Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Front Nutr ; 11: 1351433, 2024.
Article in English | MEDLINE | ID: mdl-38389793

ABSTRACT

Scope: 2´-Fucosyllactose (2´-FL), the most abundant oligosaccharide in human milk, plays an important role in numerous biological functions, including improved learning. It is not clear, however, whether 2´-FL or a cleavage product could influence neuronal cell activity. Thus, we investigated the effects of 2´-FL, its monosaccharide fucose (Fuc), and microbial fermented 2´-FL and Fuc on the parameters of neuronal cell activity in an intestinal-neuronal transwell co-culture system in vitro. Methods: Native 13C-labeled 2´-FL and 13C-Fuc or their metabolites, fermented with Bifidobacterium (B.) longum ssp. infantis and B. breve, which were taken from the lag-, log- and stationary (stat-) growth phases of batch cultures, were applied to the apical compartment of the co-culture system with Caco-2 cells representing the intestinal layer and all-trans-retinoic acid-differentiated SH-SY5Y (SH-SY5YATRA) cells mimicking neuronal-like cells. After 3 h of incubation, the culture medium in the basal compartment was monitored for 13C enrichment by using elemental analysis isotope-ratio mass spectrometry (EA-IRMS) and effects on cell viability, plasma, and mitochondrial membrane potential. The neurotransmitter activation (BDNF, GABA, choline, and glutamate) of SH-SY5YATRA cells was also determined. Furthermore, these effects were also measured by the direct application of 13C-2´-FL and 13C-Fuc to SH-SY5YATRA cells. Results: While no effects on neuronal-like cell activities were observed after intact 2´-FL or Fuc was incubated with SH-SY5YATRA cells, supernatants from the stat-growth phase of 2´-FL, fermented by B. longum ssp. infantis alone and together with B. breve, significantly induced BDNF release from SH-SY5YATRA cells. No such effects were found for 2´-FL, Fuc, or their fermentation products from B. breve. The BDNF release occurred from an enhanced vesicular release, which was confirmed by the use of the Ca2+-channel blocker verapamil. Concomitant with this event, 13C enrichment was also observed in the basal compartment when supernatants from the stat-growth phase of fermentation by B. longum ssp. infantis alone or together with B. breve were used. Conclusion: The results obtained in this study suggest that microbial products of 2´-FL rather than the oligosaccharide itself may influence neuronal cell activities.

2.
J Immunol Res ; 2023: 9603576, 2023.
Article in English | MEDLINE | ID: mdl-37545544

ABSTRACT

Background: Studies suggest that early-life gut microbiota composition and intestinal short-chain fatty acids (SCFAs) are linked to future asthma susceptibility. Furthermore, infancy offers a critical time window to modulate the microbiota and associated metabolites through diet-microbe interactions to promote infant health. Human milk oligosaccharides (HMOs), nondigestible carbohydrates abundant in breast milk, are prebiotics selectively metabolized by gut microbiota that consequently modify microbiome composition and SCFA production. Methods: Using a house dust mite mouse model of allergy, we investigated the impacts of early oral treatment of pups with biologically relevant doses of 2'-fucosyllactose (2'-FL) and 6'-sialyllactose (6'-SL), two of the most abundant HMOs in human milk, in amelioration of allergic airway disease severity. Results: We found that administration of 2'-FL and 6'-SL during early life reduced lung histopathology scores, circulating IgE, cytokine levels, and inflammatory cell infiltration, all hallmark symptoms of allergic asthma. HMO supplementation also increased the relative abundance of intestinal Bacteroidetes and Clostridia, known SCFA producers within the gut. Indeed, we detected increased SCFA concentrations in both the intestine and blood of adult mice who received HMOs prior to weaning. Conclusion: We propose a model in which orally administered HMOs delivered during early life shift the microbiota toward increased production of SCFAs, which dampens the allergic immune responses behind allergy and asthma. Overall, these data suggest the potential for HMO supplementation to protect infants against asthma development later in life, with possible benefits against additional atopic diseases such as eczema and food allergies.


Subject(s)
Asthma , Food Hypersensitivity , Gastrointestinal Microbiome , Humans , Infant , Female , Animals , Mice , Milk, Human/metabolism , Oligosaccharides/metabolism , Asthma/metabolism , Gastrointestinal Microbiome/physiology , Fatty Acids, Volatile/metabolism
3.
Nutrients ; 15(10)2023 May 17.
Article in English | MEDLINE | ID: mdl-37242222

ABSTRACT

Human milk represents an optimal source of nutrition during infancy. Milk also serves as a vehicle for the transfer of growth factors, commensal microbes, and prebiotic compounds to the immature gastrointestinal tract. These immunomodulatory and prebiotic functions of milk are increasingly appreciated as critical factors in the development of the infant gut and its associated microbial community. Advances in infant formula composition have sought to recapitulate some of the prebiotic and immunomodulatory functions of milk through human milk oligosaccharide (HMO) fortification, with the aim of promoting healthy development both within the gastrointestinal tract and systemically. Our objective was to investigate the effects of feeding formulas supplemented with the HMO 2'-fucosyllactose (2'-FL) on serum metabolite levels relative to breastfed infants. A prospective, randomized, double-blinded, controlled study of infant formulas (64.3 kcal/dL) fortified with varying levels of 2'-FL and galactooligosaccharides (GOS) was conducted [0.2 g/L 2'-FL + 2.2 g/L GOS; 1.0 g/L 2'-FL + 1.4 g/L GOS]. Healthy singleton infants age 0-5 days and with birth weight > 2490 g were enrolled (n = 201). Mothers chose to either exclusively formula-feed or breastfeed their infant from birth to 4 months of age. Blood samples were drawn from a subset of infants at 6 weeks of age (n = 35-40 per group). Plasma was evaluated by global metabolic profiling and compared to a breastfed reference group (HM) and a control formula (2.4 g/L GOS). Fortification of control infant formula with the HMO 2'-FL resulted in significant increases in serum metabolites derived from microbial activity in the gastrointestinal tract. Most notably, secondary bile acid production was broadly increased in a dose-dependent manner among infants receiving 2'-FL supplemented formula relative to the control formula. 2'-FL supplementation increased secondary bile acid production to levels associated with breastfeeding. Our data indicate that supplementation of infant formula with 2'-FL supports the production of secondary microbial metabolites at levels comparable to breastfed infants. Thus, dietary supplementation of HMO may have broad implications for the function of the gut microbiome in systemic metabolism. This trial was registered at with the U.S. National library of Medicine as NCT01808105.


Subject(s)
Microbiota , Milk, Human , Female , Humans , Infant , Infant, Newborn , Child, Preschool , Milk, Human/chemistry , Prospective Studies , Dietary Supplements , Infant Formula , Breast Feeding , Prebiotics/analysis , Oligosaccharides/pharmacology
4.
Am J Physiol Gastrointest Liver Physiol ; 325(1): G23-G41, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37120853

ABSTRACT

Necrotizing enterocolitis (NEC) is the leading cause of morbidity and mortality in premature infants. One of the most devastating complications of NEC is the development of NEC-induced brain injury, which manifests as impaired cognition that persists beyond infancy and which represents a proinflammatory activation of the gut-brain axis. Given that oral administration of the human milk oligosaccharides (HMOs) 2'-fucosyllactose (2'-FL) and 6'-sialyslactose (6'-SL) significantly reduced intestinal inflammation in mice, we hypothesized that oral administration of these HMOs would reduce NEC-induced brain injury and sought to determine the mechanisms involved. We now show that the administration of either 2'-FL or 6'-SL significantly attenuated NEC-induced brain injury, reversed myelin loss in the corpus callosum and midbrain of newborn mice, and prevented the impaired cognition observed in mice with NEC-induced brain injury. In seeking to define the mechanisms involved, 2'-FL or 6'-SL administration resulted in a restoration of the blood-brain barrier in newborn mice and also had a direct anti-inflammatory effect on the brain as revealed through the study of brain organoids. Metabolites of 2'-FL were detected in the infant mouse brain by nuclear magnetic resonance (NMR), whereas intact 2'-FL was not. Strikingly, the beneficial effects of 2'-FL or 6'-SL against NEC-induced brain injury required the release of the neurotrophic factor brain-derived neurotrophic factor (BDNF), as mice lacking BDNF were not protected by these HMOs from the development of NEC-induced brain injury. Taken in aggregate, these findings reveal that the HMOs 2'-FL and 6'-SL interrupt the gut-brain inflammatory axis and reduce the risk of NEC-induced brain injury.NEW & NOTEWORTHY This study reveals that the administration of human milk oligosaccharides, which are present in human breast milk, can interfere with the proinflammatory gut-brain axis and prevent neuroinflammation in the setting of necrotizing enterocolitis, a major intestinal disorder seen in premature infants.


Subject(s)
Brain Injuries , Cognitive Dysfunction , Enterocolitis, Necrotizing , Humans , Infant, Newborn , Infant , Female , Animals , Mice , Milk, Human/metabolism , Brain-Derived Neurotrophic Factor , Neuroinflammatory Diseases , Enterocolitis, Necrotizing/etiology , Oligosaccharides/pharmacology , Oligosaccharides/therapeutic use , Oligosaccharides/analysis , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/complications , Brain Injuries/complications , Brain Injuries/metabolism
5.
Infect Immun ; 91(2): e0057022, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36692308

