Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.905
Filtrar
1.
Nutrients ; 16(5)2024 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-38474771

RESUMO

Human milk oligosaccharides (HMOs) are a set of complex carbohydrates and the third largest solid component of human milk, after lactose and lipids. To date, over 150 HMOs have been identified and the diversity of structures produced by lactating women is influenced by maternal genetics as well as other maternal, infant, and environmental factors. While the concentrations of individual HMOs have been shown to vary between individuals and throughout the course of lactation, the variability of HMO concentration profiles following different pregnancies occurring in the same woman is presently unknown. As such, the objective of this study was to compare HMO concentrations in human milk samples provided by the same women (n = 34) following repeat pregnancies. We leveraged existing human milk samples and metadata from the UC San Diego Human Milk Research Biorepository (HMB) and measured the concentrations of the 19 most abundant HMOs using high-performance liquid chromatography with fluorescence detection (HPLC-FL). By assessing dissimilarities in HMO concentration profiles, as well as concentration trends in individual structures between pregnancies of each participant, we discovered that HMO profiles largely follow a highly personalized and predictable trajectory following different pregnancies irrespective of non-genetic influences. In conclusion, this is the first study to assess the interactions between parity and time following delivery on variations in HMO compositions.


Assuntos
Lactação , Leite Humano , Lactente , Gravidez , Humanos , Feminino , Leite Humano/química , Aleitamento Materno , Oligossacarídeos/análise , Cromatografia Líquida de Alta Pressão
2.
mSystems ; 9(4): e0029424, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38530054

RESUMO

Maternal secretor status is one of the determinants of human milk oligosaccharides (HMOs) composition, which, in turn, influences the gut microbiota composition of infants. To understand if this change in gut microbiota impacts immune cell composition, intestinal morphology, and gene expression, 21-day-old germ-free C57BL/6 mice were transplanted with fecal microbiota from infants whose mothers were either secretors (SMM) or non-secretors (NSM) or from infants consuming dairy-based formula (MFM). For each group, one set of mice was supplemented with HMOs. HMO supplementation did not significantly impact the microbiota diversity; however, SMM mice had a higher abundance of genus Bacteroides, Bifidobacterium, and Blautia, whereas, in the NSM group, there was a higher abundance of Akkermansia, Enterocloster, and Klebsiella. In MFM, gut microbiota was represented mainly by Parabacteroides, Ruminococcaceae_unclassified, and Clostrodium_sensu_stricto. In mesenteric lymph node, Foxp3+ T cells and innate lymphoid cells type 2 were increased in MFM mice supplemented with HMOs, while in the spleen, they were increased in SMM + HMOs mice. Similarly, serum immunoglobulin A was also elevated in MFM + HMOs group. Distinct global gene expression of the gut was observed in each microbiota group, which was enhanced with HMOs supplementation. Overall, our data show that distinct infant gut microbiota due to maternal secretor status or consumption of dairy-based formula and HMO supplementation impacts immune cell composition, antibody response, and intestinal gene expression in a mouse model. IMPORTANCE: Early life factors like neonatal diet modulate gut microbiota, which is important for the optimal gut and immune function. One such factor, human milk oligosaccharides (HMOs), the composition of which is determined by maternal secretor status, has a profound effect on infant gut microbiota. However, how the infant gut microbiota composition determined by maternal secretor status or consumption of infant formula devoid of HMOs impacts infant intestinal ammorphology, gene expression, and immune signature is not well explored. This study provides insights into the differential establishment of infant microbiota derived from infants fed by secretor or non-secretor mothers milk or those consuming infant formula and demonstrates that the secretor status of mothers promotes Bifidobacteria and Bacteroides sps. establishment. This study also shows that supplementation of pooled HMOs in mice changed immune cell composition in the spleen and mesenteric lymph nodes and immunoglobulins in circulation. Hence, this study highlights that maternal secretor status has a role in infant gut microbiota composition, and this, in turn, can impact host gut and immune system.


