Alternatively, SIgA supports commensal species by altering bacterial gene expression that in turn enhances growth and mucus colonization, supplying glycan residues as nutrients, and promoting overall diversity of the microbiome. Figure adapted from Yang and Palm, 2020189. Figure created with BioRender.com

IgA in Immune Exclusion, Neutralization, and Inhibition of Bacterial Colonization.

SIgA serves to protect the newborn from enteric and respiratory infections, although we are only just beginning to understand the mechanisms by which this occurs.6567 Cryo-electron microscopy indicates that SIgA assumes a V-shape, with the Fc elements tethered by their tails and the variable regions (Fab) projecting outwards.68,69 This molecular architecture explains how SIgA is so efficient at cross-linking and agglutinating viruses and bacterial cells in vitro and in vivo.70,71 SIgA-mediated agglutination results in mucus entrapment and clearance from the intestinal lumen via peristalsis, a phenomenon referred to as immune exclusion.72 Other mechanisms of immune protection by SIgA in the newborn likely include direct neutralization of toxins and microbial virulence factors, as well as inhibition of bacterial colonization and invasion.70 Maternally-derived SIgA can also suppress innate inflammatory responses by sequestering microbial pathogen-associated molecular patterns (PAMPs) and limiting their uptake into the neonatal intestinal mucosa.73,74

Some examples of human milk (HM) antibody associated protection against infectious diseases include studies showing that higher colostrum and mature milk antibody levels against enteric pathogens have been associated with protection against diarrheal diseases caused by rotavirus75,76, Shigella77, Giardia lamblia78,79, Entamoeba histolytica and Cryptosporidium spp80 and Campylobacter81. Studies of HM antibodies to respiratory pathogens have focused on the associations between maternal vaccination during pregnancy and HM IgG and IgA levels to pertussis, pneumococcus, influenza, and meningococcus.82 However, few studies have attempted to assess the degree of clinical benefit from HM antibodies on rates of disease in breastfed children. A report in Bangladeshi mothers described significantly higher vaccine-specific, viral neutralizing IgA levels to influenza A/New Caledonia in HM collected from women that were vaccinated compared to those not vaccinated.83 Furthermore, exclusive breastfeeding in the first 6 months of life significantly decreased the expected number of infant febrile respiratory illnesses, suggesting protection against influenza by these milk antibodies. Lastly, capsular antibodies to Group B Streptococcus (GBS) in HM have been associated with reduced risk of developing late-onset disease.84 More recently, neutralizing antibodies against SARS-CoV2 have been also found in HM from women that were infected with and/or vaccinated against SARS-CoV28589, although their role in protection against COVID-19 disease in breastfed infants has yet to be demonstrated. Other examples of virus-specific IgA activity playing a role in gut protection are from rotavirus and HIV-1.90 91,92

While direct evidence for a role of HM antibodies in protecting the newborn gut against enteric infections is lacking, the association of antigen-specific antibodies in HM and reduced disease incidence in infants is difficult to refute. In the case of HIV-1 infection, HM SIgA against viral envelope glycoproteins was associated with reduced postnatal transmission risk.93 Indeed, there are ongoing vaccination efforts to stimulate HIV-1 specific SIgA in HM as a means to reduce vertical transmission of the virus.94 In the case of rotavirus (RV), maternally derived RV-specific SIgA (and IgG) proved able to neutralize homologous virus in vitro.90 However, in the context of oral RV vaccination, RV-specific SIgA in HM reduced overall seroconversion rates, presumably due to “neutralization” of the vaccine itself.

Human Milk SIgA, Microbial Colonization and Immune Regulation.

SIgA in HM has profound long-term effects on the infant microbiota and immune regulation. Both poly-reactive95 and somatically hypermutated commensally-induced specific IgA96 bind commensal biota within the intestine. Mechanisms of SIgA interactions may explain the discrepancies related to binding specificity. Many IgA-commensal interactions are glycan-mediated, either from bacterial surface glycan structures like LPS on Gram negative species97, teichoic acid on Gram positive organisms98, and exopolysaccharides99, or from the SIgA glycoprotein itself, rich with N-linked glycans.100 Huus et al. offers a deep review of the many diverse mechanisms of SIgA-microbial interactions.101 In the intestine, SIgA directly impacts microbial colonization by promoting biofilm formations102 of slow-growing bacteria through enchained growth103, which enhances mucosal adhesion by glycan-glycan interactions and improves microbial cross-talk102. SIgA can also alter exopolysaccharide expression in commensal biota104 and promote niche colonization.105 In the colon, SIgA promotes colonization in the luminal but not epithelial mucosa, reducing aberrant host immune response and promoting tolerance.106 SIgA can also have a prebiotic effect, promoting growth of certain bacterial species. Some commensal microbes, like B. infantis, secrete endo-β-N-acetylglucosaminidase enzymes that can cleave the chitobiose core of N-linked glycans on SIgA, or other glycoproteins, which frees oligosaccharides that the bacteria can then use as an energy source.107,108

In mice, breeding schemes with pIgR (polymeric Ig receptor) knock-out (pigr−/− or KO) and heterozygote mating pairs provide unique opportunities to understand the impact of maternal SIgA (wt) vs lack of maternal SIgA (KO) on the developing neonate, as mouse pups do not begin endogenous SIgA production until weaning.57 Maternal SIgA was found to improve barrier protection, shape the gut microbiota, and program immune tolerance in pups, the effects of which remained through adulthood.106 In humans, maternal IgA does not cross the placenta, but infants begin producing their own SIgA sometime between 1-8 weeks of age.109,110 Longitudinal salivary IgA levels in infants have been shown to peak around 4-6 weeks with a later decline between 3-6 months of age,111113 though this pattern may differ at other mucosal sites, such as the intestine. Breastfed infants have been shown to have increased fecal IgA concentrations compared to formula-fed infants in the first 3 months of life.114,115 SIgA levels measured in infants are still considerably lower than in adults; therefore, SIgA in HM can supplement infant production during this window when infants are particularly vulnerable to infection. Additionally, Ramanan et al. found milk IgA led to coating of inflammatory commensal biota in the infant gut, which led to reduced induction of pro-inflammatory cytokines and thus induction of RORγ Tregs. This maternal IgA:infant microbiota interaction created a multigenerational transmission of immune phenotype programming.116 Collectively, information from observational studies in humans, animal studies, and in vitro investigations show maternal milk-derived SIgA as a strong driving force in the establishment and maintenance of the infant gut microbiome, although studies in human are scarce.

In the PASTURE study (Protection Against Allergy: Study in Rural Environments)117, regression analysis demonstrated an inverse relationship between the levels of IgA intake from HM feeding and the development of atopic dermatitis.118 Similar findings of higher total and cow’s milk-specific IgA levels in HM being associated with protection against cow’s milk allergy have also been reported by us.119,120 These associations do not necessarily indicate causation, although we showed HM IgA antibodies to prevent milk antigen transcytosis in a Caco2 monolayer consistent with immune exclusion. Other mechanisms could include IgA effect mediated by the gut microbiome and direct immune effects.120 Recently, we also identified differential glycosylation of IgA1 and pIgR, in addition to other glycoproteins, in HM of mothers whose infants did and did not develop atopic disease.121 These different glycan compositions could elicit direct effects on the infant immune system by controlling access to antigens or indirectly through effects on microbial composition. Indeed, SIgA is proposed to sculpt the newborns commensal microbiota though direct coating of beneficial bacterial species, while excluding harmful species, possibly in conjunction with maternally-derived IgG.104,122124 Gopalakrishna et al. found a significant inverse correlation between maternally-derived IgA coating of infant enteric bacteria and the development of necrotizing enterocolitis, a disease afflicting premature infants and a lead cause of infant mortality, indicating the essential role of milk IgA in regulating the infant gut microbial community.66

Human milk as a potential microbial reservoir for the infant gut

Findings over the last two decades support the presence of a diverse microbial ecosystem within HM.125 Staphylococcus and Streptococcus are the most prevalent and generally most dominant bacterial taxa in HM, but Gemella, Pseudomonas, Corynebacterium, Bacteroides, Veillonella, Rothia, Lactobacillus, and Actinomyces, among others, have also been consistently identified at notable abundances using 16s rRNA sequencing.126129 Origins of the HM microbiome are not entirely clear but remain an area of active investigation.125,130 Findings from studies employing 16S rRNA sequencing suggest that a large proportion of bacteria in HM arise from breast skin and the infant oral cavity,131133 the latter of which is believed to colonize HM during retrograde backflow of infant saliva into the mammary gland during suckling.134 Though these studies have not identify a large proportion of shared bacterial taxa between HM and maternal fecal bacterial communities, gut-associated, obligate anaerobes have been detected in HM, identifying the maternal gut as another possible origin site.130,135,136 Although debated, maternal gut bacteria are proposed to reach the mammary gland via immune-cell mediated translocation through an enteromammary pathway, the presence of which has been most strongly supported by evidence from rodent studies137139 as well as the detection of orally-administered probiotic strains in HM.140,141 Regardless of origin, HM remains a small, but consistent source of microorganisms for the infant gut, with an estimate that infants ingest roughly 1 x 107 – 1 x 108 bacterial cells per 800 mL of HM.142 While the bacterial composition of HM is fairly consistent within an individual, it is highly unique among mothers128,143,144 and may be one of the factors driving differences in gut microbiota composition, not only between BF and FF infants but also among infants who are exclusively BF.

