You are on page 1of 12

Carbohydrate Polymers 276 (2022) 118738

Contents lists available at ScienceDirect

Carbohydrate Polymers
journal homepage: www.elsevier.com/locate/carbpol

Human milk oligosaccharides and infant gut microbiota: Molecular


structures, utilization strategies and immune function
Bin Zhang a, b, Long-Qing Li b, Feitong Liu c, Jian-Yong Wu b, *
a
SCUT-Zhuhai Institute of Modern Industrial Innovation, School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of
Food Nutrition and Human Health, South China University of Technology, Guangzhou 510640, China
b
Research Institute for Future Food, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong,
China
c
H&H Group Global Research and Technology Center, Guangzhou 510700, China

A R T I C L E I N F O A B S T R A C T

Keywords: Human milk oligosaccharides (HMOs) are a unique class of non-digestible carbohydrates present in the mother
Human milk oligosaccharides milk, which play a key role in the development of infant gut microbiota, epithelial barrier and immune function.
Utilization strategies The deficiency of HMOs in the bovine milk-based infant formula has been widely recognized as a major culprit
Infant microbiota
for the much higher incidence of immune disorders of formula-fed infants. This report was to give an up-to-date
Immune functions
review on the structure characteristics of HMOs and the possible mechanisms, and strategies for their cellular
Commercial applications
uptake, and metabolism by the gut bacteria and the associated effects on the infant gut microbiome, and immune
function. Most previous studies have been carried out in animals or in vitro model systems on the utilization
strategies for HMOs in infant bacteria and their roles in infant microbiome, and gut immune function. A few
HMO molecules have been synthesized artificially and applied in infant formulas.

1. Introduction been identified (Petschacher & Nidetzky, 2016). Most of the HMOs are
resistant to degradation by the gastric acid and digestive enzymes in the
The gold standard for commercial infant formula milk is the close infant gastrointestinal tract, and can reach the large intestine for mi­
similarity to the composition of breast milk, which is achieved through crobial fermentation. HMOs provide a major source of carbohydrates
the design and optimization of the major ingredients such as proteins, which are selectively utilized by the gut bacteria, having a profound
fats and functional oligosaccharides. Although bovine milk as the basal impact on the microbial composition of gut microbiota.
component of formula milk can meet the basic requirements for energy The infant gut microbiota evolves with the host since birth. The
and nutrition of infant growth, it has significant differences from human microbial composition of infant gut microbiota undergoes significant
breast milk in the major ingredients and their contents. In particular, changes during the first three years of life, and can be affected by the
human milk oligosaccharides (HMOs) rank the third most abundant delivery mode, feeding pattern and environment. Vaginally delivered
group of constituents (5–15 g/L) of mature breast milk, after lactose infants on natural birth acquire bacterial communities resembling their
(55–70 g/L) and lipids (16–39 g/L). The composition of HMOs is far own mother's vaginal microbiota, dominated by Lactobacillus, Prevotella,
more diverse than the oligosaccharides in the milk of other mammals. and Sneathia spp., whereas C-section infants harbored bacterial com­
Up to date, more than 200 different molecular structures of HMOs have munities which are similar to the mother's skin surface, dominated by

Abbreviations: ABC, ATP binding cassette; CAZymes, carbohydrate active enzymes; DSLNT, Disialyllacto-N-tetraose; DP, degree of polymerization; Fuc, fucose; 2′ -
FL, 2′ -fucosyllactose; 3-FL, 3-fucosyllactose; FOS, fructo-oligosaccharides; FUT2, α1-2 fucosyltransferase; FUT3, α1-3/4 fucosyltransferases; 4′ -GL, 4′ -gal­
actosyllactose; 3′ -GL, 3′ -galactosyllactose; 6′ -GL, 6′ -galactosyllactose; Gal, galactose; GlcNAc, N-acetylglucosamine; Glc, glucose; GOS, galacto-oligosaccharides;
GPCRs, G protein-coupled receptors; HDACs, histone deacetylases; HIF, hypoxia inducible factor; HMOs, human milk oligosaccharides; IBS, irritable bowel syn­
drome; LNH, lacto-N-hexaose; LNnT, lacto-N-neotetraose; LNT, lacto-N-tetraose; LNFP I, lacto-N-fucopentaose I; LNFP II, lacto-N-fucopentaose II; LNFP III, lacto-N-
fucopentaose III; LST, sialyl-lacto-N-tetraose; NEC, necrotizing enterocolitis; SCFAs, short-chain fatty acids; Sia, sialic acid; 3′ -SL, 3′ -sialyllactose; 6′ -SL, 6′ -
sialyllactose.
* Corresponding author.
E-mail addresses: zhangb@scut.edu.cn (B. Zhang), longqing21.li@connect.polyu.hk (L.-Q. Li), erin.liu@hh.global (F. Liu), jian-yong.wu@polyu.edu.hk (J.-Y. Wu).

https://doi.org/10.1016/j.carbpol.2021.118738
Received 8 July 2021; Received in revised form 28 September 2021; Accepted 28 September 2021
Available online 9 October 2021
0144-8617/© 2021 Elsevier Ltd. All rights reserved.
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

Staphylococcus, Corynebacterium, and Propionibacterium spp. (Domi­ differences in the microbial composition of gut microbiota between
nguez-Bello et al., 2010). Bacteria present in newborn babies of a few formula-fed and breast-fed infants (Marcobal et al., 2010).
days old are usually aerobic bacteria and facultative anaerobic bacteria. Although much information is available on the HMO molecular
As oxygen in the gut is gradually consumed by the aerobic bacteria, specificity, health-promoting functions (Bode, 2012; Triantis, Bode, &
anaerobic bacteria such as Bifidobacterium and Bacteroides increasingly van Neerven, 2018; Walsh, Lane, van Sinderen, & Hickey, 2020; Yu
dominate the infant microbiota (Albenberg et al., 2014). The earliest et al., 2018), to the best of our knowledge, limited information has been
microbiome is enriched in genes which facilitate lactate utilization with documented for HMO utilization strategies of typical infant bacteria
a relatively simple structure, and mainly with degraders of simple car­ particularly Bacteroides and Lactobacillus, and the roles of HMOs in the
bohydrates such as HMOs. After weaning, solid foods provide gut bac­ development of infant microbiome and gut immune function, as well as
teria with diverse fermentation substrates, resulting in stable and the recent application of artificial HMOs in infant formula and regula­
mature microbiota community and function, with the ability to degrade tory framework. This review aims to make a critical summary on our
complex carbohydrates. Weaning is an important milestone in the current knowledge and understanding of the possible mechanisms and
development of gut microbiome, starting the transition from infancy to strategies for the cellular uptake and metabolism of HMOs by gut bac­
childhood. Ingestion of solid foods causes an increased abundance of teria and the associated effects on the infant gut microbiome and im­
Bacteroidetes, and levels of short-chain fatty acids (SCFAs) and the mune functions. It also provides an illustration on the major structure
enrichment of genes associated with carbohydrate utilization and characteristics of HMOs and a brief survey on the commercial applica­
vitamin biosynthesis (Koenig et al., 2011). After the age of three, the gut tions of HMOs in infant formulas.
microbiota has almost reached a stable community composition and gut
function, which are characteristic of the adult microbiome (Yang et al., 2. Structural characteristics of HMOs
2016).
Evidence from long-term epidemiological studies shows that The molecular structure of HMOs is a determining factor on their
formula-fed infants are more prevalent than breast-fed infants in a selective utilization by the bacteria in the gut microbiota and the
number of childhood disorders including necrotizing enterocolitis consequent effects. Regardless of the structural diversity, HMOs are
(NEC), irritable bowel syndrome (IBS), obesity, allergies, and eczema generally composed of five sugar residues with a degree of polymeri­
(He, Liu, Leone, & Newburg, 2014; Quigley, Carson, Sacker, & Kelly, zation (DP) ranging from 3 to 32. HMOs can be classified in three main
2016; Victora et al., 2016). These health problems prevalent with the structural groups, fucosylated (taking up 35–50% of the total), non-
formula-fed infants are closely associated with an aberrant gut micro­ fucosylated neutral (42–55%), and sialylated HMOs (12–14%) (Totten
biota. HMOs are widely believed to play an important role for the et al., 2012). Some terminal β1-4 chain-linked Gal can be cleaved off by

Fig. 1. HMO structures. (A) Possible linkages of HMO building blocks; (B) lactose can be fucosylated or sialylated to form HMOs; (C) lactose can be elongated
enzymatically in repeats of lacto-N-biose (type I), or N-acetyllactosamine (type II). (D) Galactosyl residues are linked to lactose through β1-3, β1-4-and β1-6 linkages
to form galactosyllactoses. (E) Elongated type I or II chains can be fucosylated or sialylated in different linkages to form a variety of structural isomers. (Abbre­
viations: 2′ -FL, 2′ -fucosyllactose; 3-FL, 3-fucosyllactose; 3′ -SL, 3′ -sialyllactose; 6′ -SL, 6′ -sialyllactose; LNnT, lacto-N-neotetraose; 3′ -GL, 3′ -galactosyllactose; 4′ -GL, 4′ -
galactosyllactose; 6′ -GL, 6′ -galactosyllactose; LNH, lacto-N-hexaose; LNFP I, lacto-N-fucopentaose I; LNFP II, lacto-N-fucopentaose II; LNFP III, lacto-N-fucopentaose
III; LST a, sialyl-lacto-N-tetraose a; LST b, sialyl-lacto-N-tetraose b; DSLNT, Disialyllacto-N-tetraose) (Cheng, Akkerman, Kong, Walvoort, & de Vos, 2021).