Assuntos
Imunidade Inata , Microbiota , Lactente , Feminino , Humanos , Animais , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos/metabolismo , Leite Humano/química , Sistema Imunitário/metabolismo , Oligossacarídeos/análise , Bifidobacterium/genética
3.
Sci Rep ; 14(1): 6730, 2024 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-38509153

RESUMO

Human milk oligosaccharides (HMOs) impact neonate immunity and health outcomes. However, the environmental factors influencing HMO composition remain understudied. This study examined the associations between ambient air pollutant (AAP) exposure and HMOs at 1-month postpartum. Human milk samples were collected at 1-month postpartum (n = 185). AAP (PM2.5, PM10, NO2) exposure included the 9-month pregnancy period through 1-month postpartum. Associations between AAP with (1) HMO diversity, (2) the sum of sialylated and fucosylated HMOs, (3) 6 a priori HMOs linked with infant health, and (4) all HMOs were examined using multivariable linear regression and principal component analysis (PCA). Exposure to AAP was associated with lower HMO diversity. PM2.5 and PM10 exposure was positively associated with the HMO 3-fucosyllactose (3FL); PM2.5 exposure was positively associated with the sum of total HMOs, sum of fucosylated HMOs, and the HMO 2'-fucosyllactose (2'FL). PCA indicated the PM2.5, PM10, and NO2 exposures were associated with HMO profiles. Individual models indicated that AAP exposure was associated with five additional HMOs (LNFP I, LNFP II, DFLNT, LNH). This is the first study to demonstrate associations between AAP and breast milk HMOs. Future longitudinal studies will help determine the long-term impact of AAP on human milk composition.


Assuntos
Poluição do Ar , Leite Humano , Lactente , Recém-Nascido , Gravidez , Feminino , Humanos , Leite Humano/química , Dióxido de Nitrogênio/análise , Oligossacarídeos/análise , Poluição do Ar/análise , Material Particulado
4.
Methods Mol Biol ; 2763: 37-44, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38347397

RESUMO

Since the core protein of mucin in the digesta of the stomach and small intestine, which is less affected by bacteria, remains intact, mucin content can be measured by enzyme-linked immunosorbent assay (ELISA). However, the mucin core protein in bacteria-rich colon digesta and feces is partially hydrolyzed by bacterial enzymes and not fully recognized by mucin antibodies, so mucin cannot be accurately quantified by ELISA. This method quantifies the glycan content linked to the mucin core protein and expresses mucin content in the colon digesta and feces as the equivalent of O-linked oligosaccharide chain. Although mucin glycans are also hydrolyzed by colonic bacteria, this method is a more accurate and simple way to measure mucin content in the digesta of the large intestine and feces than the ELISA method.


Assuntos
Mucinas , Roedores , Animais , Mucinas/metabolismo , Roedores/metabolismo , Oligossacarídeos/análise , Polissacarídeos/metabolismo , Bactérias/metabolismo , Fezes/microbiologia
5.
mSystems ; 9(3): e0071523, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38363147

RESUMO

Bifidobacterium longum subsp. infantis is a representative and dominant species in the infant gut and is considered a beneficial microbe. This organism displays multiple adaptations to thrive in the infant gut, regarded as a model for human milk oligosaccharides (HMOs) utilization. These carbohydrates are abundant in breast milk and include different molecules based on lactose. They contain fucose, sialic acid, and N-acetylglucosamine. Bifidobacterium metabolism is complex, and a systems view of relevant metabolic pathways and exchange metabolites during HMO consumption is missing. To address this limitation, a refined genome-scale network reconstruction of this bacterium is presented using a previous reconstruction of B. infantis ATCC 15967 as a template. The latter was expanded based on an extensive revision of genome annotations, current literature, and transcriptomic data integration. The metabolic reconstruction (iLR578) accounted for 578 genes, 1,047 reactions, and 924 metabolites. Starting from this reconstruction, we built context-specific genome-scale metabolic models using RNA-seq data from cultures growing in lactose and three HMOs. The models revealed notable differences in HMO metabolism depending on the functional characteristics of the substrates. Particularly, fucosyl-lactose showed a divergent metabolism due to a fucose moiety. High yields of lactate and acetate were predicted under growth rate maximization in all conditions, whereas formate, ethanol, and 1,2-propanediol were substantially lower. Similar results were also obtained under near-optimal growth on each substrate when varying the empirically observed acetate-to-lactate production ratio. Model predictions displayed reasonable agreement between central carbon metabolism fluxes and expression data across all conditions. Flux coupling analysis revealed additional connections between succinate exchange and arginine and sulfate metabolism and a strong coupling between central carbon reactions and adenine metabolism. More importantly, specific networks of coupled reactions under each carbon source were derived and analyzed. Overall, the presented network reconstruction constitutes a valuable platform for probing the metabolism of this prominent infant gut bifidobacteria.IMPORTANCEThis work presents a detailed reconstruction of the metabolism of Bifidobacterium longum subsp. infantis, a prominent member of the infant gut microbiome, providing a systems view of its metabolism of human milk oligosaccharides.


Assuntos
Fucose , Leite Humano , Lactente , Feminino , Humanos , Leite Humano/química , Fucose/análise , Lactose/análise , Oligossacarídeos/análise , Bifidobacterium/genética , Bifidobacterium longum subspecies infantis/metabolismo , Acetatos/análise , Carbono/análise , Lactatos/análise
6.
Microbiol Mol Biol Rev ; 88(1): e0009423, 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38206006

RESUMO

SUMMARYHuman milk oligosaccharides (HMOs) are complex, multi-functional glycans present in human breast milk. They represent an intricate mix of heterogeneous structures which reach the infant intestine in an intact form as they resist gastrointestinal digestion. Therefore, they confer a multitude of benefits, directly and/or indirectly, to the developing neonate. Certain bifidobacterial species, being among the earliest gut colonizers of breast-fed infants, have an adapted functional capacity to metabolize various HMO structures. This ability is typically observed in infant-associated bifidobacteria, as opposed to bifidobacteria associated with a mature microbiota. In recent years, information has been gleaned regarding how these infant-associated bifidobacteria as well as certain other taxa are able to assimilate HMOs, including the mechanistic strategies enabling their acquisition and consumption. Additionally, complex metabolic interactions occur between microbes facilitated by HMOs, including the utilization of breakdown products released from HMO degradation. Interest in HMO-mediated changes in microbial composition and function has been the focal point of numerous studies, in recent times fueled by the availability of individual biosynthetic HMOs, some of which are now commonly included in infant formula. In this review, we outline the main HMO assimilatory and catabolic strategies employed by infant-associated bifidobacteria, discuss other taxa that exhibit breast milk glycan degradation capacity, and cover HMO-supported cross-feeding interactions and related metabolites that have been described thus far.


Assuntos
Microbioma Gastrointestinal , Leite Humano , Recém-Nascido , Feminino , Humanos , Leite Humano/química , Bifidobacterium , Aleitamento Materno , Oligossacarídeos/análise , Oligossacarídeos/metabolismo
7.
Life Sci ; 339: 122420, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38218534

RESUMO

Human milk is the most valuable source of nutrition for infants. The structure and function of human milk oligosaccharides (HMOs), which are key components of human milk, have long been attracting particular research interest. Several recent studies have found HMOs to be efficacious in the prevention and treatment of necrotizing enterocolitis (NEC). Additionally, they could be developed in the future as non-invasive predictive markers for NEC. Based on previous findings and the well-defined functions of HMOs, we summarize potential protective mechanisms of HMOs against neonatal NEC, which include: modulating signal receptor function, promoting intestinal epithelial cell proliferation, reducing apoptosis, restoring intestinal blood perfusion, regulating microbial prosperity, and alleviating intestinal inflammation. HMOs supplementation has been demonstrated to be protective against NEC in both animal studies and clinical observations. This calls for mass production and use of HMOs in infant formula, necessitating more research into the safety of industrially produced HMOs and the appropriate dosage in infant formula.


Assuntos
Enterocolite Necrosante , Leite Humano , Lactente , Animais , Recém-Nascido , Humanos , Leite Humano/química , Enterocolite Necrosante/tratamento farmacológico , Enterocolite Necrosante/prevenção & controle , Intestinos , Proliferação de Células , Oligossacarídeos/farmacologia , Oligossacarídeos/uso terapêutico , Oligossacarídeos/análise
8.
Sci Rep ; 14(1): 1649, 2024 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-38238389

RESUMO

The development of a stable human gut microbiota occurs within the first year of life. Many open questions remain about how microfloral species are influenced by the composition of milk, in particular its content of human milk oligosaccharides (HMOs). The objective is to investigate the effect of the human HMO glycome on bacterial symbiosis and competition, based on the glycoside hydrolase (GH) enzyme activities known to be present in microbial species. We extracted from UniProt a list of all bacterial species catalysing glycoside hydrolase activities (EC 3.2.1.-), cross-referencing with the BRENDA database, and obtained a set of taxonomic lineages and CAZy family data. A set of 13 documented enzyme activities was selected and modelled within an enzyme simulator according to a method described previously in the context of biosynthesis. A diverse population of experimentally observed HMOs was fed to the simulator, and the enzymes matching specific bacterial species were recorded, based on their appearance of individual enzymes in the UniProt dataset. Pairs of bacterial species were identified that possessed complementary enzyme profiles enabling the digestion of the HMO glycome, from which potential symbioses could be inferred. Conversely, bacterial species having similar GH enzyme profiles were considered likely to be in competition for the same set of dietary HMOs within the gut of the newborn. We generated a set of putative biodegradative networks from the simulator output, which provides a visualisation of the ability of organisms to digest HMO and mucin-type O-glycans. B. bifidum, B. longum and C. perfringens species were predicted to have the most diverse GH activity and therefore to excel in their ability to digest these substrates. The expected cooperative role of Bifidobacteriales contrasts with the surprising capacities of the pathogen. These findings indicate that potential pathogens may associate in human gut based on their shared glycoside hydrolase digestive apparatus, and which, in the event of colonisation, might result in dysbiosis. The methods described can readily be adapted to other enzyme categories and species as well as being easily fine-tuneable if new degrading enzymes are identified and require inclusion in the model.


Assuntos
Bifidobacterium bifidum , Clostridium perfringens , Recém-Nascido , Humanos , Bifidobacterium , Mucinas/análise , Oligossacarídeos/análise , Leite Humano/química , Bactérias , Glicosídeo Hidrolases/análise , Digestão
9.
J Sep Sci ; 47(1): e2300705, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38095448

RESUMO

The sample preparation including labeling and clean-up represents a key analytical step in the analysis of oligosaccharides and glycans by either chromatographic or electrophoretic separation methods. Although the majority of labeling has been performed by neutral and/or negatively charged tags, the introduction of a positive charge into the saccharide molecule can significantly improve the analysis, especially with mass spectrometry detection. In this work, we present the evaluation of five solid-phase extraction sorbents differing in extraction chemistry for the clean-up and concentration of positively labeled maltooligosaccharides from the reaction mixtures. Maltooligosaccharides containing four to seven glucose units were labeled by cationic tags (2-aminoethyl)trimethylammonium chloride and (carboxymethyl)trimethylammonium chloride hydrazide and the extraction conditions were optimized followed by electrophoretic analysis with conductivity detection. The effects of the solid-phase extraction sorbent chemistry, extraction conditions, and sample composition are discussed. All tested sorbents were capable of cleaning up maltooligosaccharides from the reaction mixtures to some extent after optimization of the solid-phase extraction procedure (51.9%-98.9% recovery). The best-rated amide-based sorbent was used to process the sample of N-linked glycans enzymatically released from ribonuclease B.


Assuntos
Oligossacarídeos , Polissacarídeos , Oligossacarídeos/análise , Polissacarídeos/química , Eletroforese Capilar/métodos , Espectrometria de Massas , Extração em Fase Sólida
10.
Anal Chem ; 96(1): 163-169, 2024 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-38153380

RESUMO

Understanding the biological role of protein-linked glycans requires the reliable identification of glycans. Isomer separation and characterization often entail mass spectrometric detection preceded by high-performance chromatography on porous graphitic carbon. To this end, stable isotope-labeled glycans have emerged as powerful tools for retention time normalization. Hitherto, such standards were obtained by chemoenzymatic or purely enzymatic methods, which introduce, e.g., 13C-containing N-acetyl groups or galactose into native glycans. Glycan release with anhydrous hydrazine opens another route for heavy isotope introduction via concomitant de-N-acetylation. Here, we describe that de-N-acetylation can also be achieved with hydrazine hydrate, which is a more affordable and less hazardous reagent. Despite the slower reaction rate, complete conversion is achievable in 72 h at 100 °C for glycans with biantennary glycans with or without sialic acids. Shorter incubation times allow for the isolation of intermediate products with a defined degree of free amino groups, facilitating introduction of different numbers of heavy isotopes. Mass encoded glycans obtained by this versatile approach can serve a broad range of applications, e.g., as internal standards for isomer-specific studies of N-glycans, O-glycans, and human milk oligosaccharide by LC-MS on either porous graphitic carbon or─following permethylation─on reversed phase.


Assuntos
Grafite , Polissacarídeos , Humanos , Polissacarídeos/química , Espectrometria de Massas , Oligossacarídeos/análise , Carbono/química , Grafite/química , Isótopos
11.
J Agric Food Chem ; 72(1): 670-678, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38135877

RESUMO

Human milk is important for infant growth, and oligosaccharides are one of its main functional nutrients. To enable a systematic comparison of free oligosaccharide and glycoconjugate content in milk from different species, the phenol-sulfuric acid and resorcinol assays were combined to determine the content. Using real samples, the method revealed that human milk contained the highest amount of total, neutral (9.84 ± 0.31 g/L), and sialylated (3.21 ± 0.11 g/L) free oligosaccharides, followed by goat milk, with neutral (0.135 ± 0.015 g/L) and sialylated (0.192 ± 0.016 g/L) free oligosaccharides and at a distance by bovine and yak milk. The highest total glycoconjugate content was detected in yak milk (0.798 ± 0.011 g/L), followed by human, bovine, and goat milk. These findings suggest that goat milk is the best source of free oligosaccharides in infant formula and functional dairy products and yak milk is the best source of glycoconjugates.


Assuntos
Leite Humano , Leite , Lactente , Animais , Bovinos , Humanos , Leite/química , Leite Humano/química , Oligossacarídeos/análise , Glicoconjugados , Fórmulas Infantis/análise , Cabras
12.
Nutrients ; 15(21)2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37960297

RESUMO

Premature infants, given their limited reserves, heightened energy requirements, and susceptibility to nutritional deficits, require specialized care. AIM: To examine the complex interplay between nutrition and neurodevelopment in premature infants, underscoring the critical need for tailored nutritional approaches to support optimal brain growth and function. DATA SOURCES: PubMed and MeSH and keywords: preterm, early nutrition, macronutrients, micronutrients, human milk, human milk oligosaccharides, probiotics AND neurodevelopment or neurodevelopment outcomes. Recent articles were selected according to the authors' judgment of their relevance. Specific nutrients, including macro (amino acids, glucose, and lipids) and micronutrients, play an important role in promoting neurodevelopment. Early and aggressive nutrition has shown promise, as has recognizing glucose as the primary energy source for the developing brain. Long-chain polyunsaturated fatty acids, such as DHA, contribute to brain maturation, while the benefits of human milk, human milk oligosaccharides, and probiotics on neurodevelopment via the gut-brain axis are explored. This intricate interplay between the gut microbiota and the central nervous system highlights human milk oligosaccharides' role in early brain maturation. CONCLUSIONS: Individualized nutritional approaches and comprehensive nutrient strategies are paramount to enhancing neurodevelopment in premature infants, underscoring human milk's potential as the gold standard of nutrition for preterm infants.


Assuntos
Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido Prematuro , Lactente , Feminino , Recém-Nascido , Humanos , Leite Humano/química , Ácidos Graxos/análise , Micronutrientes/análise , Oligossacarídeos/análise , Glucose/análise
13.
Food Res Int ; 174(Pt 1): 113589, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37986455

RESUMO

Human milk is considered the optimal food for infants with abundant nutrients and bioactive components, which play key roles in infant health and development. Infant formulas represent appropriate substitutes for human milk. There are many brands of infant formula with different ingredient sources and functions on the market. The present study aims to quantify important bioactive components, i.e., milk oligosaccharides (MOS), sialic acids (Sia) and corticosteroids, in different infant formulas and compare these to human milk. In total, 12 different infant formulas available on the Dutch market were analyzed in this study. The concentrations of MOS and Sia were characterized by UHPLC-FLD and LC-MS, while corticosteroids were determined using established UHPLC-MS/MS methods. Among infant formulas, 15 structures of oligosaccharides were identified, of which 2'-Fucosyllactose (2'FL), 3'-Galactosyllactose (3'GL) and 6'-Galactosyllactose (6́'GL) were found in all infant formulas. The oligosaccharide concentrations differed between milk source and brands and were 3-5 times lower than in human milk. All infant formulas contained Sia, N-acetylneuraminic acid (Neu5Ac) was dominant in bovine milk-based formulas, while N-glycolylneuraminic acid (Neu5Gc) was major in goat milk-based formula. All infant formulas contained corticosteroids, yet, at lower concentrations than human milk. Insight in concentrations of bioactive components in infant formula compared to human milk may give direction to dietary advices and/or novel formula design.


Assuntos
Fórmulas Infantis , Ácidos Siálicos , Lactente , Humanos , Fórmulas Infantis/química , Ácidos Siálicos/análise , Espectrometria de Massas em Tandem , Leite Humano/química , Oligossacarídeos/análise , Corticosteroides/análise
14.
J Agric Food Chem ; 71(43): 16102-16113, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37856320

RESUMO

Four human milk oligosaccharides (HMOs), 3'-sialyllactose (3'-SL), 6'-sialyllactose (6'-SL), 2'-fucosyllactose (2'-FL), and 3-fucosyllactose (3-FL), were assessed for their possible antiviral activity against the SARS-CoV-2 spike receptor binding domain (RBD) in vitro. Among them, only 2'-FL/3-FL exhibited obvious antibinding activity against direct binding and trans-binding in competitive immunocytochemistry and enzyme-linked immunosorbent assays. The antiviral effects of 2'-FL/3-FL were further confirmed by pseudoviral assays with three SARS-Cov-2 mutants, with a stronger inhibition effect of 2'-FL than 3-FL. Then, 2'-FL/3-FL were studied with molecular docking and microscale thermophoresis analysis, showing that the binding sites of 2'-FL on RBD were involved in receptor binding, in addition to a tighter bond between them, thus enabling 2'-FL to be more effective than 3-FL. Moreover, the immunomodulation effect of 2'-FL was preliminary evaluated and confirmed in a human alveolus chip. These results would open up possible applications of 2'-FL for the prevention of SARS-CoV-2 infections by competitive binding inhibition.


Assuntos
COVID-19 , Leite Humano , Humanos , Leite Humano/química , SARS-CoV-2 , Simulação de Acoplamento Molecular , Oligossacarídeos/farmacologia , Oligossacarídeos/análise , Antivirais/farmacologia
15.
FEMS Microbiol Rev ; 47(6)2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37793834

RESUMO

A number of bacterial species are found in high abundance in the faeces of healthy breast-fed infants, an occurrence that is understood to be, at least in part, due to the ability of these bacteria to metabolize human milk oligosaccharides (HMOs). HMOs are the third most abundant component of human milk after lactose and lipids, and represent complex sugars which possess unique structural diversity and are resistant to infant gastrointestinal digestion. Thus, these sugars reach the infant distal intestine intact, thereby serving as a fermentable substrate for specific intestinal microbes, including Firmicutes, Proteobacteria, and especially infant-associated Bifidobacterium spp. which help to shape the infant gut microbiome. Bacteria utilising HMOs are equipped with genes associated with their degradation and a number of carbohydrate-active enzymes known as glycoside hydrolase enzymes have been identified in the infant gut, which supports this hypothesis. The resulting degraded HMOs can also be used as growth substrates for other infant gut bacteria present in a microbe-microbe interaction known as 'cross-feeding'. This review describes the current knowledge on HMO metabolism by particular infant gut-associated bacteria, many of which are currently used as commercial probiotics, including the distinct strategies employed by individual species for HMO utilisation.


Assuntos
Microbioma Gastrointestinal , Leite Humano , Lactente , Humanos , Leite Humano/química , Leite Humano/metabolismo , Oligossacarídeos/análise , Oligossacarídeos/metabolismo , Bactérias/genética , Bactérias/metabolismo , Açúcares/análise , Açúcares/metabolismo
16.
Nutrients ; 15(14)2023 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-37513505

RESUMO

Breastmilk is the optimal source of infant nutrition, with short-term and long-term health benefits. Some of these benefits are mediated by human milk oligosaccharides (HMOs), a unique group of carbohydrates representing the third most abundant solid component of human milk. We performed the first clinical study on infant formula supplemented with five different HMOs (5HMO-mix), comprising 2'-fucosyllactose, 3-fucosyllactose, lacto-N-tetraose, 3'-sialyllactose and 6'-sialyllactose at a natural total concentration of 5.75 g/L, and here report the analysis of the infant fecal microbiome. We found an increase in the relative abundance of bifidobacteria in the 5HMO-mix cohort compared with the formula-fed control, specifically affecting bifidobacteria that can produce aromatic lactic acids. 5HMO-mix influenced the microbial composition as early as Week 1, and the observed changes persisted to at least Week 16, including a relative decrease in species with opportunistic pathogenic strains down to the level observed in breastfed infants during the first 4 weeks. We further analyzed the functional potential of the microbiome and observed features shared between 5HMO-mix-supplemented and breastfed infants, such as a relative enrichment in mucus and tyrosine degradation, with the latter possibly being linked to the aromatic lactic acids. The 5HMO-mix supplement, therefore, shifts the infant fecal microbiome closer to that of breastfed infants.


Assuntos
Aleitamento Materno , Microbiota , Humanos , Lactente , Feminino , Leite Humano/química , Fórmulas Infantis/análise , Oligossacarídeos/análise
17.
Trends Immunol ; 44(8): 644-661, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37438187

RESUMO

Childhood allergy, including asthma, eczema, and food allergies, is a major global health burden, with prevalence increasing dramatically and novel interventions needed. Emerging research suggests that human milk oligosaccharides (HMOs), complex glycans found in breastmilk, have allergy-protective properties, indicating exciting therapeutic potential. This review evaluates current literature on the role of HMOs in allergy, assesses underlying immunological mechanisms, and discusses future research needed to translate findings into clinical implications. HMOs may mediate allergy risk through multiple structure-specific mechanisms, including microbiome modification, intestinal barrier maturation, immunomodulation, and gene regulation. Findings emphasize the importance of breastfeeding encouragement and HMO-supplemented formula milk for high allergy-risk infants. Although further investigation is necessary to determine the most efficacious structures against varying allergy phenotypes and their long-term efficacy, HMOs may represent a promising complementary tool for childhood allergy prevention.


Assuntos
Hipersensibilidade Alimentar , Leite Humano , Lactente , Feminino , Humanos , Criança , Fórmulas Infantis/química , Hipersensibilidade Alimentar/prevenção & controle , Aleitamento Materno , Oligossacarídeos/uso terapêutico , Oligossacarídeos/análise
18.
J Nutr ; 153(8): 2249-2262, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37348760

RESUMO

BACKGROUND: Early intestinal development is important to infant vitality, and optimal formula composition can promote gut health. OBJECTIVES: The objectives were to evaluate the effects of arachidonate (ARA) and/or prebiotic oligosaccharide (PRE) supplementation in formula on the development of the microbial ecosystem and colonic health parameters. METHODS: Newborn piglets were fed 4 formulas containing ARA [0.5 compared with 2.5% of dietary fatty acids (FAs)] and PRE (0 compared with 8 g/L, containing a 1:1 mixture of galactooligosaccharides and polydextrose) in a 2 x 2 factorial design for 22 d. Fecal samples were collected weekly and analyzed for relative microbial abundance. Intestinal samples were collected on day 22 and analyzed for mucosal FAs, pH, and short-chain FAs (SCFAs). RESULTS: PRE supplementation significantly increased genera within Bacteroidetes and Firmicutes, including Anaerostipes, Mitsuokella, Prevotella, Clostridium IV, and Bulleidia, and resulted in progressive separation from controls as determined by Principal Coordinates Analysis. Concentrations of SCFA increased from 70.98 to 87.37 mM, with an accompanying reduction in colonic pH. ARA supplementation increased the ARA content of the colonic mucosa from 2.35-5.34% of total FAs. PRE supplementation also altered mucosal FA composition, resulting in increased linoleic acid (11.52-16.33% of total FAs) and ARA (2.35-5.16% of total FAs). CONCLUSIONS: Prebiotic supplementation during the first 22 d of life altered the gut microbiota of piglets and increased the abundance of specific bacterial genera. These changes correlated with increased SCFA, which may benefit intestinal development. Although dietary ARA did not alter the microbiota, it increased the ARA content of the colonic mucosa, which may support intestinal development and epithelial repair. Prebiotic supplementation also increased unsaturation of FAs in the colonic mucosa. Although the mechanism requires further investigation, it may be related to altered microbial ecology or biohydrogenation of FA.


Assuntos
Microbiota , Prebióticos , Animais , Suínos , Oligossacarídeos/farmacologia , Oligossacarídeos/análise , Fezes/microbiologia , Mucosa Intestinal , Lipídeos
19.
Nutrients ; 15(11)2023 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-37299512

RESUMO

Human milk oligosaccharides (HMOs) stimulate the growth of gut commensals, prevent the adhesion of enteropathogens and modulate host immunity. The major factors influencing variations in the HMO profile are polymorphisms in the secretor (Se) or Lewis (Le) gene, which affect the activity of the enzymes fucoslytransferase 2 and 3 (FUT2 and FUT3) that lead to the formation of four major fucosylated and non-fucosylated oligosaccharides (OS). This pilot study aimed to determine the HMO profile of Israeli breastfeeding mothers of 16 term and 4 preterm infants, from a single tertiary center in the Tel Aviv area. Fifty-two human milk samples were collected from 20 mothers at three-time points: colostrum, transitional milk and mature milk. The concentrations of nine HMOs were assessed using liquid chromatography coupled with mass spectra chromatograms. Fifty-five percent of the mothers were secretors and 45% were non-secretors. Infant sex affected HMO levels depending on the maternal secretor status. Secretor mothers to boys had higher levels of FUT2-dependent OS and higher levels of disialyllacto-N-tetraose in the milk of mothers to girls, whereas non-secretor mothers to girls had higher levels of 3'-sialyllactose. In addition, the season at which the human milk samples were obtained affected the levels of some HMOs, resulting in significantly lower levels in the summer. Our findings provide novel information on the irregularity in the HMO profile among Israeli lactating women and identify several factors contributing to this variability.


Assuntos
Lactação , Leite Humano , Lactente , Masculino , Humanos , Recém-Nascido , Feminino , Leite Humano/química , Aleitamento Materno , Projetos Piloto , Israel , Recém-Nascido Prematuro , Oligossacarídeos/análise
20.
J Nutr ; 153(7): 2117-2124, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37149285

RESUMO

BACKGROUND: Understanding how human milk impacts growth requires valid analytical methods for quantifying the composition. Lactose, the most abundant constituent in human milk and a predominant source of energy, is often assessed using methods borrowed from the bovine dairy industry. However, the carbohydrate matrices of bovine and human milk are quite different, especially as they relate to human milk oligosaccharides (HMOs), each with a terminal lactose unit that may influence analytical methods. OBJECTIVES: Our goals were to determine the extent to which HMOs influence common analytical methods for measuring carbohydrates in human milk and to compare common methods for measuring lactose. METHODS: Two sets of experiments were performed. In the first set, native and HMO-spiked human milk samples (n = 16 each) were assessed and compared using 4 methods: AOAC 2006.06 (based on the Megazyme enzymatic assay), BioVision enzymatic assay, ultraperformance LC with MS, and infrared analysis. In the second set, human milk samples (n = 20) were assessed using 2 methods approved for measuring lactose in bovine milk: AOAC 984.22 that uses high-performance LC and refractive index detection and AOAC 2006.06 prepared using both volume and weighted dilutions. RESULTS: Native and HMO-spiked samples were not significantly different in lactose using AOAC 2006.06 and ultraperformance LC with MS but were significantly different using BioVision (mean difference = 0.2 g/dL; 95% CI: 0.1, 0.4; P = 0.005). Total carbohydrate measurements assessed using infrared were also higher after HMO spiking (mean difference = 0.4 g/dL; 95% CI: 0.3, 0.6; P < 0.001). Only AOAC methods 984.22 and 2006.06 for measuring lactose were very highly correlated (r > 0.90, P < 0.001). CONCLUSIONS: AOAC methods 984.22 and 2006.06 are comparable for measuring lactose in human milk and are not influenced by HMOs. HMOs influence other enzymatic methods as well as infrared analysis, which leads to an overestimate of energy values. J Nutr 2023;x:xx.


Assuntos
Lactose , Leite Humano , Humanos , Leite Humano/química , Oligossacarídeos/análise , Carboidratos da Dieta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...