Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Mammary Gland Biol Neoplasia ; 20(3-4): 149-58, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26227402

RESUMO

Breast milk is a multifunctional biofluid that provides nutrients along with highly diverse non-nutritive bioactive components such as antibodies, glycans, bacteria, and immunomodulatory proteins. Research over the past decade has confirmed the essential role of breast milk bioactives in the establishment a healthy intestinal microbiota within the infant. The intestinal microbiota of an exclusively breastfed baby is dominated by several species of Bifidobacteria - the most influential member of which is Bifidobacterium longum subspecies infantis (B. infantis) - and is referred to as the milk-oriented microbiome (MOM). MOM is associated with reduced risk of infection in infancy as well as a reduced risk of certain chronic illnesses in adulthood. Establishment and persistence of MOM is dependent on the selective digestion of complex sugar structures in breast milk that are otherwise indigestible to the infant by B. infantis and its relatives. This review focuses primarily on the influence of breast milk glycans and glycosylated proteins on the development of the intestinal microbiome, and how maternal phenotype may influence the development of MOM providing a framework to understand how variation in diet shapes a protective intestinal microbiome.


Assuntos
Bifidobacterium , Aleitamento Materno , Dieta , Intestinos/microbiologia , Microbiota , Leite Humano/metabolismo , Humanos , Imunoglobulinas , Leite Humano/imunologia , Polissacarídeos/metabolismo
2.
Annu Rev Nutr ; 34: 143-69, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24850388

RESUMO

In addition to providing complete postnatal nutrition, breast milk is a complex biofluid that delivers bioactive components for the growth and development of the intestinal and immune systems. Lactation is a unique opportunity to understand the role of diet in shaping the intestinal environment including the infant microbiome. Of considerable interest is the diversity and abundance of milk glycans that are energetically costly for the mammary gland to produce yet indigestible by infants. Milk glycans comprise free oligosaccharides, glycoproteins, glycopeptides, and glycolipids. Emerging technological advances are enabling more comprehensive, sensitive, and rapid analyses of these different classes of milk glycans. Understanding the impact of inter- and intraindividual glycan diversity on function is an important step toward interventions aimed at improving health and preventing disease. This review discusses the state of technology for glycan analysis and how specific structure-function knowledge is enhancing our understanding of early nutrition in the neonate.


Assuntos
Aleitamento Materno , Desenvolvimento Infantil , Leite Humano/metabolismo , Modelos Biológicos , Oligossacarídeos/metabolismo , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/imunologia , Bifidobacterium/metabolismo , Sequência de Carboidratos , Feminino , Glicolipídeos/análise , Glicolipídeos/química , Glicolipídeos/metabolismo , Glicopeptídeos/análise , Glicopeptídeos/química , Glicopeptídeos/metabolismo , Glicoproteínas/análise , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Imunidade Inata , Recém-Nascido , Mucosa Intestinal/crescimento & desenvolvimento , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lactação , Masculino , Proteínas do Leite/análise , Proteínas do Leite/química , Proteínas do Leite/metabolismo , Leite Humano/química , Oligossacarídeos/análise , Oligossacarídeos/química
3.
J Proteome Res ; 11(12): 6124-33, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23140396

RESUMO

Human milk oligosaccharides (HMOs), as an abundant and bioactive component of breast milk, work in many ways to promote the health of breast fed infants. The expression of HMOs has been shown to vary in accordance with Lewis blood type and secretor status, as women of different blood types differ in the expression of α1,2 fucosyltransferase (FUT2) and α1,3/4 fucosyltransferase (FUT3). In this study, HMOs were extracted from the milk of 60 women from The Gambia, Africa with various Lewis and secretor blood types. The HMOs were profiled using high resolution HPLC-Chip/TOF mass spectrometry. Notably, the amounts of fucosylation varied significantly between Le(a+b-) nonsecretors, Le(a-b+) and Le(a-b-) secretors, and Le(a-b-) nonsecretors. With higher frequency of expression of the recessive Lewis negative and nonsecretor phenotypes in West African populations, the HMO profiles of several milks from women of these phenotypes were examined, demonstrating decreased amounts of total oligosaccharide abundance and lower relative amounts of fucosylation. Also in this study, four specific fucosylated structures (2'FL, LNFP I, LDFT, and LNDFH I) were determined to be specific and sensitive glycan markers for rapidly determining secretor status without the need for serological testing.


