Next Article in Journal
Influence of Dietary Habits and Mediterranean Diet Adherence on Sleep Quality during Pregnancy. The GESTAFIT Project
Previous Article in Journal
Association between Serum Vitamin B12 and Global DNA Methylation in Colorectal Cancer Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

In Love with Shaping You—Influential Factors on the Breast Milk Content of Human Milk Oligosaccharides and Their Decisive Roles for Neonatal Development

Wacker Chemie AG, Biosolutions, Hanns-Seidel-Platz 4, 81737 München, Germany
*
Author to whom correspondence should be addressed.
Submission received: 12 October 2020 / Revised: 8 November 2020 / Accepted: 17 November 2020 / Published: 20 November 2020
(This article belongs to the Section Nutrition and Public Health)

Abstract

:
Human milk oligosaccharides (HMOs) are structurally versatile sugar molecules constituting the third major group of soluble components in human breast milk. Based on the disaccharide lactose, the mammary glands of future and lactating mothers produce a few hundreds of different HMOs implicating that their overall anabolism utilizes rather high amounts of energy. At first sight, it therefore seems contradictory that these sugars are indigestible for infants raising the question of why such an energy-intensive molecular class evolved. However, in-depth analysis of their molecular modes of action reveals that Mother Nature created HMOs for neonatal development, protection and promotion of health. This is not solely facilitated by HMOs in their indigestible form but also by catabolites that are generated by microbial metabolism in the neonatal gut additionally qualifying HMOs as natural prebiotics. This narrative review elucidates factors influencing the HMO composition as well as physiological roles of HMOs on their way through the infant body and within the gut, where a major portion of HMOs faces microbial catabolism. Concurrently, this work summarizes in vitro, preclinical and observational as well as interventional clinical studies that analyzed potential health effects that have been demonstrated by or were related to either human milk-derived or synthetic HMOs or HMO fractions.

Graphical Abstract

1. Structure of Human Milk Oligosaccharides and Human Milk Content

Human milk oligosaccharides (HMOs) are lactose-derived molecules constituting a major part of human breast milk. Even though they are highly varied in structure, the oligosaccharides are composed of only five building blocks namely glucose (Glc), galactose (Gal), fucose (Fuc), N-acetylglucosamine (GlcNAc) and N-acetylneuraminic acid (NANA, Figure 1a).
Depending on their fucose and L-N-acetylneuraminic acid (=sialic acid) content, HMOs can be further divided into the three groups of neutral fucosylated, neutral non-fucosylated and acidic sialylated HMOs. Based on studies that performed absolute quantification, these respectively make up 35–77%, 11–30% and 4–27% [1,2,3,4,5] of total HMOs and overall form nearly 200 different molecules [2]. Note that some studies state that more than 200 HMOs have already been identified. However, to the best of our knowledge, this has not been shown so far.
The mean total HMO content in human milk ranges from 4 g/L [6] to 22 g/L (Table 1), while individual mothers might produce even greater amounts of up to more than 30 g/L [5,7]. This high variability is due to several intrinsic and extrinsic factors that differ drastically between individual mother/infant pairs.
One of the most important intrinsic factors is the genetic predisposition of so-called glycosyltransferases, namely α-1,2-fucosyltransferase (FUT2) and α-1,3/4-fucosyltransferase (FUT3). These enzymes link fucose to terminal galactose residues or subterminal GlcNAc residues while different expression activities of the underlying Secretor (Se) and Lewis (Le) genes generally result in the four phenotypes frequently called milk groups: Se+Le+, Se+Le, SeLe+ and SeLe (Figure 1b).
Studies that differentiated mothers according to their milk group by means of breastmilk analysis are summarized in Table 2. Secretor- and Lewis-positive (Se+Le+) mothers have been shown to be the most dominant group (45–77%) in several populations from Europe, Asia and Africa. The second most dominant group is the non-Secretor Lewis-positive group (SeLe+, 7–34%), followed by the Secretor Lewis-negative group (Se+Le, 4–28%) and the non-Secretor Lewis-negative group (SeLe, 1–26%) [10].
In addition to this grouping, fucosyltransferases other than FUT2/3 seem to influence the HMO composition, as milk of Secretor- and Lewis-deficient mothers also contains small amounts of fucosylated HMOs [7,11,12]. This is in agreement with van Leeuwen et al., who have recently suggested that the fucosyltransferase FucTx is responsible for regional variations of the HMO composition of Lewis-negative mothers in Vietnam [13].
Newborns rapidly develop during their first months of life requiring variable nutritional intake [14]. A great number of studies have analyzed the HMO concentration during the course of lactation [5,6,8,9,12,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37]. However, only a few performed a quantification of the absolute HMO content (Table 1). Based on these studies, the mean concentration of total indigestible oligosaccharides decreases with progressive infant age regardless of the Secretor status (Figure 2). In colostrum (day 1–5) and transition milk (day 6–14), the sugars respectively make up 16–22 g/L [5,8] and 14–20 g/L [6,9] while in mature milk, the HMO concentration steadily decreases by about 38% to 65% compared to early milk (22 g/L vs. 8–13 g/L measured 18–20 weeks after birth [5,9]). In fact, the decline in concentration applies for all three different types of HMOs, while the lactose concentration is subject to an overall increase [6,9,31] On analysis of the three different HMO types, Xu et al. and Elwakiel et al. reported a decrease of neutral fucosylated and sialylated HMOs by 44–61% and 65–66% for the period of 18–20 weeks, while the neutral non-fucosylated HMO content declines most drastically by 77–86% (Figure 2) [4,5].
Obviously, the analysis of the absolute HMO content does not give detailed information about the change of individual HMOs with regard to different factors. To address such questions, a good number of studies have therefore focused on the quantification of the most important milk sugars [7,12,17,20,28,29,42,43,46,47,48]. These have for instance shown that 3FL increases up to almost threefold throughout the course of lactation regardless of the milk status and gestational age [7,12,17,28,29,32,43,47,48].
In addition, HMOs such as 6′-sialyllactose (6′SL), difucosyllacto-N-hexaose a (DFLNHa), disialyllacto-N-tetraose (DSLNT), lacto-N-tetraose (LNT), and fucosyllacto-N-hexaose II (F-LNH II) are subject to decrease. However, unlike other HMOs whose maximum is reported in colostrum, their maximum is found in late transition (week 2) or early mature milk (week 3) [12,43]. An overview of the most dominant HMOs found in term-delivering mothers of the four different milk groups is given in Figure 3. Raw data derived from the respective studies (Supplementary Table S1) are given in Supplementary Tables S2–S5. The underlying pooled mean values were derived from the studies that differentiated mothers of the four milk groups [7,17,18,29,41,43,44,49]. Respective mean values and pooled mean values for individual HMOs of mothers delivering preterm are given in Supplementary Tables S6–S9. Note that Coppa et al. 1999 and 2011 [17,41] reported high concentrations of LNDFH II and TFLNH (>2 g/L) in milk of Italian mothers. However, as these observations have not been observed independently, the concentrations of mentioned HMOs were not taken into account in Figure 3. For further discussion on the concentrations of LNDFH II and TFLNH, the reader is referred to Thurl et al. 2017 [50] who systematically reviewed the concentration of 33 oligosaccharides and compared the milk of mothers of different gestational ages. In fact, they show that the total concentration of neutral and acidic HMOs is respectively 28% higher and 54% lower in term compared to preterm mothers. Despite the lower neutral HMO concentration in preterm mothers, the concentrations of individual HMOs such as 2′FL, DF-LNH II LNT and F-LNH-II are higher than in term mothers. Conversely, all acidic HMOs under investigation were found at higher concentrations in preterm compared to term mothers [50].
Genetic predisposition, course of lactation and gestational age predominantly define the interindividual variability of human milk sugars. However, several recent studies have identified maternal [5,12,30,37,44,51,52] and infant [34,37,44,53] characteristics, as well as mode of delivery [12,37], parity [12,37,44,54], environmental [51], geographical [19,51,54,55], regional [13,51,54] and seasonal [34,54] factors to be responsible for differences in the composition of human milk sugars.

2. HMOs Shaping the Neonate

Human milk oligosaccharides already circulate in the blood of pregnant mothers at least from week 10 on [56], reaching the fetus through the amniotic fluid [57] and likely through the umbilical cord passing the placenta [58].
During the first month of life, infants consume about 500–650 mL breast milk per day, which rises to around 750–800 mL after four to five months of lactation [59]. This corresponds to absolute amounts of about 7–14 g and 6–10 g, respectively (calculated from mean values given in Figure 2).
Clearly, HMOs are consumed orally, passing the upper gastrointestinal tract, and occasionally reach the nasopharynx as well as the upper respiratory tract through aspiration. The sugars are resistant to the main digestion mechanisms of the infant such as low pH in the stomach as well as salivary, pancreatic and brush boarder enzymes and thus arrive at the distal small intestine and the colon only minimally digested (<5%) [60]. In numbers, 40–50% of HMOs are excreted in the feces [61], 1–5% are absorbed into the blood stream and excreted in the urine [62,63,64], while the remaining portion is prone to microbial catabolism. However, note that the digestion of HMOs also changes during the course of lactation. Thus, it has for instance been shown that they disappear from the neonatal feces once feeding other than mother’s milk is introduced [11].
Both, intact and catabolized HMOs fulfill crucial tasks for neonatal protection and development. Most strikingly, they support and concurrently shape the neonatal immune system by various modes of action, including pathogen protection, maturation of the gut microbiome, fostering of the intestinal barrier function and maturation of immune cells. In the following, in vitro, preclinical and clinical studies demonstrating beneficial effects on the neonatal development by either human milk-derived or synthetic HMOs or HMO fractions are summarized.

2.1. Pathogen Protection

Pathogens gain access to the human body through various ports of entry including the mucosa of the gastrointestinal, respiratory, reproductive and uroepithelial tract as well as through the skin after injury. Physical contact between host cells and many pathogens or their toxins is mediated by carbohydrate-binding proteins. These proteins (or protein motifs) are called lectins and recognize specific moieties of surface-exposed glycans either on host or pathogen cells, which resemble HMO fragments or individual HMOs. Thus, indigestible mother milk sugars act as soluble decoy receptors that interfere with the lectin-glycan association for pathogens such as Entaboeba histolytica [65], Campylobacter jejuni [66,67], Clostridium difficile [68,69], enterohaemorrhagic [70], entereopathogenic [71,72,73,74,75,76,77], enterotoxic [78], uropathogenic [78,79,80] Escherichia coli [81], Helicobacter pylori [82], L. monocytogenes [83], Neisseria Meningitidis C [84], Pseudomonas aeruginosa [75,85,86], Salmonella enterica [74,75], Staphylococcus aureus [87], Vibrio cholerae [70,74,88], human immunodeficiency virus [89], influenza virus [90,91], norovirus [92,93,94] and respiratory syncytial virus [95]. Table 3 summarizes HMOs that have been shown to block pathogen adhesion in vitro. In addition, in silico studies indicate that 3′SL and 6′SL are decoy receptors for polyomavirus VP1 [96,97], but this has not been proven in vitro so far.
Besides the competitive binding activity of HMOs to lectins, a couple of studies indicate different modes of action. For instance, Gonia et al. have shown that hyphal morphogenesis of C. albicans is disturbed by milk oligosaccharides on the cellular and gene expression level thus inhibiting pathogen-host interactions [98]. Angeloni et al. used microarray glycoprofiling and proved that 3′SL was able to modulate the glycocalyx by reduced expression of sialyltransferases, thus likely diminishing enteropathogenic E. coli adhesion sites [73]. Chen et al. showed that HMO pretreatment globally alters host signaling pathways resulting in differential subcellular localizations of L. monocytogenes [83]. L. monocytogene allowing an easier pathogen clearance. [87] Jantscher-Krenn et al. and Lin et al. presented data on the cytoprotective effect, respectively, against Entaboeba histolytica E. hystolytica [65] and uropathogenic E. coli [80], which has been related to antiapoptotic and anti-inflammatory properties of HMOs [80]. Rather recent research has attributed HMOs with antimicrobial activities against specific strains of Streptococcus agalactiae and Acinetobacter baumannii as well as antibiofilm activities against strains of Streptococcus agalactiae and Staphylococcus aureus. It was hypothesized that promiscuous incorporation of HMOs into the capsular polysaccharide and peptidoglycan/glycan binding proteins of the cell walls increases the overall membrane permeability and disturbs biofilm production [87,99,100,101,102,103]. The antimicrobial activity against individual S. agalactiae strains was shown for LNT, LNnT [104], sialylated variants of LNT [103] and fucosylated HMOs such as LNFP I–III [104] and LNDFH I [99]. This might explain why Lewis-positive mothers and their infants are less likely to be colonized with S. agalactiae and why colonization clearance in these infants has a higher probability than for infants born to Lewis-negative women [99].
Some preclinical and clinical studies corroborate the protective effects of HMOs against a few pathogens displayed in Table 3. In general, higher HMO levels in mother’s milk represented by LNFP II appear to be associated with fewer respiratory and enteric problems of newborn humans [105].
A significant reduction of C. jejuni colonization and intestinal clearance in mice has been achieved by neutral human milk oligosaccharides and 2′FL or if dams of suckling pups have been transfected with the human α1,2-fucosyltransferase gene [66]. In addition, Yu et al. demonstrated that 2′FL was able to attenuate intestinal inflammation and induction of inflammatory signaling molecules [67]. Morrow et al. analyzed Mexican mother-infant pairs and detected a positive correlation of 2′FL and LNDFH-I concentrations and the reduction of incidence and severity of diarrhea respectively related to C. jejuni and caliciviruses, which include the species of noroviruses [25,106].
Moreover, preclinical and clinical data for the protection against HIV and rotavirus exist. In mice and pigs, either 2′FL or HMO mixtures containing 2′FL, LNnT, 6′SL, 3′SL and sialic acid diminished rotavirus binding, replication, diarrhea incidence, duration and severity. These were related to a reduced gene expression of viral enterotoxin [107], changes of the gut microbiome and mucosal and systemic increase of immune cells accompanied by a balanced immune response [107,108,109,110,111]. Two observational studies on African mother-infant dyads suggested a reduction of virus transmission from HIV-infected mothers to breastfed infants and a decreased mortality of HIV-exposed babies during breastfeeding. These effects respectively correlated with higher absolute concentrations of HMOs such as LNnT [112] and fucosylated oligosaccharides (2′FL, 3FL, LNFP I–III) [113].
Furthermore, inhibition of colonization of S. pneumoniae in the nasopharynx and lungs of rabbits as well as protection from bacteremia was achieved by LNnT and its sialylated derivatives such as 3′-sialyllacto-N-neotetraose (3′SLNnT). However, 3′SLNnT was for instance not able to protect against acute otitis media related to S. pneumoniae, H. influenzae and M. catarrhalis infection in an interventional clinical study [114]. Similarly, the positive effects of 3′SL to decrease and partly cure H. pylori colonization in rhesus monkeys [115] could not be confirmed in two human interventional studies [116,117].
Idota et al. treated the intestinal loops of rabbits with the subunit B of the V. cholera toxin and observed a significant inhibition of infection-associated fluid accumulation [88]. Steenhout et al. observed a significant reduction of C. difficile toxins A and B in infants fed 2′FL/LNnT-fortified formula compared to infants receiving formula without HMOs [118]. Protection against diarrhea related to the heat-stable toxin of E. coli in mice was conferred by neutral fucosylated HMOs [119]. This was revealed by data of human neonates consuming milk with a higher proportion of 2-linked fucosyloligosaccharides that had less E. coli toxin-associated diarrhea with reduced severity compared to those consuming milk of lower 2-linked fucosyloligosaccharides levels [120]. In addition, the total HMO fraction and 2′FL were respectively able to significantly reduce EPEC colonization [76] and guard against E. coli O157 in mice [121].
Among others [109,111,122], the latter study reveals that the protective function of HMOs is not only due to their structural similarity to receptor antigens. In fact, HMOs act on the gene expression of intestinal cells enhancing the intestinal barrier function, modulate intestinal and systemic immune cells controlling immune maturation and homeostasis, and are associated with establishing a probiotic gut microbiota composition [121]. These effects will be discussed in the following chapters.

