Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Cell Host Microbe ; 31(9): 1523-1538.e10, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37657443

ABSTRACT

Manipulation of the gut microbiome using live biotherapeutic products shows promise for clinical applications but remains challenging to achieve. Here, we induced dysbiosis in 56 healthy volunteers using antibiotics to test a synbiotic comprising the infant gut microbe, Bifidobacterium longum subspecies infantis (B. infantis), and human milk oligosaccharides (HMOs). B. infantis engrafted in 76% of subjects in an HMO-dependent manner, reaching a relative abundance of up to 81%. Changes in microbiome composition and gut metabolites reflect altered recovery of engrafted subjects compared with controls. Engraftment associates with increases in lactate-consuming Veillonella, faster acetate recovery, and changes in indolelactate and p-cresol sulfate, metabolites that impact host inflammatory status. Furthermore, Veillonella co-cultured in vitro and in vivo with B. infantis and HMO converts lactate produced by B. infantis to propionate, an important mediator of host physiology. These results suggest that the synbiotic reproducibly and predictably modulates recovery of a dysbiotic microbiome.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Synbiotics , Infant , Humans , Adult , Dysbiosis , Milk, Human , Lactic Acid , Veillonella
2.
Cell Host Microbe ; 30(5): 712-725.e7, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35504279

ABSTRACT

Predictable and sustainable engraftment of live biotherapeutic products into the human gut microbiome is being explored as a promising way to modulate the human gut microbiome. We utilize a synbiotic approach pairing the infant gut microbe Bifidobacterium longum subspecies infantis (B. infantis) and human milk oligosaccharides (HMO). B. infantis, which is typically absent in adults, engrafts into healthy adult microbiomes in an HMO-dependent manner at a relative abundance of up to 25% of the bacterial population without antibiotic pretreatment or adverse effects. Corresponding changes in metabolites are detected. Germ-free mice transplanted with dysbiotic human microbiomes also successfully engraft with B. infantis in an HMO-dependent manner, and the synbiotic augments butyrate levels both in this in vivo model and in in vitro cocultures of the synbiotic with specific Firmicutes species. Finally, the synbiotic inhibits the growth of enteropathogens in vitro. Our findings point to a potential safe mechanism for ameliorating dysbioses characteristic of numerous human diseases.


Subject(s)
Microbiota , Synbiotics , Animals , Anti-Bacterial Agents/metabolism , Dysbiosis/metabolism , Dysbiosis/therapy , Humans , Infant , Mice , Milk, Human/microbiology , Oligosaccharides/metabolism
3.
Nat Commun ; 9(1): 5010, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30479342

ABSTRACT

Neonatal rotavirus infections are predominantly asymptomatic. While an association with gastrointestinal symptoms has been described in some settings, factors influencing differences in clinical presentation are not well understood. Using multidisciplinary approaches, we show that a complex interplay between human milk oligosaccharides (HMOs), milk microbiome, and infant gut microbiome impacts neonatal rotavirus infections. Validating in vitro studies where HMOs are not decoy receptors for neonatal strain G10P[11], population studies show significantly higher levels of Lacto-N-tetraose (LNT), 2'-fucosyllactose (2'FL), and 6'-siallylactose (6'SL) in milk from mothers of rotavirus-positive neonates with gastrointestinal symptoms. Further, these HMOs correlate with abundance of Enterobacter/Klebsiella in maternal milk and infant stool. Specific HMOs also improve the infectivity of a neonatal strain-derived rotavirus vaccine. This study provides molecular and translational insight into host factors influencing neonatal rotavirus infections and identifies maternal components that could promote the performance of live, attenuated rotavirus vaccines.


Subject(s)
Gastrointestinal Microbiome , Milk, Human/chemistry , Milk, Human/microbiology , Oligosaccharides/metabolism , Rotavirus Infections/microbiology , Feces/microbiology , Humans , Infant, Newborn , Rotavirus/pathogenicity , Rotavirus Infections/immunology , Rotavirus Vaccines/immunology
4.
Gut ; 67(6): 1064-1070, 2018 06.
Article in English | MEDLINE | ID: mdl-28381523