ABSTRACT

A disrupted "dysbiotic" gut microbiome engenders susceptibility to the diarrheal pathogen Clostridioides difficile by impacting the metabolic milieu of the gut. Diet, in particular the microbiota-accessible carbohydrates (MACs) found in dietary fiber, is one of the most powerful ways to affect the composition and metabolic output of the gut microbiome. As such, diet is a powerful tool for understanding the biology of C. difficile and for developing alternative approaches for coping with this pathogen. One prominent class of metabolites produced by the gut microbiome is short-chain fatty acids (SCFAs), the major metabolic end products of MAC metabolism. SCFAs are known to decrease the fitness of C. difficile in vitro, and high intestinal SCFA concentrations are associated with reduced fitness of C. difficile in animal models of C. difficile infection (CDI). Here, we use controlled dietary conditions (8 diets that differ only by MAC composition) to show that C. difficile fitness is most consistently impacted by butyrate, rather than the other two prominent SCFAs (acetate and propionate), during murine model CDI. We similarly show that butyrate concentrations are lower in fecal samples from humans with CDI than in those from healthy controls. Finally, we demonstrate that butyrate impacts growth in diverse C. difficile isolates. These findings provide a foundation for future work which will dissect how butyrate directly impacts C. difficile fitness and will lead to the development of diverse approaches distinct from antibiotics or fecal transplant, such as dietary interventions, for mitigating CDI in at-risk human populations. IMPORTANCE Clostridioides difficile is a leading cause of infectious diarrhea in humans, and it imposes a tremendous burden on the health care system. Current treatments for C. difficile infection (CDI) include antibiotics and fecal microbiota transplant, which contribute to recurrent CDIs and face major regulatory hurdles, respectively. Therefore, there is an ongoing need to develop new ways to cope with CDI. Notably, a disrupted "dysbiotic" gut microbiota is the primary risk factor for CDI, but we incompletely understand how a healthy microbiota resists CDI. Here, we show that a specific molecule produced by the gut microbiota, butyrate, is negatively associated with C. difficile burdens in humans and in a mouse model of CDI and that butyrate impedes the growth of diverse C. difficile strains in pure culture. These findings help to build a foundation for designing alternative, possibly diet-based, strategies for mitigating CDI in humans.


Subject(s)
Clostridioides difficile , Clostridium Infections , Humans , Animals , Mice , Butyrates , Permissiveness , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Fatty Acids, Volatile
6.
Nutrients ; 14(13)2022 Jun 24.
Article in English | MEDLINE | ID: mdl-35807803

ABSTRACT

Background: Five of the most abundant human milk oligosaccharides (HMOs) in human milk are 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3-FL), lacto-N-tetraose (LNT), 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL). Methods: A randomized, double-blind, controlled parallel feeding trial evaluated growth in healthy term infants fed a control milk-based formula (CF; n = 129), experimental milk-based formula (EF; n = 130) containing five HMOs (5.75 g/L; 2'-FL, 3-FL, LNT, 3'-SL and 6'-SL) or human milk (HM; n = 104). Results: No significant differences (all p ≥ 0.337, protocol evaluable cohort) were observed among the three groups for weight gain per day from 14 to 119 days (D) of age, irrespective of COVID-19 or combined non-COVID-19 and COVID-19 periods. There were no differences (p ≥ 0.05) among the three groups for gains in weight and length from D14 to D119. Compared to the CF group, the EF group had more stools that were soft, frequent and yellow and were similar to the HM group. Serious and non-serious adverse events were not different among groups, but more CF-fed infants were seen by health care professionals for illness from study entry to D56 (p = 0.044) and D84 (p = 0.028) compared to EF-fed infants. Conclusions: The study demonstrated that the EF containing five HMOs supported normal growth, gastrointestinal (GI) tolerance and safe use in healthy term infants.


Subject(s)
COVID-19 , Infant Formula , Dietary Supplements , Humans , Infant , Milk, Human , Oligosaccharides
7.
Nutrients ; 13(10)2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34684364

ABSTRACT

Breastfeeding is the best source of nutrition during infancy and is associated with a broad range of health benefits. However, there remains a significant and persistent need for innovations in infant formula that will allow infants to access a wider spectrum of benefits available to breastfed infants. The addition of human milk oligosaccharides (HMOs) to infant formulas represents the most significant innovation in infant nutrition in recent years. Although not a direct source of calories in milk, HMOs serve as potent prebiotics, versatile anti-infective agents, and key support for neurocognitive development. Continuing improvements in food science will facilitate production of a wide range of HMO structures in the years to come. In this review, we evaluate the relationship between HMO structure and functional benefits. We propose that infant formula fortification strategies should aim to recapitulate a broad range of benefits to support digestive health, immunity, and cognitive development associated with HMOs in breastmilk. We conclude that acetylated, fucosylated, and sialylated HMOs likely confer important health benefits through multiple complementary mechanisms of action.


Subject(s)
Infant Health , Milk, Human/chemistry , Oligosaccharides/metabolism , Cognition , Humans , Immunomodulation , Infant , Nutrients/analysis , Oligosaccharides/chemistry
8.
Mol Nutr Food Res ; 65(16): e2100045, 2021 08.
Article in English | MEDLINE | ID: mdl-34139057

ABSTRACT

SCOPE: To further examine the role of the human milk oligosaccharide 2'fucosyllactose (2´FL) and fucose (Fuc) in cognition. Using 13 C-labeled 2'FL,thestudy previously showed in mice that 13 C-enrichment of the brain is not caused by 13 C1 -2´FL itself, but rather by microbial metabolites. Here, the study applies 13 C1 -Fuc in the same mouse model to investigate its uptake into the brain. METHODS AND RESULTS: Mice received 13 C1 -Fuc via oral gavage (2 mmol 13 C1 -Fuc/kg-1 body weight) or intravenously (0.4 mmol/kg-1 body weight). 13 C-enrichment is measured in organs, including various brain regions, biological fluids and excrements. By EA-IRMS, the study observes an early rise of 13 C-enrichment in plasma, 30 min after oral dosing. However, 13 C-enrichment in the brain does not occur until 3-5 h post-dosing, when the 13 C-Fuc bolus has already reached the lower gut. Therefore, the researcher assume that 13 C-Fuc is absorbed in the upper small intestine but cannot cross the blood-brain barrier which is also observed after intravenous application of 13 C1 -Fuc. CONCLUSIONS: Late 13 C-enrichment in the rodent brain may be derived from 13 C1 -Fuc metabolites derived from bacterial fermentation. The precise role that Fuc or 2´FL metabolites might play in gut-brain communication needs to be investigated in further studies.


Subject(s)
Blood-Brain Barrier , Brain/metabolism , Fucose/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Brain-Gut Axis , Intestine, Small/metabolism , Male , Mice
10.
Pediatr Res ; 89(1): 91-101, 2021 01.
Article in English | MEDLINE | ID: mdl-32221473

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) develops through exaggerated toll-like receptor 4 (TLR4) signaling in the intestinal epithelium. Breast milk is rich in non-digestible oligosaccharides and prevents NEC through unclear mechanisms. We now hypothesize that the human milk oligosaccharides 2'-fucosyllactose (2'-FL) and 6'-sialyllactose (6'-SL) can reduce NEC through inhibition of TLR4 signaling. METHODS: NEC was induced in newborn mice and premature piglets and infant formula was supplemented with 2'-FL, 6'-SL, or lactose. Intestinal tissue was obtained at surgical resection. HMO inhibition of TLR4 was assessed in IEC-6 enterocytes, mice, and human tissue explants and via in silico modeling. RESULTS: Supplementation of infant formula with either 2'-FL and/or 6'-SL, but not the parent sugar lactose, reduced NEC in mice and piglets via reduced apoptosis, inflammation, weight loss, and histological appearance. Mechanistically, both 2'-FL and 6'-SL, but not lactose, reduced TLR4-mediated nuclear factor kappa light-chain enhancer of activated B cells (NF-kB) inflammatory signaling in the mouse and human intestine. Strikingly, in silico modeling revealed 2'-FL and 6'-SL, but not lactose, to dock into the binding pocket of the TLR4-MD2 complex, explaining their ability to inhibit TLR4 signaling. CONCLUSIONS: 2'-FL and 6'-SL, but not lactose, prevent NEC in mice and piglet models and attenuate NEC inflammation in the human ileum, in part through TLR4 inhibition. IMPACT: Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants that occurs in the setting of bacterial colonization of the gut and administration of formula feeds and activation by the innate immune receptor toll-like receptor 4 (TLR4). Breast milk prevents NEC through unclear mechanisms. We now show that breast milk-enriched human milk oligosaccharides (HMOs) that are derived from lactose prevent NEC through inhibition of TLR4. The human milk oligosaccharides 2'-FL and 6'-SL, but not the backbone sugar lactose, prevent NEC in mice and piglets. 2'-FL and 6'-SL but not lactose inhibited TLR4 signaling in cultured enterocytes, in enteroids derived from mouse intestine, and in human intestinal explants obtained at the time of surgical resection for patients with NEC. In seeking the mechanisms involved, 2'-FL and 6'-SL but not lactose were found to directly bind to TLR4, explaining the inhibition and protection against NEC. These findings may impact clinical practice by suggesting that administration of HMOs could serve as a preventive strategy for premature infants at risk for NEC development.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Ileum/drug effects , Intestinal Mucosa/drug effects , Lactose/analogs & derivatives , Milk, Human/chemistry , Toll-Like Receptor 4/antagonists & inhibitors , Trisaccharides/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Humans , Ileum/immunology , Ileum/metabolism , Ileum/pathology , Inflammation Mediators/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lactose/isolation & purification , Lactose/pharmacology , Mice , Molecular Docking Simulation , Signal Transduction , Sus scrofa , Toll-Like Receptor 4/metabolism , Trisaccharides/isolation & purification , Weight Loss/drug effects
11.
Cell Mol Gastroenterol Hepatol ; 9(3): 403-423, 2020.
Article in English | MEDLINE | ID: mdl-31756560