Though not predominating, Bifidobacterium has also been identified in HM in appreciable numbers using both culture-dependent and independent techniques.135,144,145 Infant-associated bifidobacterial species, such as Bifidobacterium longum, Bifidobacterium bifidum, and Bifidobacterium breve, have all been detected in both HM and infant stool of breastfeeding dyads, but counts were not associated between the two sites.145 High co-occurrences of Actinomyces, Atopobium, Rothia, and Streptococcus have been demonstrated between HM and infant fecal microbial communities,13 and other studies have further demonstrated shared taxa between sites.133 In an in-depth analysis of microbial relationships between breastfeeding dyads, it was estimated that HM contributed on average 5% of the bacteria present in the infant gut at 2 days of age but decreased to less than 1% beyond 1 month, similar to results from our own studies.44,131,132 Though estimated contributions are low, the HM microbiota may play a role in early seeding of the infant gut, which could influence later microbial succession based on the principle of priority effects.146 Additionally, it is possible that the HM microbiota may contribute to the microbiome of the proximal gastrointestinal tract thereby indirectly shaping composition of the distal colon and feces123, relationships that may have gone undetected due to the use of fecal samples as a proxy of intestinal microbial communities.

Part of the ongoing investigation of how and if the HM microbiome impacts that of the infant gut is the question of bacterial viability in HM, as much of the recent work has been done using culture-independent sequencing techniques. Recent data from Stinson et. al showed that roughly half of the DNA isolated from HM arose from non-viable cells, findings which may help to explain the lack of robust associations between taxa abundance in HM and infant feces and certainly warrant further exploration.147 Further, other components in HM may mediate the impact of the HM microbiota on the infant gut microbiome. For example, SIgA could potentially neutralize and/or inhibit colonization of HM microbiota within the infant gut148 through previously discussed mechanisms. At the same time, one study has shown that up to 40% of the bacteria in HM are IgA-coated,149 which may promote their colonization within the infant gut.148,150 There still remains a paucity of information on the functional capabilities of the bacteria or other microorganisms in HM, such as bacteriophage.151,152 This is largely due to limitations presented by the low biomass nature of HM but represent critical areas for future research.

Gut dysbiosis is associated with atopic disease

As discussed above, HM has the capacity to impact the infant gut microbiome, which plays a significant role in development of many atopic diseases. Increasing evidence indicates that gut dysbiosis in early life negatively impacts the development of the immune system and precedes development of multi-sensitized atopic disease,153 atopic dermatitis,154 food allergy,155,156 and asthma (Fig 3).157 This may be in part due to induction of Tregs. Animal models of food allergy highlight a protective role of a consortia of clostridia in the induction of Tregs158 and the MyD88/RORγt pathway in Tregs,159 important for development of oral tolerance, as well reduction in intestinal permeability and subsequent sensitization to dietary antigens.160 Similarly in human studies, clostridia were enriched in stool of infants who outgrew milk allergy by age 8 years161 and were underrepresented at 3-6 months in stool of those who later developed food sensitization.156 Bifidobacteria also appear to play a role in anti-allergic immunity as lower levels in the first 6 months of life have been associated with a higher incidence of atopic disease,162 multi-sensitized atopy,153 and cow’s milk allergy.163 Our own studies demonstrating high abundance of B. infantis in farming lifestyle populations with low rate of allergic diseases compared to urban lifestyle high-risk infants further highlight the potential protective effect of Bifidobacterium.44 This protective effect may be in part due to production of ILA as well as organic and SCFA as previously discussed.

An external file that holds a picture, illustration, etc.
Object name is nihms-1828112-f0003.jpg
Potential relationships between breastfeeding, microbiome composition, and protection against atopic disease in early life.

There are multiple potential mechanisms by which human milk components, specifically HMOs, SIgA, and microbiota, may shape the infant microbiome, metabolite production, and early immune development. Recent studies have highlighted differential abundance of several bacterial taxa that discriminate infants who do and do not develop atopic disease, though how breastfeeding specifically mediates these relationships remains unclear. The proposed mechanisms by which the early microbiome protects against atopic disease include metabolite production, such as short-chain fatty acids (SCFA) and indole-3-lactic acid (ILA), LPS immunogenicity and immunomodulation by surface polysaccharides, which can result in downstream Treg differentiation and corresponding induction of pathways associated with oral tolerance, reinforcement of the epithelial barrier, and regulation of Th1/Th2 balance. Figure created with BioRender.com

In a recent study by Depner and colleagues, the fecal microbiome at 2 months of age was associated with breastfeeding and protection against asthma; however, this protective effect was mostly associated with abundance of Bacteroides rather than Bifidobacterium.164 In the same study, a more mature microbiome at 12 months of age, characterized by higher abundances of Coprococcus and Roseburia and abundance of genes related to butyrate synthesis, were associated with protection against later asthma development. Similarly, findings from the CHILD cohort in Canada suggest differential abundance of multiple taxa in the first year of life between infants who were and were not sensitized to food and aeroallergens at 12 months of age, including a number of genera in the Clostridia class such as Coprococcus, Roseburia, and Blautia among others.165 Metagenomic data from the GUSTO cohort also recently demonstrated aberrant longitudinal development in microbial composition and function in atopic infants.166 At just 3 weeks of age, infants who developed eczema exhibited decreased abundance of Bacteroides fragilis (B. fragilis) and increased abundance of Escherichia coli and Klebsiella pneumoniae, along with increased expression of virulence genes coding for adhesion, invasion, and flagellin. All three of these species were also associated with lipopolysaccharide (LPS) biosynthesis, suggesting that differences in LPS immunogenicity among early bacterial colonizers may influence allergy development similar to in autoimmunity.167 B. fragilis also harbors some capacity to metabolize HMOs168 and produce SCFA.169Additionally, B. fragilis produces multiple surface polysaccharides, the most immunodominant of which is Polysaccharide A (PSA). A study in germ-free animals demonstrated a critical role of PSA in immune homeostasis, specifically regulation of Th1/Th2 balance170, highlighting a potential protective role of early colonization of B. fragilis against atopic disease. Subsequently at 3, 6, and 12 months, atopic infants harbored a lower abundance of multiple bacterial species known to produce short-chain fatty acids (SCFA), butyrate and propionate.

To date, multiple cohorts have demonstrated associations between low levels of fecal SCFA, propionate and butyrate, and/or genes related to biosynthesis in the first two years of life and increased risk for eczema, food allergy, atopic sensitization, and asthma.166,171173 SCFA are prominent mediators of host-microbe interactions174,175 and likely influence atopic disease risk through their roles in epithelial tight junction assembly and barrier maintenance,174,176 direct modulation of immune cell function175, and their influence on host gene expression via histone deacetylase (HDAC) inhibition.179183 Through HDAC inhibition, acetate and butyrate have been shown to upregulate FOXP3 transcription, thereby inducing Treg differentiation and subsequent prevention of allergic and airway disease177,178 as well as amelioration of colitis183 in murine models.

Does breastfeeding mediate the relationship between the gut microbiome and allergic disease?

Despite the potential for HM to impact the infant gut microbiome, the association between breastfeeding and development atopic diseases is less clear with most consistent studies showing a protective effect with exclusive breastfeeding on atopic dermatitis and recurrent wheeze/asthma.184 The conflicting findings may be due to methodological limitations including reverse causality, varying definitions of breastfeeding, diagnostic methods for atopic disease, and/or sample size with most being underpowered for food allergy. Importantly, the term ‘breastfeeding’ does not account for the complexity of and/or interactions between the many HM factors, levels of which vary between mothers with different lifestyles185 and geographic locations.186 This may lead investigators to potentially miss the many nuances of the effect of breastfeeding on the gut microbiome. Finally, randomized controlled trials in this area, which are necessary to assess the true relationships of breastfeeding on the gut microbiome, on allergic diseases and on overall immune development, are lacking for obvious ethical reasons and are difficult due to confounding effects among these and other demographic variables. However, given that allergic outcomes such as atopic dermatitis and childhood asthma could be repressed in cohorts that are exclusively breastfed early in life8,44,187 and are colonized predominantly with infant-type bifidobacteria41, it is possible that breastfeeding creates a specific “niche” in the gut microbiome or in the progression of gut colonization that is supporting anti-allergic immune development. However, due to obvious ethical issues relating to randomization of breastfeeding, this relationship is difficult to establish.

