Вы находитесь на странице: 1из 11

FOCUS

The immune benefit of breastfeeding has


been attributed in part to the diverse
bioactive components in human milk

Ann Nutr Metab 2016;69(suppl 2):42–51


Human Milk Oligosaccharides Influence Neonatal Mucosal and
Systemic Immunity
by Sharon M. Donovan and Sarah S. Comstock

Key insights
Human milk confers multiple layers of protection to the new- Gut epithelium
Gut lumen Bloodstream
born by providing bioactive components that protect the infant Goblet cell function

from pathogenic infection, facilitate intestinal and immune Crypt cell proliferation

development, and support healthy gut microbes. An important Maturation of the Cytokine
intestinal epithelium secretion
HMOs Systemic
bioactive component of human milk are the human milk oligo- Increase intestinal immunity
barrier function Bind
saccharides (HMOs). HMOs are a complex mixture of indigest- monocytes,
ible carbohydrates with a high degree of structural diversity and Affect gene lymphocytes,
expression
Promote neutrophils
represent one of the largest groups of bioactive components in a healthy Act as prebiotics
human milk. microbial
environment

Current knowledge
Local
HMOs are a family of soluble glycans that are sialylated or fu- immunity
cosylated and provide carbon sources for gut bacterial species
that colonize breastfed infants. Through their actions on the
Through diverse actions on intestinal function and the gut microbiome,
gut, HMOs directly and indirectly affect the infant’s mucosal
human milk oligosaccharides (HMOs) can modulate an infant’s local and
and systemic immunity. A large number of studies have shown systemic immunity.
that HMOs can influence the proliferation and maturation of in-
testinal cells (such as crypt cells and goblet cells). Furthermore,
HMOs can also modulate gene expression in the intestinal epi- evidence suggests that supplementing infant formulae with
thelium. Altogether, these affect the function of the intestinal HMOs is safe and beneficial for human infants. There are also
barrier, which in turn regulates local and systemic immunity. potential applications of HMOs as prophylactic and therapeu-
tic treatments for those who are immune compromised and at
Practical implications high risk of infection.
Human milk contains a higher concentration and a greater
structural diversity and degree of fucosylation compared to Recommended reading
the milk oligosaccharides in other species, including cow’s milk Kulinich A, Liu L: Human milk oligosaccharides: the role in
from which many infant formulae are derived. Commercially the fine-tuning of innate immune responses. Carbohydr Res
produced HMOs are becoming increasingly available, and 2016;432:62–70.

© 2017 S. Karger AG, Basel

E-Mail karger@karger.com
www.karger.com/anm
Human Milk: Lessons from Recent Research

Ann Nutr Metab 2016;69(suppl 2):42–51 Published online: January 20, 2017
DOI: 10.1159/000452818

Human Milk Oligosaccharides Influence


Neonatal Mucosal and Systemic Immunity
Sharon M. Donovan a Sarah S. Comstock b
a
Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, and b Department of Food Science
and Human Nutrition, Michigan State University, East Lansing, MI, USA

Key Messages ment and function. For the past decade, intense research has
• Human milk oligosaccharides (HMO) are a been directed at defining the complexity of oligosaccha-
rides in the milk of many species and is beginning to delin-
predominant component of human milk and are
comprised of diverse structures that are neutral or eate their diverse functions. These studies have shown that
acidic and some forms are sialylated or fucosylated, human milk contains a higher concentration as well as a
which contributes to their biological functions. greater structural diversity and degree of fucosylation than
• HMO protect the infant from pathogenic infections, the milk oligosaccharides in other species, particularly bo-
vine milk from which many infant formulae are produced.
facilitate the establishment of the gut microbiota,
promote intestinal development, and stimulate The commercial availability of large quantities of certain
immune maturation. HMO has furthered our understanding of the functions of
• Some types of HMO are now commercially available specific HMO, which include protecting the infant from
pathogenic infections, facilitating the establishment of the
and are being added to infant formula alone or in
combination with other prebiotics. gut microbiota, promoting intestinal development, and
stimulating immune maturation. Many of these actions are
exerted through carbohydrate-carbohydrate interactions
with pathogens or host cells. Two HMOs, 2′-fucosyllactose
(2′FL) and lacto-N-neotetraose (LNnT), have recently been
Keywords added to infant formula. Although this is a first step in nar-
Human milk oligosaccharides · Immunity · Infant rowing the compositional gap between human milk and in-
fant formula, it is unclear whether 1 or 2 HMO will recapitu-
late the complexity of actions exerted by the complex mix-
Abstract ture of HMO ingested by breastfed infants. Thus, as more
The immune system of the infant is functionally immature HMO become commercially available, either isolated from
and naïve. Human milk contains bioactive proteins, lipids, bovine milk or chemically or microbially synthesized, it is an-
and carbohydrates that protect the newborn and stimulate ticipated that more oligosaccharides will be added to infant
innate and adaptive immune development. This review will formula either alone or in combination with other prebiotics.
focus on the role human milk oligosaccharides (HMO) play © 2017 S. Karger AG, Basel
in neonatal gastrointestinal and systemic immune develop-