2
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

lactase in infant small intestine, and approximately 1% of the ingested modifications could lower the enzyme expression or enzyme activity
HMOs are absorbed directly into the systemic circulation and excreted in (Soejima et al., 2012). For example, breast milk from some non-secretor
the urine (Dotz et al., 2014). All HMOs are built on a lactose core women has a low concentration of 2′ -FL, due to the redundancy of
(Galβ1–4Glc) at the reducing end, elongated or branched with five certain pathways, or enzyme activity retained by gene mutations (Totten
monosaccharide building blocks (Fig. 1A). i.e., glucose (Glc), galactose et al., 2012).
(Gla), N-acetylglucosaminic acid (Neu5c), fucose (Fuc) and sialic acid Although mother's genetic profile was found to have a major effect
(Sia). Lactose can be fucosylated either at the terminal Gal by α1-2 on the HMO composition in the breast milk particularly fucosylated
linkage to generate 2′ -fucosyllactose (2′ -FL) or at Glc in α1-3 linkage to HMOs, stage of lactation is a key influencing factor of the HMO quantity.
form 3-fucosyllactose (3-FL) (Fig. 1B). Alternatively, lactose can be The yield of total and sialylated HMOs from the same woman is higher in
connected by sialic acids with α2-3 or α2-6 linkage to form 3′ -sia­ breast milk from the first few weeks postpartum and decreases over the
lyllactose (3′ -SL) and 6′ -sialyllactose (6′ -SL), respectively (Fig. 1B). lactation course. In addition, preterm milk is normally associated with
HMO connects galactose β-1,3-N-acetylglucosamine (Galβ1-3GlcNAc, higher HMO concentration compared with term milk (Gabrielli et al.,
type 1 chain structure) on the basis of lactose to form lacto-N-tetraose 2011).
(LNT) or linked to N-acetyllactosamine (Galβ1-4GlcNAc, type 2 chain
structure) to form lacto-N-neotetraose (lacto-N-neotetraose, LNnT), as 3. Microbial utilization strategies of HMOs in Bifidobacteria,
shown in Fig. 1C. The development and application of new and Bacteroides and Lactobacillus
advanced analytical techniques may be helpful for profiling and struc­
ture determination of HMOs, such as matrix-assisted laser desorption/ The complete degradation of HMOs with diverse molecular struc­
ionization-mass spectrometry (MALDI-MS) (Huang et al., 2020) and tures requires a series of glycoside hydrolases and/or membrane trans­
multi-dimensional nuclear magnetic resonance (NMR) spectroscopy (Shi porters. These enzymes and protein transporters are available in the
et al., 2021). infant gut microbiota and essential for the uptake, metabolism and
The genetic background and physiological status of women are utilization of HMOs by the gut bacteria. Therefore, understanding of the
among the chief factors affecting breast milk composition and cause genes encoding the required enzymes and transporters can help reveal
individual differences with race, region, diet, age, and lactation stage the mechanisms and strategies for degradation and utilization of HMOs.
(Seppo et al., 2019). The type and content of HMOs in breast milk show Many of these genes have been found and identified from various bifi­
dramatic variations with individual women and lactation stages of the dobacterial species that are among the most active species for the
same woman. The composition variation of fucosylated HMOs is metabolism of HMOs in infant gut microbiota (Walsh et al., 2020). In
determined by the activities of three or more fucosyltransferases addition, certain species of Bacteroides and Lactobacillus have also shown
(Andreas et al., 2015; Kunz et al., 2017), which are encoded by the a high HMO metabolic capacity such as Bacteroides fragilis, Bacteroides
women's secretor (Se) and Lewis (Le) blood group status. The majority of vulgatus and Lactobacillus casei (Bidart, Rodríguez-Díaz, Monedero, &
women express α1-2 fucosyltransferase (FUT2) that attach fucose resi­ Yebra, 2014; Marcobal et al., 2010; Yu, Chen, & Newburg, 2013).
dues (Fuc) to a terminal Gal through an α1-2 linkage. For example, 2′ -FL It has been found that formula-fed and breast-fed infants have
(Fucα1-2Galβ1-4Glc) and LDFT-I (Fucα1-2Galβ1-3GlcNAcβ1-3Galβ1- notable differences in the microbial diversity and the keystone species of
4Glc) are most abundant HMOs in breast milk of “secretor women”. In gut microbiota. The gut microbiota of breast-fed infants usually has a
contrast, non-secretor women do not express FUT2 and therefore these relatively low microbial diversity but a high abundance in Bifidobacte­
HMOs are absent in the breast milk. The Le gene encodes α1-3/4 fuco­ rium breve (B. breve) and Bifidobacterium longum subsp. longum
syltransferases (FUT3), which may connect to Fuc through α1-3 linkages (B. longum), while that of formula-fed or mix-fed infants has a slightly
such as LNFP-III (Galβ1-4Fucα1-3GlcNAcβ1-3Galβ1-4Glc), or through higher diversity and a higher abundancy in B. adolescentis,
α1-4 linkages such as LDFT-II (Galβ1-3Fucα1-4GlcNAcβ1-3Galβ1-4Glc) B. catenulatum, Bacteroides, Proteus and Clostridium (Azad et al., 2013;
(Moukarzel & Bode, 2017). According to the expression of FUT2 and Van Daele, Knol, & Belzer, 2019). The microbiota of breast-fed infants
FUT3 (or maternal Se and Le blood type), HMO profiles in the breast has been shown to degrade mainly the nutrients in breast milk, while
milk can be divided into four types: Se+Le+, Se− Le+, Se+Le− , and that of formula-fed infants is rich with degraders of plant-based foods
Se− Le− . The compositions of fucosylated HMOs of each type are sum­ (Koenig et al., 2011).
marized in Table 1.
Therefore, the detection of the secretion type and Lewis blood group 3.1. HMO utilization strategies of Bifidobacteria
status can be used to predict the composition and distribution of HMOs
in breast milk. However, the composition of HMOs in breast milk does HMOs can have a significant impact on the formation and develop­
not completely correspond to the detection of the above genotypes. The ment of infant gut microbiome, especially the establishment of Bifido­
expression of various glycosyltransferases is regulated by the Se and Le bacterium-dominated microbial community. Certain bifidobacterial
genes, and gene deletion mutations or amino acid sequence species can selectively utilize HMOs with specific structures. Bifidobac­
teria mostly have gene clusters encoding specific HMO degrading en­
Table 1 zymes, through the gene-encoded glycosidase and specific transporters.
Biosynthesis of fucosylated HMOs depends on Secretor and Lewis blood group Among the bifidobacterial species of breast-fed infants, B. longum and
status. B. breve are most frequently observed, whereas B. bifidum, B. longum
Gene type Fucosyltransferase Fucosylated HMOs subsp. infantis, B. pseudocatenulatum, and B. catenulatum are less frequent
HMOs (Turroni, Van Sinderen, & Ventura, 2011).
FUT2 FUT3 2′ -FL + LNFP- 3-FL + LNFP- LNFP-
The HMO utilization strategies employed by infant bifidobacteria are
I III II shown schematically in Fig. 2. There are two primary degradation
strategies for Bifidobacteria, i.e., intracellular, transport-dependent
α1-2 α1-3/4 α1-2 α1-3 α1-4
strategy and extracellular, glycosidase-dependent strategy (Katayama,
Secretor + +
2016). With extracellular digestion strategy, the HMOs are hydrolyzed
(Se)
Lewis (Le) + + + into mono- or di-saccharides outside the bacterial cells by cell-wall
Se+Le+ + + + + + associated glycosidases, and the hydrolyzed sugars are transported
Se+Le− + − + + − into the cells for further degradation in the cytoplasm. With intracellular
Se− Le+ − + − + + digestion strategy, the HMOs are directly transported into the cells by
Se− Le−
ATP binding cassette (ABC) transporters, and then hydrolyzed into
− − − − −

3
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

Fig. 2. Possible strategies for human milk oligosaccharide (HMO) consumption in B. bifidum, B. infantis, B. breve, and B. longum (Smilowitz, Lebrilla, Mills, German, &
Freeman, 2014). (Abbreviations: GNB, galacto-N-biose; LNB, lacto-N-biose).

monosaccharides or smaller oligosaccharides by glycosidases in the hydrolases (GHs) are involved in the enzymatic degradation process of
cytoplasma. HMOs, releasing monosaccharides in cells (Sela et al., 2012). After being
B. infantis shows intracellular HMO degradation mechanism through degraded into monosaccharides, HMOs are assimilated to the central
the action of many oligosaccharide transporters. Whole HMO molecules metabolic pathway of B. infantis by means of the bifid shunt, yielding
are transported into the cytoplasm through solute binding proteins acetic and lactic acids as the end products (Kim et al., 2013). A wide
(SBPs), and then metabolized intracellularly. Intracellular glycosyl variety of GHs can be secreted by B. infantis, such as α-sialidase,

Fig. 3. The role of HMOs in infant gut immune function. (a) HMOs can stimulate the growth and colonization of commensal bacteria, and (b) modulate the formation
of metabolites such as SCFAs, which can influence the gut immune system; (c) HMOs can reduce the pathogenic infections by acting as decoy receptors and (d)
stimulate epithelial cell maturation.