ABSTRACT

BACKGROUND & AIMS: Necrotizing enterocolitis (NEC) is a devastating disease of premature infants characterized by Toll-like receptor 4 (TLR4)-dependent intestinal inflammation and enterocyte death. Given that necroptosis is a proinflammatory cell death process that is linked to bacterial signaling, we investigated its potential role in NEC, and the mechanisms involved. METHODS: Human and mouse NEC intestine were analyzed for necroptosis gene expression (ie, RIPK1, RIPK3, and MLKL), and protein activation (phosphorylated RIPK3). To evaluate a potential role for necroptosis in NEC, the effects of genetic (ie, Ripk3 knockout or Mlkl knockout) or pharmacologic (ie, Nec1s) inhibition of intestinal inflammation were assessed in a mouse NEC model, and a possible upstream role of TLR4 was assessed in Tlr4-deficient mice. The NEC-protective effects of human breast milk and its constituent milk oligosaccharides on necroptosis were assessed in a NEC-in-a-dish model, in which mouse intestinal organoids were cultured as either undifferentiated or differentiated epithelium in the presence of NEC bacteria and hypoxia. RESULTS: Necroptosis was activated in the intestines of human and mouse NEC in a TLR4-dependent manner, and was up-regulated specifically in differentiated epithelium of the immature ileum. Inhibition of necroptosis genetically and pharmacologically reduced intestinal-epithelial cell death and mucosal inflammation in experimental NEC, and ex vivo in the NEC-in-a-dish system. Strikingly, the addition of human breast milk, or the human milk oligosaccharide 2 fucosyllactose in the ex vivo system, reduced necroptosis and inflammation. CONCLUSIONS: Necroptosis is activated in the intestinal epithelium upon TLR4 signaling and is required for NEC development, and explains in part the protective effects of breast milk.


Subject(s)
Enterocolitis, Necrotizing/pathology , Enterocytes/pathology , Intestinal Mucosa/pathology , Milk, Human/chemistry , Necroptosis/immunology , Animals , Disease Models, Animal , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/immunology , Enterocytes/drug effects , Enterocytes/immunology , Female , Humans , Infant, Newborn , Intestinal Mucosa/drug effects , Mice , Mice, Knockout , Necroptosis/drug effects , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Trisaccharides/pharmacology , Trisaccharides/therapeutic use , Up-Regulation
12.
Mol Nutr Food Res ; 63(13): e1900035, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31125176

ABSTRACT

SCOPE: 2´-Fucosyllactose (2´FL) is an abundant oligosaccharide in human milk. It is hypothesized that its brain enrichment is associated with improved learning. Accumulation of 2´FL in organs, biological fluids, and feces is assessed in wild-type and germ-free mice. METHODS AND RESULTS: 13 C-labelled 2´FL is applied to NMRI wild-type mice intravenously (0.2 g kg-1 ) or orally (1 g kg-1 ), while controls receive saline. Biological samples are collected (0.5-15 h) and 13 C-enrichment is measured by elemental analysis isotope ratio mass spectrometry (EA-IRMS). After oral application, 2´FL is primarily eliminated in the feces. 13 C-enrichment in organs including the brain follows the same pattern as in plasma with a maximum peak after 5 h. However, 13 C-enrichment is only detected when the 13 C-2´FL bolus reaches the colon. In contrast, in germ-free mice, the 13 C-bolus remains in the intestinal content and is expelled via the feces. Furthermore, intravenously applied 13 C-2´FL is eliminated via urine; no 13 C-enrichment of organs is observed, suggesting that intact 2´FL is not retained. CONCLUSIONS: 13 C-enrichment in brain and other organs after oral application of 13 C-2´FL in wild-type mice indicates cleaved fucose or other gut microbial 2´FL metabolites may be incorporated, as opposed to intact 2´FL.

13.
PLoS One ; 14(4): e0215151, 2019.
Article in English | MEDLINE | ID: mdl-31017915

ABSTRACT

BACKGROUND: Stress causes severe dysmotility in the mammalian gut. Almost all research done to date has concentrated on prevention of stress-induced altered gut motility but not on treatment. We had previously shown that intraluminal 2'FL could acutely moderate propulsive motility in isolated mouse colonic segments. Because 2'FL appeared to modulate enteric nervous system dependent motility, we wondered if the oligosaccharide could reverse the effects of prior restraint stress, ex vivo. We tested whether 2'FL could benefit the dysmotility of isolated jejunal and colonic segments from animals subjected to prior acute restraint stress. METHODS: Jejunal and colonic segments were obtained from male Swiss Webster mice that were untreated or subjected to 1 hour of acute restraint stress. Segments were perfused with Krebs buffer and propagating contractile clusters (PCC) digitally video recorded. 2'FL or ß-lactose were added to the perfusate at a concentration of 1 mg/ml. Spatiotemporal maps were constructed from paired before and after treatment recordings, each consisting of 20 min duration and PCC analyzed for frequency, velocity and amplitude. KEY RESULTS: Stress decreased propulsive motility in murine small intestine while increasing it in the colon. 2'FL in jejunum of previously stressed mice produced a 50% increase in PCC velocity (p = 0.0001), a 43% increase in frequency (p = 0.0002) and an insignificant decrease in peak amplitude. For stressed colon, 2'FL reduced the frequency by 23% (p = 0.017) and peak amplitude by 26% (p = 0.011), and was without effect on velocity. ß-lactose had negligible or small treatment effects. CONCLUSIONS & INFERENCES: We show that the prebiotic 2'FL may have potential as a treatment for acute stress-induced gut dysmotility, ex vivo, and that, as is the case for certain beneficial microbes, the mechanism occurs in the gut, likely via action on the enteric nervous system.


Subject(s)
Feces/chemistry , Gastrointestinal Motility/drug effects , Stress, Psychological , Trisaccharides/pharmacology , Animals , Male , Mice , Restraint, Physical
14.
Nutrients ; 10(10)2018 Oct 16.
Article in English | MEDLINE | ID: mdl-30332832

ABSTRACT

Sialic acids (Sia) are postulated to improve cognitive abilities. This study evaluated Sia effects on rat behavior when administered in a free form as N-acetylneuraminic acid (Neu5Ac) or conjugated as 6'-sialyllactose (6'-SL). Rat milk contains Sia, which peaks at Postnatal Day 9 and drops to a minimum by Day 15. To bypass this Sia peak, a cohort of foster mothers was used to raise the experimental pups. A group of pups received a daily oral supplementation of Neu5Ac to mimic the amount naturally present in rat milk, and another group received the same molar amount of Sia as 6'-SL. The control group received water. After weaning, rats were submitted to behavioral evaluation. One year later, behavior was re-evaluated, and in vivo long-term potentiation (LTP) was performed. Brain samples were collected and analyzed at both ages. Adult rats who received Sia performed significantly better in the behavioral assessment and showed an enhanced LTP compared to controls. Within Sia groups, 6'-SL rats showed better scores in some cognitive outcomes compared to Neu5Ac rats. At weaning, an effect on polysialylated-neural cell adhesion molecule (PSA-NCAM) levels in the frontal cortex was only observed in 6'-SL fed rats. Providing Sia during lactation, especially as 6'-SL, improves memory and LTP in adult rats.


Subject(s)
Dietary Supplements , Lactation , Learning/drug effects , Memory/drug effects , N-Acetylneuraminic Acid/administration & dosage , Oligosaccharides/administration & dosage , Animals , Behavior, Animal/drug effects , Female , Frontal Lobe/chemistry , Lactose/administration & dosage , Lactose/analogs & derivatives , Long-Term Potentiation/drug effects , Male , Milk/chemistry , Neural Cell Adhesion Molecule L1/analysis , Oligosaccharides/chemistry , Rats , Rats, Sprague-Dawley , Sialic Acids/analysis
15.
J Food Sci ; 83(2): 499-508, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29377120

ABSTRACT

There has been increased interest in the use of dietary ingredients, including prebiotics such as human-milk oligosaccharides (HMOs), as therapeutic strategies for food allergy. Understanding the mechanisms underlying the beneficial effects of HMOs is important to realizing their therapeutic potential. Here we demonstrate that the HMO, 6'-sialyllactose (6'SL) inhibited chemokine (IL-8 and CCL20) release from T-84 and HT-29 cells stimulated with antigen-antibody complex, TNFα or PGE2 ; an effect that was PPARγ dependent and associated with decreased activity of the transcription factors AP-1 and NFκB. In contrast, 2'-fucosyllactose (2'FL) selectively inhibited CCL20 release in response to antigen antibody complex in a PPARγ independent manner. This study reinforces the concept that structurally different oligosaccharides have distinct biological activities and identifies, for the first time, that the HMOs, 6'SL, and 2'FL, modulate human epithelial cell responses related to allergic disease. These findings encourage further investigation of the therapeutic potential of specific HMOs in food allergy. PRACTICAL APPLICATION: This study provides evidence for direct effects of HMOs in addition to their prebiotic role and demonstrates, for the first time, modulation of Ag-IgE complex activation of human epithelial cells that may have important implications for food-allergy. The study also reinforces the concept that structurally different oligosaccharides have distinct biological activities. In determining the composition of infant formula, addition of oligosaccharides with specific structures may provide direct modulation of immune responses and potentially attenuate symptoms or development of food allergy.


Subject(s)
Antigen-Antibody Complex/immunology , Chemokines/immunology , Epithelial Cells/immunology , Food Hypersensitivity/immunology , Milk, Human/chemistry , Oligosaccharides/pharmacology , Prebiotics/analysis , Chemokines/genetics , Epithelial Cells/drug effects , Food Hypersensitivity/drug therapy , HT29 Cells , Humans , Infant Formula/chemistry , Intestines/drug effects , Intestines/immunology , Trisaccharides/pharmacology
16.
PLoS One ; 11(11): e0166070, 2016.
Article in English | MEDLINE | ID: mdl-27851789

ABSTRACT

2´-fucosyllactose (2´-FL) is an abundant human milk oligosaccharide (HMO) in human milk with diverse biological effects. We recently reported ingested 2´-FL stimulates central nervous system (CNS) function, such as hippocampal long term potentiation (LTP) and learning and memory in rats. Conceivably the effect of 2´-FL on CNS function may be via the gut-brain axis (GBA), specifically the vagus nerve, and L-fucose (Fuc) may play a role. This study had two aims: (1) determine if the effect of ingested 2´-FL on the modulation of CNS function is dependent on the integrity of the molecule; and (2) confirm if oral 2´-FL modified hippocampal LTP and associative learning related skills in rats submitted to bilateral subdiaphragmatic vagotomy. Results showed that 2´-FL but not Fuc enhanced LTP, and vagotomy inhibited the effects of oral 2´-FL on LTP and associative learning related paradigms. Taken together, the data show that dietary 2´-FL but not its Fuc moiety affects cognitive domains and improves learning and memory in rats. This effect is dependent on vagus nerve integrity, suggesting GBA plays a role in 2´-FL-mediated cognitive benefits.


Subject(s)
Brain/physiology , Conditioning, Operant/drug effects , Diet , Digestive System/drug effects , Long-Term Potentiation/drug effects , Trisaccharides/pharmacology , Vagus Nerve/physiology , Administration, Oral , Animals , Brain/drug effects , Fucose/administration & dosage , Fucose/pharmacology , Male , Rats, Sprague-Dawley , Trisaccharides/administration & dosage , Vagotomy , Vagus Nerve/drug effects
17.
J Nutr ; 146(12): 2559-2566, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27798337

ABSTRACT

BACKGROUND: Evidence suggests that human milk oligosaccharides (HMOs) provide multiple benefits to infants, including prebiotic effects, gut maturation, antimicrobial activities, and immune modulation. Clinical intervention studies with HMOs are required to confirm these benefits in infants. OBJECTIVE: Our objective was to investigate the effects of feeding formulas supplemented with the HMO 2'-fucosyllactose (2'-FL) on biomarkers of immune function in healthy term infants. METHODS: We performed a substudy nested within a randomized, double-blind, controlled growth and tolerance study in healthy singleton infants (birth weight ≥2490 g) who were enrolled by 5 d of life and exclusively formula-fed (n = 317) or breastfed (n = 107) from enrollment to 4 mo of age. Formula-fed infants were randomly assigned to receive 1 of 3 formulas, all containing 2.4 g total oligosaccharides/L [control: galacto-oligosaccharides (GOS) only; experimental formulas: GOS + 0.2 or 1.0 g 2'-FL/L], and compared with a breastfed reference group. For this substudy, blood samples were drawn from infants at 6 wk of age (n = 31-42/group). Peripheral blood mononuclear cells (PBMCs) were isolated for cellular phenotyping and stimulated ex vivo with phytohemagglutinin for proliferation and cell cycle progression or respiratory syncytial virus (RSV). Cytokine concentrations were measured in plasma and in ex vivo-stimulated culture supernatants. RESULTS: Breastfed infants and infants fed either of the experimental formulas with 2'-FL were not different but had 29-83% lower concentrations of plasma inflammatory cytokines than did infants fed the control formula [interleukin (IL) receptor antagonist (IL-1ra), IL-1α, IL-1ß, IL-6, and tumor necrosis factor α (TNF-α)] (P ≤ 0.05). In ex vivo RSV-stimulated PBMC cultures, breastfed infants were not different than either of the groups fed formula with 2'-FL, but they had lower concentrations of TNF-α (31%) and interferon γ (IFN-γ 54%) (P ≤ 0.05) and tended to have lower IL-1ra (25%) and IL-6 (38%) (unadjusted P ≤ 0.05) and IL-1ß (30%) (unadjusted P = 0.06) than did infants fed the control formula. CONCLUSIONS: Our data indicate that infants fed formula supplemented with 2'-FL exhibit lower plasma and ex vivo inflammatory cytokine profiles, similar to those of a breastfed reference group. This trial was registered at clinicaltrials.gov as NCT01808105.


Subject(s)
Breast Feeding , Cytokines/blood , Infant Formula/chemistry , Trisaccharides/pharmacology , Cell Proliferation , Cytokines/metabolism , Double-Blind Method , Gene Expression Regulation , Humans , Infant , Infant, Newborn , Inflammation/blood , Inflammation/metabolism , Leukocytes, Mononuclear/metabolism , Respiratory Syncytial Viruses/isolation & purification , Trisaccharides/administration & dosage , Trisaccharides/chemistry , Viral Load
18.
Br J Nutr ; 116(7): 1175-1187, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27609061

ABSTRACT

Necrotising enterocolitis (NEC) is a common disease in premature infants characterised by intestinal ischaemia and necrosis. The only effective preventative strategy against NEC is the administration of breast milk, although the protective mechanisms remain unknown. We hypothesise that an abundant human milk oligosaccharide (HMO) in breast milk, 2'-fucosyllactose (2'FL), protects against NEC by enhancing intestinal mucosal blood flow, and we sought to determine the mechanisms underlying this protection. Administration of HMO-2'FL protected against NEC in neonatal wild-type mice, resulted in a decrease in pro-inflammatory markers and preserved the small intestinal mucosal architecture. These protective effects occurred via restoration of intestinal perfusion through up-regulation of the vasodilatory molecule endothelial nitric oxide synthase (eNOS), as administration of HMO-2'FL to eNOS-deficient mice or to mice that received eNOS inhibitors did not protect against NEC, and by 16S analysis HMO-2'FL affected the microbiota of the neonatal mouse gut, although these changes do not seem to be the primary mechanism of protection. Induction of eNOS by HMO-2'FL was also observed in cultured endothelial cells, providing a link between eNOS and HMO in the endothelium. These data demonstrate that HMO-2'FL protects against NEC in part through maintaining mesenteric perfusion via increased eNOS expression, and suggest that the 2'FL found in human milk may be mediating some of the protective benefits of breast milk in the clinical setting against NEC.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Infant, Premature, Diseases/physiopathology , Milk, Human/chemistry , Splanchnic Circulation/drug effects , Trisaccharides/administration & dosage , Animals , Animals, Newborn , Disease Models, Animal , Enterocolitis, Necrotizing/physiopathology , Female , Gene Expression/drug effects , Humans , Infant, Newborn , Intestinal Mucosa/blood supply , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/drug effects , Nitric Oxide/analysis , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/physiology
19.
J Nutr Biochem ; 31: 20-7, 2016 05.
Article in English | MEDLINE | ID: mdl-27133420

ABSTRACT

Human milk oligosaccharides have been proposed to exert beneficial effects on brain development. During the last decades, most of the studies have focused on the evaluation of sialylated structures but recent experiments have also tested fucosylated oligosaccharides, i.e. 2'-fucosyllactose (2'-FL). The present study aimed to determine whether oral 2'-FL has an effect on the development of newborn brain, contributing to enhance cognitive skills later in life. Rat pups received an oral supplementation of 2'-FL (2'-FL group) or water (control group) during the lactation period. Thereafter, animals were maintained on a rodent standard diet. Rats (n=12 rats/group) were evaluated twice, at age 4-6weeks and again at age 1year, using classical behavioral tests. In vivo long-term potentiation (LTP) was also performed at the same ages (n=10 rats/group). Both groups showed similar behavior when the animals were assessed just after weaning (age 4-6weeks), although the 2'-FL group seemed to perform slightly better in Morris Water Maze. At age 1year, 2'-FL rats performed significantly better in the Novel Object Recognition and Y maze paradigms, when compared to controls. In addition, LTP was more intense and longer lasting in the rats supplemented with 2'-FL than in control animals, both in young and adult animals. Oral administration of 2'-FL exclusively during lactation enhanced cognitive abilities, not only in childhood but also in adulthood.


Subject(s)
Trisaccharides/administration & dosage , Administration, Oral , Animals , Behavior, Animal/drug effects , Female , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Pregnancy , Rats
20.
Biochem J ; 473(10): 1343-53, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26976925

ABSTRACT

Human milk glycans (HMGs) are prebiotics, pathogen receptor decoys and regulators of host physiology and immune responses. Mechanistically, human lectins (glycan-binding proteins, hGBP) expressed by dendritic cells (DCs) are of major interest, as these cells directly contact HMGs. To explore such interactions, we screened many C-type lectins and sialic acid-binding immunoglobulin-like lectins (Siglecs) expressed by DCs for glycan binding on microarrays presenting over 200 HMGs. Unexpectedly, DC-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) showed robust binding to many HMGs, whereas other C-type lectins failed to bind, and Siglec-5 and Siglec-9 showed weak binding to a few glycans. By contrast, most hGBP bound to multiple glycans on other microarrays lacking HMGs. An α-linked fucose residue was characteristic of HMGs bound by DC-SIGN. Binding of DC-SIGN to the simple HMGs 2'-fucosyl-lactose (2'-FL) and 3-fucosyl-lactose (3-FL) was confirmed by flow cytometry to beads conjugated with 2'-FL or 3-FL, as well as the ability of the free glycans to inhibit DC-SIGN binding. 2'-FL had an IC50 of ∼1 mM for DC-SIGN, which is within the physiological concentration of 2'-FL in human milk. These results demonstrate that DC-SIGN among the many hGBP expressed by DCs binds to α-fucosylated HMGs, and suggest that such interactions may be important in influencing immune responses in the developing infant.


Subject(s)
Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Milk, Human/chemistry , Polysaccharides/metabolism , Receptors, Cell Surface/metabolism , Dendritic Cells/metabolism , Humans , Protein Array Analysis , Protein Binding , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Trisaccharides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...