© 2017 S. Karger AG, Basel Sharon M. Donovan, PhD, RD


Department of Food Science and Human Nutrition, University of Illinois
339 Bevier Hall, 905 S. Goodwin Avenue
E-Mail karger@karger.com
Urbana, IL 61801 (USA)
www.karger.com/anm
E-Mail sdonovan @ illinois.edu
Background
The human infant enters the world with a functionally
naïve immune system affecting both adaptive and innate
immune responses [1], which leaves the newborn at high
Immune
risk for common infections. Postnatal immune matura- maturation and
tion is stimulated by antigenic exposures and host-mi- function
crobe interactions [1, 2]. How and what the infant is fed
influences the development and competence of the im-
mune system [3–5]. Human milk protects the infant dur-
Intestinal
ing this vulnerable period by providing bioactive compo- development
nents that protect the infant from pathogenic infection,
support intestinal development, promote barrier func-
Bioactive components
tion, stimulate immune development, facilitate immune in human milk
tolerance, and feed gut microbes [2–5]. Thus, human
milk supplies multiple layers of protection for the infant Establishment
(Fig. 1). of the
microbiome
Breastfeeding, particularly exclusive breastfeeding for
6 months or more, relative to formula-feeding, decreases
the incidence and/or severity of infectious diseases [6].
Many diseases with infectious and immune components
in their etiology, including diarrhea, respiratory and uri-
Fig. 1. Human milk may orchestrate gastrointestinal, immune, and
nary tract infections, otitis media, bacteremia, and necro- microbiota development. The intestinal ecosystem represents a
tizing enterocolitis occur less often in breast- than formu- complex, interactive environment in which human milk influenc-
la-fed infants [6, 7]. Breastfeeding has also been impli- es intestinal development, establishment of gut microbiota, and
cated in reducing the incidence of other diseases involving maturation of the gut mucosal and systemic immune system. In
the immune system and immune tolerance, such as in- turn, signals from the microbiota stimulate maturation and speci-
ficity of the mucosal and systemic immune systems. In addition,
flammatory bowel disease, celiac disease, asthma, allergy, the immune system and microbiota promote intestinal develop-
type 1 diabetes, as well as acute lymphoblastic and acute ment. Human milk contains bioactive nutrients and other compo-
myeloblastic leukemias [6, 8]. These benefits may be me- nents that are modulators of these processes, of which the oligosac-
diated in part through effects of breastfeeding on the in- charides are a key component.
testinal microbiota [8, 9], which in turn stimulates matu-
ration and specificity of the neonatal mucosal and sys-
temic immune systems [2].
The immune benefit of breastfeeding has been attrib- exceptions, all HMO carry lactose (Galβ1–4Glc) at the
uted in part to the diverse bioactive components in hu- reducing end, which can be elongated in β1–3 or β1–6
man milk [2–5]. A strong case can be made for a key role linkage by 2 different disaccharides, either Galβ1–
of human milk oligosaccharides (HMO) in neonatal im- 3GlcNAc (type 1 chain) or Galβ1–4GlcNAc (type 2 chain)
mune defense and maturation. As will be described be- [11].
low, HMO are present in high concentrations in human The HMO content has been reported in the range of
milk, exist in an incredible structural diversity [10–13], 1–10 g/L in mature milk and 15–23 g/L in colostrum [10–
and confer host protection and mediate immune re- 13]. In term breast milk, ∼35–50% of HMO are fucosyl-
sponses through a number of mechanisms [14, 15]. ated, 12–14% are sialylated, and 42–55% are nonfucosyl-
ated neutral HMO [10–13] (Table  1). However, HMO
composition is influenced by maternal genetics, includ-
HMO Content and Composition ing secretor and Lewis Blood Group status [10, 11]. HMO
HMO are complex soluble glycans that are predomi- fucosylation is mediated by the 2 fucosyltransferases
nantly present in free form in milk. These glycans are FUT2 (secretor gene) and FUT3 (Lewis gene). Nonsecre-
synthesized from 5 basic monosaccharides: galactose, tor mothers, who lack a functional FUT2 enzyme and
glucose, N-acetylglucosamine, fucose, and the sialic acid represent about 30% of women worldwide, produce milk
derivative N-acetylneuraminic acid [10, 11]. With few lacking in α1-2-fucosylated oligosaccharides like 2′-fu-

HMO Influence Neonatal Mucosal and Ann Nutr Metab 2016;69(suppl 2):42–51 43
Systemic Immunity DOI: 10.1159/000452818
Table 1. Concentrations of major HMO in
human milk [10 – 13] Categories of HMO (% total) Oligosaccharide Mean concentration
(range), g/L

Fucosylated (35 – 50%) 2′FL 2.7 (1.88 – 4.9)


3′FL 0.5 (0.25 – 0.86)
LNFP I 0.122 (0.106 – 0.145)
LNFP II + III 0.156 (0.120 – 0.161)
Sialylated (12 – 14%) 3′SL 0.2 (0.1 – 0.3)
6′SL 0.5 (0.2 – 1.22)
Nonfucosylated neutral (42 – 55%) LNnT 0.3 (0.17 – 0.45)

2′FL, 2′-fucosyllactose; 3′SL, 3′-sialyllactose; 6′SL, 6′-sialyllactose; HMO, human milk


oligosaccharides; LNFP, lacto-N-fucopentaose; LNnT, lacto-N-neotetraose.

cosyllactose (2′FL) and lacto-N-fucopentaose (LNFP) I ceae were less abundant in infants fed formula with 2′FL
[10, 11]. The absence of these compounds may have func- and LNnT compared to infants fed nonsupplemented
tional consequences. For example, infants consuming formula, and these levels were closer to those observed in
milk produced by women who are nonsecretors exhibit breastfed infants [20]. Furthermore, the concentrations
delayed colonization of bifidobacteria, higher abundance of several important metabolites in stool (propionate, bu-
of Streptococcus taxa, and have functional differences in tyrate, and lactate) in infants fed the HMO-supplemented
the metabolic activity of their microbiota [16]. Infants fed formula were more similar to those of breastfed infants
milk from nonsecretor mothers are at higher risk for diar- [20].
rheal diseases [17]. Previously, we have shown that HMO fermentation by
neonatal pig microbiota produced short-chain fatty acids
and promoted the growth of beneficial bacteria in vitro
HMO and the Microbiome [21] and in vivo [22]. Gut bacteria and the immune re-
The development of the infant gut microbiota is a se- sponse, particularly the gastrointestinal immune re-
quential process that begins in utero and continues dur- sponse, are tightly interrelated [23]. Thus, in this animal
ing the first 2–3 years of life. Microbial composition and model, HMO-induced changes in the gut bacterial popu-
diversity is shaped by host genetics and multiple environ- lations of the pigs could alter the course of an intestinal
mental factors, of which diet is a principal contributor [8, infection [24] which in turn would alter the immune re-
9]. Studies conducted over the past decade have shown sponse [22]. Alternatively, the change in the gut bacteria
that specific Bacteroides and Bifidobacterium species that could directly affect the immune system of these animals
commonly colonize breastfed infants efficiently utilize [2]. Additional ways whereby HMO may mediate neona-
HMO as carbon sources. This is particularly true of B. tal immunity are summarized in the following section.
longum ssp. infantis (B. infantis), which is a predominant
gut microbe in most breastfed infants [18]. The discovery
of a genomic island in B. infantis that encodes specific HMO as Immune Modulators
enzymes for the metabolism of HMO supports an adapta- Summarized in Figure 2 are the results of an accumu-
tion of this species to the intestinal milieu of the breastfed lating body of evidence showing that HMO indirectly and
infant [18, 19]. Indeed, a recent study in human infants directly influence infant mucosal and systemic immune
fed formula supplemented with 2′FL (1 g/L) and LNnT function. In general, intestinal health and barrier func-
(0.5 g/L) demonstrated that the global microbiota com- tion are considered a first line of defense in innate immu-
position of infants fed formulae with 2′FL and LNnT was nity. Cell proliferation takes place in the crypts, and cells
significantly different to that of infants fed nonsupple- differentiate as they migrate up the villus-crypt axis, with
mented formula (p < 0.001) at the genus level and closer the exception of Paneth cells, which migrate down to the
to that of breastfed infants at 3 months of age [20]. In ad- base of the crypt. HMO reduce intestinal crypt cell prolif-
dition, Bifidobacterium was more abundant (p < 0.01), eration [25, 26], increase intestinal cell maturation [26],
whereas Escherichia and unclassified Peptostreptococca- and increase barrier function [26] (indicated by 1–3 in

44 Ann Nutr Metab 2016;69(suppl 2):42–51 Donovan/Comstock


DOI: 10.1159/000452818
Mucin Epithelial Lamina
Intestinal lumen Circulation
layer layer propria

Commensal
microbiota

11
2’FL 8

6’SL
6 6’SL
10

Peyer’s patch
Pathogen

Bifidobacterium

7 Microfold 2’FL
LNT cell

12

2
5
Tight junction
Crypt 1
LNT

Goblet 4 9
cell

Epithelial
cell and villi

Dendritic Naïve
Macrophage Neutrophil T cell T cell Platelet Cytokines
cell

Fig. 2. Potential mechanisms whereby human milk oligosaccha- prebiotics to promote the growth of healthy bacteria, including
rides (HMO) influence host immune function. HMO affects in- Bifidobacteria and Bacteroides species (7), and HMO inhibit infec-
nate immunity through the epithelial barrier: HMO reduce intes- tions by bacteria and viruses by either binding to the pathogen in
tinal crypt cell proliferation (1), increase intestinal cell maturation the lumen or by inhibiting binding to cell-surface glycan receptors
(2), increase barrier function (3), and may influence goblet cell (8). HMO affect immune cell populations and cytokine secretion
function (4), as has been shown for galacto-oligosaccharides. In (9). HMO are also absorbed into the blood (10), where they affect
addition, HMO affect epithelial immune gene expression both di- binding of monocytes, lymphocytes, and neutrophils to endothe-
rectly (5) and indirectly through the microbiota (6). HMO serve as lial cells (11) and formation of platelet-neutrophil complexes (12).

HMO Influence Neonatal Mucosal and Ann Nutr Metab 2016;69(suppl 2):42–51 45
Systemic Immunity DOI: 10.1159/000452818
Fig. 2). A layer formed by mucus glycoproteins or mucins Lectins are carbohydrate-binding proteins on the sur-
produced by goblet cells acts as a lubricant and a protec- faces of mammalian cells that translate recognition of
tive physical barrier between the intestinal epithelium specific motifs and the spatial presentation of those mo-
and the luminal contents (indicated by 4 in Fig. 2). HMO tifs into action. Lectins are grouped according to their
may influence goblet cell function, as has been shown for carbohydrate recognition domains (CRD) [42, 43]. There
galacto-oligosaccharides (GOS) [27]. HMO affects epi- are at least a dozen CRD identified in mammals, but 3
thelial immune gene expression both directly [28–30] and classes of lectins related to the influence of HMO on im-
indirectly through the microbiota [31] (indicated by 5 mune responses are C-type lectins, siglecs (sialic acid-
and 6 in Fig. 2, respectively). As noted above, HMO serve binding Ig-like lectins), and galectins.
as prebiotics to promote the growth of healthy bacteria, C-type lectins require calcium to function and include
including Bifidobacteria and Bacteroides genera [32] (in- selectins, mannose-binding lectin, and dendritic cell-spe-
dicated by 7 in Fig.  2), and HMO inhibit infections by cific intercellular adhesion molecule 3-grabbing non-in-
bacteria and viruses by either binding to the pathogen in tegrin (DC-SIGN). C-type lectin receptors on the surface
the lumen or by inhibiting binding to cell-surface glycan of dendritic cells (DC) determine whether the cell will
receptors [14–15, 22] (indicated by 8 in Fig. 2). Addition- induce tolerance rather than lymphocyte activation [44].
ally, dietary oligosaccharides decorate the intestinal lin- DC-SIGN is of particular interest with regard to mecha-
ing contributing to the intestinal glycan repertoire [33]. nisms by which HMO can influence immunity because it
HMO also contribute to epithelial barrier function by has a CRD specific for fucose units. Furthermore, DC-
supporting the growth of B. infantis in the infant gut [10, SIGN is expressed by cells in the gastrointestinal tracts of
18]. B. infantis produces peptides that have been shown infants [45]. These intestinal cells are likely antigen-pre-
to normalize intestinal permeability through enhanced senting cells as DC-SIGN is expressed by antigen-pre-
tight junction protein expression in a mouse model of senting cells, specifically DC [43]. Although interactions
colitis [34]. It is likely that HMO support other bacterial between fucosylated ligands and DC-SIGN contribute to
species that are important for the maintenance of gut in- immune tolerance, the cellular response ultimately de-
tegrity. These changes in intestinal barrier function pends upon the other ligand-receptor reactions occurring
would, in turn, alter both the local and systemic immune simultaneously [43].
system [35]. HMO affect immune cell populations and Siglecs are sialic acid-binding lectins most commonly
cytokine secretion [22, 36] (indicated by 9 in Fig. 2). Some found on subsets of immune cells [46]. There are at least
HMO are also absorbed into the blood stream [37–39] 16 siglecs expressed by different leukocyte populations,
(indicated by 10 in Fig. 2), where they exert systemic ef- which include sialoadhesin (siglec-1), CD22 (siglec-2),
fects by binding of monocytes, lymphocytes, and neutro- myelin-associated glycoprotein (MAG, siglec-4), si-
phils to endothelial cells [40] (indicated by 11 in Fig. 2) glec-15, and CD33-related siglecs. Siglec specificity de-
and formation of platelet-neutrophil complexes [41] (in- rives from differences in secondary binding sites [43]. Si-
dicated by 12 in Fig. 2). Readers are referred to a recent glecs are endocytic cell surface receptors that carry cargo
review by Kulinich and Liu [15] for additional discussion between the cell surface and intracellular vesicles; these
of this topic. receptors are mainly expressed on cells involved in anti-
gen processing and presentation [43]. Furthermore, sialic
acid-containing molecules can gain entry to macrophages
Carbohydrate Binding as a Potential by binding to siglecs on the cell surface [46]. On mam-
Mechanism of HMO in the Immune System malian cells, some sialic acid-containing glycans function
Carbohydrates and carbohydrate-binding proteins as self-associated molecular patterns and prevent im-
play an important role in immune responses. Cells have mune responses to nonpathogenic stimuli. Ligation of
unique glycan signatures made from combinations of particular siglecs stimulates the production of the immu-
specific glycan motifs that are engaged when a cell con- noregulatory cytokine interleukin (IL)-10 [47].
tacts another cell or other components of its environ- Galectins are important for cell turnover and immune
ment [42, 43]. However, many of the glycan motifs regulation. The CRD of galectins is specific for β-galac-
found on mammalian cells are also found on microbes tosides. When cells are desialylated, the density of ex-
and in food, including human milk. These similarities posed galactose moieties on the cell surface increases. For
provide opportunities for host-microbe-HMO interac- example, naïve T cells express CD45 with an α-2,6-linked
tions. sialic acid. The amount of α-2,6-linked sialic acid is re-

