Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Trials ; 25(1): 238, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38576033

RESUMO

BACKGROUND: Functional constipation (FC) in children is a common gastrointestinal disorder with a worldwide-pooled prevalence of 9.5%. Complaints include infrequent bowel movements, painful defecation due to hard and/or large stools, faecal incontinence, and abdominal pain. Prebiotic oligosaccharides have been shown to relieve constipation symptoms in young adults and elderly. However, sufficient evidence is lacking linking additional prebiotic intake to improve symptoms in children with FC. We hypothesise that prebiotic oligosaccharides are able to relieve symptoms of constipation in young children as well. METHODS: In the present randomised, double-blind, placebo-controlled, multi-centre study, we will study the effects of two prebiotic oligosaccharides in comparison to placebo on constipation symptoms in children of 1-5 years (12 to 72 months) of age diagnosed with FC according to the Rome IV criteria for functional gastrointestinal disorders. The primary outcome measure will be change in stool consistency. Secondary outcomes include stool frequency and stool consistency in a number of cases (%). Tertiary outcomes include among others painful defecation, use of rescue medication, and quality of life. In addition, the impact on gut microbiome outcomes such as faecal microbiota composition and metabolites will be investigated. Participants start with a run-in period, after which they will receive supplements delivered in tins with scoops for 8 weeks, containing one of the two prebiotic oligosaccharides or placebo, followed by a 4-week wash-out period. DISCUSSION: This randomised double-blind, placebo-controlled multi-centre study will investigate the effectiveness of prebiotic oligosaccharides in children aged 1-5 years with FC. TRIAL REGISTRATION: ClinicalTrials.gov NCT04282551. Registered on 24 February 2020.


Assuntos
Defecação , Microbioma Gastrointestinal , Criança , Adulto Jovem , Idoso , Humanos , Pré-Escolar , Prebióticos , Qualidade de Vida , Constipação Intestinal/diagnóstico , Constipação Intestinal/tratamento farmacológico , Oligossacarídeos/efeitos adversos , Hábitos , Método Duplo-Cego , Resultado do Tratamento , Ensaios Clínicos Controlados Aleatórios como Assunto , Estudos Multicêntricos como Assunto
2.
J Pediatr ; 261: 113559, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37331467

RESUMO

OBJECTIVE: To summarize available data on defecation frequency and stool consistency of healthy children up to age 4 in order to estimate normal references values. STUDY DESIGN: Systematic review including cross-sectional, observational, and interventional studies published in English, that reported on defecation frequency and/or stool consistency in healthy children 0-4 years old. RESULTS: Seventy-five studies were included with 16 393 children and 40 033 measurements of defecation frequency and/or stool consistency. Based on visual inspection of defecation frequency data, a differentiation was made between two age categories: young infants (0-14 weeks old) and young children (15 weeks-4 years old). Young infants had a mean defecation frequency of 21.8 per week (95 % CI, 3.9-35.2) compared with 10.9 (CI, 5.7-16.7) in young children (P < .001). Among young infants, human milk-fed (HMF) infants had the highest mean defecation frequency per week (23.2 [CI, 8.8-38.1]), followed by formula-fed (FF) infants (13.7 [CI 5.4-23.9]), and mixed-fed (MF) infants (20.7 [CI, 7.0-30.2]). Hard stools were infrequently reported in young infants (1.5%) compared with young children (10.5%), and a reduction in the frequency of soft/watery stools was observed with higher age (27.0% in young infants compared with 6.2% in young children). HMF young infants had softer stools compared with FF young infants. CONCLUSIONS: Young infants (0-14 weeks old) have softer and more frequent stools compared with young children (15 weeks-4 years old).


Assuntos
Defecação , Leite Humano , Lactente , Humanos , Criança , Pré-Escolar , Recém-Nascido , Estudos Transversais , Diarreia , Alimentos Formulados , Fezes
3.
J Pediatr Gastroenterol Nutr ; 73(2): 210-216, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34402810

RESUMO

OBJECTIVE: The aim of the study was to assess whether the modified Bristol Stool Form Scale (m-BSFS) is reliable, valid and user-friendly to use by parents, grandparents, and day childcare employees to evaluate stool consistency in toilet and nontoilet-trained toddlers in the Netherlands. STUDY DESIGN: Translation to Dutch and validity of the m-BSFS (scoring 32 general stool pictures) for 1 to 3 year old toddlers (n = 89) was evaluated by parents, grandparents, and day childcare employees. A subgroup of participants scored an additional 7 pictures of stools in a diaper to validate the m-BSFS for non-toilet-trained toddlers (n = 16). To determine inter-rater reliability, 2-way random effects single-rater intraclass correlation coefficient (ICC)consistency was used. Intra-rater reliability was measured by Cohen kappa (κ) by rating the same pictures in random order twice, with at least 1 week between the first and second scoring. RESULTS: Inter- and intra-rater reliability of the m-BSFS were above recommended minimal standards of 0.61 for the 32 general stool pictures as well as for the 7 pictures of stools in a diaper. ICCconsistency for the general stool pictures of the first and second ratings were 0.71 (n = 89) and 0.79 (n = 77), respectively, with a κ of 0.71 (n = 77). ICCconsistency for the stools in diaper pictures of the first and second ratings were 0.93 (n = 16) and 0.93 (n = 15), respectively, with a κ of 0.77 (n = 15). CONCLUSIONS: The m-BSFS is reliable, valid and user-friendly to use by Dutch-speaking parents, grandparents, and day childcare workers to evaluate stool consistency in both toilet- and nontoilet-trained toddlers in the Netherlands.


Assuntos
Aparelho Sanitário , Pré-Escolar , Fezes , Humanos , Lactente , Pais , Reprodutibilidade dos Testes , Irrigação Terapêutica
4.
Expert Rev Gastroenterol Hepatol ; 11(11): 1031-1045, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28737484

RESUMO

INTRODUCTION: The bacteria received upon birth are the start of colonization of the approximately 1014 bacteria that are present in the mature human gastrointestinal tract, better known as the microbiota. The gut microbiota is implicated in gastrointestinal health, nutrient metabolism and benefits such as prevention of infection. Dietary fiber, including prebiotics, escape digestion in the small intestine and reach the colon intact, where they are partially or completely fermented by the gut microbiota. Areas covered: The possible interactions between dietary fiber, prebiotics and microbiota are discussed as well as how this relates to functional gastrointestinal disorders. During the first years of life the microbiota have not yet reached a stable state and is sensitive to disturbance by environmental factors. An imbalance in the microbiota early in life is found to be associated with several functional gastrointestinal disorders such as colic, functional abdominal pain, irritable bowel syndrome and constipation. Expert commentary: A better understanding of how gut microbial changes in early-life can impact gastrointestinal health might lead to new treatments or disease prevention. Nutritional strategies with fiber or prebiotics may support health due to modification of colonic microbiota composition and metabolic activity, for example by growth stimulation of Bifidobacterium and Lactobacillus.


Assuntos
Fibras na Dieta/administração & dosagem , Gastroenteropatias/terapia , Microbioma Gastrointestinal , Intestinos/microbiologia , Prebióticos/administração & dosagem , Adolescente , Fenômenos Fisiológicos da Nutrição do Adolescente , Fatores Etários , Criança , Pré-Escolar , Fibras na Dieta/efeitos adversos , Fibras na Dieta/metabolismo , Digestão , Gastroenteropatias/epidemiologia , Gastroenteropatias/microbiologia , Gastroenteropatias/fisiopatologia , Humanos , Lactente , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido , Intestinos/fisiopatologia , Estado Nutricional , Prebióticos/efeitos adversos , Recomendações Nutricionais , Resultado do Tratamento
5.
Eur J Nutr ; 56(5): 1919-1930, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27295033

RESUMO

PURPOSE: The direct effects of galacto-oligosaccharides (GOS), including Vivinal® GOS syrup (VGOS) and purified Vivinal® GOS (PGOS), on the epithelial integrity and corresponding interleukin-8 (IL-8/CXCL8) release were examined in a Caco-2 cell model for intestinal barrier dysfunction. To investigate structure-activity relationships, the effects of individual DP fractions of VGOS were evaluated. Moreover, the obtained results with GOS were compared with Caco-2 monolayers incubated with fructo-oligosaccharides (FOS) and inulin. METHODS: Caco-2 monolayers were pretreated (24 h) with or without specific oligosaccharides or DP fractions of VGOS (DP2 to DP6) before being exposed for 12 or 24 h to the fungal toxin deoxynivalenol (DON). Transepithelial electrical resistance and lucifer yellow permeability were measured to investigate barrier integrity. A calcium switch assay was used to study the reassembly of tight junction proteins. Release of CXCL8, a typical marker for inflammation, was quantified by ELISA. RESULTS: In comparison with PGOS, FOS and inulin, VGOS showed the most pronounced protective effect on the DON-induced impairment of the monolayer integrity, acceleration of the tight junction reassembly and the subsequent CXCL8 release. DP2 and DP3 in concentrations occurring in VGOS prevented the DON-induced epithelial barrier disruption, which could be related to their high prevalence in VGOS. However, no effects of the separate DP GOS fractions were observed on CXCL8 release. CONCLUSIONS: This comparative study demonstrates the direct, microbiota-independent effects of oligosaccharides on the intestinal barrier function and shows the differences between individual galacto- and fructo-oligosaccharides. This microbiota-independent effect of oligosaccharides depends on the oligosaccharide structure, DP length and concentration.


Assuntos
Células Epiteliais/efeitos dos fármacos , Microbioma Gastrointestinal , Intestinos/citologia , Oligossacarídeos/farmacologia , Células CACO-2 , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Humanos , Interleucina-8/metabolismo , Intestinos/microbiologia , Inulina/farmacologia , Relação Estrutura-Atividade , Tricotecenos/toxicidade
6.
Br J Nutr ; 116(2): 294-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27212112

RESUMO

Necrotising enterocolitis (NEC) is one of the most frequent and fatal intestinal disorders in preterm infants and has very limited treatment options. Breast-fed infants are at a 6-10-fold lower NEC risk than formula-fed infants, and we have previously shown that human milk oligosaccharides (HMO) improved survival and reduced pathology in a rat NEC model. The HMO disialyllacto-N-tetraose (DSLNT) was most effective, and sialylation was shown to be essential for its protective effect. Galacto-oligosaccharides (GOS), currently added to some infant formula, but not containing sialic acid, had no effect. In addition to DSLNT, our previous work also showed that the neutral HMO fraction, which contains high concentrations of 2'-fucosyllactose (2'FL), slightly improved pathology scores. Here, we assessed the in vivo efficacy of 2'FL, as well as of GOS that we enzymatically sialylated (Sia-GOS). Neonatal rats were randomised into the following study groups - dam-fed (DF), formula-fed (FF), FF containing pooled HMO (10 mg/ml), GOS (8 mg/ml), Sia-GOS (500 µm) or 2'FL (2 mg/ml) - and subjected to the established NEC protocol. The DF and HMO groups had the lowest pathology scores with mean values of 0·67 (sd 0·34) and 0·90 (sd 0·47), respectively. The FF group had significantly elevated pathology scores of 2·02 (sd 0·63). Although the addition of GOS to the formula had no protective effect and generated scores of 2·00 (sd 0·63), the addition of Sia-GOS or 2'FL significantly lowered pathology scores to 1·32 (sd 0·56) (P<0·0034) and 1·43 (sd 0·51) (P<0·0040), respectively. The results warrant further studies to investigate the underlying mechanisms and to assess safety and efficacy in human neonates.


Assuntos
Enterocolite Necrosante/tratamento farmacológico , Galactose/uso terapêutico , Fórmulas Infantis/química , Leite Humano/química , Oligossacarídeos/uso terapêutico , Ácidos Siálicos/uso terapêutico , Trissacarídeos/uso terapêutico , Animais , Animais Recém-Nascidos , Aleitamento Materno , Feminino , Galactose/metabolismo , Galactose/farmacologia , Humanos , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido , Intestinos/efeitos dos fármacos , Intestinos/patologia , Oligossacarídeos/metabolismo , Oligossacarídeos/farmacologia , Distribuição Aleatória , Ratos Sprague-Dawley , Ácidos Siálicos/metabolismo , Ácidos Siálicos/farmacologia , Trissacarídeos/farmacologia
7.
J Agric Food Chem ; 63(50): 10862-72, 2015 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-26621571

RESUMO

Human milk oligosaccharides (HMOs) are absorbed into the blood (about 1% of the HMO intake) and subsequently excreted in urine, where they may protect the infant from pathogen infection. As dietary galacto-oligosaccharides (GOS) have partial structural similarities with HMOs, this study investigated the presence of GOS and oligosaccharides originating from milk replacer in blood serum, urine, and cecal and fecal samples of piglets, as a model for human infants. Using liquid chromatography-mass spectrometry and capillary electrophoresis with fluorescence detection, oligosaccharides originating from piglet diet including 3'-sialyllactose and specific GOS ranging from degree of polymerization 3 to 6 were detected in blood serum and in urine of piglets. In blood serum, GOS levels ranged from 16 to 23 µg/mL, representing about 0.1% of the GOS daily intake. In urine, approximately 0.85 g of GOS/g of creatinine was found. Cecum digesta and feces contained low amounts of oligosaccharides, suggesting an extensive GOS intestinal fermentation in piglets.


Assuntos
Fezes/química , Galactose/farmacocinética , Substitutos do Leite/química , Oligossacarídeos/farmacocinética , Sus scrofa , Animais , Dieta/veterinária , Fermentação , Galactose/administração & dosagem , Galactose/análise , Absorção Intestinal , Intestino Delgado/metabolismo , Oligossacarídeos/sangue , Oligossacarídeos/urina
8.
J Nutr ; 145(7): 1604-13, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26019243

RESUMO

BACKGROUND: The integrity of the epithelial layer in the gastrointestinal tract protects organisms from exposure to luminal antigens, which are considered the primary cause of chronic intestinal inflammation and allergic responses. The common wheat-associated fungal toxin deoxynivalenol acts as a specific disruptor of the intestinal tight junction network and hence might contribute to the pathogenesis of inflammatory bowel diseases. OBJECTIVE: The aim of the current study was to assess whether defined galacto-oligosaccharides (GOSs) can prevent deoxynivalenol-induced epithelial dysfunction. METHODS: Human epithelial intestinal Caco-2 cells, pretreated with different concentrations of GOSs (0.5%, 1%, and 2%) for 24 h, were stimulated with 4.2-µM deoxynivalenol (24 h), and 6/7-wk-old male B6C3F1 mice were fed a diet supplemented with 1% GOSs for 2 wk before being orally exposed to deoxynivalenol (25 mg/kg body weight, 6 h). Barrier integrity was determined by measuring transepithelial electrical resistance (TEER) and intestinal permeability to marker molecules. A calcium switch assay was conducted to study the assembly of epithelial tight junction proteins. Alterations in tight junction and cytokine expression were assessed by quantitative reverse transcriptase-polymerase chain reaction, Western blot analysis, or ELISA, and their localization was visualized by immunofluorescence microscopy. Sections of the proximal and distal small intestine were stained with hematoxylin/eosin for histomorphometric analysis. RESULTS: The in vitro data showed that medium supplemented with 2% GOSs improved tight junction assembly reaching an acceleration of 85% after 6 h (P < 0.05). In turn, GOSs prevented the deoxynivalenol-induced loss of epithelial barrier function as measured by TEER (114% of control), and paracellular flux of Lucifer yellow (82.7% of prechallenge values, P < 0.05). Moreover, GOSs stabilized the expression and cellular distribution of claudin3 and suppressed by >50% the deoxynivalenol-induced synthesis and release of interleukin-8 [IL8/chemokine CXC motif ligand (CXCL8)] (P < 0.05). In mice, GOSs prevented the deoxynivalenol-induced mRNA overexpression of claudin3 (P = 0.022) and CXCL8 homolog keratinocyte hemoattractant (Kc) (Cxcl1) (P = 0.06) as well as the deoxynivalenol-induced morphologic defects. CONCLUSIONS: The results demonstrate that GOSs stimulate the tight junction assembly and in turn mitigate the deleterious effects of deoxynivalenol on the intestinal barrier of Caco-2 cells and on villus architecture of B6C3F1 mice.


Assuntos
Oligossacarídeos/farmacologia , Junções Íntimas/efeitos dos fármacos , Tricotecenos/toxicidade , Animais , Células CACO-2 , Claudina-3/genética , Claudina-3/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/etiologia , Interleucina-8/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Permeabilidade , Junções Íntimas/metabolismo
9.
Nutr Rev ; 72(6): 377-89, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24828428

RESUMO

Human milk is a rich source of oligosaccharides. Acidic oligosaccharides, such as sialyllactose (SL), contain sialic acid (SA) residues. In human milk, approximately 73% of SA is bound to oligosaccharides, whereas only 3% is present in free form. Oligosaccharides are highly resistant to hydrolysis in the gastrointestinal tract. Only a small portion of the available oligosaccharides in breast milk is absorbed in the neonatal small intestine. SL and sialylated oligosaccharides are thought to have significant health benefits for the neonate, because of their roles in supporting resistance to pathogens, gut maturation, immune function, and cognitive development. The need for SA to allow proper development during the neonatal period is thought to exceed the endogenous synthesis. Therefore, these structures are important nutrients for the neonate. Based on the potential benefits, SL and sialylated oligosaccharides may be interesting components for application in infant nutrition. Once the hurdle of limited availability of these oligosaccharides has been overcome, their functionality can be explored in more detail, and supplementation of infant formula may become feasible.


Assuntos
Dieta , Trato Gastrointestinal/metabolismo , Fenômenos Fisiológicos da Nutrição do Lactente , Lactose/análogos & derivados , Leite Humano/química , Necessidades Nutricionais , Oligossacarídeos/metabolismo , Ácidos Siálicos/metabolismo , Animais , Aleitamento Materno , Suplementos Nutricionais , Humanos , Lactente , Fórmulas Infantis/química , Lactose/metabolismo
10.
FEMS Microbiol Ecol ; 87(1): 41-51, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23909489

RESUMO

Prebiotics are considered to have potential to reduce disturbances in the gut microbiota induced by antibiotics. Results in literature are, however, not consistent. The current in vitro study conducted in a fermentation screening platform allowed to unambiguously compare the impact of galacto-oligosaccharides (GOS) on adult gut microbiota composition and activity upon treatment with four antibiotics at two doses. The changes in relative abundance of bacteria upon antibiotic treatment and the growth of Bifidobacterium and Lactobacillus upon GOS addition were antibiotic and dose dependent. This conclusion explains discrepancies in literature and indicates that particular combinations of GOS antibiotic should be studied. The combination GOS-Amoxicillin was especially of interest as, after decrease in Bifidobacterium levels, a recovery of mainly Bifidobacterium longum was observed and could be correlated with specific degradation patterns of GOS. Next to different degradation profiles of individual GOS, an accumulation of monosaccharides and intermediate organic acids was observed in antibiotic-treated microbiota as compared to nontreated microbiota. This showed that although GOS were utilized and beneficial bacteria could grow in 3 of 4 antibiotics tested, the metabolic activity of an antibiotic-treated microbiota was still disturbed as compared to the nontreated microbiota.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/metabolismo , Galactose/metabolismo , Trato Gastrointestinal/microbiologia , Microbiota/efeitos dos fármacos , Oligossacarídeos/metabolismo , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/genética , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Fermentação , Trato Gastrointestinal/metabolismo , Humanos , Lactobacillus/efeitos dos fármacos , Lactobacillus/genética , Lactobacillus/crescimento & desenvolvimento , Lactobacillus/metabolismo , Prebióticos/análise , Prebióticos/microbiologia
11.
Br J Nutr ; 110(7): 1292-303, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23507173

RESUMO

Adolescence is a time for rapid growth that represents an opportunity to influence peak bone mass. Prebiotic agents, such as galacto-oligosaccharides (GOS), increase Ca absorption in animal models and postmenopausal women. The objectives of the present study were to investigate the dose-response relationship of GOS supplementation on Ca absorption during growth and to assess changes in colonic microbiota to better understand the mechanism by which GOS is acting. A total of thirty-one healthy adolescent girls aged 10-13 years consumed smoothie drinks twice daily with 0, 2·5 or 5 g GOS for three 3-week periods in a random order. Fractional Ca absorption was determined from urinary Ca excretion over 48 h at the end of each 3-week period using a dual stable isotope method. Faecal microbiota and bifidobacteria were assessed by PCR-denaturing gradient gel electrophoresis and quantitative PCR. Fractional Ca absorption after the 48 h treatment with control, 5 and 10 g GOS/d was 0·393 (SD 0·092), 0·444 (SD 0·086) and 0·419 (SD 0·099), respectively. Significant improvements in Ca absorption were seen with both low and high doses of GOS compared with the control (P,0·02), but itwas not a dose-response relationship. The increase in absorption was greatest in the urine collected after 24 h, which is consistent with lower gut absorption. Faecal bifidobacteria increased (control 10·89 (SD 13·86), 5 g GOS 22·80 (SD 15·74) and 10 g GOS 11·54 (SD 14·20)) with the GOS treatment (P,0·03). The results suggest that daily consumption of 5 g GOS increases Ca absorption, which may be mediated by the gut microbiota, specifically bifidobacteria.


Assuntos
Bifidobacterium , Cálcio da Dieta/metabolismo , Cálcio/metabolismo , Fezes/microbiologia , Galactose/farmacologia , Intestinos/microbiologia , Oligossacarídeos/farmacologia , Adolescente , Cálcio/urina , Cálcio da Dieta/urina , Criança , Método Duplo-Cego , Feminino , Humanos , Absorção Intestinal
12.
Br J Nutr ; 109(7): 1338-48, 2013 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-22850280

RESUMO

Galacto-oligosaccharides (GOS) are carbohydrates that are fermented by colonic microbiota. The present study examined effects of a 3-week dietary enrichment with 6 % (w/w) GOS on parameters of energy balance in forty-three male Wistar rats. GOS was tested with two doses of calcium phosphate (30 and 100 mmol/kg), known to differently affect colonic fermentation. After 17 d, isoenergetic test meals were presented and plasma responses of ghrelin, glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) were measured. On day 21 (study termination) epididymal fat pads and caecum were weighed. Additionally, gastrointestinal mucosal samples and proximal colonic contents were analysed for gene expression (ghrelin, proglucagon and PYY) and fermentation metabolites (SCFA and lactate), respectively. GOS reduced energy intake most prominently during the first week, without provoking compensatory overeating later on (average intake reduction: 14 %). The GOS-fed rats showed increased caecal and reduced fat-pad weight and increased gene expression of the satiety-related peptides, PYY (1.7-fold) and proglucagon (3.5-fold). Pre-meal baseline and post-meal plasma levels of PYY, but not of ghrelin or GLP-1, were higher in GOS-fed rats than in control rats. Ca enrichment resulted in higher energy intake (average 4.5 %). GOS diets increased lactic acid levels and slightly reduced butyric acid in proximal colonic contents. Ca abolished the GOS-related elevation of lactic acid, while increasing propionic acid levels, but did not inhibit GOS-related effects on energy intake, fat-pad weight or gene expression. These results indicate that dietary GOS stimulate a number of physiological mechanisms that can reduce energy intake, regardless of the calcium phosphate content of the diet.


Assuntos
Cálcio da Dieta/metabolismo , Fibras na Dieta/metabolismo , Mucosa Gástrica/metabolismo , Hormônios Gastrointestinais/metabolismo , Mucosa Intestinal/metabolismo , Oligossacarídeos/metabolismo , Resposta de Saciedade , Animais , Depressores do Apetite/química , Depressores do Apetite/metabolismo , Fosfatos de Cálcio/administração & dosagem , Cálcio da Dieta/administração & dosagem , Fibras na Dieta/análise , Ingestão de Energia , Fermentação , Galactose/química , Galactose/metabolismo , Hormônios Gastrointestinais/sangue , Hormônios Gastrointestinais/genética , Regulação da Expressão Gênica , Mucosa Intestinal/microbiologia , Masculino , Oligossacarídeos/química , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Organismos Livres de Patógenos Específicos , Aumento de Peso
13.
J Nutr ; 142(7): 1205-12, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22623395

RESUMO

Galacto-oligosaccharides (GOS) are considered to be prebiotic, although the contribution of specific members of the microbiota to GOS fermentation and the exact microbial metabolites that are produced upon GOS fermentation are largely unknown. We aimed to determine this using uniformly (13)C-labeled GOS. The normal (control) medium and unlabeled or (13)C-labeled GOS was added to a dynamic, validated, in vitro model of the large-intestine containing an adult-type microbiota. Liquid-chromatography MS was used to measure the incorporation of (13)C label into metabolites. 16S-rRNA stable isotope probing coupled to a phylogenetic micro-array was used to determine label incorporation in microbial biomass. The primary members within the complex microbiota that were directly involved in GOS fermentation were shown to be Bifidobacterium longum, B. bifidum, B. catenulatum, Lactobacillus gasseri, and L. salivarius, in line with the prebiotic effect of GOS, although some other species incorporated (13)C label also. GOS fermentation led to an increase in acetate (+49%) and lactate (+23%) compared with the control. Total organic acid production was 8.50 and 7.52 mmol/g of carbohydrate fed for the GOS and control experiments, respectively. At the same time, the cumulative production of putrefactive metabolites (branched-chain fatty acids and ammonia) was reduced by 55%. Cross-feeding of metabolites from primary GOS fermenters to other members of the microbiota was observed. Our findings support a prebiotic role for GOS and its potential to act as a synbiotic in combination with certain probiotic strains.


Assuntos
Bifidobacterium/efeitos dos fármacos , Colo/efeitos dos fármacos , Galactose/farmacologia , Lactobacillus/efeitos dos fármacos , Oligossacarídeos/farmacologia , Prebióticos , Probióticos , Ácido Acético/metabolismo , Amônia/metabolismo , Bifidobacterium/metabolismo , Isótopos de Carbono/metabolismo , Colo/metabolismo , Colo/microbiologia , Ácidos Graxos/biossíntese , Fermentação , Galactose/metabolismo , Ácido Láctico/metabolismo , Lactobacillus/metabolismo , Análise em Microsséries , Modelos Biológicos , Oligossacarídeos/metabolismo , Filogenia , RNA Ribossômico 16S , Coloração e Rotulagem , Simbióticos
14.
J Agric Food Chem ; 59(12): 6501-10, 2011 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-21553845

RESUMO

Galactooligosaccharides (GOS), prebiotic nondigestible oligosaccharides derived from lactose, have the potential for improving mineral balance and bone properties. This study examined the dose-response effect of GOS supplementation on calcium and magnesium absorption, mineral retention, bone properties, and gut microbiota in growing rats. Seventy-five 4-week-old male Sprague-Dawley rats were randomized into one of five treatment groups (n = 15/group) and fed a diet containing 0, 2, 4, 6, or 8% GOS by weight for 8 weeks. Dietary GOS significantly decreased cecal pH and increased cecal wall weight and content weight in a dose-dependent manner (p < 0.0001). Fingerprint patterns of the 16S rRNA gene PCR-DGGE from fecal DNA indicated the variance of bacterial community structure, which was primarily explained by GOS treatments (p = 0.0001). Quantitative PCR of the samples revealed an increase in the relative proportion of bifidobacteria with GOS (p = 0.0001). Net calcium absorption was increased in a dose-response manner (p < 0.01) with GOS supplementation. Dietary GOS also increased (p < 0.02) net magnesium absorption, femur 45Ca uptake, calcium and magnesium retention, and femur and tibia breaking strength. Distal femur total and trabecular volumetric bone mineral density (vBMD) and area and proximal tibia vBMD increased (p < 0.02) with GOS supplementation. Trabecular-rich bones, that is, those that rapidly turn over, were most benefited. Regression modeling showed that GOS benefited calcium and magnesium utilization and vBMD through decreased cecal pH, increased cecal wall and content weight, and increased proportion of bifidobacteria.


Assuntos
Desenvolvimento Ósseo , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Minerais/metabolismo , Oligossacarídeos/metabolismo , Prebióticos/análise , Absorção , Animais , Bifidobacterium/genética , Bifidobacterium/isolamento & purificação , Bifidobacterium/metabolismo , Densidade Óssea , Osso e Ossos/fisiologia , Cálcio/metabolismo , Fermentação , Humanos , Magnésio/metabolismo , Masculino , Modelos Animais , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
15.
Br J Nutr ; 105(4): 489-95, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20875217

RESUMO

Previous animal and human studies have shown protective effects of Ca on the resistance to enteropathogenic infections. Most interventions were performed with calcium phosphate and little is known about the protective effect of other dietary sources of Ca. Therefore, we investigated the efficacy of several Ca salts to enhance intestinal resistance to Salmonella enteritidis infection. Rats (n 7-8 per group) were fed a high-fat, Western human-style, purified diet with a low Ca content (20 mmol calcium phosphate/kg; negative control group) or the same diet supplemented with either (extra) calcium phosphate, milk Ca, calcium chloride or calcium carbonate (total of 100 mmol Ca supplement/kg). Diets contained Cr-EDTA for assessment of incremental changes in intestinal permeability. After an adaptation period of 2 weeks, animals were orally infected with S. enteritidis to mimic a human-relevant foodborne infection. Ca supplement-induced changes on faecal lactobacilli and enterobacteria were studied before infection. Changes in intestinal permeability were determined by measuring urinary Cr with time. Persistence of Salmonella was determined by studying faecal excretion of this pathogen in time. Overall, all Ca salts increased resistance towards Salmonella. After infection, body weight gain and food intake were higher in the calcium phosphate group. Calcium phosphate and milk Ca decreased faecal enterobacteria before infection. All Ca salts decreased infection-induced intestinal permeability and persistence of Salmonella. Calcium phosphate, milk Ca, calcium carbonate and calcium chloride are able to enhance the intestinal resistance to Salmonella in rats.


Assuntos
Cálcio/farmacologia , Intestinos/microbiologia , Infecções por Salmonella/microbiologia , Infecções por Salmonella/prevenção & controle , Sais/farmacologia , Ração Animal , Ciências da Nutrição Animal , Animais , Peso Corporal , Cálcio/química , Fosfatos de Cálcio/química , Ácido Edético/química , Íons , Masculino , Permeabilidade , Ratos , Ratos Wistar , Infecções por Salmonella/metabolismo , Salmonella enteritidis/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...