4
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

α-fucosidase, β-galactosidases and β-N-acetylhexosaminidases, and the Schwab, 2016).


enzyme affinity covers almost all HMOs in breast milk, including 3′ -SL, The HMO degradation strategies and metabolic pathways are highly
6′ -SL, 2′ -FL, 3-FL, LNnT, and LacNAc (Garrido, Ruiz-Moyano, & Mills, dependent on the specific Bifidobacteria species or strains in view of the
2012; Sela et al., 2011; Sela et al., 2012). For example, B. infantis ATCC obvious differences in the gene clusters related to the HMO digestion.
15697 is a typical strain with strong HMO utilization capacity (Ward, Likewise, a specific species or strain of Bifidobacterium can have different
Niñonuevo, Mills, Lebrilla, & German, 2007), due to the certain genes of utilization capabilities for specific types of HMOs (Lawson et al., 2019).
this bacteria encoded Family 1 SBPs and a large number of glycosyl The digestion of certain HMO structures often involves multiple gene
hydrolases (GHs) (Garrido, Dallas, & Mills, 2013). Family 1 SBPs have clusters, and small differences between highly similar homologous se­
extremely high abundance in these microbial cells (Garrido, Kim, quences can also lead to differential expression of related enzymes and
German, Raybould, & Mills, 2011), part of which contributed by the transporters. It has been reported that Bifidobacteria strains lacking ABC
expression of various SBPs-related genes induced by HMOs (Garrido transporters may express extracellular glycosidases to utilize HMOs
et al., 2015). SBPs are members of the ABC transporter superfamily for (Matsuki et al., 2016; Zabel et al., 2019).
oligosaccharides, showing affinity for many different HMOs (Garrido
et al., 2011) (Fig. 3). 3.2. HMO utilization strategies of Bacteroides and Lactobacillus
B. bifidum consumes HMOs through an extracellular glycosidase-
dependent molecular mechanism, utilizing various extracellular GHs Bacteroides can ferment or catabolize not only a variety of plant-
to achieve the initial degradation of HMOs outside the cells (Kitaoka, based dietary fibers, but also host-derived glycoconjugates from the
2012). Extracellular GHs of B. bifidum have strong HMO degradation mucus layers of gut (Bjursell, Martens, & Gordon, 2006; Salyers, Ver­
capacity, similar to the endogenous GHs of B. infantis. The intermediate cellotti, West, & Wilkins, 1977). Carbohydrates that can be degraded
products (e.g., monosaccharides, disaccharides) bind to SBPs and enter and consumed by Bacteroides are similar in the sugar units to HMOs,
the cells. Two unique extracellular GHs have been found in B. bifidum, i. including N-acetylglucosamine, galactose, fucose, sialic acid, and N-
e., lacto-N-biosidase and endo-N-acetylgalactosaminidase. For example, acetylgalactosamine, suggesting the possibility of HMOs being degraded
the lacto-N-biosidase cleaves LNT to produce lacto-N-biose (LNB) and by Bacteroides.
lactose, then LNB combines with SBPs and enters B. bifidum cells (Wada For example, Bacteroides fragilis has been found having the same
et al., 2008). The related genes that control this process are located on genes that are related to mucus glycoconjugate metabolism in the HMO
the LNB/GNB gene cluster, which is present in most bifidobacteria and degradation process (Marcobal et al., 2011). Bacteroides have a variety
related to the LNT degradation, and its expression is induced by LNT or of strategies for degrading HMOs to various degrees, and the mechanism
other HMOs (Garrido et al., 2015; Kitaoka, Tian, & Nishimoto, 2005; of HMO utilization as the sole carbon source is connected to the in­
Nishimoto & Kitaoka, 2007). In addition, the intermediate products of duction by HMOs of specific gene expressions. Through the analysis of
HMO degradation such as some disaccharides enter the cells first and the annotated genes of related carbohydrate active enzymes (CAZymes)
may then be excreted from the cells as monosaccharides after being in the genomes of Bacteroides fragilis ATCC25285 and Bacteroides vul­
hydrolyzed by intracellular enzymes. B. bifidum SC555 was cultured gatus ATCC8482, Marcobal et al. (2010) attained multiple CAZyme in­
under the conditions of 2′ -FL, 3-FL or 6′ -SL, but could not digest fucose formation regarding the glycoside hydrolase families that may be
and sialic acid when lactose was the sole carbon source (Garrido et al., involved in the HMO degradation, including GH2 (α-galactosidase),
2015). This may have been attributed to the lack of metabolic pathways GH3 (β-N-acetylgalactosaminidase), GH20 (β-hexosaminidase), GH27
for consuming fucose and sialic acid. During its growth, fucose and sialic (α-N-acetylgalactosaminidase), GH29 (α-L-fucosidase), GH33 (siali­
acid may be released out of the cells and used by other bacteria or even dase), GH42 (β-galactosidase) and GH95 (α-1,2-fucosidase). The type
pathogens in the gut, causing adverse effects to the host (Egan, Moth­ and number of GHF genes are different in the Bacteroides fragilis
erway, Ventura, & van Sinderen, 2014; Egan, Motherway, Ventura, & ATCC25285 and Bacteroides vulgatus ATCC8482, which further explains
van Sinderen, 2014). the differences in the HMO degradation capacity and metabolic strategy
B. breve is one of the dominant species in infant gut. Various B. breve among different species/strains of Bacteroides. For example, Bacteroides
strains have different degradation capabilities for HMOs, depending on fragilis ATCC25285 showed a higher degradation capacity for non-
the type of GHs encoded by the related genes. Table 2 summarizes the fucosylated HMOs and long-chain oligosaccharides compared with
HMO degradation profiles by selected strains. Although B. breve encodes Bacteroides vulgatus ATCC8482 (Xu et al., 2007). It appears that each
GH95 α-fucosidase, only strains encoding GH29 α-fucosidase can utilize bacterium has adapted during evolution to retain a unique metabolic
2′ -FL, LNFP I, LNFP III and other fucosylated HMOs (Ruiz-Moyano, niche, which can provide growth advantages to compete the mixed HMO
Totten, Garrido, Smilowitz, & Mills, 2013). Certain B. breve strains show substrates (Sela & Mills, 2010).
a high degradation capability for sialylated HMOs, such as sialyl-LNT. These is very limited information about the molecular utilization
Compared to neutral HMOs, B. breve SC95 prefers to utilize sialylated strategies of Lactobacillus species for HMOs. It has been reported that
HMOs (Thomson, Medina, & Garrido, 2018). B. breve UCC2003 gene L. casei could degrade HMOs with different transport systems and
clusters encode sialic acid-ABC transporters and intracellular α-siali­ metabolic pathways compared to Bifidobacterium (Bidart, Rodríguez-
dases and other enzymes for utilization of sialic acids from HMOs or the Díaz, Monedero, & Yebra, 2014). L. casei contains a special gene cluster
ones other bifidobacteria excreted (Egan, Motherway, Ventura, & van gnbREFGBCDA, which participates in the metabolism of GNB, LNB and
Sinderen, 2014; Egan, Motherway, Kilcoyne, et al., 2014). As for the N-acetylgalactosamine (Bidart et al., 2014). However, during the
neutral HMOs such as LNT and LNnT, B. breve can utilize these through metabolism process, the N-acetylgalactosamine parts of LNB and LGB
the above two degradation strategies. enter different pathways, GnbG (phospho-β-galactosidase) participates
Most B. longum strains consume both fucosylated HMOs (e.g., 2′ -FL, in the hydrolysis of LNB and GNB, but GnbF (N-acetylgalactosamine-6P
3-FL, LDFT) but also sialylated HMOs (e.g., 3′ -SL, 6′ -SL) as summarized deacetylase) and GnbE (galactosamine-6P isomerase/deaminase) are
in Table 2. However, the degradation strategy differs among different only related to GNB degradation, while the utilization of LNB by L. casei
strains (Yu, Chen, & Newburg, 2013). The HMO degradation strategy of depends on nagA (N-acetylglucosamine-6P deacetylase). The galactose
B. longum SC596 is similar to that of B. infantis, and the new gene cluster degradation of LNB and LGB is accomplished through the tagatose-6P
of B. longum SC596 can encode intracellular GHs, related ABC trans­ pathway (Bidart et al., 2014).
porters, and SBPs with a unique capability for binding and degrading
LNT, SL, FL and other acidic and neutral HMOs (Garrido et al., 2016). In 3.3. Cross-feeding effect between HMO degraders
addition, some B. longum strains isolated from the intestinal tract of
infants can only consume 2′ -FL, 3′ -FL and LNnT (Bunesova, Lacroix, & In the infant colon, HMOs are partially or completely degraded by

5
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

Table 2
Degradation of HMOs by selected strains of gut bacteria.
Species Strain number Glc/Lac 2′ -FL 3′ -FL 3′ -SL 6′ -SL LNnT LDFT Reference

B. infantis
BSM28-1 + + + + + Bunesova et al. (2016)
TPY6-2 + + + Bunesova et al. (2016)
DSM20082 + + + + Bunesova et al. (2016)
ATCC25962 + + + + + Garrido et al. (2015)
ATCC17930 + + + + + Garrido et al. (2015)
ATCC15697 Ward et al. (2007)
B. adolescenlis ATCC15703 + − − − − − Thongaram, Hoeflinger, Chow, and Miller (2017)

B. animalis
ATCC25527 + − − − − − Thongaram et al. (2017)
Bb-12 + − − − − − Thongaram et al. (2017)
Bf-6 + − − − − − Thongaram et al. (2017)
DSM10140 + − − − − − Thongaram et al. (2017)
JCM10602 + − − − − Garrido et al. (2015)

B. breve
DSM20213 − − − − − Bunesova et al. (2016)
TPY10-1 − − + Bunesova et al. (2016)
ATCC15700 + − − − − + Thongaram et al. (2017)
M-16V + − − − − − Thongaram et al. (2017)

B. bifidum
ATCC29521 + − − − − − Thongaram et al. (2017)
JCM1254 + + + − + Garrido et al. (2015)
JCM1255 + − − − − Garrido et al. (2015)
JCM1209 − + + − + Garrido et al. (2015)
JCM7002 + + + + + Garrido et al. (2015)

B. kashiwanohense
DSM20213 + + − Bunesova et al. (2016)
TPY11-1 + + − − − Bunesova et al. (2016)

B. longum
DSM20088 + + − − + Bunesova et al. (2016)
BRS8-2 + + + − + Bunesova et al. (2016)
TPY12-1 + + + − + Bunesova et al. (2016)
DSM20219 − − − − − Bunesova et al. (2016)
BSM11-5 + + − − − Bunesova et al. (2016)
BB536 + − − − − − Thongaram et al. (2017)
M-63 + + + + + + Thongaram et al. (2017)
ATCC15697 + + + + + + + Thongaram et al. (2017); Yu et al. (2013)
ATCC15708 + + + + + Yu, Chen, Kling, et al. (2013)
JCM7010 + + + + + Yu, Chen, Kling, et al. (2013)
JCM11347 + + + + + Yu, Chen, Kling, et al. (2013)

B. pseudolongum
DSM20092 − − − − − Bunesova et al. (2016)
BSM8-1 − − − − − Bunesova et al. (2016)
PV8-2 − − − − − Bunesova et al. (2016)

L. acidophilus
La-5 + − − − − + Thongaram et al. (2017)
NCFM + − − − − + Thongaram et al. (2017)
L. delbrueckii ATCC7830 + + − + − Yu, Chen, Kling, et al. (2013)
L. fermentum CECT5716 + − − − − − Thongaram et al. (2017)
L. gasseri ATCC33323 + − − − − − Thongaram et al. (2017)
L. jensenii ATCC25258 + − − − − − Thongaram et al. (2017)

L. johnsonii
ATCC11506 + − − − − − Thongaram et al. (2017)
ACD-1 + − − − − − Thongaram et al. (2017)
L. paracasei LCV-1 + − − − − − Thongaram et al. (2017)
L. plantarum LP-66 + − − − − + Thongaram et al. (2017)
L. reuteri DSM17938 + − − − − + Thongaram et al. (2017)

L. rhamnosus
DR20 + − − − − − Thongaram et al. (2017)
ATCC53103 + − − − − − − Thongaram et al. (2017); Yu, Chen, Kling, et al. (2013)
B. vulgatus ATCC8482 + + + + + Yu, Chen, Kling, et al. (2013)
B. fragilis ATCC25285 + + + + + Yu, Chen, Kling, et al. (2013)
B. thetaiotaomicron ATCC29148 + + + + − Yu, Chen, Kling, et al. (2013)
Clostridium perfringens ATCC13124 − − − − − Yu, Chen, Kling, et al. (2013)
Clostridium leptum ATCC29065 − − − − − Yu, Chen, Kling, et al. (2013)

Plus (+) indicating degradation of HMO and minus (− ) no degradation.

6
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

certain bacteria. The incompletely degraded HMO products can be Bifidobacteria. Because of the structural similarity of HMOs to intestinal
further degraded and utilized as the carbon and energy sources by other epithelial glycoconjugates, some SBPs can bind to the intestinal
bacteria. The cross-feeding effect of infant gut bacteria is particularly epithelium and the epithelial-associative SBPs enhance the binding of
prominent among bifidobacterial species/strains. The HMO utilization Bifidobacteria to intestinal epithelial cells. In addition, Bifidobacteria
by certain bifidobacterial strains can affect the growth of other strains by metabolism of HMOs can increase the expression of anti-inflammatory
sharing the degradation products. Cooperation with other bacteria cytokines and tight junction proteins (Chichlowski, De Lartigue,
(including both HMO consumers and non-HMO consumers) to promote German, Raybould, & Mills, 2012).
the degradation and metabolism of HMOs to the greatest extent can
contribute to increasing the diversity and attaining a dominant position 4.2. The impact of HMOs on infant microbiota changes
of Bifidobacteria (Lawson et al., 2019). A study has found a cross-feeding
mechanism for HMO degradation among four bifidobacteria strains with The intestinal microbiota has been co-evolved with the host since
complete HMO degradation gene clusters, which can theoretically birth. The composition of infant microbiota undergoes significant
metabolize all structural types of HMOs, but some HMO degradation changes during the first three years of life, and then gradually stabilizes
products were not further degraded in the in vitro culture (Gotoh et al., to form an adult-like shape. Infants begin to contact with a variety of
2018). When these strains were added to the infant fecal inocula, these microorganisms from mother's body and environment after being
HMO degradation products were further degraded by other bifido­ delivered, but not all microorganisms can colonize the infant gut. Only
bacterial species/strains, which significantly stimulated the growth of those microorganisms that adapt to the baby's intestinal environment
non-HMO consumers, and promoted the overall advantage of bifido­ can survive. Aerobic and facultative anaerobic bacteria usually exist in
bacteria in the microbial community. The cross-feeding effect of bifi­ newborn babies such as Streptococcus and Enterobacteria, and the species
dobacteria is very dependent on the extracellular degradation by are closely related to the delivery mode and environmental factors.
ectoenzymes. Only when the cells are lysed or the metabolites are Naturally delivered infants have similar gut bacterial varieties to the
secreted by the cells, can the HMO degradation products be utilized and mother's vagina, such as Lactobacilli, while newborn babies with C-sec­
metabolized through the cross-feeding effect (Mee, Collins, Church, & tion have more bacteria that are similar to those from the mother's skin
Wang, 2014; Nishiyama et al., 2018; Nishiyama et al., 2017). In addi­ and external environment, such as Staphylococcus, Streptococcus and
tion, other non-bifidobacterial species can also benefit from the cross- Propionibacterium (Dominguez-Bello et al., 2010). With growing age,
feeding mechanism. oxygen in the infant intestine is gradually consumed by aerobic bacteria,
and the decrease in oxygen content allows the growth of anaerobic
4. Effects of HMOs on infant microbiome and immune functions bacteria. For example, Bifidobacterium and Bacteroides gradually domi­
nate the infant microbiota (Albenberg et al., 2014). Low diversity and
4.1. The role of HMOs in the development of infant microbiome high variability are the major characteristics of the infant microbiota,
and the differences in the shape and composition of microbiota among
HMOs can play an important role in the development and maturation individual infants could be even higher than the adults (Chu et al.,
of infant immune function through their strong influence on the infant 2017).
gut microbiota and the gut epithelial barrier. HMOs can contribute to During the first six months of life, the infant microbiota has a single
the development infant microbiome ecology from the colonization of structure and a small number of species, mainly degraders of simple
aerobic, and facultative anaerobic bacteria (e.g., Escherichia coli, carbohydrates. Bifidobacterium comprises 90% of the gut microbiota in
Enterococcus, Streptococcus, Staphylococcus, etc.) and anaerobic bacteria breast-fed infants, whereas less Bifidobacterium and more Bacteroides are
(e.g., Bifidobacterium, Bacteroides, Clostridium, etc.). The infant intestinal typical of formula-fed infants (Underwood et al., 2015). Breast-fed in­
mucosal immune system gradually matures in the process of continuous fants normally have a lower microbial diversity in the gut compared
contact, colonization and stimulation of the mucosal epithelial surface with the formula-fed infants (Azad et al., 2013; Van Daele et al., 2019).
by these gut bacteria. Certain beneficial or commensal bacteria specif­ Bifidobacterium dominates the breast-fed infant microbiota, due to the
ically bind to the surface of infant mucosal epithelial cells to inhibit high capability of HMO utilization. HMOs are the preferred substrate for
pathogens through space-occupying effect, nutritional competition, or B. infantis and other bifidobacteria, which can stimulate the growth of
secretion of antimicrobial metabolites such as bacteriocins, or directly beneficial bacteria and prevent harmful bacteria from HMO utilization.
stimulate the immune cells in the lamina propria of intestinal mucosa to Bacterial fermentation of HMOs produces SCFAs, resulting in a low-pH
induce the maturation of the immune system (Bode & Jantscher-Krenn, environment in the colon, which is conducive to the growth of benefi­
2012). For example, Bifidobacterium, a common dominant bacterium in cial bacteria and inhibits potential pathogens (Yu, Chen, & Newburg,
the intestinal tract of breast-fed infants, can stimulate immune cells to 2013). Bifidobacterium has an advantage in competing with pathogens
secrete more IL-1 and IL-6, while IL-1 can promote Helper T cells to for limited nutrients, and its colonization helps reduce the proportion of
secrete IL-2 and B lymphocytes to secrete antibodies (Maslowski & pathogens and other harmful bacteria.
Mackay, 2011). In the meantime, IL-6 promotes the differentiation and A recent clinical study has shown that the secretor status of women is
maturation of B lymphocytes, making them secrete antibodies, and can closely associated with the abundance of Bifidobacterium species in the
also directly induce the proliferation of T lymphocytes (Maslowski & infant gut, whereas non-secretor breast milk delays the colonization of
Mackay, 2011). bifidobacteria, with promotion of Clostridium and Enterobacteria (Lewis
The HMO molecular utilization strategy of bifidobacteria and the et al., 2015). The bifidobacteria isolated from feces of secretor breast
microbial cross-feeding mechanism provide strong support for the milk-fed infants were able to utilize 2′ -FL as the sole carbon source,
establishment and balancing of infant microbiome, and HMOs may also indicating a more pronounced bifidobacterial metabolic activity tar­
use their specific structures to influence immune functions (Bode & geting fucosylated HMOs (Lewis et al., 2015).
Jantscher-Krenn, 2012). While increasing the diversity of the gut mi­ Numerous studies have shown that HMOs can selectively stimulate
crobial population is generally considered beneficial for adults, it is not the growth of beneficial strains such as B. infantis, B. breve, B. bifidum and
the case for infants, as bifidobacteria are mainly beneficial to infant B. longum (Asakuma et al., 2011; Bunesova et al., 2016; Thomson et al.,
nutrition absorption, disease prevention, and growth promotion (Bel­ 2018; Xiao et al., 2010). For example, B. longum and B. breve are more
kaid & Hand, 2014; Huda et al., 2014). HMOs promote the establish­ active to digest LNT, whereas B. infantis is prone to utilize LNnT, and
ment of gut barrier function and the development of the intestinal B. infantis and B. bifidum have shown a preference to utilize fucosylated
immune system in infants by affecting the microbiota composition and HMOs (Suzuki et al., 2008; Wada et al., 2008). In addition, the molec­
their metabolites. HMOs can induce genes encoding HMOs-type SBPs in ular structure of HMOs probably also affects the colonic fermentation