Assuntos
Lactação/metabolismo , Leite Humano/química , Oligossacarídeos/análise , Biomarcadores/metabolismo , Cromatografia Líquida de Alta Pressão , Feminino , Gâmbia , Genótipo , Humanos , Antígenos do Grupo Sanguíneo de Lewis/genética , Espectrometria de Massas/métodos , Oligossacarídeos/metabolismo , Fenótipo , Sensibilidade e Especificidade , Trissacarídeos/análise , Trissacarídeos/metabolismo
4.
Sci Rep ; 11(1): 1472, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33479326

RESUMO

The gut microbiome plays an important role in early life, protecting newborns from enteric pathogens, promoting immune system development and providing key functions to the infant host. Currently, there are limited data to broadly assess the status of the US healthy infant gut microbiome. To address this gap, we performed a multi-state metagenomic survey and found high levels of bacteria associated with enteric inflammation (e.g. Escherichia, Klebsiella), antibiotic resistance genes, and signatures of dysbiosis, independent of location, age, and diet. Bifidobacterium were less abundant than generally expected and the species identified, including B. breve, B. longum and B. bifidum, had limited genetic capacity to metabolize human milk oligosaccharides (HMOs), while B. infantis strains with a complete capacity for HMOs utilization were found to be exceptionally rare. Considering microbiome composition and functional capacity, this survey revealed a previously unappreciated dysbiosis that is widespread in the contemporary US infant gut microbiome.


Assuntos
Bifidobacterium/genética , Microbioma Gastrointestinal , Metagenômica/métodos , Bifidobacterium/isolamento & purificação , DNA Bacteriano/química , DNA Bacteriano/metabolismo , Bases de Dados Factuais , Dieta , Farmacorresistência Bacteriana/genética , Disbiose , Fezes/microbiologia , Humanos , Lactente , Recém-Nascido , Leite Humano/metabolismo , Oligossacarídeos/metabolismo , Estados Unidos
5.
Appl Environ Microbiol ; 75(23): 7319-25, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19801485

RESUMO

Galacto-oligosaccharides (GOS) are versatile food ingredients that possess prebiotic properties. However, at present there is a lack of precise analytical methods to demonstrate specific GOS consumption by bifidobacteria. To better understand the role of GOS as prebiotics, purified GOS (pGOS) without disaccharides and monosaccharides was prepared and used in bacterial fermentation experiments. Growth curves showed that all bifidobacteria assayed utilized and grew on pGOS preparations. We used a novel mass spectrometry approach involving matrix-assisted laser desorption ionization-Fourier transform ion cyclotron resonance (MALDI-FTICR) to determine the composition of oligosaccharides in GOS syrup preparations. MALDI-FTICR analysis of spent fermentation media demonstrated that there was preferential consumption of selected pGOS species by different bifidobacteria. The approach described here demonstrates that MALDI-FTICR is a rapid-throughput tool for comprehensive profiling of oligosaccharides in GOS mixtures. In addition, the selective consumption of certain GOS species by different bifidobacteria suggests a means for targeting prebiotics to enrich select bifidobacterial species.


Assuntos
Bifidobacterium/metabolismo , Meios de Cultura/química , Oligossacarídeos/metabolismo , Bifidobacterium/química , Bifidobacterium/crescimento & desenvolvimento , Fermentação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos
6.
J Agric Food Chem ; 56(2): 618-26, 2008 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18088092

RESUMO

Human milk is a complex biological fluid that provides not only primary nourishment for infants but also protection against pathogens and influences their metabolic, immunologic, and even cognitive development. The presence of oligosaccharides in remarkable abundance in human milk has been associated to provide diverse biological functions including directing the development of an infant's intestinal microflora and immune system. Recent advances in analytical tools offer invaluable insights in understanding the specific functions and health benefits these biomolecules impart to infants. Oligosaccharides in human milk samples obtained from five different individual donors over the course of a 3 month lactation period were isolated and analyzed using HPLC-Chip/TOF-MS technology. The levels and compositions of oligosaccharides in human milk were investigated from five individual donors. Comparison of HPLC-Chip/TOF-MS oligosaccharides profiles revealed heterogeneity among multiple individuals with no significant variations at different stages of lactation within individual donors.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas , Técnicas Analíticas Microfluídicas , Leite Humano/química , Oligossacarídeos/análise , Feminino , Fucose/análise , Humanos , Lactação , Ácido N-Acetilneuramínico/análise , Fatores de Tempo
7.
J Agric Food Chem ; 55(22): 8914-9, 2007 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-17915960

RESUMO

The molecular basis by which human breast milk supports the development of a protective intestinal microbiome in infants is unknown. After lactose and lipids, human milk oligosaccharides (HMOs) are quantitatively the third largest and most diverse component of breast milk. In this work, glycomic profiling of HMO consumption by bifidobacteria using Fourier transform ion cyclotron resonance mass spectrometry reveals that one species, Bifidobacterium longum biovar infantis ATCC 15697, an isolate from the infant gut, preferentially consumes small mass oligosaccharides, representing 63.9% of the total HMOs available. These HMOs were detected in human breast milk at the onset and constantly through the first month of lactation by use of high performance liquid chromatography-chip time-of-flight mass spectrometry. Further characterization revealed that strain ATCC 15697 possesses both fucosidase and sialidase activities not present in the other tested strains. This work provides evidence that these small mass HMOs are selectively metabolized by select bifidobacterial strains and represent a potential new class of bioactive molecules functioning as prebiotics to facilitate a protective gut colonization in breast-fed newborns.


Assuntos
Bifidobacterium/metabolismo , Leite Humano/química , Oligossacarídeos/metabolismo , Polissacarídeos/metabolismo , Feminino , Humanos , Lactação , Especificidade da Espécie , Fatores de Tempo
8.
mSphere ; 2(6)2017.
Artigo em Inglês | MEDLINE | ID: mdl-29242832

RESUMO

Attempts to alter intestinal dysbiosis via administration of probiotics have consistently shown that colonization with the administered microbes is transient. This study sought to determine whether provision of an initial course of Bifidobacterium longum subsp. infantis (B. infantis) would lead to persistent colonization of the probiotic organism in breastfed infants. Mothers intending to breastfeed were recruited and provided with lactation support. One group of mothers fed B. infantis EVC001 to their infants from day 7 to day 28 of life (n = 34), and the second group did not administer any probiotic (n = 32). Fecal samples were collected during the first 60 postnatal days in both groups. Fecal samples were assessed by 16S rRNA gene sequencing, quantitative PCR, mass spectrometry, and endotoxin measurement. B. infantis-fed infants had significantly higher populations of fecal Bifidobacteriaceae, in particular B. infantis, while EVC001 was fed, and this difference persisted more than 30 days after EVC001 supplementation ceased. Fecal milk oligosaccharides were significantly lower in B. infantis EVC001-fed infants, demonstrating higher consumption of human milk oligosaccharides by B. infantis EVC001. Concentrations of acetate and lactate were significantly higher and fecal pH was significantly lower in infants fed EVC001, demonstrating alterations in intestinal fermentation. Infants colonized by Bifidobacteriaceae at high levels had 4-fold-lower fecal endotoxin levels, consistent with observed lower levels of Gram-negative Proteobacteria and Bacteroidetes. IMPORTANCE The gut microbiome in early life plays an important role for long-term health and is shaped in large part by diet. Probiotics may contribute to improvements in health, but they have not been shown to alter the community composition of the gut microbiome. Here, we found that breastfed infants could be stably colonized at high levels by provision of B. infantis EVC001, with significant changes to the overall microbiome composition persisting more than a month later, whether the infants were born vaginally or by caesarean section. This observation is consistent with previous studies demonstrating the capacity of this subspecies to utilize human milk glycans as a nutrient and underscores the importance of pairing a probiotic organism with a specific substrate. Colonization by B. infantis EVC001 resulted in significant changes to fecal microbiome composition and was associated with improvements in fecal biochemistry. The combination of human milk and an infant-associated Bifidobacterium sp. shows, for the first time, that durable changes to the human gut microbiome are possible and are associated with improved gut function.

9.
J Agric Food Chem ; 54(20): 7471-80, 2006 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-17002410

RESUMO

Oligosaccharides in human milk represent a group of bioactive molecules that have evolved to be an abundant and diverse component of human milk, even though they have no direct nutritive value to the infant. A recent hypothesis proposes that they could be substrates for the development of the intestinal microflora and the mucosal immune system. The inability to determine the exact composition of these oligosaccharides limits research and the ability to understand their biological functions. Oligosaccharides isolated from the lipids and proteins of individual human milk samples were analyzed by a combination of techniques including microchip liquid chromatography mass spectrometry (HPLC-Chip/MS) and matrix-assisted laser desorption/ionization Fourier transform ion cyclotron resonance mass spectrometry (MALDI-FT ICR MS). Accurate mass measurements obtained using an orthogonal time-of-flight (o-TOF) mass spectrometry provided oligosaccharide composition for approximately 200 individual molecular species. Comparison of HPLC-Chip/MS profiles from five different women revealed variations in milk oligosaccharide compositions. HPLC-Chip/MS profiling provides a method for routinely identifying milk oligosaccharides. Tandem MS in combination with exoglycosidase digestion provides unambiguous differentiation of structural isomers.


Assuntos
Leite Humano/química , Oligossacarídeos/análise , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Lipídeos/química , Espectrometria de Massas , Procedimentos Analíticos em Microchip , Proteínas do Leite/química , Estrutura Molecular , Oligossacarídeos/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
10.
Trials ; 17(1): 340, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27449926

RESUMO

BACKGROUND: The development of probiotics as therapies to cure or prevent disease lags far behind that of other investigational medications. Rigorously designed phase I clinical trials are nearly non-existent in the field of probiotic research, which is a contributing factor to this disparity. As a consequence, how to appropriately dose probiotics to study their efficacy is unknown. Herein we propose a novel phase I ascending dose trial of Bifidobacterium longum subsp. infantis (B. infantis) to identify the dose required to produce predominant gut colonisation in healthy breastfed infants at 6 weeks of age. METHODS/DESIGN: This is a parallel-group, placebo-controlled, randomised, double-blind ascending dose phase I clinical trial of dietary supplementation with B. infantis in healthy breastfed infants. The objective is to determine the pharmacologically effective dose (ED) of B. infantis required to produce predominant (>50 %) gut colonisation in breastfed infants at 6 weeks of age. Successively enrolled infant groups will be randomised to receive two doses of either B. infantis or placebo on days 7 and 14 of life. Stool samples will be used to characterise the gut microbiota at increasing doses of B. infantis. DISCUSSION: Probiotic supplementation has shown promising results for the treatment of a variety of ailments, but evidence-based dosing regimes are currently lacking. The ultimate goal of this trial is to establish a recommended starting dose of B. infantis for further efficacy-testing phase II trials designed to evaluate B. infantis for the prevention of atopic dermatitis and food allergies in at-risk children. TRIAL REGISTRATION: Clinicaltrials.gov # NCT02286999 , date of trial registration 23 October 2014.


Assuntos
Bifidobacterium longum subspecies infantis/crescimento & desenvolvimento , Aleitamento Materno , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Probióticos/uso terapêutico , Fatores Etários , California , Protocolos Clínicos , Método Duplo-Cego , Fezes/microbiologia , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Probióticos/efeitos adversos , Projetos de Pesquisa , Fatores de Tempo , Resultado do Tratamento
13.
Ann N Y Acad Sci ; 1190: 97-103, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20388140

RESUMO

With the progressive aging of the world's population, immunosenescence is rapidly becoming a clinical concern as it accounts for a higher incidence of severe infections and poor response to vaccines. To identify nutritional approaches that may counteract immunosenescence is of obvious importance in clinical practice. Dairy products in general and whey proteins in particular share the capacity to stimulate the immune system within the digestive tract while the antibody response to Streptococcus pneumoniae vaccine is a good marker of the immune function. We performed a controlled, randomized, double-blind pilot study to determine if an eight-week supplementation with whey protein (or soy protein used as control) could enhance the serum response to pneumococcal vaccine in healthy senior citizens. Out of 127 volunteers, 17 subjects were eligible and completed the study receiving the vaccine after four weeks of supplementation. Antibody levels were measured at baseline and the end of the study against 14 pneumococcal types and a detailed nutritional questionnaire was administered to all subjects. Subjects receiving whey protein manifested a serum response higher compared to the control soy supplementation against 12/14 bacterial types. In particular, whey led to a higher frequency of response to all four more virulent types (4, 9, 14, and 23). Calorie and protein intake data suggest a better nutritional status in the whey group. Whey protein supplementation is a promising supplement to stimulate the immune response to vaccine in senior citizens and possibly to counteract immunosenescence while larger studies are warranted.


Assuntos
Adjuvantes Imunológicos/farmacologia , Formação de Anticorpos/efeitos dos fármacos , Proteínas do Leite/farmacologia , Vacinas Pneumocócicas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Idoso , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas do Leite/administração & dosagem , Projetos Piloto , Placebos , Proteínas do Soro do Leite
14.
Microb Biotechnol ; 2(3): 333-42, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-21261928

RESUMO

Human milk contains approximately 200 complex oligosaccharides believed to stimulate the growth and establishment of a protective microbiota in the infant gut. The lack of scalable analytical techniques has hindered the measurement of bacterial metabolism of these and other complex prebiotic oligosaccharides. An in vitro, multi-strain, assay capable of measuring kinetics of bacterial growth and detailed oligosaccharide consumption analysis by FTICR-MS was developed and tested simultaneously on 12 bifidobacterial strains. For quantitative consumption, deuterated and reduced human milk oligosaccharide (HMO) standards were used. A custom software suite developed in house called Glycolyzer was used to process the large amounts of oligosaccharide mass spectra automatically with (13)C corrections based on de-isotoping protocols. High growth on HMOs was characteristic of Bifidobacterium longum biovar infantis strains, which consumed nearly all available substrates, while other bifidobacterial strains tested, B. longum bv. longum, B. adolescentis, B. breve and B. bifidum, showed low or only moderate growth ability. Total oligosaccharide consumption ranged from a high of 87% for B. infantis JCM 7009 to only 12% for B. adolescentis ATCC 15703. A detailed analysis of consumption glycoprofiles indicated strain-specific capabilities towards differential metabolism of milk oligosaccharides. This method overcomes previous limitations in the quantitative, multi-strain analysis of bacterial metabolism of HMOs and represents a novel approach towards understanding bacterial consumption of complex prebiotic oligosaccharides.


Assuntos
Bifidobacterium/metabolismo , Leite Humano/metabolismo , Oligossacarídeos/metabolismo , Bifidobacterium/química , Bifidobacterium/crescimento & desenvolvimento , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Humanos , Leite Humano/química , Modelos Biológicos , Oligossacarídeos/química , Software
15.
J Agric Food Chem ; 56(16): 7446-50, 2008 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-18656925

RESUMO

Human milk fat globules (HMFGs) are considered to constitute a triglyceride-rich source of fat and energy. However, milk contains lipid particles at different sizes ranging from tens of micrometers to less than 1 microm. In particular, the physical, chemical, and biological properties of submicron sized particles are poorly described. Individual HMFGs were analyzed using laser trapping confocal Raman spectroscopy, and their chemical signature was obtained and compared to 1, 5, and 10 microm globules. Significant differences in both lipid composition and relative lipid content were found between the classes of particles with different diameters. A strong Raman peak at 1742 cm(-1) corresponding to the triacylglycerol core was detected in the 5 and 10 microm diameter globules, whereas in the smaller HMFGs no detectable peak was found. In addition, the submicron particles produced Raman signals consistent with large quantities of unsaturated fatty acids. Moreover, cis and trans isomers of unsaturated fatty acids were found to be unequally distributed between large and small milk fat globules. Interestingly, trans unsaturated fatty acids were found only in 1 and 5 microm globules although more prominent in the 5 microm diameter range. This is the first evidence for size related differential lipid composition of various diameter classes of HMFGs. The results suggest that the milk fat globule size distribution determines milk lipid composition. In addition, large portions of the HMFGs are secreted into milk conspicuously not for fat delivery. Thus, small HMFGs may offer novel metabolic and nutritional functions.


Assuntos
Glicolipídeos/química , Glicoproteínas/química , Lipídeos/análise , Lipídeos/química , Humanos , Gotículas Lipídicas , Tamanho da Partícula , Análise Espectral Raman
16.
Nestle Nutr Workshop Ser Pediatr Program ; 62: 205-18; discussion 218-22, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18626202

RESUMO

Human milk contains a high concentration of diverse soluble oligosaccharides, carbohydrate polymers formed from a small number of monosaccharides. Novel methods combining liquid chromatography with high resolution mass spectrometry have identified approximately 200 unique oligosaccharides structures varying from 3 to 22 sugars. The increasing complexity of oligosaccharides follows the general pattern of mammalian evolution though the concentration and diversity of these structures in homo sapiens are strikingly. There is also diversity among human mothers in oligosaccharides. Milks from randomly selected mothers contain as few as 23 and as many as 130 different oligosaccharides. The functional implications of this diversity are not known. Despite the role of milk to serve as a sole nutrient source for mammalian infants, the oligosaccharides in milk are not digestible by human infants. This apparent paradox raises questions about the functions of these oligosaccharides and how their diverse molecular structures affect their functions. The nutritional function most attributed to milk oligosaccharides is to serve as prebiotics - a form of indigestible carbohydrate that is selectively fermented by desirable gut microflora. This function was tested by purifying human milk oligosaccharides and providing these as the sole carbon source to various intestinal bacteria. Indeed, the selectively of providing the complex mixture of oligosaccharides pooled from human milk samples is remarkable. Among a variety of Bifidobacteria tested only Bifidobacteria longum biovar infantis was able to grow extensively on human milk oligosaccharides as sole carbon source. The genomic sequence of this strain revealed approximately 700 genes that are unique to infantis, including a variety of co-regulated glycosidases, relative to other Bifidobacteria, implying a co-evolution of human milk oligosaccharides and the genetic capability of select intestinal bacteria to utilize them. The goal of ongoing research is to assign specific functions to the combined oligosaccharide-bacteria-host interactions that emerged from this evolutionary pressure.


Assuntos
Bactérias/metabolismo , Fenômenos Fisiológicos da Nutrição do Lactente/fisiologia , Intestinos/microbiologia , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Leite Humano/química , Oligossacarídeos/análise , Bactérias/crescimento & desenvolvimento , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Feminino , Fermentação , Humanos , Lactente , Recém-Nascido , Probióticos , Solubilidade
17.
Anal Chem ; 80(1): 159-65, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18044961

RESUMO

Inulin is a class of fructooligosaccharide (FOS) derived from plants, which is often used as a natural food ingredient. Inulin is currently used as an additive in baked goods, dairy products, infant formula, and dietary supplements as a result of its purported health-promoting properties. The growth of health-promoting lactobacilli and bifidobacteria is supported by FOS, giving it the classification of a prebiotic; however, its ability to selectivity stimulate only beneficial bacteria has not been demonstrated. In order to better understand the role of inulin and FOS as prebiotics, matrix-assisted laser desorption/ionization Fourier transform ion cyclotron resonance mass spectrometry has been used for qualitative and quantitative analysis on bacterial growth. A method using an internal standard has been developed to quantify the consumption of FOS by Bifidobacterium longum bv. infantis using a calibration curve. Due to the differential consumption of FOS, the calibration curve was modified to include intensity components for each polymer unit in order to achieve more accurate quantitation. The method described was designed to be more rapid, precise, and robust for quantitative analysis when compared to existing methods.


Assuntos
Ciclotrons , Análise de Fourier , Oligossacarídeos/análise , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Bifidobacterium/metabolismo , Calibragem , Fermentação , Insulina/análise , Óptica e Fotônica , Padrões de Referência
18.
Anal Biochem ; 361(1): 15-23, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17181994

RESUMO

Oligosaccharides are the third most abundant component in human milk. In the past decades, it became apparent that they would be able to protect against pathogens and participate in the development of the gut microflora for infants. However, their role in infants' nutrition and development remains poorly understood. To better understand this function, it is extremely important to have a quantitative tool for profiling oligosaccharides. In this article, we show the development of a method to quantitatively differentiate the relative amounts of oligosaccharides fermented by different intestinal bacteria. To determine the oligosaccharide consumption, bacteria were grown in a medium using human milk oligosaccharides (HMOs) as the only carbon source purified from breast milk and further analyzed by matrix-assisted laser desorption/ionization-Fourier transform ion cyclotron resonance mass spectrometry (MALDI-FTICR MS). A method using an internal deuterium-labeled standard was developed and compared with an external standard method, with the internal standard method giving better precision and unambiguous measurements than the external standard method and providing to be a novel and robust tool for following bacterial fermentation of milk oligosaccharides.


Assuntos
Leite Humano/química , Oligossacarídeos/análise , Fenômenos Fisiológicos Bacterianos , Bifidobacterium/fisiologia , Cromatografia Líquida de Alta Pressão , Fermentação , Espectrometria de Massas/métodos , Bancos de Leite Humano , Oligossacarídeos/química , Oligossacarídeos/isolamento & purificação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Estados Unidos
19.
Gastroenterology ; 131(1): 97-107, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16831594

RESUMO

BACKGROUND & AIMS: Release of 5-hydroxytryptamine (5-HT) from mucosal enterochromaffin cells and activation of 5-HT(3) receptors (5-HT(3)Rs) on neurons in the gut wall is important in the response of the gut to the luminal environment. Intestinal inflammation is associated with increased levels of mucosal 5-HT. The aims of the study were to determine the following: (1) if 5-HT(3)R undergoes ligand-induced internalization in myenteric neurons, and (2) the effect of long-term increase of mucosal 5-HT on 5-HT(3)Rs. METHODS: Acute effects of exogenous 5-HT or endogenous release of 5-HT by luminal glucose on cellular localization of 5-HT(3)Rs was determined by immunohistochemistry and confocal microscopy. Treatment with the serotonin re-uptake inhibitor, fluoxetine, for 6 days (20 mg/kg daily orally) was used to increase mucosal 5-HT chronically in rats. Net ileal fluid movement was measured in anesthetized rats by the weight change of a 2.5% agarose cylinder. RESULTS: Acute increases in 5-HT induced by exogenous or endogenous 5-HT decreased 5-HT(3)R immunoreactivity at the neuronal cell membrane by 70% and 60%, respectively. Chronic fluoxetine treatment increased mucosal levels of 5-HT and decreased membrane 5-HT(3)R immunoreactivity by 27%. Net fluid absorption was decreased by a 5-HT(3)R agonist or by luminal glucose; this was attenuated 88% and 99%, respectively, by fluoxetine treatment. CONCLUSIONS: Long-term increase in 5-HT in the intestinal mucosa results in increased 5-HT(3)R internalization in myenteric neurons. Chronic changes in mucosal 5-HT may alter gastrointestinal secretory and motor function via ongoing loss of receptor from neuronal membrane, causing a mismatch between luminal content and absorption.


Assuntos
Mucosa Intestinal/inervação , Plexo Mientérico/metabolismo , Neurônios/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo , Animais , Western Blotting , Fluoxetina/farmacologia , Íleo/inervação , Íleo/metabolismo , Imuno-Histoquímica , Absorção Intestinal/efeitos dos fármacos , Absorção Intestinal/fisiologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Plexo Mientérico/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ondansetron/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores 5-HT3 de Serotonina/efeitos dos fármacos , Antagonistas da Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa