7,324
Views
12
CrossRef citations to date
0
Altmetric
Review

Comparing early life nutritional sources and human milk feeding practices: personalized and dynamic nutrition supports infant gut microbiome development and immune system maturation

, & ORCID Icon
Article: 2190305 | Received 19 Sep 2022, Accepted 06 Mar 2023, Published online: 13 Apr 2023

ABSTRACT

Exclusive breastfeeding is recommended for the first six months of life, but many infants receive pumped milk, formula, donor human milk, or other nutritional sources during this critical period. Substantive evidence shows early nutrition influences development of the microbiome and immune system, affecting lifelong health. However, the underlying mechanisms are unclear and the nuances of human milk feeding are rarely considered. This review synthesizes evidence from human studies and model systems to discuss the impact of different nutritional sources on co-development of the gut microbiome, antigen tolerance, and immunity. We highlight two key mechanisms: epigenetics and the so-called “weaning reaction”. Collectively, this evidence highlights i) the fundamental role of parents’ own milk, fed directly at the breast, as a dynamic and personalized nutrition source that drives developmental programming, and ii) the deficiencies of alternative nutritional sources and priority research areas for improving these alternatives when direct breastfeeding is not possible.

Plain Language Summary

Before they begin eating solid foods, infants may be fed a variety of nutritional sources such as breast milk (“parent’s own milk”, fed directly at the breast or pumped and fed from a bottle), commercial infant formula, or sterilized “donor human milk” from a certified milk bank. Early nutrition affects the infant’s gut microbiome (bacteria living in the intestinal tract), development of their immune system, and their health throughout life. However, it is unclear how different forms of early nutrition affect these important processes. Parent’s own milk contains compounds that support gut microbes and stimulate development of the infant immune system, changing over time to meet infant needs. For many of these compounds, donor human milk contains a lesser amount, and formula contains even less – or none at all. Some compounds are also affected by pumping and storing parents’ own milk. This review highlights how differences among these nutritional sources influence gene expression and gut development to shape the infant microbiome and immune system. Current evidence shows that parent’s own milk, fed at the breast, offers unique benefits that are not replicated by other forms of early nutrition. This review also outlines how early life nutrition research can help us understand human development and develop new ways to provide the best possible start to life for all infants.

Introduction

Immune system structures and functions are immature, or differentially adapted, at birth.Citation1,Citation2 Compared to adults, neonatal neutrophils, monocytes, and dendritic cells have reduced functional capacity to prevent and clear pathogenic infections.Citation3–5 Furthermore, intestinal barriers have increased permeability for up to 6 months, allowing ingested macromolecules, including immunoglobulins (Igs), to translocate across the immature intestinal epithelium.Citation2,Citation6 Activated and functional T and B cells are detected around 12 weeks after birth, but at this stage they are more likely to induce tolerance for an antigen rather than clearing it from the infant system.Citation7–9 In parallel, the infant microbiome rapidly matures in the first few weeks and months of life. Gut microbiota play a key role in shaping the immune system by protecting the infant from harmful pathogens, facilitating mucosal immune structure stimulation, and inducing antigen tolerance.Citation10

During this critical period of early development, infant nutrition influences developmental processes that drive the maturation of the gut microbiome and infant immune system toward the composition and capabilities of a healthy adult.Citation11–13 Deciphering how different sources of early nutrition – namely, human milk in various forms, and human milk substitutes of various formulations – influence gut microbiome development and immune system maturation is essential to understanding how to provide the best start to life for all infants.

Gut microbiome and immune development

Gut microbiome composition varies throughout life and has been implicated in the pathophysiology of many health conditions including inflammatory bowel disease (IBD), colorectal cancer, obesity, diabetes, and neurological disorders.Citation14,Citation15 Early life gut colonization is a critical process that has been associated with lifelong gut microbiome composition.Citation11 The gut is initially colonized at birth and evolves in terms of diversity and abundance of microbes over time. This process is affected by factors such as birth mode, diet (nutrition and feeding practices), presence of siblings and household pets, antibiotic exposure, and geographical location.Citation1,Citation10 Many studies have consistently shown that human milk feeding (i.e. breastfeeding) is the strongest predictor of gut microbiota composition in the first months of lifeCitation16–20 – although very few have distinguished among different forms of human milk feeding.

Gut microbiota confer many helpful and protective immune functions to their host throughout life. Commensal microbes help prevent pathogenic gut colonization and intestinal barrier breaches by competitively excluding pathogens and stimulating epithelial production of antimicrobial peptides.Citation21 Furthermore, commensal gut microbes in early life (e.g. Bifidobacterium infantis, Bifidobacterium bifidum, Lactobacillus rhamnosus) can enhance gut barrier development by strengthening tight junctions in intestinal epithelial cells.Citation22,Citation23 Other common gut microbes, such as Bacteroides thetaiotaomicron and Lactobacillus reuteri, dampen host inflammatory responses by inhibiting nuclear factor-κB (NF-κB) activation.Citation24 This promotes immune system homeostasis throughout life by regulating pro- and anti-inflammatory immune system responses.Citation25 Furthermore, gut microbial antigen – immune system interactions in early life are critical for infant antigen tolerance development.Citation26 Defects in tolerance induction have been associated with the development of type 1 diabetes, allergy, IBD, and cancer.Citation26,Citation27

Certain gut microbes (e.g. some Bifidobacterium and Bacteroides species) can metabolize dietary oligosaccharides that are non-digestible to the host, including human milk oligosaccharides (HMOs). Microbial products generated from this metabolism (e.g. short-chain fatty acids, acetate, propionate, butyrate, folate, amino acids, vitamins, hydrogen, and carbon dioxide) have nutritional value to the host and can also be used as substrates for enzymes including acetylases, methylases, and glucuronidases.Citation24,Citation28,Citation29 These metabolites have also been associated with antigen tolerance regulation, intestinal barrier maturation, and epigenetic modifications within intestinal cells and peripheral tissues – processes that, when disrupted, are associated with the development of pathologies including IBD, obesity, and diabetes.Citation29,Citation30

During early life, irregular colonization or antibiotic-induced dysregulation of the microbiome can bias the infant immune system to a hypersensitive or allergic state.Citation31 The absence of adequate colonization in early life can lead to an increased likelihood of developing immune-mediated diseases, such as allergy, autoimmune conditions, and type 1 diabetes.Citation31 Thus, understanding how early life nutrition influences the gut microbiome is crucial to developing methods for correcting microbiome “dysbiosis” and preventing immune-mediated diseases.

Forms of early life nutrition

The most common forms of infant nutrition are the birthing parent’s own milk (POM, also known as “mother’s own milk”) and commercial infant formula, typically made from cow’s milk. POM can be fed directly at the breast (DPOM), or expressed and fed from a bottle (EPOM). A less common but increasingly available POM alternative is donor human milk (DHM), which is received, pasteurized, and distributed by a human milk bank.Citation32 Informal milk sharing through unregulated networks is another increasingly common practice, but is not discussed in this review.Citation32 These nutritional sources vary somewhat in their nutrient profiles and greatly in their dynamism and content of microbial and immunomodulatory factors.Citation33

Parents’ own milk (POM) – direct or expressed

POM is the ‘gold standard’ for infant nutrition. Exclusive POM feeding is associated with a lower incidence of respiratory infections in infancy and childhood, and healthier growth profiles, compared to formula feeding.Citation34 A complex and dynamic biological system,Citation35,Citation36 POM contains protective and immunomodulatory components, adapts dynamically to infant needs and contains sufficient macro and micronutrients (excluding Vitamin K and D) to fully support healthy infant development for the first six months of life (, ).Citation9,Citation33 For about five days after delivery, lactating parents produce colostrum, an early form of POM that is low in fat, high in protein, and rich in immunomodulatory components (e.g. bioactive enzymes and immune cells) ().Citation33,Citation37 POM then transitions to a higher fat, lower protein mature milk (>14 days after birth) that generally contains lower levels of immunomodulatory components.Citation37 Most immunomodulatory component concentrations within POM continue to gradually decrease until the cessation of POM feeding.Citation9,Citation33 These dynamic changes allow the developing infant immune system to gain more “responsibility” and independence in managing and mounting immune responses, setting up healthy lifelong immune function.

Figure 1. Associations between human milk components and physiological processes that influence infant immune development: epigenetics, microbiome, and gut integrity.

Figure 1. Associations between human milk components and physiological processes that influence infant immune development: epigenetics, microbiome, and gut integrity.

Table 1. Milk components associated with infant immune system and/or gut microbiome development, and their relative concentrations in different nutritional sources: parents own milk (POM, distinguishing between colostrum and mature milk), frozen expressed POM (EPOM), donor human milk (DHM), and commercial infant formula.

POM also contains live microbes that may influence the infant gut microbiome composition.Citation74 The composition of POM and its microbiota depend on many dynamic factors, including method of feeding (e.g. directly from the breast/chest, or pumped/expressed and bottled), lactation stage, lactating parent BMI, mode of delivery, infant sex, and geographical location.Citation75,Citation76

Interestingly, the level of macrophages and TNF-α within DPOM have been shown to increase when nursing infants are mounting an immune response against an infection, even when the lactating parent is asymptomatic, indicating that DPOM can adapt to the immunological needs of the infant.Citation38 The authors hypothesize that this dynamic ability is facilitated by a bi-directional exchange of immune factors that occurs through direct suckling at the breast.Citation38 EPOM does not appear to adapt in this manner, presumably because it is collected without direct infant contact and/or due to storage practices that may change its bioactive profile.Citation38,Citation77

Donor human milk (DHM)

DHM is considered the next best option when POM is unavailable, particularly for vulnerable neonates at high risk of severe morbidity and mortality. The World Health Organization recommends DHM for low- and very-low-birthweight infants, as well as small and sick neonates that cannot receive DPOM.Citation78 The eligibility for infants to receive DHM varies regionally and among health care institutions, and the guidelines and policies for the quality, safety, and ethical standards of DHM vary between countries.Citation79 There are an estimated 756 milk banks in 66 countries that manage the collection, processing, and distribution of DHM globally.

Although methodologies vary, all certified human milk banks process donated milk to destroy non-spore forming microbes.Citation41 The Holder pasteurization process (62.5°C for 30 min) is commonly used to eliminate potential pathogens from donated milk; however, this unfortunately eliminates beneficial milk microbes and immune cells, and lowers the activity of many signaling proteins and bioactive enzymes, such as lactoferrin, lipase and amylase (). Despite these modifications, pasteurized DHM contains viable forms of many heat-stable immunomodulatory components present in POM, such as Igs and HMOs.Citation39,Citation59 Like POM, DHM composition and nutrient content is influenced by donor factors (e.g. lactation stage, diet, age, ethnicity).Citation33 Improving DHM processing technology to maximize the preservation of immunomodulatory functions is an active area of research.Citation80

Infant formula

Commercial infant formula, typically made from cow’s milk, is an option for parents that cannot readily provide POM. While composition varies to some extent between manufacturers, all contain similar nutrient profiles because infant formulas are tightly regulated compared to other food products, although regulations vary among countries.Citation81,Citation82 Currently, these regulations do not require the addition of immunomodulatory components (e.g. signaling proteins, bioactive enzymes, immunoglobulins, oligosaccharides, probiotic microbes, postbiotic microbial products)Citation82, reflecting the uncertainty regarding the benefits, risks and feasibility of including such additives. Some immunomodulatory components (e.g. oligosaccharides, lactoferrin, and microRNAs) are present or added to some formulas; however, their structures are not identical to the human varieties, and their levels and bioactivity are impacted by the many steps involved in processing cow’s milk to generate infant formula (i.e. pasteurization, homogenization, fractionation, heat treatment, mixing, and emulsification).Citation56,Citation60,Citation70,Citation83,Citation84

In addition to its compositional differences from human milk, formula does not have the dynamic ability of POM, so it cannot be optimally matched to the evolving stages of infant development. Reflecting these deficits compared to POM, formula feeding is associated with an increased risk of developing necrotizing enterocolitis (NEC), respiratory infections, asthma, obesity, diabetes, and IBD compared to exclusive POM feeding.Citation33,Citation60 Thus, improving infant formula and investigating the short and long-term effects of incorporating human milk components is an important field of infant nutrition research,Citation60 alongside supporting, protecting, and promoting POM feeding as the ideal scenario.Citation85

Review aim

This review discusses differences in early life nutritional sources – including POM (DPOM and EPOM), DHM, and commercial formula – and how they impact gut microbiome development and infant immune system maturation. We also comment on the impacts of these nutritional sources and gut microbiota on the weaning reaction and early life epigenetics.

Compositional differences among different early life nutrition sources that influence microbiome and immune development

POM, DHM, and commercial formula all contain the essential macro and micronutrients required for healthy infant development and growth; however, only the POM nutrient profile changes over time as the infant ages. Non-nutrient components including immune cells, immunoglobulins, signaling proteins, and bioactive enzymes are provided through POM (), variably present (often at relatively lower levels) in DHM, and mostly absent from formula (). These differences are important to elucidate because these bioactive components are known to influence infant gut microbiome and immune system development,Citation57,Citation86 as discussed below.

Figure 2. Suggested mechanisms of action and interactions among parents’ own milk (POM) components, infant gut microbiota, and the infant immune system. Adapted from “intestinal immune system (small intestine)”, by Biorender.Com (2023). Retrieved from https://app.biorender.com/biorender-templates.

Figure 2. Suggested mechanisms of action and interactions among parents’ own milk (POM) components, infant gut microbiota, and the infant immune system. Adapted from “intestinal immune system (small intestine)”, by Biorender.Com (2023). Retrieved from https://app.biorender.com/biorender-templates.

Immune cells

Immune cells (also known as leukocytes) are uniquely present in POM and predominantly consist of dendritic cells, monocytes, macrophages, neutrophils, innate lymphoid cells, and lymphocytes (T cells and B cells) ().Citation87 Their levels follow a predictable pattern after delivery, with concentrations and effector functions of immune cells being highest in colostrum, and gradually decreasing as milk matures.Citation9,Citation37 The high buffering capacity of human milk protects these cells from infant saliva-mediated enzymatic digestion and enables them to survive in the infant digestive tract.Citation87 Once they reach the infant gut, POM immune cells can infiltrate the permeable neonatal intestinal barrier, allowing them to become activated and motile in the infant.Citation40 For example, POM cytotoxic T cells and plasma B cells elicit cytotoxic effects and secrete IgG in the infant, respectively, which may compensate for the decreased activity of immature infant immune cells.Citation88,Citation89 Additionally, POM macrophages are phagocytic, secrete cytolytic and inflammatory mediators, and express activation markers (e.g. CD11c) ().Citation90

Human milk plasma B cells produce immunoglobulins (discussed further below), which in turn, have roles in blocking pathogenic infection, regulating gut inflammation, and promoting antigen tolerance.Citation91,Citation92 This POM B cell activity indirectly regulates the development of infant RAR-related orphan receptor gamma expressing regulatory T cells (RORγt+ Tregs) – cells that contribute to colonic immune system control by regulating T helper cell effector functions and subsequent inflammatory responses ().Citation93 The critical role of RORγt+ Tregs is illustrated in mice deficient for these cells, who display gut microbiome dysbiosis, increased amounts of inflammatory Th17 cells, and increased risk of colitis.Citation93–95

Innate lymphoid cells produce an array of effector cytokines associated with intestinal microbiome development and are present in POM.Citation96,Citation97 In mice, innate lymphoid cells are major producers of IL-22 - a cytokine associated with gut barrier maintenance and the expression of antimicrobial proteins (e.g Reg3, S100) at mucosal surfaces.Citation96 IL-22 producing innate lymphoid cells have been associated with preventing commensal gut microbiota dissemination and subsequent systemic inflammation in mouse pups, therefore, it is reasonable to speculate that POM innate lymphoid cells may act similarly within infants ().Citation87

Thus, immune cells in POM support the development of infant immune system machinery and promote the development of tolerance to microbial species.Citation87 Establishing this host-microbial symbiosis in infancy relieves the burden of constant immune system stimulation by gut microbes, and can lower the likelihood of developing NEC or other inflammatory conditions.Citation57 Immune cells are inactivated during DHM processing and formula manufacturing.Citation39 The lack of functional immune cells in formula likely contributes to the increased likelihood of these conditions as well as gastrointestinal infections among formula-fed infants.Citation34

Additionally, both formula and DHM feeding circumvent the bidirectional ‘feedback loop’ enabled by suckling at the breast, where it has been shown that DPOM macrophage concentrations increase during infant infections.Citation38 This is relevant to immune development because it suggests that direct feeding allows infants to communicate their immunological needs to their lactating parent.Citation38 The same ‘feedback loop’ deficiency applies to EPOM feeding, which likely also suffers from reduced immune cell activity, depending on milk storage conditions.Citation77

Signaling proteins and bioactive enzymes

POM contains various cytokines, chemokines, growth factors and receptors including transforming growth factor-ß1 (TGF-ß1), TGF-ß2, interleukins IL-10, IL-6, IL-1ß, insulin-like growth factor-1 (IGF-1), IL-7, tumor necrosis factor-α (TNF-α), and granulocyte colony-stimulating factor (GCSF) ().Citation9,Citation33 These signaling proteins are associated with both gut microbiome and immune system development, and can act locally in the intestine through binding cell surface receptors, or systemically by permeating the intestinal barrier ().Citation33

The TGF-ß family is the most abundant family of cytokines in POM.Citation33 TGF-ß proteins induce the differentiation of naïve T cells to antigen-specific Tregs that help maintain intestinal homeostasis, induce tolerance, and regulate inflammation.Citation98 POM TGF-ß1 and TGF-ß2 have been associated with neonatal gut microbial colonization and composition.Citation99 POM TGF-ß2 has been shown to inhibit pro-inflammatory cytokine production in immature intestinal epithelia, decrease gut permeability, and stimulate epithelial repair mechanisms, suggesting they contribute to shaping the intestinal environment and regulating gut microbiota colonization ().Citation99,Citation100 TGF-ß proteins may also interact with other bioactive components (e.g. microbes, oligosaccharides, bioactive enzymes) in POM to influence gut microbiome composition.

Several interleukins in POM (i.e. IL-10, IL-6, and IL-1ß) have been associated with the development of neonatal oral tolerance to foods, although it remains unclear if this results from the direct cytokine activity on the infant gastrointestinal tract, or if these cytokines are markers of another immunomodulatory mechanism transmitted through POM.Citation101 Additionally, GCSF contributes to intestinal epithelial development by increasing intestinal epithelial cell villi, crypt depth, and proliferation in mice pups.Citation102 IGF-1 levels in POM have been associated with both the morphological and functional development of the gastrointestinal tract ().Citation51 IL-7 levels in POM have been associated with infant thymus size and lymphocyte output.Citation103

TNF-α is an inflammatory cytokine normally produced by macrophages/monocytes during acute inflammation,Citation38 contributing to immune system reactivity, microbiota-induced immune responses, and moderation of gut microbiota composition moderation.Citation104 Interestingly, TNF-α levels in DPOM increase when the infant is mounting an immune response to an infection – a dynamic function that is not possible in DHM or formula, and is likely disrupted in EPOM.Citation38

Lactoferrin is present at ~5 g/L in colostrum and decreases to ~2 g/L in mature milk.Citation105 Lactoferrin is only produced in small amounts by neonates; therefore, POM is the main source of lactoferrin in POM-fed neonates.Citation106 This antimicrobial enzyme contributes to selective infant gut colonization, and can prevent pathogenic intestinal bacterial growth by i) chelating iron with a high affinity and ii) altering gram-negative bacterial outer membranes ().Citation107,Citation108 Lactoferrin administration to mice pups and human infants has been shown to increase Bifidobacterium levels in both the gut and feces, indicating lactoferrin has bifidogenic activity.Citation109 Low lactoferrin levels in early life may increase infant susceptibility to aberrant gut microbiome development, sepsis, NEC and enteric infections, as well as diabetes and obesity in later life.Citation106,Citation108 Infant formula contains low levels (~1.4 g/L) of bovine lactoferrin, which has a similar, but decreased ability to chelate iron.Citation56,Citation110 Additional bovine lactoferrin is added to commercial formula in some countries, and is generally recognized as safe by the Food and Drug Administration.Citation106

The pasteurization process involved in DHM preparation leads to 40–85% loss in immunologically detectable concentrations of lactoferrin, and IGF-1.Citation39,Citation41,Citation51 This suggests lactoferrin, and potentially other signaling proteins and bioactive enzymes present in DHM, are denatured or lose bioactive activity.Citation39 The bioactivity of signaling proteins and enzymes in EPOM compared to DPOM has not been widely studied, but likely varies according to each component’s kinetics and sensitivity to different storage conditions.Citation45 Bovine signaling proteins and enzymes are largely destroyed by infant formula manufacturing processes (e.g. enzymatic hydrolysis, fermentation, heat treatment, radiation).Citation83

Immunoglobulins

Immunoglobulins (Igs) - including IgA, soluble IgA [SIgA], IgG, IgM, IgE, and IgD – are present in POM, where they resist infant digestive pathways and maintain function in infants ().Citation111 These functions are especially critical during the early neonatal period because Ig-containing and producing host cells are not present at birth; appearing between 10 days to eight weeks of age depending on Ig.Citation112,Citation113 Thus, young infants rely on innate immune structures (e.g. physical barriers, innate immune cells) and POM antibodies to avoid infection (reviewed in ref.Citation9). Igs transferred in milk can support innate structures by conferring the same function as host-made Igs (i.e. providing ‘passive’ immunity), and by indirectly influencing gut microbiome development, as described below.Citation92 The most well-studied POM Igs in terms of microbiome and immune development are IgA and IgG.Citation92

SIgA is the most abundant Ig in POM and is capable of binding to antigens present on toxins, viruses, and both commensal and pathogenic microbes.Citation92,Citation114 SIgA-microbe interactions contribute to ‘beneficial’ early life microbe (e.g. bifidobacteria, lactobacilli) colonization and maintenance within the infant gut.Citation114,Citation115 SIgA-microbe interactions are also associated with the prevention of both commensal and pathogenic gut microbe translocation across the mucosal epithelium,Citation92 as demonstrated in a mouse model where increased penetration of commensal microbes into mesenteric lymph nodes was observed in pups fed POM lacking IgA, when compared to pups fed POM with IgA.Citation116 Treg activity has also been associated with the level of IgA-microbe coating, suggesting IgA-microbe interactions may have a role in modulating how microbes are sensed by the immune system.Citation114,Citation117

Furthermore, POM IgG and IgA have been shown to dampen mucosal T helper cell responses in mice, limiting adaptive immune responses to commensal antigens.Citation118 Verhasselt et al.Citation27 have also proposed that human milk IgG-antigen immune complexes can transfer across intestinal barriers and mediate inflammatory responses by promoting the formation of infant Tregs. POM Ig microbiota exclusion and immune system regulation promotes intestinal homeostasis and helps prevent excessive immune stimulation as the infant gut is colonized by microbes.Citation92

DHM contains a lower concentration of functional Igs when compared to POM, attributable to DHM pasteurization and freeze-thaw processes.Citation39,Citation111 EPOM Igs may also be affected by freezing, although this has not been widely studied. The lack of immunoglobulins within formula may contribute to the increased levels of gut inflammation observed in formula-fed infants.Citation60,Citation119 Additionally, compared to controls, mouse pups that did not receive SIgA in POM had different gut microbiota compositions persisting into adulthood, and higher expression of genes associated with IBD and inflammatory disease, suggesting that lower levels of functional Igs in DHM and formula may have implications for infant development and lifelong health.Citation92,Citation120 Understanding the composition and impact of POM Igs, both alone and in conjunction with other POM components, on early life immune system development is an important future research direction.

