Materials-Methods-Analysis-Hmo-Composition-Majority-Participants-Supplements-d

Материал из ТОГБУ Компьютерный Центр
Версия от 09:01, 22 марта 2024; Enemycatsup62 (обсуждение | вклад) (Materials-Methods-Analysis-Hmo-Composition-Majority-Participants-Supplements-d)

(разн.) ← Предыдущая | Текущая версия (разн.) | Следующая → (разн.)
Перейти к: навигация, поиск

Results In an unadjusted bivariate model, there was no difference in individual HMO composition, total HMO-bound fucose and HMO-bound sialic acid, or diversity and evenness scores by diet group. When adjusting for factors that significantly differed between groups (maternal BMI and lactation stage), no differences in HMO composition were observed. Secretor status was significant for 13 of the outcome variables with the strongest positive relationship with total HMO (β =22) and HMO-bound fucose (β =), and the strongest negative relationship with sialyl-lacto-N-tetraose b (LSTb) (β = 44). Lactation stage was significant for eight analytes, with the strongest positive impact on 3'sialyllactose (3'SL) HMO composition (β =13) and 3'SL (β =25). Conclusions Lactating women who consume plant-based diets do not produce different breast milk as it relates Efficient production of lacto-N-fucopentaose III in engineered Escherichia coli using α1,3-fucosyltransferase from Parabacteroides goldsteinii.Lacto-N-fucopentaose III (LNFP III) is a human milk oligosaccharide (HMO) with potential health benefits in infants, including in immune development and modulation of the intestinal environment.

Low-cost fermentative production of various HMOs from lactose by engineered Escherichia coli has attracted attention, but few reports have investigated long-chain HMO production, such as of the pentasaccharide LNFP III. LNFP III is synthesized by α1,3-fucosyltransfer reaction to the glucosamine (GlcNAc) moiety in the lacto-N-neotetraose (LNnT) skeleton by α1,3-fucosyltransferase (α1,3-FucT). However, the known α1,3-FucTs also transfer fucose to the reducing terminal glucose moiety of LNnT or the starting material lactose, resulting in various byproducts. Here, we found a useful α1,3-FucT from Parabacteroides goldsteinii (PgsFucT), which is only reactive for GlcNAc in the N-acetyllactosamine (LacNAc) skeleton in vivo. On the basis of sequence alignment with a FucT of known structure, we also generated α1,3-FucT variants with altered reactivity for LacNAc or lactose. An E. coli strain heterologously expressing PgsFucT accumulated 34 gL of LNFP III after 48 h of culture in a 3-L jar-fermenter.

lacto n neotetraose of various byproduct sugars were remarkably decreased compared with a strain expressing the previously characterized α1,3-fucT from Bacteroides fragilis.Conflict of interest statement Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported here.Investigations of the in vitro transport of human milk oligosaccharides by a Caco-2 monolayer using a novel high performance liquid chromatography-mass Complex lactose-derived oligosaccharides belong to the main components of human milk and are believed to exert multiple functions in the breast-fed infant. Therefore, we investigated the transepithelial transport of human milk oligosaccharides over Caco-2 monolayers. Main human milk oligosaccharides (HMOs) in the apical, basolateral, or intracellular compartment were separated by high performance liquid chromatography using a Hypercarb(TM) column and analyzed on line by mass spectrometry. This method allowed the identification and quantification of these components in intra- and extracellular fractions without prior purification. Using this technique we were able to show that acidic and neutral HMOs cross the epithelial barrier.

lacto n neotetraose of neutral, but not acidic, oligosaccharides was temperature-sensitive and partly inhibited by brefeldin A and bafilomycin A. Furthermore, net flux from the apical to the basolateral compartment was only observed for the neutral components. Similarly, apical cellular uptake was only found for neutral components but not for acidic oligosaccharides. Intracellular concentrations of neutral HMOs were significantly increased by inhibitors of transcytosis such as brefeldin A, N-ethylmaleimide, or bafilomycin A. The cellular uptake was saturable, and an apparent K(m) for lacto-N-fucopentaose I of 1 +- mmolliter and for lacto-N-tetraose of 1 +- mmolliter was determined. Furthermore, the uptake of lacto-N-fucopentaose I could be inhibited by the addition of the stereoisomer lacto-N-fucopentaose II but not by lacto-N-tetraose. These findings suggest that neutral HMOs are transported across the intestinal epithelium by receptor-mediated transcytosis as well as via paracellular pathways, whereas translocation of acidic HMOs solely represents paracellular flux.

Milk oligosaccharide profiles by reversed-phase HPLC of their perbenzoylated Human milk is rich in oligosaccharides, some of which inhibit toxins and pathogens involved in diseases of infants.