Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Mechanisms underlying immune effects of dietary oligosaccharides1–4

Prescilla V Jeurink, Betty CAM van Esch, Anneke Rijnierse, Johan Garssen, and Le´on MJ Knippels

ABSTRACT health” (1), whereas probiotics according to the FAO/WHO are


The WHO refers to human milk as the nutritional gold standard for “live micro-organisms which, when administered in adequate
term infants. Human milk contains many immunomodulatory com- amounts, confer a health benefit on the host” (2).

Downloaded from https://academic.oup.com/ajcn/article/98/2/572S/4577367 by guest on 18 October 2021


pounds, including oligosaccharides. Human-milk oligosaccharides In this review, an overview of the prebiotic role of dietary
can serve as prebiotics because the nondigestible oligosaccharides oligosaccharides on the microbiota as well as the microbiota-
present in human milk show a clear bifidogenic effect on the gut independent immune modulation by these prebiotics is provided.
microbiota. Dietary oligosaccharide structures that have prebiotic In addition, recent publications that report on the pathways by
effects similar to human-milk oligosaccharides include galacto- which pre- and probiotics might exert their immunomodulatory
oligosaccharides, fructo-oligosaccharides, and pectin-derived acidic effect are summarized.
oligosaccharides. Both animal studies and human clinical trials
showed that dietary intervention with these dietary oligosaccharides
in early life could lead to the prevention of atopic dermatitis, food HOST HOMEOSTASIS
allergy, and allergic asthma. The immune-modulating effects of these The gastrointestinal tract is the largest immune organ in the
oligosaccharides are likely assisted via alteration of the intestinal body. It contains w65% of the overall immunologic tissues and
microbiota or in a microbiota-independent manner by direct interac- #80% of the immunoglobulin-producing tissues of the body.
tion on immune cells or both. In this review, an overview of the pre- These immunologic areas in the gastrointestinal tract are in-
biotic role of dietary oligosaccharides on the microbiota and the volved in managing explicit pathogenic threats and in preventing
microbiota-independent immune modulation by these prebiotics is the induction of inflammation by damage-associated molecular
provided. In addition, recent publications that report on the pathways patterns. Recently, it has become clear that many gut-related
by which the oligosaccharides might exert their direct immunomod- disorders are correlated with an imbalance in the microbiota and
ulatory effect are summarized. Am J Clin Nutr 2013;98 the immune system. Well-known examples include inflamma-
(suppl):572S–7S. tory bowel diseases such as Crohn disease and ulcerative colitis
(3, 4). In addition, impaired immune functioning is frequently
INTRODUCTION associated with irritable bowel syndrome, which is characterized
by the frequent occurrence of gut discomfort symptoms such as
Hippocrates (460–377 BC), considered as the father of West-
diarrhea, constipation, or bloating (5–7). It is also recognized
ern medicine, stated, “Let medicine be thy food and food be thy
that in other disease pathologies such as chronic heart failure
medicine,” which indicates that the medicinal properties of
(8), autism (9), allergy (10–12), and HIV (13), the microbiota
various nutritional components have been appreciated since
and general gut integrity play a role.
ancient times. Hippocrates also documented medicinal proper-
The composition and development of the intestinal microbiota
ties of a powder made from willow bark and leaves containing
in infants are influenced by genetic factors, exposure to microbes,
salicylic acid, which is now the most well-known painkiller
and mode of delivery but also to a large extent by the composition
found in almost every home (aspirin). This shows how nature
of human milk, which also contributes to the maturation of the
delivers a chemical structure for medication such as aspirin. To
infants’ immune system (14–16). Clinical data show that
date, almost 70% of available drugs originate from chemical
compounds found in vegetables, plants, and fruit. In addition, 1
From the Danone Research–Centre for Specialised Nutrition, Immunol-
there is a large body of evidence that indicates that human health ogy Platform, Wageningen, Netherlands (PVJ, BCAMvE, AR, JG, and
is also modulated through interactions with microbes present in LMJK), and Utrecht Institute for Pharmaceutical Sciences, Faculty of Sci-
the intestine. The beneficial effect of specific microbes was ence, Utrecht University, Utrecht, Netherlands (PVJ, BCAMvE, AR, JG, and
postulated by the Nobel Prize winner Elie Metchnikoff .100 y LMJK).
2
ago. He stated that certain lactic acid bacteria present in yogurt Presented at the symposium “Bringing Science to Early Life Nutrition”
were beneficial to health and had life-prolonging properties. held in Utrecht, Netherlands, 25 October 2011.
3
This notion has been validated by a broad array of products that Supported by the non-profitable Dutch Top Institute Pharma Grant of the
project “Immune Modulation and Tolerance Induction, Prevention, and In-
aim to improve human health through direct administration of
hibition of Inflammatory Diseases (T1-214).”
prebiotics or probiotics. Generally, prebiotics are described as “a 4
Address correspondence to PV Jeurink, Danone Research–Centre for
selectively fermented ingredient that allows specific changes, Specialised Nutrition, PO Box 7005, 6700 CA Wageningen, Netherlands.
both in the composition and/or activity in the gastrointestinal E-mail: prescilla.jeurink@danone.com.
microflora that confers benefits upon host well-being and First published online July 3, 2013; doi: 10.3945/ajcn.112.038596.

572S Am J Clin Nutr 2013;98(suppl):572S–7S. Printed in USA. Ó 2013 American Society for Nutrition
IMMUNE EFFECTS OF OLIGOSACCHARIDES 573S
breastfed children have high numbers of the health-promoting been suggested that maternal factors such as diet have a larger
lactobacilli and bifidobacteria, whereas children that are bottle- influence on human-milk composition in later lactation stages
fed with a cow-milk formula have significantly lower numbers than in the first few months (20).
of these intestinal bacteria (17). This large impact of human
milk on an infant’s microbiota is important to reestablish the
immunologic balance that is skewed during pregnancy to avoid MICROBIOTA-INDEPENDENT IMMUNE MODULATION
rejection of the fetus by the mother’s body (15) and to set the Apart from their prebiotic effect related to microbiota changes,
metabolic homeostasis (16). HMOS can act as receptor analogs to inhibit the adhesion of
pathogens on the epithelial surface (21). There is increasing
evidence that HMOS act directly on the immune system, most
HUMAN MILK likely via specific sugar receptors on immune cells (22–25).
The WHO refers to human milk as the nutritional gold standard HMOS have been shown to interfere with in vitro leukocyte
for term infants. From a nutritional point of view, human-milk recruitment to sites of inflammation and to inhibit cell-cell in-
compounds are therefore interesting targets for immunomodu- teractions of lymphocytes via selectins (24), whereas other

Downloaded from https://academic.oup.com/ajcn/article/98/2/572S/4577367 by guest on 18 October 2021


latory research. Several studies have shown the antiinfective HMOS inhibit adhesion to endothelial cells and leukocyte roll-
properties of human milk, which reduces the incidence of gas- ing (22). Moreover, oligosaccharides have been shown to in-
trointestinal and nonenteric infections in infants (15). In addition terfere with T helper (Th)1/Th2 skewing in cord blood–derived
to these antiinfective properties, antiinflammatory properties of mononuclear cells (more interferon-g producing CD4+ T cells)
human milk have been described (15). This is mainly of im- and to affect the Th2-type immune response of allergen-specific
portance during the bacterial colonization of the mucosal surfaces T cells from peanut-allergic individuals (24, 25). These publi-
of newborns. For example, in the skin and gut, large quantities of cations support the in vitro evidence for epithelial transport of
microbial components are in direct contact with the sterile ne- HMOS (24). Interestingly, Naarding et al (23) showed that
onate. Coordination of the inflammatory response occurring after HMOS are able to bind specifically to the lectin receptor DC-
this first contact is vital. The epithelial layer, together with the SIGN [dendritic cell (DC)–specific intercellular adhesion mol-
intraepithelial and immune-competent cells in the lamina propria, ecule-3-grabbing non-integrin] expressed by DCs. DC-SIGN
is the most important player in regulating the recognition of interacts with a variety of pathogens, including HIV-1, and
microorganisms and the maintenance of gut homeostasis. As binding of HMOS inhibited the transfer of HIV-1 to CD4+ T
described in more detail by Oozeer et al (14) elsewhere in this lymphocytes. The above-described data suggest that oligosac-
supplement, this gut-ecophysiology is important for metabolic, charides act systemically and are thereby modulating the im-
immunologic, and even neurologic development, which em- mune response in a microbiota-independent manner.
phasizes the diverse activity of human-milk compounds.
Human milk contains many immunomodulatory compounds,
including IgG, IgM, and isoforms of immunoglobulins (secretory DIETARY OLIGOSACCHARIDES WITH FUNCTIONS
IgA), nucleotides, specific amino acids (taurine, polyamines), SIMILAR TO HMOSs
PUFAs (EPA, DHA), monoglycerides, leuric acid, linoleic acid, In human milk, the proportion of prebiotic carbohydrates is
cytokines and chemokines, soluble receptors (CD14, Toll-like substantial [12–15 g/L (26)], whereas prebiotic oligosaccharides
receptor 2), antibacterial proteins/peptides (lactoferrin, lyso- in cow milk are present only in trace amounts. In addition, the
syme, b-lactoglobulin, casein), intact immune cells, and car- HMOS composition is very complex because .200 different
bohydrates. Carbohydrates in human milk, such as lactose, HMOS structures have been determined (26). The HMOS
glycoconjugates, and oligosaccharides, display a multifunctional structures are often divided into 2 main categories: neutral and
activity spectrum as described in detail by Stahl (18) elsewhere acidic oligosaccharides (27). Within the neutral core structures
in this supplement. In addition to being an energy source, hu- and fucosylated carbohydrates, 4 molecular masses represent
man-milk oligosaccharides (HMOS)5 can also serve as pre- #70% of the total molecules, including isomeres of lacto-N-
biotics because the nondigestible HMOS present in human milk tetraose, lacto-N-neotetraose, lacto-n-hexaose , monofucosyl-
show a clear bifidogenic effect on the gut microbiota. However, lacto-N-hexaose, and difucosyl lacto-N-hexaose (26). The acidic
the HMOS composition of human milk cannot be generalized oligosaccharides (AOS) contain sialic acids or sulfate groups,
because the mother’s secretor status and Lewis blood type de- are present in human milk in relatively low amounts (1.5–3.3
termine the fucosylation pattern and thereby the types of HMOS g/L), and mainly consist of 5-N-acetyl-neuraminic acid (28).
present in human milk. Furthermore, Albrecht et al indicated Oligosaccharide structures that display prebiotic effects sim-
that the composition of human milk shows considerable differ- ilar to HMOS include galacto-oligosaccharides (GOS), fructo-
ences in the carbohydrate content during different phases of oligosaccharides (FOS), and AOS. Several types of GOS with
lactation (19). The literature on the effect of maternal diet on the different chemical characteristics are used in immunomodulatory
nutrient composition of human milk is scarce, although it has research. GOS produced by glycosylation of lactose, by using
b-galactosidase enzymes, is usually referred to as b-linked GOS
(29). Short-chain GOS (scGOS) has not been studied widely
5
Abbreviations used: AOS, acidic oligosaccharides; DC, dendritic cell; dp, with respect to its immunomodulating effects as a single dietary
degree of polymerization; FOS, fructo-oligosaccharides; Foxp3, forkhead box agent. Although antiallergic and antiinflammatory effects were
p3; GOS, galacto-oligosaccharides; HMOS, human-milk oligosaccharides;
lcFOS, long-chain fructo-ligosaccharides; MLN, mesenteric lymph node;
described for a-GOS (30, 31) and raffinose (31, 32), more work
pAOS, pectin-derived acidic oligosaccharides; scGOS, short-chain galacto- is needed to clarify the effects of GOS and compare these
oligosaccharides; Th, helper T cell; Treg, regulatory T cell. with similar effects of other (nondigestible) oligosaccharides.
574S JEURINK ET AL

Definitions of fructans, such as various types of FOS and inulin, case of food allergy, the allergic response most likely occurs
vary widely in the literature. Most described experiments were because of either a failure in establishing oral tolerance or
performed with unprocessed chicory inulin or fructans derived a breakdown of existing tolerance. In addition, food allergy and
from chicory inulin. Unprocessed chicory inulin is mainly eczema are strongly correlated in children ,1 y of age, and
composed of fructans with a degree of polymerization (dp) approximately one-third of them display atopic dermatitis
ranging from 2 to 60, ending with a terminal glucose monomer. symptoms as a consequence of sensitization to food allergens
Partially hydrolyzed inulin (short-chain FOS) has a typical dp (45–47). Food allergens can exaggerate these atopic dermatitis
range of 2–8, and there are more molecules that end without symptoms by activation of Th2 cells in the skin because the
a terminal glucose monomer compared with inulin. The physical gastrointestinal DCs are able to transport the allergen to the
removal of short-chain fructans from chicory inulin (33, 34) skin where they can degranulate mast cells (48).
leads to a mixture of fructans with terminal glucose monomers A large, prospective, randomized, double-blind, placebo-
and an approximate average dp of 22 [long-chain FOS (lcFOS)]. controlled study in which healthy infants received a mixture of
To stimulate the entire microbiota, a great variation of oli- scGOS+lcFOS+pAOS (ratio of 9:1:2) showed prevention of the
gosaccharide structures is needed, because HMOS comprise development of atopic dermatitis (49). Moreover, infants who

Downloaded from https://academic.oup.com/ajcn/article/98/2/572S/4577367 by guest on 18 October 2021


many different oligosaccharides. Therefore, AOS such as were fed with scGOS+lcFOS (ratio of 9:1) showed increased
methylated pectin–derived AOS (pAOS) are under investigation fecal secretory IgA, suggesting a positive effect on the immune
(35, 36). The addition of pAOS facilitates the required oligo- system (11). The direct beneficial effect on the immune system
saccharide diversity, because it is known that AOS act very was also shown in a study in infants at risk (ie, a family history of
specifically to their acidity, enabling them to interact with sur- atopy). In this study by Moro et al (50), the at-risk infants re-
faces and to prevent the adhesion of pathogens on the intestinal ceived a hydrolyzed formula either supplemented with the scGOS
epithelium. +lcFOS mixture or not supplemented during the first 6 mo of life.
Like HMOS, scGOS and lcFOS can complete the entire The data indicate that the incidence of atopic dermatitis and
gastrointestinal passage because they are detectable in the feces infectious diseases was reduced in infants who received the
of infants fed scGOS+lcFOS-supplemented formula (37). Several prebiotic mixture during these first 6 mo (50). This protection
clinical trials showed that feeding infants a formula supple- lasted, because the early-life intervention with these prebiotics
mented with a specific mixture of scGOS (Vivinal GOS) and also resulted in significantly lower allergic symptoms at 24 mo of
lcFOS (Beneo HP) (ratio of 9:1; Immunofortis) resulted in an age (51). Even after 5 y, preliminary observations in a subset
intestinal microbiota similar to that found in breastfed infants— indicated that scGOS+lcFOS consumption early in life has
ie, higher amounts of Bifidobacterium and Lactobacillus species a protective effect on atopic dermatitis and allergic rhinitis (52). It
(17, 38, 39). Term infants who were fed a formula containing is therefore suggested that intervention with prebiotics early in
pAOS with or without scGOS+lcFOS showed no differences in life induces a prolonged preventive effect on the development of
growth, crying, vomiting, and regurgitation patterns compared atopic disorders.
with the placebo group (35). Also in preterm infants, enteral In addition to the effect on the allergic response, van Hoffen
supplementation of a prebiotic mixture consisting of scGOS et al (53) showed that scGOS+lcFOS supplementation of high-
+lcFOS+pAOS lowered the stool pH and stool viscosity with risk infants reduced the total immunoglobulin response, modu-
a trend toward a higher stool frequency (40). The effects of lating the allergic response away from the symptom-inducing
dietary oligosaccharides in infant formula on the microbiota and IgE, whereas the vaccination response to DTP (diphtheria, tet-
their influence on human health are extensively described by anus, and polio) remained equally effective. Several other studies
Oozeer et al (14) in this supplement issue. also showed a reduced incidence of infections in combination
The first postnatal year of life seems to be a key period for with a reduction in allergic disease outcomes after supplemen-
programming the immune system. Feeding (ie, human milk) and tation with scGOS+lcFOS-containing nutrition (54–56). In HIV-
other factors to which a newborn is subjected (ie, antibiotics) 1–infected individuals who were not receiving highly active
may have an influence on the indigenous gut microbiota, in- antiretroviral therapy, dietary scGOS+lcFOS+pAOS application
testinal epithelial integrity, and the immune system. It is fur- resulted in increased bifidobacteria amounts; reduced numbers
thermore known that infants are more prone to develop Th2 of Clostridium histolyticus, Eubacterium rectale, and Clostrid-
disorders, which might be partly associated with the immature ium coccoides; reduced CD4+ T cell activation; and improved
gut. Therefore, an imbalance in the immune system can lead to natural killer cell cytotoxicity (57).
immunologic disorders, with allergies and atopic dermatitis as
the most prevalent disorders (36). Chicken egg (4–5%), cow
milk (2–3%), and peanut (.1%) are the 3 most important di- ROLE OF REGULATORY T CELLS IN IMMUNE
agnosed food allergies in young children (41, 42). Although it is RESPONSES
established that there is a strong genetic influence in the de- The maintenance of a proper balance within the immune re-
velopment of food hypersensitivity, the answer to the question sponses requires a well-orchestrated regulation. In food allergy,
of which factors are responsible for the rising number of al- the subtle balance between hypersensitivity and oral tolerance is
lergic children might also lie in the changing westernized arranged by regulatory cells, including IL-10–producing Tr1
lifestyle, nutritional habits, higher consumption of food pre- cells, natural killer T cells, and transforming growth factor
servatives and additives, environmental conditions, and air b–producing Th3 cells (58–60). Moreover, naturally occurring
pollution (43). Furthermore, the antigen type and dose, and the CD4+CD25+Foxp3+ regulatory T cells (Tregs) have shown their
age at which the first exposure occurs, are all mentioned as key importance in the maintenance of peripheral tolerance and the
players in the subsequently evoked immune response (44). In suppression of CD4+ and CD8+ T cell proliferation (61, 62).
IMMUNE EFFECTS OF OLIGOSACCHARIDES 575S
Interestingly, these Tregs can be generated de novo in, eg, the sults were obtained in a study that used another cow-milk
intestinal mucosa (63). Approximately 5–10% of the CD4+ T protein, casein. Dietary scGOS+lcFOS+pAOS before and dur-
cell population are regulatory T cells, and most of them display ing oral casein sensitization showed a strong reduction in the
the transcription factor forkhead box p3 (Foxp3). Foxp3 is allergic effector response compared with allergic control mice,
considered to be a necessary gene for Treg generation and which could be abrogated by in vivo depletion of CD25+ Tregs
maintenance (63). However, naturally occurring Tregs are (75). Overall, these results indicate active regulation of the
plastic, which means that they are capable of losing Foxp3 and mucosal immune response that occurs on sensitization with
their regulatory function and, conversely, capable of acquiring casein or whey as a consequence of dietary intervention with
various effector functions (64). In addition to their crucial role in scGOS+lcFOS+pAOS.
oral tolerance development, Tregs are also increasingly prom- Flinterman et al (76) showed that there is a considerable
inent in vaccine strategies because they serve to limit activation, chance of developing acute allergic reactions to cow milk after its
trafficking, and/or effector functions of both CD4+ and CD8+ T elimination in children with atopic eczema dermatitis syndrome
cells (65, 66). During a primary immune response, the expansion who previously did not experience problems after cow-milk
and attraction of conventional and Treg populations occur in intake. A possible explanation for this was published by van Esch

Downloaded from https://academic.oup.com/ajcn/article/98/2/572S/4577367 by guest on 18 October 2021


synchrony (67). More important, the relative accumulation of (77) who showed in a mouse model for cow-milk allergy that
Tregs at peak response significantly exceeds that of conventional partial and not extensive whey hydrolysates retained the putative
T cells, reflecting an extensive relation between Tregs and capacity to prevent allergic symptoms when given orally before
conventional T cells, which control the magnitude of the re- sensitization. In addition, the protective effects coincided with
sponse (68). Little is known about the intracellular factors and elevated numbers of Tregs in the intestine and could be adop-
external cues for the differentiation and function of distinct Treg tively transferred by using mesenteric lymph node (MLN) cells.
populations. Recently, it has been shown that both Foxp3 as well In a subsequent study, they postulated that unique mucosal
as T-bet (a transcription factor both necessary and sufficient for CD103+ DCs, present in the lamina propria and MLNs, and
Th1 differentiation) act jointly within Tregs to produce unique capable of sensing external stimuli present in the gastrointestinal
functional outcomes in Th1 type of inflammation (69), sug- tract, possess the capability to induce Tregs that are responsible
gesting the possibility of specific immune response–related Treg for the above-described tolerance induction. Because systemic
development. oral tolerance to food antigens may be generated via the in-
The effects of scGOS+lcFOS have also been studied exten- duction of Tregs in the MLNs by antigen-presenting migratory
sively in mouse models for influenza vaccination (70), allergic DCs arriving from the intestinal mucosa, the effect of dietary
asthma (71), and cow milk allergy (12). Although the influenza- scGOS+lcFOS+pAOS intervention in a cow-milk allergy model
specific vaccination response and fecal bifidobacteria and on CD103+ DCs and CD4+CD25+Foxp3+ Tregs in MLN cells
lactobacilli proportions in scGOS+lcFOS-fed mice were signifi- was investigated. It was shown that scGOS+lcFOS+pAOS could
cantly enhanced, dietary intervention with scGOS+lcFOS+pAOS improve the tolerizing capacity of a partial whey hydrolysate,
was even more effective (72). Moreover, van’t Land et al (73) and improved tolerance response coincided with increased per-
showed that dietary scGOS1lcFOS1pAOS clearly induces centages of CD11c+CD103+ DCs and Foxp3+ Tregs in MLN
a more pronounced Th1 responsiveness, as shown by increased cells (78).
Influenza-specific, delayed-type hypersensitivity responses and
T-bet expression levels. Furthermore, CD25+ Tregs played
a prominent role in the development of prebiotic scGOS+lcFOS CONCLUSIONS
+pAOS-induced immune modulation toward enhanced Th1 In this review, the prebiotic function of dietary oligosaccha-
vaccine responsiveness, because in vivo depletion of CD25+ cells rides was compared with the functionality of HMOS. Both animal
completely diminished scGOS+lcFOS+pAOS-induced immune studies and human clinical trials show that dietary intervention
modulation toward control levels. with oligosaccharides in early life could lead to the prevention of
Dietary scGOS+lcFOS+pAOS was also investigated in an atopic dermatitis, food allergy, and/or allergic asthma. It is
experimental allergic asthma model in mice (71) and showed thought that the immune-modulating effects of these oligosac-
a significant suppression of ovalbumin-induced airway in- charides are mediated via modification of the intestinal micro-
flammation and hyperresponsiveness. A similar allergy-reducing biota and/or in a microbiota-independent manner by direct
effect of pAOS was observed in a model for cow-milk allergy to interaction on immune cells. These direct “pharmaceutical
whey, in which the severity of allergic symptoms, as determined properties” of oligosaccharides are very promising and should
by a strongly reduced acute allergic skin response, in mice fed be studied further. Because most studies to date are performed in
the prebiotic diet during whey sensitization was ameliorated a preventive setup (ie, applying the prebiotics before induction
(74). This reduced allergic skin response was accompanied by of the disease), studies investigating treatment protocols are
a slight reduction in Th2-type Ig concentrations (whey-specific required to establish the capacity of oligosaccharides to reduce,
IgE and IgG1) and unaltered high concentrations of Th1-type eg, established allergies. Moreover, the studies on the role of
IgG2a. Because naturally occurring or de novo–induced CD25+ Tregs indicate that dietary intervention induces Tregs that are (at
Tregs are important for the acquisition of oral tolerance (61– least in large part) responsible for the abrogation of allergic
63), the contribution of these cells to the protective effect of the symptoms and tolerance induction. In addition to the role in the
prebiotic diet was studied by transferring spleen cells. The reduction of Th2-mediated allergic responses, Tregs also play
beneficial diet-induced reduction in the allergic response was a prominent role in the development of prebiotic-induced
transferable, and ex vivo depletion of CD25+ cells from the immune modulation toward enhanced Th1 Influenza-specific-
transferred spleen cells abolished this effect (74). Similar re- vaccination response.
576S JEURINK ET AL
We acknowledge Paul Vos, Belinda van’t Land, and Bastiaan Schouten for 17. Moro G, Minoli I, Mosca M, Fanaro S, Jelinek J, Stahl B, Boehm G.
their expertise in prebiotics. The non-profitable Dutch Top Institute Pharma Dosage-related bifidogenic effects of galacto- and fructooligosaccharides
is acknowledged for providing assistance in the public-private partnership in formula-fed term infants. J Pediatr Gastroenterol Nutr 2002;34:291–5.
project “Immune Modulation and Tolerance Induction, Prevention, and In- 18. Georgi G, Bartke N, Wiens F, Stahl B. Functional glycans and glyco-
hibition of Inflammatory Diseases (T1-214)” and for funding part of the conjugates in human milk. Am J Clin Nutr 2013;98(suppl):578S–85S.
19. Albrecht S, Schols HA, van den Heuvel EG, Voragen AG, Gruppen H.
work described.
Occurrence of oligosaccharides in feces of breast-fed babies in their
The authors’ responsibilities were as follows—BCAMvE and AR: con- first six months of life and the corresponding breast milk. Carbohydr
tributed equally to the review of the manuscript. JG is head of the Division of Res 2011;346:2540–50.
Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of 20. Nommsen LA, Lovelady CA, Heinig MJ, Lonnerdal B, Dewey KG.
Science at the Utrecht University; and LMJK is Group Leader of Immunol- Determinants of energy, protein, lipid, and lactose concentrations in
ogy at Danone Research–Centre for Specialised Nutrition. All authors, as human milk during the first 12 mo of lactation: the DARLING study.
indicated by the affiliations, are part of a strategic alliance between Utrecht Am J Clin Nutr 1991;53:457–65.
University and Danone Research–Centre for Specialised Nutrition. JG and 21. Boehm G, Stahl B. Oligosaccharides. In: Mattila-Sandholm T, ed.
BCAMvE are partly employed and AR, LK, and PJ fully employed by Functional dairy products. Cambridge, United Kingdom: Woodhead
Danone Research. Publishing, 2003:203–43.
22. Bode L, Kunz C, Muhly-Reinholz M, Mayer K, Seeger W, Rudloff S.

Downloaded from https://academic.oup.com/ajcn/article/98/2/572S/4577367 by guest on 18 October 2021


Inhibition of monocyte, lymphocyte, and neutrophil adhesion to en-
dothelial cells by human milk oligosaccharides. Thromb Haemost
REFERENCES 2004;92:1402–10.
1. Roberfroid M. Prebiotics: the concept revisited. J Nutr 2007;137 23. Naarding MA, Ludwig IS, Groot F, Berkhout B, Geijtenbeek TB,
(suppl):830S–7S. Pollakis G, Paxton WA. Lewis X component in human milk binds DC-
2. FAO/WHO. Joint FAO/WHO expert consultation: health and nu- SIGN and inhibits HIV-1 transfer to CD4+ T lymphocytes. J Clin In-
tritional properties of probiotics in food including powder milk vest 2005;115:3256–64.
with live lactic acid bacteria. Available from: http://www.who.int/ 24. Eiwegger T, Stahl B, Haidl P, Schmitt J, Boehm G, Dehlink E, Urbanek
foodsafety/publications/fs_management/en/probiotics.pdf (cited 29 R, Szepfalusi Z. Prebiotic oligosaccharides: in vitro evidence for gas-
March 2012). trointestinal epithelial transfer and immunomodulatory properties.
3. Sartor RB. Microbial influences in inflammatory bowel diseases.
Pediatr Allergy Immunol 2010;21:1179–88.
Gastroenterology 2008;134:577–94.
25. Eiwegger T, Stahl B, Schmitt J, Boehm G, Gerstmayr M, Pichler J,
4. Strober W, Fuss I, Mannon P. The fundamental basis of inflammatory
Dehlink E, Loibichler C, Urbanek R, Szepfalusi Z. Human milk–derived
bowel disease. J Clin Invest 2007;117:514–21.
oligosaccharides and plant-derived oligosaccharides stimulate cytokine
5. Clavel T, Haller D. Molecular interactions between bacteria, the epi-
production of cord blood T-cells in vitro. Pediatr Res 2004;56:536–40.
thelium, and the mucosal immune system in the intestinal tract: im-
26. Garrido D, Kim JH, German JB, Raybould HE, Mills DA. Oligosac-
plications for chronic inflammation. Curr Issues Intest Microbiol 2007;
charide binding proteins from Bifidobacterium longum subsp. infantis
8:25–43.
reveal a preference for host glycans. PLoS ONE 2011;6:e17315.
6. Kassinen A, Krogius-Kurikka L, Makivuokko H, Rinttila T, Paulin L,
27. Nakhla T, Fu D, Zopf D, Brodsky NL, Hurt H. Neutral oligosaccharide
Corander J, Malinen E, Apajalahti J, Palva A. The fecal microbiota of
irritable bowel syndrome patients differs significantly from that of content of preterm human milk. Br J Nutr 1999;82:361–7.
healthy subjects. Gastroenterology 2007;133:24–33. 28. Thurl S, Munzert M, Henker J, Boehm G, Muller-Werner B, Jelinek J,
7. Ohman L, Lindmark AC, Isaksson S, Posserud I, Strid H, Sjovall H, Stahl B. Variation of human milk oligosaccharides in relation to milk
Simren M. B-cell activation in patients with irritable bowel syndrome groups and lactational periods. Br J Nutr 2010;104:1261–71.
(IBS). Neurogastroenterol Motil 2009;21:644–50, e27. 29. Yamashita A, Hashimoto H, Fujita K, Okada M, Mori S, Kitahata S.
8. Sandek A, Anker SD, von Haehling S. The gut and intestinal bacteria Reverse reaction of Aspergillus niger APC-9319 alpha-galactosidase in
in chronic heart failure. Curr Drug Metab 2009;10:22–8. a supersaturated substrate solution: production of alpha-linked gal-
9. White JF. Intestinal pathophysiology in autism. Exp Biol Med (May- actooligosaccharide (alpha-GOS). Biosci Biotechnol Biochem 2005;
wood) 2003;228:639–49. 69:1381–8.
10. Hougee S, Vriesema AJ, Wijering SC, Knippels LM, Folkerts G, 30. Abe C, Fujita K, Kikuchi E, Hirano S, Kuboki H, Yamashita A, Ha-
Nijkamp FP, Knol J, Garssen J. Oral treatment with probiotics reduces shimoto H, Mori S, Okada M. Effects of alpha-linked gal-
allergic symptoms in ovalbumin-sensitized mice: a bacterial strain actooligosaccharide on adjuvant-induced arthritis in Wistar rats and
comparative study. Int Arch Allergy Immunol 2010;151:107–17. type II collagen-induced arthritis in DBA/1J mice. Int J Tissue React
11. Scholtens PA, Alliet P, Raes M, Alles MS, Kroes H, Boehm G, 2004;26:65–73.
Knippels LM, Knol J, Vandenplas Y. Fecal secretory immunoglobulin 31. Sonoyama K, Watanabe H, Watanabe J, Yamaguchi N, Yamashita A,
A is increased in healthy infants who receive a formula with short- Hashimoto H, Kishino E, Fujita K, Okada M, Mori S, et al. Allergic
chain galacto-oligosaccharides and long-chain fructo-oligosaccharides. airway eosinophilia is suppressed in ovalbumin-sensitized Brown
J Nutr 2008;138:1141–7. Norway rats fed raffinose and alpha-linked galactooligosaccharide. J
12. Schouten B, van Esch BC, Hofman GA, van Doorn SA, Knol J, Nauta Nutr 2005;135:538–43.
AJ, Garssen J, Willemsen LE, Knippels LM. Cow milk allergy 32. Watanabe H, Sonoyama K, Watanabe J, Yamaguchi N, Kikuchi H,
symptoms are reduced in mice fed dietary synbiotics during oral sen- Nagura T, Aritsuka T, Fukumoto K, Kasai T. Reduction of allergic
sitization with whey. J Nutr 2009;139:1398–403. airway eosinophilia by dietary raffinose in Brown Norway rats. Br J
13. Hofer U, Speck RF. Disturbance of the gut-associated lymphoid tissue Nutr 2004;92:247–55.
is associated with disease progression in chronic HIV infection. Semin 33. Coussement PA. Inulin and oligofructose: safe intakes and legal status.
Immunopathol 2009;31:257–66. J Nutr 1999;129:1412S–7S.
14. Oozeer R, van Limpt K, Ludwig T, Ben Amor K, Martin R, Wind RD, 34. van Loo J, Coussement P, de Leenheer L, Hoebregs H, Smits G. On the
Boehm G, Knol J. Intestinal microbiology in early life: specific pre- presence of inulin and oligofructose as natural ingredients in the
biotics can have similar functionalities as human-milk oligosaccha- Western diet. Crit Rev Food Sci Nutr 1995;35:525–52.
rides. Am J Clin Nutr 2013;98(suppl):561S–71S. 35. Fanaro S, Jelinek J, Stahl B, Boehm G, Kock R, Vigi V. Acidic oli-
15. Van’t Land B, Boehm G, Garssen J. Breast milk: components with gosaccharides from pectin hydrolysate as new component for infant
immune modulating potential and their possible role in immune me- formulae: effect on intestinal flora, stool characteristics, and pH. J
diated disease resistance. In: Watson R, Zibadi S, Preedy V, eds. Di- Pediatr Gastroenterol Nutr 2005;41:186–90.
etary components and immune function. Chapter 25. Dordrecht, 36. Rijnierse A, Jeurink PV, van Esch BC, Garssen J, Knippels LM. Food-
Netherlands: Springer Science, 2010:25–41. derived oligosaccharides exhibit pharmaceutical properties. Eur J
16. Nauta AJ, Ben Amor K, Knol J, Garssen J, van der Beek EM. Rele- Pharmacol 2011;668(suppl 1):S117–23.
vance of pre- and postnatal nutrition to development and interplay 37. Moro GE, Stahl B, Fanaro S, Jelinek J, Boehm G, Coppa GV. Dietary
between the microbiota and metabolic and immune systems. Am J Clin prebiotic oligosaccharides are detectable in the faeces of formula-fed
Nutr 2013;98(suppl):586S–93S. infants. Acta Paediatr Suppl 2005;94:27–30.
IMMUNE EFFECTS OF OLIGOSACCHARIDES 577S
38. Knol J, Scholtens P, Kafka C, Steenbakkers J, Gro S, Helm K, Klarczyk 59. O’Garra A, Vieira P. Regulatory T cells and mechanisms of immune
M, Schopfer H, Bockler HM, Wells J. Colon microflora in infants fed system control. Nat Med 2004;10:801–5.
formula with galacto- and fructo-oligosaccharides: more like breast-fed 60. van Wijk F, Wehrens EJ, Nierkens S, Boon L, Kasran A, Pieters R,
infants. J Pediatr Gastroenterol Nutr 2005;40:36–42. Knippels LM. CD4+CD25+ T cells regulate the intensity of hyper-
39. Veereman-Wauters G, Staelens S, Van de Broek H, Plaskie K, Wesling sensitivity responses to peanut, but are not decisive in the induction of
F, Roger LC, McCartney AL, Assam P. Physiological and bifidogenic oral sensitization. Clin Exp Allergy 2007;37:572–81.
effects of prebiotic supplements in infant formulae. J Pediatr Gastro- 61. Izcue A, Powrie F. Special regulatory T-cell review: regulatory T cells and
enterol Nutr 2011;52:763–71. the intestinal tract—patrolling the frontier. Immunology 2008;123:6–10.
40. Westerbeek EA, Hensgens RL, Mihatsch WA, Boehm G, Lafeber HN, 62. Kashyap M, Thornton AM, Norton SK, Barnstein B, Macey M,
van Elburg RM. The effect of neutral and acidic oligosaccharides on Brenzovich J, Shevach E, Leonard WJ, Ryan JJ. Cutting edge: CD4 T
stool viscosity, stool frequency and stool pH in preterm infants. Acta cell-mast cell interactions alter IgE receptor expression and signaling. J
Paediatr 2011;100:1426–31. Immunol 2008;180:2039–43.
41. Wang J, Sampson HA. Food allergy. J Clin Invest 2011;121:827–35. 63. Huehn J, Polansky JK, Hamann A. Epigenetic control of FOXP3 ex-
42. Branum AM, Lukacs SL. Food allergy among U.S. children: trends in pression: the key to a stable regulatory T-cell lineage? Nat Rev Im-
prevalence and hospitalizations. NCHS data brief no. 10. Hyattsville, munol 2009;9:83–9.
MD: National Center for Health Statistics, 2008:1–8. 64. Murai M, Krause P, Cheroutre H, Kronenberg M. Regulatory T-cell
43. Lack G. Epidemiologic risks for food allergy. J Allergy Clin Immunol stability and plasticity in mucosal and systemic immune systems.

Downloaded from https://academic.oup.com/ajcn/article/98/2/572S/4577367 by guest on 18 October 2021


2008;121:1331–6. Mucosal Immunol 2010;3:443–9.
44. Strobel S. Immunity induced after a feed of antigen during early life: 65. Shevach EM, DiPaolo RA, Andersson J, Zhao DM, Stephens GL,
oral tolerance v. sensitisation. Proc Nutr Soc 2001;60:437–42. Thornton AM. The lifestyle of naturally occurring CD4+ CD25+
45. Werfel T, Breuer K. Role of food allergy in atopic dermatitis. Curr Foxp3+ regulatory T cells. Immunol Rev 2006;212:60–73.
Opin Allergy Clin Immunol 2004;4:379–85. 66. Vignali DA, Collison LW, Workman CJ. How regulatory T cells work.
46. Hill DJ, Heine RG, Hosking CS, Brown J, Thiele L, Allen KJ, Su J, Nat Rev Immunol 2008;8:523–32.
Varigos G, Carlin JB. IgE food sensitization in infants with eczema 67. Haribhai D, Lin W, Relland LM, Truong N, Williams CB, Chatila TA.
attending a dermatology department. J Pediatr 2007;151:359–63. Regulatory T cells dynamically control the primary immune response
47. Hauk PJ. The role of food allergy in atopic dermatitis. Curr Allergy to foreign antigen. J Immunol 2007;178:2961–72.
Asthma Rep 2008;8:188–94. 68. Belkaid Y, Rouse BT. Natural regulatory T cells in infectious disease.
48. Leung DY. Atopic dermatitis: new insights and opportunities for Nat Immunol 2005;6:353–60.
therapeutic intervention. J Allergy Clin Immunol 2000;105:860–76. 69. Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH. A
49. Grüber C, van Stuijvenberg M, Mosca F, Moro G, Chirico G, Braegger novel transcription factor, T-bet, directs Th1 lineage commitment. Cell
CP, Riedler J, Boehm G, Wahn U. Reduced occurrence of early atopic 2000;100:655–69.
dermatitis because of immunoactive prebiotics among low-atopy-risk 70. Vos AP, Haarman M, Buco A, Govers M, Knol J, Garssen J, Stahl B,
infants. J Allergy Clin Immunol 2010;126:791–7. Boehm G, M’Rabet L. A specific prebiotic oligosaccharide mixture
50. Moro G, Arslanoglu S, Stahl B, Jelinek J, Wahn U, Boehm G. A stimulates delayed-type hypersensitivity in a murine influenza vacci-
mixture of prebiotic oligosaccharides reduces the incidence of atopic nation model. Int Immunopharmacol 2006;6:1277–86.
dermatitis during the first six months of age. Arch Dis Child 2006;91: 71. Vos AP, van Esch BC, Stahl B, M’Rabet L, Folkerts G, Nijkamp FP,
814–9. Garssen J. Dietary supplementation with specific oligosaccharide
51. Arslanoglu S, Moro GE, Schmitt J, Tandoi L, Rizzardi S, Boehm G. mixtures decreases parameters of allergic asthma in mice. Int Im-
Early dietary intervention with a mixture of prebiotic oligosaccharides munopharmacol 2007;7:1582–7.
reduces the incidence of allergic manifestations and infections during 72. Vos AP, Haarman M, van Ginkel JW, Knol J, Garssen J, Stahl B,
the first two years of life. J Nutr 2008;138:1091–5. Boehm G, M’Rabet L. Dietary supplementation of neutral and acidic
52. Arslanoglu S, Moro GE, Boehm G, Wienz F, Stahl B, Bertino E, oligosaccharides enhances Th1-dependent vaccination responses in
Boehm G. Early neutral prebiotic oligosaccharide supplementation mice. Pediatr Allergy Immunol 2007;18:304–12.
reduces the incidence of some allergic manifestations in the first 5 73. van’t Land B, Schijf M, van Esch BC, van Bergenhenegouwen J,
years of life. J Biol Regul Homeost Agents 2012;26(suppl):49S- Bastiaans J, Schouten B, Boon L, Garssen J. Regulatory T-cells have
59S. a prominent role in the immune modulated vaccine response by specific
53. van Hoffen E, Ruiter B, Faber J, M’Rabet L, Knol EF, Stahl B, oligosaccharides. Vaccine 2010;28:5711–7.
Arslanoglu S, Moro G, Boehm G, Garssen J. A specific mixture of 74. Schouten B, van Esch BC, Hofman GA, Boon L, Knippels LM,
short-chain galacto-oligosaccharides and long-chain fructo-oligosac- Willemsen LE, Garssen J. Oligosaccharide-induced whey-specific
charides induces a beneficial immunoglobulin profile in infants at high CD25(+) regulatory T-cells are involved in the suppression of cow milk
risk for allergy. Allergy 2009;64:484–7. allergy in mice. J Nutr 2010;140:835–41.
54. Boehm G, Jelinek J, Knol J, M’Rabet L, Stahl B, Vos P, Garssen J. 75. Schouten B, van Esch BC, Hofman GA, de Kivit S, Boon L, Knippels
Prebiotics and immune responses. J Pediatr Gastroenterol Nutr 2004;39 LM, Garssen J, Willemsen LE. A potential role for CD25+ regulatory
(suppl 3):S772–3. T-cells in the protection against casein allergy by dietary non-digestible
55. Coppa GV, Zampini L, Galeazzi T, Gabrielli O. Prebiotics in human carbohydrates. Br J Nutr 2012;107:96–105.
milk: a review. Dig Liver Dis 2006;38(suppl 2):S291–4. 76. Flinterman AE, Knulst AC, Meijer Y, Bruijnzeel-Koomen CA,
56. Morrow AL, Rangel JM. Human milk protection against infectious Pasmans SG. Acute allergic reactions in children with AEDS after
diarrhea: implications for prevention and clinical care. Semin Pediatr prolonged cow’s milk elimination diets. Allergy 2006;61:370–4.
Infect Dis 2004;15:221–8. 77. van Esch BC, Schouten B, de Kivit S, Hofman GA, Knippels LM,
57. Gori A, Rizzardini G, Van’t Land B, Amor KB, van Schaik J, Torti C, Willemsen LE, Garssen J. Oral tolerance induction by partially hy-
Quirino T, Tincati C Bandera A, Knol J, et al. Specific prebiotics drolyzed whey protein in mice is associated with enhanced numbers of
modulate gut microbiota and immune activation in HAART-naive HIV- Foxp3(+) regulatory T-cells in the mesenteric lymph nodes. Pediatr
infected adults: results of the “COPA” pilot randomized trial. Mucosal Allergy Immunol 2011;22:820–6.
Immunol 2011;4:554–63. 78. van Esch BC. Avoidance versus tolerance: new concepts for allergy
58. Akbari O, Umetsu DT. Role of regulatory dendritic cells in allergy and management. PhD dissertation. University of Utrecht, Utrecht, Neth-
asthma. Curr Opin Allergy Clin Immunol 2004;4:533–8. erlands, 2011.

You might also like