Conclusions

The connection between HM, the infant gut microbiome, physiology and health is an active area of scientific inquiry. In addition to providing essential nutrients for growth, HM has evolved to contribute to guiding the assembly of the bacterial communities that colonize the gut during early life. As an example, the complexity of HMO biosynthesis in the mammary gland 23 matched with similarly complex utilization of HMO by bifidobacteria108, especially B. infantis, suggests a remarkable evolutionary adaptation to select bacterial species for early gut microbiome. This alone suggests that there is a specific benefit from these bacterial species, and that HM has also evolved to indirectly support the infant also indirectly by supporting a specific microbial composition. While the relationships between HMOs and members of the infant gut microbiota are fairly well understood, the potential mechanisms by which other components, those discussed in this review as well as others, shape bacterial communities in early life are only now emerging.

Similarly, while studies have identified relationships between bacterial taxa and metabolites with allergic disease, it is unclear how breastfeeding may mediate this relationship. In addition to deeper sequencing of the infant gut microbiome to gain better taxonomic resolution and insight into functional capacity, there is a need to characterize how HM components act synergistically to influence microbial development and metabolic output. Importantly, there it will be critical to conduct these studies longitudinally as the microbiome and infant diet rapidly change over the first year of life. Though not discussed in this review, assembly of other microorganisms, such as viruses, in the infant gut microbiome “niche” are shaped by breastfeeding and hold the potential to influence infant immune development, either independently or through interkingdom interactions with bacteria.188 Furthering our knowledge of the dynamic relationships between HM, the infant microbiome, and immune development will open the door for the development of microbiota-tailored interventions to improve human health.

Funding:

ECD is supported by USDA NIFA 67012-35010. DAS is supported by NICHD of the National Institutes of Health under award number R01HD106554. SL is supported by NIH R21AI154680. KMJ and AS are supported by U01 AI131344 from the National Institute of Infectious and Allergic Diseases. The content is solely the responsibility of the authors and does not necessarily represent the official views of the USDA or National Institutes of Health.

Conflict of Interest:

VC is an employee of TurtleTree LLC. KJM has received consulting fees from Merck, DBV Technologies and Janssen R&D. She has also received funding from NIH, Bill and Melinda Gates Foundation, Aimmune, and Janssen R&D for unrelated work. All other authors report no potential conflicts of interest.

Abbreviations

BFbreastfed
FFformula-fed
GIgastrointestinal
HMhuman milk
HMOhuman milk oligosaccharide
ILAIndol-3-lactic acid
PAMPSpathogen-associated molecular patterns
PCplasma cells
pIgRpolymeric immunoglobulin receptor
SCFAshort-chain fatty acids
SIgAsecretory immunoglobulin A
SCsecretory component

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

1. Collado MC, Cernada M, Baüerl C, Vento M & Pérez-Martínez G Microbial ecology and host-microbiota interactions during early life stages. Gut Microbes 3, 352–365 (2012). [Europe PMC free article] [Abstract] [Google Scholar]
2. Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe 17, 690–703 (2015). [Abstract] [Google Scholar]
3. Wang M, Monaco MH & Donovan SM Impact of early gut microbiota on immune and metabolic development and function. Semin Fetal Neonatal Med 21, 380–387 (2016). [Abstract] [Google Scholar]
4. Stewart CJ, Ajami NJ, O’Brien JL, Hutchinson DS, Smith DP, Wong MC, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature 562, 583–588 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
5. Davis EC, Wang M & Donovan SM The role of early life nutrition in the establishment of gastrointestinal microbial composition and function. Gut Microbes 8, 143–171 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
6. Le Doare K, Holder B, Bassett A & Pannaraj PS Mother’s Milk: A Purposeful Contribution to the Development of the Infant Microbiota and Immunity. Front Immunol 9, 361 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
7. German JB, Freeman SL, Lebrilla CB & Mills DA Human milk oligosaccharides: evolution, structures and bioselectivity as substrates for intestinal bacteria. Nestle Nutr Workshop Ser Pediatr Program 62, 205–218; discussion 218-222 (2008). [Europe PMC free article] [Abstract] [Google Scholar]
8. Victora CG, Bahl R, Barros AJ, França GV, Horton S, Krasevec J, et al. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet 387, 475–490 (2016). [Abstract] [Google Scholar]
9. Ho NT, Li F, Lee-Sarwar KA, Tun HM, Brown BP, Pannaraj PS, et al. Meta-analysis of effects of exclusive breastfeeding on infant gut microbiota across populations. Nat Commun 9, 4169 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
10. Rodríguez JM, Murphy K, Stanton C, Ross RP, Kober OI, Juge N, et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb Ecol Health Dis 26, 26050 (2015). [Europe PMC free article] [Abstract] [Google Scholar]
11. Davis EC, Dinsmoor AM, Wang M & Donovan SM Microbiome Composition in Pediatric Populations from Birth to Adolescence: Impact of Diet and Prebiotic and Probiotic Interventions. Dig Dis Sci 65, 706–722 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
12. Subramanian S, Huq S, Yatsunenko T, Haque R, Mahfuz M, Alam MA, et al. Persistent gut microbiota immaturity in malnourished Bangladeshi children. Nature 510, 417–421 (2014). [Europe PMC free article] [Abstract] [Google Scholar]
13. Fehr K, Moossavi S, Sbihi H, Boutin RCT, Bode L, Robertson B, et al. Breastmilk Feeding Practices Are Associated with the Co-Occurrence of Bacteria in Mothers’ Milk and the Infant Gut: the CHILD Cohort Study. Cell Host Microbe 28, 285–297 e284 (2020). [Abstract] [Google Scholar]
14. Forbes JD, Azad MB, Vehling L, Tun HM, Konya TB, Guttman DS, 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 172, e181161 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
15. Pannaraj PS, Li F, Cerini C, Bender JM, Yang S, Rollie A, et al. Association Between Breast Milk Bacterial Communities and Establishment and Development of the Infant Gut Microbiome. JAMA Pediatr 171, 647–654 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
16. Bergström A, Skov TH, Bahl MI, Roager HM, Christensen LB, Ejlerskov KT, et al. Establishment of intestinal microbiota during early life: a longitudinal, explorative study of a large cohort of Danish infants. Appl Environ Microbiol 80, 2889–2900 (2014). [Europe PMC free article] [Abstract] [Google Scholar]
17. Brink LR, Mercer KE, Piccolo BD, Chintapalli SV, Elolimy A, Bowlin AK, et al. Neonatal diet alters fecal microbiota and metabolome profiles at different ages in infants fed breast milk or formula. Am J Clin Nutr 111, 1190–1202 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
18. Sillner N, Walker A, Lucio M, Maier TV, Bazanella M, Rychlik M, et al. Longitudinal Profiles of Dietary and Microbial Metabolites in Formula- and Breastfed Infants. Front Mol Biosci 8, 660456 (2021). [Europe PMC free article] [Abstract] [Google Scholar]
19. Alnouti Y Bile Acid sulfation: a pathway of bile acid elimination and detoxification. Toxicol Sci 108, 225–246 (2009). [Abstract] [Google Scholar]
20. Bridgman SL, Azad MB, Field CJ, Haqq AM, Becker AB, Mandhane PJ, et al. Fecal Short-Chain Fatty Acid Variations by Breastfeeding Status in Infants at 4 Months: Differences in Relative versus Absolute Concentrations. Front Nutr 4, 11 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
21. den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud DJ & Bakker BM The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 54, 2325–2340 (2013). [Europe PMC free article] [Abstract] [Google Scholar]
22. Chen X Human Milk Oligosaccharides (HMOS): Structure, Function, and Enzyme Catalyzed Synthesis. Adv Carbohydr Chem Biochem 72, 113–190 (2015). [Europe PMC free article] [Abstract] [Google Scholar]
23. Kellman BP, Richelle A, Yang J-Y, Chapla D, Chiang AWT, Najera JA, et al. Elucidating Human Milk Oligosaccharide biosynthetic genes through network-based multi-omics integration. Nature Communications 13, 2455 (2022). [Europe PMC free article] [Abstract] [Google Scholar]
24. Ninonuevo MR, Park Y, Yin H, Zhang J, Ward RE, Clowers BH, et al. A strategy for annotating the human milk glycome. J Agric Food Chem 54, 7471–7480 (2006). [Abstract] [Google Scholar]
25. Tao N, Wu S, Kim J, An HJ, Hinde K, Power ML, et al. Evolutionary glycomics: characterization of milk oligosaccharides in primates. J Proteome Res 10, 1548–1557 (2011). [Europe PMC free article] [Abstract] [Google Scholar]
26. Coppa GV, Pierani P, Zampini L, Carloni I, Carlucci A & Gabrielli O Oligosaccharides in human milk during different phases of lactation. Acta Paediatr Suppl 88, 89–94 (1999). [Abstract] [Google Scholar]
27. Kunz C, Rudloff S, Baier W, Klein N & Strobel S Oligosaccharides in human milk: structural, functional, and metabolic aspects. Annu Rev Nutr 20, 699–722 (2000). [Abstract] [Google Scholar]
28. Engfer MB, Stahl B, Finke B, Sawatzki G & Daniel H Human milk oligosaccharides are resistant to enzymatic hydrolysis in the upper gastrointestinal tract. Am J Clin Nutr 71, 1589–1596 (2000). [Abstract] [Google Scholar]
29. Gnoth MJ, Kunz C, Kinne-Saffran E & Rudloff S Human milk oligosaccharides are minimally digested in vitro. J Nutr 130, 3014–3020 (2000). [Abstract] [Google Scholar]
30. Marcobal A & Sonnenburg JL Human milk oligosaccharide consumption by intestinal microbiota. Clin Microbiol Infect 18 Suppl 4, 12–15 (2012). [Europe PMC free article] [Abstract] [Google Scholar]
31. Sela DA, Chapman J, Adeuya A, Kim JH, Chen F, Whitehead TR, et al. The genome sequence of Bifidobacterium longum subsp. infantis reveals adaptations for milk utilization within the infant microbiome. Proc Natl Acad Sci U S A 105, 18964–18969 (2008). [Europe PMC free article] [Abstract] [Google Scholar]
32. Turroni F, Bottacini F, Foroni E, Mulder I, Kim JH, Zomer A, et al. Genome analysis of Bifidobacterium bifidum PRL2010 reveals metabolic pathways for host-derived glycan foraging. Proc Natl Acad Sci U S A 107, 19514–19519 (2010). [Europe PMC free article] [Abstract] [Google Scholar]
33. Li M, Bauer LL, Chen X, Wang M, Kuhlenschmidt TB, Kuhlenschmidt MS, et al. Microbial composition and in vitro fermentation patterns of human milk oligosaccharides and prebiotics differ between formula-fed and sow-reared piglets. J Nutr 142, 681–689 (2012). [Europe PMC free article] [Abstract] [Google Scholar]
34. Wong JM, de Souza R, Kendall CW, Emam A & Jenkins DJ Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol 40, 235–243 (2006). [Abstract] [Google Scholar]
35. Aragozzini F, Ferrari A, Pacini N & Gualandris R Indole-3-lactic acid as a tryptophan metabolite produced by Bifidobacterium spp. Appl Environ Microbiol 38, 544–546 (1979). [Europe PMC free article] [Abstract] [Google Scholar]
36. Meng D, Sommella E, Salviati E, Campiglia P, Ganguli K, Djebali K, et al. Indole-3-lactic acid, a metabolite of tryptophan, secreted by Bifidobacterium longum subspecies infantis is anti-inflammatory in the immature intestine. Pediatr Res 88, 209–217 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
37. Lanis JM, Alexeev EE, Curtis VF, Kitzenberg DA, Kao DJ, Battista KD, et al. Tryptophan metabolite activation of the aryl hydrocarbon receptor regulates IL-10 receptor expression on intestinal epithelia. Mucosal Immunol 10, 1133–1144 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
38. Kominsky DJ, Campbell EL, Ehrentraut SF, Wilson KE, Kelly CJ, Glover LE, et al. IFN-γ-mediated induction of an apical IL-10 receptor on polarized intestinal epithelia. J Immunol 192, 1267–1276 (2014). [Europe PMC free article] [Abstract] [Google Scholar]
39. Newburg DS, Ruiz-Palacios GM & Morrow AL Human milk glycans protect infants against enteric pathogens. Annu Rev Nutr 25, 37–58 (2005). [Abstract] [Google Scholar]
40. Davis JC, Lewis ZT, Krishnan S, Bernstein RM, Moore SE, Prentice AM, et al. Growth and Morbidity of Gambian Infants are Influenced by Maternal Milk Oligosaccharides and Infant Gut Microbiota. Sci Rep 7, 40466 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
41. Turroni F, Peano C, Pass DA, Foroni E, Severgnini M, Claesson MJ, et al. Diversity of bifidobacteria within the infant gut microbiota. PLoS One 7, e36957 (2012). [Europe PMC free article] [Abstract] [Google Scholar]
42. Adlerberth I, Strachan DP, Matricardi PM, Ahrne S, Orfei L, Aberg N, et al. Gut microbiota and development of atopic eczema in 3 European birth cohorts. J Allergy Clin Immunol 120, 343–350 (2007). [Abstract] [Google Scholar]
43. Chen YY, Zhao X, Moeder W, Tun HM, Simons E, Mandhane PJ, et al. Impact of Maternal Intrapartum Antibiotics, and Caesarean Section with and without Labour on Bifidobacterium and Other Infant Gut Microbiota. Microorganisms 9(2021). [Europe PMC free article] [Abstract] [Google Scholar]
44. Seppo AE, Bu K, Jumabaeva M, Thakar J, Choudhury RA, Yonemitsu C, et al. Infant gut microbiome is enriched with Bifidobacterium longum ssp. infantis in Old Order Mennonites with traditional farming lifestyle. Allergy (2021). [Europe PMC free article] [Abstract] [Google Scholar]
45. Tannock GW, Lawley B, Munro K, Gowri Pathmanathan S, Zhou SJ, Makrides M, et al. Comparison of the compositions of the stool microbiotas of infants fed goat milk formula, cow milk-based formula, or breast milk. Appl Environ Microbiol 79, 3040–3048 (2013). [Europe PMC free article] [Abstract] [Google Scholar]
46. Henrick BM, Rodriguez L, Lakshmikanth T, Pou C, Henckel E, Arzoomand A, et al. Bifidobacteria-mediated immune system imprinting early in life. Cell 184, 3884–3898.e3811 (2021). [Abstract] [Google Scholar]
47. Taft DH, Lewis ZT, Nguyen N, Ho S, Masarweh C, Dunne-Castagna V, et al. Bifidobacterium Species Colonization in Infancy: A Global Cross-Sectional Comparison by Population History of Breastfeeding. Nutrients 14(2022). [Europe PMC free article] [Abstract] [Google Scholar]
48. Haarman M & Knol J Quantitative real-time PCR assays to identify and quantify fecal Bifidobacterium species in infants receiving a prebiotic infant formula. Appl Environ Microbiol 71, 2318–2324 (2005). [Europe PMC free article] [Abstract] [Google Scholar]
49. O’Hara AM, O’Regan P, Fanning A, O’Mahony C, Macsharry J, Lyons A, et al. Functional modulation of human intestinal epithelial cell responses by Bifidobacterium infantis and Lactobacillus salivarius. Immunology 118, 202–215 (2006). [Abstract] [Google Scholar]
50. Huda MN, Ahmad SM, Alam MJ, Khanam A, Kalanetra KM, Taft DH, et al. Bifidobacterium Abundance in Early Infancy and Vaccine Response at 2 Years of Age. Pediatrics 143(2019). [Europe PMC free article] [Abstract] [Google Scholar]
51. Sjögren YM, Tomicic S, Lundberg A, Böttcher MF, Björkstén B, Sverremark-Ekström E, et al. Influence of early gut microbiota on the maturation of childhood mucosal and systemic immune responses. Clin Exp Allergy 39, 1842–1851 (2009). [Abstract] [Google Scholar]
52. Tobias J, Olyaei A, Laraway B, Jordan BK, Dickinson SL, Golzarri-Arroyo L, et al. Bifidobacteriumlongum subsp. infantis EVC001 Administration Is Associated with a Significant Reduction in the Incidence of Necrotizing Enterocolitis in Very Low Birth Weight Infants. J Pediatr (2022). [Europe PMC free article] [Abstract] [Google Scholar]
53. Lueschow SR, Boly TJ, Frese SA, Casaburi G, Mitchell RD, Henrick BM, et al. Bifidobacterium longum Subspecies infantis Strain EVC001 Decreases Neonatal Murine Necrotizing Enterocolitis. Nutrients 14(2022). [Europe PMC free article] [Abstract] [Google Scholar]
54. Costeloe K, Bowler U, Brocklehurst P, Hardy P, Heal P, Juszczak E, et al. A randomised controlled trial of the probiotic Bifidobacterium breve BBG-001 in preterm babies to prevent sepsis, necrotising enterocolitis and death: the Probiotics in Preterm infantS (PiPS) trial. Health Technol Assess 20, 1–194 (2016). [Abstract] [Google Scholar]
55. Brandtzaeg P The secretory immune system of lactating human mammary glands compared with other exocrine organs. Ann N Y Acad Sci 409, 353–382 (1983). [Abstract] [Google Scholar]
56. Demers-Mathieu V, Underwood MA, Beverly RL, Nielsen SD & Dallas DC Comparison of Human Milk Immunoglobulin Survival during Gastric Digestion between Preterm and Term Infants. Nutrients 10(2018). [Europe PMC free article] [Abstract] [Google Scholar]
57. Johansen FE, Pekna M, Norderhaug IN, Haneberg B, Hietala MA, Krajci P, et al. Absence of epithelial immunoglobulin A transport, with increased mucosal leakiness, in polymeric immunoglobulin receptor/secretory component-deficient mice. J Exp Med 190, 915–922 (1999). [Europe PMC free article] [Abstract] [Google Scholar]
58. Roux ME, McWilliams M, Phillips-Quagliata JM, Weisz-Carrington P & Lamm ME Origin of IgA-secreting plasma cells in the mammary gland. J Exp Med 146, 1311–1322 (1977). [Europe PMC free article] [Abstract] [Google Scholar]
59. Li H, Limenitakis JP, Greiff V, Yilmaz B, Schären O, Urbaniak C, et al. Mucosal or systemic microbiota exposures shape the B cell repertoire. Nature 584, 274–278 (2020). [Abstract] [Google Scholar]
60. Hanson LA & Korotkova M The role of breastfeeding in prevention of neonatal infection. Seminars in neonatology : SN 7, 275–281 (2002). [Abstract] [Google Scholar]
61. Nickerson SC, Pankey JW & Boddie NT Distribution, location, and ultrastructure of plasma cells in the uninfected, lactating bovine mammary gland. J Dairy Res 51, 209–217 (1984). [Abstract] [Google Scholar]
62. Butler JE, Rainard P, Lippolis J, Salmon H & Kacskovics I The Mammary Gland in Mucosal and Regional Immunity. Mucosal Immunology, 2269–2306 (2015). [Google Scholar]
63. Fisher MM, Nagy B, Bazin H & Underdown BJ Biliary transport of IgA: role of secretory component. Proc Natl Acad Sci U S A 76, 2008–2012 (1979). [Europe PMC free article] [Abstract] [Google Scholar]
64. Huang J, Guerrero A, Parker E, Strum JS, Smilowitz JT, German JB, et al. Site-specific glycosylation of secretory immunoglobulin A from human colostrum. J Proteome Res 14, 1335–1349 (2015). [Europe PMC free article] [Abstract] [Google Scholar]
65. Brandtzaeg P The mucosal immune system and its integration with the mammary glands. J Pediatr 156, S8–15 (2010). [Abstract] [Google Scholar]
66. Gopalakrishna KP, Macadangdang BR, Rogers MB, Tometich JT, Firek BA, Baker R, et al. Maternal IgA protects against the development of necrotizing enterocolitis in preterm infants. Nat Med 25, 1110–1115 (2019). [Europe PMC free article] [Abstract] [Google Scholar]
67. Atyeo C & Alter G The multifaceted roles of breast milk antibodies. Cell 184, 1486–1499 (2021). [Abstract] [Google Scholar]
68. Stadtmueller BM, Huey-Tubman KE, Lopez CJ, Yang Z, Hubbell WL & Bjorkman PJ The structure and dynamics of secretory component and its interactions with polymeric immunoglobulins. Elife 5(2016). [Europe PMC free article] [Abstract] [Google Scholar]
69. Kumar N, Arthur CP, Ciferri C & Matsumoto ML Structure of the secretory immunoglobulin A core. Science 367, 1008–1014 (2020). [Abstract] [Google Scholar]
70. Richards AF, Baranova DE, Pizzuto MS, Jaconi S, Willsey GG, Torres-Velez FJ, et al. Recombinant Human Secretory IgA Induces Salmonella Typhimurium Agglutination and Limits Bacterial Invasion into Gut-Associated Lymphoid Tissues. ACS Infect Dis 7, 1221–1235 (2021). [Europe PMC free article] [Abstract] [Google Scholar]
71. Binsker U, Lees JA, Hammond AJ & Weiser JN Immune exclusion by naturally acquired secretory IgA against pneumococcal pilus-1. J Clin Invest 130, 927–941 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
72. Stokes CR, Soothill JF & Turner MW Immune exclusion is a function of IgA. Nature 255, 745–746 (1975). [Abstract] [Google Scholar]
73. Boullier S, Tanguy M, Kadaoui KA, Caubet C, Sansonetti P, Corthesy B, et al. Secretory IgA-mediated neutralization of Shigella flexneri prevents intestinal tissue destruction by down-regulating inflammatory circuits. J Immunol 183, 5879–5885 (2009). [Abstract] [Google Scholar]
74. Cullender TC, Chassaing B, Janzon A, Kumar K, Muller CE, Werner JJ, et al. Innate and adaptive immunity interact to quench microbiome flagellar motility in the gut. Cell Host Microbe 14, 571–581 (2013). [Europe PMC free article] [Abstract] [Google Scholar]
75. Pickering LK, Morrow AL, Herrera I, O’Ryan M, Estes MK, Guilliams SE, et al. Effect of maternal rotavirus immunization on milk and serum antibody titers. J Infect Dis 172, 723–728 (1995). [Abstract] [Google Scholar]
76. Chen MY, Kirkwood CD, Bines J, Cowley D, Pavlic D, Lee KJ, et al. Rotavirus Specific Maternal Antibodies and Immune Response to RV3-BB Neonatal Rotavirus Vaccine in New Zealand. Hum Vaccin Immunother, 0 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
77. Hayani KC, Guerrero ML, Morrow AL, Gomez HF, Winsor DK, Ruiz-Palacios GM, et al. Concentration of milk secretory immunoglobulin A against Shigella virulence plasmid-associated antigens as a predictor of symptom status in Shigella-infected breast-fed infants. J Pediatr 121, 852–856 (1992). [Abstract] [Google Scholar]
78. Walterspiel JN, Morrow AL, Guerrero ML, Ruiz-Palacios GM & Pickering LK Secretory anti-Giardia lamblia antibodies in human milk: protective effect against diarrhea. Pediatrics 93, 28–31 (1994). [Abstract] [Google Scholar]
79. Noguera-Obenza M, Ochoa TJ, Gomez HF, Guerrero ML, Herrera-Insua I, Morrow AL, et al. Human milk secretory antibodies against attaching and effacing Escherichia coli antigens. Emerg Infect Dis 9, 545–551 (2003). [Europe PMC free article] [Abstract] [Google Scholar]
80. Korpe PS, Liu Y, Siddique A, Kabir M, Ralston K, Ma JZ, et al. Breast milk parasite-specific antibodies and protection from amebiasis and cryptosporidiosis in Bangladeshi infants: a prospective cohort study. Clin Infect Dis 56, 988–992 (2013). [Europe PMC free article] [Abstract] [Google Scholar]
81. Ruiz-Palacios GM, Calva JJ, Pickering LK, Lopez-Vidal Y, Volkow P, Pezzarossi H, et al. Protection of breast-fed infants against Campylobacter diarrhea by antibodies in human milk. J Pediatr 116, 707–713 (1990). [Abstract] [Google Scholar]
82. Maertens K, De Schutter S, Braeckman T, Baerts L, Van Damme P, De Meester I, et al. Breastfeeding after maternal immunisation during pregnancy: providing immunological protection to the newborn: a review. Vaccine 32, 1786–1792 (2014). [Abstract] [Google Scholar]
83. Schlaudecker EP, Steinhoff MC, Omer SB, McNeal MM, Roy E, Arifeen SE, et al. IgA and neutralizing antibodies to influenza a virus in human milk: a randomized trial of antenatal influenza immunization. PLoS One 8, e70867 (2013). [Europe PMC free article] [Abstract] [Google Scholar]
84. Dangor Z, Khan M, Kwatra G, Izu A, Nakwa F, Ramdin T, et al. The Association Between Breast Milk Group B Streptococcal Capsular Antibody Levels and Late-onset Disease in Young Infants. Clin Infect Dis 70, 1110–1114 (2020). [Abstract] [Google Scholar]
85. Young BE, Seppo AE, Diaz N, Rosen-Carole C, Nowak-Wegrzyn A, Cruz Vasquez JM, et al. Association of Human Milk Antibody Induction, Persistence, and Neutralizing Capacity With SARS-CoV-2 Infection vs mRNA Vaccination. JAMA Pediatr 176, 159–168 (2022). [Europe PMC free article] [Abstract] [Google Scholar]
86. Pace RM, Williams JE, Jarvinen KM, Belfort MB, Pace CDW, Lackey KA, et al. Characterization of SARS-CoV-2 RNA, Antibodies, and Neutralizing Capacity in Milk Produced by Women with COVID-19. mBio 12(2021). [Europe PMC free article] [Abstract] [Google Scholar]
87. Collier AY, McMahan K, Yu J, Tostanoski LH, Aguayo R, Ansel J, et al. Immunogenicity of COVID-19 mRNA Vaccines in Pregnant and Lactating Women. JAMA 325, 2370–2380 (2021). [Europe PMC free article] [Abstract] [Google Scholar]
88. Fox A, Marino J, Amanat F, Krammer F, Hahn-Holbrook J, Zolla-Pazner S, et al. Robust and Specific Secretory IgA Against SARS-CoV-2 Detected in Human Milk. iScience 23, 101735 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
89. Perl SH, Uzan-Yulzari A, Klainer H, Asiskovich L, Youngster M, Rinott E, et al. SARS-CoV-2-Specific Antibodies in Breast Milk After COVID-19 Vaccination of Breastfeeding Women. JAMA 325, 2013–2014 (2021). [Europe PMC free article] [Abstract] [Google Scholar]
90. Kazimbaya KM, Chisenga CC, Simuyandi M, Phiri CM, Laban NM, Bosomprah S, et al. In-vitro inhibitory effect of maternal breastmilk components on rotavirus vaccine replication and association with infant seroconversion to live oral rotavirus vaccine. PLoS One 15, e0240714 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
91. Fouda GG, Eudailey J, Kunz EL, Amos JD, Liebl BE, Himes J, et al. Systemic administration of an HIV-1 broadly neutralizing dimeric IgA yields mucosal secretory IgA and virus neutralization. Mucosal Immunol (2016). [Europe PMC free article] [Abstract] [Google Scholar]
92. Tay MZ, Kunz EL, Deal A, Zhang L, Seaton KE, Rountree W, et al. Rare Detection of Antiviral Functions of Polyclonal IgA Isolated from Plasma and Breast Milk Compartments in Women Chronically Infected with HIV-1. J Virol 93(2019). [Europe PMC free article] [Abstract] [Google Scholar]
93. Pollara J, McGuire E, Fouda GG, Rountree W, Eudailey J, Overman RG, et al. Association of HIV-1 Envelope-Specific Breast Milk IgA Responses with Reduced Risk of Postnatal Mother-to-Child Transmission of HIV-1. J Virol 89, 9952–9961 (2015). [Europe PMC free article] [Abstract] [Google Scholar]
94. Nelson CS, Pollara J, Kunz EL, Jeffries TL Jr., Duffy R, Beck C, et al. Combined HIV-1 Envelope Systemic and Mucosal Immunization of Lactating Rhesus Monkeys Induces a Robust Immunoglobulin A Isotype B Cell Response in Breast Milk. J Virol 90, 4951–4965 (2016). [Europe PMC free article] [Abstract] [Google Scholar]
95. Bunker JJ, Erickson SA, Flynn TM, Henry C, Koval JC, Meisel M, et al. Natural polyreactive IgA antibodies coat the intestinal microbiota. Science 358(2017). [Europe PMC free article] [Abstract] [Google Scholar]
96. Kabbert J, Benckert J, Rollenske T, Hitch TCA, Clavel T, Cerovic V, et al. High microbiota reactivity of adult human intestinal IgA requires somatic mutations. J Exp Med 217(2020). [Europe PMC free article] [Abstract] [Google Scholar]
97. Raskova Kafkova L, Brokesova D, Krupka M, Stehlikova Z, Dvorak J, Coufal S, et al. Secretory IgA N-glycans contribute to the protection against E. coli O55 infection of germ-free piglets. Mucosal Immunol 14, 511–522 (2021). [Europe PMC free article] [Abstract] [Google Scholar]
98. Mathias A & Corthésy B Recognition of gram-positive intestinal bacteria by hybridoma- and colostrum-derived secretory immunoglobulin A is mediated by carbohydrates. J Biol Chem 286, 17239–17247 (2011). [Europe PMC free article] [Abstract] [Google Scholar]
99. Joglekar P, Ding H, Canales-Herrerias P, Pasricha PJ, Sonnenburg JL & Peterson DA Intestinal IgA Regulates Expression of a Fructan Polysaccharide Utilization Locus in Colonizing Gut Commensal Bacteroides thetaiotaomicron. mBio 10(2019). [Europe PMC free article] [Abstract] [Google Scholar]
100. Arnold JN, Wormald MR, Sim RB, Rudd PM & Dwek RA The impact of glycosylation on the biological function and structure of human immunoglobulins. Annu Rev Immunol 25, 21–50 (2007). [Abstract] [Google Scholar]
101. Huus KE, Petersen C & Finlay BB Diversity and dynamism of IgA-microbiota interactions. Nat Rev Immunol 21, 514–525 (2021). [Abstract] [Google Scholar]
102. Bollinger RR, Everett ML, Palestrant D, Love SD, Lin SS & Parker W Human secretory immunoglobulin A may contribute to biofilm formation in the gut. Immunology 109, 580–587 (2003). [Abstract] [Google Scholar]
103. Moor K, Diard M, Sellin ME, Felmy B, Wotzka SY, Toska A, et al. High-avidity IgA protects the intestine by enchaining growing bacteria. Nature 544, 498–502 (2017). [Abstract] [Google Scholar]
104. Nakajima A, Vogelzang A, Maruya M, Miyajima M, Murata M, Son A, et al. IgA regulates the composition and metabolic function of gut microbiota by promoting symbiosis between bacteria. J Exp Med 215, 2019–2034 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
105. Donaldson GP, Ladinsky MS, Yu KB, Sanders JG, Yoo BB, Chou WC, et al. Gut microbiota utilize immunoglobulin A for mucosal colonization. Science 360, 795–800 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
106. Rogier EW, Frantz AL, Bruno ME & Kaetzel CS Secretory IgA is Concentrated in the Outer Layer of Colonic Mucus along with Gut Bacteria. Pathogens 3, 390–403 (2014). [Europe PMC free article] [Abstract] [Google Scholar]
107. Karav S, Le Parc A, Leite Nobrega de Moura Bell JM, Frese SA, Kirmiz N, Block DE, et al. Oligosaccharides Released from Milk Glycoproteins Are Selective Growth Substrates for Infant-Associated Bifidobacteria. Appl Environ Microbiol 82, 3622–3630 (2016). [Europe PMC free article] [Abstract] [Google Scholar]
108. Barratt MJ, Nuzhat S, Ahsan K, Frese SA, Arzamasov AA, Sarker SA, et al. Bifidobacterium infantis treatment promotes weight gain in Bangladeshi infants with severe acute malnutrition. Sci Transl Med 14, eabk1107 (2022). [Europe PMC free article] [Abstract] [Google Scholar]
109. Haworth JC & Dilling L Concentration of gamma-A-globulin in serum, saliva, and nasopharyngeal secretions of infants and children. J Lab Clin Med 67, 922–933 (1966). [Abstract] [Google Scholar]
110. Rognum TO, Thrane S, Stoltenberg L, Vege A & Brandtzaeg P Development of intestinal mucosal immunity in fetal life and the first postnatal months. Pediatr Res 32, 145–149 (1992). [Abstract] [Google Scholar]
111. Gleeson M, Cripps AW, Clancy RL, Hensley MJ, Dobson AJ & Firman DW Breast feeding conditions a differential developmental pattern of mucosal immunity. Clin Exp Immunol 66, 216–222 (1986). [Abstract] [Google Scholar]
112. Burgio GR, Lanzavecchia A, Plebani A, Jayakar S & Ugazio AG Ontogeny of secretory immunity: levels of secretory IgA and natural antibodies in saliva. Pediatr Res 14, 1111–1114 (1980). [Abstract] [Google Scholar]
113. Gahnberg L, Smith DJ, Taubman MA & Ebersole JL Salivary-IgA antibody to glucosyltransferase of oral microbial origin in children. Arch Oral Biol 30, 551–556 (1985). [Abstract] [Google Scholar]
114. Koutras AK & Vigorita VJ Fecal secretory immunoglobulin A in breast milk versus formula feeding in early infancy. J Pediatr Gastroenterol Nutr 9, 58–61 (1989). [Abstract] [Google Scholar]
115. Bridgman SL, Konya T, Azad MB, Guttman DS, Sears MR, Becker AB, et al. High fecal IgA is associated with reduced Clostridium difficile colonization in infants. Microbes Infect 18, 543–549 (2016). [Abstract] [Google Scholar]
116. Ramanan D, Sefik E, Galván-Peña S, Wu M, Yang L, Yang Z, et al. An Immunologic Mode of Multigenerational Transmission Governs a Gut Treg Setpoint. Cell 181, 1276–1290.e1213 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
117. von Mutius E & Schmid S The PASTURE project: EU support for the improvement of knowledge about risk factors and preventive factors for atopy in Europe. Allergy 61, 407–413 (2006). [Abstract] [Google Scholar]
118. Orivuori L, Loss G, Roduit C, Dalphin JC, Depner M, Genuneit J, et al. Soluble immunoglobulin A in breast milk is inversely associated with atopic dermatitis at early age: the PASTURE cohort study. Clin Exp Allergy 44, 102–112 (2014). [Abstract] [Google Scholar]
119. Järvinen KM, Laine ST, Järvenpää AL & Suomalainen HK Does low IgA in human milk predispose the infant to development of cow’s milk allergy? Pediatr Res 48, 457–462 (2000). [Abstract] [Google Scholar]
120. Jarvinen KM, Westfall JE, Seppo MS, James AK, Tsuang AJ, Feustel PJ, et al. Role of maternal elimination diets and human milk IgA in the development of cow’s milk allergy in the infants. Clin Exp Allergy 44, 69–78 (2014). [Europe PMC free article] [Abstract] [Google Scholar]
121. Holm M, Saraswat M, Joenväárä S, Seppo A, Looney RJ, Tohmola T, et al. Quantitative glycoproteomics of human milk and association with atopic disease. PLoS One 17, e0267967 (2022). [Europe PMC free article] [Abstract] [Google Scholar]
122. Planer JD, Peng Y, Kau AL, Blanton LV, Ndao IM, Tarr PI, et al. Development of the gut microbiota and mucosal IgA responses in twins and gnotobiotic mice. Nature 534, 263–266 (2016). [Europe PMC free article] [Abstract] [Google Scholar]
123. Koch MA, Reiner GL, Lugo KA, Kreuk LS, Stanbery AG, Ansaldo E, et al. Maternal IgG and IgA Antibodies Dampen Mucosal T Helper Cell Responses in Early Life. Cell 165, 827–841 (2016). [Europe PMC free article] [Abstract] [Google Scholar]
124. Sterlin D, Fadlallah J, Adams O, Fieschi C, Parizot C, Dorgham K, et al. Human IgA binds a diverse array of commensal bacteria. J Exp Med 217(2020). [Europe PMC free article] [Abstract] [Google Scholar]
125. Ruiz L, García-Carral C & Rodriguez JM Unfolding the Human Milk Microbiome Landscape in the Omics Era. Frontiers in Microbiology 10(2019). [Europe PMC free article] [Abstract] [Google Scholar]
126. Cabrera-Rubio R, Collado MC, Laitinen K, Salminen S, Isolauri E & Mira A The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery. Am J Clin Nutr 96, 544–551 (2012). [Abstract] [Google Scholar]
127. Williams JE, Carrothers JM, Lackey KA, Beatty NF, York MA, Brooker SL, et al. Human Milk Microbial Community Structure Is Relatively Stable and Related to Variations in Macronutrient and Micronutrient Intakes in Healthy Lactating Women. J Nutr 147, 1739–1748 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
128. Moossavi S, Sepehri S, Robertson B, Bode L, Goruk S, Field CJ, et al. Composition and Variation of the Human Milk Microbiota Are Influenced by Maternal and Early-Life Factors. Cell Host Microbe 25, 324–335 e324 (2019). [Abstract] [Google Scholar]
129. Demmelmair H, Jimenez E, Collado MC, Salminen S & McGuire MK Maternal and Perinatal Factors Associated with the Human Milk Microbiome. Curr Dev Nutr 4, nzaa027 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
130. Fernandez L, Langa S, Martin V, Maldonado A, Jimenez E, Martin R, et al. The human milk microbiota: origin and potential roles in health and disease. Pharmacol Res 69, 1–10 (2013). [Abstract] [Google Scholar]
131. Davis EC, Wang M & Donovan SM Microbial Interrelationships across Sites of Breastfeeding Mothers and Infants at 6 Weeks Postpartum. Microorganisms 10, 1155 (2022). [Europe PMC free article] [Abstract] [Google Scholar]
132. Williams JE, Carrothers JM, Lackey KA, Beatty NF, Brooker SL, Peterson HK, et al. Strong Multivariate Relations Exist Among Milk, Oral, and Fecal Microbiomes in Mother-Infant Dyads During the First Six Months Postpartum. J Nutr 149, 902–914 (2019). [Europe PMC free article] [Abstract] [Google Scholar]
133. Biagi E, Quercia S, Aceti A, Beghetti I, Rampelli S, Turroni S, et al. The Bacterial Ecosystem of Mother’s Milk and Infant’s Mouth and Gut. Frontiers in Microbiology 8(2017). [Europe PMC free article] [Abstract] [Google Scholar]
134. Ramsay DT, Kent JC, Owens RA & Hartmann PE Ultrasound imaging of milk ejection in the breast of lactating women. Pediatrics 113, 361–367 (2004). [Abstract] [Google Scholar]
135. Jost T, Lacroix C, Braegger CP, Rochat F & Chassard C Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding. Environ Microbiol 16, 2891–2904 (2014). [Abstract] [Google Scholar]
136. Martin R, Langa S, Reviriego C, Jiminez E, Marin ML, Xaus J, et al. Human milk is a source of lactic acid bacteria for the infant gut. J Pediatr 143, 754–758 (2003). [Abstract] [Google Scholar]
137. Rodríguez JM The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation? Adv Nutr 5, 779–784 (2014). [Europe PMC free article] [Abstract] [Google Scholar]
138. Perez PF, Dore J, Leclerc M, Levenez F, Benyacoub J, Serrant P, et al. Bacterial imprinting of the neonatal immune system: lessons from maternal cells? Pediatrics 119, e724–732 (2007). [Abstract] [Google Scholar]
139. Treven P, Mrak V, Bogovic Matijasic B, Horvat S & Rogelj I Administration of probiotics Lactobacillus rhamnosus GG and Lactobacillus gasseri K7 during pregnancy and lactation changes mouse mesenteric lymph nodes and mammary gland microbiota. J Dairy Sci 98, 2114–2128 (2015). [Abstract] [Google Scholar]
140. Arroyo R, Martín V, Maldonado A, Jiménez E, Fernández L & Rodríguez JM Treatment of infectious mastitis during lactation: antibiotics versus oral administration of Lactobacilli isolated from breast milk. Clin Infect Dis 50, 1551–1558 (2010). [Abstract] [Google Scholar]
141. Jiménez E, Fernández L, Maldonado A, Martín R, Olivares M, Xaus J, et al. Oral administration of Lactobacillus strains isolated from breast milk as an alternative for the treatment of infectious mastitis during lactation. Appl Environ Microbiol 74, 4650–4655 (2008). [Europe PMC free article] [Abstract] [Google Scholar]
142. Boix-Amoros A, Collado MC & Mira A Relationship between Milk Microbiota, Bacterial Load, Macronutrients, and Human Cells during Lactation. Front Microbiol 7, 492 (2016). [Europe PMC free article] [Abstract] [Google Scholar]
143. Hunt KM, Foster JA, Forney LJ, Schutte UM, Beck DL, Abdo Z, et al. Characterization of the diversity and temporal stability of bacterial communities in human milk. PLoS One 6, e21313 (2011). [Europe PMC free article] [Abstract] [Google Scholar]
144. Murphy K, Curley D, O’Callaghan TF, O’Shea CA, Dempsey EM, O’Toole PW, et al. The Composition of Human Milk and Infant Faecal Microbiota Over the First Three Months of Life: A Pilot Study. Sci Rep 7, 40597 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
145. Grönlund MM, Gueimonde M, Laitinen K, Kociubinski G, Grönroos T, Salminen S, et al. Maternal breast-milk and intestinal bifidobacteria guide the compositional development of the Bifidobacterium microbiota in infants at risk of allergic disease. Clin Exp Allergy 37, 1764–1772 (2007). [Abstract] [Google Scholar]
146. Sprockett D, Fukami T & Relman DA Role of priority effects in the early-life assembly of the gut microbiota. Nat Rev Gastroenterol Hepatol 15, 197–205 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
147. Stinson LF, Trevenen ML & Geddes DT The Viable Microbiome of Human Milk Differs from the Metataxonomic Profile. Nutrients 13(2021). [Europe PMC free article] [Abstract] [Google Scholar]
148. Donald K, Petersen C, Turvey SE, Finlay BB & Azad MB Secretory IgA: Linking microbes, maternal health, and infant health through human milk. Cell Host & Microbe 30, 650–659 (2022). [Abstract] [Google Scholar]
149. Dzidic M, Mira A, Artacho A, Abrahamsson TR, Jenmalm MC & Collado MC Allergy development is associated with consumption of breastmilk with a reduced microbial richness in the first month of life. Pediatr Allergy Immunol 31, 250–257 (2020). [Abstract] [Google Scholar]
150. Orndorff PE, Devapali A, Palestrant S, Wyse A, Everett ML, Bollinger RR, et al. Immunoglobulin-Mediated Agglutination of and Biofilm Formation by <i>Escherichia coli</i> K-12 Require the Type 1 Pilus Fiber. Infection and Immunity 72, 1929–1938 (2004). [Europe PMC free article] [Abstract] [Google Scholar]
151. Pannaraj PS, Ly M, Cerini C, Saavedra M, Aldrovandi GM, Saboory AA, et al. Shared and Distinct Features of Human Milk and Infant Stool Viromes. Front Microbiol 9, 1162 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
152. Milani C, Casey E, Lugli GA, Moore R, Kaczorowska J, Feehily C, et al. Tracing mother-infant transmission of bacteriophages by means of a novel analytical tool for shotgun metagenomic datasets: METAnnotatorX. Microbiome 6, 145 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
153. Fujimura KE, Sitarik AR, Havstad S, Lin DL, Levan S, Fadrosh D, et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat Med 22, 1187–1191 (2016). [Europe PMC free article] [Abstract] [Google Scholar]
154. Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L & Jenmalm MC Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin Immunol 129, 434–440, 440.e431-432 (2012). [Abstract] [Google Scholar]
155. Azad MB, Konya T, Guttman DS, Field CJ, Sears MR, HayGlass KT, et al. Infant gut microbiota and food sensitization: associations in the first year of life. Clin Exp Allergy 45, 632–643 (2015). [Abstract] [Google Scholar]
156. Savage JH, Lee-Sarwar KA, Sordillo J, Bunyavanich S, Zhou Y, O’Connor G, et al. A prospective microbiome-wide association study of food sensitization and food allergy in early childhood. Allergy 73, 145–152 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
157. Arrieta MC, Stiemsma LT, Dimitriu PA, Thorson L, Russell S, Yurist-Doutsch S, et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci Transl Med 7, 307ra152 (2015). [Abstract] [Google Scholar]
158. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337–341 (2011). [Europe PMC free article] [Abstract] [Google Scholar]
159. Abdel-Gadir A, Stephen-Victor E, Gerber GK, Noval Rivas M, Wang S, Harb H, et al. Microbiota therapy acts via a regulatory T cell MyD88/RORgammat pathway to suppress food allergy. Nat Med 25, 1164–1174 (2019). [Europe PMC free article] [Abstract] [Google Scholar]
160. Stefka AT, Feehley T, Tripathi P, Qiu J, McCoy K, Mazmanian SK, et al. Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci U S A 111, 13145–13150 (2014). [Europe PMC free article] [Abstract] [Google Scholar]
161. Bunyavanich S, Shen N, Grishin A, Wood R, Burks W, Dawson P, et al. Early-life gut microbiome composition and milk allergy resolution. J Allergy Clin Immunol 138, 1122–1130 (2016). [Europe PMC free article] [Abstract] [Google Scholar]
162. Kalliomäki M, Kirjavainen P, Eerola E, Kero P, Salminen S & Isolauri E Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy Clin Immunol 107, 129–134 (2001). [Abstract] [Google Scholar]
163. Dong P, Feng JJ, Yan DY, Lyu YJ & Xu X Early-life gut microbiome and cow’s milk allergy- a prospective case - control 6-month follow-up study. Saudi J Biol Sci 25, 875–880 (2018). [Europe PMC free article] [Abstract] [Google Scholar]
164. Depner M, Taft DH, Kirjavainen PV, Kalanetra KM, Karvonen AM, Peschel S, et al. Maturation of the gut microbiome during the first year of life contributes to the protective farm effect on childhood asthma. Nat Med 26, 1766–1775 (2020). [Abstract] [Google Scholar]
165. Petersen C, Dai DLY, Boutin RCT, Sbihi H, Sears MR, Moraes TJ, et al. A rich meconium metabolome in human infants is associated with early-life gut microbiota composition and reduced allergic sensitization. Cell Rep Med 2, 100260 (2021). [Europe PMC free article] [Abstract] [Google Scholar]
166. Ta LDH, Chan JCY, Yap GC, Purbojati RW, Drautz-Moses DI, Koh YM, et al. A compromised developmental trajectory of the infant gut microbiome and metabolome in atopic eczema. Gut Microbes 12, 1–22 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
167. Vatanen T, Kostic AD, d’Hennezel E, Siljander H, Franzosa EA, Yassour M, et al. Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell 165, 1551 (2016). [Abstract] [Google Scholar]
168. Yu ZT, Chen C & Newburg DS Utilization of major fucosylated and sialylated human milk oligosaccharides by isolated human gut microbes. Glycobiology 23, 1281–1292 (2013). [Europe PMC free article] [Abstract] [Google Scholar]
169. Rios-Covian D, Salazar N, Gueimonde M & de Los Reyes-Gavilan CG Shaping the Metabolism of Intestinal Bacteroides Population through Diet to Improve Human Health. Front Microbiol 8, 376 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
170. Mazmanian SK, Liu CH, Tzianabos AO & Kasper DL An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122, 107–118 (2005). [Abstract] [Google Scholar]
171. Roduit C, Frei R, Ferstl R, Loeliger S, Westermann P, Rhyner C, et al. High levels of butyrate and propionate in early life are associated with protection against atopy. Allergy 74, 799–809 (2019). [Abstract] [Google Scholar]
172. Cait A, Cardenas E, Dimitriu PA, Amenyogbe N, Dai D, Cait J, et al. Reduced genetic potential for butyrate fermentation in the gut microbiome of infants who develop allergic sensitization. J Allergy Clin Immunol 144, 1638–1647.e1633 (2019). [Abstract] [Google Scholar]
173. 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 141, 1334–1342.e1335 (2018). [Abstract] [Google Scholar]
174. Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 17, 662–671 (2015). [Europe PMC free article] [Abstract] [Google Scholar]
175. Correa-Oliveira R, Fachi JL, Vieira A, Sato FT & Vinolo MA Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunology 5, e73 (2016). [Europe PMC free article] [Abstract] [Google Scholar]
176. Peng L, Li ZR, Green RS, Holzman IR & Lin J Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J Nutr 139, 1619–1625 (2009). [Europe PMC free article] [Abstract] [Google Scholar]
177. Brand S, Teich R, Dicke T, Harb H, Yildirim AO, Tost J, et al. Epigenetic regulation in murine offspring as a novel mechanism for transmaternal asthma protection induced by microbes. J Allergy Clin Immunol 128, 618–625 e611-617 (2011). [Abstract] [Google Scholar]
178. Thorburn AN, McKenzie CI, Shen S, Stanley D, Macia L, Mason LJ, et al. Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat Commun 6, 7320 (2015). [Abstract] [Google Scholar]
179. Licciardi PV, Ververis K & Karagiannis TC Histone deacetylase inhibition and dietary short-chain Fatty acids. ISRN Allergy 2011, 869647 (2011). [Europe PMC free article] [Abstract] [Google Scholar]
180. Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson J, et al. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol 8, 80–93 (2015). [Europe PMC free article] [Abstract] [Google Scholar]
181. Yang W, Yu T, Huang X, Bilotta AJ, Xu L, Lu Y, et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat Commun 11, 4457 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
182. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504, 451–455 (2013). [Europe PMC free article] [Abstract] [Google Scholar]
183. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446–450 (2013). [Abstract] [Google Scholar]
184. Greer FR, Sicherer SH & Burks AW The Effects of Early Nutritional Interventions on the Development of Atopic Disease in Infants and Children: The Role of Maternal Dietary Restriction, Breastfeeding, Hydrolyzed Formulas, and Timing of Introduction of Allergenic Complementary Foods. Pediatrics 143(2019). [Abstract] [Google Scholar]
185. Seppo AE, Choudhury R, Pizzarello C, Palli R, Fridy S, Rajani PS, et al. Traditional Farming Lifestyle in Old Older Mennonites Modulates Human Milk Composition. Front Immunol 12, 741513 (2021). [Europe PMC free article] [Abstract] [Google Scholar]
186. Ruiz L, Espinosa-Martos I, Garcia-Carral C, Manzano S, McGuire MK, Meehan CL, et al. What’s Normal? Immune Profiling of Human Milk from Healthy Women Living in Different Geographical and Socioeconomic Settings. Front Immunol 8, 696 (2017). [Europe PMC free article] [Abstract] [Google Scholar]
187. Gungor D, Nadaud P, LaPergola CC, Dreibelbis C, Wong YP, Terry N, et al. Infant milk-feeding practices and food allergies, allergic rhinitis, atopic dermatitis, and asthma throughout the life span: a systematic review. Am J Clin Nutr 109, 772S–799S (2019). [Europe PMC free article] [Abstract] [Google Scholar]
188. Liang G, Zhao C, Zhang H, Mattei L, Sherrill-Mix S, Bittinger K, et al. The stepwise assembly of the neonatal virome is modulated by breastfeeding. Nature 581, 470–474 (2020). [Europe PMC free article] [Abstract] [Google Scholar]
189. Yang Y & Palm NW Immunoglobulin A and the microbiome. Curr Opin Microbiol 56, 89–96 (2020). [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/135397877
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/135397877

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1016/j.jaci.2022.07.014

Supporting
Mentioning
Contrasting
0
18
0

Article citations


Go to all (21) article citations

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

Bill and Melinda Gates Foundation

    Eunice Kennedy Shriver National Institute of Child Health and Human Development

      Janssen Research and Development

        NIAID NIH HHS (2)

        NICHD NIH HHS (1)

        National Institute of Allergy and Infectious Diseases (1)

        National Institutes of Health (2)

        U.S. Department of Agriculture (1)