2.2. Natural Prebiotics

The development of the intestinal flora is interconnected with metabolic and immune maturation, determining behavior and (patho-)physiology of the host [130,131].
Currently, it is under debate whether the first seed for colonization is already planted in utero, contradicting the traditional notion that this happens during birth [132,133]. In any case, prenatal and perinatal factors such as maternal diet, disease state, biodiversity of the environment, use of antibiotics, mode of delivery and feeding practices “imprint a specific hallmark” on the gut microbiota of babies [132].
Only a few bacterial families within the phyla of Firmicutes and Proteobacteria colonize the immature gastrointestinal tract of babies [134]. With progressive age, these inhabitants are displaced by Bacteroidetes and Actinobacteria leading to a microbial colonization of increased alpha diversity [135,136,137,138,139]. The early successive gut ecology is mainly dominated by Bifidobacterium [34,140,141,142], which can make up more than 90% in breastfed infants compared to about 50% in formula-fed infants [136,143,144]; an effect that is also observed in piglets [131,145]. In part, this is due to initial seeding before and during birth as well as maternal transmission of commensal bacteria present in mother’s milk [133,146]. However, it also indicates prebiotic mother’s milk contents. This is corroborated by the facts that fucosylated HMOs alleviate the depletion of bifidobacteria in C-section born infants [147] and that weaning accelerates microbial aging towards an adult-like composition [131] of noticeable decrease in Bifidobacterium [134,136,148].
The presence of a “bifidus factor” in human milk that supports the growth of commensal bacteria was postulated almost a century ago [149] and was proven to be composed of oligosaccharides [150,151,152,153,154]. A higher total HMO content [155,156] in mothers of milk groups 1, 2 and 3 compared to milk group 4 [41] as well as individual HMOs such as 2′FL [156] correlates with increased amounts of bifidobacteria in neonatal guts. In addition, infants of Secretor mothers establish earlier and more frequent bifidobacterial colonization [157], which appears to positively influence the amount of Bifidobacterium in children even at later ages of 2 to 3 years [158]. An intervention with HMO and 2′FL respectively showed an increase of Actinobacteria in fecal [159] samples and in a semi-continuous colon simulator model that was accounted for by a decrease of Proteobacteria in the latter study [160]. Notably, a microbiota shift towards probiotic dominance was recently achieved by feeding infants with 2′FL/LNnT-supplemented formula for 6 months [161] and even in healthy adults that received 2′FL and/or LNnT for only two weeks [162]. Taking these results together, it appears that Secretor milk is more beneficial for bifidobacterial growth than non-Secretor milk.
In order to elucidate the prebiotic molecular mechanism of HMOs, several bacteria have been tested for their ability to grow on individual or total HMOs as the sole carbon source in vitro. Mainly Bifidobacterium [134,141,157,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183] and individual strains of Bacteroides [167,177], Enterococcus [177], Lactobacillus [177,184], Streptococcus, and Clostridium cluster IV/XIVa [185] are adapted to utilize oligosaccharides. Intriguingly, the extent of neonatal HMO digestion correlates with these genera [139], overall indicating that mother’s milk induces a selective pressure towards commensal gut colonization.
Among HMO utilizers, bifidobacteria co-evolved with humans and developed the most complex pangenome for the digestion of human milk oligosaccharides. This serves efficient protocooperation [141,183,186] within its own but also with other commensal genera [133,187]. In fact, their well-organized use of HMOs was corroborated by De Leoz et al. [137] and Borewicz et al. [142], who showed a negative correlation of fecal HMO concentration and, among other genera, bifidobacterial abundance. However, HMO consumption differs drastically even within the same (sub-)species, which is related to the fact that individual strains evolved a very specific genetic reservoir to fit into defined catabolic niches. Still, either an extra- or an intracellular HMO assimilation strategy can be generally described for the main bifidobacterial species of the neonatal gut, namely, B. bifidum, B. breve, B. longum subsp. longum, B. longum subsp. infantis, B. infantis, B. pseudocatenulatum and B. kashiwanohense [188]. An excellent overview of the genetic repertoire of these species was recently given by Sakanaka et al. [188]. Glycoprofiling of oligosaccharide degradation products and genomic analysis discovered that most B. longum infantis strains are equipped with a whole machinery of enzymes for intracellular catabolism [166,168,169,170,178,179,189]. The relevant genes are highly conserved in a cluster consisting of ATP-binding cassette transporter (ABC) molecules with solute-binding proteins (SBPs) of high HMO affinity and intracellular glycosyl hydrolases such as fucosidases, sialidases, galactosidases and N-acetylglucosaminidases. Similarly, B. breve, B. pseudocatenulatum and B. kashiwanohense [190] also import HMOs for intracellular degradation, but with reduced substrate bandwidth [134,175,176,179]. Furthermore, the majority of B. breve preferably assimilate LNnT, LNT and its degradation product lacto-N-biose (LNB) [169,170,176,179,181], a metabolic pathway that is also conserved in B. pseudocatenulatum [134,169] and B. kashiwanohense [179]. Like B. infantis, most B. bifidum bacteria are able to assimilate most HMOs, but individual strains rather rely on the extracellular assimilation strategy. B. longum strains are capable of disintegrating a limited number of fucosylated [176,188] but mainly non-fucosylated neutral HMOs in the extra- and intracellular space [180]. It thereby shares the possibility with B. bifidum of disintegrating LNT by lacto-N-biosidase, yielding lactose and LNB [169,170,180], which is taken up by both for further catabolism. Intriguingly, some B. bifidum strains altruistically leave extracellular HMO digestion products unconsumed for neighboring species or strains within the bifidobacterial ecosystem [141,183,191]. This cross-feeding strategy is believed to maximize nutrient exploitation to foster the dominance of Bifidobacterium [141].
Commensal gut bacteria whose growth is nurtured by HMOs (Bacteroidetes, Enterococcus, Lactobacillus, Streptococcus, Clostridium cluster IV/XIVa and mainly Bifidobacterium) produce several molecules that are beneficial for infants including B and K vitamins as well as bacteriocins and antivirals [133,192,193,194,195]. Moreover, their carbohydrate catabolism yields multiple metabolites among which short chain fatty acids (SCFAs) like acetate, butyrate and propionate are the most important. In fact, an increase in the number of these commensals concurrent with enhanced fecal SCFA production was achieved by total and individual HMOs such as 2′FL, 3FL and DFL in in vitro fermentation studies of infant feces [159,196,197,198]. Similar observations were made in newborn mice [199], rats [122] and pigs [108,124,145,200] supplemented with LNnt, 2′FL, a HMO mix (2′FL, LNT, LNnT, 6′SL) or total HMOs. SCFAs derived from HMOs exert numerous positive effects on the neonatal host. A detailed description of their molecular modes of action is not within the scope of this review, but key functions should be mentioned. These small organic acids contribute to the host energy metabolism, influencing several organs and providing up to 10% of total caloric requirements and up to 70% of energy for colonocytes thus supporting colonic function and health [201]. They trigger the proliferation and differentiation of colonocytes, enhance the intestinal epithelial barrier function [199] by induction of mucin 2 expression and differentiation of goblet cells, regulate and modulate immune function [202] and are thought to be the key sensing molecules of the microbiota-gut-brain axis affecting development and homeostasis of the central nervous system [194,203,204,205,206,207].
In addition, microbial degradation of HMOs not only yields SCFAs but also lactate and concurrently reduces the intestinal pH [159,197]. On the one hand, this facilitates the uptake of essential nutrients by the host. On the other hand, SCFAs are toxic to several detrimental bacteria in their nonionized form prevalent at low pH [208]. This HMO-mediated competitive exclusion of pathogens is even further promoted by milk oligosaccharides such as 3′SL and 6′SL that induce a transcriptomic and physiological response of some bifidobacterial strains for a physical contact with intestinal cells [209,210,211,212]. At this interface, commensals directly influence intestinal function and shape the architecture of the gastrointestinal immune system [211] by stimulating the maintenance of the sterile mucus layer [213], immunomodulatory cytokines, the epithelial secretion of antimicrobial peptides and the gastrointestinal lymphoid tissue [138].
This chapter revealed that crucial processes of neonatal development are tightly associated with the establishment of a healthy gut microbiota nourished by oligosaccharides from human milk. However, HMOs themselves not only act as natural prebiotics and soluble decoy receptors for pathogens. They are even able to directly affect critical physiological processes such as maturation and maintenance of the intestinal barrier function as well as immune development and homeostasis; topics that will be covered in the next section.

2.3. Maturation and Maintenance of the Intestinal Barrier and Function

Intestinal epithelial cells form a physical barrier in the gastrointestinal tract and are in close cooperation with commensal microbiota, immune and stromal cells to defend the body from pathogens at the forefront. Multipotent stems cells continuously differentiate from the crypt along the villus axis into nutrient-absorbing enterocytes, secretory goblet, enteroendocrine and Paneth cells as well as microfold and tuft cells that communicate with the intestinal immune system. This cellular barrier is reinforced by tight junctions as well as the glycocalyx and the mucus layer. Tight junctions control the paracellular permeability of the epithelial surface, while the glycocalyx, a complex meshwork of glycolipids and glycoproteins, is responsible for microbial adhesion, recognition, communication and toxin repulsion on the apical side. The mucus covers the inner surface of the gastrointestinal tract and provides a barrier function against pathogens and exogenous molecules [214,215].
Direct modulatory effects of HMOs on intestinal cells and structures are sparse. However, 2′FL supplementation was shown to increase villus areas and heights [122], crypt depths [216], elevated expression of brush boarder enzymes [124], rodents [122,216] or pigs [124], suggesting supporting effects for intestinal absorption. In vitro studies with non-transformed fetal crypt cells, preconfluent and postconfluent absorptive and secretory intestinal cells detected HMO-mediated cell maturation from the crypt to the villus [217,218,219,220]. Representatives of fucosylated, neutral non-fucosylated and sialylated HMOs inhibited the proliferation of immature secretory crypt cells through cell cycle arrest [218,219,221] or apoptosis [218,219,220,221] and enhanced their differentiation in the presence of 2′FL [219] and 6′SL [217]. In contrast, tested HMOs and LNnT respectively increased the nutrient absorption and barrier function of mature absorptive villal cells [219,220]. As mentioned above, Angeloni et al. detected that 3′SL is able to reduce the expression of sialyltransferases in intestinal cells thus modulating their glycocalyx for reduced enteropathogenic E. coli adherence [73]. Kong et al. proved that 2′FL and 3FL enhance the stability of the glycocalyx and its barrier function and integrity [222]. The same two HMOs induce the transcription of mucins (MUC2) in goblet cells [223], which was even confirmed in HMO-supplemented mice [224]. Furthermore, elevated mucin expression concurrent with reduced pathogenic adhesion and membrane permeability in 2′FL [121] or 3′SL-supplemented rats [225] as well as an increase of the mucosal proportion in 2′FL-supplemented pigs challenged with E. coli is in agreement with these observations [124]. Finally, Chleilat et al. reported that HMOs promote reduced intestinal permeability in female rats related to increased mRNA levels of tight junction proteins such as zonula occludens 1 and occludin [225].

2.4. Intestinal Immune Development and Homeostasis

Intrauterine and perinatal mucosal and systemic immunity deviates from mature immunity characterized by a propensity towards Th2 helper cells dominated by anti-inflammatory cytokine production. Rapidly after birth, the proportions of pro-inflammatory Th1 and regulatory T cells increase to achieve Th1 and TH17 homeostasis with Th2 in order to tolerate commensal colonization while protecting against extracellular and intracellular antigenic substances or pathogens [132,226]. HMOs have thereby been ascribed to mediate this “vigilant establishment” [227] by interaction with intestinal and immune cell receptor molecules such as C-type lectins [228,229], galectins [230], selectins [231], siglecs (sialic acid-binding Ig-like lectins) [228,232] and toll-like receptors [229,233,234,235].
In the gut, the intestinal epithelium is in steady concert with mucosal immune cells of the Peyer’s patches and the mesenteric lymph nodes to guarantee an appropriate response to multifaceted external stimuli and protect against overshooting reactions. At this forefront of host defense, milk sugars attenuate mucosal expression of inflammatory cytokine and take care of a balanced immune response as was evaluated in vitro and in vivo [236].
For instance, 3′SL-induced peroxisome proliferator-activated receptor γ (PPARγ) in Caco-2 cells that regulates peptidoglycan recognition protein 3, which in turn reduced inflammation (NFκB ↓; IL-8 ↓, IL-12 ↓, TNF α ↓, with ↓ meaning reduced expression) [237]. Later, Zehra et al. confirmed the PPARy-mediated anti-inflammatory action of sialylated HMOs with 6′SL showing IL-8 and MIP-3α reduction upon pro-inflammatory stimuli related to reduced AP1 and NFκB activity. In contrast, the same study showed PPARy independent immune attenuation by 2′FL [238]. Furthermore, active mimicry of inflammation [200], presence of pathogen-associated molecular patterns [234] as well as C. jejuni [67], E. coli [239] or Salmonella [240] were also attenuated by a reduced expression of inflammatory markers (IL-1β, IL-8, I L-12, MCP-1/2, MIP-3α) in the presence of specific colostral HMOs like 2′galactosyloliogosaccharide (2′GL) as well as total and individual HMOs like 2′FL, 3FL, 3′SL, 6′SL and LNFP I [67,234,239,240]. In one of these studies, elevated cytokines involved in tissue repair and Th1/Th2 homeostasis were detected [234]. Furthermore, exposure of intestinal epithelial cells to bacterial DNA boosted inflammatory Th1 IFNγ and regulatory IL-10, reduced Th2 type IL-13 and induced galectin release that attracts migratory dendritic cells. Intriguingly, intestinal cells imprinted with this pathogenic stimulus instructed naïve T helper cells towards a similar response (IFNγ, IL10) via dendritic cells suggesting a HMO-dependent development of adaptive immune response [241].
Several preclinical studies corroborated the effects of HMOs observed for intestinal cells in vitro and indicate that milk sugars promote lymphocyte proliferation (B cell, regulatory T cells, Th1 cells) [242] and Th1/Th2 balancing in the mucosa. In a murine model of chronic colitis, 3′SL resembling some pathogen surface structures induced Th1/ Th17-dependent inflammation via dendritic cells of the mesenteric lymph nodes potentially educating the infant’s immune system about foreign antigens [243]. In pigs, HMO supplementation decreased pro-inflammatory IL-8 expression, increased Th1 cytokines (IL-12, INF-γ) balanced by regulatory IL-10 while attenuating the immune response to rotavirus infection [108]. Likewise, the intestinal inflammation in mice caused by C. jejuni attenuated an inflammatory response on the gene expression and protein level (IL-1β ↓, TNF α ↓, IL-6 ↓, MIP-2 ↓), while unchanged IL-17 levels indicated no induction of adaptive immune response [67]. Evidence that 2′FL exhibits positive immunomodulation was further shown in healthy and rotavirus infected rats. Both increased intestinal toll-like receptor expression upon supplementation likely facilitating an improved communication with the mucosal immune system as well as viral clearance [111]. Furthermore, in a mouse model of food allergy, 2′FL and 6′SL reduced allergy symptoms and proved attenuative modes of action reducing mast cell numbers and inhibiting their degranulation while increasing regulatory T cells (CD4+, CD25+, IL-10+) in Peyer’s patches and mesenteric lymph nodes [244]. Overall, the presented studies indicate supportive functions of HMOs achieving immune balance upon pathogen stimuli. However, note that there are some conflicting data. Hester et al. detected no effect of HMOs on intestinal Th1/Th2 homeostasis [107], which is in agreement with data from 2′FL-supplemented healthy rats exhibiting a parallel reduction of pro- and anti-inflammatory mucosal cytokines without tendency towards a Th1 increase [122].
The etiology of chronic inflammatory diseases of the gastrointestinal tract like colitis ulcerosa and necrotizing enterocolitis (NEC) are not well understood but are likely related to dysbiosis. NEC is a devastating disorder that mainly affects preterm infants of very low birth weight. Comparison of breast-fed and formula-fed infants indicated a lower incidence of NEC related to HMOs, which was corroborated by the fact that children of mothers with a low Secretor phenotype [245], low HMO diversity [246], low LNDFH I [246] but mainly DSLNT [246,247] concentration are more prone to the disease. In fact, the most dominant sialylated HMO of mothers delivering preterm infants is DSLNT as shown in Supplementary Tables S6–S9. These observational results were underlined by interventional studies with HMOs in rodents [248,249,250,251]. In addition, 2′FL and 3FL [224,252] but not 3′SL or 6′SL fostered [253] mice are more resistant to DSS-induced colitis of reduced severity associated with decreased markers of inflammation, histological and diarrhea scores.
Even though observed effects have been mainly linked to the prebiotic effects of HMOs, their immune attenuation and support of the intestinal barrier might play a role as well. This was proven in murine NEC models, which exhibited a decrease of pro-inflammatory markers [250,254], preservation of the intestinal mucosal architecture by gavage of 2′FL [250] and preservation of mucin-expressing goblet cells by the gavage of total HMOs [224]. In an interventional study in pigs, neither a reduction of NEC lesions nor an anti-inflammatory effect could be achieved by a mixture of four HMOs or more than 25 HMOs. Thus, further studies appear to be necessary to clarify the roles of HMOs in the development of NEC [200].

2.5. Systemic Effects

About 1–5% [62,63,64] of ingested HMOs are absorbed into the blood stream where they can exert balancing effects on the systemic immune system. This became evident on analysis of peripheral blood mononuclear cells (PBMCs) that were isolated from breastfed and formula-fed infants either receiving formula with or without 2′FL. Notably, 2′FL-supplemented babies showed similar anti-inflammatory cytokine levels as breastfed ones, which were ~30% to ~80% lower than in infants receiving no 2′FL [255]. Moreover, 2′FL and a mix of 4 HMOs (2′FL, LNnT, 3′SL, 6′SL) respectively increased splenic lymphocytes in mice [256] and the population of PMBCs in piglets, while increasing Th1 IFNγ in both cases [109]. In fact, in vitro studies on PBMCs including dendritic cells, lymphocytes and macrophages corroborated the observation that HMOs directly interact with systemic immune cells and affect their proliferation, differentiation and maturation. Xiao et al. have shown that total HMOs promote immune tolerance by acting on DCs that communicate with T cells through the presentation of antigens. The presence of milk oligosaccharides induced maturation of DCs, elevated regulatory IL-10 and IL-27 as well as pro-inflammatory IL6 but not TNF-α and caused the differentiation of naïve to regulatory T cells. Furthermore, the homeostatic function of HMOs via DCs upon lipopolysaccharide (LPS) stimulation was expressed by the release of IL-10 and IL-27, the reduction of pro-inflammatory markers (IL-12p70, IL-6 and TNF-α), an increase of regulatory T cells and a decrease of the Th1 frequency [229].
Data supporting HMO-mediated immune balancing via dendritic cells were obtained in non-obese diabetic mice, which are characterized by a Th1/Th17 polarized immune response. Here, HMOs induced anti-diabetogenic cytokine levels and the differentiation of dendritic cells towards a regulatory/tolerogenic phenotype thus reducing the incidence of diabetes and pancreatic insulitis later in life. While these positive effects were related to an alleviation of microbial dysbiosis and SCFAs in parallel, in vitro studies underlined that HMOs are even solely able to modulate blood mononuclear dendritic cells towards a regulatory cytokine microenvironment triggering an increase of regulatory T cells [257]. However, as 2′FL and 6′SL have not been able to modulate human dendritic cells, further studies are necessary to discover the active single sugars [258].
Studies using cord blood mononuclear cells (CBMCs) and PBMCs further reveal Th1/Th2 balancing of the neonate by acidic HMOs while increasing the regulatory T cell population [259,260]. In addition, fucosylated HMOs support regulatory immune response by inhibition of IFN-γ and IL-12 production [261], reduction of cell proliferation and IL-10 expression under non-stimulating conditions [261,262]. Upon respiratory syncytial virus infection, PBMCs have an anti-inflammatory effect in the presence of milk oligosaccharides [95]. In contrast, HMOs tend to potentiate the immune response upon co-stimulation with LPS and phytohemagglutinin by increasing immune cell proliferation and inflammatory TNF-α as well as decreasing T-cell population [262].
Direct immune modulatory effects on macrophages have also been shown for neutral HMOs [261,263,264]. These increase the production of prostaglandins as well as the expression of nitric oxide synthase, cyclooxygenase, pro-inflammatory but also regulatory cytokines [235,263,264]. Aside from this, Atochina et al. [263] discovered that LNFP III-stimulated macrophages activate natural killer cells. Finally, Bode et al. indicated that acidic HMOs protect tissues from leukocyte (monocyte, lymphocyte and neutrophil) infiltration [265]. This is an initial key event of inflammatory diseases like NEC, whereas neutrophil activation through formation of platelet-neutrophil complexes is crucial for progression [266]. Both events can be hindered by systemically circulating HMOs that bind to selectin surface receptors necessary for molecular docking [267].
Little evidence exists that individual HMOs are able to interact with the enteric and central nervous systems without relation to their prebiotic role in the microbiota gut-brain axis as was proposed by Tarr et al. and Jacobi et al. [203,204]. For instance, Bienenstock et al. verified that 2′FL and 3FL diminish colon motor contractions potentially by stimulating enteric neurons that in turn affect migrating motor complexes [268]. Data of Krug et al. [269], Matthies et al. [270] and Vázquez et al. [271] indicate that fucosyllactose improves long-term potentiation [269,270] and learning capabilities [271]. These were respectively assessed on hippocampal slices of rats [270], freely moving rats receiving intrahippocampal 2′FL [269] and rats that orally ingested 2′FL [271]. Among other speculated underlying mechanisms [269,270], 2′FL increased the expression of proteins in the hippocamus, stratium and frontal cortex that are fundamental for synaptic function related to learning and memory formation [271]. These molecular mechanisms were partly corroborated by Wu et al., who even attributed neuroprotective abilities to 2′FL [272], even though relations to prebiotic effects were not excluded. Observational human studies have also linked breastfeeding [273,274,275,276] and 2′FL to a better cognitive development of infants [276]. However, more studies are required to understand the modes of action in greater depth and to differentiate between HMOs that act on the brain either directly or via the microbiota-gut-brain axis.

3. Conclusions

Mother Nature went to great efforts to evolve the molecular class of human milk oligosaccharides, which contains a large variety of sugar molecules that are composed of five building blocks only. Their composition in human milk changes in interplay of the mother and her offspring and on demand of the growing infant to fulfill critical tasks for neonatal development and protection. This review collected in vitro, preclinical and clinical studies to elucidate that HMOs not only serve as soluble decoy receptors for pathogen protection. In fact, they actively shape immature gut microbiota, develop the intestinal and systemic immune systems, while supporting the function of the enteric and central nervous systems.
The recent advent of chemical and biotechnological production techniques has made several individual HMOs such as 2′FL, 3FL, DFL, LNT, LNnt, 3′SL and 6′SL available for research and mothers that (must) rely on formula feeding. In the future, this will allow larger (interventional) studies to further dig into the molecular modes of actions of HMOs and will hopefully trigger the industrial production of a greater variety of such promotive sugars to serve the market with personalized infant formula.

Supplementary Materials

Supplementary information are available online at https://0-www-mdpi-com.brum.beds.ac.uk/2072-6643/12/11/3568/s1. Table S1: Study information, HMO breastmilk content of term-delivering mothers, Table S2: Mean and pooled mean values of individual HMOs for mothers delivering at term belonging to milk group 1 (Se+Le+), Table S3: Mean and pooled mean values of individual HMOs for mothers delivering at term belonging to milk group 2 (SeLe+), Table S4: Mean and pooled mean values of individual HMOs for mothers delivering at term belonging to milk group 3 (Se+Le), Table S5: Mean and pooled mean values of individual HMOs for mothers delivering at term belonging to milk group 4 (SeLe), Table S6: Mean and pooled mean values of individual HMOs for mothers delivering preterm belonging to milk group 1 (Se+Le+), Table S7: Mean and pooled mean values of individual HMOs for mothers delivering preterm belonging to milk group 2 (SeLe+), Table S8: Mean and pooled mean values of individual HMOs for mothers delivering preterm belonging to milk group 3 (Se+Le), Table S9: Mean and pooled mean values of individual HMOs for mothers delivering preterm belonging to milk group 4 (SeLe).

Author Contributions

Writing—original draft preparation, C.H.; writing—review and editing, C.H., O.M.; supervision, O.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We kindly thank Norbert Sprenger for providing raw data of the concentration of human milk oligosaccharides.

Conflicts of Interest

C. Hundshammer and O. Minge are employed at Wacker Chemie AG declaring commercial interests.

References

  1. Kunz, C.; Rudloff, S.; Schad, W.; Braun, D. Lactose-derived oligosaccharides in the milk of elephants: Comparison with human milk. Br. J. Nutr. 1999, 82, 391–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ninonuevo, M.R.; Park, Y.; Yin, H.; Zhang, J.; Ward, R.E.; Clowers, B.H.; German, J.B.; Freeman, S.L.; Killeen, K.; Grimm, R.; et al. A strategy for annotating the human milk glycome. J. Agric. Food Chem. 2006, 54, 7471–7480. [Google Scholar] [CrossRef] [PubMed]
  3. Totten, S.M.; Zivkovic, A.M.; Wu, S.; Ngyuen, U.; Freeman, S.L.; Ruhaak, L.R.; Darboe, M.K.; German, J.B.; Prentice, A.M.; Lebrilla, C.B. Comprehensive profiles of human milk oligosaccharides yield highly sensitive and specific markers for determining secretor status in lactating mothers. J. Proteome Res. 2012, 11, 6124–6133. [Google Scholar] [CrossRef] [PubMed]
  4. Xu, G.; Davis, J.C.; Goonatilleke, E.; Smilowitz, J.T.; German, J.B.; Lebrilla, C.B. Absolute Quantitation of Human Milk Oligosaccharides Reveals Phenotypic Variations during Lactation. J. Nutr. 2017, 147, 117–124. [Google Scholar] [CrossRef] [Green Version]
  5. Elwakiel, M.; Hageman, J.A.; Wang, W.; Szeto, I.M.; van Goudoever, J.B.; Hettinga, K.A.; Schols, H.A. Human Milk Oligosaccharides in Colostrum and Mature Milk of Chinese Mothers: Lewis Positive Secretor Subgroups. J. Agric. Food Chem. 2018, 66, 7036–7043. [Google Scholar] [CrossRef]
  6. Viverge, D.; Grimmonprez, L.; Cassanas, G.; Bardet, L.; Bonnet, H.; Solere, M. Variations of lactose and oligosaccharides in milk from women of blood types secretor A or H, secretor Lewis, and secretor H/nonsecretor Lewis during the course of lactation. Ann. Nutr. Metab. 1985, 29, 1–11. [Google Scholar] [CrossRef]
  7. Gabrielli, O.; Zampini, L.; Galeazzi, T.; Padella, L.; Santoro, L.; Peila, C.; Giuliani, F.; Bertino, E.; Fabris, C.; Coppa, G.V. Preterm milk oligosaccharides during the first month of lactation. Pediatrics 2011, 128, e1520–e1531. [Google Scholar] [CrossRef]
  8. Viverge, D.; Grimmonprez, L.; Cassanas, G.; Bardet, L.; Solere, M. Variations in oligosaccharides and lactose in human milk during the first week of lactation. J. Pediatr. Gastroenterol. Nutr. 1990, 11, 361–364. [Google Scholar] [CrossRef]
  9. Coppa, G.V.; Gabrielli, O.; Pierani, P.; Catassi, C.; Carlucci, A.; Giorgi, P.L. Changes in carbohydrate composition in human milk over 4 months of lactation. Pediatrics 1993, 91, 637–641. [Google Scholar]
  10. Urashima, T.; Hirabayashi, J.; Sato, S.; Kobata, A. Human Milk Oligosaccharides as Essential Tools for Basic and Application Studies on Galectins. Trends Glycosci. Glycotechnol. 2018, 30, SE51–SE65. [Google Scholar] [CrossRef] [Green Version]
  11. Bode, L. Human milk oligosaccharides: Every baby needs a sugar mama. Glycobiology 2012, 22, 1147–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Samuel, T.M.; Binia, A.; de Castro, C.A.; Thakkar, S.K.; Billeaud, C.; Agosti, M.; Al-Jashi, I.; Costeira, M.J.; Marchini, G.; Martínez-Costa, C.; et al. Impact of maternal characteristics on human milk oligosaccharide composition over the first 4 months of lactation in a cohort of healthy European mothers. Sci. Rep. 2019, 9, 11767. [Google Scholar] [CrossRef] [PubMed]
  13. van Leeuwen, S.S.; Stoutjesdijk, E.; Ten Kate, G.A.; Schaafsma, A.; Dijck-Brouwer, J.; Muskiet, F.A.J.; Dijkhuizen, L. Regional variations in human milk oligosaccharides in Vietnam suggest FucTx activity besides FucT2 and FucT3. Sci. Rep. 2018, 8, 16790. [Google Scholar] [CrossRef] [PubMed]
  14. Dror, D.K.; Allen, L.H. Overview of Nutrients in Human Milk. Adv. Nutr. 2018, 9, 278S–294S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Sabharwal, H.; Sjoblad, S.; Lundblad, A. Sialylated oligosaccharides in human milk and feces of preterm, full-term, and weaning infants. J. Pediatr. Gastroenterol. Nutr. 1991, 12, 480–484. [Google Scholar] [CrossRef]
  16. Miller, J.B.; Bull, S.; Miller, J.; McVeagh, P. The oligosaccharide composition of human milk: Temporal and individual variations in monosaccharide components. J. Pediatr. Gastroenterol. Nutr. 1994, 19, 371–376. [Google Scholar] [CrossRef]
  17. Coppa, G.V.; Pierani, P.; Zampini, L.; Carloni, I.; Carlucci, A.; Gabrielli, O. Oligosaccharides in human milk during different phases of lactation. Acta Paediatr. 1999, 88, 89–94. [Google Scholar] [CrossRef]
  18. Nakhla, T.; Fu, D.; Zopf, D.; Brodsky, N.L.; Hurt, H. Neutral oligosaccharide content of preterm human milk. Br. J. Nutr. 1999, 82, 361–367. [Google Scholar] [CrossRef] [Green Version]
  19. Erney, R.M.; Malone, W.T.; Skelding, M.B.; Marcon, A.A.; Kleman-Leyer, K.M.; O’Ryan, M.L.; Ruiz-Palacios, G.; Hilty, M.D.; Pickering, L.K.; Prieto, P.A. Variability of human milk neutral oligosaccharides in a diverse population. J. Pediatr. Gastroenterol. Nutr. 2000, 30, 181–192. [Google Scholar] [CrossRef]
  20. Chaturvedi, P.; Warren, C.D.; Altaye, M.; Morrow, A.L.; Ruiz-Palacios, G.; Pickering, L.K.; Newburg, D.S. Fucosylated human milk oligosaccharides vary between individuals and over the course of lactation. Glycobiology 2001, 11, 365–372. [Google Scholar] [CrossRef]
  21. Wang, B.; Brand-Miller, J.; McVeagh, P.; Petocz, P. Concentration and distribution of sialic acid in human milk and infant formulas. Am. J. Clin. Nutr. 2001, 74, 510–515. [Google Scholar] [CrossRef] [PubMed]
  22. Martin-Sosa, S.; Martin, M.J.; Garcia-Pardo, L.A.; Hueso, P. Sialyloligosaccharides in human and bovine milk and in infant formulas: Variations with the progression of lactation. J. Dairy Sci. 2003, 86, 52–59. [Google Scholar] [CrossRef]
  23. Sumiyoshi, W.; Urashima, T.; Nakamura, T.; Arai, I.; Saito, T.; Tsumura, N.; Wang, B.; Brand-Miller, J.; Watanabe, Y.; Kimura, K. Determination of each neutral oligosaccharide in the milk of Japanese women during the course of lactation. Br. J. Nutr. 2003, 89, 61–69. [Google Scholar] [CrossRef] [PubMed]
  24. Davidson, B.; Meinzen-Derr, J.K.; Wagner, C.L.; Newburg, D.S.; Morrow, A.L. Fucosylated oligosaccharides in human milk in relation to gestational age and stage of lactation. Adv. Exp. Med. Biol. 2004, 554, 427–430. [Google Scholar] [CrossRef] [PubMed]
  25. Morrow, A.L.; Ruiz-Palacios, G.M.; Altaye, M.; Jiang, X.; Guerrero, M.L.; Meinzen-Derr, J.K.; Farkas, T.; Chaturvedi, P.; Pickering, L.K.; Newburg, D.S. Human milk oligosaccharides are associated with protection against diarrhea in breast-fed infants. J. Pediatr. 2004, 145, 297–303. [Google Scholar] [CrossRef]
  26. Asakuma, S.; Akahori, M.; Kimura, K.; Watanabe, Y.; Nakamura, T.; Tsunemi, M.; Arai, I.; Sanai, Y.; Urashima, T. Sialyl oligosaccharides of human colostrum: Changes in concentration during the first three days of lactation. Biosci. Biotechnol. Biochem. 2007, 71, 1447–1451. [Google Scholar] [CrossRef] [Green Version]
  27. Asakuma, S.; Urashima, T.; Akahori, M.; Obayashi, H.; Nakamura, T.; Kimura, K.; Watanabe, Y.; Arai, I.; Sanai, Y. Variation of major neutral oligosaccharides levels in human colostrum. Eur. J. Clin. Nutr. 2008, 62, 488–494. [Google Scholar] [CrossRef] [Green Version]
  28. Leo, F.; Asakuma, S.; Fukuda, K.; Senda, A.; Urashima, T. Determination of sialyl and neutral oligosaccharide levels in transition and mature milks of Samoan women, using anthranilic derivatization followed by reverse phase high performance liquid chromatography. Biosci. Biotechnol. Biochem. 2010, 74, 298–303. [Google Scholar] [CrossRef]
  29. Thurl, S.; Munzert, M.; Henker, J.; Boehm, G.; Muller-Werner, B.; Jelinek, J.; Stahl, B. Variation of human milk oligosaccharides in relation to milk groups and lactational periods. Br. J. Nutr. 2010, 104, 1261–1271. [Google Scholar] [CrossRef] [Green Version]
  30. Van Niekerk, E.; Autran, C.A.; Nel, D.G.; Kirsten, G.F.; Blaauw, R.; Bode, L. Human milk oligosaccharides differ between HIV-infected and HIV-uninfected mothers and are related to necrotizing enterocolitis incidence in their preterm very-low-birth-weight infants. J. Nutr. 2014, 144, 1227–1233. [Google Scholar] [CrossRef] [Green Version]
  31. Spevacek, A.R.; Smilowitz, J.T.; Chin, E.L.; Underwood, M.A.; German, J.B.; Slupsky, C.M. Infant Maturity at Birth Reveals Minor Differences in the Maternal Milk Metabolome in the First Month of Lactation. J. Nutr. 2015, 145, 1698–1708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Austin, S.; De Castro, C.A.; Benet, T.; Hou, Y.; Sun, H.; Thakkar, S.K.; Vinyes-Pares, G.; Zhang, Y.; Wang, P. Temporal Change of the Content of 10 Oligosaccharides in the Milk of Chinese Urban Mothers. Nutrients 2016, 8, 346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Grote, V.; Verduci, E.; Scaglioni, S.; Vecchi, F.; Contarini, G.; Giovannini, M.; Koletzko, B.; Agostoni, C. Breast milk composition and infant nutrient intakes during the first 12 months of life. Eur. J. Clin. Nutr. 2016, 70, 250–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Davis, J.C.; Lewis, Z.T.; Krishnan, S.; Bernstein, R.M.; Moore, S.E.; Prentice, A.M.; Mills, D.A.; Lebrilla, C.B.; Zivkovic, A.M. Growth and Morbidity of Gambian Infants are Influenced by Maternal Milk Oligosaccharides and Infant Gut Microbiota. Sci. Rep. 2017, 7, 40466. [Google Scholar] [CrossRef] [PubMed]
  35. Sprenger, N.; Lee, L.Y.; De Castro, C.A.; Steenhout, P.; Thakkar, S.K. Longitudinal change of selected human milk oligosaccharides and association to infants’ growth, an observatory, single center, longitudinal cohort study. PLoS ONE 2017, 12, e0171814. [Google Scholar] [CrossRef] [Green Version]
  36. McJarrow, P.; Radwan, H.; Ma, L.; MacGibbon, A.K.H.; Hashim, M.; Hasan, H.; Obaid, R.S.; Naja, F.; Mohamed, H.J.J.; Al Ghazal, H.; et al. Human Milk Oligosaccharide, Phospholipid, and Ganglioside Concentrations in Breast Milk from United Arab Emirates Mothers: Results from the MISC Cohort. Nutrients 2019, 11, 2400. [Google Scholar] [CrossRef] [Green Version]
  37. Wang, M.; Zhao, Z.; Zhao, A.; Zhang, J.; Wu, W.; Ren, Z.; Wang, P.; Zhang, Y. Neutral Human Milk Oligosaccharides Are Associated with Multiple Fixed and Modifiable Maternal and Infant Characteristics. Nutrients 2020, 12, 826. [Google Scholar] [CrossRef] [Green Version]
  38. Thurl, S.; Henker, J.; Siegel, M.; Tovar, K.; Sawatzki, G. Detection of four human milk groups with respect to Lewis blood group dependent oligosaccharides. Glycoconj. J. 1997, 14, 795–799. [Google Scholar] [CrossRef]
  39. Stahl, B.; Thurl, S.; Henker, J.; Siegel, M.; Finke, B.; Sawatzki, G. Detection of four human milk groups with respect to Lewis-blood-group-dependent oligosaccharides by serologic and chromatographic analysis. Adv. Exp. Med. Biol. 2001, 501, 299–306. [Google Scholar] [CrossRef]
  40. Musumeci, M.; Simpore, J.; D’Agata, A.; Sotgiu, S.; Musumeci, S. Oligosaccharides in colostrum of Italian and Burkinabe women. J. Pediatr. Gastroenterol. Nutr. 2006, 43, 372–378. [Google Scholar] [CrossRef] [Green Version]
  41. Coppa, G.V.; Gabrielli, O.; Zampini, L.; Galeazzi, T.; Ficcadenti, A.; Padella, L.; Santoro, L.; Soldi, S.; Carlucci, A.; Bertino, E.; et al. Oligosaccharides in 4 different milk groups, Bifidobacteria, and Ruminococcus obeum. J. Pediatr. Gastroenterol. Nutr. 2011, 53, 80–87. [Google Scholar] [CrossRef] [PubMed]
  42. Kunz, C.; Meyer, C.; Collado, M.C.; Geiger, L.; Garcia-Mantrana, I.; Bertua-Rios, B.; Martinez-Costa, C.; Borsch, C.; Rudloff, S. Influence of Gestational Age, Secretor, and Lewis Blood Group Status on the Oligosaccharide Content of Human Milk. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 789–798. [Google Scholar] [CrossRef] [PubMed]
  43. Austin, S.; De Castro, C.A.; Sprenger, N.; Binia, A.; Affolter, M.; Garcia-Rodenas, C.L.; Beauport, L.; Tolsa, J.F.; Fischer Fumeaux, C.J. Human Milk Oligosaccharides in the Milk of Mothers Delivering Term versus Preterm Infants. Nutrients 2019, 11, 1282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Tonon, K.M.; Morais, B.d.M.; Abrao, C.F.V.A.; Miranda, A.; Morais, T.B. Maternal and Infant Factors Associated with Human Milk Oligosaccharides Concentrations According to Secretor and Lewis Phenotypes. Nutrients 2019, 11, 1358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Mank, M.; Hauner, H.; Heck, A.J.R.; Stahl, B. Targeted LC-ESI-MS2 characterization of human milk oligosaccharide diversity at 6 to 16 weeks post-partum reveals clear staging effects and distinctive milk groups. Anal. Bioanal. Chem. 2020. [Google Scholar] [CrossRef]
  46. Wu, J.; Wu, S.; Huo, J.; Ruan, H.; Xu, X.; Hao, Z.; Wei, Y.A. Systematic Characterization and Longitudinal Study Reveal Distinguishing Features of Human Milk Oligosaccharides in China. Curr. Dev. Nutr. 2020, 4, nzaa113. [Google Scholar] [CrossRef]
  47. Paganini, D.; Uyoga, M.A.; Kortman, G.A.M.; Boekhorst, J.; Schneeberger, S.; Karanja, S.; Hennet, T.; Zimmermann, M.B. Maternal Human Milk Oligosaccharide Profile Modulates the Impact of an Intervention with Iron and Galacto-Oligosaccharides in Kenyan Infants. Nutrients 2019, 11, 2596. [Google Scholar] [CrossRef] [Green Version]
  48. Ferreira, A.L.; Alves, R.; Figueiredo, A.; Alves-Santos, N.; Freitas-Costa, N.; Batalha, M.; Yonemitsu, C.; Manivong, N.; Furst, A.; Bode, L.; et al. Human Milk Oligosaccharide Profile Variation throughout Postpartum in Healthy Women in a Brazilian Cohort. Nutrients 2020, 12, 790. [Google Scholar] [CrossRef] [Green Version]
  49. Olivares, M.; Albrecht, S.; De Palma, G.; Ferrer, M.D.; Castillejo, G.; Schols, H.A.; Sanz, Y. Human milk composition differs in healthy mothers and mothers with celiac disease. Eur. J. Nutr. 2015, 54, 119–128. [Google Scholar] [CrossRef]
  50. Thurl, S.; Munzert, M.; Boehm, G.; Matthews, C.; Stahl, B. Systematic review of the concentrations of oligosaccharides in human milk. Nutr. Rev. 2017, 75, 920–933. [Google Scholar] [CrossRef] [Green Version]
  51. McGuire, M.K.; Meehan, C.L.; McGuire, M.A.; Williams, J.E.; Foster, J.; Sellen, D.W.; Kamau-Mbuthia, E.W.; Kamundia, E.W.; Mbugua, S.; Moore, S.E.; et al. What’s normal? Oligosaccharide concentrations and profiles in milk produced by healthy women vary geographically. Am. J. Clin. Nutr. 2017, 105, 1086–1100. [Google Scholar] [CrossRef] [PubMed]
  52. Quin, C.; Vicaretti, S.D.; Mohtarudin, N.A.; Garner, A.M.; Vollman, D.M.; Gibson, D.L.; Zandberg, W.F. Influence of sulfonated and diet-derived human milk oligosaccharides on the infant microbiome and immune markers. J. Biol. Chem. 2020. [Google Scholar] [CrossRef] [PubMed]
  53. Alderete, T.L.; Autran, C.; Brekke, B.E.; Knight, R.; Bode, L.; Goran, M.I.; Fields, D.A. Associations between human milk oligosaccharides and infant body composition in the first 6 mo of life. Am. J. Clin. Nutr. 2015, 102, 1381–1388. [Google Scholar] [CrossRef] [PubMed]
  54. Azad, M.B.; Robertson, B.; Atakora, F.; Becker, A.B.; Subbarao, P.; Moraes, T.J.; Mandhane, P.J.; Turvey, S.E.; Lefebvre, D.L.; Sears, M.R.; et al. Human Milk Oligosaccharide Concentrations Are Associated with Multiple Fixed and Modifiable Maternal Characteristics, Environmental Factors, and Feeding Practices. J. Nutr. 2018, 148, 1733–1742. [Google Scholar] [CrossRef]
  55. Newburg, D.S. Human milk oligosaccharides vary among populations. Am. J. Clin. Nutr. 2017, 105, 1027–1028. [Google Scholar] [CrossRef] [Green Version]
  56. Jantscher-Krenn, E.; Aigner, J.; Reiter, B.; Köfeler, H.; Csapo, B.; Desoye, G.; Bode, L.; van Poppel, M.N.M. Evidence of human milk oligosaccharides in maternal circulation already during pregnancy: A pilot study. Am. J. Physiol. Metab. 2019, 316, e347–e357. [Google Scholar] [CrossRef]
  57. Wise, A.; Robertson, B.; Choudhury, B.; Rautava, S.; Isolauri, E.; Salminen, S.; Bode, L. Infants Are Exposed to Human Milk Oligosaccharides Already in utero. Front. Pediatr. 2018, 6, 270. [Google Scholar] [CrossRef] [Green Version]
  58. Hirschmugl, B.; Brandl, W.; Csapo, B.; van Poppel, M.; Köfeler, H.; Desoye, G.; Wadsack, C.; Jantscher-Krenn, E. Evidence of Human Milk Oligosaccharides in Cord Blood and Maternal-to-Fetal Transport across the Placenta. Nutrients 2019, 11, 2640. [Google Scholar] [CrossRef] [Green Version]
  59. Neville, M.C.; Keller, R.; Seacat, J.; Lutes, V.; Neifert, M.; Casey, C.; Allen, J.; Archer, P. Studies in human lactation: Milk volumes in lactating women during the onset of lactation and full lactation. Am. J. Clin. Nutr. 1988, 48, 1375–1386. [Google Scholar] [CrossRef]
  60. Gnoth, M.J.; Kunz, C.; Kinne-Saffran, E.; Rudloff, S. Human Milk Oligosaccharides Are Minimally Digested in Vitro. J. Nutr. 2000, 130, 3014–3020. [Google Scholar] [CrossRef]
  61. Coppa, G.V.; Pierani, P.; Zampini, L.; Bruni, S.; Carloni, I.; Gabrielli, O. Characterization of oligosaccharides in milk and feces of breast-fed infants by high-performance anion-exchange chromatography. Adv. Exp. Med. Biol. 2001, 501, 307–314. [Google Scholar] [CrossRef] [PubMed]
  62. Rudloff, S.; Pohlentz, G.; Diekmann, L.; Egge, H.; Kunz, C. Urinary excretion of lactose and oligosaccharides in preterm infants fed human milk or infant formula. Acta Paediatr. 1996, 85, 598–603. [Google Scholar] [CrossRef] [PubMed]
  63. Rudloff, S.; Pohlentz, G.; Borsch, C.; Lentze, M.J.; Kunz, C. Urinary excretion of in vivo 13C-labelled milk oligosaccharides in breastfed infants. Br. J. Nutr. 2012, 107, 957–963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Goehring, K.C.; Kennedy, A.D.; Prieto, P.A.; Buck, R.H. Direct evidence for the presence of human milk oligosaccharides in the circulation of breastfed infants. PLoS ONE 2014, 9, e101692. [Google Scholar] [CrossRef] [Green Version]
  65. Jantscher-Krenn, E.; Lauwaet, T.; Bliss, L.A.; Reed, S.L.; Gillin, F.D.; Bode, L. Human milk oligosaccharides reduce Entamoeba histolytica attachment and cytotoxicity in vitro. Br. J. Nutr. 2012, 108, 1839–1846. [Google Scholar] [CrossRef] [Green Version]
  66. Ruiz-Palacios, G.M.; Cervantes, L.E.; Ramos, P.; Chavez-Munguia, B.; Newburg, D.S. Campylobacter jejuni binds intestinal H(O) antigen (Fuc alpha 1, 2Gal beta 1, 4GlcNAc), and fucosyloligosaccharides of human milk inhibit its binding and infection. J. Biol. Chem. 2003, 278, 14112–14120. [Google Scholar] [CrossRef] [Green Version]
  67. Yu, Z.T.; Nanthakumar, N.N.; Newburg, D.S. The Human Milk Oligosaccharide 2′-Fucosyllactose Quenches Campylobacter jejuni-Induced Inflammation in Human Epithelial Cells HEp-2 and HT-29 and in Mouse Intestinal Mucosa. J. Nutr. 2016, 146, 1980–1990. [Google Scholar] [CrossRef]
  68. El-Hawiet, A.; Kitova, E.N.; Kitov, P.I.; Eugenio, L.; Ng, K.K.; Mulvey, G.L.; Dingle, T.C.; Szpacenko, A.; Armstrong, G.D.; Klassen, J.S. Binding of Clostridium difficile toxins to human milk oligosaccharides. Glycobiology 2011, 21, 1217–1227. [Google Scholar] [CrossRef] [Green Version]
  69. Nguyen, T.T.; Kim, J.W.; Park, J.S.; Hwang, K.H.; Jang, T.S.; Kim, C.H.; Kim, D. Identification of Oligosaccharides in Human Milk Bound onto the Toxin A Carbohydrate Binding Site of Clostridium difficile. J. Microbiol. Biotechnol. 2016, 26, 659–665. [Google Scholar] [CrossRef] [Green Version]
  70. El-Hawiet, A.; Kitova, E.N.; Klassen, J.S. Recognition of human milk oligosaccharides by bacterial exotoxins. Glycobiology 2015, 25, 845–854. [Google Scholar] [CrossRef] [Green Version]
  71. Cravioto, A.; Tello, A.; Villafan, H.; Ruiz, J.; del Vedovo, S.; Neeser, J.R. Inhibition of localized adhesion of enteropathogenic Escherichia coli to HEp-2 cells by immunoglobulin and oligosaccharide fractions of human colostrum and breast milk. J. Infect. Dis. 1991, 163, 1247–1255. [Google Scholar] [CrossRef] [PubMed]
  72. Crane, J.K.; Azar, S.S.; Stam, A.; Newburg, D.S. Oligosaccharides from human milk block binding and activity of the Escherichia coli heat-stable enterotoxin (STa) in T84 intestinal cells. J. Nutr. 1994, 124, 2358–2364. [Google Scholar] [CrossRef]
  73. Angeloni, S.; Ridet, J.L.; Kusy, N.; Gao, H.; Crevoisier, F.; Guinchard, S.; Kochhar, S.; Sigrist, H.; Sprenger, N. Glycoprofiling with micro-arrays of glycoconjugates and lectins. Glycobiology 2005, 15, 31–41. [Google Scholar] [CrossRef] [PubMed]
  74. Coppa, G.V.; Zampini, L.; Galeazzi, T.; Facinelli, B.; Ferrante, L.; Capretti, R.; Orazio, G. Human milk oligosaccharides inhibit the adhesion to Caco-2 cells of diarrheal pathogens: Escherichia coli, Vibrio cholerae, and Salmonella fyris. Pediatr. Res. 2006, 59, 377–382. [Google Scholar] [CrossRef] [Green Version]
  75. Weichert, S.; Jennewein, S.; Hufner, E.; Weiss, C.; Borkowski, J.; Putze, J.; Schroten, H. Bioengineered 2′-fucosyllactose and 3-fucosyllactose inhibit the adhesion of Pseudomonas aeruginosa and enteric pathogens to human intestinal and respiratory cell lines. Nutr. Res. 2013, 33, 831–838. [Google Scholar] [CrossRef] [PubMed]
  76. Manthey, C.F.; Autran, C.A.; Eckmann, L.; Bode, L. Human milk oligosaccharides protect against enteropathogenic Escherichia coli attachment in vitro and EPEC colonization in suckling mice. J. Pediatr. Gastroenterol. Nutr. 2014, 58, 165–168. [Google Scholar] [CrossRef] [PubMed]
  77. Facinelli, B.; Marini, E.; Magi, G.; Zampini, L.; Santoro, L.; Catassi, C.; Monachesi, C.; Gabrielli, O.; Coppa, G.V. Breast milk oligosaccharides: Effects of 2′-fucosyllactose and 6′-sialyllactose on the adhesion of Escherichia coli and Salmonella fyris to Caco-2 cells. J. Matern. Neonatal Med. 2019, 32, 2950–2952. [Google Scholar] [CrossRef] [PubMed]
  78. Martin-Sosa, S.; Martin, M.J.; Hueso, P. The sialylated fraction of milk oligosaccharides is partially responsible for binding to enterotoxigenic and uropathogenic Escherichia coli human strains. J. Nutr. 2002, 132, 3067–3072. [Google Scholar] [CrossRef]
  79. Coppa, G.V.; Gabrielli, O.; Giorgi, P.; Catassi, C.; Montanari, M.P.; Varaldo, P.E.; Nichols, B.L. Preliminary study of breastfeeding and bacterial adhesion to uroepithelial cells. Lancet 1990, 335, 569–571. [Google Scholar] [CrossRef]
  80. Lin, A.E.; Autran, C.A.; Espanola, S.D.; Bode, L.; Nizet, V. Human milk oligosaccharides protect bladder epithelial cells against uropathogenic Escherichia coli invasion and cytotoxicity. J. Infect. Dis. 2014, 209, 389–398. [Google Scholar] [CrossRef] [Green Version]
  81. Virkola, R.; Parkkinen, J.; Hacker, J.; Korhonen, T.K. Sialyloligosaccharide chains of laminin as an extracellular matrix target for S fimbriae of Escherichia coli. Infect. Immun. 1993, 61, 4480–4484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Simon, P.M.; Goode, P.L.; Mobasseri, A.; Zopf, D. Inhibition of Helicobacter pylori binding to gastrointestinal epithelial cells by sialic acid-containing oligosaccharides. Infect. Immun. 1997, 65, 750–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Chen, P.; Reiter, T.; Huang, B.; Kong, N.; Weimer, B.C. Prebiotic Oligosaccharides Potentiate Host Protective Responses against L. monocytogenes Infection. Pathogens 2017, 6, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Hakkarainen, J.; Toivanen, M.; Leinonen, A.; Frangsmyr, L.; Stromberg, N.; Lapinjoki, S.; Nassif, X.; Tikkanen-Kaukanen, C. Human and bovine milk oligosaccharides inhibit Neisseria meningitidis pili attachment in vitro. J. Nutr. 2005, 135, 2445–2448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Devaraj, N.; Sheykhnazari, M.; Warren, W.S.; Bhavanandan, V.P. Differential binding of Pseudomonas aeruginosa to normal and cystic fibrosis tracheobronchial mucins. Glycobiology 1994, 4, 307–316. [Google Scholar] [CrossRef]
  86. Perret, S.; Sabin, C.; Dumon, C.; Pokorna, M.; Gautier, C.; Galanina, O.; Ilia, S.; Bovin, N.; Nicaise, M.; Desmadril, M.; et al. Structural basis for the interaction between human milk oligosaccharides and the bacterial lectin PA-IIL of Pseudomonas aeruginosa. Biochem. J. 2005, 389, 325–332. [Google Scholar] [CrossRef] [Green Version]
  87. Ackerman, D.L.; Craft, K.M.; Doster, R.S.; Weitkamp, J.H.; Aronoff, D.M.; Gaddy, J.A.; Townsend, S.D. Antimicrobial and Antibiofilm Activity of Human Milk Oligosaccharides against Streptococcus agalactiae, Staphylococcus aureus, and Acinetobacter baumannii. ACS Infect. Dis. 2018, 4, 315–324. [Google Scholar] [CrossRef]
  88. Idota, T.; Kawakami, H.; Murakami, Y.; Sugawara, M. Inhibition of cholera toxin by human milk fractions and sialyllactose. Biosci. Biotechnol. Biochem. 1995, 59, 417–419. [Google Scholar] [CrossRef]
  89. Hong, P.; Ninonuevo, M.R.; Lee, B.; Lebrilla, C.; Bode, L. Human milk oligosaccharides reduce HIV-1-gp120 binding to dendritic cell-specific ICAM3-grabbing non-integrin (DC-SIGN). Br. J. Nutr. 2009, 101, 482–486. [Google Scholar] [CrossRef] [Green Version]
  90. Matrosovich, M.N.; Gambaryan, A.S.; Tuzikov, A.B.; Byramova, N.E.; Mochalova, L.V.; Golbraikh, A.A.; Shenderovich, M.D.; Finne, J.; Bovin, N.V. Probing of the receptor-binding sites of the H1 and H3 influenza A and influenza B virus hemagglutinins by synthetic and natural sialosides. Virology 1993, 196, 111–121. [Google Scholar] [CrossRef]
  91. Pandey, R.P.; Kim, D.H.; Woo, J.; Song, J.; Jang, S.H.; Kim, J.B.; Cheong, K.M.; Oh, J.S.; Sohng, J.K. Broad-spectrum neutralization of avian influenza viruses by sialylated human milk oligosaccharides: In vivo assessment of 3′-sialyllactose against H9N2 in chickens. Sci. Rep. 2018, 8, 2563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Weichert, S.; Koromyslova, A.; Singh, B.K.; Hansman, S.; Jennewein, S.; Schroten, H.; Hansman, G.S. Structural Basis for Norovirus Inhibition by Human Milk Oligosaccharides. J. Virol. 2016, 90, 4843–4848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Koromyslova, A.; Tripathi, S.; Morozov, V.; Schroten, H.; Hansman, G.S. Human norovirus inhibition by a human milk oligosaccharide. Virology 2017, 508, 81–89. [Google Scholar] [CrossRef] [PubMed]
  94. Hanisch, F.G.; Hansman, G.S.; Morozov, V.; Kunz, C.; Schroten, H. Avidity of alpha-fucose on human milk oligosaccharides and blood group-unrelated oligo/polyfucoses is essential for potent norovirus-binding targets. J. Biol. Chem. 2018, 293, 11955–11965. [Google Scholar] [CrossRef] [Green Version]
  95. Duska-McEwen, G.; Senft, A.; Ruetschilling, T.; Barrett, E.; Buck, R. Human Milk Oligosaccharides Enhance Innate Immunity to Respiratory Syncytial Virus and Influenza in Vitro. Food Nutr. Sci. 2014, 5, 1383–1395. [Google Scholar] [CrossRef] [Green Version]
  96. Neu, U.; Khan, Z.M.; Schuch, B.; Palma, A.S.; Liu, Y.; Pawlita, M.; Feizi, T.; Stehle, T. Structures of B-lymphotropic polyomavirus VP1 in complex with oligosaccharide ligands. PLOS Pathog. 2013, 9, e1003714. [Google Scholar] [CrossRef] [Green Version]
  97. Ströh, L.J.; Gee, G.V.; Blaum, B.S.; Dugan, A.S.; Feltkamp, M.C.; Atwood, W.J.; Stehle, T. Trichodysplasia spinulosa-Associated Polyomavirus Uses a Displaced Binding Site on VP1 to Engage Sialylated Glycolipids. PLoS Pathog. 2015, 11, e1005112. [Google Scholar] [CrossRef]
  98. Gonia, S.; Tuepker, M.; Heisel, T.; Autran, C.; Bode, L.; Gale, C.A. Human Milk Oligosaccharides Inhibit Candida albicans Invasion of Human Premature Intestinal Epithelial Cells. J. Nutr. 2015, 145, 1992–1998. [Google Scholar] [CrossRef]
  99. Andreas, N.J.; Al-Khalidi, A.; Jaiteh, M.; Clarke, E.; Hyde, M.J.; Modi, N.; Holmes, E.; Kampmann, B.; Mehring Le Doare, K. Role of human milk oligosaccharides in Group B Streptococcus colonisation. Clin. Transl. Immunol. 2016, 5, e99. [Google Scholar] [CrossRef] [Green Version]
  100. Ackerman, D.L.; Doster, R.S.; Weitkamp, J.H.; Aronoff, D.M.; Gaddy, J.A.; Townsend, S.D. Human Milk Oligosaccharides Exhibit Antimicrobial and Antibiofilm Properties against Group B Streptococcus. ACS Infect. Dis. 2017, 3, 595–605. [Google Scholar] [CrossRef] [Green Version]
  101. Lin, A.E.; Autran, C.A.; Szyszka, A.; Escajadillo, T.; Huang, M.; Godula, K.; Prudden, A.R.; Boons, G.J.; Lewis, A.L.; Doran, K.S.; et al. Human milk oligosaccharides inhibit growth of group B Streptococcus. J. Biol. Chem. 2017, 292, 11243–11249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Craft, K.M.; Gaddy, J.A.; Townsend, S.D. Human Milk Oligosaccharides (HMOs) Sensitize Group B Streptococcus to Clindamycin, Erythromycin, Gentamicin, and Minocycline on a Strain Specific Basis. ACS Chem. Biol. 2018, 13, 2020–2026. [Google Scholar] [CrossRef] [PubMed]
  103. Craft, K.M.; Thomas, H.C.; Townsend, S.D. Sialylated variants of lacto-N-tetraose exhibit antimicrobial activity against Group B Streptococcus. Org. Biomol. Chem. 2019, 17, 1893–1900. [Google Scholar] [CrossRef] [PubMed]
  104. Craft, K.M.; Thomas, H.C.; Townsend, S.D. Interrogation of Human Milk Oligosaccharide Fucosylation Patterns for Antimicrobial and Antibiofilm Trends in Group B Streptococcus. ACS Infect. Dis. 2018, 4, 1755–1765. [Google Scholar] [CrossRef]
  105. Stepans, M.B.; Wilhelm, S.L.; Hertzog, M.; Rodehorst, T.K.; Blaney, S.; Clemens, B.; Polak, J.J.; Newburg, D.S. Early consumption of human milk oligosaccharides is inversely related to subsequent risk of respiratory and enteric disease in infants. Breastfeed. Med. 2006, 1, 207–215. [Google Scholar] [CrossRef]
  106. Morrow, A.L.; Ruiz-Palacios, G.M.; Altaye, M.; Jiang, X.; Guerrero, M.L.; Meinzen-Derr, J.K.; Farkas, T.; Chaturvedi, P.; Pickering, L.K.; Newburg, D.S. Human milk oligosaccharide blood group epitopes and innate immune protection against campylobacter and calicivirus diarrhea in breastfed infants. Adv. Exp. Med. Biol. 2004, 554, 443–446. [Google Scholar] [CrossRef]
  107. Hester, S.N.; Chen, X.; Li, M.; Monaco, M.H.; Comstock, S.S.; Kuhlenschmidt, T.B.; Kuhlenschmidt, M.S.; Donovan, S.M. Human milk oligosaccharides inhibit rotavirus infectivity in vitro and in acutely infected piglets. Br. J. Nutr. 2013, 110, 1233–1242. [Google Scholar] [CrossRef] [Green Version]
  108. Li, M.; Monaco, M.H.; Wang, M.; Comstock, S.S.; Kuhlenschmidt, T.B.; Fahey, G.C., Jr.; Miller, M.J.; Kuhlenschmidt, M.S.; Donovan, S.M. Human milk oligosaccharides shorten rotavirus-induced diarrhea and modulate piglet mucosal immunity and colonic microbiota. ISME J. 2014, 8, 1609–1620. [Google Scholar] [CrossRef] [Green Version]
  109. Comstock, S.S.; Li, M.; Wang, M.; Monaco, M.H.; Kuhlenschmidt, T.B.; Kuhlenschmidt, M.S.; Donovan, S.M. Dietary Human Milk Oligosaccharides but Not Prebiotic Oligosaccharides Increase Circulating Natural Killer Cell and Mesenteric Lymph Node Memory T Cell Populations in Noninfected and Rotavirus-Infected Neonatal Piglets. J. Nutr. 2017, 147, 1041–1047. [Google Scholar] [CrossRef] [Green Version]
  110. Azagra-Boronat, I.; Massot-Cladera, M.; Knipping, K.; Van’t Land, B.; Stahl, B.; Garssen, J.; Rodriguez-Lagunas, M.J.; Franch, A.; Castell, M.; Perez-Cano, F.J. Supplementation With 2′-FL and scGOS/lcFOS Ameliorates Rotavirus-Induced Diarrhea in Suckling Rats. Front. Cell. Infect. Microbiol. 2018, 8, 372. [Google Scholar] [CrossRef]
  111. Azagra-Boronat, I.; Massot-Cladera, M.; Knipping, K.; Van’t Land, B.; Tims, S.; Stahl, B.; Knol, J.; Garssen, J.; Franch, A.; Castell, M.; et al. Oligosaccharides Modulate Rotavirus-Associated Dysbiosis and TLR Gene Expression in Neonatal Rats. Cells 2019, 8, 876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Bode, L.; Kuhn, L.; Kim, H.Y.; Hsiao, L.; Nissan, C.; Sinkala, M.; Kankasa, C.; Mwiya, M.; Thea, D.M.; Aldrovandi, G.M. Human milk oligosaccharide concentration and risk of postnatal transmission of HIV through breastfeeding. Am. J. Clin. Nutr. 2012, 96, 831–839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kuhn, L.; Kim, H.Y.; Hsiao, L.; Nissan, C.; Kankasa, C.; Mwiya, M.; Thea, D.M.; Aldrovandi, G.M.; Bode, L. Oligosaccharide composition of breast milk influences survival of uninfected children born to HIV-infected mothers in Lusaka, Zambia. J. Nutr. 2015, 145, 66–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ukkonen, P.; Varis, K.; Jernfors, M.; Herva, E.; Jokinen, J.; Ruokokoski, E.; Zopf, D.; Kilpi, T. Treatment of acute otitis media with an antiadhesive oligosaccharide: A randomised, double-blind, placebo-controlled trial. Lancet 2000, 356, 1398–1402. [Google Scholar] [CrossRef]
  115. Mysore, J.V.; Wigginton, T.; Simon, P.M.; Zopf, D.; Heman-Ackah, L.M.; Dubois, A. Treatment of Helicobacter pylori infection in rhesus monkeys using a novel antiadhesion compound. Gastroenterology 1999, 117, 1316–1325. [Google Scholar] [CrossRef]
  116. Opekun, A.R.; El-Zaimaity, H.M.; Osato, M.S.; Gilger, M.A.; Malaty, H.M.; Terry, M.; Headon, D.R.; Graham, D.Y. Novel therapies for Helicobacter pylori infection. Aliment. Pharmacol. Ther. 1999, 13, 35–42. [Google Scholar] [CrossRef]
  117. Parente, F.; Cucino, C.; Anderloni, A.; Grandinetti, G.; Bianchi Porro, G. Treatment of Helicobacter pylori infection using a novel antiadhesion compound (3′sialyllactose sodium salt). A double blind, placebo-controlled clinical study. Helicobacter 2003, 8, 252–256. [Google Scholar] [CrossRef]
  118. Steenhout, P.; Sperisen, P.; Martin, F.-P.; Sprenger, N.; Wernimont, S.; Pecquet, S.; Berger, B. Term Infant Formula Supplemented with Human Milk Oligosaccharides (2′Fucosyllactose and Lacto-N-neotetraose) Shifts Stool Microbiota and Metabolic Signatures Closer to that of Breastfed Infants. FASEB J. 2016, 30, 275.7. [Google Scholar] [CrossRef]
  119. Newburg, D.S.; Pickering, L.K.; McCluer, R.H.; Cleary, T.G. Fucosylated oligosaccharides of human milk protect suckling mice from heat-stabile enterotoxin of Escherichia coli. J. Infect. Dis. 1990, 162, 1075–1080. [Google Scholar] [CrossRef]
  120. Newburg, D.S.; Ruiz-Palacios, G.M.; Altaye, M.; Chaturvedi, P.; Guerrero, M.L.; Meinzen-Derr, J.K.; Morrow, A.L. Human milk alphal,2-linked fucosylated oligosaccharides decrease risk of diarrhea due to stable toxin of E. coli in breastfed infants. Biol. Mammary Gland 2004, 554, 457–461. [Google Scholar] [CrossRef]
  121. Wang, Y.; Zou, Y.; Wang, J.; Ma, H.; Zhang, B.; Wang, S. The Protective Effects of 2′-Fucosyllactose against E. coli O157 Infection Are Mediated by the Regulation of Gut Microbiota and the Inhibition of Pathogen Adhesion. Nutrients 2020, 12, 1284. [Google Scholar] [CrossRef] [PubMed]
  122. Azagra-Boronat, I.; Massot-Cladera, M.; Mayneris-Perxachs, J.; Knipping, K.; Van’t Land, B.; Tims, S.; Stahl, B.; Garssen, J.; Franch, A.; Castell, M.; et al. Immunomodulatory and Prebiotic Effects of 2′-Fucosyllactose in Suckling Rats. Front. Immunol. 2019, 10, 1773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Bouchara, J.P.; Sanchez, M.; Chevailler, A.; Marot-Leblond, A.; Lissitzky, J.C.; Tronchin, G.; Chabasse, D. Sialic acid-dependent recognition of laminin and fibrinogen by Aspergillus fumigatus conidia. Infect. Immun. 1997, 65, 2717–2724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Cilieborg, M.S.; Sangild, P.T.; Jensen, M.L.; Ostergaard, M.V.; Christensen, L.; Rasmussen, S.O.; Morbak, A.L.; Jorgensen, C.B.; Bering, S.B. alpha1,2-Fucosyllactose Does Not Improve Intestinal Function or Prevent Escherichia coli F18 Diarrhea in Newborn Pigs. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 310–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Yue, H.; Han, Y.; Yin, B.; Cheng, C.; Liu, L. Comparison of the antipathogenic effect toward Staphylococcus aureus of N-linked and free oligosaccharides derived from human, bovine, and goat milk. J. Food Sci. 2020, 85, 2329–2339. [Google Scholar] [CrossRef]
  126. Andersson, B.; Porras, O.; Hanson, L.A.; Lagergard, T.; Svanborg-Eden, C. Inhibition of attachment of Streptococcus pneumoniae and Haemophilus influenzae by human milk and receptor oligosaccharides. J. Infect. Dis. 1986, 153, 232–237. [Google Scholar] [CrossRef]
  127. Idanpaan-Heikkila, I.; Simon, P.M.; Zopf, D.; Vullo, T.; Cahill, P.; Sokol, K.; Tuomanen, E. Oligosaccharides interfere with the establishment and progression of experimental pneumococcal pneumonia. J. Infect. Dis. 1997, 176, 704–712. [Google Scholar] [CrossRef]
  128. Barthelson, R.; Mobasseri, A.; Zopf, D.; Simon, P. Adherence of Streptococcus pneumoniae to respiratory epithelial cells is inhibited by sialylated oligosaccharides. Infect. Immun. 1998, 66, 1439–1444. [Google Scholar] [CrossRef] [Green Version]
  129. Laucirica, D.R.; Triantis, V.; Schoemaker, R.; Estes, M.K.; Ramani, S. Milk Oligosaccharides Inhibit Human Rotavirus Infectivity in MA104 Cells. J. Nutr. 2017, 147, 1709–1714. [Google Scholar] [CrossRef] [Green Version]
  130. Cox, L.M.; Yamanishi, S.; Sohn, J.; Alekseyenko, A.V.; Leung, J.M.; Cho, I.; Kim, S.G.; Li, H.; Gao, Z.; Mahana, D.; et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 2014, 158, 705–721. [Google Scholar] [CrossRef] [Green Version]
  131. Stewart, C.J.; Ajami, N.J.; O’Brien, J.L.; Hutchinson, D.S.; Smith, D.P.; Wong, M.C.; Ross, M.C.; Lloyd, R.E.; Doddapaneni, H.; Metcalf, G.A.; et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nat. Cell Biol. 2018, 562, 583–588. [Google Scholar] [CrossRef] [PubMed]
  132. Dzidic, M.; Boix-Amorós, A.; Selma-Royo, M.; Mira, A.; Collado, M.C. Gut Microbiota and Mucosal Immunity in the Neonate. Med. Sci. 2018, 6, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Wong, C.B.; Odamaki, T.; Xiao, J.-Z. Insights into the reason of Human-Residential Bifidobacteria (HRB) being the natural inhabitants of the human gut and their potential health-promoting benefits. FEMS Microbiol. Rev. 2020, 44, 369–385. [Google Scholar] [CrossRef] [PubMed]
  134. Matsuki, T.; Yahagi, K.; Mori, H.; Matsumoto, H.; Hara, T.; Tajima, S.; Ogawa, E.; Kodama, H.; Yamamoto, K.; Yamada, T.; et al. A key genetic factor for fucosyllactose utilization affects infant gut microbiota development. Nat. Commun. 2016, 7, 11939. [Google Scholar] [CrossRef] [PubMed]
  135. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef] [PubMed]
  136. 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, 690–703. [Google Scholar] [CrossRef] [Green Version]
  137. De Leoz, M.L.; Kalanetra, K.M.; Bokulich, N.A.; Strum, J.S.; Underwood, M.A.; German, J.B.; Mills, D.A.; Lebrilla, C.B. Human milk glycomics and gut microbial genomics in infant feces show a correlation between human milk oligosaccharides and gut microbiota: A proof-of-concept study. J. Proteome Res. 2015, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
  138. Houghteling, P.D.; Walker, W.A. Why is initial bacterial colonization of the intestine important to infants’ and children’s health? J. Pediatric Gastroenterol. Nutr. 2015, 60, 294–307. [Google Scholar] [CrossRef] [Green Version]
  139. Borewicz, K.; Gu, F.; Saccenti, E.; Arts, I.C.W.; Penders, J.; Thijs, C.; van Leeuwen, S.S.; Lindner, C.; Nauta, A.; van Leusen, E.; et al. Correlating Infant Faecal Microbiota Composition and Human Milk Oligosaccharide Consumption by Microbiota of One-Month Old Breastfed Infants. Mol. Nutr. Food Res. 2019, e1801214. [Google Scholar] [CrossRef]
  140. Davis, J.C.; Totten, S.M.; Huang, J.O.; Nagshbandi, S.; Kirmiz, N.; Garrido, D.A.; Lewis, Z.T.; Wu, L.D.; Smilowitz, J.T.; German, J.B.; et al. Identification of Oligosaccharides in Feces of Breast-fed Infants and Their Correlation with the Gut Microbial Community. Mol. Cell. Proteom. 2016, 15, 2987–3002. [Google Scholar] [CrossRef] [Green Version]
  141. Lawson, M.A.E.; O’Neill, I.J.; Kujawska, M.; Gowrinadh Javvadi, S.; Wijeyesekera, A.; Flegg, Z.; Chalklen, L.; Hall, L.J. Breast milk-derived human milk oligosaccharides promote Bifidobacterium interactions within a single ecosystem. ISME J. 2019. [Google Scholar] [CrossRef] [Green Version]
  142. Borewicz, K.; Gu, F.; Saccenti, E.; Hechler, C.; Beijers, R.; de Weerth, C.; van Leeuwen, S.S.; Schols, H.A.; Smidt, H. The association between breastmilk oligosaccharides and faecal microbiota in healthy breastfed infants at two, six, and twelve weeks of age. Sci. Rep. 2020, 10, 4270. [Google Scholar] [CrossRef] [PubMed]
  143. Harmsen, H.J.M.; Wildeboer–Veloo, A.C.M.; Raangs, G.C.; Wagendorp, A.A.; Klijn, N.; Bindels, J.G.; Welling, G.W. Analysis of Intestinal Flora Development in Breast-Fed and Formula-Fed Infants by Using Molecular Identification and Detection Methods. J. Pediatr. Gastroenterol. Nutr. 2000, 30, 61–67. [Google Scholar] [CrossRef] [PubMed]
  144. Boesten, R.; Schuren, F.; Ben Amor, K.; Haarman, M.; Knol, J.; de Vos, W.M. Bifidobacterium population analysis in the infant gut by direct mapping of genomic hybridization patterns: Potential for monitoring temporal development and effects of dietary regimens. Microb. Biotechnol. 2011, 4, 417–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Li, M.; Bauer, L.L.; Chen, X.; Wang, M.; Kuhlenschmidt, T.B.; Kuhlenschmidt, M.S.; Fahey, G.C., Jr.; Donovan, S.M. Microbial composition and in vitro fermentation patterns of human milk oligosaccharides and prebiotics differ between formula-fed and sow-reared piglets. J. Nutr. 2012, 142, 681–689. [Google Scholar] [CrossRef] [Green Version]
  146. Cabrera-Rubio, R.; Kunz, C.; Rudloff, S.; Garcia-Mantrana, I.; Crehua-Gaudiza, E.; Martinez-Costa, C.; Collado, M.C. Association of Maternal Secretor Status and Human Milk Oligosaccharides with Milk Microbiota: An Observational Pilot Study. J. Pediatr. Gastroenterol. Nutr. 2019, 68, 256–263. [Google Scholar] [CrossRef]
  147. Korpela, K.; Salonen, A.; Hickman, B.; Kunz, C.; Sprenger, N.; Kukkonen, K.; Savilahti, E.; Kuitunen, M.; de Vos, W.M. Fucosylated oligosaccharides in mother’s milk alleviate the effects of caesarean birth on infant gut microbiota. Sci. Rep. 2018, 8, 13757. [Google Scholar] [CrossRef] [Green Version]
  148. Odamaki, T.; Kato, K.; Sugahara, H.; Hashikura, N.; Takahashi, S.; Xiao, J.-z.; Abe, F.; Osawa, R. Age-related changes in gut microbiota composition from newborn to centenarian: A cross-sectional study. BMC Microbiol. 2016, 16, 90. [Google Scholar] [CrossRef] [Green Version]
  149. Schönfeld, H. Über die Beziehungen der einzelnen Bestandteile der Frauenmilch zur Bifidusflora. Jahrbuch Kinderh. 1926, 113, 19–60. [Google Scholar]
  150. Gauhe, A.; György, P.; Hoover, J.R.E.; Kuhn, R.; Rose, C.S.; Ruelius, H.W.; Zilliken, F. Bifidus factor. IV. Preparations obtained from human milk. Arch. Biochem. Biophys. 1954, 48, 214–224. [Google Scholar] [CrossRef]
  151. György, P.; Hoover, J.R.E.; Kuhn, R.; Rose, C.S. Bifidus factor. III. The rate of dialysis. Arch. Biochem. Biophys. 1954, 48, 209–213. [Google Scholar] [CrossRef]
  152. György, P.; Norris, R.F.; Rose, C.S. Bifidus factor. I. A variant of Lactobacillus bifidus requiring a special growth factor. Arch. Biochem. Biophys. 1954, 48, 193–201. [Google Scholar] [CrossRef]
  153. Rose, C.S.; Kuhn, R.; Zilliken, F.; György, P. Bifidus factor. V. The activity of α- and β-methyl-N-acetyl-d-glucosaminides. Arch. Biochem. Biophys. 1954, 49, 123–129. [Google Scholar] [CrossRef]
  154. Gyorgy, P.; Kuhn, R.; Rose, C.S.; Zilliken, F. Bifidus factor. II. Its occurrence in milk from different species and in other natural products. Arch. Biochem. Biophys. 1954, 48, 202–208. [Google Scholar] [CrossRef]
  155. Aakko, J.; Kumar, H.; Rautava, S.; Wise, A.; Autran, C.; Bode, L.; Isolauri, E.; Salminen, S. Human milk oligosaccharide categories define the microbiota composition in human colostrum. Benef. Microbes 2017, 8, 563–567. [Google Scholar] [CrossRef]
  156. Rudloff, S.; Kuntz, S.; Ostenfeldt Rasmussen, S.; Roggenbuck, M.; Sprenger, N.; Kunz, C.; Sangild, P.T.; Brandt Bering, S. Metabolism of Milk Oligosaccharides in Preterm Pigs Sensitive to Necrotizing Enterocolitis. Front. Nutr. 2019, 6, 23. [Google Scholar] [CrossRef]
  157. Lewis, Z.T.; Totten, S.M.; Smilowitz, J.T.; Popovic, M.; Parker, E.; Lemay, D.G.; Van Tassell, M.L.; Miller, M.J.; Jin, Y.S.; German, J.B.; et al. Maternal fucosyltransferase 2 status affects the gut bifidobacterial communities of breastfed infants. Microbiome 2015, 3, 13. [Google Scholar] [CrossRef] [Green Version]
  158. Smith-Brown, P.; Morrison, M.; Krause, L.; Davies, P.S. Mothers Secretor Status Affects Development of Childrens Microbiota Composition and Function: A Pilot Study. PLoS ONE 2016, 11, e0161211. [Google Scholar] [CrossRef]
  159. Yu, Z.T.; Chen, C.; Kling, D.E.; Liu, B.; McCoy, J.M.; Merighi, M.; Heidtman, M.; Newburg, D.S. The principal fucosylated oligosaccharides of human milk exhibit prebiotic properties on cultured infant microbiota. Glycobiology 2013, 23, 169–177. [Google Scholar] [CrossRef]
  160. Salli, K.; Anglenius, H.; Hirvonen, J.; Hibberd, A.A.; Ahonen, I.; Saarinen, M.T.; Tiihonen, K.; Maukonen, J.; Ouwehand, A.C. The effect of 2′-fucosyllactose on simulated infant gut microbiome and metabolites; a pilot study in comparison to GOS and lactose. Sci. Rep. 2019, 9, 13232. [Google Scholar] [CrossRef] [Green Version]
  161. Berger, B.; Porta, N.; Foata, F.; Grathwohl, D.; Delley, M.; Moine, D.; Charpagne, A.; Siegwald, L.; Descombes, P.; Alliet, P.; et al. Linking Human Milk Oligosaccharides, Infant Fecal Community Types, and Later Risk to Require Antibiotics. mBio 2020, 11, e03196-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Elison, E.; Vigsnaes, L.K.; Rindom Krogsgaard, L.; Rasmussen, J.; Sorensen, N.; McConnell, B.; Hennet, T.; Sommer, M.O.; Bytzer, P. Oral supplementation of healthy adults with 2′-O-fucosyllactose and lacto-N-neotetraose is well tolerated and shifts the intestinal microbiota. Br. J. Nutr. 2016, 116, 1356–1368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Ward, R.E.; Ninonuevo, M.; Mills, D.A.; Lebrilla, C.B.; German, J.B. In vitro fermentation of breast milk oligosaccharides by Bifidobacterium infantis and Lactobacillus gasseri. Appl. Environ. Microbiol. 2006, 72, 4497–4499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Ward, R.E.; Ninonuevo, M.; Mills, D.A.; Lebrilla, C.B.; German, J.B. In vitro fermentability of human milk oligosaccharides by several strains of bifidobacteria. Mol. Nutr. Food Res. 2007, 51, 1398–1405. [Google Scholar] [CrossRef]
  165. LoCascio, R.G.; Ninonuevo, M.R.; Freeman, S.L.; Sela, D.A.; Grimm, R.; Lebrilla, C.B.; Mills, D.A.; German, J.B. Glycoprofiling of bifidobacterial consumption of human milk oligosaccharides demonstrates strain specific, preferential consumption of small chain glycans secreted in early human lactation. J. Agric. Food Chem. 2007, 55, 8914–8919. [Google Scholar] [CrossRef]
  166. Locascio, R.G.; Ninonuevo, M.R.; Kronewitter, S.R.; Freeman, S.L.; German, J.B.; Lebrilla, C.B.; Mills, D.A. A versatile and scalable strategy for glycoprofiling bifidobacterial consumption of human milk oligosaccharides. Microb. Biotechnol. 2009, 2, 333–342. [Google Scholar] [CrossRef] [Green Version]
  167. Marcobal, A.; Barboza, M.; Froehlich, J.W.; Block, D.E.; German, J.B.; Lebrilla, C.B.; Mills, D.A. Consumption of human milk oligosaccharides by gut-related microbes. J. Agric. Food Chem. 2010, 58, 5334–5340. [Google Scholar] [CrossRef] [Green Version]
  168. LoCascio, R.G.; Desai, P.; Sela, D.A.; Weimer, B.; Mills, D.A. Broad Conservation of Milk Utilization Genes in Bifidobacterium longum subsp. infantis as Revealed by Comparative Genomic Hybridization. Appl. Environ. Microbiol. 2010, 76, 7373–7381. [Google Scholar] [CrossRef] [Green Version]
  169. Xiao, J.-z.; Takahashi, S.; Nishimoto, M.; Odamaki, T.; Yaeshima, T.; Iwatsuki, K.; Kitaoka, M. Distribution of in Vitro Fermentation Ability of Lacto-N-Biose I, a Major Building Block of Human Milk Oligosaccharides, in Bifidobacterial Strains. Appl. Environ. Microbiol. 2010, 76, 54–59. [Google Scholar] [CrossRef] [Green Version]
  170. Asakuma, S.; Hatakeyama, E.; Urashima, T.; Yoshida, E.; Katayama, T.; Yamamoto, K.; Kumagai, H.; Ashida, H.; Hirose, J.; Kitaoka, M. Physiology of consumption of human milk oligosaccharides by infant gut-associated bifidobacteria. J. Biol. Chem. 2011, 286, 34583–34592. [Google Scholar] [CrossRef] [Green Version]
  171. Garrido, D.; Kim, J.H.; German, J.B.; Raybould, H.E.; Mills, D.A. Oligosaccharide binding proteins from Bifidobacterium longum subsp. infantis reveal a preference for host glycans. PLoS ONE 2011, 6, e17315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Rockova, S.; Rada, V.; Marsik, P.; Vlkova, E.; Bunesova, V.; Sklenar, J.; Splichal, I. Growth of bifidobacteria and clostridia on human and cow milk saccharides. Anaerobe 2011, 17, 223–225. [Google Scholar] [CrossRef] [PubMed]
  173. Sela, D.A.; Li, Y.; Lerno, L.; Wu, S.; Marcobal, A.M.; German, J.B.; Chen, X.; Lebrilla, C.B.; Mills, D.A. An infant-associated bacterial commensal utilizes breast milk sialyloligosaccharides. J. Biol. Chem. 2011, 286, 11909–11918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Sela, D.A.; Garrido, D.; Lerno, L.; Wu, S.; Tan, K.; Eom, H.J.; Joachimiak, A.; Lebrilla, C.B.; Mills, D.A. Bifidobacterium longum subsp. infantis ATCC 15697 alpha-fucosidases are active on fucosylated human milk oligosaccharides. Appl. Environ. Microbiol. 2012, 78, 795–803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Strum, J.S.; Kim, J.; Wu, S.; De Leoz, M.L.A.; Peacock, K.; Grimm, R.; German, J.B.; Mills, D.A.; Lebrilla, C.B. Identification and Accurate Quantitation of Biological Oligosaccharide Mixtures. Anal. Chem. 2012, 84, 7793–7801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Ruiz-Moyano, S.; Totten, S.M.; Garrido, D.A.; Smilowitz, J.T.; German, J.B.; Lebrilla, C.B.; Mills, D.A. Variation in consumption of human milk oligosaccharides by infant gut-associated strains of Bifidobacterium breve. Appl. Environ. Microbiol. 2013, 79, 6040–6049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Yu, Z.T.; Chen, C.; Newburg, D.S. Utilization of major fucosylated and sialylated human milk oligosaccharides by isolated human gut microbes. Glycobiology 2013, 23, 1281–1292. [Google Scholar] [CrossRef] [Green Version]
  178. Garrido, D.; Ruiz-Moyano, S.; Lemay, D.G.; Sela, D.A.; German, J.B.; Mills, D.A. Comparative transcriptomics reveals key differences in the response to milk oligosaccharides of infant gut-associated bifidobacteria. Sci. Rep. 2015, 5, 13517. [Google Scholar] [CrossRef]
  179. Bunesova, V.; Lacroix, C.; Schwab, C. Fucosyllactose and L-fucose utilization of infant Bifidobacterium longum and Bifidobacterium kashiwanohense. BMC Microbiol. 2016, 16, 248. [Google Scholar] [CrossRef] [Green Version]
  180. Garrido, D.; Ruiz-Moyano, S.; Kirmiz, N.; Davis, J.C.; Totten, S.M.; Lemay, D.G.; Ugalde, J.A.; German, J.B.; Lebrilla, C.B.; Mills, D.A. A novel gene cluster allows preferential utilization of fucosylated milk oligosaccharides in Bifidobacterium longum subsp. longum SC596. Sci. Rep. 2016, 6, 35045. [Google Scholar] [CrossRef] [Green Version]
  181. James, K.; Motherway, M.O.; Bottacini, F.; van Sinderen, D. Bifidobacterium breve UCC2003 metabolises the human milk oligosaccharides lacto-N-tetraose and lacto-N-neo-tetraose through overlapping, yet distinct pathways. Sci. Rep. 2016, 6, 38560. [Google Scholar] [CrossRef] [PubMed]
  182. Thongaram, T.; Hoeflinger, J.L.; Chow, J.; Miller, M.J. Human milk oligosaccharide consumption by probiotic and human-associated bifidobacteria and lactobacilli. J. Dairy Sci. 2017, 100, 7825–7833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Gotoh, A.; Katoh, T.; Sakanaka, M.; Ling, Y.; Yamada, C.; Asakuma, S.; Urashima, T.; Tomabechi, Y.; Katayama-Ikegami, A.; Kurihara, S.; et al. Sharing of human milk oligosaccharides degradants within bifidobacterial communities in faecal cultures supplemented with Bifidobacterium bifidum. Sci. Rep. 2018, 8, 13958. [Google Scholar] [CrossRef] [PubMed]
  184. Schwab, C.; Gänzle, M. Lactic acid bacteria fermentation of human milk oligosaccharide components, human milk oligosaccharides and galactooligosaccharides. FEMS Microbiol. Lett. 2011, 315, 141–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Pichler, M.J.; Yamada, C.; Shuoker, B.; Alvarez-Silva, C.; Gotoh, A.; Leth, M.L.; Schoof, E.; Katoh, T.; Sakanaka, M.; Katayama, T.; et al. Butyrate producing colonic Clostridiales metabolise human milk oligosaccharides and cross feed on mucin via conserved pathways. Nat. Commun. 2020, 11, 3285. [Google Scholar] [CrossRef]
  186. Nishiyama, K.; Nagai, A.; Uribayashi, K.; Yamamoto, Y.; Mukai, T.; Okada, N. Two extracellular sialidases from Bifidobacterium bifidum promote the degradation of sialyl-oligosaccharides and support the growth of Bifidobacterium breve. Anaerobe 2018, 52, 22–28. [Google Scholar] [CrossRef]
  187. De Vuyst, L.; Leroy, F. Cross-feeding between bifidobacteria and butyrate-producing colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas production. Int. J. Food Microbiol. 2011, 149, 73–80. [Google Scholar] [CrossRef]
  188. Sakanaka, M.; Gotoh, A.; Yoshida, K.; Odamaki, T.; Koguchi, H.; Xiao, J.-Z.; Kitaoka, M.; Katayama, T. Varied Pathways of Infant Gut-Associated Bifidobacterium to Assimilate Human Milk Oligosaccharides: Prevalence of the Gene Set and Its Correlation with Bifidobacteria-Rich Microbiota Formation. Nutrients 2019, 12, 71. [Google Scholar] [CrossRef] [Green Version]
  189. Sela, D.A.; Chapman, J.; Adeuya, A.; Kim, J.H.; Chen, F.; Whitehead, T.R.; Lapidus, A.; Rokhsar, D.S.; Lebrilla, C.B.; German, J.B.; et al. The genome sequence of Bifidobacterium longum subsp. infantis reveals adaptations for milk utilization within the infant microbiome. Proc. Natl. Acad. Sci. USA 2008, 105, 18964–18969. [Google Scholar] [CrossRef] [Green Version]
  190. James, K.; Bottacini, F.; Contreras, J.I.S.; Vigoureux, M.; Egan, M.; Motherway, M.O.; Holmes, E.; van Sinderen, D. Metabolism of the predominant human milk oligosaccharide fucosyllactose by an infant gut commensal. Sci. Rep. 2019, 9, 15427. [Google Scholar] [CrossRef] [Green Version]
  191. Centanni, M.; Ferfuson, S.A.; Sims, L.M.; Biswas, A.; Tannock, G.W. Bifidobacterium bifidum ATCC 15696 and Bifidobacterium breve 24b Metabolic Interaction Based on 2′-O-Fucosyl-Lactose Studied in Steady-State Cultures in a Freter-Style Chemostat. Appl. Environ. Microbiol. 2019, 85, e02783-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. LeBlanc, J.G.; Milani, C.; de Giori, G.S.; Sesma, F.; van Sinderen, D.; Ventura, M. Bacteria as vitamin suppliers to their host: A gut microbiota perspective. Curr. Opin. Biotechnol. 2013, 24, 160–168. [Google Scholar] [CrossRef] [PubMed]
  193. Martinez, F.A.; Balciunas, E.M.; Converti, A.; Cotter, P.D.; de Souza Oliveira, R.P. Bacteriocin production by Bifidobacterium spp. A review. Biotechnol. Adv. 2013, 31, 482–488. [Google Scholar] [CrossRef] [PubMed]
  194. LeBlanc, J.G.; Chain, F.; Martín, R.; Bermúdez-Humarán, L.G.; Courau, S.; Langella, P. Beneficial effects on host energy metabolism of short-chain fatty acids and vitamins produced by commensal and probiotic bacteria. Microb. Cell Fact. 2017, 16, 1–10. [Google Scholar] [CrossRef] [Green Version]
  195. Bondue, P.; Milani, C.; Arnould, E.; Ventura, M.; Daube, G.; LaPointe, G.; Delcenserie, V. Bifidobacterium mongoliense genome seems particularly adapted to milk oligosaccharide digestion leading to production of antivirulent metabolites. BMC Microbiol. 2020, 20, 111. [Google Scholar] [CrossRef]
  196. Shen, Q.; Tuohy, K.M.; Gibson, G.R.; Ward, R.E. In vitro measurement of the impact of human milk oligosaccharides on the faecal microbiota of weaned formula-fed infants compared to a mixture of prebiotic fructooligosaccharides and galactooligosaccharides. Lett. Appl. Microbiol. 2011, 52, 337–343. [Google Scholar] [CrossRef]
  197. Musilova, S.; Modrackova, N.; Hermanova, P.; Hudcovic, T.; Svejstil, R.; Rada, V.; Tejnecky, V.; Bunesova, V. Assessment of the synbiotic properites of human milk oligosaccharides and Bifidobacterium longum subsp. infantis in vitro and in humanised mice. Benef. Microbes 2017, 8, 281–289. [Google Scholar] [CrossRef]
  198. Abbeele, P.V.D.; Duysburgh, C.; Vazquez, E.; Chow, J.; Buck, R.; Marzorati, M. 2′-Fucosyllactose alters the composition and activity of gut microbiota from formula-fed infants receiving complementary feeding in a validated intestinal model. J. Funct. Foods 2019, 61, 103484. [Google Scholar] [CrossRef]
  199. Grabinger, T.; Glaus Garzon, J.F.; Hausmann, M.; Geirnaert, A.; Lacroix, C.; Hennet, T. Alleviation of Intestinal Inflammation by Oral Supplementation with 2-Fucosyllactose in Mice. Front. Microbiol. 2019, 10, 1385. [Google Scholar] [CrossRef]
  200. Rasmussen, S.O.; Martin, L.; Ostergaard, M.V.; Rudloff, S.; Roggenbuck, M.; Nguyen, D.N.; Sangild, P.T.; Bering, S.B. Human milk oligosaccharide effects on intestinal function and inflammation after preterm birth in pigs. J. Nutr. Biochem. 2017, 40, 141–154. [Google Scholar] [CrossRef]
  201. den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.-J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Corrêa-Oliveira, R.; Fachi, J.L.; Vieira, A.; Sato, F.T.; Vinolo, M.A.R. Regulation of immune cell function by short-chain fatty acids. Clin. Transl. Immunol. 2016, 5, e73. [Google Scholar] [CrossRef] [PubMed]
  203. Tarr, A.J.; Galley, J.D.; Fisher, S.E.; Chichlowski, M.; Berg, B.M.; Bailey, M.T. The prebiotics 3′ Sialyllactose and 6′ Sialyllactose diminish stressor-induced anxiety-like behavior and colonic microbiota alterations: Evidence for effects on the gut-brain axis. Brain Behav. Immun. 2015, 50, 166–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Jacobi, S.K.; Yatsunenko, T.; Li, D.; Dasgupta, S.; Yu, R.K.; Berg, B.M.; Chichlowski, M.; Odle, J. Dietary Isomers of Sialyllactose Increase Ganglioside Sialic Acid Concentrations in the Corpus Callosum and Cerebellum and Modulate the Colonic Microbiota of Formula-Fed Piglets. J. Nutr. 2016, 146, 200–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol. Hepatol 2019, 16, 461–478. [Google Scholar] [CrossRef] [PubMed]
  206. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [Green Version]
  207. Zuurveld, M.; van Witzenburg, N.P.; Garssen, J.; Folkerts, G.; Stahl, B.; Van’t Land, B.; Willemsen, L.E.M. Immunomodulation by Human Milk Oligosaccharides: The Potential Role in Prevention of Allergic Diseases. Front. Immunol. 2020, 11, 801. [Google Scholar] [CrossRef]
  208. Sun, Y.; O’Riordan, M.X.D. Regulation of bacterial pathogenesis by intestinal short-chain Fatty acids. Adv. Appl. Microbiol. 2013, 85, 93–118. [Google Scholar] [CrossRef] [Green Version]
  209. Chichlowski, M.; De Lartigue, G.; German, J.B.; Raybould, H.E.; Mills, D.A. Bifidobacteria isolated from infants and cultured on human milk oligosaccharides affect intestinal epithelial function. J. Pediatr. Gastroenterol. Nutr. 2012, 55, 321–327. [Google Scholar] [CrossRef] [Green Version]
  210. Kavanaugh, D.W.; O’Callaghan, J.; Butto, L.F.; Slattery, H.; Lane, J.; Clyne, M.; Kane, M.; Joshi, L.; Hickey, R.M. Exposure of Bifidobacterium longum subsp. infantis to Milk Oligosaccharides Increases Adhesion to Epithelial Cells and Induces a Substantial Transcriptional Response. PLoS ONE 2013, 8, e67224. [Google Scholar] [CrossRef] [Green Version]
  211. Wickramasinghe, S.; Pacheco, A.R.; Lemay, D.G.; Mills, D.A. Bifidobacteria grown on human milk oligosaccharides downregulate the expression of inflammation-related genes in Caco-2 cells. BMC Microbiol. 2015, 15, 172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Musilova, S.; Modrackova, N.; Doskocil, I.; Svejstil, R.; Rada, V. Influence of human milk oligosaccharides on adherence of bifidobacteria and clostridia to cell lines. Acta Microbiol. Immunol. Hung. 2017, 64, 415–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Ewaschuk, J.B.; Diaz, H.; Meddings, L.; Diederichs, B.; Dmytrash, A.; Backer, J.; Looijer-van Langen, M.; Madsen, K.L. Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function. Am. J. Physiol. Physiol. Liver Physiol. 2008, 295, G1025–G1034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Gerbe, F.; Jay, P. Intestinal tuft cells: Epithelial sentinels linking luminal cues to the immune system. Mucosal Immunol. 2016, 9, 1353–1359. [Google Scholar] [CrossRef]
  215. Strzyz, P. Bend it like glycocalyx. Nat. Rev. Mol. Cell Biol. 2019, 20, 388. [Google Scholar] [CrossRef]
  216. Mezoff, E.A.; Hawkins, J.A.; Ollberding, N.J.; Karns, R.; Morrow, A.L.; Helmrath, M.A. The human milk oligosaccharide 2′-fucosyllactose augments the adaptive response to extensive intestinal. Am. J. Physiol. Liver Physiol. 2016, 310, G427–G438. [Google Scholar] [CrossRef] [Green Version]
  217. Kuntz, S.; Rudloff, S.; Kunz, C. Oligosaccharides from human milk influence growth-related characteristics of intestinally transformed and non-transformed intestinal cells. Br. J. Nutr. 2008, 99, 462–471. [Google Scholar] [CrossRef] [Green Version]
  218. Hester, S. Individual and Combined Effects of Nucleotides and Human Milk Oligosaccharides on Proliferation, Apoptosis and Necrosis in a Human Fetal Intestinal Cell Line. Food Nutr. Sci. 2012, 03, 1567–1576. [Google Scholar] [CrossRef] [Green Version]
  219. Holscher, H.D.; Davis, S.R.; Tappenden, K.A. Human milk oligosaccharides influence maturation of human intestinal Caco-2Bbe and HT-29 cell lines. J. Nutr. 2014, 144, 586–591. [Google Scholar] [CrossRef] [Green Version]
  220. Holscher, H.D.; Bode, L.; Tappenden, K.A. Human Milk Oligosaccharides Influence Intestinal Epithelial Cell Maturation in Vitro. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 296–301. [Google Scholar] [CrossRef]
  221. Kuntz, S.; Kunz, C.; Rudloff, S. Oligosaccharides from human milk induce growth arrest via G2/M by influencing growth-related cell cycle genes in intestinal epithelial cells. Br. J. Nutr. 2009, 101, 1306–1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Kong, C.; Elderman, M.; Cheng, L.; de Haan, B.J.; Nauta, A.; de Vos, P. Modulation of Intestinal Epithelial Glycocalyx Development by Human Milk Oligosaccharides and Non-Digestible Carbohydrates. Mol. Nutr. Food Res. 2019, 63, e1900303. [Google Scholar] [CrossRef] [PubMed]
  223. Cheng, L.; Kong, C.; Walvoort, M.T.C.; Faas, M.M.; de Vos, P. Human Milk Oligosaccharides Differently Modulate Goblet Cells Under Homeostatic, Proinflammatory Conditions and ER Stress. Mol. Nutr. Food Res 2020, 64, 1900976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Wu, R.Y.; Li, B.; Koike, Y.; Maattanen, P.; Miyake, H.; Cadete, M.; Johnson-Henry, K.C.; Botts, S.R.; Lee, C.; Abrahamsson, T.R.; et al. Human Milk Oligosaccharides Increase Mucin Expression in Experimental Necrotizing Enterocolitis. Mol. Nutr. Food Res. 2019, 63, e1800658. [Google Scholar] [CrossRef] [PubMed]
  225. Chleilat, F.; Klancic, T.; Ma, K.; Schick, A.; Nettleton, J.E.; Reimer, R.A. Human Milk Oligosaccharide Supplementation Affects Intestinal Barrier Function and Microbial Composition in the Gastrointestinal Tract of Young Sprague Dawley Rats. Nutrients 2020, 12, 1532. [Google Scholar] [CrossRef] [PubMed]
  226. Berger, A. Th1 and Th2 responses: What are they? BMJ 2000, 321, 424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Yu, J.C.; Khodadadi, H.; Malik, A.; Davidson, B.; Salles, É.d.S.L.; Bhatia, J.; Hale, V.L.; Baban, B. Innate Immunity of Neonates and Infants. Front. Immunol. 2018, 9, 1759. [Google Scholar] [CrossRef]
  228. Noll, A.J.; Yu, Y.; Lasanajak, Y.; Duska-McEwen, G.; Buck, R.H.; Smith, D.F.; Cummings, R.D. Human DC-SIGN binds specific human milk glycans. Biochem. J. 2016, 473, 1343–1353. [Google Scholar] [CrossRef] [Green Version]
  229. Xiao, L.; van De Worp, W.R.; Stassen, R.; van Maastrigt, C.; Kettelarij, N.; Stahl, B.; Blijenberg, B.; Overbeek, S.A.; Folkerts, G.; Garssen, J.; et al. Human milk oligosaccharides promote immune tolerance via direct interactions with human dendritic cells. Eur. J. Immunol. 2019, 49, 1001–1014. [Google Scholar] [CrossRef] [Green Version]
  230. Kitova, E.N.; El-Hawiet, A.; Klassen, J.S. Screening carbohydrate libraries for protein interactions using the direct ESI-MS assay. Applications to libraries of unknown concentration. J. Am. Soc. Mass Spectrom. 2014, 25, 1908–1916. [Google Scholar] [CrossRef]
  231. Rudloff, S.; Stefan, C.; Pohlentz, G.; Kunz, C. Detection of ligands for selectins in the oligosaccharide fraction of human milk. Eur. J. Nutr. 2002, 41, 85–92. [Google Scholar] [CrossRef] [PubMed]
  232. Koliwer-Brandl, H.; Siegert, N.; Umnus, K.; Kelm, A.; Tolkach, A.; Kulozik, U.; Kuballa, J.; Cartellieri, S.; Kelm, S. Lectin inhibition assays for the analysis of bioactive milk sialoglycoconjugates. Int. Dairy J. 2011, 21, 413–420. [Google Scholar] [CrossRef]
  233. Asakuma, S.; Yokoyama, T.; Kimura, K.; Watanabe, Y.; Nakamura, T.; Fukuda, K.; Urashima, T. Effect of Human Milk Oligosaccharides on Messenger Ribonucleic Acid Expression of Toll-like Receptor 2 and 4, and of MD2 in the Intestinal Cell Line HT-29. J. Appl. Glycosci. 2010, 57, 177–183. [Google Scholar] [CrossRef] [Green Version]
  234. He, Y.; Liu, S.; Leone, S.; Newburg, D.S. Human colostrum oligosaccharides modulate major immunologic pathways of immature human intestine. Mucosal Immunol. 2014, 7, 1326–1339. [Google Scholar] [CrossRef] [PubMed]
  235. Cheng, L.; Kiewiet, M.B.G.; Groeneveld, A.; Nauta, A.; de Vos, P. Human milk oligosaccharides and its acid hydrolysate LNT2 show immunomodulatory effects via TLRs in a dose and structure-dependent way. J. Funct. Foods 2019, 59, 174–184. [Google Scholar] [CrossRef]
  236. Lane, J.A.; O’Callaghan, J.; Carrington, S.D.; Hickey, R.M. Transcriptional response of HT-29 intestinal epithelial cells to human and bovine milk oligosaccharides. Br. J. Nutr. 2013, 110, 2127–2137. [Google Scholar] [CrossRef]
  237. Zenhom, M.; Hyder, A.; de Vrese, M.; Heller, K.J.; Roeder, T.; Schrezenmeir, J. Prebiotic oligosaccharides reduce proinflammatory cytokines in intestinal Caco-2 cells via activation of PPARgamma and peptidoglycan recognition protein 3. J. Nutr 2011, 141, 971–977. [Google Scholar] [CrossRef] [Green Version]
  238. Zehra, S.; Khambati, I.; Vierhout, M.; Mian, M.F.; Buck, R.; Forsythe, P. Human Milk Oligosaccharides Attenuate Antigen-Antibody Complex Induced Chemokine Release from Human Intestinal Epithelial Cell Lines. J. Food Sci. 2018, 83, 499–508. [Google Scholar] [CrossRef]
  239. He, Y.; Liu, S.; Kling, D.E.; Leone, S.; Lawlor, N.T.; Huang, Y.; Feinberg, S.B.; Hill, D.R.; Newburg, D.S. The human milk oligosaccharide 2′-fucosyllactose modulates CD14 expression in human enterocytes, thereby attenuating LPS-induced inflammation. Gut 2016, 65, 33–46. [Google Scholar] [CrossRef] [Green Version]
  240. Newburg, D.S.; Ko, J.S.; Leone, S.; Nanthakumar, N.N. Human Milk Oligosaccharides and Synthetic Galactosyloligosaccharides Contain 3′-, 4-, and 6′-Galactosyllactose and Attenuate Inflammation in Human T84, NCM-460, and H4 Cells and Intestinal Tissue ex Vivo. J. Nutr. 2016, 146, 358–367. [Google Scholar] [CrossRef]
  241. Ayechu-Muruzabal, V.; Overbeek, S.A.; Kostadinova, A.I.; Stahl, B.; Garssen, J.; Van’t Land, B.; Willemsen, L.E.M. Exposure of Intestinal Epithelial Cells to 2′-Fucosyllactose and CpG Enhances Galectin Release and Instructs Dendritic Cells to Drive Th1 and Regulatory-Type Immune Development. Biomolecules 2020, 10, 784. [Google Scholar] [CrossRef] [PubMed]
  242. Xiao, L.; Engen, P.A.; Leusink-Muis, T.; van Ark, I.; Stahl, B.; Overbeek, S.A.; Garssen, J.; Naqib, A.; Green, S.J.; Keshavarzian, A.; et al. The Combination of 2′-Fucosyllactose with Short-Chain Galacto-Oligosaccharides and Long-Chain Fructo-Oligosaccharides that Enhance Influenza Vaccine Responses Is Associated with Mucosal Immune Regulation in Mice. J. Nutr. 2019, 149, 856–869. [Google Scholar] [CrossRef] [PubMed]
  243. Kurakevich, E.; Hennet, T.; Hausmann, M.; Rogler, G.; Borsig, L. Milk oligosaccharide sialyl(alpha2,3)lactose activates intestinal CD11c+ cells through TLR4. Proc. Natl. Acad. Sci. USA 2013, 110, 17444–17449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Castillo-Courtade, L.; Han, S.; Lee, S.; Mian, F.M.; Buck, R.; Forsythe, P. Attenuation of food allergy symptoms following treatment with human milk oligosaccharides in a mouse model. Allergy 2015, 70, 1091–1102. [Google Scholar] [CrossRef] [PubMed]
  245. Morrow, A.L.; Meinzen-Derr, J.; Huang, P.; Schibler, K.R.; Cahill, T.; Keddache, M.; Kallapur, S.G.; Newburg, D.S.; Tabangin, M.; Warner, B.B.; et al. Fucosyltransferase 2 non-secretor and low secretor status predicts severe outcomes in premature infants. J. Pediatr. 2011, 158, 745–751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Wejryd, E.; Marti, M.; Marchini, G.; Werme, A.; Jonsson, B.; Landberg, E.; Abrahamsson, T.R. Low Diversity of Human Milk Oligosaccharides is Associated with Necrotising Enterocolitis in Extremely Low Birth Weight Infants. Nutrients 2018, 10, 1556. [Google Scholar] [CrossRef] [Green Version]
  247. Autran, C.A.; Kellman, B.P.; Kim, J.H.; Asztalos, E.; Blood, A.B.; Spence, E.C.H.; Patel, A.L.; Hou, J.; Lewis, N.E.; Bode, L. Human milk oligosaccharide composition predicts risk of necrotising enterocolitis in preterm infants. Gut 2018, 67, 1064–1070. [Google Scholar] [CrossRef]
  248. Jantscher-Krenn, E.; Zherebtsov, M.; Nissan, C.; Goth, K.; Guner, Y.S.; Naidu, N.; Choudhury, B.; Grishin, A.V.; Ford, H.R.; Bode, L. The human milk oligosaccharide disialyllacto-N-tetraose prevents necrotising enterocolitis in neonatal rats. Gut 2012, 61, 1417–1425. [Google Scholar] [CrossRef]
  249. Autran, C.A.; Schoterman, M.H.; Jantscher-Krenn, E.; Kamerling, J.P.; Bode, L. Sialylated galacto-oligosaccharides and 2′-fucosyllactose reduce necrotising enterocolitis in neonatal rats. Br. J. Nutr. 2016, 116, 294–299. [Google Scholar] [CrossRef] [Green Version]
  250. Good, M.; Sodhi, C.P.; Yamaguchi, Y.; Jia, H.; Lu, P.; Fulton, W.B.; Martin, L.Y.; Prindle, T.; Nino, D.F.; Zhou, Q.; et al. The human milk oligosaccharide 2′-fucosyllactose attenuates the severity of experimental necrotising enterocolitis by enhancing mesenteric perfusion in the neonatal intestine. Br. J. Nutr. 2016, 116, 1175–1187. [Google Scholar] [CrossRef] [Green Version]
  251. Wang, C.; Zhang, M.; Guo, H.; Yan, J.; Chen, L.; Teng, W.; Ren, F.; Li, Y.; Wang, X.; Luo, J.; et al. Human Milk Oligosaccharides Activate Epidermal Growth Factor Receptor and Protect Against Hypoxia-Induced Injuries in the Mouse Intestinal Epithelium and Caco2 Cells. J. Nutr 2020, 150, 756–762. [Google Scholar] [CrossRef] [PubMed]
  252. Weiss, G.A.; Chassard, C.; Hennet, T. Selective proliferation of intestinal Barnesiella under fucosyllactose supplementation in mice. Br. J. Nutr 2014, 111, 1602–1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Fuhrer, A.; Sprenger, N.; Kurakevich, E.; Borsig, L.; Chassard, C.; Hennet, T. Milk sialyllactose influences colitis in mice through selective intestinal bacterial colonization. J. Exp. Med. 2010, 207, 2843–2854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. He-Yang, J.; Zhang, W.; Liu, J.; Xue, P.; Zhou, X. Human breast milk oligosaccharides attenuate necrotizing enterocolitis in rats by suppressing mast cell accumulation, DPPI activity and TLR4 expression in ileum tissue, and regulating mitochondrial damage of Caco-2 cells. J. Exp. Med. 2020, 88, 106881. [Google Scholar] [CrossRef]
  255. Goehring, K.C.; Marriage, B.J.; Oliver, J.S.; Wilder, J.A.; Barrett, E.G.; Buck, R.H. Similar to Those Who Are Breastfed, Infants Fed a Formula Containing 2′-Fucosyllactose Have Lower Inflammatory Cytokines in a Randomized Controlled Trial. J. Nutr. 2016, 146, 2559–2566. [Google Scholar] [CrossRef] [Green Version]
  256. Xiao, L.; Leusink-Muis, T.; Kettelarij, N.; van Ark, I.; Blijenberg, B.; Hesen, N.A.; Stahl, B.; Overbeek, S.A.; Garssen, J.; Folkerts, G.; et al. Human Milk Oligosaccharide 2′-Fucosyllactose Improves Innate and Adaptive Immunity in an Influenza-Specific Murine Vaccination Model. Front. Immunol. 2018, 9, 452. [Google Scholar] [CrossRef] [Green Version]
  257. Xiao, L.; van’t Land, B.; Engen, P.A.; Naqib, A.; Green, S.J.; Nato, A.; Leusink-Muis, T.; Garssen, J.; Keshavarzian, A.; Stahl, B.; et al. Human milk oligosaccharides protect against the development of autoimmune diabetes in NOD-mice. Sci. Rep. 2018, 8, 3829. [Google Scholar] [CrossRef] [Green Version]
  258. Perdijk, O.; van Neerven, R.J.J.; van den Brink, E.; Savelkoul, H.F.J.; Brugman, S. The oligosaccharides 6′-sialyllactose, 2′-fucosyllactose or galactooligosaccharides do not directly modulate human dendritic cell differentiation or maturation. PLoS ONE 2018, 13, e0200356. [Google Scholar] [CrossRef]
  259. Eiwegger, T.; Stahl, B.; Schmitt, J.; Boehm, G.; Gerstmayr, M.; Pichler, J.; Dehlink, E.; Loibichler, C.; Urbanek, R.; Szepfalusi, Z. Human milk--derived oligosaccharides and plant-derived oligosaccharides stimulate cytokine production of cord blood T-cells in vitro. Pediatr. Res. 2004, 56, 536–540. [Google Scholar] [CrossRef] [Green Version]
  260. Eiwegger, T.; Stahl, B.; Haidl, P.; Schmitt, J.; Boehm, G.; Dehlink, E.; Urbanek, R.; Szepfalusi, Z. Prebiotic oligosaccharides: In vitro evidence for gastrointestinal epithelial transfer and immunomodulatory properties. Allergy Immunol. 2010, 21, 1179–1188. [Google Scholar] [CrossRef]
  261. Sotgiu, S.; Arru, G.; Fois, M.L.; Sanna, A.; Musumeci, M.; Rosati, G.; Musumeci, S. Immunomodulation of fucosyl-lactose and lacto-N-fucopentaose on mononuclear cells from multiple sclerosis and healthy subjects. Int. J. Biomed. Sci. 2006, 2, 114–120. [Google Scholar] [PubMed]
  262. Comstock, S.S.; Wang, M.; Hester, S.N.; Li, M.; Donovan, S.M. Select human milk oligosaccharides directly modulate peripheral blood mononuclear cells isolated from 10-d-old pigs. Br. J. Nutr. 2014, 111, 819–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Atochina, O.; Harn, D. LNFPIII/LeX-stimulated macrophages activate natural killer cells via CD40-CD40L interaction. Clin. Diagn. Lab. Immunol. 2005, 12, 1041–1049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Zhang, W.; Yan, J.; Wu, L.; Yu, Y.; Ye, R.D.; Zhang, Y.; Liang, X. In vitro immunomodulatory effects of human milk oligosaccharides on murine macrophage RAW264.7 cells. Carbohydr. Polym. 2019, 207, 230–238. [Google Scholar] [CrossRef]
  265. Bode, L.; Kunz, C.; Muhly-Reinholz, M.; Mayer, K.; Seeger, W.; Rudloff, S. Inhibition of monocyte, lymphocyte, and neutrophil adhesion to endothelial cells by human milk oligosaccharides. Thromb. Haemost. 2004, 92, 1402–1410. [Google Scholar] [CrossRef]
  266. Bode, L.; Rudloff, S.; Kunz, C.; Strobel, S.; Klein, N. Human milk oligosaccharides reduce platelet-neutrophil complex formation leading to a decrease in neutrophil beta 2 integrin expression. J. Leukoc. Biol. 2004, 76, 820–826. [Google Scholar] [CrossRef]
  267. Bode, L. Human Milk Oligosaccharides in the Prevention of Necrotizing Enterocolitis: A Journey from in vitro and in vivo Models to Mother-Infant Cohort Studies. Front. Pediatr. 2018, 6, 385. [Google Scholar] [CrossRef]
  268. Bienenstock, J.; Buck, R.H.; Linke, H.; Forsythe, P.; Stanisz, A.M.; Kunze, W.A. Fucosylated but not sialylated milk oligosaccharides diminish colon motor contractions. PLoS ONE 2013, 8, e76236. [Google Scholar] [CrossRef] [Green Version]
  269. Krug, M.; Wagner, M.; Staak, S.; Smalla, K.-H. Fucose and fucose-containing sugar epitopes enhance hippocampal long-term potentiation in the freely moving rat. Brain Res. 1994, 643, 130–135. [Google Scholar] [CrossRef]
  270. Matthies, H.; Staak, S.; Krug, M. Fucose and fucosyllactose enhance in-vitro hippocampal long-term potentiation. Brain Res. 1996, 725, 276–280. [Google Scholar] [CrossRef]
  271. Vazquez, E.; Barranco, A.; Ramirez, M.; Gruart, A.; Delgado-Garcia, J.M.; Martinez-Lara, E.; Blanco, S.; Martin, M.J.; Castanys, E.; Buck, R.; et al. Effects of a human milk oligosaccharide, 2′-fucosyllactose, on hippocampal long-term potentiation and learning capabilities in rodents. J. Nutr. Biochem. 2015, 26, 455–465. [Google Scholar] [CrossRef]
  272. Wu, K.-J.; Chen, Y.-H.; Bae, E.-K.; Song, Y.; Min, W.; Yu, S.-J. Human Milk Oligosaccharide 2′-Fucosyllactose Reduces Neurodegeneration in Stroke Brain. Transl. Stroke Res. 2020. [Google Scholar] [CrossRef]
  273. Anderson, J.W.; Johnstone, B.M.; Remley, D.T. Breast-feeding and cognitive development: A meta-analysis. Am. J. Clin. Nutr. 1999, 70, 525–535. [Google Scholar] [CrossRef]
  274. Kramer, M.S.; Aboud, F.; Mironova, E.; Vanilovich, I.; Platt, R.W.; Matush, L.; Igumnov, S.; Fombonne, E.; Bogdanovich, N.; Ducruet, T.; et al. Breastfeeding and Child Cognitive Development: New Evidence From a Large Randomized Trial. Arch. Gen. Psychiatry 2008, 65, 578–584. [Google Scholar] [CrossRef]
  275. Belfort, M.B.; Rifas-Shiman, S.L.; Kleinman, K.P.; Guthrie, L.B.; Bellinger, D.C.; Taveras, E.M.; Gillman, M.W.; Oken, E. Infant feeding and childhood cognition at ages 3 and 7 years: Effects of breastfeeding duration and exclusivity. JAMA Pediatr. 2013, 167, 836–844. [Google Scholar] [CrossRef] [Green Version]
  276. Berger, P.K.; Plows, J.F.; Jones, R.B.; Alderete, T.L.; Yonemitsu, C.; Poulsen, M.; Ryoo, J.H.; Peterson, B.S.; Bode, L.; Goran, M.I. Human milk oligosaccharide 2′-fucosyllactose links feedings at 1 month to cognitive development at 24 months in infants of normal and overweight mothers. PLoS ONE 2020, 15, e0228323. [Google Scholar] [CrossRef]
Figure 1. (a) Basic building blocks of human milk oligosaccharides; (b) effect of Secretor and Lewis genes on the composition of HMOs; and (c) main neutral fucosylated, neutral non-fucosylated and acidic HMOs. Abbreviations: 2′FL: 2′-fucosyllactose; 3FL: 3-fucosyllactose; LNFP I, II, III, IV, V, VI: lacto-N-fucopentaose I, II, II, IV, V, IV; F-LNH I, II: fucosyl-lacto-N-hexaose I, II; A-Tetra: A-tetrasaccharide; DFL: difucosyllactose; LNDFH I, II: lacto-N-difucosylhexaose I, II; LNnDFH: lacto-N-neodifucosylhexaose; DFLNH I, II: difucosyllacto-N-hexaose I, II; DFLNnH: difucosyllacto-N-neohexaose; DF-p-LNH: difucosyl-para-lacto-N-hexaose; DF-p-LNnH: difucosyl-para-lacto-N-neohexaose; TF-LNH: trifucosyl-lacto-N-hexaose; LNT: lacto-N-tetraose; LNnT: lacto-N-neotetraose; LNH: lacto-N-hexaose; LNnH: lacto-N-neohexaose; 3′/6′GL: 3′-/6′-galactosyloligosaccharide; 3′/6′SL: 3′-/6′- sialyllactose; LST a, b, c: sialyllacto-N-tetraose a, b, c; DSLNT: disialyllacto-N-tetraose.
Figure 1. (a) Basic building blocks of human milk oligosaccharides; (b) effect of Secretor and Lewis genes on the composition of HMOs; and (c) main neutral fucosylated, neutral non-fucosylated and acidic HMOs. Abbreviations: 2′FL: 2′-fucosyllactose; 3FL: 3-fucosyllactose; LNFP I, II, III, IV, V, VI: lacto-N-fucopentaose I, II, II, IV, V, IV; F-LNH I, II: fucosyl-lacto-N-hexaose I, II; A-Tetra: A-tetrasaccharide; DFL: difucosyllactose; LNDFH I, II: lacto-N-difucosylhexaose I, II; LNnDFH: lacto-N-neodifucosylhexaose; DFLNH I, II: difucosyllacto-N-hexaose I, II; DFLNnH: difucosyllacto-N-neohexaose; DF-p-LNH: difucosyl-para-lacto-N-hexaose; DF-p-LNnH: difucosyl-para-lacto-N-neohexaose; TF-LNH: trifucosyl-lacto-N-hexaose; LNT: lacto-N-tetraose; LNnT: lacto-N-neotetraose; LNH: lacto-N-hexaose; LNnH: lacto-N-neohexaose; 3′/6′GL: 3′-/6′-galactosyloligosaccharide; 3′/6′SL: 3′-/6′- sialyllactose; LST a, b, c: sialyllacto-N-tetraose a, b, c; DSLNT: disialyllacto-N-tetraose.
Nutrients 12 03568 g001
Figure 2. Change of the HMO content during the course of lactation. The mean content of total HMOs (yellow) decreases by up to 67% during the course of lactation, which has been shown by Viverge et al. [6] and Coppa et al. [9] (top images). Differentiation of HMOs into the three groups of neutral fucosylated, neutral non-fucosylated and sialylated HMOs shows a similar trend for the individual groups (Xu et al. [4] and Elwakiel et al. [5]). Compared to colostrum, these respectively decrease by up to 61%, 86% and 66% in mature milk.
Figure 2. Change of the HMO content during the course of lactation. The mean content of total HMOs (yellow) decreases by up to 67% during the course of lactation, which has been shown by Viverge et al. [6] and Coppa et al. [9] (top images). Differentiation of HMOs into the three groups of neutral fucosylated, neutral non-fucosylated and sialylated HMOs shows a similar trend for the individual groups (Xu et al. [4] and Elwakiel et al. [5]). Compared to colostrum, these respectively decrease by up to 61%, 86% and 66% in mature milk.
Nutrients 12 03568 g002
Figure 3. Most dominant HMOs of term-delivering mothers differentiated by the Secretor status. The ten most dominant neutral fucosylated (except for SeLe), the four most important neutral non-fucosylated and the six most important acidic HMOs are respectively displayed in red to light orange, blue and violet to pink.
Figure 3. Most dominant HMOs of term-delivering mothers differentiated by the Secretor status. The ten most dominant neutral fucosylated (except for SeLe), the four most important neutral non-fucosylated and the six most important acidic HMOs are respectively displayed in red to light orange, blue and violet to pink.
Nutrients 12 03568 g003
Table 1. Studies that quantified absolute human milk oligosaccharide concentrations.
Table 1. Studies that quantified absolute human milk oligosaccharide concentrations.
HMO Content/g∙L−1Period of Observation/dLocation of Studyn (mothers)Reference
3.5–18.51–90FranceN/A[6]
15.2–16.72–7France15[8]
12.9–20.94–120Italy46[9]
7.92–28Germany10[1]
8.6–19.610–120USA45[4]
5.3–6.5180Malawi88[4]
7.8–22.41–140China30[5]
Table 2. Secretor and Lewis genetic predisposition of different populations. Note that not all studies differentiated between all groups.
Table 2. Secretor and Lewis genetic predisposition of different populations. Note that not all studies differentiated between all groups.
Se+Le+/%SeLe+/%Se+Le/%SeLe/%Location of Studyn (Mothers/Children)Reference
69 20.0 9.0 1.0 Germany50[38,39]
71.724.53.8 Burkina Faso53[40]
73.323.33.3 Italy50[40]
73.017.010.0Germany30[29]
45.010.028.026.0Gambia60[3]
55.628.611.14.8Italy63[41]
46.9 34.418.8Spain32[42]
73.020.07.0-China30[5]
67.024.05.04.0Vietnam141[13]
75.019.04.02.0Switzerland25[43]
70.020.07.0 3.0Switzerland290[12]
75.611.511.51.3Brazil78[44]
76.717.24.31.7China116[37]
77.07.013.03.0Germany60[45]
58.320.816.74.2China24[46]
Table 3. Effects of HMOs on different pathogens. If not stated differently, the antiadhesive activity of HMOs describes the competitive interference of lectin-glycan association.
Table 3. Effects of HMOs on different pathogens. If not stated differently, the antiadhesive activity of HMOs describes the competitive interference of lectin-glycan association.
PathogenStrain/SubtypeHMO/HMO FractionActivityReference
Aspergillus fumigatusCBS 113.263′SLAntiadhesive[123]
Candida albicansSC5314Total HMOCellular and gene expression of hyphal morphogenesis[98]
Entamoeba histolyticaHM-1:IMSSTotal HMO, neutral non-fucosylated HMO, LNT, LNFP II, LNFP IIIAntiadhesive, cytoprotective[65]
Acinetobacter baumanniiATCC 19606Total HMOAntimicrobial[87]
Campylobacter jejuni81–176, 287ip, 84sp, 166ip, 10sp, 57spTotal HMO, 2′FLInvasion protection, antiadhesive[66,67]
Clostridium difficileN/ATotal HMO, neutral non-fucosylated HMO, 8 individual HMOsTcdA toxin inhibition[68,69]
Clostridium difficileN/A11 individual HMOsTcdB toxin inhibition[68]
Escherichia coliO18:K13′SLAntiadhesive[81]
Enterohemorrhagic Escherichia coliN/A20 individual HMOsHLT, Stx1, Stx2 toxin inhibition[70]
Enteropathogenic Escherichia coliO119, E2348/69, 01163, 0111:H2, 01736, 0119:H6, 851/71, O142:H6Total HMO, neutral HMO, fucosylated HMO, acidic HMO, 2′FL, 3FL, DFL, LNFP I, LNFP II, LNFP III, LNT, LNnT, 3′SL, 6′SLSta toxin inhibition, antiadhesive[71,72,73,74,75,76,77]
Enterotoxic Escherichia coliH4 CFA/I, 23 CFA/II, F18Acidic HMO, 2′FL, 3′SL, 6′SL, DSLNT, LST a, 3′SL, 3FLAntiadhesive[78,124]
Uropathogenic Escherichia coliCTF073, FVL 25 Fimbria P-likeTotal HMO, neutral, tri-, penta-, high molecular weight HMO, sialylated HMO, 3′SL, 6′SL, DSLNT, LST a, 3′SL3FLAntiadhesive, antiinvasive, cytoprotective[78,79,80]
Pseudomonas aeruginosaDSM17072′FL, 3FL, LNFP II, LNnFP V, LNnDFH II, 3′SLAntiadhesive[75,85,86]
Helicobacter pylori1832, CP22, 1351,3′SLAntiadhesive[82]
L. monocytogenesEGD-eTotal HMOAntiadhesive[83]
Neissereria Meningitidis C8013Acidic and neutral HMOAntiadhesive[84]
Salmonella entericaSerovar fyrisAcidic, neutral low weight HMO, 6′SL, 2′FL, 3FLAntiadhesive[74,75]
Staphylococcuss aureusUSA300Total HMOAntimicrobial, antibiofilm[87,125]
Streptococcus agalactiaeCNCTC 10/84, GB590, GB2, ST III (COH-1, A909, NCTC)Total HMO, neutral HMO, fucosylated HMO, LNT, LNnT, DFL, LNFP I, II, III, LNnH, LNDFHI, LST a, LST c, DSLNT, 3′SL, 6′SAntimicrobial, antibiofilm[87,99,100,101,102,103]
Streptococcus pneumoniaeR6, SIII, AIILNT, LNnT, 3′SL, 6′SL, LSTc, 3′SLNnT, 3′GLAntiadhesive[126,127,128]
Vibrio choleraeN/AAcidic HMO, neutral high. mol. weight HMO, 18 individual neutral and sialylated HMOs, 3′SLAntiadhesive, cholera toxin B inhibition[70,74,88]
Influenza virusH1N1, H1Nx, H9N23′SL, 6′SLAntiadhesive[90,91,95]
Human immunodeficiency virusN/ATotal HMOAntiadhesive[89]
NorovirusG.I, GII.4, GII.10, G.172′FL, 3FL, LNFP IAntiadhesive[92,93,94]
Respiratory syncytial virusNM2322′FL, 3′SL, LNnTAntiadhesive[95]
RotavirusOSU; G1P, G2P3FL, 3′SL, 6′SLAntiadhesive[107,129]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hundshammer, C.; Minge, O. In Love with Shaping You—Influential Factors on the Breast Milk Content of Human Milk Oligosaccharides and Their Decisive Roles for Neonatal Development. Nutrients 2020, 12, 3568. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12113568

AMA Style

Hundshammer C, Minge O. In Love with Shaping You—Influential Factors on the Breast Milk Content of Human Milk Oligosaccharides and Their Decisive Roles for Neonatal Development. Nutrients. 2020; 12(11):3568. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12113568

Chicago/Turabian Style

Hundshammer, Christian, and Oliver Minge. 2020. "In Love with Shaping You—Influential Factors on the Breast Milk Content of Human Milk Oligosaccharides and Their Decisive Roles for Neonatal Development" Nutrients 12, no. 11: 3568. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12113568

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop