Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Carbohydr Polym ; 259: 117734, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33673995

ABSTRACT

Oligosaccharides are one of the most important components in mammalian milk. Milk oligosaccharides can promote colonization of gut microbiota and protect newborns from infections. The diversity and structures of MOs differ among mammalian species. MOs in human and farm animals have been well-documented. However, the knowledge on MOs in rat and mouse have been very limited even though they are the most-widely used models for studies of human physiology and disease. Herein, we use a high-sensitivity online solid-phase extraction and HILIC coupled with electrospray tandem mass spectrometry to analyze the acidic MOs in rat and mouse. Among the fifteen MOs identified, twelve were reported for the first time in rat and mouse together with two novel sulphated oligosaccharides. The complete list of acidic oligosaccharides present in rat and mouse milk is the baseline information of these animals and should contribute to biological/biomedical studies using rats and mice as models.


Subject(s)
Milk/metabolism , Oligosaccharides/analysis , Spectrometry, Mass, Electrospray Ionization , Animals , Chromatography, High Pressure Liquid , Hydrophobic and Hydrophilic Interactions , Mice , Oligosaccharides/isolation & purification , Rats , Solid Phase Extraction
2.
Front Microbiol ; 11: 1097, 2020.
Article in English | MEDLINE | ID: mdl-32528455

ABSTRACT

The fucosylated carbohydrate moieties on intestinal epithelial cells (IECs) are involved in the creation of an environmental niche for commensal and pathogenic bacteria. Core fucosylation catalyzed by fucosyltransferase 8 (Fut8) is the major fucosylation pattern on the N-glycans of the surface glycoproteins on IECs, however, the role of IECs core fucosylation during infection remains unclear. This study was conducted to investigate the interaction between IECs core fucosylation and gut microbiota, and the effects of this interaction on protecting Salmonella enterica subsp. enterica serovar Typhi (S. Typhi) infection. Firstly, the Fut8 +/+ and Fut8 +/- mice were infected with S. Typhi. The level of IECs core fucosylation and protein expression of intestinal mucosa were then detected by LCA blot and Western blot, respectively. The gut microbiota of Fut8 +/+ and Fut8 +/- mice before and after S. Typhi infection was assessed by 16S rRNA sequencing. Our results showed that core fucosylation was ubiquitous expressed on the intestinal mucosa of mice and had significant effects on their gut microbiota. Fut8+/- mice was more susceptive to S. Typhi infection than Fut8+/+ mice. Interestingly, infection of S. Typhi upregulated the core fucosylation level of IECs and increased the abundances of beneficial microorganisms such as Lactobacillus and Akkermansia spp. Further in vitro and in vivo studies demonstrated that Wnt/ß-catenin signaling pathway mediated the elevation of IECs core fucosylation level upon infection of S. Typhi. Taken together, our data in this study revealed that the IECs core fucosylation plays an important role in protecting against S. Typhi infection via up-regulating the biological antagonism of intestinal microbiota.

3.
Int J Clin Exp Pathol ; 13(3): 411-424, 2020.
Article in English | MEDLINE | ID: mdl-32269678

ABSTRACT

Sporothrix schenckii induced sporotrichosis has gained importance in recent years because of its worldwide prevalence. The dimorphic switching process is required for the pathogenesis of S. schenckii. Previously, we found that STE20-like protein kinase (SsSte20) was overexpressed in the early yeast stage, but not in the mycelial stage of S. schenckii, which suggested its involvement in morphogenesis of this fungal pathogen. It remains unclear, however, whether SsSte20 is essential for dimorphic switching of S. schenckii and what are its related genes. In this study, the function of SsSte20 was investigated using double-stranded RNA interference (dsRNAi) mediated by Agrobacterium tumefaciens. We evaluated its effects on normal asexual development, yeast-phase cell formation, and cell wall composition and integrity. In addition, by transcriptome analysis of the SsSte20 knockdown (SsSte20-i) mutant and the standard S. schenckii strain, we further investigated the genes and pathways that were affected by SsSte20. Our results showed that inactivation of SsSte20 significantly affected the growth and internal components of S. schenckii conidia and impaired the dimorphic switching process. RNA transcriptome analysis of the standard S. schenckii strain and the SsSte20-i mutant revealed that SsSte20 inhibition affected the genes that were not only involved in the biological process, but also in the cellular component, and the molecular functions of S. schenckii. It mainly affected the expression of iron/ion-binding transporter genes, oxidation-reduction-related genes, 1, 3-beta-glucosidase, and methylsterol monooxygenase, which are highly associated with environmental information processing and the biosynthesis of cell wall components. Overall, our research supports the claim that SsSte20 plays an essential role in the dimorphism of S. schenckii and affects its global transcriptome.

