In vitro comparative evaluation of the impact of lacto-N-biose I, a major building block of human milk oligosaccharides, on the fecal microbiota of infants

Anaerobe ◽  
2013 ◽  
Vol 19 ◽  
pp. 50-57 ◽  
Author(s):  
Takumi Satoh ◽  
Toshitaka Odamaki ◽  
Mariko Namura ◽  
Takashi Shimizu ◽  
Keiji Iwatsuki ◽  
...  
2009 ◽  
Vol 76 (1) ◽  
pp. 54-59 ◽  
Author(s):  
Jin-zhong Xiao ◽  
Sachiko Takahashi ◽  
Mamoru Nishimoto ◽  
Toshitaka Odamaki ◽  
Tomoko Yaeshima ◽  
...  

ABSTRACT This study investigated the potential utilization of lacto-N-biose I (LNB) by individual strains of bifidobacteria. LNB is a building block for the human milk oligosaccharides, which have been suggested to be a factor for selective growth of bifidobacteria. A total of 208 strains comprising 10 species and 4 subspecies were analyzed for the presence of the galacto-N-biose/lacto-N-biose I phosphorylase (GLNBP) gene (lnpA) and examined for growth when LNB was used as the sole carbohydrate source. While all strains of Bifidobacterium longum subsp. longum, B. longum subsp. infantis, B. breve, and B. bifidum were able to grow on LNB, none of the strains of B. adolescentis, B. catenulatum, B. dentium, B. angulatum, B. animalis subsp. lactis, and B. thermophilum showed any growth. In addition, some strains of B. pseudocatenulatum, B. animalis subsp. animalis, and B. pseudolongum exhibited the ability to utilize LNB. With the exception for B. pseudocatenulatum, the presence of lnpA coincided with LNB utilization in almost all strains. These results indicate that bifidobacterial species, which are the predominant species found in infant intestines, are potential utilizers of LNB. These findings support the hypothesis that GLNBP plays a key role in the colonization of bifidobacteria in the infant intestine.


Nutrients ◽  
2020 ◽  
Vol 12 (10) ◽  
pp. 3047
Author(s):  
Jane M. Natividad ◽  
Andreas Rytz ◽  
Sonia Keddani ◽  
Gabriela Bergonzelli ◽  
Clara L. Garcia-Rodenas

Breastfeeding is integral in the proper maturation of the intestinal barrier and protection against inflammatory diseases. When human milk (HM) is not available, supplementation with HM bioactives like Human Milk Oligosaccharides (HMOs) may help in providing breastfeeding barrier-protective benefits. An increasing HMO variety is becoming industrially available, enabling approaching the HMO complexity in HM. We aimed at assessing the impact of blends of available HMOs on epithelial barrier function in vitro. The capacity of individual [2′-Fucosyllactose (2′FL), Difucosyllactose, Lacto-N-tetraose, Lacto-N-neotetraose, 3′-Siallylactose and 6′-Siallylactose] or varying combinations of 3, 5 and 6 HMOs to modulate fluorescein-isothiocyanate (FITC)-labelled Dextran 4 KDa (FD4) translocation and/or transepithelial resistance (TEER) was characterized in Caco-2: HT29- methotrexate (MTX) cell line monolayers before and after an inflammatory challenge with TNF-α and IFN-γ. The six HMO blend (HMO6) dose-dependently limited the cytokine-induced FD4 translocation and TEER decrease and increased TEER values before challenge. Similarly, 3 and 5 HMO blends conferred a significant protection against the challenge, with 2′FL, one of the most abundant but most variable oligosaccharides in HM, being a key contributor. Overall, our results suggest differential ability of specific HMOs in modulating the intestinal barrier and support the potential of supplementation with combinations of available HMOs to promote gut health and protect against intestinal inflammatory disorders.


2021 ◽  
Vol Publish Ahead of Print ◽  
Author(s):  
Cilius Esmann Fonvig ◽  
Ingvild Dybdrodt Amundsen ◽  
Louise Kristine Vigsnæs ◽  
Nikolaj Sørensen ◽  
Christine Frithioff-Bøjsøe ◽  
...  

2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Ioannis Kostopoulos ◽  
Janneke Elzinga ◽  
Noora Ottman ◽  
Jay T. Klievink ◽  
Bernadet Blijenberg ◽  
...  

Glycobiology ◽  
2020 ◽  
Vol 30 (10) ◽  
pp. 774-786 ◽  
Author(s):  
Sara Porfirio ◽  
Stephanie Archer-Hartmann ◽  
G Brett Moreau ◽  
Girija Ramakrishnan ◽  
Rashidul Haque ◽  
...  

Abstract Human breast milk is an incredibly rich and complex biofluid composed of proteins, lipids and complex carbohydrates, including a diverse repertoire of free human milk oligosaccharides (HMOs). Strikingly, HMOs are not digested by the infant but function as prebiotics for bacterial strains associated with numerous benefits. Considering the broad variety of beneficial effects of HMOs, and the vast number of factors that affect breast milk composition, the analysis of HMO diversity and complexity is of utmost relevance. Using human milk samples from a cohort of Bangladeshi mothers participating in a study on malnutrition and stunting in children, we have characterized breast milk oligosaccharide composition by means of permethylation followed by liquid chromatography coupled with high-resolution tandem mass spectrometry (LC-MS/MS) analysis. This approach identified over 100 different glycoforms and showed a wide diversity of milk composition, with a predominance of fucosylated and sialylated HMOs over nonmodified HMOs. We observed that these samples contain on average 80 HMOs, with the highest permethylated masses detected being >5000 mass units. Here we report an easily implemented method developed for the separation, characterization and relative quantitation of large arrays of HMOs, including higher molecular weight sialylated HMOs. Our ultimate goal is to create a simple, high-throughput method, which can be used for full characterization of sialylated and/or fucosylated HMOs. These results demonstrate how current analytical techniques can be applied to characterize human milk composition, providing new tools to help the scientific community shed new light on the impact of HMOs during infant development.


2012 ◽  
Vol 108 (10) ◽  
pp. 1839-1846 ◽  
Author(s):  
Evelyn Jantscher-Krenn ◽  
Tineke Lauwaet ◽  
Laura A. Bliss ◽  
Sharon L. Reed ◽  
Frances D. Gillin ◽  
...  

Human milk oligosaccharides (HMO), complex sugars that are highly abundant in breast milk, block viral and bacterial attachment to the infant's intestinal epithelium and lower the risk of infections. We hypothesised that HMO also prevent infections with the protozoan parasiteEntamoeba histolytica,as its major virulence factor is a lectin that facilitates parasite attachment and cytotoxicity and binds galactose (Gal) andN-acetyl-galactosamine. HMO contain Gal, are only minimally digested in the small intestine and reach the colon, the site ofE. histolyticainfection. The objective of the present study was to investigate whether HMO reduceE. histolyticaattachment and cytotoxicity. Ourin vitroresults show that physiological concentrations of isolated, pooled HMO detachE. histolyticaby more than 80 %. In addition, HMO rescueE. histolytica-induced destruction of human intestinal epithelial HT-29 cells in a dose-dependent manner. The cytoprotective effects were structure-specific. Lacto-N-tetraose with its terminal Gal rescued up to 80 % of the HT-29 cells, while HMO with fucose α1–2-linked to the terminal Gal had no effect. Galacto-oligosaccharides (GOS), which also contain terminal Gal and are currently added to infant formula to mimic some of the beneficial effects of HMO, completely abolishedE. histolyticaattachment and cytotoxicity at 8 mg/ml. Although our results need to be confirmedin vivo, they may provide one explanation for why breast-fed infants are at lower risk ofE. histolyticainfections. HMO and GOS are heat tolerant, stable, safe and in the case of GOS, inexpensive, which could make them valuable candidates as alternative preventive and therapeutic anti-amoebic agents.


2022 ◽  
Vol 9 ◽  
Author(s):  
Sarah Bajorek ◽  
Rebbeca M. Duar ◽  
Maxwell Corrigan ◽  
Christa Matrone ◽  
Kathryn A. Winn ◽  
...  

Not all infants carry specialized gut microbes, meaning they cannot digest human milk oligosaccharides and therefore do not receive complete benefits from human milk. B. infantis EVC001 is equipped to convert the full array of complex oligosaccharides into compounds usable by the infant, making it an ideal candidate to stabilize gut function and improve nutrition in preterm infants. A prospective, open-label study design was used to evaluate the tolerability of B. infantis EVC001 and its effects on the fecal microbiota in preterm infants in a Neonatal Intensive Care Unit. Thirty preterm infants <1,500 g and/or <33 weeks gestation at birth were divided into two matched groups, and control infants were enrolled and discharged prior to enrolling EVC001 infants to prevent cross-colonization of B. infantis: (1) fifteen control infants received no EVC001, and (2) fifteen infants received once-daily feedings of B. infantis EVC001 (8.0 x 109 CFU) in MCT oil. Clinical information regarding medications, growth, nutrition, gastrointestinal events, diagnoses, and procedures was collected throughout admission. Infant stool samples were collected at baseline, Study Days 14 and 28, and 34-, 36-, and 38-weeks of gestation. Taxonomic composition of the fecal microbiota, functional microbiota analysis, B. infantis, and human milk oligosaccharides (HMOs) in the stool were determined or quantified using 16S rRNA gene sequencing, metagenomic sequencing, qPCR, and mass spectrometry, respectively. No adverse events or tolerability issues related to EVC001 were reported. Control infants had no detectable levels of B. infantis. EVC001 infants achieved high levels of B. infantis (mean = 9.7 Log10 CFU/μg fecal DNA) by Study Day 14, correlating with less fecal HMOs (ρ = −0.83, P < 0.0001), indicating better HMO utilization in the gut. In this study, B. infantis EVC001 was shown to be safe, well-tolerated, and efficient in colonizing the preterm infant gut and able to increase the abundance of bifidobacteria capable of metabolizing HMOs, resulting in significantly improved utilization of human milk.Clinical Trial Registration:https://clinicaltrials.gov/ct2/show/NCT03939546, identifier: NCT03939546.


Sign in / Sign up

Export Citation Format

Share Document