7
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

rate and SCFA production. Some studies have shown that fermentation directly act on intestinal epithelial cells. By regulating the gene
of SL and 2′ -FL increased the concentration of SCFAs and lactic acid, expression of epithelial cells, the glycans on the cell surface are changed,
though different HMOs had distinct SCFA profiles during the fermen­ thereby preventing pathogenic microorganisms from contacting intes­
tation. For example, Perdijk et al. (2019) established an in vitro tinal epithelial cells (Kunz, Rudloff, Baier, Klein, & Strobel, 2000).
fermentation model and found that SL can greatly increase the con­ It has been suggested that HMOs modulate the immune functions and
centration of propionic acid compared with GOS. Salli et al. (2019) regulate the responses of epithelial cells and immune cells by altering
found that 2′ -FL can increase the concentration of lactic acid in the in cell proliferation, differentiation, and apoptosis processes, as well as cell
vitro fermentation model compared with GOS. However, there have signaling pathways and cell surface glycosylation. The intestinal
been very few comparative studies to reveal the structure-microbial epithelium lining in the small intestine and colon is a physical barrier
fermentation outcomes of various HMOs. between the intestinal lumen and the circulatory system and the first
Jing, Chen, Yu, He, and Newburg (2016) have carried out in vitro line of defense for innate immunity. The permeability of intestinal
experiments to evaluate the 2′ -FL utilization by single strains (B. longum, epithelium mainly depends on the structure and expression of tight
L. acidophilus, etc.), and found that 2′ -FL, as the sole substrate for junction proteins. Nutrients can be absorbed by epithelial cells or be­
anaerobic fermentation of certain strains, was readily consumed, pro­ tween tight junctions. Perturbation of the intestinal barrier integrity can
ducing high concentrations of total SCFAs. Yu, Chen, Kling, et al. (2013) quickly lead to a series of immune responses, including gastrointestinal
found that Bifidobacterium effectively consumed 2′ -FL and 3-FL during infection, inflammation, and allergies (Vancamelbeke & Vermeire,
the in vitro anaerobic fermentation, and the increase of the lactic acid 2017). HMOs with different structures can have a direct effect on
and SCFA concentration significantly inhibited the growth of E. coli and different cells of the intestinal epithelial barrier. For example, Cheng,
Clostridium perfringens that can hardly use fucosylated HMOs. In a ran­ Kong, Walvoort, Faas, and de Vos (2020) used the LS174 T goblet cell
domized double-blind controlled multicenter clinical trial (Berger et al., line to test the effects of 2′ -FL and 3-FL on the intestinal mucus barrier
2020), it was found that after three-month intervention, infant formula function and found that 3-FL regulates IL-13 and TNF-α to improve
containing 2′ -FL and LNnT significantly increased the abundance of MUC2 gene expression. Similarly, HMO intervention increased the
Bifidobacterium and Streptococcus, and turned the microbiota of expression of MUC2 and decreased the permeability of the intestinal
caesarean section infant group of 0–6-month-old to the vaginal delivery epithelium in a neonatal mice model (Wu et al., 2019).
infants. The glycocalyx acts as the backbone of proteoglycans, which pro­
Short-chain fatty acids play an important role in connecting micro­ vides binding sites for microorganisms and shows intestinal barrier
biome with the host immune system. Acetic, propionic, and butyric acids function. Some in vitro studies have shown that HMOs can enhance the
are commonly found in the colon with relatively high concentrations, intestinal barrier function by changing the expression of glycocalyx.
and are absorbed by intestinal epithelial cells. SCFAs can modify cell Kong et al. (2019) proved that fucosylated HMO (2′ -FL and 3-FL)
gene expression, differentiation, proliferation and apoptosis and other significantly increased the thickness of absorbed albumin, and 3-FL
processes to change cell functions, interact with the activation of G increased the area coverage of albumin, heparan sulfate and hyaluron­
protein-coupled receptors (GPCRs), and inhibit histone deacetylases ic acid in the glycocalyx of Caco-2 cells, with enhanced stability of the
(HDACs) activity and other signal transduction pathways related to the glycocalyx and reduced adhesion of pathogenic bacteria. The immature
stabilization of hypoxia inducible factor (HIF). Microbiota and the im­ glycocalyx layer is associated with changes in the expression of heparan
mune system are connected through regulating GPCRs and the activity sulfate and hyaluronic acid and may lead to gastrointestinal diseases
of enzymes and transcription factors to further modulate the develop­ such as inflammatory bowel disease (IBD) (Kong et al., 2019).
ment, survival and function of IECS and leukocytes. Each SCFA has its HMOs can directly interact with infant intestinal epithelial cells,
special functions to the infant health. Both acetic and propionic acid affecting their gene expression, cell cycle, and cell surface glycosylation
have been suggested to reduce the risk of infant asthma (Fukuda et al., and regulating their growth, differentiation and apoptosis (Kuntz,
2011). Butyric acid can stimulate intestinal epithelial metabolism, Rudloff, & Kunz, 2008). He et al. (2014)) assessed the effect of HMOs on
consume intracellular oxygen, stabilize HIF, and enhance epithelial gene expression in fetal immature intestinal mucosa, and identified
barrier function (Kelly et al., 2015). In addition, butyric acid has been several immune-related pathways, such as immune cell communication,
shown to enhance regulatory T cells (Tregs) activity and proliferation, homeostasis, and immune differentiation. Sialyllactose can promote the
and inhibit the pro-inflammatory activity of a variety of other immune differentiation and growth of intestinal epithelial cells by up-regulating
cells, and reduce the risk of diarrhea by improving the gut barrier the expression of alkaline phosphatase, indicating that sialyllactose can
function (Thorburn et al., 2015). In addition to SCFAs, other metabolites be recognized by the receptors of the epithelial cells and beneficial to
derived from gut microbiota such as bacteriocins, amino acids and vi­ promote intestine barrier function (Kuntz et al., 2008). It has been re­
tamins, play a vital role in activating the intestinal immune response, ported that 3′ -SL had anti-inflammation effect by reducing the IL-12 and
thereby resisting external pathogens (Li et al., 2018). IL-8 expression in Caco-2 cells mediated by the NF-κB pathway and the
anti-inflammatory nuclear receptor PPARg (Zenhom et al., 2011).
4.3. The impact of HMOs on the gut immune function of infants Apart from regulating the mucus and glycocalyx, HMOs can stimu­
late the maturation of epithelial cells. Holscher, Davis, and Tappenden
The gut immune function in newborn infants is immature, with (2014)) used HT-29 and Caco-2Be mixed cells to model different dif­
characteristics of weak barrier function and low microbiota diversity as ferentiation patterns along the crypt-villi axis, and evaluated the effects
well as immature immune response (Yu et al., 2018). The HMO mole­ of LNnT, 2′ -FL and 6′ -SL on the differentiation, apoptosis and prolifer­
cules may modulate neonatal immunity indirectly, by altering host gut ation of epithelial cells. It was found that LNnT and 6′ -SL lead to a
microbiota composition and the metabolites as mentioned in the pre­ decrease in cell proliferation, and LNnT can reduce the permeability of
vious section, and also directly, by affecting host epithelial and immune epithelial cells (Holscher et al., 2014). In a later report from the same
cell responses, or systemic immune function in the intestine and other research group, Holscher, Bode, and Tappenden (2017)) found that 2′ -
sites. A recent study has suggested that some neutral HMO fractions FL, 3-FL and 6′ -SL alone or in combination can reduce cell proliferation,
produced significant immunomodulation activities in RAW264.7 increase the differentiation of intestinal epithelial HT-29 and Caco-2Be
macrophage cells via the upregulated expression of nitric oxide synthase cells, and promote the maturation of intestinal epithelium.
and cy-clooxygenase 2 (Zhang et al., 2019). HMOs have structural
similarity to the surface glycans of intestinal epithelial cells, and could
prevent infections by binding pathogens to intestinal epithelial cells
(Morozov, Hansman, Hanisch, Schroten, & Kunz, 2018). HMOs can also

8
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

5. Application of artificial HMOs in infant formula and and LNnT (0.6 g/L). The addition of 2′ -FL alone can also be applied to
regulatory framework food products for babies over 1 year old, or added with LNnT at a ratio of
2:1, with the maximum dosage of 2′ -FL (1.2 g/L) and LNnT (0.6 g/L). In
For many years, it has been difficult to isolate or synthesize human 2016, biotechnologically synthetized 2′ -FL and LNnT from Glycom A/S
milk oligosaccharides with defined molecular structures and to apply received Novel Food status in EU and GRAS notification in US. Since
these oligosaccharides in infant formula. Instead, alternative prebiotic 2018, Glycosyn and Inbiose have manufactured 2′ -FL with receiving
oligosaccharides such as galacto-oligosaccharides (GOS) and fructo- GRAS notification from US FDA. In 2018, LNT and DFL from Glycom A/S
oligosaccharides (FOS) have been supplemented to infant formula to and 3′ -SL from Genechem received the GRAS notification from the US
mimic HMOs. However, several studies have shown that these oligo­ FDA (Bych et al., 2019). In 2019, US FDA stipulated that the maximum
saccharides cannot mimic the prebiotic effects or biological function of dosage of 2′ -FL in infant formula for 0 to 12 months old infants is 2.4 g/
HMOs. For example, 2′ -FL has the advantages over FOS in reducing L, and the maximum dosage of LNnT in infant formula is 0.6 g/L. In the
intestinal pH and SCFA production, and is preferentially utilized by same year, Food Standards Australia and New Zealand (FSANZ) rec­
Bifidobacterium and Lactobacilli to colonize and form dominant bacteria ommended that the maximum dosage of 2′ -FL and LNnT in infant for­
in infant gut (Yu, Chen, Kling, et al., 2013). It has been reported that mula is 96 mg/100 kJ if only 2′ -FL is added, or 24 mg/100 kJ for LNnT
GOS can be utilized non-selectively by various bacteria, including and 96 mg/100 kJ for the sum of 2′ -FL and LNnT if both are added. The
pathogenic bacteria (Yu, Chen, Kling, et al., 2013). It is unlikely that establishment of valid and official standards by regulatory and profes­
GOS, FOS or other non-human oligosaccharides with different structures sional authorities has safeguarded the safety and nutrition quality of
from HMOs can mimic the structure-specific effects of HMOs (Bode & infant formula and other infant food products but also paved the way for
Jantscher-Krenn, 2012). their commercial applications.
The increasing awareness of the significance of HMOs in infant Among a variety of HMOs, 2′ -FL, 3′ -SL, 6′ -SL, 3′ -GL, LNnT and DFL
health and development has stimulated both research and commercial have been most widely used as ingredients of infant formula, dietary
interests world-wide in the synthesis and manufacture of HMOs or their supplements, and health foods (Fig. 4). The first infant formula products
active analogues as supplements to infant formula. In recent years, appeared in 2016 and more products have continued to emerge. For
major technology breakthroughs have been made for production of example, Abbott has applied 2′ -FL to the infant formula (Similac® Pro-
HMO molecules such as 2′ -FL, 3′ -SL, 6′ -SL, LNT, LNnT and DFL through Advance) with positive health benefits for brain development, reduction
cell engineering, chemoenzymatic and chemical synthetic processes. of infection, inflammation and NEC through microbiome modulation. In
This has brought revolutionary changes to the infant nutraceutical in­ addition, Aptamil applied 2′ -FL and 3′ -GL with the combination of FOS
dustry so that several companies have acquired the capability for and GOS to the infant formula to mimic HMO formula in breast milk.
manufacturing these HMOs at large scale such as Glycom A/S, Abbott Wyeth also uses HMO in a variety of infant formula products. For
Laboratories, Glycosyn LLC, Inbiose, Jennewein, and GeneChem etc. In example, 0.25 g/L 2′ -FL is added to S-26® infant formula stage 1–4; in
2015, chemically synthesized 2′ -FL and LNnT from Glycom A/S and the Illuma series, 2′ -FL and LNnT are added to 1–4 stage of infant for­
biotechnologically synthetized 2′ -FL from Jennewein received the first mula. In Biostime HMO infant formula, every 800 g of milk powder
regulatory approvals as GRAS from US Food and Drug Administration contains 2′ -FL in a dose as high as 3.12 g. In addition, Mead Johnson,
(FDA) (Salminen, 2017). The supplementation of synthetic or artificial Nestle, etc. have also applied HMOs to their infant formula products. In
HMO compounds to infant foods has already been approved by US, EU, Nestle's NAN formula milk, the addition amount of 2′ -FL is 0.26 g/L, and
Australia and other regulatory agencies (Bych et al., 2019). The Euro­ the addition amount of LNnT is 0.13 g/L; in Nestle's BEBA formulas, 2′ -
pean Food Safety Authority approved the use of 2′ -FL and LNnT in infant FL is added up to 1 g/L and LNnT is added up to 0.5 g/L. With the
formula and other infant food products in 2015, and indicated that the gradual expansion of HMOs in the field of infant formula, applications
addition of 2′ -FL and LNnT at a ratio of 2:1 (w/w) to infant foods is safe have also expanded to dietary supplements and medical foods. For
for babies under 1 year old, with the maximum dosage of 2′ -FL (1.2 g/L) example, Dupont added 2′ -FL and 3-FL into UHT milk and yogurt

Fig. 4. Overview over the commercial application in the last three decades of HMOs in infant formula products by some major food companies.

9
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

products (Christensen, Skov, Lendal, & Hornshoj, 2020). Biostime used Belkaid, Y., & Hand, T. W. (2014). Role of the microbiota in immunity and inflammation.
Cell, 157(1), 121–141.
freeze-drying technology to combine 2′ -FL with a variety of highly
Berger, B., Porta, N., Foata, F., Grathwohl, D., Delley, M., Moine, D., Charpagne, A.,
active probiotics, and has launched a new product for children aged 3 to Siegwald, L., Descombes, P., Alliet, P., et al. (2020). Linking human milk
12 and newborns from 1 to 6 months. oligosaccharides, infant fecal community types, and later risk to require antibiotics.
mBio, 11(2), e03196–e03219.
Bidart, G. N., Rodríguez-Díaz, J., Monedero, V., & Yebra, M. J. (2014). A unique gene
6. Concluding remarks and future directions cluster for the utilization of the mucosal and human milk-associated glycans galacto-
N-biose and lacto-N-biose in lactobacillus casei. Molecular Microbiology, 93(3),
There is accumulating evidence that HMOs play a key role in the 521–538.
Bjursell, M. K., Martens, E. C., & Gordon, J. I. (2006). Functional genomic and metabolic
development of the gut microbiome, and the immune system in early life studies of the adaptations of a prominent adult human gut symbiont, bacteroides
and are also beneficial to infant health and wellbeing. The beneficial thetaiotaomicron, to the suckling period. Journal of Biological Chemistry, 281(47),
effects of HMOs on the infant health are strongly dependent on their 36269–36279.
Bode, L. (2012). Human milk oligosaccharides: Every baby needs a sugar mama.
cellular uptake and metabolism by the gut bacteria and the associated Glycobiology, 22(9), 1147–1162.
effects on the infant gut microbiome and immune function. The uptake Bode, L., & Jantscher-Krenn, E. (2012). Structure-function relationships of human milk
of HMOs is structure-dependent and selective by the gut bacterial spe­ oligosaccharides. Advances in Nutrition, 3(3), 383S–391S.
Bunesova, V., Lacroix, C., & Schwab, C. (2016). Fucosyllactose and L-fucose utilization of
cies. This review has summarized the HMO utilization strategies of infant Bifidobacterium longum and Bifidobacterium kashiwanohense. BMC Microbiology,
major infant bacteria such as Bifidobacterium, Bacteroides and Lactoba­ 16, 248.
cillus, and their functions in the development of infant microbiome and Bych, K., Mikš, M. H., Johanson, T., Hederos, M. J., Vigsnæs, L. K., & Becker, P. (2019).
Production of HMOs using microbial hosts—From cell engineering to large scale
gut immune function. Currently only a few HMO molecules have been production. Current Opinion in Biotechnology, 56, 130–137.
synthesized artificially and found commercial application in infant and Cheng, L., Akkerman, R., Kong, C., Walvoort, M. T. C., & de Vos, P. (2021). More than
baby foods. sugar in the milk: Human milk oligosaccharides as essential bioactive molecules in
breast milk and current insight in beneficial effects. Critical Review in Food Science
Although significant research progress and technology break­
and Nutrition, 61, 1184–1200.
throughs have been achieved on the function and application of HMOs Cheng, L., Kong, C., Walvoort, M. T. C., Faas, M. M., & de Vos, P. (2020). Human milk
in infant nutrition and health, much more effort is needed for the oligosaccharides differently modulate goblet cells under homeostatic,
research and development in several important areas as listed below. proinflammatory conditions and ER stress. Molecular Nutrition & Food Research, 64
(5), Article e1900976.
Chichlowski, M., De Lartigue, G., German, J. B., Raybould, H. E., & Mills, D. A. (2012).
1. Only a few HMO molecules are available from commercial suppliers Bifidobacteria isolated from infants and cultured on human milk oligosaccharides
for research and even fewer for infant formula applications. affect intestinal epithelial function. Journal of Pediatric Gastroenterology and Nutrition,
55(3), 321–327.
2. Systematic studies at the molecular level through various -omics Christensen, A. S., Skov, S. H., Lendal, S. E., & Hornshoj, B. H. (2020). Quantifying the
approaches, e.g. genomic, metabolomic, proteomic, and glycomic, human milk oligosaccharides 2′ -fucosyllactose and 3-fucosyllactose in different food
are most helpful to reveal the molecular strategies and mechanisms applications by high-performance liquid chromatography with refractive index
detection. Journal of Food Science, 85(2), 332–339.
for the cellular uptake and metabolism by different microbes and the Chu, D. M., Ma, J., Prince, A. L., Antony, K. M., Seferovic, M. D., & Aagaard, K. M.
immune function of HMOs. (2017). Maturation of the infant microbiome community structure and function
3. The current knowledge and understanding of HMOs in infant health across multiple body sites and in relation to mode of delivery. Nature Medicine, 23
(3), 314–326.
has been mainly based on findings from animal or in vitro experi­ Dominguez-Bello, M. G., Costello, E. K., Contreras, M., Magris, M., Hidalgo, G., Fierer, N.,
ments. Further studies on human subjects or humanized animal & Knight, R. (2010). Delivery mode shapes the acquisition and structure of the initial
models are needed with consideration of the gene background and microbiota across multiple body habitats in newborns. Proceedings of the National
Academy of Sciences, 107(26), 11971–11975.
physiological status of mothers to ascertain the functions of the
Dotz, V., Rudloff, S., Blank, D., Lochnit, G., Geyer, R., & Kunz, C. (2014). 13C-labeled
mixed HMO formulation for the different individuals. oligosaccharides in breastfed infants'urine: Individual-, structure- and time-
4. More rigorous and placebo-controlled interventions with the HMOs dependent differences in the excretion. Glycobiology, 24(2), 185–194.
in human subjects are needed to justify the epidemiological evidence Egan, M., Motherway, O.’. C., Kilcoyne, M., Kane, M., Joshi, L., Ventura, M., & Van
Sinderen, D. (2014). Cross-feeding by Bifidobacterium breve UCC2003 during co-
and the significance for infant nutrition and immune function. cultivation with Bifidobacterium bifidum PRL2010 in a mucin-based medium. BMC
Microbiology, 14(1), 282.
Egan, M., O'Connell Motherway, M., Ventura, M., and van Sinderen, D. (2014).
Metabolism of sialic acid by Bifidobacterium breve UCC2003. Applied Environmental
Declaration of competing interest Microbiology, 80(14), 4414–4426.
Fukuda, S., Toh, H., Hase, K., Oshima, K., Nakanishi, Y., Yoshimura, K., , … Taylor, T. D.,
et al. (2011). Bifidobacteria can protect from enteropathogenic infection through
All authors declare there is no conflict of interest. production of acetate. Nature, 469, 543–791.
Gabrielli, O., Zampini, L., Galeazzi, T., Padella, L., Santoro, L., Peila, C., … Coppa, G. V.
Acknowledgements (2011). Preterm milk oligosaccharides during the first month of lactation. Pediatrics,
128(6), e1520–e1531.
Garrido, D., Dallas, D. C., & Mills, D. A. (2013). Consumption of human milk
This work was financially supported by the Hong Kong Scholar glycoconjugates by infant-associated bifidobacteria: Mechanisms and implications.
Program (XJ2019049), The Hong Kong Polytechnic University, and the Microbiology, 159(4), 649–664.
Garrido, D., Kim, J. H., German, J. B., Raybould, H. E., & Mills, D. A. (2011).
111 Project (B17018). Oligosaccharide binding proteins from Bifidobacterium longum subsp. infantis reveal a
preference for host glycans. PLoS One, 6(3), Article e17315.
References Garrido, D., Ruiz-Moyano, S., Kirmiz, N., Davis, J. C., Totten, S. M., Lemay, D. G., …
Mills, D. A. (2016). A novel gene cluster allows preferential utilization of fucosylated
milk oligosaccharides in Bifidobacterium longum subsp. longum SC596. Scientific
Albenberg, L., Esipova, T. V., Judge, C. P., Bittinger, K., Chen, J., Laughlin, A., , … Li, H.,
Reports, 6, 35045.
et al. (2014). Correlation between intraluminal oxygen gradient and radial
Garrido, D., Ruiz-Moyano, S., Lemay, D. G., Sela, D. A., German, J. B., & Mills, D. A.
partitioning of intestinal microbiota. Gastroenterology, 147(5), 1055–1063.e1058.
(2015). Comparative transcriptomics reveals key differences in the response to milk
Andreas, N. J., Hyde, M. J., Gomez-Romero, M., Lopez-Gonzalvez, M. A., Villasenor, A.,
oligosaccharides of infant gut-associated bifidobacteria. Scientific Reports, 5, 13517.
Wijeyesekera, A., … Garcia-Perez, I. (2015). Multiplatform characterization of
Garrido, D., Ruiz-Moyano, S., & Mills, D. A. (2012). Release and utilization of N-acetyl-D-
dynamic changes in breast milk during lactation. Electrophoresis, 36(18), 2269–2285.
glucosamine from human milk oligosaccharides by Bifidobacterium longum subsp.
Asakuma, S., Hatakeyama, E., Urashima, T., Yoshida, E., Katayama, T., Yamamoto, K.,
infantis. Anaerobe, 18(4), 430–435.
Kumagai, H., Ashida, H., Hirose, J., & Kitaoka, M. (2011). Physiology of
Gotoh, A., Katoh, T., Sakanaka, M., Ling, Y., Yamada, C., Asakuma, S., Urashima, T.,
consumption of human milk oligosaccharides by infant gut-associated
Tomabechi, Y., Katayama-Ikegami, A., et al. (2018). Sharing of human milk
Bifidobacteria. Journal of Biological Chemistry, 286(40), 34583–34592.
oligosaccharides degradants within bifidobacterial communities in faecal cultures
Azad, M. B., Konya, T., Maughan, H., Guttman, D. S., Field, C. J., Chari, R. S., …
supplemented with Bifidobacterium bifidum. Scientific Reports, 8(1), 13958.
Kozyrskyj, A. L. (2013). Gut microbiota of healthy Canadian infants: Profiles by
mode of delivery and infant diet at 4 months. Canadian Medical Association Journal,
185(5), 385–394.

