Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Fucosylated but Not Sialylated Milk Oligosaccharides Diminish Colon Motor Contractions

  • John Bienenstock ,

    bienens@mcmaster.ca

    Affiliations McMaster Brain-Body Institute at St Joseph’s Healthcare, Hamilton, Ontario, Canada, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada

  • Rachael H. Buck,

    Affiliation Abbott Nutrition, a Division of Abbott Laboratories, Columbus, Ohio, United States of America

  • Hawley Linke,

    Affiliation Abbott Nutrition, a Division of Abbott Laboratories, Columbus, Ohio, United States of America

  • Paul Forsythe,

    Affiliations Department of Medicine, McMaster University, Hamilton, Ontario, Canada, Firestone Institute for Respiratory Health, Hamilton, Ontario, Canada

  • Andrew M. Stanisz,

    Affiliation McMaster Brain-Body Institute at St Joseph’s Healthcare, Hamilton, Ontario, Canada

  • Wolfgang A. Kunze

    Affiliations McMaster Brain-Body Institute at St Joseph’s Healthcare, Hamilton, Ontario, Canada, Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada, Farncombe Family Digestive Health Institute, Hamilton, Ontario, Canada

Abstract

Human milk oligosaccharides (HMO) are being studied by different groups exploring a broad range of potential beneficial effects to the breastfed infant. Many of these effects have been attributed to a growth promotion effect on certain gut organisms such as bifidobacteria. Additionally, evidence indicates that HMO are able to directly promote positive changes in gut epithelium and immune responses under certain conditions. This study utilizes a standardized ex vivo murine colon preparation to examine the effects of sialylated, fucosylated and other HMO on gut motor contractions. Only the fucosylated molecules, 2’FL and 3’FL, decreased contractility in a concentration dependent fashion. On the basis of IC50 determinations 3’FL was greater than 2 times more effective than 2’FL. The HMO 3’SL and 6’SL, lacto-N-neotetraose (LNnT), and galactooligosaccharides (GOS) elicited no effects. Lactose was used as a negative control. Fucosylation seems to underlie this functional regulation of gut contractility by oligosaccharides, and L-fucose, while it was also capable of reducing contractility, was substantially less effective than 3’FL and 2’FL. These results suggest that specific HMO are unlikely to be having these effects via bifidogenesis, but though direct action on neuronally dependent gut migrating motor complexes is likely and fucosylation is important in providing this function, we cannot conclusively shown that this is not indirectly mediated. Furthermore they support the possibility that fucosylated sugars and fucose might be useful as therapeutic or preventative adjuncts in disorders of gut motility, and possibly also have beneficial central nervous system effects.

Introduction

Human milk oligosaccharides (HMO) constitute a repertoire of more than 100 soluble glycan structures. Their potential beneficial effects for the breastfed infant have been studied by several researchers but only one clinical trial has been reported [1-3]. The possibility to examine biological effects of HMO has increased due to technical advances which now offer for exploration previously unavailable synthetic carbohydrates. Milk oligosaccharide content varies amongst different mammals, however, they are a major molecular class in human breast milk while other species have limited and less diverse repertoires. For example, bovine milk has been shown to contain less complex, diverse and generally less abundant structures than human milk [4] and it has been generally agreed that fucosylated oligosaccharides such as 2’FL are not present. However a recent paper by Sundekilde, Barile, Meyrand, Poulsen, and Larsen 2012 [5] have shown for the first time the presence, albeit at low concentration, of some more polymerized fucosylated milk glycans. Accordingly, infant formulas and nutritional supplements derived from bovine milk lack the glycan diversity present in mother’s milk.

The observation that the feces of breastfed infants differed in their microbial content from formula fed infants supported early attempts to explain certain of the perceived advantages of breastfeeding (for review see Bode [1]). Several recent studies have identified the ability of specific, generally desirable, gut microbiota (e.g., bifidobacteria) to metabolize both sialylated and fucosylated HMO and to flourish when HMO are available as fermentation substrates in vitro and in vivo [6-8]. Additionally, Chichlowski, et al, have demonstrated HMO- associated effects on colonic cell lines that are mediated via bifidobacteria [9]. Nevertheless, it is still broadly assumed that HMO are advantageous to the host as a result of indirect action of different groups of gut bacteria, which include bifidobacteria and lactic acid bacteria, whose growth and activity they promote [10]. These observations have led to the introduction of the term prebiotics which are said to be beneficial to the host through this action (9).

However HMO can have direct effects on intestinal epithelial structure and function [11,12], and can interfere with the adhesion of infectious bacteria such as Campylobacter jejuni [13], HIV [14] and protozoan parasites such as Entamoeba histolytica [15]. Effects of HMO on the immune system have been shown by studies with both mouse and human T-lymphocytes and dendritic cells, and are mediated by interactions with cell surface C-type lectins such as P- and E-selectins, and DC-SIGN. The latter specifically binds high mannose and/or fucose-containing glycans [1]. We have found no evidence in the literature for HMO effects upon gut contractility.

Certain lactobacilli have direct effects within minutes on the enteric nervous system and on neuronally dependent gut motor contractions [16-18]. The amplitude of such peristaltic contractions of small and large intestines induced by increased pressure was reduced within minutes of luminal administration of a specific Lactobacillus strain (JB-1) but not by a Lactobacillus salivarius [16].

In a subsequent recent publication we showed that a capsular exopolysaccharide of another symbiotic but Gram negative bacteria (Bacteroides fragilis) in the gut lumen has similar effects to the parent bacteria and JB-1. This suggests the possibility that other glycans may also be functionally effective in this physiological system. We have therefore turned our attention to the examination of HMO on neuronally dependent colonic contractions [18,19].

We surmised that these effects would be easier to observe and measure in the absence of confounding factors introduced by the nutritional context and processing by the gut microbiome that may occur after oral administration.

Materials and Methods

Endotoxin free Krebs buffer was constituted as previously described [16]. Test sugars were obtained as a gift from Abbott Nutrition (Columbus, OH, USA). Purity and endotoxin concentrations are summarized in Table 1. HMO "purity" was established by high performance ion chromatography with pulsed amperometric detection (IC-PAD) using relative peak area comparisons. Moisture content was determined separately using the Karl Fischer method for moisture determination. 3’-Sialyllactose (3’ SL), 6’Sialyllactose (6’SL) and 2’Fucosylactose (2’FL) were all derived from bacterial synthesis. 3’-Fucosylactose (3’ FL) was chemically synthesized and lacto-N-neoTetraose (LNnT) was synthesized from a yeast fermentation system and purified by crystallization [3]. The galactooligosaccharides (GOS) preparation consisted of galactose (1.23%), glucose (15.8%), lactose (9.26%), 48.4% GOS and 25.3% water. L-fucose (catalogue number F2252) and β-lactose were obtained from Sigma (St Louis, MO, USA). Endotoxin levels were estimated by limulus assay (Limulus Amebocyte Lysate (LAL) QCL-1000, Lonza catalogue number 50-647U, Wilmington, MA, USA) and lipopolysaccharide (LPS) 500,000 EU/mg from Sigma (catalogue number L2637, St. Louis, MO, USA).

Adult male Swiss Webster mice were obtained from Charles River (Raleigh, NC, USA). Handling of animals and all experimental procedures were conducted in accordance with the guidelines of The Canadian Council on Animal Care and approved by the McMaster University Animal Research Ethics Board.

Organ bath intraluminal pressure recordings:

The method we used has recently been published [18] and is similar to that described by us [16] for jejunal motility. It follows the technique described by Keating et al [20] for measurements of colon. Briefly, mouse distal colon 4cm segments were excised and flushed with Krebs buffer under a 2 hPa gravity pressure head. Designated “oral” and “anal” ends of each segment were cannulated, mounted in a 20mL organ bath chamber and submerged in oxygenated Krebs. The lumen was gravity perfused at 0.5mL.min-1 with carbogen-gassed Krebs (95% O2 and 5% CO2 at room temp). The organ chamber (serosal compartment) was perfused with the same pre-warmed (34°C) Krebs buffer at 5mL.min-1. This temperature was chosen since it preserves stable gut function for up to 2 hours. At the beginning of the experiment, intraluminal pressure of 5 hPa was obtained by adjusting the heights of inflow and outflow tubes, and recordings were made at this pressure. Test materials were applied by switching the oral luminal inflow from Krebs to Krebs plus test substance by closing and opening the appropriate stopcocks.

Recordings were analyzed off-line using the Clampfit module of PClamp 9 software (Molecular Devices) as previously described for jejunal motility analysis. Intraluminal pressure changes were measured at the midpoint of the longitudinal axis of the gut segment and the pressure signals were amplified, digitized, and stored on a PC computer. Peak pressures (PPr) for individual migrating motor complexes (MMC) were measured using the cursors in Clampfit as the difference between baseline and the maximal pressure reached during the PPr. Control PPr was calculated as the average from at least 6 successive motor complexes with Krebs perfusing the lumen just before the intraluminal perfusate was switched to one containing an HMO. Then, a further 6 PPr were measured between 15 to 30 min after beginning the HMO application and after which the effects on PPr had plateaued. The latter 6 measurements were averaged to provide the “after” PPr value in the paired “before and after” experiments.

For each experiment HMO were only applied once at a particular concentration because when a given HMO altered PPr the effect did not fully wash out even after switching the luminal perfusate to Krebs buffer for 2 h. Responses each single experiment were displayed as connected lines rather than bar graphs to emphasize the before and after nature of the experimental design. Concentration-response relations were plotted using GraphPad Prism 6.0 (GraphPad Software, San Diego, CA, USA) from the pooled data of individual before and after experiments. Log (HMO)-PPr plots were fitted by a 3-point logistic (Hill) equation of the form Y = bottom + (top - bottom)/(1 + 10X - log IC50), where IC50 is the concentration of the HMO that produces 50% inhibition.

Video Recordings.

We also employed a recently developed video imaging system to record peristalsis of colonic motor contractions [18] to confirm the results we obtained in peak pressure recordings. These allow an analysis in real time of MMC and relaxation of the gut wall. These can be converted to colour in a heat map format, and additionally provide the opportunity to quantitate frequency of contractions and their time course, thus allowing calculations of velocity.

Statistics.

All statistics were calculated using GraphPad and descriptive statistics given as means +/- SE, and significance tests made using the Wilcoxon matched-pairs (before/after) signed rank test. The statistically discernible difference for tests of significance was set at P = 0.05; all tests were 2-tailed. Significance is indicated on graphs using conventional markers: *P = < 0.05; ns: P > 0.05

Results

Lactose did not alter PPr (P = 0.8, n = 6) at 1mg/mL or over a concentration range 0.5-3.0mg/mL and was thus used as a negative control when compared to Krebs buffer by itself as shown in Figure 1, and was used as such throughout the study.

thumbnail
Figure 1. Effects of lactose on peak pressures in colon motility experiments.

Lack of effect of β-Lactose (1mg/mL) on peak preasures of migrating motor complexes in 'before and after' experiments (n=6). ns=not significant.

https://doi.org/10.1371/journal.pone.0076236.g001

HMO and Monosaccharides% PurityEndotoxin Concentration EU/mg
3’Sialyllactose (3’SL)97.1%0.015
6’Sialyllactose (6’SL)96.6%0.496
2’-Fucosylactose (2’FL)95.3%0.375
3’-Fucosylactose (3’FL)99.7%Not Tested
Lacto-N-neotetraose (LNnT)95.2%0.34
Galactooligosaccharides (GOS)48.4%0.58
L-Fucose99.0%Not Tested
Lactose99.0%0.002

Table 1. Characteristics of HMO and Monosaccharides

CSV
Download CSV

The recorded propagated MMC were neuronally dependent since they were inhibited entirely by prior addition of tetrodotoxin (TTX), a specific neuron inhibitor (Figure S1), and as also shown in this figure, TTX inhibited the diminished contractions after the addition of HMO in keeping with our previous observations with jejunal preparations [16]. Of all the glycans tested (see Table 1 for list), only two (2’FL and 3’FL) demonstrated effects on PPr. Figure 2 shows the magnitude of the responses for the two effective HMOs tested (Figure 2 A& B) as well as the monosaccharide L-Fucose(Figure 2C). The onset of PPr reduction ranged from 5-15 min and did not wash out after switching the intraluminal perfusate back to one containing only Krebs buffer for up to 2 h. The IC50 for 3’FL was 420µg/mL, for 2’FL 1073µg/mL (Figure 2A and B) and 3264 µg/mL for L-Fucose. Since the effects were only observed with glycans containing fucose, we examined if L-fucose alone could modulate PPr of motor complexes in this system. As shown in Figure 2C, fucose also diminished PPr, but to a significantly lesser extent. The L-fucose effect was exhibited at a higher concentrations (IC50 =3264 µg/mL) than required for the fucosylated HMO. These results are summarized in Figure 2 inset table.

thumbnail
Figure 2. Concentration response curves for effects of fucosylated HMO.

Effects of 3’FL (A), 2’FL (B) and L-Fucose (C) on peak pressure. Inset table indicates the number of points fitted to the curve and IC50 values. * p values <0.05.

https://doi.org/10.1371/journal.pone.0076236.g002

We wished to confirm these results using a recently developed video imaging system [18] and applied this to testing the effects of 2’FL and fucose. As seen in the heat map of the effects of 2’FL at 0.5mg/mL (Figure 3) there is a statistically significant decrease in both frequency of contractions and their velocity in before and after experiments.

thumbnail
Figure 3. Effects of 0.5 mg/mL intraluminal 2’FL on colon motility.

Heat maps derived from black/white spatio-temporal video recordings of colon motility. (A) Shows regular migrating motor complexes (MMCs) during control recording. (B) Addition of 2’FL to the lumen decreased both the slope (MMC velocity) of the valleys and MMC frequency. (C, D) Summary statistics of before and after experiments showing that 2’FL significantly reduced both MMC velocity (C) and frequency (D).

https://doi.org/10.1371/journal.pone.0076236.g003

The results from experiments when sialylated HMO 3’ SL (P = 0.8, n = 6) and 6’SL (P = 0.6, n =6), as well as LNnT (P = 0.2, n = 6) and GOS (P = 0.2, n = 6) were applied for up to 1 h are shown in Figure 4. The highest concentration evaluated for any of these HMO was 5mg/mL and no significant effects on PPr were observed at any lower concentration (>0.5mg/mL). Since the glycan preparations were all contaminated with LPS, albeit at minimal concentrations, we tested the effects of its addition to the luminal perfusion fluid, at 100 and 500 EU/mL, concentrations, significantly in excess of the endotoxin levels recorded for the oligosaccharide preparations (as indicated in Table 1). LPS demonstrated no effects at either concentration (data not shown).

thumbnail
Figure 4. Before and after results of effects of non-fucosylated HMO.

Lack of effects of 3’ SL (A), 6’SL (B), LNnT (C) and GOS (D). n=>6/oligosaccharide tested. ns= not significant. Concentrations of all HMO shown at 1mg/mL for comparison purposes. None had any effect on motility up to 5mg/mL.

https://doi.org/10.1371/journal.pone.0076236.g004

Discussion

The health benefits of breastfeeding for the infant are well documented. Attempts to delineate roles for the myriad components putatively responsible for these effects have been influenced by the fact that while infant formulas differ substantially from breast milk, efforts are continually being made to improve their composition. One example has been supplementation with non-HMO glycans such as GOS and fructo-oligosaccharides (FOS) which themselves have significant positive effects on immune responses [21].

HMO are widely thought to provide a number of health benefits through their activity and interactions with immune and endocrine systems [1]. They are resistant to gastric acid and the small intestine environment and are mostly unaltered by the time they reach the large intestine [22-26], however this appears to depend on the age of the infant, maturity of gut adaptation, and the concomitant ingestion of alternative nutrients [25,26]. Evidence of growth promoting effects on bifidobacteria and lactic acid bacteria and other alterations to the colonic microbiome suggest a causal relationship with health benefits [1,10]. However specific HMO have clear direct effects on host tissues and their component cells such as epithelium [11,12]. Our observations in the ex vivo mouse colon model of peristalsis clearly show that simple fucosylated molecules have immediate effects within 5-15 minutes upon colonic neuronally dependent smooth muscle contractions. The further observation that L-fucose itself had similar functional effects (decreased amplitude of intraluminal filling pressure induced migrating motor complexes), suggests a potential role for this monosaccharide and other fucosylated molecules such as 2’FL and 3’FL, in the regulation of gut motility. On a relative concentration basis, 2’FL is almost three times more active than L-fucose, and 3’FL is additionally greater than two times more effective than 2’FL. Interestingly, while 3’FL is a normal constituent of mouse milk, 2’FL is not [27]. Mouse intestine, therefore, may not be as functionally adapted to respond to an oligosaccharide, which is not normally delivered in maternal milk. It should also be noted that unlike the neutral 2’ and 3’ fucosylactoses, the neutral HMO LNnT and the acidic sialylactose examples, 3’ SL and 6’SL showed no demonstrable effect in the motility model. Taken together these results may point to at least two mechanistic hypotheses; on the one hand, the effect of free fucose, albeit small, suggests that the monosaccharide may be limited in intestinal tissue and that its sudden availability fosters the local synthesis of fucosylated glycoconjugates that in turn attenuate neuronal activity. This suggestion would apply more to the in vivo situation than the ex vivo model we have used. However, the fact that specific fucosylated glycoconjugates such as 2’FL and 3’FL are able to elicit a substantial reduction of gut motility suggests a specific interaction of fucose and/or fucose residues with tissue receptors that in turn regulate gut motility.

The best interpretation of the demonstrated modulation of neuronally dependent migrating motor complexes predicts that fucosylated molecules could demonstrate anti-nociceptive activity as well. Large amplitude motor complexes appear to be essential for the perception of visceral pain [28], so that a reduction in amplitude as demonstrated by fucosylated HMO ought to moderate the nociceptive stimulus. The further exploration of the potential use of these particular HMO in conditions associated with disordered motility and gut pain, such as functional gut disorders and infantile colic, appears warranted.

The mechanism(s) whereby certain simple sugar components of breast milk may directly influence the migrating motor complexes is not known. “Bifidogenesis” is improbable because preparation of the colon segments removes most non-adherent contents. The rapid response (5-15 min) of gut musculature to exposure to the luminally perfused oligosaccharides suggests that the very limited number of existing bacteria still present in adherent mucus, are extremely unlikely to have been responsible for the motility effects. While we can rule out bifidogenesis we cannot rule out indirect effects from the few bacteria present in mucus. Furthermore, we have recently shown that a complex glycan, a bacterial exopolysaccharide, was neuronally active within seconds of placement into the lumen of an intact gut segment [17]. Again, this evidence does not rule out possible indirect effects through primary effects on epithelium or immune cells below the epithelium. However, both simple and complex glycans can influence the ENS almost immediately. While HMO do not appear to be absorbed across the intestinal epithelium intact in vivo, experiments with monolayers of epithelial cell lines have shown that both acidic and neutral HMO can be transported [29,30]. Neutral HMO are transported both transcellularly and via paracellular pathways [30], leaving open the possibility that the effects seen are indirectly mediated.

While neurons do express glycan receptors which do not bind fucose, such as galectins [31] and TLR [32-34], the presence of DC-SIGN which binds fucose and mannose with equal high affinity has not been reported. Free fucose has been shown to have a direct effect on differentiated Caco-2 epithelial cells, which appear unable to metabolize the sugar, although it promotes a TLR-2-like signaling pathway [35]. Alternatively, simple diffusion or transport of the oligosaccharides themselves across the epithelium may promote neuronal interaction since axonal terminals are known to be present in lateral intercellular spaces and immediately below the basal surfaces of the epithelium. Fucose itself has been described in the older literature as being highly immunoregulatory both in vivo and in vitro [36,37] and such actions have also been recorded for 2’FL (Sotgiu et al 2006) [38]. It is therefore plausible to postulate that it may be exerting similar direct effects on the enteric nervous system as it does on epithelial and immune cells.

The use of a mouse colon segment as a model for potential human application may be intuitively questioned. However, this assay has been validated as a potential screening method for pharmacological agents with known or potential therapeutic effects on human gut [20]. The authors’ conclusions are also reflected in the title of the paper: “The validation of an in vitro colonic motility assay as a biomarker for gastrointestinal adverse reactions”. The motor complexes which we record, are entirely neuronally dependent, since they are completely abolished by the specific sodium channel blocker and neurotoxin TTX (Figure S1).

Fucosylated molecules regulate the synapse function and development as well as neuronal morphology in primary hippocampal neuron culture [39]. They are also involved in cognitive aspects of brain function such as task-specific learning and long-term potentiation [40-42]. The concentrations of specific HMO in breast milk vary according to secretor status and time after parturition [43]. In the first 3 months of lactation the highest concentration of 2’FL in 12 donors approximated 3g/L [44] which thereafter declined to a mean of 1.2g/L. The concentrations of HMO which we have tested therefore fall into the likely physiological ranges occurring in the intestinal lumen in breast-fed infants. Given the possibility that small amounts of HMO may be transported or translocated intact across the intestinal epithelium, it is possible that in infancy, dietary fucosylated oligosaccharides and their degradation products such as fucose, may play an important role in the development and robust function of the central and enteric nervous systems.

Our results support further investigations of fucose, and fucosylated carbohydrates such as 2’FL and 3’FL as specific adjuncts to improve function of the enteric nervous system, and the preventative or therapeutic treatment of disorders involving gut nociception, contractility and motility. These suggestions are supported for 2’FL by our observations with video recordings showing decreased frequency, reduction of amplitude and velocity of colonic motor contractions. Since the effects of fucosylated oligosaccharides clearly occur through interactions, directly or indirectly with the ENS, we speculate that they could well also be exerting a positive effect on the brain via the vagus nerve [45,46] in supporting cognition and memory [47,48].

Supporting Information

Figure S1.

Lack of direct effect of 0.5 mg/mL 2’FL on colon muscle contractions when neurons are silenced. (A) Representative intraluminal pressure trace showing that addition of the specific neurotoxin, tetrodotoxin (TTX) at 0.3µM to the solution perfusing the gut segments rapidly abolished MMC pressure waves, leaving only contractile (ripples) that are entirely dependent on the musculature. Intraluminal 2’FL was applied after 15 minute recording with TTX and this had no effect on contractility in the absence of neural activity. (B) Summary statistics for 6 experiments showing that 2’FL had no statistically significant effect on the frequency with which ripples occurred.

https://doi.org/10.1371/journal.pone.0076236.s001

(TIFF)

Acknowledgments

The authors wish to thank Pedro A. Prieto, PhD for editorial review of the manuscript.

Author Contributions

Conceived and designed the experiments: JB WAK PF. Performed the experiments: AMS. Analyzed the data: WAK AMS. Contributed reagents/materials/analysis tools: RHB HL. Wrote the manuscript: JB WAK.

References

  1. 1. Bode L (2012) Human milk oligosaccharides: Every baby needs a sugar mama. Glycobiology 22: 1147-1162. doi:https://doi.org/10.1093/glycob/cws074. PubMed: 22513036.
  2. 2. Kobata A (2010) Structures and application of oligosaccharides in human milk. Proc Japan Academy B Phys Biol Sci 86: 731-747. doi:https://doi.org/10.2183/pjab.86.731. PubMed: 20689231.
  3. 3. Prieto PA (2005) In Vitro and Clinical Experiences with a Human Milk Oligosaccharide, Lacto-NneoTetraose, and Fructooligosaccharides. Foods Foods Ingredient J JAPAN 219: 1018-1030.
  4. 4. Zivkovic AM, Barile D (2011) Bovine milk as a source of functional oligosaccharides for improving human health. Advances Nutr 2: 284-289. doi:https://doi.org/10.3945/an.111.000455. PubMed: 22332060.
  5. 5. Sundekilde UK, Barile D, Meyrand M, Poulsen NA, Larsen LB et al. (2012) Natural variability in bovine milk oligosaccharides from Danish Jersey and Holstein-Friesian breeds. J Agric Food Chem 60: 6188-6196. doi:https://doi.org/10.1021/jf300015j. PubMed: 22632419.
  6. 6. Asakuma S, Hatakeyama E, Urashima T, Yoshida E, Katayama T et al. (2011) Physiology of consumption of human milk oligosaccharides by infant gut-associated bifidobacteria. J Biol Chem 286: 34583-34592. doi:https://doi.org/10.1074/jbc.M111.248138. PubMed: 21832085.
  7. 7. Koropatkin NM, Cameron EA, Martens EC (2012) How glycan metabolism shapes the human gut microbiota. Nat Rev Microbiol 10: 323-335. PubMed: 22491358.
  8. 8. Sela DA, Garrido D, Lerno L, Wu S, Tan K et al. (2012) Bifidobacterium longum subsp. infantis ATCC 15697 alpha-fucosidases are active on fucosylated human milk oligosaccharides. Appl Environ Microbiol 78: 795-803. doi:https://doi.org/10.1128/AEM.06762-11. PubMed: 22138995.
  9. 9. Chichlowski M, De Lartigue G, German JB, Raybould HE, Mills DA (2012) Bifidobacteria isolated from infants and cultured on human milk oligosaccharides affect intestinal epithelial function. J Pediatr Gastroenterol Nutr 55: 321-327. doi:https://doi.org/10.1097/MPG.0b013e31824fb899. PubMed: 22383026.
  10. 10. Gibson GR, Roberfroid MB (1995) Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics. J Nutr 125: 1401-1412. PubMed: 7782892.
  11. 11. Kuntz S, Rudloff S, Kunz C (2008) Oligosaccharides from human milk influence growth-related characteristics of intestinally transformed and non-transformed intestinal cells. Br J Nutr 99: 462-471. PubMed: 17925055.
  12. 12. Kuntz S, Kunz C, Rudloff S (2009) Oligosaccharides from human milk induce growth arrest via G2/M by influencing growth-related cell cycle genes in intestinal epithelial cells. Br J Nutr 101: 1306-1315. doi:https://doi.org/10.1017/S0007114508079622. PubMed: 19079835.
  13. 13. Ruiz-Palacios GM, Cervantes LE, Ramos P, Chavez-Munguia B, Newburg DS (2003) Campylobacter jejuni binds intestinal H(O) antigen (Fuc alpha 1, 2Gal beta 1, 4GlcNAc), and fucosyloligosaccharides of human milk inhibit its binding and infection. J Biol Chem 278: 14112-14120. doi:https://doi.org/10.1074/jbc.M207744200. PubMed: 12562767.
  14. 14. Kuhn L, Aldrovandi GM, Sinkala M, Kankasa C, Semrau K et al. (2008) Effects of early, abrupt weaning on HIV-free survival of children in Zambia. N Engl J Med 359: 130-141. doi:https://doi.org/10.1056/NEJMoa073788. PubMed: 18525036.
  15. 15. Jantscher-Krenn E, Lauwaet T, Bliss LA, Reed SL, Gillin FD et al. (2012) Human milk oligosaccharides reduce Entamoeba histolytica attachment and cytotoxicity in vitro. Br J Nutr, 108: 1-8. PubMed: 22264879.
  16. 16. Wang B, Mao YK, Diorio C, Pasyk M, Wu RY et al. (2010) Luminal administration ex vivo of a live Lactobacillus species moderates mouse jejunal motility within minutes. FASEB J 24: 4078-4088. doi:https://doi.org/10.1096/fj.09-153841. PubMed: 20519636.
  17. 17. Mao YK, Wang B, Forsythe P, Bienenstock J, Kunze WA (2013) Bacteroides fragilis polysaccharide A is necessary and sufficeient for acute activation oif intestinal sensory neurons. Nature communications In Press.
  18. 18. Wu RY, Pasyk M, Wang B, Forsythe P, Bienenstock J et al. (2013) Spatiotemporal maps reveal regional differences in the effects on gut motility for Lactobacillus reuteri and rhamnosus strains. Neurogastroenterol Motil 25: e205-e214. doi:https://doi.org/10.1111/nmo.12072. PubMed: 23316914.
  19. 19. Mao YK, Kasper DL, Wang B, Forsythe P, Bienenstock J et al. (2013) Bacteroides fragilis polysaccharide A is necessary and sufficient for acute activation of intestinal sensory neurons. Nat Commun 4: 1465. doi:https://doi.org/10.1038/ncomms2478. PubMed: 23403566.
  20. 20. Keating C, Martinez V, Ewart L, Gibbons S, Grundy L et al. (2010) The validation of an in vitro colonic motility assay as a biomarker for gastrointestinal adverse drug reactions. Toxicol Appl Pharmacol 245: 299-309. doi:https://doi.org/10.1016/j.taap.2010.03.014. PubMed: 20350559.
  21. 21. de Kivit S, Kraneveld AD, Garssen J, Willemsen LE (2011) Glycan recognition at the interface of the intestinal immune system: target for immune modulation via dietary components. Eur J Pharmacol 668 Suppl 1: S124-S132. doi:https://doi.org/10.1016/j.ejphar.2011.05.086. PubMed: 21816141.
  22. 22. Rudloff S, Pohlentz G, Borsch C, Lentze MJ, Kunz C (2012) Urinary excretion of in vivo (1)(3)C-labelled milk oligosaccharides in breastfed infants. Br J Nutr 107: 957-963. doi:https://doi.org/10.1017/S0007114511004016. PubMed: 21888740.
  23. 23. Rudloff S, Pohlentz G, Diekmann L, Egge H, Kunz C (1996) Urinary excretion of lactose and oligosaccharides in preterm infants fed human milk or infant formula. Acta Paediatr 85: 598-603. doi:https://doi.org/10.1111/j.1651-2227.1996.tb14095.x. PubMed: 8827106.
  24. 24. Coppa GV, Pierani P, Zampini L, Bruni S, Carloni I et al. (2001) Characterization of oligosaccharides in milk and feces of breast-fed infants by high-performance anion-exchange chromatography. Adv Exp Med Biol 501: 307-314. doi:https://doi.org/10.1007/978-1-4615-1371-1_38. PubMed: 11787695.
  25. 25. Albrecht S, Schols HA, van den Heuvel EG, Voragen AG, Gruppen H (2011) Occurrence of oligosaccharides in feces of breast-fed babies in their first six months of life and the corresponding breast milk. Carbohydr Res 346: 2540-2550. doi:https://doi.org/10.1016/j.carres.2011.08.009. PubMed: 21962590.
  26. 26. Albrecht S, Schols HA, van Zoeren D, van Lingen RA, Groot Jebbink LJ et al. (2011) Oligosaccharides in feces of breast- and formula-fed babies. Carbohydr Res 346: 2173-2181. doi:https://doi.org/10.1016/j.carres.2011.06.034. PubMed: 21782161.
  27. 27. Prieto PA, Mukerji P, Kelder B, Erney R, Gonzalez D et al. (1995) Remodeling of mouse milk glycoconjugates by transgenic expression of a human glycosyltransferase. J Biol Chem 270: 29515-29519. doi:https://doi.org/10.1074/jbc.270.49.29515. PubMed: 7493992.
  28. 28. Sarna SK (2007) Enteric descending and afferent neural signaling stimulated by giant migrating contractions: essential contributing factors to visceral pain. Am J Physiol Gastrointest Liver Physiol 292: G572-G581. PubMed: 16990445.
  29. 29. Eiwegger T, Stahl B, Haidl P, Schmitt J, Boehm G et al. (2010) Prebiotic oligosaccharides: in vitro evidence for gastrointestinal epithelial transfer and immunomodulatory properties. Pediatr Allergy Immunol 21: 1179-1188. doi:https://doi.org/10.1111/j.1399-3038.2010.01062.x. PubMed: 20444147.
  30. 30. Gnoth MJ, Rudloff S, Kunz C, Kinne RK (2001) Investigations of the in vitro transport of human milk oligosaccharides by a Caco-2 monolayer using a novel high performance liquid chromatography-mass spectrometry technique. J Biol Chem 276: 34363-34370. doi:https://doi.org/10.1074/jbc.M104805200. PubMed: 11423546.
  31. 31. Gensel JC, Kigerl KA, Mandrekar-Colucci SS, Gaudet AD, Popovich PG (2012) Achieving CNS axon regeneration by manipulating convergent neuro-immune signaling. Cell Tissue Res 349: 201-213. doi:https://doi.org/10.1007/s00441-012-1425-5. PubMed: 22592625.
  32. 32. Okun E, Griffioen KJ, Mattson MP (2011) Toll-like receptor signaling in neural plasticity and disease. Trends Neurosci 34: 269-281. doi:https://doi.org/10.1016/j.tins.2011.02.005. PubMed: 21419501.
  33. 33. Liu T, Gao YJ, Ji RR (2012) Emerging role of Toll-like receptors in the control of pain and itch. Neurosci Bull 28: 131-144. doi:https://doi.org/10.1007/s12264-012-1219-5. PubMed: 22466124.
  34. 34. Côté M, Drouin-Ouellet J, Cicchetti F, Soulet D (2011) The critical role of the MyD88-dependent pathway in non-CNS MPTP-mediated toxicity. Brain Behav Immun 25: 1143-1152. doi:https://doi.org/10.1016/j.bbi.2011.02.017. PubMed: 21376805.
  35. 35. Chow WL, Lee YK (2008) Free fucose is a danger signal to human intestinal epithelial cells. Br J Nutr 99: 449-454. PubMed: 17697405.
  36. 36. Baba T, Yoshida T, Cohen S (1979) Suppression of cell-mediated immune reactions by monosaccharides. J Immunol 122: 838-841. PubMed: 156214.
  37. 37. Rocklin RE (1976) Role of monosaccharides in the interaction of two lymphocyte mediators with their target cells. J Immunol 116: 816-820. PubMed: 768376.
  38. 38. Sotgiu S, Arru G, Fois ML, Sanna A, Musumeci M et al. (2006) Immunomodulation of fucosyl-lactose and lacto-N-fucopentaose on mononuclear cells from multiple sclerosis and healthy subjects. Int J Biomed Sci 2: 114-120. PubMed: 23674973.
  39. 39. Murrey HE, Gama CI, Kalovidouris SA, Luo WI, Driggers EM et al. (2006) Protein fucosylation regulates synapsin Ia/Ib expression and neuronal morphology in primary hippocampal neurons. Proc Natl Acad Sci U S A 103: 21-26. doi:https://doi.org/10.1073/pnas.0503381102. PubMed: 16373512.
  40. 40. McCabe NR, Rose SP (1985) Passive avoidance training increases fucose incorporation into glycoproteins in chick forebrain slices in vitro. Neurochem Res 10: 1083-1095. doi:https://doi.org/10.1007/BF00965883. PubMed: 4058654.
  41. 41. Pohle W, Acosta L, Rüthrich H, Krug M, Matthies H (1987) Incorporation of [3H]fucose in rat hippocampal structures after conditioning by perforant path stimulation and after LTP-producing tetanization. Brain Res 410: 245-256. doi:https://doi.org/10.1016/0006-8993(87)90321-0. PubMed: 3594237.
  42. 42. Matthies H, Staak S, Krug M (1996) Fucose and fucosyllactose enhance in-vitro hippocampal long-term potentiation. Brain Res 725: 276-280. doi:https://doi.org/10.1016/S0006-8993(96)00406-4. PubMed: 8836537.
  43. 43. Coppa GV, Gabrielli O, Pierani P, Catassi C, Carlucci A et al. (1993) Changes in carbohydrate composition in human milk over 4 months of lactation. Pediatrics 91: 637-641. PubMed: 8441573.
  44. 44. Chaturvedi P, Warren CD, Altaye M, Morrow AL, Ruiz-Palacios G et al. (2001) Fucosylated human milk oligosaccharides vary between individuals and over the course of lactation. Glycobiology 11: 365-372. doi:https://doi.org/10.1093/glycob/11.5.365. PubMed: 11425797.
  45. 45. Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM et al. (2011) Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A 108: 16050-16055. doi:https://doi.org/10.1073/pnas.1102999108. PubMed: 21876150.
  46. 46. Bonaz B, Picq C, Sinniger V, Mayol JF, Clarençon D (2013) Vagus nerve stimulation: from epilepsy to the cholinergic anti-inflammatory pathway. Neurogastroenterol Motil 25: 208-221. doi:https://doi.org/10.1111/nmo.12076. PubMed: 23360102.
  47. 47. Matthies H, Schroeder H, Smalla KH, Krug M (2000) Enhancement of glutamate release by L-fucose changes effects of glutamate receptor antagonists on long-term potentiation in the rat hippocampus. Learn Mem 7: 227-234. doi:https://doi.org/10.1101/lm.7.4.227. PubMed: 10940323.
  48. 48. Nelson ED, Ramberg JE, Best T, Sinnott RA (2012) Neurologic effects of exogenous saccharides: a review of controlled human, animal, and in vitro studies. Nutr Neurosci 15: 149-162. doi:https://doi.org/10.1179/1476830512Y.0000000004. PubMed: 22417773.