Microbes and probiotics

POM contains microbes, including several taxa that populate the infant gut in early life (e.g. Bifidobacterium and Streptococcus) ().Citation76,Citation121 The diversity and amount of POM microbes change over timeCitation58 and some taxa are shared between POM and infant stool,Citation76 suggesting that milk microbiota may contribute to infant gut colonization ().Citation16–18 However, the sources and functions of milk microbiota are not well understood, in part because it is methodologically challenging to study milk microbes due to their relatively low abundance. It is also important to note that DNA sequencing methods are not able to differentiate between viable and non-viable bacteria; therefore, alternate methods are required to investigate live microbiota in POM and better understand their functional roles.Citation122

Milk from lactating parents who feed EPOM has a different microbial profile than those who do not, suggesting the method of POM feeding (directly at the breast vs. pumped and bottled) is a key factor influencing POM microbiota composition, presumably due to the contribution of exogenous bacteria (e.g. from the infant oral cavity and/or pumping equipment).Citation123,Citation124 Investigating the impact of different breast pump cleaning practices and milk storage conditions will help elucidate the relevance of these processes to immune system development.Citation74,Citation122

DHM pasteurization and formula processing destroy 99% of non-spore forming microbes.Citation39,Citation57 Despite this, DHM feeding is associated with preterm infant gut microbiota composition,Citation125 indicating that other factors present in DHM – such as HMOs and Igs – contribute to infant gut microbiota composition. Research is underway to investigate if incubating pasteurized DHM with a sample of POM can amplify POM microbes in DHM, although the clinical feasibility and safety of this method has yet to be examined. Potentially, this method could assist parents struggling to express adequate quantities of milk by introducing their infants to a beneficial cocktail of POM microbes.Citation126 Collecting multiple POM samples and incubating in DHM over time could also allow for changes in POM microbiota over time to be reflected in DHM.Citation127

Commercial probiotic supplements can be used to add microbes to DHM and formula. Bifidobacterium and Lactobacillus are commonly included in early life probiotic supplements and are generally recognized as safe by the Food and Drug Administration.Citation57,Citation128 Some lactobacilli and bifidobacterial probiotic strains are capable of fermenting HMOs into simpler sugars that can be utilized as an energy source by the host.Citation129,Citation130 Additionally, 6–12 month old infants receiving formula supplemented with Lactobacillus fermentum (and galactooligosaccharide) have a decreased incidence of gastrointestinal and upper respiratory infections when compared to age-matched control infants (receiving formula supplemented with galactooligosaccharides only).Citation131 Bifidobacterium lactis formula supplementation has also been associated with enrichment of beneficial early-life bacteria (Bifidobacterium and Lactobacillus) in low birth weight infants.Citation132 Accordingly, probiotics have been proposed as a strategy to improve gut microbiota colonization in low-birth weight infants exposed to antibiotics early in life;Citation133 however, there are concerns that large doses of probiotics administered to immunologically immature infants could overwhelm the immune system and lead to infection and sepsis.Citation134,Citation135

The optimal composition and safe dosage of probiotic microbes for infants at different stages of development is an important area of ongoing research.Citation136 Notably, human milk could be an important source of inspiration for this research because at least some members of the human milk microbiome appear to have co-evolved with our species to survive digestion and seed the infant gut.Citation76

Oligosaccharides

Prebiotics are compounds that are fermented by beneficial microbiota and stimulate their growth. Human milk oligosaccharides (HMOs), the third most abundant component of human milk, serve as prebiotics in the infant gut where they are the preferred substrate for bifidobacteria and other select taxa.Citation137 HMOs also act as soluble receptor decoys that bind to pathogenic bacteria and prevent them from infecting the host, and can act locally on mucosa-associated lymphoid tissue to inhibit the expression of inflammatory genes ().Citation137 More than 200 HMOs have been identified in POM; their concentrations vary during lactation with the majority being most abundant in colostrum (average 9–22 g/L) and gradually decreasing (6–15 g/L in mature milk) until weaning ().Citation138 Two exceptions are 3-fucosyllactose and 3’ sialyllactose, which increase in concentration over the course of lactation.Citation33,Citation138 Like other POM components, HMO concentration changes likely occur to match changing infant needs over time. HMO concentration is also influenced by country of origin, genetics, lactating parent health and environmental factors.Citation138,Citation139 Unlike many other POM components, HMO concentrations and patterns are not affected by pasteurization or freezing; thus, HMOs are largely preserved in EPOM and DHM.Citation59

Recognizing the importance of HMOs to the infant microbiome and health, formula manufacturers are increasingly incorporating prebiotic oligosaccharides into their products. Initially these were limited to fructooligosaccharides (FOS) and galactooligosaccharides (GOS), which are oligomers derived from lactose and inulin, respectively.Citation140 In randomized clinical trials of formula-fed infants comparing GOS and/or FOS-enriched formulas to control formulas without these additives, GOS has been shown to enrich Bifidobacterium in infant stoolCitation84 and GOS/FOS reduced the rate of infections in the first six months of life.Citation140 Bovine oligosaccharides have also been added to infant formulas, eliciting higher counts of infant stool Bifidobacterium and Lactobacillus compared with control formula.Citation141 More recently, some formulas have begun to incorporate HMOs including synthetic 2’-fucosyllactose and lacto-N-neotetraose, with initial studies focused primarily on establishing the safety of these additives.Citation142 More research is required to understand the clinical benefits of adding these ingredients to infant formula, and to explore the value of other combinations of different oligosaccharides. It is important to note that GOS and FOS are structurally and functionally very distinct from HMOs naturally found in human milk,Citation33,Citation137 and that even HMO-containing formulas have vastly different HMO compositions compared to human milk (i.e. just one or a few HMOs at relatively low concentrations in formula, compared to the dozens of different and highly abundant HMOs in human milk).Citation137

Fatty acids

Human milk contains fat globules (also known as mature lipid droplets) predominantly composed of fatty acid (FA) bound triacylglycerols.Citation143 Mature human milk FA content consists of<1% short-chain FAs, ~12% medium-chain FAs, ~82% long-chain FAs, and~1% “essential” polyunsaturated long-chain FAs. Essential FAs cannot be biologically synthesized by humans and must be acquired from an individual’s diet.Citation60,Citation143 Human milk FAs serve as a major energy source for the infant and have essential biological functions contributing to neurological and immune system development, as well as gut microbiota colonization in early life ().Citation144,Citation145

FAs are antimicrobial – their amphiphilic properties allow them to disrupt or remodel microbial cell membranes by acting as a detergent and solubilizing cell membrane materials, or by inserting into a microbial cell membrane.Citation143,Citation146 These mechanisms lead to the inhibition of microbial growth, observed in vitro when microbial taxa such as Lactobacillus, Bifidobacterium, Escherichia, and Clostridium are cultured with medium-chain or long-chain polyunsaturated FAs.Citation145,Citation147,Citation148 Despite uncertain nuances of FA-microbe interactions, including structure-dependent antimicrobial activity differences and potentially positive effects of absorbed FAs on microbial activities,Citation145 this evidence suggests that POM-derived long and medium-chain FAs may contribute to the regulation of microbial growth in the gut ().

POM FAs have been associated with infant immune function. Laitinen et al.Citation149 show that POM received by infants with atopic eczema contained a lower amount of polyunsaturated long-chain FAs compared to control infants. Furthermore, essential polyunsaturated long-chain FAs present in POM, including arachidonic acid and docosahexaenoic acid, induce anti-inflammatory cytokine expression and Treg activation.Citation150 These FAs are also important substrates for the synthesis of eicosanoids – signaling molecules with critical roles in immune system processes including platelet aggregation and cell proliferation.Citation144 In addition short-chain FAs in POM can serve as energy sources for epithelial cells and stimulate lamina propria Treg proliferation and anti-inflammatory cytokine expression.Citation151

Human milk FA profiles change with lactation stage – the concentrations of short and medium-chain FAs increase over time, and the concentrations of essential polyunsaturated long-chain FAs (i.e. docosahexaenoic acid and arachidonic acid) decrease over time.Citation60,Citation63,Citation143 The human milk FA profile is also influenced by geographic location, maternal diet and maternal BMI.Citation63 The association between POM FAs and immune system activity indicate FA profile changes may occur in a manner suited to the infant’s evolving gut microbiome, immune system, and dietary source.Citation63

Human milk FA profiles are not impacted by Holder pasteurizationCitation64 and appear to remain stable when human milk is frozen at −60°C.Citation152 In contrast, freezing human milk at −20°C leads to a decrease in fat content in as few as two daysCitation65, which has implications for EPOM and DHM as parents and milk banks and often freeze human milk at −20°C.Citation67 FA profiles are further impacted by lactation stage – a particularly important consideration for DHM because human milk donors are often term-delivering parents donating mature milk.Citation111 In samples collected from an Iowa milk bank, docosahexaenoic acid levels were 53% lower in DHM, compared to colostrum, indicating there may be an increased risk of essential FA deficiency in infants receiving DHM.Citation66 To address this concern, it has been suggested that human milk banks could supplement DHM with docosahexaenoic acid, or encourage human milk donors to supplement their diet with docosahexaenoic acid.Citation66,Citation148

Most infant formula is derived from cow’s milk and supplemented with vegetable fat blends (e.g. palm, coconut, sunflower, and/or soy oil), as well as manufactured docosahexaenoic acid and arachidonic acid, in an attempt to create a FA profile that closely resembles human milk.Citation60,Citation153 However, most vegetable FAs are attached to triglycerides at different structural positions (sn-1 and sn-3) compared to human milk FAs (sn-2).Citation154 One study has shown that infants receiving synthetic sn-2 positioned palmitic acid for six weeks had an increased abundance of fecal Lactobacillus and Bifidobacterium when compared to infants receiving a control formula.Citation155 Optimizing the FA profile of human milk substitutes is an active area of research.

Postbiotics

Postbiotics refer to non-viable microbes (intact or broken) and/or microbial metabolites that confer a health benefit on the host. This includes cell wall fragments, microbial cell fractions, short-chain fatty acids, enzymes, vitamins, and other bacterial metabolites.Citation68,Citation156 Postbiotics have some similar functions to probiotics; for example, they can adhere to gut mucosa, bind to cell receptors, transduce receptor signals, and exclude pathogens from colonizing the gut microbiome. However, they do not actively metabolize prebiotic substrates.Citation157 In gnotobiotic mice, Bifidobacterium breve postbiotics have been shown to suppress pro-inflammatory cytokine production in the spleen, although live strains had greater impact on the regulation of intestinal metabolism.Citation158 Mice receiving infant formula supplemented with postbiotics derived from Bifidobacterium breve and Streptococcus thermophilus show prolonged dendritic cell survival and maturation, improved epithelial barrier function, and increased anti-inflammatory cytokine (i.e. IL-10) production ().Citation68 Additionally, metabolic products released by Lactobacillus paracasei fermentation, when added to formula, have been shown to inhibit immune cell inflammation and can protect against colitis in mice.Citation68,Citation159 Further research is needed to determine if similar effects are seen in humans, but it has been suggested that postbiotics could serve as a safer option to probiotics for particularly vulnerable infants at risk for infection or NEC.Citation68,Citation156

Postbiotics can be present in any solution containing microbes, including all forms of human milk (). Presumably, differences in EPOM and DPOM microbiota composition (described above) would lead to differences in postbiotic composition – although to our knowledge, this has not been studied. The DHM pasteurization process kills viable microbes, suggesting the level of postbiotics in DHM may be higher compared to POM. DHM postbiotic activity may contribute to the maintained protection and stabilization of infants at risk of or suffering from NEC. At the time of this review, however, little has been reported on DHM postbiotics.Citation160 Finally, while raw cow’s milk contains postbiotics, formula processing techniques result in protein proteolysis and denaturation, as well as epitope destruction, which likely impact postbiotic presence and functionality.Citation39

Further research is required to understand the function of postbiotic compounds and confirm the safety and utility of postbiotic supplementation. This includes investigating their influence on gut microbiome development and immune system maturation.

Critical processes in early life immune system development affected by early nutrition and the microbiome

Epigenetic modifications and the so-called “weaning reaction” are two critical processes that are affected by early nutrition and gut microbiota, and contribute to immune development. In this section, we describe these key processes and discuss how they may be influenced by different nutritional sources during infancy.

The weaning reaction

Weaning refers to the dietary shift from POM to other foods. Mouse studies show that, during a specific time window, weaning pups undergo vigorous gut microbiota-induced immune stimulation leading to changes in gene expression and microbiome composition in the intestinal tract ().Citation17,Citation161 Al Nabhani et al.Citation161 have shown that this weaning reaction contributes to RORγt+ Treg generation and the prevention of “pathological imprinting” (susceptibility to inflammatory pathologies later in life, including allergy, autoimmunity, IBD, and chronic inflammation). Although mostly studied in mice thus far, the weaning reaction may have a critical role in human immune system development.Citation26,Citation161,Citation162

Figure 3. Approximate changes in milk composition and other factors associated with the weaning reaction over time, in mice. Adapted from Hornef and Torow (2019).Citation1

Figure 3. Approximate changes in milk composition and other factors associated with the weaning reaction over time, in mice. Adapted from Hornef and Torow (2019).Citation1

Overview of the weaning reaction

Weaning is accompanied by the withdrawal of POM components from the infant system and changes to gut microbiome composition induced by new nutritional sources (). During this transition, the loss of POM components appears to induce the formation of goblet cell associated passages (GAPs) in the colon and small intestine.Citation26,Citation162 Although other routes of translocation across the luminal epithelium exist, GAPs are the main pathway for delivering luminal substances to the colonic and small intestine lamina propria (the connective tissue that underlies the gut mucosal epithelium).Citation162 These luminal substances, including microbial and dietary antigens, stimulate lamina propria immune cells (e.g. mononuclear phagocytes) and facilitate the generation and maintenance of RORγt+ Tregs,Citation26,Citation162 which further regulate effector T cells to promote antigen tolerance ().Citation93 This sequence of developmental processes has lifelong implications, as early-life insufficiency of RORγt+ Tregs is associated with an increased susceptibility to colitis, allergic inflammation, and cancer across the lifecourse.Citation161

The weaning reaction also induces widespread changes in gene expression.Citation161 Genes induced following the recognition of luminal antigens by innate immune receptors in the gut epithelium include many associated with anti-microbial immunity, such as Reg3 defensins, CC and CXC chemokines, and cytokine receptors. Additionally, the weaning reaction stimulates TNF-α and IFN-γ expression in T cells ().

Throughout life, GAPs facilitate the creation and maintenance of lamina propria Tregs by “sampling” luminal antigensCitation162, but this process is inhibited periodically when antigen exposure may be harmful to the host. For example, it has been shown in mice that GAP inhibition occurs as a defense mechanism to limit inflammation when the host is infected with Salmonella.Citation163 Similarly, POM-induced inhibition of GAP formation in early life appears to be an important mechanism for preventing excessive immune system stimulation in young offspring who may be unable to efficiently clear or respond to luminal antigen exposures.Citation164

EGF and other EGFR ligands in POM have been proposed as the master regulators of GAP inhibition prior to weaning (). In POM-fed mice, who typically wean around 3 weeks of age, POM-derived EGF is thought to inhibit GAP formation in the first 10 days by binding to goblet cell EGFRs ().Citation26,Citation165 This is supported by i) evidence showing limited presence of luminal antigens in the normal mouse gut during this period,Citation26 ii) a cross-fostering study showing that “early milk” blocked the weaning reaction in mouse pups while mature milk did not,Citation161 and iii) experiments showing that the weaning reaction was blocked by EGF exposure and induced by EGF receptor inhibition.Citation161 Additionally, EGF supplementation has been shown to increase intestinal barrier strength and reduce NEC severity in preterm rats.Citation166

Further evidence of the time-sensitive weaning reaction comes from experiments manipulating the timing of microbial exposures in germ-free mice.Citation161 Germ-free mouse pups introduced early to microbes that normally expand during weaning (e.g. Clostridia, Bacteroidia) are pathologically imprinted (i.e. have an increased susceptibility to inflammatory pathologies) when compared to pups that are exposed to the same microbes during the developmentally appropriate critical time window. Furthermore, germ-free pups introduced to the same microbes after the completion of weaning do not express TNF-α and IFN-γ - two markers of the weaning reaction. Thus, the introduction of microbes before or after the window of opportunity appears problematic for host development. Aside from the timing of exposure, the type of microbial stimulation also appears to be important, as gram-positive anaerobic microbes were necessary to induce the weaning reaction in these germ-free mouse experiments. Together, this evidence indicates that important microbiome-mediated developmental imprinting occurs during a clearly-defined time window that “opens” in response to the withdrawal of EGF in POM (), and “closes” after weaning is complete, though the mechanisms of this closure remain to be determined.Citation26,Citation161

The weaning reaction has not been clearly defined in humans, but similar to mice, EGF is present at high concentrations in human colostrum (25–40 ng/mL) and gradually decreases as POM matures (5–12 ng/mL in mature milk).Citation54 It has been hypothesized that when POM EGF levels drop below the threshold required for GAP inhibition, GAPs form and allow vigorous microbial dissemination into both the small intestine and colonic lamina propria (), triggering a critical shift in gene expression and cascade of immune system maturation processes required for optimal development and health.Citation161 Notably, this hypothesis suggests that the “weaning reaction” is stimulated by natural changes in POM composition, and could be triggered by the premature withdrawal of POM components, but does not necessarily require complete cessation of POM consumption.

Nutritional impacts on the weaning reaction

While EPOM and DHM preparation processes do not have a significant impact on EGF concentrations in milk,Citation39,Citation45 they might impact the weaning reaction by disrupting the “chronobiology” of POM feeding.Citation164 Human milk donors are most often term-delivering parents donating mature milk,Citation111 with relatively lower EGF levels than would be present in POM produced for premature newborns at early stages of development, which may not effectively inhibit GAP formation. Indeed, this has been demonstrated in mice where “early milk” blocked the weaning reaction in recipient pups, whereas mature milk did not.Citation161 This supports the proposal by Knoop et al.Citation164 that due to temporal changes in POM composition, DHM from donors closer to parturition may provide superior protection compared to DHM expressed later in lactation.

Formula contains little or no EGF.Citation55 This is reflected in infant stool, where EGF concentrations are lower over the first 60 days of life among exclusively formula-fed vs. exclusively human milk-fed infants.Citation164 While the weaning reaction was not characterized in this human study, it has been shown that mice with low EGF exposure prior to weaning (mimicking the exposure of formula-fed children) have accelerated colon GAP formation, increased bacterial translocation, and an elevated risk of enteric pathogen sepsis.Citation164 This suggests the lack of EGF in formula may have implications for health in humans. Indeed, brief periods of formula feeding have been associated with an increased risk of NEC, when compared to infants receiving exclusive POM.Citation167 These brief periods without POM EGF may induce GAP formation, luminal antigen exposure among colon and small intestine lamina propria immune cells, and inflammatory responses, potentially explaining the increased risk of NEC associated with brief periods of neonatal formula feeding.

Finally, considering that gut microbes are key mediators of the weaning reaction,Citation161 it is also possible that different early life nutritional sources could indirectly influence the weaning reaction through their strong and differential impacts on the gut microbiome in early life. Understanding the strain-specificity of the weaning reaction could help elucidate the role of the gut microbiota in immune system development.

Early life epigenetic modifications

Early life epigenetic processes regulate gene expression and impact lifelong health, serving as a primary mechanism for developmental programming. The term epigenetics means “above” or “on top of” genetics and refers to the systems that control gene expression without transforming genomic sequences. Epigenetics control gene expression through i) covalent modifications to DNA and histones (e.g. methylation, acetylation and phosphorylation) – known as epigenetic imprintingCitation168,Citation169 and ii) microRNA (miRNA) interference, where miRNAs bind to mRNA and disrupt the translation process or signal mRNA degradation. Epigenetic imprinting occurs to the greatest degree from the time of conception to the infant’s second birthday (the first 1000 days of life),Citation169 and has been associated with both early life nutrition and the development of conditions such as asthma, allergy, obesity and diabetes later in life.Citation29 Gut microbiota influence epigenetic processes that mediate inflammation and intestinal barrier integrity in early life.Citation169 Thus, the interplay between infant nutrition, the gut microbiome, and epigenetic modifications is an important relationship to consider when investigating immune system development.

Early life nutrition and epigenetics

Nutritional components and by-products (e.g. folate, methionine, choline, vitamin B-12) can modify epigenetic reactions, and the diet is an important source of epigenetically-active miRNAs. Therefore, the variable nutrient profiles and miRNA content of different early life nutritional sources are key determinants of epigenetic programming during infancy.Citation29,Citation71,Citation168 Indeed, early life nutritional exposures have been associated with epigenetic modification of genes vital for immune system development and growth, such as FOXP3 (Treg development), FTO (obesity), INS (diabetes), IGF1 (growth).Citation29,Citation71,Citation169 In pigs, formula feeding has been associated with decreased amounts of methylation, and increased amounts of hyperacetylation at various histone protein sites responsible for inflammatory gene (IL-8, toll-like receptor (TLR) 4) expression, when compared to POM feeding.Citation170 In human intestinal cells, human milk was shown to inhibit the activation pathway of NF-κB.Citation71,Citation171 Furthermore, the expression of NF-κB and Foxp3 is down and up-regulated, respectively, by human milk miRNAs.Citation29,Citation172 These transcription factors target thousands of other genes, including many that regulate Treg proliferation and immune system development.Citation173 Additionally, new evidence shows that intake of POM by very low birth weight infants is associated with DNA methylation patterns at 5.5 years of age.Citation174 The epigenetic signatures of other infant feeding practices among full term infants has yet to be established but may provide a deeper understanding of how POM shapes immune system development.

miRNA content and composition vary among human milk sources and substitutes. POM contains over 1400 miRNAs originating from the lactating gland; with concentrations decreasing over the course of lactation ().Citation71 The composition of miRNAs in POM depends on the lactating parent’s diet, gut microbiota, genetics and time postpartum,Citation71,Citation175 presumably in response to infant feeding patterns and needs.Citation176 These POM miRNAs have been associated with the expression of over 4000 DNA CpG sites – including sites in genes involved in development and function of the immune system (). For example, MiR-223 is a common POM miRNA that regulates transcription factors that influence the proliferation of T cells and granulocytes.Citation71 Similarly, MiR-155 is also found in POM and regulates Treg development indirectly by regulating Foxp3 transcription factor expression.Citation172 Determining additional targets and functions of POM miRNAs is an important direction for future research.

miRNA differences between EPOM and DPOM have not been studied. miRNAs are present in DHM and are resistant to degradation characteristic of pasteurization and milk bank storage.Citation71 miRNAs have been detected in cow milk-based formula, but at very low concentrations relative to human POM.Citation70,Citation177

Early life gut microbiota and epigenetics

Early nutritional exposures can also influence epigenetic modifications through their impact on the gut microbiome.Citation169 Two major mechanisms have been suggested to explain how microbiota influence epigenome variation.Citation178

(I) Gut microbes can impact host epigenetic modification pathways by interacting directly with components that mediate epigenetic mechanisms.Citation169,Citation179 For example, commensal microbiota are recognized by pattern-recognition receptors (PRRs) (e.g. Toll-Like Receptors [TLRs]) on intestinal epithelial cells, triggering the expression of genes with epigenetic activity, such as Pigp, Hdac9, H3Kac, H4Kac, and Tollip (reviewed in ref.Citation180)(). Additionally, methylation of the TLR4 gene is lower in germ-free vs. conventionally housed mice,Citation181 indicating a role for gut microbiota in mediating this epigenetic modification and the downstream expression of TLR4. These findings highlight a potential mechanism of microbiota-epigenetic-induced tolerance in early life, because TLRs are involved in sensing and stimulating an immune response to environmental stimuli.Citation181 Further investigation is required to understand other pattern recognition receptor signal transduction pathways induced by different microbes and elucidate their downstream epigenetic effects. Further research is also required to investigate epigenetic modifications resulting from the incorporation of foreign, microbial genetic material into the host.Citation178

(II) Microbial metabolites can alter the availability and function of epigenetic machinery (reviewed in ref.Citation182) (). For example, butyrate produced by microbes (e.g. select Lactobacillus, Bifidobacterium, and Clostridium species) can act as a histone deacetylase (HDAC) inhibitor.Citation183 Butyrate HDAC inhibition can suppress pro-inflammatory NF-κB activation and decrease IFN-γ production.Citation184 Lactobacilli and bifidobacteria also produce folate, a cofactor necessary for the transfer of carbons in DNA methylation processes.Citation183 Folate and butyrate are also present in POM and DHM ().Citation33 Without these microbial metabolites, infants can be predisposed to intestinal and systemic inflammation,Citation178 which are hallmarks of conditions such as NEC, asthma and allergies.Citation179 Early life nutritional sources that promote lactobacilli and bifidobacteria colonization of the infant gut, such as POM and DHM, can regulate inflammation levels in this way.Citation168,Citation169,Citation179

Alongside this evidence that gut microbes affect host epigenetics, there is also evidence that epigenetic regulation of epithelial barrier development and intestinal metabolite processing can affect gut colonization. This indicates the presence of a host-microbe cross-talk mechanism in which microbiota influence host epigenetic signatures, and host epigenetics influence microbiota composition – both of which are clearly impacted by early life nutrition and feeding practices, as described above ().Citation178 More research is needed to gain a deeper understanding of how early life nutrition affects host-microbe cross-talk and it’s resulting impact on host epigenetics and immune development.

Conclusion

The collective evidence in this review suggests early life POM feeding optimally regulates the colonization and composition of the infant gut microbiome, with both short and long-term health implications. POM components also mediate the interplay between the infant gut microbiome and immune system stimulation – a process critical for tolerance induction, healthy immune system development, and prevention of pathological imprinting. Many POM components – from nutrients and enzymes to cells and microbes – have been directly or indirectly linked to these key developmental processes. Temporal changes in POM allow the maturing infant immune system to gradually gain more “responsibility and independence” over time. When fed directly at the breast, POM appears to adapt to the infant’s immunological needs through bi-directional parent-infant signaling. Fully characterizing the extent and significance of POM composition and functionality as a biological systemCitation35, including its dynamism and adaptability, will help us further understand the fundamental influence of nutrition on additional processes that were beyond the scope of this review, including metabolism and neurodevelopment, as well as overall human development.

This review also highlights the limitations of DPOM substitutes from the perspectives of gut microbiome and immune development. Commercial formula lacks most of the immunomodulatory components found in POM, and is not a dynamic nor personalized form of nutrition. While DHM retains some of these factors, processing steps alter their bioactivity and current DHM provision strategies are not personalized or dynamic. Finally, EPOM feeding is the closest alternative to DPOM and offers many benefits, but affects milk microbiota composition and interferes with the natural dynamism and adaptability of DPOM feeding. Throughout the review, we have identified priority research areas (summarized in Box 1) with the goal of improving all forms of infant feeding to support lifelong immune health.

Box 1. Priority areas for future research on early life nutrition, gut microbiota and immune development.

Abbreviations

POM=

parent’s own milk

Ig=

immunoglobulin

HMO=

human milk oligosaccharide

NEC=

necrotizing enterocolitis

DPOM=

direct parent’s own milk

EPOM=

expressed parent’s own milk

DHM=

donor human milk

TGF=

transforming growth factor

IL=

interleukin

IGF=

insulin-like growth factor

EGF=

epithelial growth factor

GCSF=

granulocyte colony-stimulating factor

IBD=

inflammatory bowel disease

TNF-α=

tumor necrosis factor alpha

TLR=

toll like receptor

NF-κB=

nuclear factor-κB

RORγt+ Treg=

RAR-related orphan receptor gamma expressing regulatory T cell

FOS=

fructooligosaccharide

GOS=

galactooligosaccharides

FA=

fatty acid

Disclosure statement

M.B.A. receives research funding from the Canadian Institutes of Health Research, US National Institutes of Health, Research Manitoba, the Canada Foundation for Innovation, the Bill and Melinda Gates Foundation, the Manitoba Children’s Hospital Foundation, CIFAR, and the Garfield Weston Foundation. She has previously received funding from Prolacta Biosciences. She has contributed without remuneration to online courses on breast milk and the infant microbiome produced by Microbiome Courses. She serves in a volunteer capacity for the International Society for Research on Human Milk and Lactation and as a member of the US Canada Joint Human Milk Initiative. She has consulted for DSM Nutritional Products and serves on the Scientific Advisory Boards of TinyHealth and MalaikaVx.

Additional information

Funding

This review was supported, in part, by Canadian Institutes of Health Research Canadian Microbiome Initiative 2 Team Grant (MRT-168044) and by the Canada Research Chairs Program. M.B.A. is supported as a Canada Research Chair in the Developmental Origins of Chronic Disease and is a Fellow of the CIFAR Humans & Microbiome Program. S.R.A is supported by a CIHR Canada Graduate Scholarships - Master’s award.

Unknown widget #5d0ef076-e0a7-421c-8315-2b007028953f

of type scholix-links

References

  • Hornef MW, Torow N. ‘Layered immunity’ and the ‘neonatal window of opportunity’ – timed succession of non-redundant phases to establish mucosal host–microbial homeostasis after birth. Immunology. 2020;159(1):15–25. doi:10.1111/imm.13149.
  • Weström B, Arévalo Sureda E, Pierzynowska K, Pierzynowski SG, Pérez-Cano F-J. The immature gut barrier and its importance in establishing immunity in newborn mammals. Front Immunol. 2020;11:1153. doi:10.3389/fimmu.2020.01153.
  • Nussbaum C, Gloning A, Pruenster M, Frommhold D, Bierschenk S, Genzel-Boroviczeny O, von Andrian UH, Quackenbush E, Sperandio M. Neutrophil and endothelial adhesive function during human fetal ontogeny. J Leukoc Biol. 2013;93(2):175–184. doi:10.1189/jlb.0912468.
  • Sadeghi K, Berger A, Langgartner M, Prusa A, Hayde M, Herkner K, Pollak A, Spittler A, Förster-waldl E. Immaturity of infection control in preterm and term newborns is associated with impaired toll-like receptor signaling. J Infect Dis. 2007;195(2):296–302. doi:10.1086/509892.
  • Willems F, Vollstedt S, Suter M. Phenotype and function of neonatal DC. Eur J Immunol. 2009;39(1):26–35. doi:10.1002/eji.200838391.
  • Brambell FWR. The transmission of immunity from mother to young and the catabolism of immunoglobulins. The Lancet. 1966;2(7473):1087–1093. doi:10.1016/S0140-6736(66)92190-8.
  • Haynes BF, Martin ME, Kay HH, Kurtzberg J. Early events in human T cell ontogeny. Phenotypic characterization and immunohistologic localization of T cell precursors in early human fetal tissues. J Exp Med. 1988;168(3):1061–1080. doi:10.1084/jem.168.3.1061.
  • Mold JE, Venkatasubrahmanyam S, Burt TD, Michaëlsson J, Rivera JM, Galkina SA, Weinberg K, Stoddart CA, McCune JM. Fetal and adult hematopoietic stem cells give rise to distinct t cell lineages in humans. Science. 2010;330(6011):1695–1699. doi:10.1126/science.1196509.
  • Dawod B, Marshall JS, Azad MB. Breastfeeding and the developmental origins of mucosal immunity: how human milk shapes the innate and adaptive mucosal immune systems. Curr Opin Gastroenterol. 2021;37(6):547–556. doi:10.1097/MOG.0000000000000778.
  • Yao Y, Cai X, Ye Y, Wang F, Chen F, Zheng C. The role of microbiota in infant health: from early life to adulthood. Front Immunol. 2021;12:708472. doi:10.3389/fimmu.2021.708472.
  • Houghteling PD, Walker WA. Why is initial bacterial colonization of the intestine important to infants’ and children’s health? J Pediatr Gastroenterol Nutr. 2015;60(3):294–307. doi:10.1097/MPG.0000000000000597.
  • He X, Parenti M, Grip T, Domellöf M, Lönnerdal B, Hernell O, Timby N, Slupsky CM. Metabolic phenotype of breast-fed infants, and infants fed standard formula or bovine MFGM supplemented formula: a randomized controlled trial. Sci Rep. 2019;9(1):339. doi:10.1038/s41598-019-48858-y.
  • Isaacs EB, Fischl BR, Quinn BT, Chong WK, Gadian DG, Lucas A. Impact of breast milk on intelligence quotient, brain size, and white matter development. Pediatr Res. 2010;67(4):357–362. doi:10.1203/PDR.0b013e3181d026da.
  • Cryan JF, O’riordan KJ, Sandhu K, Peterson V, Dinan TG. The gut microbiome in neurological disorders. Lancet Neurol. 2020;19(2):179–194. doi:10.1016/S1474-4422(19)30356-4.
  • Hills RD, Pontefract BA, Mishcon HR, Black CA, Sutton SC, Theberge CR. Gut microbiome: profound implications for diet and disease. Nutrients. 2019;11(7):1613. doi:10.3390/nu11071613.
  • Stewart CJ, Ajami NJ, O’brien JL, Hutchinson DS, Smith DP, Wong MC, Ross MC, Lloyd RE, Doddapaneni HV, Metcalf GA, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018;562(7728):583–588. doi:10.1038/s41586-018-0617-x.
  • 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.
  • 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. 2020;65(3):706–722. doi:10.1007/s10620-020-06092-x.
  • 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.
  • Ho NT, Li F, Lee-Sarwar KA, Tun HM, Brown BP, Pannaraj PS, Bender JM, Azad MB, Thompson AL, Weiss ST, et al. Meta-analysis of effects of exclusive breastfeeding on infant gut microbiota across populations. Nat Commun. 2018;9(1):4169. doi:10.1038/s41467-018-06473-x.
  • Panwar RB, Sequeira RP, Clarke TB. Microbiota-mediated protection against antibiotic-resistant pathogens. Genes Immun. 2021;22(5-6):255–267. doi:10.1038/s41435-021-00129-5.
  • 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.
  • Peng L, Li Z-R, 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. 2009;139(9):1619–1625. doi:10.3945/jn.109.104638.
  • Zhang S, Paul S, Kundu P. NF-κB regulation by gut microbiota decides homeostasis or disease outcome during ageing. Front Cell Dev Biol. 2022;10:874940. doi:10.3389/fcell.2022.874940.
  • Frost BL, Jilling T, Caplan MS. The importance of pro-inflammatory signaling in neonatal necrotizing enterocolitis. Semin Perinatol. 2008;32(2):100–106. doi:10.1053/j.semperi.2008.01.001.
  • Knoop KA, Gustafsson JK, McDonald KG, Kulkarni DH, Coughlin PE, McCrate S, Kim D, Hsieh C-S, Hogan SP, Elson CO, et al. Microbial antigen encounter during a preweaning interval is critical for tolerance to gut bacteria. Sci Immunol. 2017;2(18): eaao1314. doi:10.1126/sciimmunol.aao1314.
  • Verhasselt V. Neonatal tolerance under breastfeeding influence. Curr Opin Immunol. 2010;22(5):623–630. doi:10.1016/j.coi.2010.08.008.
  • Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, Tuohy K. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr. 2018;57(1):1–24. doi:10.1007/s00394-017-1445-8.
  • van Esch BC, Porbahaie M, Abbring S, Garssen J, Potaczek DP, Savelkoul HFJ, van Neerven RJJ. The impact of milk and its components on epigenetic programming of immune function in early life and beyond: implications for allergy and asthma. Front Immunol. 2020;11:2141. doi:10.3389/fimmu.2020.02141.
  • Stiemsma LT, Turvey SE. Asthma and the microbiome: defining the critical window in early life. Allergy Asthma Clin Immunol. 2017;13:3. doi:10.1186/s13223-016-0173-6.
  • Sarkar A, Yoo JY, Dutra SVO, Morgan KH, Groer M. The association between early-life gut microbiota and long-term health and diseases. J Clin Med. 2022;10(3):459. doi:10.3390/jcm10030459.
  • Palmquist AEL, Doehler K. Human milk sharing practices in the U.S. Matern Child Nutr. 2016;12(2):278–290. doi:10.1111/mcn.12221.
  • Ballard O, Morrow AL. Human milk composition: nutrients and bioactive factors. Pediatr Clin North Am. 2013;60(1):49–74. doi:10.1016/j.pcl.2012.10.002.
  • Victora CG, Bahl R, Barros AJD, França GVA, Horton S, Krasevec J, Murch S, Sankar MJ, Walker N, Rollins NC, et al. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. The Lancet. 2016;387(10017):475–490. doi:10.1016/S0140-6736(15)01024-7.
  • Shenhav L, Azad MB, Gilbert JA. Using community ecology theory and computational microbiome methods to study human milk as a biological system. mSystems. 2022;7(1): e0113221. doi:10.1128/msystems.01132-21.
  • Christian P, Smith ER, Lee SE, Vargas AJ, Bremer AA, Raiten DJ. The need to study human milk as a biological system. Am J Clin Nutr. 113(5);1063–1072. doi:10.1093/ajcn/nqab075.
  • Kulski J, Hartmann P. Changes in human milk composition during the initiation of lactation. Aust J Exp Biol Med Sci. 1981;59(1):101–114. doi:10.1038/icb.1981.6.
  • Riskin A, Almog M, Peri R, Halasz K, Srugo I, Kessel A. Changes in immunomodulatory constituents of human milk in response to active infection in the nursing infant. Pediatr Res. 2012;71(2):220–225. doi:10.1038/pr.2011.34.
  • Ewaschuk JB, Unger S, Harvey S, O’connor DL, Field CJ. Effect of pasteurization on immune components of milk: implications for feeding preterm infants. Appl Physiol Nutr Metab. 2011;36(2):175–182. doi:10.1139/h11-008.
  • Witkowska-Zimny M, Kaminska-El-Hassan E. Cells of human breast milk. Cell Mol Biol Lett. 2017;22:11. doi:10.1186/s11658-017-0042-4.
  • Dussault N, Cayer MP, Landry P, de Grandmont MJ, Cloutier M, Thibault L, Girard M. Comparison of the effect of holder pasteurization and high-pressure processing on human milk bacterial load and bioactive factors preservation. J Pediatr Gastroenterol Nutr. 2021;72(5):756–762. doi:10.1097/MPG.0000000000003065.
  • Ahrabi AF, Handa D, Codipilly CN, Shah S, Williams JE, McGuire MA, Potak D, Aharon GG, Schanler RJ. Effects of extended freezer storage on the integrity of human milk. J Pediatr. 2016;177:140–143. doi:10.1016/j.jpeds.2016.06.024.
  • Czosnykowska-Łukacka M, Lis-Kuberka J, Królak-Olejnik B, Orczyk-Pawiłowicz M. Changes in human milk immunoglobulin profile during prolonged lactation. Front Pediatr. 2020;8:428. doi:10.3389/fped.2020.00428.
  • Clyne PS, Kulczycki A. Human breast milk contains bovine lgG. Relationship to infant colic? Pediatrics. 1991;87(4):439–444. doi:10.1542/peds.87.4.439.
  • Ramírez-Santana C, Pérez-Cano FJ, Audí C, Castell M, Moretones MG, López-Sabater MC, Castellote C, Franch A. Effects of cooling and freezing storage on the stability of bioactive factors in human colostrum. J Dairy Sci. 2012;95(5):2319–2325. doi:10.3168/jds.2011-5066.
  • Peroni DG, Piacentini GL, Bodini A, Pigozzi R, Boner AL. Transforming growth factor-β1 is elevated in unpasteurized cow’s milk. Pediatr Allergy Immunol. 2009;20(1):42–44. doi:10.1111/j.1399-3038.2008.00737.x.
  • Frost BL, Jilling T, Lapin B, Maheshwari A, Caplan MS. Maternal breast milk transforming growth factor-beta and feeding intolerance in preterm infants. Pediatr Res. 2014;76(4):386–393. doi:10.1038/pr.2014.96.
  • Dawod B, Marshall JS. Cytokines and soluble receptors in breast milk as enhancers of oral tolerance development. Front Immunol. 2019;10:16. doi:10.3389/fimmu.2019.00016.
  • Untalan PB, Keeney SE, Palkowetz KH, Rivera A, Goldman AS. Heat susceptibility of interleukin-10 and other cytokines in donor human milk. Breastfeed Med. 2009;4(3):137–144. doi:10.1089/bfm.2008.0145.
  • Giorgi MV, Codipilly CN, Potak D, Heiman HS, Schanler RJ. Pasteurization preserves IL-8 in human milk. Front Pediatr. 2018;6:281. doi:10.3389/fped.2018.00281.
  • Hoeflich A, Meyer Z. Functional analysis of the IGF-system in milk. Best Pract Res Clin Endocrinol Metab. 2017;31(4):409–418. doi:10.1016/j.beem.2017.10.002.
  • Ngom PT, Collinson AC, Pido-Lopez J, Henson SM, Prentice AM, Aspinall R. Improved thymic function in exclusively breastfed infants is associated with higher interleukin 7 concentrations in their mothers’ breast milk. Am J Clin Nutr. 2004;80(3):722–728. doi:10.1093/ajcn/80.3.722.
  • Ewaschuk JB, Unger S, O’connor DL, Stone D, Harvey S, Clandinin MT, Field CJ. Effect of pasteurization on selected immune components of donated human breast milk. J Perinatol. 2011;31(9):593–598. doi:10.1038/jp.2010.209.
  • Cummins AG, Thompson FM. Effect of breast milk and weaning on epithelial growth of the small intestine in humans. Gut. 2002;51(5):748–754. doi:10.1136/gut.51.5.748.
  • Yagi H, Suzuki S, Noji T, Nagashima K, Kuroume T. Epidermal growth factor in cow’s milk and milk formulas. Acta Paediatr Scand. 1986;75(2):233–235. doi:10.1111/j.1651-2227.1986.tb10190.x.
  • Satue-Gracia MT, Frankel EN, Rangavajhyala N, German JB. Lactoferrin in infant formulas: effect on oxidation. J Agric Food Chem. 2000;48(10):4984–4990. doi:10.1021/jf0002490.
  • Kapourchali FR, Cresci GAM. Early-life gut microbiome—the importance of maternal and infant factors in its establishment. Nutr Clin Pract. 2020;35(3):386–405. doi:10.1002/ncp.10490.
  • Khodayar-Pardo P, Mira-Pascual L, Collado MC, Martínez-Costa C. Impact of lactation stage, gestational age and mode of delivery on breast milk microbiota. J Perinatol. 2014;34(8):599–605. doi:10.1038/jp.2014.47.
  • Hahn W, Kim J, Song S, Park S, Kang NM. The human milk oligosaccharides are not affected by pasteurization and freeze-drying. J Matern Fetal Neonatal Med. 2019;32:985–991. doi:10.1080/14767058.2017.1397122.
  • Almeida CC, Mendonça Pereira BF, Leandro KC, Costa MP, Spisso BF, Conte-Junior CA. Bioactive compounds in infant formula and their effects on infant nutrition and health: a systematic literature review. Int J Food Sci. 2021;2021:8850080. doi:10.1155/2021/8850080.
  • Bührer C, Ensenauer R, Jochum F, Kalhoff H, Koletzko B, Lawrenz B, Mihatsch W, Posovszky C, Rudloff S. Infant formulas with synthetic oligosaccharides and respective marketing practices. Mol Cell Pediatr. 2022;9(1):14. doi:10.1186/s40348-022-00147-x.
  • Plows JF, Berger PK, Jones RB, Alderete TL, Yonemitsu C, Najera JA, Khwajazada S, Bode L, Goran MI. Longitudinal changes in Human Milk Oligosaccharides (HMOs) over the course of 24 months of lactation. J Nutr. 2021;151(4):876–882. doi:10.1093/jn/nxaa427.
  • Floris LM, Stahl B, Abrahamse-Berkeveld M, Teller IC. Human milk fatty acid profile across lactational stages after term and preterm delivery: a pooled data analysis. Prostaglandins Leukot Essent Fatty Acids. 2020;156:102023. doi:10.1016/j.plefa.2019.102023.
  • Fidler N, Sauerwald TU, Demmelmair H, Koletzko B. Fat content and fatty acid composition of fresh, pasteurized, or sterilized human milk. Adv Exp Med Biol. 2001;501:485–495 doi:10.1007/978-1-4615-1371-1_60.
  • Chang YC, Chen CH, Lin MC. The macronutrients in human milk change after storage in various containers. Pediatr Neonatol. 2012;53(3):205–209. doi:10.1016/j.pedneo.2012.04.009.
  • Castillo F, Castillo-Ferrer FJ, Cordobilla B, Domingo JC. Inadequate content of Docosahexaenoic Acid (DHA) of donor human milk for feeding preterm infants: a comparison with mother’s own milk at different stages of lactation. Nutrients. 2021;13(4):1300. doi:10.3390/nu13041300.
  • García-Lara NR, Escuder-Vieco D, García-Algar O, de La Cruz J, Lora D, Pallás-Alonso C. Effect of freezing time on macronutrients and energy content of breastmilk. Breastfeed Med. 2012;7(4):295–301. doi:10.1089/bfm.2011.0079.
  • Wegh CAM, Geerlings SY, Knol J, Roeselers G, Belzer C. Postbiotics and their potential applications in early life nutrition and beyond. Int J Mol Sci. 2019;20(19):4673. doi:10.3390/ijms20194673.
  • Scano P, Murgia A, Demuru M, Consonni R, Caboni P. Metabolite profiles of formula milk compared to breast milk. Food Res Int. 2016;87:76–82. doi:10.1016/j.foodres.2016.06.024.
  • Kim SY, Yi DY. Components of human breast milk: from macronutrient to microbiome and microRNA. Clin Exp Pediatr. 2020;63(8):301–309. doi:10.3345/cep.2020.00059.
  • Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Alshaer W, Hasan H, Albakri KA, Alkhafaji E, Issa NN, Al-Holy MA, Abderrahman SM, et al. Immunomodulatory properties of human breast milk: microRNA contents and potential epigenetic effects. Biomed. 2022;10(6):1219. doi:10.3390/biomedicines10061219.
  • Matias-Garcia PR, Wilson R, Mussack V, Reischl E, Waldenberger M, Gieger C, Anton G, Peters A, Kuehn-Steven A. Impact of long-term storage and freeze-thawing on eight circulating microRnas in plasma samples. PLoS One. 2020;15(1):e0227648. doi:10.1371/journal.pone.0227648.
  • Howard KM, Jati Kusuma R, Baier SR, Friemel T, Markham L, Vanamala J, Zempleni J. Loss of miRnas during processing and storage of cow’s (Bos taurus) milk. J Agric Food Chem. 2015;63(2):588–592. doi:10.1021/jf505526w.
  • Moossavi S, Azad MB. Origins of human milk microbiota: new evidence and arising questions. Gut Microbes. 2020;12(1):1667722. doi:10.1080/19490976.2019.1667722.
  • Mantziari A, Rautava S. Factors influencing the microbial composition of human milk. Semin Perinatol. 2021;45(8):151507 doi:10.1016/j.semperi.2021.151507.
  • Fehr K, Moossavi S, Sbihi H, Boutin RCT, Bode L, Robertson B, Yonemitsu C, Field CJ, Becker AB, Mandhane PJ, et al. Breastmilk feeding practices are associated with the co-occurrence of bacteria in mothers’ milk and the infant gut: the CHILD cohort study. Cell Host Microbe. 2020;28(2):285–297. doi:10.1016/j.chom.2020.06.009.
  • Pittard WB, Geddes KM, Brown S, Mintz S, Hulsey TC. Bacterial contamination of human milk: container type and method of expression. Am J Perinatol. 1991;8(1):25–27. doi:10.1055/s-2007-999332.
  • World Health Organization. Guidelines on optimal feeding of low birth-weight infants in low- and middle-income countries. World Health Organization. Geneva: WHO; 2011.
  • Fang MT, Chatzixiros E, Grummer-Strawn L, Engmann C, Israel-Ballard K, Mansen K, O’connor DL, Unger S, Herson M, Weaver G, et al. Developing global guidance on human milk banking. Bull World Health Organ. 2021;99(12):892–900. doi:10.2471/BLT.21.286943.
  • Permanyer M, Castellote C, Ramírez-Santana C, Audí C, Pérez-Cano FJ, Castell M, López-Sabater MC, Franch À. Maintenance of breast milk immunoglobulin a after high-pressure processing. J Dairy Sci. 2010;93(3):877–883. doi:10.3168/jds.2009-2643.
  • Martin CR, Ling PR, Blackburn GL. Review of Infant Feeding: key Features of Breast Milk and Infant Formula. Nutrients. 2016;8(5):279. doi:10.3390/nu8050279.
  • Codex Alimentarius Commission. Standard for infant formula and formulas for special medical purposes intended for infants. Codex Stan 72 - 1981. Rome, Codex Alimentarius Commission; 2007. p. 1–18.
  • Maryniak NZ, Sancho AI, Hansen EB, Bøgh KL. Alternatives to Cow’s Milk-Based Infant Formulas in the Prevention and Management of Cow’s Milk Allergy. Foods. 2022;11(7):926. doi:10.3390/foods11070926.
  • Neumer F, Urraca O, Alonso J, Palencia J, Varea V, Theis S, Rodriguez-palmero M, Moreno-muñoz JA, Guarner F, Veereman G, et al. Long-term safety and efficacy of prebiotic enriched infant formula—a randomized controlled trial. Nutrients. 2021;13(4):1276. doi:10.3390/nu13041276.
  • Azad MB, Nickel NC, Bode L, Brockway M, Brown A, Chambers C, Goldhammer C, Hinde K, McGuire M, Munblit D, et al. Breastfeeding and the origins of health: interdisciplinary perspectives and priorities. Matern Child Nutr. 2021;17(2):e13109. doi:10.1111/mcn.13109.
  • Sanidad KZ, Zeng MY. Neonatal gut microbiome and immunity. Curr Opin Microbiol. 2020;56:30–37. doi:10.1016/j.mib.2020.05.011.
  • Laouar A. Maternal Leukocytes and Infant Immune Programming during Breastfeeding. Trends Immunol. 2020;41(3):225–239. doi:10.1016/j.it.2020.01.005.
  • Cabinian A, Sinsimer D, Tang M, Zumba O, Mehta H, Toma A, Sant’angelo D, Laouar Y, Laouar A. Transfer of Maternal Immune Cells by Breastfeeding: maternal Cytotoxic T Lymphocytes Present in Breast Milk Localize in the Peyer’s Patches of the Nursed Infant. PLoS One. 2016;11(6):e0156762. doi:10.1371/journal.pone.0156762.
  • Arvola M, Gustafsson E, Svensson L, Jansson L, Holmdahl R, Heyman B, Okabe M, Mattsson R. Immunoglobulin-Secreting Cells of Maternal Origin Can Be Detected in B Cell-Deficient Mice1. Biol Reprod. 2000;63(6):1817–1824. doi:10.1095/biolreprod63.6.1817.
  • Field CJ. The Immunological Components of Human Milk and Their Effect on Immune Development in Infants. J Nutr. 2005;135(1):1–4. doi:10.1093/jn/135.1.1.
  • Tuaillon E, Valea D, Becquart P, Al TY, Meda N, Bollore K, van de PP, Vendrell J-P. Human Milk-Derived B Cells: a Highly Activated Switched Memory Cell Population Primed to Secrete Antibodies. J Immunol. 2009;182(11):7155–7162. doi:10.4049/jimmunol.0803107.
  • Atyeo C, Alter G. The multifaceted roles of breast milk antibodies. Cell. 2021;184(6):1486–1499. doi:10.1016/j.cell.2021.02.031.
  • Sefik E, Geva-Zatorsky N, Oh S, Konnikova L, Zemmour D, McGuire AM, Burzyn D, Ortiz-Lopez A, Lobera M, Yang J, et al. Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells. Science. 2015;349(6251):993–997. doi:10.1126/science.aaa9420.
  • Xu M, Pokrovskii M, Ding Y, Yi R, Au C, Harrison OJ, Galan C, Belkaid Y, Bonneau R, Littman DR. C-MAF-dependent regulatory T cells mediate immunological tolerance to a gut pathobiont. Nature. 2018;554(7692):373–377. doi:10.1038/nature25500.
  • Ohnmacht C, Park J-H, Cording S, Wing JB, Atarashi K, Obata Y, Gaboriau-Routhiau V, Marques R, Dulauroy S, Fedoseeva M, et al. The microbiota regulates type 2 immunity through RORγt + T cells. Science. 2015;349(6251):989–993. doi:10.1126/science.aac4263.
  • Sonnenberg GF, Artis D. Innate Lymphoid Cell Interactions with Microbiota: implications for Intestinal Health and Disease. Immunity. 2012;37(4):601–610. doi:10.1016/j.immuni.2012.10.003.
  • Baban B, Malik A, Bhatia J, Yu JC. Presence and Profile of Innate Lymphoid Cells in Human Breast Milk. JAMA Pediatr. 2018;172(6):594–596. doi:10.1001/jamapediatrics.2018.0148.
  • Oddy WH, Rosales F. A systematic review of the importance of milk TGF-β on immunological outcomes in the infant and young child. Pediatr Allergy Immunol. 2010;21:47–59. doi:10.1111/j.1399-3038.2009.00913.x.
  • Sitarik AR, Bobbitt KR, Havstad SL, Fujimura KE, Levin AM, Zoratti EM, Kim H, Woodcroft KJ, Wegienka G, Ownby DR, et al. Breast Milk Transforming Growth Factor β is Associated with Neonatal Gut Microbial Composition. J Pediatr Gastroenterol Nutr. 2017;65(3):e60–e67. doi:10.1097/MPG.0000000000001585.
  • Rautava S, Nanthakumar NN, Dubert-Ferrandon A, Lu L, Rautava J, Walker WA. Breast Milk-Transforming Growth Factor-β2 Specifically Attenuates IL-1β-Induced Inflammatory Responses in the Immature Human Intestine via an SMAD6- and ERK-Dependent Mechanism. Neonatology. 2011;99(3):192–201. doi:10.1159/000314109.
  • Järvinen KM, Suárez-Fariñas M, Savilahti E, Sampson HA, Berin MC. Immune factors in breast milk related to infant milk allergy are independent of maternal atopy. J Allergy Clin Immunol. 2015;135(5):1390–1393. doi:10.1016/j.jaci.2014.10.051.
  • Gersting JA, Christensen RD, Calhoun DA. Effects of enterally administering granulocyte colony-stimulating factor to suckling mice. Pediatr Res. 2004;55(5):802–806. doi:10.1203/01.PDR.0000117846.51197.7C.
  • Aspinall R, Prentice AM, Ngom PT, Ryffel B. Interleukin 7 from Maternal Milk Crosses the Intestinal Barrier and Modulates T-Cell Development in Offspring. PLoS One. 2011;6(6):e20812. doi:10.1371/journal.pone.0020812.
  • Jones-Hall YL, Nakatsu CH. The Intersection of TNF, IBD and the Microbiome. Gut Microbes. 2016;7(1):58–62. doi:10.1080/19490976.2015.1121364.
  • Czosnykowska-Łukacka M, Orczyk-Pawiłowicz M, Broers B, Królak-Olejnik B. Lactoferrin in Human Milk of Prolonged Lactation. Nutrients. 2019;11(10):2350. doi:10.3390/nu11102350.
  • Manzoni P. Clinical Benefits of Lactoferrin for Infants and Children. J Pediatr. 2016;173:S43–52. doi:10.1016/j.jpeds.2016.02.075.
  • Ellison RT, Giehl TJ. Killing of gram-negative bacteria by lactoferrin and lysozyme. J Clin Invest. 1991;88(4):1080–1091. doi:10.1172/JCI115407.
  • Vega-Bautista A, de la Garza M, Carrero JC, Campos-Rodríguez R, Godínez-Victoria M, Drago-Serrano ME. The Impact of Lactoferrin on the Growth of Intestinal Inhabitant Bacteria. Int J Mol Sci. 2019;20(19):4707. doi:10.3390/ijms20194707.
  • Oda H, Wakabayashi H, Yamauchi K, Abe F. Lactoferrin and bifidobacteria. Biometals. 2014;27(5):915–922. doi:10.1007/s10534-014-9741-8.
  • Lönnerdal B, Jiang R, Du X. Bovine lactoferrin can be taken up by the human intestinal lactoferrin receptor and exert bioactivities. J Pediatr Gastroenterol Nutr. 2011;53(6):606–614. https://pubmed.ncbi.nlm.nih.gov/21832946/.
  • Demers-Mathieu V, Huston RK, Markell AM, McCulley EA, Martin RL, Spooner M, Dallas DC. Differences in Maternal Immunoglobulins within Mother’s Own Breast Milk and Donor Breast Milk and across Digestion in Preterm Infants. Nutrients. 2019;11(4):920. doi:10.3390/nu11040920.
  • Takemura T, Eishi Y. Distribution of secretory component and immunoglobulins in the developing lung. Am Rev Respir Dis. 1985;131(1):125–130. doi:10.1164/arrd.1985.131.1.125.
  • Granato A, Chen Y, Wesemann DR. Primary immunoglobulin repertoire development: time and space matter. Curr Opin Immunol. 2015;33:126–131. doi:10.1016/j.coi.2015.02.011.
  • 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. 2022;30(5):650–659. doi:10.1016/j.chom.2022.02.005.
  • Dzidic M, Mira A, Artacho A, Abrahamsson TR, Jenmalm MC, Collado MC, Ebisawa M. Allergy development is associated with consumption of breastmilk with a reduced microbial richness in the first month of life. Pediatr Allergy Immunol. 2020;31(3):250–257. doi:10.1111/pai.13176.
  • Kramer DR, Cebra JJ. Role of maternal antibody in the induction of virus specific and bystander IgA responses in peyer’s patches of suckling mice. Int Immunol. 1995;7(6):911–918. doi:10.1093/intimm/7.6.911.
  • Ramanan D, Sefik E, Galván-Peña S, Wu M, Yang L, Yang Z, Kostic A, Golovkina TV, Kasper DL, Mathis D, et al. An Immunologic Mode of Multigenerational Transmission Governs a Gut Treg Setpoint. Cell. 2020;181(6):1276–1290. doi:10.1016/j.cell.2020.04.030.
  • Koch MA, Reiner GL, Lugo KA, Kreuk LSM, Stanbery AG, Ansaldo E, Seher TD, Ludington WB, Barton GM. Maternal IgG and IgA Antibodies Dampen Mucosal T Helper Cell Responses in Early Life. Cell. 2016;165(4):827–841. doi:10.1016/j.cell.2016.04.055.
  • Thai JD, Gregory KE. Bioactive Factors in Human Breast Milk Attenuate Intestinal Inflammation during Early Life. Nutrients. 2020;12(2):581. doi:10.3390/nu12020581.
  • Rogier EW, Frantz AL, Bruno MEC, Wedlund L, Cohen DA, Stromberg AJ, Kaetzel CS Secretory antibodies in breast milk promote long-term intestinal homeostasis by regulating the gut microbiota and host gene expression. Proc Natl Acad Sci USA. 2014;111(8):3074–3079. doi:10.1073/pnas.1315792111.
  • Kalbermatter C, Fernandez Trigo N, Christensen S, Ganal-Vonarburg SC. Maternal microbiota, early life colonization and breast milk drive immune development in the newborn. Front Immunol. 2021;12:683022. doi:10.3389/fimmu.2021.683022.
  • Stinson LF, Trevenen ML, Geddes DT. The viable microbiome of human milk differs from the metataxonomic profile. Nutrients. 2021;13(12):4445. doi:10.3390/nu13124445.
  • Moossavi S, Sepehri S, Robertson B, Bode L, Goruk S, Field CJ, Lix LM, de Souza RJ, Becker AB, Mandhane PJ, et al. Composition and variation of the human milk microbiota are influenced by maternal and early-life factors. Cell Host Microbe. 2019;25(2):324–335.e4. doi:10.1016/j.chom.2019.01.011.
  • Treven P, Mahnič A, Rupnik M, Golob M, Pirš T, Matijašić BB, Lorbeg PM. Evaluation of human milk microbiota by 16S rRNA Gene Next-Generation Sequencing (NGS) and Cultivation/MALDI-TOF mass spectrometry identification. Front Microbiol. 2019;10:2612. doi:10.3389/fmicb.2019.02612.
  • Piñeiro-Ramos JD, Parra-Llorca A, Ten-Doménech I, Gormaz M, Ramón-Beltrán A, Cernada M, Quintás G, Collado MC, Kuligowski J, Vento M. Effect of donor human milk on host-gut microbiota and metabolic interactions in preterm infants. Clin Nutr. 2021;40(3):1296–1309. doi:10.1016/j.clnu.2020.08.013.
  • Cacho NT, Harrison NA, Parker LA, Padgett KA, Lemas DJ, Marcial GE, Li N, Carr LE, Neu J, Lorca GL. Personalization of the microbiota of donor human milk with mother’s own milk. Front Microbiol. 2017;8:1470. doi:10.3389/fmicb.2017.01470.
  • Marin-Gómez W, Grande MJ, Pérez-Pulido R, Galvez A, Lucas R. Changes in the bacterial diversity of human milk during late lactation period (weeks 21 to 48). Foods. 2020;9(9):1184. doi:10.3390/foods9091184.
  • Gil-Campos M, López MÁ, Rodriguez-Benítez MV, Romero J, Roncero I, Linares MD, Maldonado J, López-Huertas E, Berwind R, Ritzenthaler KL, et al. Lactobacillus fermentum CECT 5716 is safe and well tolerated in infants of 1–6 months of age: a randomized controlled trial. Pharmacol Res. 2012;65(2):231–238. doi:10.1016/j.phrs.2011.11.016.
  • Thongaram T, Hoeflinger JL, Chow JM, Miller MJ. Human milk oligosaccharide consumption by probiotic and human-associated bifidobacteria and lactobacilli. J Dairy Sci. 2017;100(10):7825–7833. doi:10.3168/jds.2017-12753.
  • Chong HY, Tan LTH, Law JWF, Hong KW, Ratnasingam V, Ab Mutalib NS, Lee LH, Letchumanan V. Exploring the potential of human milk and formula milk on infants’ gut and health. Nutrients. 2022;14(17):3554. doi:10.3390/nu14173554.
  • Maldonado J, Cañabate F, Sempere L, Vela F, Sánchez AR, Narbona E, López-Huertas E, Geerlings A, Valero AD, Olivares M, et al. Human milk probiotic lactobacillus fermentum CECT5716 reduces the incidence of gastrointestinal and upper respiratory tract infections in infants. J Pediatr Gastroenterol Nutr. 2012;54(1):55–61. doi:10.1097/MPG.0b013e3182333f18.
  • Chi C, Xue Y, Liu R, Wang Y, Lv N, Zeng H, Buys N, Zhu B, Sun J, Yin C. Effects of a formula with a probiotic Bifidobacterium lactis supplement on the gut microbiota of low birth weight infants. Eur J Nutr. 2019;59(4):1493–1503. doi:10.1007/s00394-019-02006-4.
  • Esaiassen E, Hjerde E, Cavanagh JP, Pedersen T, Andresen JH, Rettedal SI, Støen R, Nakstad B, Willassen NP, Klingenberg C. Effects of probiotic supplementation on the gut microbiota and antibiotic resistome development in preterm infants. Front Pediatr. 2018;6:347. doi:10.3389/fped.2018.00347.
  • Doron S, Snydman DR. Risk and safety of probiotics. Clin Infect Dis. 2015;60:S129–S134. doi:10.1093/cid/civ085.
  • Quin C, Estaki M, Vollman DM, Barnett JA, Gill SK, Gibson DL. Probiotic supplementation and associated infant gut microbiome and health: a cautionary retrospective clinical comparison. Sci Rep. 2018;8(1):8283. doi:10.1038/s41598-018-26423-3.
  • Samara J, Moossavi S, Alshaikh B, Ortega VA, Pettersen VK, Ferdous T, Hoops SL, Soraisham A, Vayalumkal J, Dersch-Mills D, et al. Supplementation with a probiotic mixture accelerates gut microbiome maturation and reduces intestinal inflammation in extremely preterm infants. Cell Host Microbe. 2022;30(5):696–711.e5. doi:10.1016/j.chom.2022.04.005.
  • Bode L. Human milk oligosaccharides: every baby needs a sugar mama. Glycobiology. 2012;22(9):1147–1162. doi:10.1093/glycob/cws074.
  • Thum C, Wall CR, Weiss GA, Wang W, Szeto IMY, Day L. Changes in HMO concentrations throughout lactation: influencing factors, health effects and opportunities. Nutri. 2021;13(7):2272. doi:10.3390/nu13072272.
  • Azad MB, Robertson B, Atakora F, Becker AB, Subbarao P, Moraes TJ, Mandhane PJ, Turvey SE, Lefebvre DL, Sears MR, et al. Human milk oligosaccharide concentrations are associated with multiple fixed and modifiable maternal characteristics, environmental factors, and feeding practices. J Nutr. 2018;148(11):1733–1742. doi:10.1093/jn/nxy175.
  • Arslanoglu S, Moro GE, Boehm G. Early supplementation of prebiotic oligosaccharides protects formula-fed infants against infections during the first 6 months of life. J Nutr. 2007;137(11):2420–2424. doi:10.1093/jn/137.11.2420.
  • Meli F, Puccio G, Cajozzo C, Ricottone GL, Pecquet S, Sprenger N, Steenhout P. Growth and safety evaluation of infant formulae containing oligosaccharides derived from bovine milk: a randomized, double-blind, noninferiority trial. BMC Pediatr. 2014;14:306. doi:10.1186/s12887-014-0306-3.
  • Vandenplas Y, Berger B, Carnielli VP, Ksiazyk J, Lagström H, Luna MS, Migacheva N, Mosselmans JM, Picaud JC, 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.
  • Lee H, Padhi E, Hasegawa Y, Larke J, Parenti M, Wang A, Hernell O, Lönnerdal B, Slupsky C. Compositional dynamics of the milk fat globule and its role in infant development. Front Pediatr. 2018;6:313. doi:10.3389/fped.2018.00313.
  • Calder PC. Functional roles of fatty acids and their effects on human health. J Parenter Enteral Nutr. 2015;39:18S–32S. doi:10.1177/0148607115595980.
  • Moossavi S, Atakora F, Miliku K, Sepehri S, Robertson B, Duan QL, Becker AB, Mandhane PJ, Turvey SE, Moraes TJ, et al. Integrated analysis of human milk microbiota with oligosaccharides and fatty acids in the child cohort. Front Nutr. 2019;6:58. doi:10.3389/fnut.2019.00058.
  • Churchward CP, Alany RG, Snyder LAS. Alternative antimicrobials: the properties of fatty acids and monoglycerides. Crit Rev Microbiol. 2018;44(5):561–570. doi:10.1080/1040841X20181467875.
  • Perez-Burillo S, Rajakaruna S, Paliy O. Growth of Bifidobacterium species is inhibited by free fatty acids and bile salts but not by glycerides. AIMS Microbiol. 2022;8(1):53–60. doi:10.3934/microbiol.2022005.
  • Bobiński R, Bobińska J. Fatty acids of human milk – a review. Int J Vitam Nutr Res. 2020;92(3-4):280–291. doi:10.1024/0300-9831/a000651.
  • Laitinen K, Hoppu U, Hämäläinen M, Linderborg K, Moilanen E, Isolauri E. Breast milk fatty acids may link innate and adaptive immune regulation: analysis of soluble CD14, prostaglandin E2, and fatty acids. Pediatr Res. 2006;59(5):723–727. doi:10.1203/01.pdr.0000203158.31452.9e.
  • Harbige LS. Fatty acids, the immune response, and autoimmunity: a question of n−6 essentiality and the balance between n−6 and n−3. Lipids. 2003;38(4):323–341. doi:10.1007/s11745-003-1067-z.
  • Walker WA, Iyengar RS. Breast milk, microbiota, and intestinal immune homeostasis. Pediatr Res. 2015;77(1):220–228. doi:10.1038/pr.2014.160.
  • Zhang L, Qu J, Huppertz T, Liu J, Sun Z, Zhou P. Effects of different freeze-thaw processes on the bioactivity and digestibility of human milk. LWT. 2022;156:113025. doi:10.1016/j.lwt.2021.113025.
  • Kent G. Regulating fatty acids in infant formula: critical assessment of U.S. policies and practices. Int Breastfeed J. 2014;9(1):2. doi:10.1186/1746-4358-9-2.
  • Looijesteijn E, Brouwer RWW, Schoemaker RJW, Ulfman LH, Ham SL, Jeurink P, Karaglani E, van IJcken WFJ, Manios Y. Effect of bovine milk fat-based infant formulae on microbiota, metabolites and stool parameters in healthy term infants in a randomized, crossover, placebo-controlled trial. BMC Nutr. 2022;8(1):93. doi:10.1186/s40795-022-00575-y.
  • Schmelzle H, Wirth S, Skopnik H, Radke M, Knol J, Böckler HM, Wells J, Fusch C. Randomized double-blind study of the nutritional efficacy and bifidogenicity of a new infant formula containing partially hydrolyzed protein, a high β-palmitic acid level, and nondigestible oligosaccharides. J Pediatr Gastroenterol Nutr. 2003;36(3):343–351. doi:10.1097/00005176-200303000-00008.
  • Żółkiewicz J, Marzec A, Ruszczyński M, Feleszko W. Postbiotics—a step beyond pre- and probiotics. Nutrients. 2020;12(8):2189. doi:10.3390/nu12082189.
  • Kataria J, Li N, Wynn JL, Neu J. Probiotic microbes: do they need to be alive to be beneficial? Nutr Rev. 2009;67:546–550. doi:10.1111/j.1753-4887.2009.00226.x.
  • Sugahara H, Yao R, Odamaki T, Xiao JZ. Differences between live and heat-killed bifidobacteria in the regulation of immune function and the intestinal environment. Benef Microbes. 2017;8(3):463–472. doi:10.3920/BM2016.0158.
  • Zagato E, Mileti E, Massimiliano L, Fasano F, Budelli A, Penna G, Rescigno M . Lactobacillus paracasei CBA L74 metabolic products and fermented milk for infant formula have anti-inflammatory activity on dendritic cells in vitro and protective effects against colitis and an enteric pathogen in vivo. PLoS One. 2014;9(2):e87615. doi:10.1371/journal.pone.0087615.
  • Beghetti I, Barone M, de Fazio L, Laderchi E, Biagi E, Turroni S, Brigidi P, Pession A, Corvaglia L, Aceti A. A pilot study on donor human milk microbiota: a comparison with preterm human milk microbiota and the effect of pasteurization. Nutrients. 2022;14(12):2483. doi:10.3390/nu14122483.
  • Al Nabhani Z, Dulauroy S, Marques R, Cousu C, Al Bounny S, Déjardin F, Sparwasser T, Bérard M, Cerf-Bensussan N, Eberl G. A weaning reaction to microbiota is required for resistance to immunopathologies in the adult. Immunity. 2019;50(5):1276–1288.e5. doi:10.1016/j.immuni.2019.02.014.
  • Kulkarni DH, Gustafsson JK, Knoop KA, McDonald KG, Bidani SS, Davis JE, Floyd AN, Hogan SP, Hsieh C-S, Newberry RD. Goblet cell associated antigen passages support the induction and maintenance of oral tolerance. Mucosal Immunol. 2020;13(2):271–282. doi:10.1038/s41385-019-0240-7.
  • Kulkarni DH, McDonald KG, Knoop KA, Gustafsson JK, Kozlowski KM, Hunstad DA, Miller MJ, Newberry RD. Goblet cell associated antigen passages are inhibited during Salmonella typhimurium infection to prevent pathogen dissemination and limit responses to dietary antigens. Mucosal Immunol. 2018;11(4):1103–1113. doi:10.1038/s41385-018-0007-6.
  • Knoop KA, Coughlin PE, Floyd AN, Ndao IM, Hall-Moore C, Shaikh N, Gasparrini AJ, Rusconi B, Escobedo M, Good M, et al. Maternal activation of the EGFR prevents translocation of gut-residing pathogenic Escherichia coli in a model of late-onset neonatal sepsis. Proc Natl Acad Sci U S A. 2020;117(14):7941–7949. doi:10.1073/pnas.1912022117.
  • Knoop KA, Newberry RD. Goblet cells: multifaceted players in immunity at mucosal surfaces. Mucosal Immunol. 2018;11(6):1551–1557. doi:10.1038/s41385-018-0039-y.
  • Clark JA, Doelle SM, Halpern MD, Saunders TA, Holubec H, Dvorak K, Boitano SA, Dvorak B. Intestinal barrier failure during experimental necrotizing enterocolitis: protective effect of EGF treatment. Am J Physiol Gastrointest Liver Physiol. 2006;291(5):G938–G949. doi:10.1152/ajpgi.00090.2006.
  • Siggers J, Sangild PT, Jensen TK, Siggers RH, Skovgaard K, Støy ACF, Jensen BB, Thymann T, Bering SB, Boye M. Transition from parenteral to enteral nutrition induces immediate diet-dependent gut histological and immunological responses in preterm neonates. Am J Physiol Gastrointest Liver Physiol. 2011;301(3):G435–G445. doi:10.1152/ajpgi.00400.2010.
  • Choi SW, Friso S. Epigenetics: a new bridge between nutrition and health. Adv Nutri. 2010;1(1):8–16. doi:10.3945/an.110.1004.
  • Indrio F, Martini S, Francavilla R, Corvaglia L, Cristofori F, Mastrolia SA, Neu J, Rautava S, Russo Spena G, Raimondi F, et al. Epigenetic matters: the link between early nutrition microbiome, and long-term health development. Front Pediatr. 2017;5:178. doi:10.3389/fped.2017.00178.
  • Willems R, Krych L, Rybicki V, Jiang P, Sangild PT, Shen RL, Hensel KO, Wirth S, Postberg J, Jenke AC. Introducing enteral feeding induces intestinal subclinical inflammation and respective chromatin changes in preterm pigs. Epigenomics. 2015;7(4):553–565. doi:10.2217/epi.15.13.
  • Minekawa R, Takeda T, Sakata M, Hayashi M, Isobe A, Yamamoto T, Tasaka K, Murata Y. Human breast milk suppresses the transcriptional regulation of IL-1β-induced NF-κB signaling in human intestinal cells. Am J Physiol Cell Physiol. 2004;287(5):C1404–C1411. doi:10.1152/ajpcell.00471.2003.
  • Melnik BC, John SM, Carrera-Bastos P, Schmitz G. Milk: a postnatal imprinting system stabilizing FoxP3 expression and regulatory T cell differentiation. Clin Transl Allergy. 2016;6:18. doi:10.1186/s13601-016-0108-9.
  • Liu T, Zhang L, Joo D, Sun S-C. NF-κB signaling in inflammation. Signal Transuct Target Ther. 2017;2:17023–. doi:10.1038/sigtrans.2017.23.
  • Xu J, Shin J, McGee M, Unger S, Bando N, Sato J, Vandewouw M, Patel Y, Branson HM, Paus T, et al. Intake of mother’s milk by very-low-birth-weight infants and variation in DNA methylation of genes involved in neurodevelopment at 5.5 years of age. Am J Clin Nutr. 2022;116(4):1038–1048. doi:10.1093/ajcn/nqac221.
  • Benmoussa A, Provost P. Milk MicroRNAs in health and disease. Compr Rev Food Sci Food Saf. 2019;18(3):703–722. doi:10.1111/1541-4337.12424.
  • Alsaweed M, Lai CT, Hartmann PE, Geddes DT, Kakulas F, Zheng Y. Human milk cells and lipids conserve numerous known and novel miRnas, some of which are differentially expressed during lactation. PLoS One. 2016;11(4):e0152610. doi:10.1371/journal.pone.0152610.
  • Vélez-Ixta JM, Benítez-Guerrero T, Aguilera-Hernández A, Martínez-Corona H, Corona-Cervantes K, Juárez-Castelán CJ, Rangel-Calvillo MN, García-Mena J. Detection and quantification of immunoregulatory miRnas in human milk and infant milk formula. Biotech (Basel). 2022;11(2):11. doi:10.3390/biotech11020011.
  • Cortese R, Lu L, Yu Y, Ruden D, Claud EC. Epigenome-microbiome crosstalk: a potential new paradigm influencing neonatal susceptibility to disease. Epigenetics. 2016;11(3):205–215. doi:10.1080/15592294.2016.1155011.
  • Fofanova TY, Petrosino JF, Kellermayer R. Microbiome–epigenome interactions and the environmental origins of inflammatory bowel diseases. J Pediatr Gastroenterol Nutr. 2016;62(2):208–219. doi:10.1097/MPG.0000000000000950.
  • Zhang Q, Cao X. Epigenetic regulation of the innate immune response to infection. Nat Rev Immunol. 2019;19(7):417–432. doi:10.1038/s41577-019-0151-6.
  • Takahashi K, Sugi Y, Nakano K, Tsuda M, Kurihara K, Hosono A, Kaminogawa S. Epigenetic control of the host gene by commensal bacteria in large intestinal epithelial cells. J Biol Chem. 2011;286(41):35755–35762. doi:10.1074/jbc.M111.271007.
  • Nagy-Szakal D, Kellermayer R. Colonic mucosal DNA methylation, immune response, and microbiome patterns in Toll-like receptor 2-knockout mice. Gut Microbes. 2011;2(3):178–182. doi:10.4161/gmic.2.3.16107.
  • Sharma M, Li Y, Stoll ML, Tollefsbol TO. The epigenetic connection between the gut microbiome in obesity and diabetes. Front Genet. 2020;10:1329. doi:10.3389/fgene.2019.01329.
  • Uozaki H, Fukayama M. Epstein-Barr virus and gastric carcinoma–viral carcinogenesis through epigenetic mechanisms. Int J Clin Exp Pathol. 2008;1(3):198–216.