4.
FASEB J ; 34(3): 3715-3731, 2020 03.
Article in English | MEDLINE | ID: mdl-31944389

ABSTRACT

Gestational diabetes mellitus (GDM) is significantly associated with allergen sensitization in early childhood, and this may influence the gut microbiome and immune system of the children. In addition to mother-to-child transmission of microbes, milk glycans play a pivotal role in shaping the gut microbiome of infants. A previous study has demonstrated alterations in the major milk N-glycans of mothers with GDM. However, the impact of these changes on the gut microbiome and immune response of the neonates has yet to be studied. Here, we aimed to compare the glycosylation levels of various milk glycans between normal and GDM mice, and to characterize the intestinal microbiome and immune responses of the offspring after weaning. We found that GDM mouse milk contained significantly higher concentrations of fucosylated and sialylated N-glycans than control mice, but there was no difference in the concentration of milk oligosaccharides between the groups. The differences in milk N-glycans had direct effects on the intestinal microbiome of the offspring, which in turn affected their immune response upon challenge with ovalbumin (OVA), with disruptions in the Th1/Th2 and Th17/Treg cell balances. This study lays the foundation for further research and development of specific nutritional care for the offspring of GDM mothers.


Subject(s)
Diabetes, Gestational/metabolism , Diabetes, Gestational/microbiology , Gastrointestinal Microbiome/physiology , Milk/chemistry , Polysaccharides/metabolism , Animals , Bacteroides/physiology , Female , Male , Mice , Polymerase Chain Reaction , Pregnancy , RNA, Ribosomal, 16S/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
5.
mBio ; 10(2)2019 04 02.
Article in English | MEDLINE | ID: mdl-30940702

ABSTRACT

The maternal milk glycobiome is crucial for shaping the gut microbiota of infants. Although high core fucosylation catalyzed by fucosyltransferase 8 (Fut8) is a general feature of human milk glycoproteins, its role in the formation of a healthy microbiota has not been evaluated. In this study, we found that the core-fucosylated N-glycans in milk of Chinese mothers selectively promoted the colonization of specific gut microbial groups, such as Bifidobacterium spp. and Lactobacillus spp. in their breast-fed infants during lactation. Compared with Fut8+/+ (WT) mouse-fed neonates, the offspring fed by Fut8+/- maternal mice had a distinct gut microbial profile, which was featured by a significant reduction of Lactobacillus spp., Bacteroides spp., and Bifidobacterium spp. and increased abundance of members of the Lachnospiraceae NK4A136 group and Akkermansia spp. Moreover, these offspring mice showed a lower proportion of splenic CD19+ CD69+ B lymphocytes and attenuated humoral immune responses upon ovalbumin (OVA) immunization. In vitro studies demonstrated that the chemically synthesized core-fucosylated oligosaccharides possessed the ability to promote the growth of tested Bifidobacterium and Lactobacillus strains in minimal medium. The resulting L-fucose metabolites, lactate and 1,2-propanediol, could promote the activation of B cells via the B cell receptor (BCR)-mediated signaling pathway.IMPORTANCE This study provides novel evidence for the critical role of maternal milk protein glycosylation in shaping early-life gut microbiota and promoting B cell activation of neonates. The special core-fucosylated oligosaccharides might be promising prebiotics for the personalized nutrition of infants.


Subject(s)
B-Lymphocytes/immunology , Bifidobacterium/metabolism , Fucose/metabolism , Gastrointestinal Tract/immunology , Lactobacillus/metabolism , Milk, Human/chemistry , Polysaccharides/metabolism , Animals , China , Female , Gastrointestinal Tract/microbiology , Humans , Infant , Lymphocyte Activation , Mice , Polysaccharides/chemistry
6.
Nutrition ; 62: 63-73, 2019 06.
Article in English | MEDLINE | ID: mdl-30852460

ABSTRACT

OBJECTIVE: High fructose consumption exacerbates purine degradation and intestinal dysbiosis, which are closely related to the development of hyperuricemia. Probiotics are powerful weapons to combat metabolic disturbance and intestinal dysbiosis. Previously we isolated a Lactobacillus strain named DM9218 that could reduce the serum uric acid (UA) level by assimilating purine nucleosides. The present study aimed to evaluate the effects of DM9218 on high-fructose-induced hyperuricemia and to elucidate the underlying mechanisms. METHODS: Mice were fed a normal diet, a high-fructose diet, or high-fructose diet with DM9218. Metabolic parameters, fructose- and UA-related metabolites, and fecal microbiota were investigated. Whole-genome sequencing of strain DM9218 was also conducted. In addition, an inosine hydrolase from DM9218 was heterologously expressed in Escherichia coli, and its inosine-degrading activity was detected. RESULTS: Our results indicated that DM9218 could decrease serum UA level and hepatic xanthine oxidase activity in fructose-fed mice. It could protect against high-fructose-induced liver damage and retard UA accumulation by degrading inosine. The modulation effect of DM9218 on high-fructose-induced intestinal dysbiosis resulted in enhancement of intestinal barrier function and reduction of liver lipopolysaccharide, which was closely correlated with the down-regulation of inflammatory cytokine-stimulated xanthine oxidase expression and activity. CONCLUSIONS: Lactobacillus brevis DM9218 is a probiotic strain with the potential to ameliorate fructose-induced hyperuricemia.


Subject(s)
Dysbiosis/drug therapy , Fructose/administration & dosage , Gastrointestinal Microbiome/drug effects , Hyperuricemia/drug therapy , Inosine/metabolism , Levilactobacillus brevis , Animals , Diet/adverse effects , Diet/methods , Disease Models, Animal , Dysbiosis/metabolism , Dysbiosis/urine , Hyperuricemia/etiology , Hyperuricemia/urine , Inosine/urine , Intestines/drug effects , Intestines/microbiology , Male , Mice , Mice, Inbred BALB C , Probiotics
7.
mSystems ; 3(6)2018.
Article in English | MEDLINE | ID: mdl-30637338

ABSTRACT

The milk glycobiome has a significant impact on the gut microbiota of infants, which plays a pivotal role in health and development. Fucosylated human milk oligosaccharides (HMOs) and N-glycans on milk proteins are beneficial for the development of healthy gut microbiota, and the fucosylation levels of these glycans can be affected by the maternal fucosyltransferase 2 gene (FUT2). Here, we present results of longitudinal research on paired milk and stool samples from 56 Chinese mothers (CMs) and their breast-fed children. Changes of HMOs and fucosylated N-glycans in milk of CMs at different lactation stages were detected, which allowed characterization of the major differences in milk glycans and consequential effects on the gut microbiome of infants according to maternal FUT2 status. Significant differences in the abundance of total and fucosylated HMOs between secretor and nonsecretor CMs were noted, especially during early lactation. Despite a tendency toward decreasing milk protein concentrations, the fucosylation levels of milk N-glycans increased during late lactation. The changes in the levels of fucosylated HMOs and milk N-glycans were highly correlated with the growth of Bifidobacterium spp. and Lactobacillus spp. in the gut of infants during early and later lactation, respectively. Enriched expression of genes encoding glycoside hydrolases, glycosyl transferases, ATP-binding cassette (ABC) transporters, and permeases in infants fed by secretor CMs contributed to the promotion of these bacteria in infants. Our data highlight the important role of fucosylated milk glycans in shaping the gut microbiome of infants and provide a solid foundation for development of "personalized" nutrition for Chinese infants. IMPORTANCE Human milk glycans provide a broad range of carbon sources for gut microbes in infants. Levels of protein glycosylation in human milk vary during lactation and may also be affected by the stages of gestation and lactation and by the secretor status of the mother. This was the first study to evaluate systematically dynamic changes in human milk oligosaccharides and fucosylated N-glycans in the milk of Chinese mothers with different secretor statuses during 6 months of lactation. Given the unique single nucleotide polymorphism site (rs1047781, A385T) on the fucosyltransferase 2 gene among Chinese populations, our report provides a specific insight into the milk glycobiome of Chinese mothers, which may exert effects on the gut microbiota of infants that differ from findings from other study cohorts.

SELECTION OF CITATIONS
SEARCH DETAIL
...