10
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

He, Y., Liu, S., Leone, S., & Newburg, D. S. (2014). Human colostrum oligosaccharides Nishiyama, K., Nagai, A., Uribayashi, K., Yamamoto, Y., Mukai, T., & Okada, N. (2018).
modulate major immunologic pathways of immature human intestine. Mucosal Two extracellular sialidases from Bifidobacterium bifidum promote the degradation
Immunology, 7(6), 1326–1339. of sialyl-oligosaccharides and support the growth of Bifidobacterium breve.
Holscher, H. D., Bode, L., & Tappenden, K. A. (2017). Human milk oligosaccharides Anaerobe, 52, 22–28.
influence intestinal epithelial cell maturation in vitro. Journal of Pediatric Nishiyama, K., Yamamoto, Y., Sugiyama, M., Takaki, T., Urashima, T., Fukiya, S.,
Gastroenterology and Nutrition, 64(2), 296–301. Yokota, A., Okada, N., & Mukai, T. (2017). Bifidobacterium bifidum extracellular
Holscher, H. D., Davis, S. R., & Tappenden, K. A. (2014). Human milk oligosaccharides sialidase enhances adhesion to the mucosal surface and supports carbohydrate
influence maturation of human intestinal Caco-2Bbe and HT-29 cell lines. Journal of assimilation. mBio, 8(5), 917–928.
Nutrition, 144(5), 586–591. Perdijk, O., van Baarlen, P., Fernandez-Gutierrez, M. M., van den Brink, E.,
Huang, C. C., Sun, S. W., Yan, J. Y., Wang, H., Zhou, J. Y., Gao, H. Y., Xie, W. C., Li, Y., & Schuren, F. H. J., Brugman, S., … van Neerven, R. J. J. (2019). Sialyllactose and
Chai, W. G. (2020). Identification of carbohydrate peripheral epitopes important for galactooligosaccharides promote epithelial barrier functioning and distinctly
recognition by positive-ion MALDI multistage mass spectrometry. Carbohydrate modulate microbiota composition and short-chain fatty acid production in vitro.
Polymers, 229, Article 115528. Frontiers in Immunology, 10, 94.
Huda, M., Lewis, Z., Kalanetra, K., Rashid, M., Ahmad, S., Raqib, R., … Stephensen, C. Petschacher, B., & Nidetzky, B. (2016). Biotechnological production of fucosylated
(2014). Stool microbiota and vaccine responses of infants. Pediatrics, 134(2), human milk oligosaccharides: Prokaryotic fucosyltransferases and their use in
362–372. biocatalytic cascades or whole cell conversion systems. Journal of Biotechnology, 235,
Jing, W., Chen, C., Yu, Z., He, Y., Newburg, D. S., and Technology. (2016). Relative 61–83.
fermentation of oligosaccharides from human milk and plants by gut microbes. Quigley, M. A., Carson, C., Sacker, A., & Kelly, Y. (2016). Exclusive breastfeeding
European Food Research, 243(1), 1–14. duration and infant infection. European Journal of Clinical Nutrition, 70(12),
Katayama, T. (2016). Host-derived glycans serve as selected nutrients for the gut 1420–1427.
microbe: Human milk oligosaccharides and bifidobacteria. Bioscience, Biotechnology, Ruiz-Moyano, S., Totten, S. M., Garrido, D. A., Smilowitz, J. T., & Mills, D. A. (2013).
and Biochemistry, 80(4), 621–632. Variation in consumption of human milk oligosaccharides by infant gut-associated
Kelly, C. J., Zheng, L., Campbell, E. L., Saeedi, B. J., Scholz, C. C., Bayless, A. J., , … strains of Bifidobacterium breve. Applied Environmental Microbiology, 79(19),
Magnuson, A., et al. (2015). Crosstalk between microbiota-derived short-chain fatty 6040–6049.
acids and intestinal epithelial HIF augments tissue barrier function. Cell Host & Salli, K., Anglenius, H., Hiryonen, J., Hibberd, A. A., Ahonen, I., Saarinen, M. T., …
Microbe, 17(5), 662–671. Ouwehand, A. C. (2019). The effect of 2′ -fucosyllactose on simulated infant gut
Kim, J. H., An, H. J., Garrido, D., German, J. B., Lebrilla, C. B., & Mills, D. A. (2013). microbiome and metabolites; a pilot study in comparison to GOS and lactose.
Proteomic analysis of Bifidobacterium longum subsp. infantis reveals the metabolic Scientific Reports, 9, 13232.
insight on consumption of prebiotics and host glycans. PLoS One, 8(2), Article Salminen, S. (2017). Regulatory aspects of human milk oligosaccharides. In E. Isolauri,
e57535. P. M. Sherman, & W. A. Walker (Eds.), Nestle Nutrition Institute Workshop Series: Vol.
Kitaoka, M. (2012). Bifidobacterial enzymes involved in the metabolism of human milk 88. Intesinal microbiome: Functional aspects in health and disease (pp. 161–170).
oligosaccharides. Advances in Nutrition, 3(3), 422S–429S. Salyers, A. A., Vercellotti, J. R., West, S. E., & Wilkins, T. D. (1977). Fermentation of
Kitaoka, M., Tian, J., & Nishimoto, M. (2005). Novel putative galactose operon involving mucin and plant polysaccharides by strains of Bacteroides from the human colon.
lacto-N-biose phosphorylase in Bifidobacterium longum. Applied Environmental Applied Environmental Microbiology, 33(2), 319–322.
Microbiology, 71(6), 3158–3162. Sela, D., Li, Y., Lerno, L., Wu, S., Marcobal, A., German, J., … Mills, D. A. (2011). An
Koenig, J. E., Spor, A., Scalfone, N., Fricker, A. D., Stombaugh, J., Knight, R., … Ley, R. E. infant-associated bacterial commensal utilizes breast milk sialyloligosaccharides.
(2011). Succession of microbial consortia in the developing infant gut microbiome. Journal of Biological Chemistry, 286(14), 11909–11918.
Proceedings of the National Academy of Sciences, 108, 4578–4585. Sela, D. A., Garrido, D., Lerno, L., Wu, S., Tan, K., Eom, H. J., … Mills, D. A. (2012).
Kong, C., Elderman, M., Cheng, L., de Haan, B. J., Nauta, A., & de Vos, P. (2019). Bifidobacterium longum subsp. infantis ATCC 15697-fucosidases are active on
Modulation of intestinal epithelial glycocalyx development by human milk fucosylated human milk oligosaccharides. Applied and Environmental Microbiology, 78
oligosaccharides and non-digestible carbohydrates. Molecular Nutrition & Food (3), 795–803.
Research, 63(17), Article e1900303. Sela, D. A., & Mills, D. A. (2010). Nursing our microbiota: Molecular linkages between
Kuntz, S., Rudloff, S., & Kunz, C. (2008). Oligosaccharides from human milk influence bifidobacteria and milk oligosaccharides. Trends in Microbiology, 18(7), 298–307.
growth-related characteristics of intestinally transformed and non-transformed Seppo, A. E., Kukkonen, A. K., Kuitunen, M., Savilahti, E., Yonemitsu, C., Bode, L., &
intestinal cells. British Journal of Nutrition, 99(3), 462–471. Järvinen, K. M. (2019). Association of maternal probiotic supplementation with
Kunz, C., Meyer, C., Collado, M. C., Geiger, L., Garcia-Mantrana, I., Bertua-Rios, B., … human milk oligosaccharide composition. JAMA Pediatrics, 173(3), 286–288.
Rudloff, S. (2017). Influence of gestational age, secretor, and Lewis blood group Shi, Q., Yan, J., Jiang, B., Chi, X., Wang, J., Liang, X., & Ai, X. (2021). A general strategy
status on the oligosaccharide content of human milk. Journal of Pediatric for the structural determination of carbohydrates by multi-dimentional NMR
Gastroenterology and Nutrition, 64(5), 789–798. spectroscopies. Carbohydrate Polymers, 267, Article 118218.
Kunz, C., Rudloff, S., Baier, W., Klein, N., & Strobel, S. (2000). Oligosaccharides in Smilowitz, J. T., Lebrilla, C. B., Mills, D. A., German, J. B., & Freeman, S. L. (2014).
human milk: Structural, functional, and metabolic aspects. Annual Review of Breast milk oligosaccharides: Structure-function relationships in the neonate. Annual
Nutrition, 20(1), 699–722. Review of Nutrition, 34, 143–169.
Lawson, M. A. E., O’Neill, I. J., Kujawska, M., Gowrinadh Javvadi, S., Wijeyesekera, A., Soejima, M., Fujimoto, R., Agusa, T., Iwata, H., Fujihara, J., Takeshita, H., , …
Flegg, Z., … Hall, L. J. (2019). Breast milk-derived human milk oligosaccharides Nakajima, T., et al. (2012). Genetic variation of FUT2 in a Vietnamese population:
promote Bifidobacterium interactions within a single ecosystem. ISME Journal, 14, Identification of two novel Se enzyme-inactivating mutations. Transfusion, 52(6),
635–648. 1268–1275.
Lewis, Z. T., Totten, S. M., Smilowitz, J. T., Popovic, M., Parker, E., Lemay, D. G., , … Suzuki, R., Wada, J., Katayama, T., Fushinobu, S., Wakagi, T., Shoun, H., Sugimoto, H.,
German, J. B., et al. (2015). Maternal fucosyltransferase 2 status affects the gut Tanaka, A., Kumagai, H., Ashida, H., Kitaoka, M., & Yamamoto, K. (2008). Structural
bifidobacterial communities of breastfed infants. Microbiome, 3, 13. and thermodynamic analyses of solute-binding protein from Bifidobacterium longum
Li, Z., Quan, G., Jiang, X., Yang, Y., Ding, X., Zhang, D., … Zhu, G. (2018). Effects of specific for core 1 disaccharide and lacto-N-biose I. Journal of Biological Chemistry,
metabolites derived from gut microbiota and hosts on pathogens. Frontiers in Cellular 283, 13165–13173.
and Infection Microbiology, 8, 314. Thomson, P., Medina, D. A., & Garrido, D. (2018). Human milk oligosaccharides and
Marcobal, A., Barboza, M., Froehlich, J. W., Block, D. E., German, J. B., Lebrilla, C. B., & infant gut bifidobacteria: Molecular strategies for their utilization. Food Microbiology,
Mills, D. A. (2010). Consumption of human milk oligosaccharides by gut-related 75, 37–46.
microbes. Journal of Agricultural and Food Chemistry, 58(9), 5334–5340. Thongaram, T., Hoeflinger, J. L., Chow, J., & Miller, M. J. (2017). Human milk
Marcobal, A., Barboza, M., Sonnenburg, E. D., Pudlo, N. A., Martens, E. C., Desai, P. T., , oligosaccharide consumption by probiotic and human-associated bifidobacteria and
… German, J. B., et al. (2011). Bacteroides in the infant gut consume milk lactobacilli. Journal of Dairy Science, 100(10), 7825–7833.
oligosaccharides via mucus-utilization pathways. Cell Host & Microbe, 10(5), Thorburn, A. N., McKenzie, C. I., Shen, S., Stanley, D., Macia, L., Mason, L. J., , …
507–514. Robert, R., et al. (2015). Evidence that asthma is a developmental origin disease
Maslowski, K. M., & Mackay, C. R. (2011). Diet, gut microbiota and immune responses. influenced by maternal diet and bacterial metabolites. Nature Communications, 6,
Nature Immunology, 12(1), 5–9. 7320.
Matsuki, T., Yahagi, K., Mori, H., Matsumoto, H., Hara, T., Tajima, S., … Kurokawa, K. Totten, S. M., Zivkovic, A. M., Wu, S., Ngyuen, U., Freeman, S. L., Ruhaak, L. R., …
(2016). A key genetic factor for fucosyllactose utilization affects infant gut Lebrilla, C. B. (2012). Comprehensive profiles of human milk oligosaccharides yield
microbiota development. Nature Communications, 7, 11939. highly sensitive and specific markers for determining secretor status in lactating
Mee, M. T., Collins, J. J., Church, G. M., & Wang, H. H. (2014). Syntrophic exchange in mothers. Journal of Proteome Research, 11(12), 6124–6133.
synthetic microbial communities. Proceedings of the National Academy of Sciences, 111 Triantis, V., Bode, L., & van Neerven, R. J. J. (2018). Immunological effects of human
(20), 2149–2156. milk oligosaccharides. Frontiers in Pediatrics, 6, 190.
Morozov, V., Hansman, G., Hanisch, F.-G., Schroten, H., & Kunz, C. (2018). Human milk Turroni, F., Van Sinderen, D., & Ventura, M. (2011). Genomics and ecological overview
oligosaccharides as promising antivirals. Molecular Nutrition & Food Research, 62(6), of the genus Bifidobacterium. International Journal of Food Microbiology, 149(1),
Article 1700679. 37–44.
Moukarzel, S., & Bode, L. (2017). Human milk oligosaccharides and the preterm infant: A Underwood, M. A., Gaerlan, S., De Leoz, M. L. A., Dimapasoc, L., Kalanetra, K. M.,
journey in sickness and in health. Clinics in Perinatology, 44(1), 193–207. Lemay, D. G., … Lebrilla, C. B. (2015). Human milk oligosaccharides in premature
Nishimoto, M., & Kitaoka, M. (2007). Identification of N-acetylhexosamine 1-kinase in infants: Absorption, excretion, and influence on the intestinal microbiota. Pediatric
the complete lacto-N-biose I/galacto-N-biose metabolic pathway in Bifidobacterium Research, 78(6), 670–677.
longum. Applied Environmental Microbiology, 73(20), 6444–6449.

11
B. Zhang et al. Carbohydrate Polymers 276 (2022) 118738

Van Daele, E., Knol, J., & Belzer, C. (2019). Microbial transmission from mother to child: Xu, J., Mahowald, M. A., Ley, R. E., Lozupone, C. A., Hamady, M., Martens, E. C., , …
Improving infant intestinal microbiota development by identifying the obstacles. Latreille, P., et al. (2007). Evolution of symbiotic bacteria in the distal human
Critical Review in Microbiology, 45(5–6), 613–648. intestine. PLoS Biology, 5(7), 1574–1586.
Vancamelbeke, M., & Vermeire, S. (2017). The intestinal barrier: A fundamental role in Yang, I., Corwin, E. J., Brennan, P. A., Jordan, S., Murphy, J. R., & Dunlop, A. (2016).
health and disease. Expert Review of Gastroenterology & Hepatology, 11(9), 821–834. The infant microbiome. Nursing Research, 65(1), 76–88.
Victora, C., Bahl, R., Barros, A., França, G., Horton, S., Krasevec, J., , … Rollins, N., et al. Yu, J. C., Khodadadi, H., Malik, A., Davidson, B., Salles, E., Bhatia, J., … Baban, B.
(2016). Breastfeeding in the 21st century: Epidemiology, mechanisms, and lifelong (2018). Innate immunity of neonates and infants. Frontiers in Immunology, 9, 1759.
effect. Lancet, 387, 475–490. Yu, Z.-T., Chen, C., Kling, D. E., Liu, B., McCoy, J. M., Merighi, M., … Newburg, D. S.
Wada, J., Ando, T., Kiyohara, M., Ashida, H., Kitaoka, M., Yamaguchi, M., Kumagai, H., (2013). The principal fucosylated oligosaccharides of human milk exhibit prebiotic
Katayama, T., & Yamamoto, K. (2008). Bifidobacterium bifidum lacto-N-biosidase, a properties on cultured infant microbiota. Glycobiology, 23(2), 169–177.
critical enzyme for the degradation of human milk oligosaccharides with a type 1 Yu, Z. T., Chen, C., & Newburg, D. S. (2013). Utilization of major fucosylated and
structure. Applied and Environmental Microbiology, 74(13), 3996–4004. sialylated human milk oligosaccharides by isolated human gut microbes.
Walsh, C., Lane, J. A., van Sinderen, D., & Hickey, R. M. (2020). Human milk Glycobiology, 23(11), 1281–1292.
oligosaccharides: Shaping the infant gut microbiota and supporting health. Journal of Zabel, B., Yde, C. C., Roos, P., Marcussen, J., Jensen, H. M., Salli, K., … Morovic, W.
Functional Foods, 72, Article 104074. (2019). Novel genes and metabolite trends in bifidobacterium longum subsp. infantis
Ward, R. E., Niñonuevo, M., Mills, D. A., Lebrilla, C. B., & German, J. B. (2007). In vitro Bi-26 metabolism of human milk oligosaccharide 2′ -fucosyllactose. Scientific Reports,
fermentability of human milk oligosaccharides by several strains of bifidobacteria. 9(1), 7983.
Molecular Nutrition & Food Research, 51(11), 1398–1405. Zenhom, M., Hyder, A., de Vrese, M., Heller, K. J., Roeder, T., & Schrezenmeir, J. (2011).
Wu, R. Y., Li, B., Koike, Y., Maattanen, P., Miyake, H., Cadete, M., , … Prebiotic oligosaccharides reduce proinflammatory cytokines in intestinal Caco-2
Abrahamsson, T. R., et al. (2019). Human milk oligosaccharides increase mucin cells via activation of PPARgamma and peptidoglycan recognition protein 3. Journal
expression in experimental necrotizing enterocolitis. Molecular Nutrition & Food of Nutrition, 141(5), 971–977.
Research, 63(3), Article e1800658. Zhang, W., Yan, J., Wu, L., Yu, Y., Ye, R. D., Zhang, Y., & Liang, X. (2019). In vitro
Xiao, J.-Z., Takahashi, S., Nishimoto, M., Odamaki, T., Yaeshima, T., Iwatsuki, K., & immunomodulatory effects of human milk oligosaccharides onmurine macrophage
Kitaoka, M. (2010). Distribution of in vitro fermentation ability of lacto-N-biose I, a RAW264.7 cells. Carbohydrate Polymers, 207, 230–238.
major building block of human milk oligosaccharides, in bifidobacterial strains.
Applied and Environmental Microbiology, 76(1), 54–59.

12

You might also like