46 Ann Nutr Metab 2016;69(suppl 2):42–51 Donovan/Comstock


DOI: 10.1159/000452818
Table 2. Immune-related outcomes of HMO feeding studies

Species Study design Major findings Ref.

Human infants Healthy singleton infants enrolled by 5 days of life and fed Breastfed infants and infants fed either formula with 53
formulae to 4 months of age 2′FL were similar and had lower plasma inflammatory
– Breastfed cytokines than infants fed the control formula
– Formula + 2.4 g/L GOS In ex vivo RSV-stimulated PBMC cultures, cells from
– Formula + 2.2 g/L GOS + 0.2 g/L 2′FL breastfed infants were not different from those of
– Formula + 1.4 g/L GOS + 1.0 g/L 2´FL either of the groups fed formula with 2′FL, but
PBMC isolated at 6 weeks of age secreted less TNF-α and IFN-γ and tended to have
lower IL-1Ra, IL-6 and IL-1β than cells from infants fed
the control formula
Human infants Healthy singleton infants enrolled by 14 days of life and fed Infants fed HMO-supplemented formula had 54
experimental formulae to 6 months of age, and standard significantly fewer parental reports of:
follow-up formula to 12 months – Bronchitis through 4, 6, and 12 months
– Formula – Lower respiratory tract infection through 12 months
– Formula + 1.0 g/L 2′FL + 0.5 g/L LNnT – Antipyretics use through 4 months
– Antibiotics use through 6 and 12 months

Colostrum-deprived 15-day feeding study HMO resulted in shorter duration of diarrhea and 22
piglets – Formula higher ileal IFN-γ and IL-10 mRNA expression than
– Formula + 4 g/L HMO (40% 2′FL; 35% LNnT; 10% 6′SL; formula, but similar concentrations of RV-specific IgG
5% 3′SL; 10% free SA) and IgM as formula
Infected with OSU strain RV on day 10 and analyzed on day 15
Adult female E. coli infection model 2′FL prevented body weight loss and reduced AIEC 29
C57BL/6 mice – 0.25% DSS orally for days 0 – 3 colonization, colonic inflammation, crypt cell CD14
– 2′FL (100 mg or vehicle by oral gavage days 0 – 4) expression, and IL-6, IL-17, and TNF-α production in
– 20 mg streptomycin by oral gavage on day 4 response to AIEC infection
Infected with AIEC on day 5 and analyzed on day 9
Adult male Food allergy treatment model 2′FL and 6′SL attenuated diarrhea and hypothermia 51
Balb/c mice – IP sensitized to OVA induced by OVA challenge, reduced intestinal mast
– 2 weeks later (day 27), oral gavage daily (1 mg in 200 μl PBS) cell numbers and passive cutaneous anaphylaxis,
2′FL and increased Peyer’s patch T regulatory cells and
6′SL CD11c+CD103+ DC
Lactose 6′SL increased OVA-specific IgG2a and MLN
– Day 28 oral challenge with OVA (50 mg) every 3 days until T regulatory cells
day 43 Splenocytes from 6′SL-treated mice produced more
IFN-γ and IL-10 but less TNF
Splenocytes from 2′FL-treated mice produced less
IFN-γ

2′FL, 2′-fucosyllactose; 3′SL, 3′-sialyllactose; 6′SL, 6′-sialyllactose; AIEC, adherent-invasive E. coli, DC, dendritic cells; DSS, dextran sodium sulfate;
GOS, galacto-oligosaccharides; HMO, human milk oligosaccharides; IFN, interferon; IP, intraperitoneal infection; Ig, immunoglobulin; IL, interleukin;
LNnT, lacto-N-neotetraose; MLN, mesenteric lymph nodes; OSU, Ohio State University; OVA, ovalbumin; PBMC, peripheral blood mononuclear cells;
PBS, phosphate-buffered saline; RSV, respiratory syncytial virus; RV, rotavirus; SA, sialic acid; TNF, tumor necrosis factor.

duced following T-cell activation. The decrease in α-2,6- charides (LPS) to model a bacterial infection [29]. Co-
linked sialic acid renders the activated T cells susceptible incubation of Bifidobacterium with cells of the Caco-2
to galectin-1-mediated apoptosis [48]. Thus, binding of intestinal cell line and HMO resulted in downregulation
sialyated HMO to cells may prevent apoptosis. of intestinal cell genes related to chemokine activity com-
pared to co-incubation with glucose or lactose [29]. Con-
versely, in the absence of a bacterial co-stimulant, HMO
HMO as Modulators of Mucosal Immunity increased expression of several chemokines by the HT-
Intestinal cell lines have been used to determine ef- 29 cell line [28]. Additional work in T84 and HCT8 in-
fects of HMO on immune-related gene expression and testinal cell lines showed that complex mixtures of HMO
protein production. These cells have been co-incubated as well as 2′FL reduced signatures of intestinal inflamma-
with oligosaccharides [28], bacteria [48], or lipopolysac- tion [29].

HMO Influence Neonatal Mucosal and Ann Nutr Metab 2016;69(suppl 2):42–51 47
Systemic Immunity DOI: 10.1159/000452818
HMO have been demonstrated to affect the course of a [55] have been fed 2′FL, but only growth and toxicologi-
gastrointestinal viral infection. In an acute rotavirus (RV) cal outcomes were reported. A recently published paper
infection model where a 21-day-old piglet’s ileum was iso- described immune outcomes in human infants fed for-
lated in situ, intestinal loops co-treated with HMO and RV mula containing 2.4 g/L GOS, 2.2 g/L GOS + 0.2 g/L 2′FL,
had reduced non-structural protein-4 (NSP-4) mRNA ex- or 1.4 g/L GOS + 1.0 g/L 2′FL compared to a breastfed
pression indicating that HMO can reduce RV replication reference control [53]. Infants were fed the formula from
[49]. Intestinal cytokine and chemokine expression, how- 5 days to 4 months of age, and blood samples were ob-
ever, was not affected. Both neutral and acidic HMO de- tained at 6 weeks of age for cytokine analysis, immune cell
creased NSP4 intestinal mRNA expression in the in situ phenotyping, and ex vivo stimulation of isolated PBMC.
model, whereas only acidic HMO effectively inhibited RV Breastfed infants and infants fed either formula with 2′FL
infectivity in an in vitro model [49]. were similar and had lower plasma inflammatory cyto-
kines than infants fed the control formula. In addition,
cytokine secretion by PBMC from breastfed infants and
HMO as Modulators of Systemic Immunity and infants fed either 2′FL-containing formula that were
Protection from Infection stimulated ex vivo with respiratory syncytial virus was
HMO are detected in the plasma of infants fed human similar and secreted less tumor necrosis factor-α and
milk at concentrations of 1–133 mg/L [37, 39], suggesting interferon-γ and tended to have lower IL-1Ra, IL-6, and
the potential for dietary HMO to directly affect immune IL-1β than cells from infants fed the control formula [53].
cells circulating in the blood. As discussed above, many Another recent study in human infants evaluated the
immune receptors recognize the oligosaccharide struc- effect of formula supplemented with both 2′FL (1.0 g/L)
tures of their glycoprotein ligands [14, 15]. Because a sub- and LNnT (0.5 g/L) compared to an unsupplemented for-
set of HMO is structurally similar to selectin ligands [14], mula. Infants were fed the formulae from 14 days to 6
it is likely that HMO can bind directly to immune cells months of age, after which they were switched to a stan-
and trigger signaling that results in changes to immune dard follow-on formula and followed until 12 months of
cell populations and functions. For instance, the P- and age. Infants fed the HMO-supplemented formula had sig-
E-selectins recognize sialyl-Lewis x (sLeX), a glycan moi- nificantly fewer parental reports (p = 0.004–0.047) of:
ety of several HMO [11]. Additionally, fucosylation and bronchitis through 4 months (2.3 vs. 12.6%), 6 months
sialylation, 2 enzymatic modifications common to HMO, (6.8 vs. 21.8%), and 12 months (10.2 vs. 27.6%); lower re-
enable binding to selectins [50]. HMO-induced disrup- spiratory tract infection (AE cluster) through 12 months
tion of immune protein-carbohydrate interactions re- (19.3 vs. 34.5%); antipyretics use through 4 months (15.9
duced neutrophil rolling [40] and activation [41]. HMO vs. 29.9%); and antibiotics use through 6 months (34.1 vs.
directly affect immune cell proliferation and cytokine 49.4%) and 12 months (42.0 vs. 60.9%) than those fed the
production in ex vivo experiments with peripheral blood standard formula [54].
mononuclear cells (PBMC) from neonatal pigs [36]. Several studies in animal models support the reduced
Stimulation with isolated HMO stimulated production of incidence of infection in human infants fed formula with
the regulatory cytokine IL-10 [36]. Others observed that HMO. In mice infected with Escherichia coli, once daily
the acidic HMO induce IL-10 production; additionally, oral gavage with 100 mg, 2′FL prevented body weight loss
they found that acidic HMO induce IFN-γ from ex vivo and reduced colonization with adherent-invasive E. coli,
stimulated human cord blood mononuclear cells [45]. colonic inflammation, crypt cell CD14 expression, as well
Isolated HMO enhanced proliferation of PBMC stimu- as IL-6, IL-17, and tumor necrosis factor-α production in
lated with a T-cell mitogen, phytohemagglutinin (PHA), response to adherent-invasive E. coli infection compared
and sialylated HMO enhanced proliferation of PBMC to mice treated with vehicle [29]. Mice fed 2′FL and sub-
stimulated with the B-cell mitogen LPS [36]. In contrast, jected to ileocecal resection gained more weight and had
2′FL inhibited proliferation of unstimulated PBMC cul- greater crypt depth and villus height at the site of transec-
tured for 3 days. Thus, the response to HMO may depend tion than nonsupplemented mice [30]. The 2′FL-fed mice
on the state of the infant. In the unstimulated state, HMO also had upregulated mucosal immune response genes in
dampen proliferation, whereas HMO enhance prolifera- the distal small bowel [30]. The studies where pigs and
tion in response to a mitogenic stimuli. human infants were fed the HMO have focused on 2′FL,
To date, very few studies have fed HMO and analyzed which is readily available in large quantities at reasonable
immune outcomes [22, 29, 30, 51–54] (Table 2). Piglets cost, and fucosylated oligosaccharides have been shown

48 Ann Nutr Metab 2016;69(suppl 2):42–51 Donovan/Comstock


DOI: 10.1159/000452818
to feed specific beneficial classes of bacteria during intes- and in human milk is the same. 3′SL is recognized by DC
tinal inflammatory events [56]. Given what is known and generates an immune response through the TLR4
about the effects of other HMO, these compounds also signaling pathway [61]. These results suggest that the
should be used in feeding studies when available in suf- presence of 3′SL increases the inflammatory response
ficient quantities. through direct effects on DC. When TLR4 was absent,
Only 1 in vivo study used a complex mixture of HMO 3′SL was less effective at inducing DC activation. How-
and assessed immune outcomes. In that report, neonatal ever, those DC also demonstrated a minimal increase in
pigs fed a diet containing 4 g/L HMO, consisting of 40% CD40 expression suggesting that at least one other 3′SL-
2′FL, 10% 6′-sialyllactose (6′SL), 35% lacto-N-neote- sensing mechanism, albeit much less efficient than the
traose (LNnT), 5%, 3′-sialyllactose (3′SL), and 10% free TLR4 pathway, exists on DC. TLR4 is the receptor for E.
sialic acid, had a reduced duration of diarrhea, in re- coli LPS. Another link between 3′SL and TLR4 is ex-
sponse to RV infection to 48.8 ± 9.8 h versus 80.6 ± 4.5 h plained in a newer paper, where it is demonstrated that
in pigs fed nonsupplemented formula [22]. Ileal tissue 3′SL stimulates the proliferation of the intestinal E. coli
from the pigs fed HMO contained greater IFN-γ (pro- population and that this overgrowth of E. coli is respon-
duced by Th1 cells) and IL-10 (an anti-inflammatory cy- sible for exacerbation of dextran sulfate sodium colitis
tokine) mRNA than that from pigs fed formula [22]. through release of proinflammatory cytokines from in-
In a mouse model of food allergy, 2′FL and 6′SL admin- testinal DC [62]. These examples demonstrate the com-
istered via oral gavage reduced symptoms in mice sensi- plexity of the relationships between oligosaccharides, the
tized to ovalbumin, an egg protein [51]. Specifically, oval- gut bacteria and the immune system.
bumin-stimulated splenocytes from mice treated with 6′SL
produced more IL-10 and less IFN-γ than those from un-
treated mice. Furthermore, 2′FL- or 6′SL-treated mice had Conclusion
more regulatory immune cells in their intestinal immune The rich diversity of HMO has the potential to modu-
tissues than untreated mice. Interestingly, neither 2′FL nor late both innate and adaptive neonatal immunity. Find-
6′SL affected intestinal T regulatory cells when adminis- ings from in vitro experiments and animal models show
tered to nonsensitized mice [51]. This exemplifies the ne- that HMO directly interact with gastrointestinal epithe-
cessity of identifying an appropriate challenge model to as- lial cells as well as with mucosal and systemic immune
sess the effects of dietary compounds on the immune sys- cells to modulate immune function. HMO also benefi-
tem. In mice, the milk oligosaccharides LNFP III and LNnT cially shape the microbiome of the breastfed infant. The
are Th2-biasing and suppress Th1 responses [57]. Recently, increased availability of HMO from commercial sources
it has been reported that human infants who were fed hu- as well as accumulating evidence demonstrating that for-
man milk with low LNFP III concentrations (<60 μM) were mula supplemented with HMO is safe and may confer
6.7-times (95% CI 2.0–22) more likely to become affected benefits for human infants have led to the recent addition
with cow’s milk allergy when compared to infants receiving of 2′FL alone or in combination with LNnT to infant for-
milk with high LNFP III concentrations [58]. mulae. In addition, due to their beneficial effects on im-
Another approach using knockout mice showed that mune function and host defense, HMO may also be ben-
SL-containing compounds can directly affect gastrointes- eficial for other segments of the population who are im-
tinal mucosal immunity [52, 59]. In one study, the pres- mune compromised or at high risk for infection. There
ence of 3′SL in the milk increased the number of immune are limited studies in which animals or humans have been
cells infiltrating the gut in IL-10 null mice [52]. Further- fed HMO. Additionally, few studies have assessed the ef-
more, supplementation with 3′SL increased colitis sever- fects of feeding complex mixtures of HMO on the im-
ity in newborn IL-10 and St3gal4 (the enzyme that syn- mune response. Thus, future research is needed to delin-
thesizes 3′SL) null mice, and cross-fostering wildtype eate mechanisms and to fully realize the potential for
mice to deficient dams reduced colitis severity. One ca- HMO to benefit infant immune function.
veat of this work is that it was conducted in the absence
of endogenous IL-10 production, whereas other in vivo
studies have demonstrated that some HMO increase in- Disclosure Statement
testinal IL-10 [22, 51]. 3′SL is a product of several patho- Sharon Donovan has received grant funding from Nestlé Nu-
trition R&D. Sarah Comstock has no disclosures.
genic bacteria [60] and the conformation (α2,3-link be-
tween sialic acid and galactose) on pathogenic bacteria

HMO Influence Neonatal Mucosal and Ann Nutr Metab 2016;69(suppl 2):42–51 49
Systemic Immunity DOI: 10.1159/000452818
References
1 Levy O, Wynn JL: A prime time for trained 16 Lewis ZT, Totten SM, Smilowitz JT, Popovic 27 Bhatia S, Prabhu PN, Benefiel AC, Miller MJ,
immunity: innate immune memory in new- M, Parker E, Lemay DG, Van Tassell ML, Chow J, Davis SR, Gaskins HR: Galacto-oli-
borns and infants. Neonatology 2014; 105: Miller MJ, Jin YS, German JB, Lebrilla CB, gosaccharides may directly enhance intesti-
136–141. Mills DA: Maternal fucosyltransferase 2 sta- nal barrier function through the modulation
2 Walker WA, Iyengar RS: Breast milk, micro- tus affects the gut bifidobacterial communi- of goblet cells. Mol Nutr Food Res 2015; 59:
biota, and intestinal immune homeostasis. ties of breastfed infants. Microbiome 2015; 566–573.
Pediatr Res 2015;77:220–228. 3:13. 28 Lane JA, O’Callaghan J, Carrington SD,
3 Andreas NJ, Kampmann B, Mehring Le- 17 Newburg DS, Ruiz-Palacios GM, Altaye M, Hickey RM: Transcriptional response of HT-
Doare K: Human breast milk: a review on its Chaturvedi P, Meinzen-Derr J, Guerrero 29 intestinal epithelial cells to human and
composition and bioactivity. Early Hum Dev MDL, et al: Innate protection conferred by bovine milk oligosaccharides. Br J Nutr
2015;91:629–635. fucosylated oligosaccharides of human milk 2013;110:2127–2137.
4 Turfkruyer M, Verhasselt V: Breast milk and against diarrhea in breastfed infants. Glyco- 29 He Y, Liu S, Kling DE, Leone S, Lawlor NT,
its impact on maturation of the neonatal im- biology 2004;14:253–263. Huang Y, Feinberg SB, Hill DR, Newburg
mune system. Curr Opin Infect Dis 2015;28: 18 Underwood MA, German JB, Lebrilla CB, DS: The human milk oligosaccharide 2′-fu-
199–206. Mills DA: Bifidobacterium longum subspe- cosyllactose modulates CD14 expression in
5 Donovan SM: Role of human milk compo- cies infantis: champion colonizer of the in- human enterocytes, thereby attenuating
nents in gastrointestinal development: cur- fant gut. Pediatr Res 2015;77:229–235. LPS-induced inflammation. Gut 2016; 65:
rent knowledge and future needs. J Pediatr 19 Garrido D, Barile D, Mills DA: A molecular 33–46.
2006;149:S49–S61. basis for bifidobacterial enrichment in the 30 Mezoff EA, Hawkins JA, Ollberding NJ,
6 American Academy of Pediatrics, Section on infant gastrointestinal tract. Adv Nutr 2012; Karns R, Morrow AL, Helmrath MA: The
Breastfeeding: Breastfeeding and the use of 3:415S–421S. human milk oligosaccharide 2′-fucosyllac-
human milk. Pediatrics 2012;129;e827–e841. 20 Steenhout P, Sperisen P, Martin F-P, Sprenger tose augments the adaptive response to ex-
7 Horta BL, Victora CG: Long-term Effects of N, Wernimont S, Pecquet S, Berger B: Term tensive intestinal resection. Am J Physiol
Breastfeeding. Geneva, World Health Orga- infant formula supplemented with human Gastrointest Liver Physiol 2016; 310:G427–
nization, 2013. http://apps.who.int/iris/bitst milk oligosaccharides (2′fucosyllactose and G438.
ream/10665/79198/1/9789241505307_eng.pdf lacto-N-neotetraose) shifts stool microbiota 31 Wickramasinghe S, Pacheco AR, Lemay DG,
(accessed September 15, 2016). and metabolic signatures closer to that of Mills DA: Bifidobacteria grown on human
8 Li M, Wang M, Donovan SM: Early develop- breastfed infants. FASEB J 2016;30 (suppl 1): milk oligosaccharides downregulate the ex-
ment of the gut microbiome and immune- 275.7 pression of inflammation-related genes in
mediated childhood disorders. Semin Re- 21 Li M, Bauer LL, Chen X, Wang M, Kuhlen- Caco-2 cells. BMC Microbiol 2015;15:172.
prod Med 2014;32:74–86. schmidt TB, Kuhlenschmidt MS, Fahey GC 32 Marcobal A, Sonnenburg JL: Human milk
9 Wang M, Monaco MH, Donovan SM: Impact Jr, Donovan SM: Microbial composition and oligosaccharide consumption by intestinal
of early gut microbiota on immune and met- in vitro fermentation patterns of human microbiota. Clin Microbiol Infect 2012;
abolic development and function. Semin Fe- milk oligosaccharides and prebiotics differ 18(suppl 4):12–15.
tal Neonat Med 2016, Epub ahead of print. between formula-fed and sow-reared piglets. 33 Kavanaugh D, O’Callaghan J, Kilcoyne M,
10 Smilowitz JT, Lebrilla CB, Mills DA, Ger- J Nutr 2012;142:681–689. Kane M, Joshi L, Hickey RM: The intestinal
man JB, Freeman SL: Breast milk oligosac- 22 Li M, Monaco MH, Wang M, Comstock SS, glycome and its modulation by diet and nu-
charides: structure-function relationships in Kuhlenschmidt TB, Fahey GC Jr, Miller MJ, trition. Nutr Rev 2015;73:359–375.
the neonate. Annu Rev Nutr 2014; 34: 143– Kuhlenschmidt MS, Donovan SM: Human 34 Ewaschuk JB, Diaz H, Meddings L, Dieder-
169. milk oligosaccharides shorten rotavirus-in- ichs B, Dmytrash A, Backer J, Looijer-van
11 Kunz C, Meyer C, Collado MC, Geiger L, duced diarrhea and modulate piglet mucosal Langen M, Madsen KL: Secreted bioactive
García-Mantrana I, Bertua-Ríos B, Mar- immunity and colonic microbiota. ISME J factors from Bifidobacterium infantis en-
tínez-Costa C, Borsch C, Rudloff S: Influ- 2014;8:1609–1620. hance epithelial cell barrier function. Am J
ence of gestational age, secretor and Lewis 23 Goto Y, Kiyono H: Epithelial barrier: an in- Physiol Gastrointest Liver Physiol 2008; 295:
blood group status on the oligosaccharide terface for the cross-communication be- 1025–1034.
content of human milk. J Pediatr Gastroen- tween gut flora and immune system. Immu- 35 Macpherson AJ, Geuking MB, McCoy KD:
terol Nutr 2016, Epub ahead of print. nol Rev 2012;245:147–163. Immune responses that adapt the intestinal
12 Thurl S, Munzert M, Henker J, Boehm G, 24 Sassone-Corsi M, Raffatellu M: No vacancy: mucosa to commensal intestinal bacteria.
Müller-Werner B, Jelinek J, Stahl B: Varia- how beneficial microbes cooperate with im- Immunology 2005;115:153–162.
tion of human milk oligosaccharides in rela- munity to provide colonization resistance to 36 Comstock SS, Wang M, Hester SN, Li M,
tion to milk groups and lactational periods. pathogens. J Immunol 2015;194:4081–4087. Donovan SM: Select human milk oligosac-
Br J Nutr 2010;104:1261–1271. 25 Hester SN, Donovan SM: Individual and charides directly modulate peripheral blood
13 Martín-Sosa S, Martín MJ, García-Pardo combined effects of nucleotides and human mononuclear cells isolated from 10-d-old
LA, Hueso P: Sialyloligosaccharides in hu- milk oligosaccharides on proliferation, apo- pigs. Br J Nutr 2014;111:819–828.
man and bovine milk and in infant formulas: ptosis and necrosis in a human fetal intesti- 37 Goehring KC, Kennedy AD, Prieto PA, Buck
variations with the progression of lactation. nal cell line. Food Nutr Sci 2012; 3: 1567– RH: Direct evidence for the presence of
J Dairy Sci 2003;86:52–59. 1576. human milk oligosaccharides in the circula-
14 Newburg DS, He Y: Neonatal gut microbiota 26 Holscher HD, Davis SR, Tappenden KA: Hu- tion of breastfed infants. PLoS One 2014; 9:
and human milk glycans cooperate to atten- man milk oligosaccharides influence matu- e101692.
uate infection and inflammation. Clin Ob- ration of human intestinal Caco-2Bbe and 38 Marriage BJ, Buck RH, Goehring KC, Oliver
stet Gynecol 2015;58:814–826. HT-29 cell lines. J Nutr 2014;144:586–591. JS, Williams JA: Infants fed a lower calorie
15 Kulinich A, Liu L: Human milk oligosaccha- formula with 2′-fucosyllactose (2′FL) show
rides: the role in the fine-tuning of innate im- growth and 2′FL uptake like breast-fed in-
mune responses. Carbohydr Res 2016; 432: fants. J Pediatr Gastroenterol Nutr 2015; 61:
62–70. 649–658.

50 Ann Nutr Metab 2016;69(suppl 2):42–51 Donovan/Comstock


DOI: 10.1159/000452818
39 Ruhaak LR, Stroble C, Underwood MA, Le- 48 Earl LA, Bi S, Baum LG: N- and O-glycans 55 Hanlon PR, Thorsrud BA: A 3-week pre-
brilla CB: Detection of milk oligosaccharides modulate galectin-1 binding, CD45 signal- clinical study of 2′-fucosyllactose in farm
in plasma of infants. Anal Bioanal Chem ing, and T cell death. J Biol Chem 2010; 285: piglets. Food Chem Toxicol 2014; 74: 343–
2014;406:5775–5784. 2232–2244. 348.
40 Bode L, Kunz C, Muhly-Reinholz M, Mayer 49 Hester SN, Chen X, Li M, Monaco MH, 56 Kashyap PC, Marcobal A, Ursell LK, Smits
K, Seeger W, Rudloff S: Inhibition of mono- Comstock SS, Kuhlenschmidt TB, Kuhlen- SA, Sonnenburg ED, Costello EK, Higgin-
cyte, lymphocyte, and neutrophil adhesion schmidt MS, Donovan SM: Human milk oli- bottom SK, Domino SE, Holmes SP, Relman
to endothelial cells by human milk oligosac- gosaccharides inhibit rotavirus infectivity in DA, Knight R, Gordon JI, Sonnenburg JI:
charides. Thromb Haemost 2004; 92: 1402– vitro and in acutely infected piglets. Br J Nutr Genetically dictated change in host mucus
1410. 2013;110:1233–1242. carbohydrate landscape exerts a diet-depen-
41 Bode L, Rudloff S, Kunz C, Strobel S, Klein 50 Luhn K, Wild MK: Human deficiencies of dent effect on the gut microbiota. Proc Natl
N: Human milk oligosaccharides reduce fucosylation and sialylation affecting selec- Acad Sci USA 2013;110:17059–17064.
platelet-neutrophil complex formation lead- tin ligands. Semin Immunopathol 2012; 34: 57 Okano M, Satoskar AR, Nishizaki K, Harn
ing to a decrease in neutrophil beta 2 integrin 383–399. DA Jr: Lacto-N-fucopentaose III found on
expression. J Leukoc Biol 2004; 76:820–826. 51 Castillo-Courtade L, Han S, Lee S, Mian FM, Schistosoma mansoni egg antigens functions
42 Rabinovich GA, Croci DO: Regulatory cir- Buck R, Forsythe P: Attenuation of food al- as adjuvant for proteins by inducing Th2-
cuits mediated by lectin-glycan interactions lergy symptoms following treatment with type response. J Immunol 2001; 167: 442–
in autoimmunity and cancer. Immunity human milk oligosaccharides in a mouse 450.
2012;36:322–335. model. Allergy 2015;70:1091–1102. 58 Seppo AE, Autran CA, Bode L, Järvinen KM:
43 Schnaar RL: Glycans and glycan-binding 52 Kurakevich E, Hennet T, Hausmann M, Human milk oligosaccharides and develop-
proteins in immune regulation: a concise in- Rogler G, Borsig L: Milk oligosaccharide ment of cow’s milk allergy in infants. J Al-
troduction to glycobiology for the allergist. sialyl(α2,3)lactose activates intestinal lergy Clin Immunol 2016, Epub ahead of
J Allergy Clin Immunol 2015;135:609–615. CD11c+ cells through TLR4. Proc Natl Acad print.
44 Geijtenbeek TB, van Vliet SJ, Engering A, Sci USA 2013;110:17444–17449. 59 Huang YL, Chassard C, Hausmann M, von
‘t Hart BA, van Kooyk Y: Self- and nonself- 53 Goerhring KC, Marriage BJ, Oliver JS, Wild- Itzstein M, Hennet T: Sialic acid catabolism
recognition by C-type lectins on dendritic er JA, Barrett EG, Buck RH: Similar to those drives intestinal inflammation and micro-
cells. Annu Rev Immunol 2004;22:33–54. who are breastfed, infants fed a formula bial dysbiosis in mice. Nat Commun 2015;6:
45 Koning N, Kessen SF, Van Der Voorn JP, Ap- containing 2′-fucosyllactose have lower in- 8141.
pelmelk BJ, Jeurink PV, Knippels LM, Gars- flammatory cytokines in a randomized 60 Severi E, Hood DW, Thomas GH: Sialic acid
sen J, Van Kooyk Y: Human milk blocks DC- controlled trial. J Nutr 2016, DOI: 10.3945/ utilization by bacterial pathogens. Microbi-
SIGN-pathogen interaction via MUC1. Front jn.116.236919. ology 2007;153:2817–2822.
Immunol 2015;6:112. 54 Puccio G, Alliet P, Cajozzo C, Janssens E, 61 Audry M, Jeanneau C, Imberty A, Harduin-
46 Macauley MS, Crocker PR, Paulson JC: Si- Corsello G, Wernimont S, Egli D, Gosoniu L, Lepers A, Delannoy P, Breton C: Current
glec-mediated regulation of immune cell Sprenger N, Steenhout P: Effects of infant trends in the structure-activity relationships
function in disease. Nat Rev Immunol 2014; formula with human milk oligosaccharides of sialyltransferases. Glycobiology 2011; 21:
14:653–666. on growth and morbidity: a randomized 716–726.
47 Stephenson HN, Mills DC, Jones H, Milioris multicenter trial. JPGN, in press. 62 Kuijf ML, Samsom JN, van Rijs W, Bax M,
E, Copland A, Dorrell N, Wren BW, Crocker Huizinga R, Heikema AP, van Doorn PA, van
PR, Escors D, Bajaj-Elliott M: Pseudaminic Belkum A, van Kooyk Y, Burgers PC, Luider
acid on Campylobacter jejuni flagella modu- TM, Endtz HP, Nieuwenhuis EE, Jacobs BC:
lates dendritic cell IL-10 expression via Si- TLR4-mediated sensing of Campylobacter
glec-10 receptor: a novel flagellin-host inter- jejuni by dendritic cells is determined by
action. J Infect Dis 2014;210:1487–1498. sialylation. J Immunol 2010;185:748–755.

HMO Influence Neonatal Mucosal and Ann Nutr Metab 2016;69(suppl 2):42–51 51
Systemic Immunity DOI: 10.1159/000452818

Вам также может понравиться