ABSTRACT

OBJECTIVE: Necrotising enterocolitis (NEC) is one of the most common and often fatal intestinal disorders in preterm infants. Markers to identify at-risk infants as well as therapies to prevent and treat NEC are limited and urgently needed. NEC incidence is significantly lower in breast-fed compared with formula-fed infants. Infant formula lacks human milk oligosaccharides (HMO), such as disialyllacto-N-tetraose (DSLNT), which prevents NEC in neonatal rats. However, it is unknown if DSLNT also protects human preterm infants. DESIGN: We conducted a multicentre clinical cohort study and recruited 200 mothers and their very low birthweight infants that were predominantly human milk-fed. We analysed HMO composition in breast milk fed to infants over the first 28 days post partum, matched each NEC case with five controls and used logistic regression and generalised estimating equation to test the hypothesis that infants who develop NEC receive milk with less DSLNT than infants who do not develop NEC. RESULTS: Eight infants in the cohort developed NEC (Bell stage 2 or 3). DSLNT concentrations were significantly lower in almost all milk samples in NEC cases compared with controls, and its abundance could identify NEC cases prior to onset. Aggregate assessment of DSLNT over multiple days enhanced the separation of NEC cases and control subjects. CONCLUSIONS: DSLNT content in breast milk is a potential non-invasive marker to identify infants at risk of developing NEC, and screen high-risk donor milk. In addition, DSLNT could serve as a natural template to develop novel therapeutics against this devastating disorder.


Subject(s)
Enterocolitis, Necrotizing/etiology , Milk, Human/chemistry , Oligosaccharides/analysis , Breast Feeding , Case-Control Studies , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Infant, Premature , Infant, Very Low Birth Weight , Male , Prospective Studies , Risk , Risk Assessment
5.
J Org Chem ; 82(24): 13152-13160, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29124935

ABSTRACT

Necrotizing enterocolitis (NEC) is one of the most common and devastating intestinal disorders in preterm infants. Therapies to meet the clinical needs for this special and highly vulnerable population are extremely limited. A specific human milk oligosaccharide (HMO), disialyllacto-N-tetraose (DSLNT), was shown to contribute to the beneficial effects of breastfeeding as it prevented NEC in a neonatal rat model and was associated with lower NEC risk in a human clinical cohort study. Herein, gram-scale synthesis of two DSLNT analogs previously shown to have NEC preventing effect is described. In addition, four novel disialyl glycans have been designed and synthesized by enzymatic or chemoenzymatic methods. Noticeably, two disialyl tetraoses have been produced by enzymatic sialylation of chemically synthesized thioethyl ß-disaccharides followed by removal of the thioethyl aglycon. Dose-dependent and single-dose comparison studies showed varying NEC-preventing effects of the disialyl glycans in neonatal rats. This study helps to refine the structure requirement of the NEC-preventing effect of disialyl glycans and provides important dose-dependent information for using DSLNT analogs as potential therapeutics for NEC prevention in preterm infants.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Oligosaccharides/chemistry , Polysaccharides/chemistry , Animals , Humans , Infant, Newborn , Models, Animal , Oligosaccharides/pharmacology , Polysaccharides/pharmacology , Rats
6.
J Biol Chem ; 292(27): 11243-11249, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28416607

ABSTRACT

Streptococcus agalactiae (group B Streptococcus, GBS) is a leading cause of invasive bacterial infections in newborns, typically acquired vertically during childbirth secondary to maternal vaginal colonization. Human milk oligosaccharides (HMOs) have important nutritional and biological activities that guide the development of the immune system of the infant and shape the composition of normal gut microbiota. In this manner, HMOs help protect against pathogen colonization and reduce the risk of infection. In the course of our studies of HMO-microbial interactions, we unexpectedly uncovered a novel HMO property to directly inhibit the growth of GBS independent of host immunity. By separating different HMO fractions through multidimensional chromatography, we found the bacteriostatic activity to be confined to specific non-sialylated HMOs and synergistic with a number of conventional antibiotic agents. Phenotypic screening of a GBS transposon insertion library identified a mutation within a GBS-specific gene encoding a putative glycosyltransferase that confers resistance to HMOs, suggesting that HMOs may function as an alternative substrate to modify a GBS component in a manner that impairs growth kinetics. Our study uncovers a unique antibacterial role for HMOs against a leading neonatal pathogen and expands the potential therapeutic utility of these versatile molecules.


Subject(s)
Anti-Bacterial Agents/pharmacology , Milk, Human/chemistry , Oligosaccharides/pharmacology , Streptococcus agalactiae/growth & development , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Female , Humans , Oligosaccharides/chemistry , Oligosaccharides/isolation & purification , Streptococcus agalactiae/genetics
8.
Br J Nutr ; 116(2): 294-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27212112

ABSTRACT

Necrotising enterocolitis (NEC) is one of the most frequent and fatal intestinal disorders in preterm infants and has very limited treatment options. Breast-fed infants are at a 6-10-fold lower NEC risk than formula-fed infants, and we have previously shown that human milk oligosaccharides (HMO) improved survival and reduced pathology in a rat NEC model. The HMO disialyllacto-N-tetraose (DSLNT) was most effective, and sialylation was shown to be essential for its protective effect. Galacto-oligosaccharides (GOS), currently added to some infant formula, but not containing sialic acid, had no effect. In addition to DSLNT, our previous work also showed that the neutral HMO fraction, which contains high concentrations of 2'-fucosyllactose (2'FL), slightly improved pathology scores. Here, we assessed the in vivo efficacy of 2'FL, as well as of GOS that we enzymatically sialylated (Sia-GOS). Neonatal rats were randomised into the following study groups - dam-fed (DF), formula-fed (FF), FF containing pooled HMO (10 mg/ml), GOS (8 mg/ml), Sia-GOS (500 µm) or 2'FL (2 mg/ml) - and subjected to the established NEC protocol. The DF and HMO groups had the lowest pathology scores with mean values of 0·67 (sd 0·34) and 0·90 (sd 0·47), respectively. The FF group had significantly elevated pathology scores of 2·02 (sd 0·63). Although the addition of GOS to the formula had no protective effect and generated scores of 2·00 (sd 0·63), the addition of Sia-GOS or 2'FL significantly lowered pathology scores to 1·32 (sd 0·56) (P<0·0034) and 1·43 (sd 0·51) (P<0·0040), respectively. The results warrant further studies to investigate the underlying mechanisms and to assess safety and efficacy in human neonates.


Subject(s)
Enterocolitis, Necrotizing/drug therapy , Galactose/therapeutic use , Infant Formula/chemistry , Milk, Human/chemistry , Oligosaccharides/therapeutic use , Sialic Acids/therapeutic use , Trisaccharides/therapeutic use , Animals , Animals, Newborn , Breast Feeding , Female , Galactose/metabolism , Galactose/pharmacology , Humans , Infant Nutritional Physiological Phenomena , Infant, Newborn , Intestines/drug effects , Intestines/pathology , Oligosaccharides/metabolism , Oligosaccharides/pharmacology , Random Allocation , Rats, Sprague-Dawley , Sialic Acids/metabolism , Sialic Acids/pharmacology , Trisaccharides/pharmacology
9.
J Nutr ; 144(8): 1227-33, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24919691

ABSTRACT

The heavy burden of maternal HIV infection has resulted in a high prevalence of premature birth and associated necrotizing enterocolitis (NEC). Human milk oligosaccharides (HMOs) were recently associated with HIV infection and transmission through breastfeeding and were also shown to reduce NEC in an animal model, particularly the HMO disialyllacto-N-tetraose (DSLNT). The primary aim of this study was to verify differences in HMO composition between HIV-infected and HIV-uninfected women. The secondary aim was to assess whether the HMO composition in the milk of mothers whose infants were diagnosed with NEC differs from that of mothers whose infants did not develop NEC. This study forms part of a larger clinical trial conducted at the Tygerberg Children's Hospital, Cape Town, South Africa, which recruited HIV-infected and HIV-uninfected mothers and their preterm infants (<34 wk gestation; ≥500 and ≤1250 g). Eighty-two mother-infant pairs were selected for the substudy. Mother-infant pairs were stratified according to the mother's HIV (infected/uninfected) and secretor status (secretor/nonsecretor). HMOs in 4- and 28-d postpartum milk samples were analyzed by HPLC and compared between groups. Our results confirm previous reports that HIV-infected mothers have higher relative abundances of 3'-sialyllactose in their milk compared with HIV-uninfected mothers (10.7% vs. 6.8%; P < 0.01). Most intriguingly, the data also indicated that low concentrations of DSLNT in the 4-d milk samples in the mother's milk increased the infant's risk of NEC (200 ± 126 vs. 345 ± 186 µg/mL; P < 0.05), which is in accordance with results from previously published animal studies and warrants further investigation. This trial was registered at clinicaltrials.gov as NCT01868737.


Subject(s)
Enterocolitis, Necrotizing/epidemiology , HIV Infections/metabolism , Milk, Human/chemistry , Oligosaccharides/analysis , Premature Birth/epidemiology , Adult , Breast Feeding , Enterocolitis, Necrotizing/etiology , Female , HIV Infections/complications , Humans , Incidence , Infant, Premature/metabolism , Infant, Very Low Birth Weight/metabolism , Male , Mothers , Pregnancy , Premature Birth/etiology , Randomized Controlled Trials as Topic , South Africa , Young Adult
10.
Angew Chem Int Ed Engl ; 53(26): 6687-91, 2014 Jun 23.
Article in English | MEDLINE | ID: mdl-24848971

ABSTRACT

Two novel synthetic α2-6-linked disialyl hexasaccharides, disialyllacto-N-neotetraose (DSLNnT) and α2-6-linked disialyllacto-N-tetraose (DS'LNT), were readily obtained by highly efficient one-pot multienzyme (OPME) reactions. The sequential OPME systems described herein allowed the use of an inexpensive disaccharide and simple monosaccharides to synthesize the desired complex oligosaccharides with high efficiency and selectivity. DSLNnT and DS'LNT were shown to protect neonatal rats from necrotizing enterocolitis (NEC) and are good therapeutic candidates for preclinical experiments and clinical application in treating NEC in preterm infants.


Subject(s)
Enterocolitis, Necrotizing/drug therapy , Oligosaccharides/chemical synthesis , Protective Agents/therapeutic use , Animals , Bifidobacterium/enzymology , Drug Evaluation, Preclinical , Multienzyme Complexes/metabolism , Oligosaccharides/chemistry , Oligosaccharides/therapeutic use , Protective Agents/chemical synthesis , Protective Agents/chemistry , Rats
11.
J Infect Dis ; 209(3): 389-98, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23990566

ABSTRACT

The invasive pathogen uropathogenic Escherichia coli (UPEC) is the primary cause of urinary tract infections (UTIs). Recurrent infection that can progress to life-threatening renal failure has remained as a serious global health concern in infants. UPEC adheres to and invades bladder epithelial cells to establish infection. Studies have detected the presence of human milk oligosaccharides (HMOs) in urine of breast-fed, but not formula-fed, neonates. We investigated the mechanisms HMOs deploy to elicit protection in human bladder epithelial cells infected with UPEC CFT073, a prototypic urosepsis-associated strain. We found a significant reduction in UPEC internalization into HMO-pretreated epithelial cells without observing any significant effect in UPEC binding to these cells. This event coincides with a rapid decrease in host cell cytotoxicity, recognized by LIVE/DEAD staining and cell detachment, but independent of caspase-mediated or mitochondrial-mediated programmed cell death pathways. Further investigation revealed HMOs, and particularly the sialic acid-containing fraction, reduced UPEC-mediated MAPK and NF-κB activation. Collectively, our results indicate that HMOs can protect bladder epithelial cells from deleterious cytotoxic and proinflammatory effects of UPEC infection, and may be one contributing mechanism underlying the epidemiological evidence of reduced UTI incidence in breast-fed infants.


Subject(s)
Endocytosis/drug effects , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Immunologic Factors/metabolism , Milk, Human/chemistry , Oligosaccharides/metabolism , Uropathogenic Escherichia coli/physiology , Bacterial Adhesion/drug effects , Cell Survival/drug effects , Humans , Infant , Uropathogenic Escherichia coli/drug effects
12.
J Pediatr Gastroenterol Nutr ; 58(2): 165-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24048169

ABSTRACT

Breast-feeding reduces the risk of enteric bacterial infections in newborns in part because of human milk oligosaccharides (HMOs), complex glycans that are present in human milk, but not in infant formula. Enteropathogenic Escherichia coli (EPEC) are attaching/effacing pathogens that cause serious diarrheal illness with potentially high mortality in infants. We isolated HMOs from pooled human milk and found that they significantly reduce EPEC attachment to cultured epithelial cells. In suckling mice, administration of HMOs significantly reduced colonization with EPEC compared with untreated controls. These data suggest an essential role for HMOs in the prevention of EPEC infections in human infants.


Subject(s)
Anti-Infective Agents/therapeutic use , Bacterial Adhesion/drug effects , Diarrhea/prevention & control , Enteropathogenic Escherichia coli/drug effects , Escherichia coli Infections/prevention & control , Milk, Human/chemistry , Oligosaccharides/therapeutic use , Animals , Anti-Infective Agents/pharmacology , Breast Feeding , Diarrhea/etiology , Diarrhea/microbiology , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Humans , Mice, Inbred C57BL , Oligosaccharides/isolation & purification , Oligosaccharides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL