Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
1.
Nutrients ; 16(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38931218

ABSTRACT

BACKGROUND: The purpose of this research was to assess the growth, tolerance, and compliance outcomes associated with the consumption of a hydrolyzed rice infant formula (HRF) enriched with 2'-Fucosyllactose (2'-FL) a Human Milk Oligosaccharide (HMO), and nucleotides in an intended population of infants. METHODS: This was a non-randomized single-group, multicenter study. The study formula was a hypoallergenic HRF with 2'-FL, Docosahexaenoic acid (DHA), Arachidonic acid (ARA), and nucleotides. Infants 0-90 days of age who were formula fed and experiencing persistent feeding intolerance symptoms, symptoms of suspected food protein (milk and/or soy) allergy, or other conditions where an extensively hydrolyzed infant formula was deemed an appropriate feeding option were recruited by pediatricians from their local populations. The primary outcome was maintenance of weight-for-age z-score. Weight, length, head circumference, formula intake, tolerance measures, clinical symptoms and questionnaires were collected. Thirty-three infants were enrolled, and 27 completed the study, on study product. RESULTS: Weight-for-age z-scores of infants showed a statistically significant improvement from Visit 1 to Visit 4 (p = 0.0331). There was an adequate daily volume intake of 762 ± 28 mL/day, average daily number of stools of 2.1 ± 0.3, and mean rank stool consistency of 2.38 ± 0.18. After 28 days of switching to a HRF, 86.8 ± 5.9% of the symptoms resolved or got better by Visit 4 as reported by parents. CONCLUSIONS: HRF with 2'-FL HMO was safe, well tolerated, and supported weight gain in infants with suspected cow's milk allergy or persistent feeding intolerance.


Subject(s)
Infant Formula , Milk, Human , Oligosaccharides , Oryza , Trisaccharides , Humans , Infant Formula/chemistry , Trisaccharides/administration & dosage , Infant , Milk, Human/chemistry , Oryza/chemistry , Female , Male , Oligosaccharides/administration & dosage , Infant, Newborn , Infant Nutritional Physiological Phenomena
2.
Nutrients ; 13(9)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34578862

ABSTRACT

Insulin resistance leads to the onset of medical conditions such as type 2 diabetes, and its development is associated with the alteration in the gut microbiota. Although it has been demonstrated that supplementation with prebiotics modulates the gut microbiota, limited evidence is available for effects of prebiotics on insulin resistance, especially for humans. We investigated the prebiotic effect of 1-kestose supplementation on fasting insulin concentration in obesity-prone humans and rats. In the preliminary study using rats, the hyperinsulinemia induced by high-fat diet was suppressed by intake of water with 2% (w/v) 1-kestose. In the clinical study using obese-prone volunteers, the fasting serum insulin level was significantly reduced from 6.5 µU/mL (95% CI, 5.5-7.6) to 5.3 (4.6-6.0) by the 12-week intervention with supplementation of 10 g 1-kestose/day, whereas it was not changed by the intervention with placebo (6.2 µU/mL (5.4-7.1) and 6.5 (5.5-7.6) before and after intervention, respectively). The relative abundance of fecal Bifidobacterium was significantly increased to 0.3244 (SD, 0.1526) in 1-kestose-supplemented participants compared to that in control participants (0.1971 (0.1158)). These results suggest that prebiotic intervention using 1-kestose may potentially ameliorate insulin resistance in overweight humans via the modulation of the gut microbiota. UMIN 000028824.


Subject(s)
Dietary Supplements , Gastrointestinal Microbiome/drug effects , Glucose/metabolism , Obesity/metabolism , Trisaccharides/pharmacology , Adult , Animals , Disease Models, Animal , Double-Blind Method , Female , Humans , Japan , Male , Middle Aged , Rats , Rats, Sprague-Dawley , Trisaccharides/administration & dosage
3.
Carbohydr Polym ; 270: 118389, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34364630

ABSTRACT

Galactooligosaccharides have been known to have many health benefits as prebiotic ingredients. In this study, we examined the anti-inflammatory activity of the galactooligosaccharide, NeoGOS-P70 (Korean commercial product), in a dextran sodium sulfate-induced colitis model. Next, we performed compositional characterization of NeoGOS-P70, which confirmed that it was a 77.4% high-purity GOS products, including a large amount of 4'-galactosyllactose. Further experiments in DSS-induced colitis model showed that oral administration of NeoGOS-P70 could significantly improve DSS-induced colitis symptoms, such as weight loss, reduction in colon shortening, and suppression of inflammatory mediators, including interleukin-6, tumor necrosis factor-α, and myeloperoxidase secretion from colon of ulcerative colitis mice. Histological analysis of mucin expression in colon tissue revealed the protective effects of NeoGOS-P70. These results suggest the potential of the novel GOS, NeoGOS-P70, as an anti-ulcerative colitis agent that could regulate inflammatory responses.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bacillus , Colitis/drug therapy , Trisaccharides/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Colitis/chemically induced , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colon/metabolism , Dextran Sulfate/adverse effects , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred BALB C , Peroxidase/metabolism , Prebiotics/administration & dosage , Trisaccharides/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
4.
Food Funct ; 12(18): 8681-8693, 2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34351342

ABSTRACT

This study examined the impact of chitobiose (GlcN)2 and chitotriose (GlcN)3 on lipid accumulation modification and their inhibitory functionalities. (GlcN)2 and (GlcN)3 significantly inhibited the total cholesterol (TC), triglyceride (TG), and low-density lipid cholesterol (LDL-c) levels in the liver of the ob/ob-/- mice fed a non-high-fat diet. This phenomenon was associated with a reduction in the mRNA and protein expression of TG synthesis and fatty acid uptake-related signaling, significantly affecting the cluster of differentiation 36 (CD36) and diacylglycerol acyltransferase 2 (DGAT2). Furthermore, the CD36 and DGAT2 genes were overexpressed by constructing a plasmid and transfecting it into HepG2 cells, after which the phenotypic traits of lipid accumulation were assessed in vitro. Consequently, it was evident that (GlcN)2 and (GlcN)3 reduced the overexpression of these proteins and relieved cellular lipid accumulation. In conclusion, these results indicated that (GlcN)2 and (GlcN)3 acted positively against NAFLD while regulating steatosis in the non-high-fat diet NAFLD model. The potential NAFLD treatment strategies, such as targeting CD36 and DGAT2 signaling, could provide scientific insight into further applying food-derived ingredients to reduce the risk of high-fat metabolism.


Subject(s)
CD36 Antigens/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Disaccharides/administration & dosage , Fatty Acids/administration & dosage , Non-alcoholic Fatty Liver Disease/therapy , Triglycerides/biosynthesis , Trisaccharides/administration & dosage , Animals , CD36 Antigens/genetics , Diacylglycerol O-Acyltransferase/genetics , Diet , Dietary Fats/administration & dosage , Gene Expression Regulation , Hep G2 Cells , Humans , Lipid Metabolism/genetics , Lipids/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction , Simvastatin/pharmacology
5.
Food Funct ; 12(18): 8507-8521, 2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34308934

ABSTRACT

2'-Fucosyllactose (2'-FL) is one of the predominant oligosaccharides found in human milk and has several well-established beneficial effects in the host. It has previously been shown that 2'-FL can improve the metabolic phenotype in high-fat (HF)-fed mice. Here we investigated whether dietary supplementation with 2'-FL was associated with improved intestinal barrier integrity, signaling in the vagal afferent pathway and cognitive function. Mice were fed either a low-fat (LF, 10% fat per kcal) or HF (45% fat per kcal) diet with or without supplementation of 2'-FL (10% w/w) in the diet for 8 weeks. Body weight, energy intake, fat and lean mass, intestinal permeability (ex vivo in Ussing chambers), lipid profiles, gut microbiome and microbial metabolites, and cognitive functions were measured. Vagal afferent activity was measured via immunohistochemical detection of c-Fos protein in the brainstem in response to peripheral administration of cholecystokinin (CCK). 2'-FL significantly attenuated the HF-induced increase in fat mass and energy intake. 2'-FL significantly reduced intestinal permeability and significantly increased expression of interleukin (IL)-22, a cytokine known for its protective role in the intestine. Additionally, 2'-FL led to changes in the gut microbiota composition and in the associated microbial metabolites. Signaling in the vagal afferent pathway was improved but there was no effect on cognitive function. In conclusion, 2'-FL supplementation improved the metabolic profiles, gut barrier integrity, lipid metabolism and signaling in the vagal afferent pathway. These findings support the utility of 2'-FL in the control of gut barrier function and metabolic homeostasis under a metabolic challenge.


Subject(s)
Afferent Pathways/physiology , Brain-Gut Axis/physiology , Dietary Supplements , Intestinal Mucosa/physiology , Milk, Human/chemistry , Trisaccharides/administration & dosage , Vagus Nerve/physiology , Animals , Bacteria/classification , Bacteria/growth & development , Bacteria/metabolism , Brain/metabolism , Cecum/metabolism , Cecum/microbiology , Diet, Fat-Restricted , Diet, High-Fat , Gastrointestinal Microbiome , Lipid Metabolism , Male , Metabolome , Mice , Mice, Inbred C57BL , Signal Transduction , Trisaccharides/blood
6.
Sci Rep ; 11(1): 8302, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859330

ABSTRACT

Current research implicates pre- and probiotic supplementation as a potential tool for improving symptomology in physical and mental ailments, which makes it an attractive concept for clinicians and consumers alike. Here we focus on the transitional period of late adolescence and early adulthood during which effective interventions, such as nutritional supplementation to influence the gut microbiota, have the potential to offset health-related costs in later life. We examined multiple indices of mood and well-being in 64 healthy females in a 4-week double blind, placebo controlled galacto-oligosaccharides (GOS) prebiotic supplement intervention and obtained stool samples at baseline and follow-up for gut microbiota sequencing and analyses. We report effects of the GOS intervention on self-reported high trait anxiety, attentional bias, and bacterial abundance, suggesting that dietary supplementation with a GOS prebiotic may improve indices of pre-clinical anxiety. Gut microbiota research has captured the imagination of the scientific and lay community alike, yet we are now at a stage where this early enthusiasm will need to be met with rigorous research in humans. Our work makes an important contribution to this effort by combining a psychobiotic intervention in a human sample with comprehensive behavioural and gut microbiota measures.


Subject(s)
Anti-Anxiety Agents , Anxiety/prevention & control , Dietary Supplements , Gastrointestinal Microbiome/drug effects , Healthy Volunteers , Prebiotics , Trisaccharides/pharmacology , Adolescent , Adult , Female , Humans , Prebiotics/administration & dosage , Trisaccharides/administration & dosage , Young Adult
7.
Nutrients ; 14(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35011074

ABSTRACT

Human milk is rich in oligosaccharides that influence intestinal development and serve as prebiotics for the infant gut microbiota. Probiotics and 2'-fucosyllactose (2'-FL) added individually to infant formula have been shown to influence infant development, but less is known about the effects of their synbiotic administration. Herein, the impact of formula supplementation with 2'-fucosyllactose (2'-FL) and Bifidobacterium longum subsp. infantis Bi-26 (Bi-26), or 2'-FL + Bi-26 on weight gain, organ weights, and intestinal development in piglets was investigated. Two-day-old piglets (n = 53) were randomized in a 2 × 2 design to be fed a commercial milk replacer ad libitum without (CON) or with 1.0 g/L 2'-FL. Piglets in each diet were further randomized to receive either glycerol stock alone or Bi-26 (109 CFU) orally once daily. Body weights and food intake were monitored from postnatal day (PND) 2 to 33/34. On PND 34/35, animals were euthanized and intestine, liver and brain weights were assessed. Intestinal samples were collected for morphological analyses and measurement of disaccharidase activity. Dry matter of cecum and colon contents and Bifidobacterium longum subsp. infantis abundance by RT-PCR were also measured. All diets were well tolerated, and formula intake did not differ among the treatment groups. Daily body weights were affected by 2'-FL, Bi-26, and day, but no interaction was observed. There was a trend (p = 0.075) for greater total body weight gain in CON versus all other groups. Jejunal and ascending colon histomorphology were unaffected by treatment; however, there were main effects of 2'-FL to increase (p = 0.040) and Bi-26 to decrease (p = 0.001) ileal crypt depth. The addition of 2'-FL and/or Bi-26 to milk replacer supported piglet growth with no detrimental effects on body and organ weights, or intestinal structure and function.


Subject(s)
Animals, Newborn/growth & development , Bifidobacterium longum subspecies infantis , Intestines/growth & development , Organ Size/drug effects , Swine/growth & development , Trisaccharides/administration & dosage , Animals , Bifidobacterium longum subspecies infantis/isolation & purification , Diet/veterinary , Disaccharidases/metabolism , Intestines/drug effects , Intestines/microbiology , Male , Milk Substitutes , Probiotics/administration & dosage , Swine/microbiology , Symbiosis , Weight Gain/drug effects
8.
Nutr Hosp ; 37(4): 698-706, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32698596

ABSTRACT

Introduction: Introduction: human milk oligosaccharides (HMOs) are an important component of human milk supporting the development of a balanced intestinal microbiota and immune protection in breastfed infants. Randomized controlled trials (RCTs) have demonstrated that infant formulas supplemented with the HMOs 2'-fucosyllactose (2'FL) and lacto-N-neotetraose (LNnT) are safe, well-tolerated, and support normal growth. This Real-World Evidence (RWE) study aimed to evaluate growth and tolerance in infants consuming a formula supplemented with 1 g/L of 2'FL and 0.5 g/L of LNnT, and included a mixed-feeding group never studied before in RCTs. Participants and methods: this open-label, prospective study was conducted at six centers in Spain, and included healthy, exclusively breastfed infants (BF group), an exclusively formula-fed group (FF) who received a milk-based formula with 2' FL and LNnT, and a group mixed fed with both formula and human milk (MF), for 8 weeks. Co-primary outcomes were growth (anthropometry) and gastrointestinal tolerance (Infant Gastrointestinal Symptom Questionnaire, IGSQ). Secondary outcomes included formula satisfaction and adverse events (AEs). Results: 159 infants completed the study (66 FF, 48 MF, and 45 BF). Mean z-scores for growth were similar between all groups and within ± 0.5 of WHO medians at week 8. Composite IGSQ scores demonstrated low GI distress in all groups, with no significant group differences at baseline, week 4, or week 8. Incidence of AEs was low overall, and comparable across groups. Conclusions: in this RWE study examining a HMO-supplemented infant formula, growth and tolerance outcomes were similar to RCT findings, supporting the effectiveness of this early feeding option.


Introducción: Introducción: los oligosacáridos de la leche materna (HMO) contribuyen a desarrollar la inmunoprotección y la microbiota intestinal. Los ensayos aleatorizados (RCT) han demostrado que las fórmulas enriquecidas con 2'fucosilactosa (2'FL) y lacto-N-neotetraosa (LNnT) son seguras, bien toleradas y favorecen el crecimiento. El objetivo de este estudio ha sido valorar el crecimiento, la seguridad y la tolerancia digestiva en lactantes alimentados con una fórmula enriquecida con 1 g/L de 2'FL y 0,5 g/L de LNnT, con datos de la vida real (RWE), incluyendo un grupo de alimentación mixta no estudiado antes en los RCT. Participantes y métodos: estudio prospectivo abierto en seis hospitales españoles que incluyó lactantes sanos alimentados con leche materna (BF), con fórmula enriquecida en 2'FL y LNnT (FF) o con mezcla de ambas (MF), durante ocho semanas. Se valoraron el crecimiento (antropometría), la tolerancia gastrointestinal (cuestionario IGSQ) y los acontecimientos adversos. Resultados: 159 lactantes completaron el estudio (66, 48 y 45, en los grupos FF, MF y BF, respectivamente). Las puntuaciones Z antropométricas a la semana 8 fueron similares entre los grupos y se hallaron dentro del rango de ± 0,5 de la normalidad. Las puntuaciones IGSQ compuestas mostraron un bajo malestar digestivo, sin diferencias significativas entre los grupos, al inicio y en las semanas 4 y 8. La incidencia de eventos adversos fue baja y comparable entre los grupos. Conclusiones: en este estudio RWE que evaluó una fórmula para lactantes enriquecida en HMO, los resultados sobre el crecimiento, la tolerancia y la seguridad fueron similares a los obtenidos en los RCT, respaldando su eficacia como alimentación temprana opcional.


Subject(s)
Infant Formula/chemistry , Oligosaccharides/administration & dosage , Trisaccharides/administration & dosage , Female , Humans , Infant , Infant, Newborn , Male , Milk, Human/chemistry , Oligosaccharides/analysis , Prospective Studies , Trisaccharides/analysis
9.
Nutrients ; 12(7)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32709093

ABSTRACT

Mounting evidence suggests that dietary oligosaccharides promote brain development. This study assessed the capacity of oligofructose (OF) alone or in combination with 2'-fucosyllactose (2'-FL) to alter recognition memory, structural brain development, and hippocampal gene expression. Beginning on postnatal day (PND) 2, male pigs received one of three milk replacers formulated to contain OF, OF + 2'-FL, or no oligosaccharides (CON). Pigs were tested on the novel object recognition task using delays of 1 or 48 h at PND 22. At PND 32-33, magnetic resonance imaging (MRI) procedures were used to assess structural brain development and hippocampal tissue was collected for analysis of mRNA expression. Pigs that consumed the OF diet demonstrated increased recognition memory after a 1 h delay, whereas those consuming diets containing OF + 2'-FL displayed increased recognition memory after a 48 h delay. Pigs fed OF or OF + 2'-FL exhibited a larger relative volume of the olfactory bulbs compared with CON pigs. Provision of OF or OF + 2'-FL altered gene expression related to dopaminergic, GABAergic, cholinergic, cell adhesion, and chromatin remodeling processes. Collectively, these data indicate that dietary OF and OF + 2'-FL differentially improve cognitive performance and affect olfactory bulb structural development and hippocampal gene expression.


Subject(s)
Hippocampus/metabolism , Oligosaccharides/administration & dosage , Trisaccharides/administration & dosage , Animals , Diet , Gene Expression Regulation , Linear Models , Magnetic Resonance Imaging , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recognition, Psychology/drug effects , Swine
10.
Biochem Biophys Res Commun ; 529(1): 64-69, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32560820

ABSTRACT

RegIIIß and RegIIIγ are antimicrobial peptides expressed in intestinal epithelial cells. Expression of these peptides is reportedly decreased by high-fat diet (HFD) and increased by indigestible oligosaccharides in mice. Clearly, these dietary regimens change the structure of intestinal microbiota. We employed an intestinal microbiota transplantation (IMT) to test whether diet-induced changes in the expression of these peptides are mediated by gut microbiota. C57BL/6J mice were fed either a normal-fat diet (NFD), a HFD, or a NFD supplemented with or without 1-kestose (KES), an indigestible oligosaccharide. Ileal RegIIIß and RegIIIγ mRNA levels were lower in mice receiving IMT from HFD-fed mice than in those receiving NFD-fed mice and higher in mice receiving IMT from KES-supplemented mice than in those receiving the mice without KES supplementation. Western blot analysis showed that serum RegIIIß levels changed in parallel with the ileal mRNA levels. We propose that HFD- and KES-induced changes in the ileal RegIIIß and RegIIIγ expression and in the circulating RegIIIß levels are mediated, at least in part, by intestinal microbiota.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Pancreatitis-Associated Proteins/blood , Pancreatitis-Associated Proteins/genetics , Animals , Diet , Diet, High-Fat , Ileum/metabolism , Interleukins/genetics , Interleukins/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trisaccharides/administration & dosage , Interleukin-22
11.
Nutrients ; 12(5)2020 May 25.
Article in English | MEDLINE | ID: mdl-32466125

ABSTRACT

Human milk oligosaccharides (HMOs) are chief maternal milk constituents that feed the intestinal microbiota and drive maturation of the infant gut. Our objective was to determine whether supplementing individual HMOs to a weanling diet alters growth and gut health in rats. Healthy three-week-old Sprague Dawley rat pups were randomized to control, 2'-O-fucosyllactose (2'FL)- and 3'sialyllactose (3'SL)-fortified diets alone or in combination at physiological doses for eight weeks. Body composition, intestinal permeability, serum cytokines, fecal microbiota composition, and messenger RNA (mRNA) expression in the gastrointestinal tract were assessed. Males fed a control diet were 10% heavier and displayed elevated interleukin (IL-18) (p = 0.01) in serum compared to all HMO-fortified groups at week 11. No differences in body composition were detected between groups. In females, HMOs did not affect body weight but 2'FL + 3'SL significantly increased cecum weight. All female HMO-fortified groups displayed significant reductions in intestinal permeability compared to controls (p = 0.02). All HMO-fortified diets altered gut microbiota composition and mRNA expression in the gastrointestinal tract, albeit differently according to sex. Supplementation with a fraction of the HMOs found in breast milk has a complex sex-dependent risk/benefit profile. Further long-term investigation of gut microbial profiles and supplementation with other HMOs during early development is warranted.


Subject(s)
Dietary Supplements , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/microbiology , Milk, Human/drug effects , Oligosaccharides/administration & dosage , Animals , Biomarkers/blood , Body Weight , Cecum/drug effects , Cecum/metabolism , Cecum/microbiology , Feces/microbiology , Female , Gastrointestinal Tract/drug effects , Interleukin-18/blood , Lactose/administration & dosage , Lactose/analogs & derivatives , Leptin/blood , Male , Milk, Human/chemistry , Organ Size/drug effects , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/isolation & purification , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA , Sialic Acids/administration & dosage , Trisaccharides/administration & dosage
12.
Pediatr Allergy Immunol ; 31(7): 745-754, 2020 10.
Article in English | MEDLINE | ID: mdl-32426882

ABSTRACT

BACKGROUND: Bioactive proteins and human milk oligosaccharides (HMOs), important ingredients in breast milk, that protect against infections are lacking in young child formula (YCF). This study investigated the effects of new YCFs on respiratory and gastrointestinal infections in toddlers. METHODS: Four hundred and sixty one healthy Chinese children aged 1-2.5 years were recruited in this randomized, controlled, double-blind, parallel-group clinical trial of different YCFs. They were randomly assigned to either standard milk formula (YCF-Ref) or one of three new YCFs containing bioactive proteins and/or the HMO 2'-fucosyllactose (2'-FL) and/or milk fat for six months. Primary outcomes were incidence of upper respiratory tract infection (URTI) and duration of gastrointestinal tract infections (GITI). RESULTS: There were no significant between-group differences in primary outcomes. For secondary outcomes, subjects receiving 2'-FL-supplemented YCF had longer URTI. Subjects receiving YCF supplemented with milk fat and intact bioactive proteins, and 2'-FL at levels found in breast milk, had more GITI episodes and shorter time to first GITI but similar effects on URTI duration than YCF-Ref recipients. No effects on URTI and GITI were observed in toddlers receiving YCF with bioactive proteins at lower levels than breast milk. Occurrence of adverse events and anthropometry were similar in all groups. CONCLUSIONS: All three YCFs supplemented with different combinations of intact bioactive proteins, 2'-FL, and milk fat are safe in toddlers. No difference is detected among YCFs on URTI incidence and GITI duration. Further studies are needed to verify these findings especially in infants who may benefit most from the immune-boosting effects of bioactive proteins and HMOs.


Subject(s)
Gastrointestinal Diseases/epidemiology , Infant Formula/chemistry , Respiratory Tract Infections/epidemiology , Asian People , Child, Preschool , Dietary Supplements , Double-Blind Method , Female , Gastrointestinal Diseases/prevention & control , Humans , Incidence , Infant , Male , Milk, Human/chemistry , Oligosaccharides/administration & dosage , Oligosaccharides/chemistry , Respiratory Tract Infections/prevention & control , Treatment Outcome , Trisaccharides/administration & dosage , Trisaccharides/chemistry
13.
J Vet Med Sci ; 82(7): 866-874, 2020 Jul 10.
Article in English | MEDLINE | ID: mdl-32389951

ABSTRACT

1-kestose is a structural component of fructo-oligosaccharides and is composed of 2 fructose residues bound to sucrose through ß2-1 bonds. In the present study, the influence of the ingestion of 1-kestose on the intestinal microbiota was investigated in cats. Six healthy cats were administered 1 g/day of 1-kestose for 8 weeks followed by a 2-week wash-out period. Fecal samples were collected from cats after 0, 4, 8, and 10 weeks. The intestinal microbiota was examined by a 16S rRNA gene metagenomic analysis and real-time PCR. Short-chain fatty acids were measured by GC/MS. The results suggested that the intestinal bacterial community structure in feline assigned to this study was divided into 2 types: one group mainly composed of the genus Lactobacillus (GA) and the other mainly composed of the genus Blautia with very few bacteria of Lactobacillus (GB). Furthermore, the number of Bifidobacterium slightly increased after the administration of 1-kestose (at 4 and 8 weeks) (P<0.1). The administration of 1-kestose also increased the abundance of Megasphaera, the butyric acid-producing bacteria, at 4 and 8 weeks (P<0.1). Furthermore, an increase in butyric acid levels was observed after the administration of 1-kestose for 4 weeks (P<0.1). These results suggest that 1-kestose activated butyrate-producing bacteria as well as bifidobacteria and propose its potential as a new generation prebiotic.


Subject(s)
Cats/microbiology , Gastrointestinal Microbiome/drug effects , Trisaccharides/administration & dosage , Animal Feed/analysis , Animals , Bacteria/classification , Bacteria/genetics , Bifidobacterium/isolation & purification , Butyrates/metabolism , Diet/veterinary , Fatty Acids, Volatile/metabolism , Feces/microbiology , Female , Lactobacillus/isolation & purification , Male , Megasphaera/isolation & purification , Prebiotics/administration & dosage , RNA, Ribosomal, 16S/genetics
14.
Nutrients ; 12(4)2020 Apr 05.
Article in English | MEDLINE | ID: mdl-32260563

ABSTRACT

Obesity is characterized by fat accumulation, chronic inflammation and impaired satiety signaling, which may be due in part to gut microbial dysbiosis. Manipulations of the gut microbiota and its metabolites are attractive targets for obesity treatment. The predominant oligosaccharide found in human milk, acts as a prebiotic with beneficial effects on the host. However, little is known about the beneficial effects of 2'-FL in obesity. The aim of this study was to determine the beneficial effects of 2'-FL supplementation on the microbiota-gut-brain axis and the diet-induced obese phenotype in high fat (HF)-fed mice. Male C57/BL6 mice (n = 6/group; six weeks old) were counter-balanced into six weight-matched groups and fed either a low-fat (LF; 10% kcal as fat), HF (45% kcal as fat) or HF diet with 2'-FL (HF_2'-FL) at 1, 2, 5 and 10% (w/v) in drinking water for six weeks. General phenotypes (body weight, energy intake, fat and lean mass), cecal microbiome and metabolites, gut-brain signaling, intestinal permeability and inflammatory and lipid profiles were assessed. Only 10% 2'-FL, but not 1, 2 or 5%, decreased HF diet-induced increases in energy intake, fat mass and body weight gain. A supplementation of 10% 2'-FL changed the composition of cecal microbiota and metabolites compared to LF- and HF-fed mice with an increase in Parabacteroides abundance and lactate and pyruvate, respectively, whose metabolic effects corresponded to our study findings. In particular, 10% 2'-FL significantly reversed the HF diet-induced impairment of cholecystokinin-induced inhibition of food intake. Gene expressions of interleukin (IL)-1ß, IL-6, and macrophage chemoattractant protein-1 in the cecum were significantly downregulated by 10% 2'-FL compared to the HF group. Furthermore, 10% 2'-FL suppressed HF diet-induced upregulation of hepatic peroxisome proliferator-activated receptor gamma, a transcription factor for adipogenesis, at the gene level. In conclusion, 10% 2'-FL led to compositional changes in gut microbiota and metabolites associated with improvements in metabolic profiles and gut-brain signaling in HF-fed mice. These findings support the use of 2'-FL for modulating the hyperphagic response to HF diets and improving the microbiota-gut-brain axis.


Subject(s)
Brain/physiology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/physiology , Obesity/chemically induced , Trisaccharides/administration & dosage , Animal Feed/analysis , Animals , Brain/drug effects , Diet/veterinary , Diet, High-Fat , Dietary Supplements , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Inflammation/chemically induced , Inflammation/drug therapy , Mice , Obesity/drug therapy
15.
Nat Commun ; 11(1): 1250, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144257

ABSTRACT

Currently, there are no non-invasive tools to accurately diagnose wound and surgical site infections before they become systemic or cause significant anatomical damage. Fluorescence and photoacoustic imaging are cost-effective imaging modalities that can be used to noninvasively diagnose bacterial infections when paired with a molecularly targeted infection imaging agent. Here, we develop a fluorescent derivative of maltotriose (Cy7-1-maltotriose), which is shown to be taken up in a variety of gram-positive and gram-negative bacterial strains in vitro. In vivo fluorescence and photoacoustic imaging studies highlight the ability of this probe to detect infection, assess infection burden, and visualize the effectiveness of antibiotic treatment in E. coli-induced myositis and a clinically relevant S. aureus wound infection murine model. In addition, we show that maltotriose is an ideal scaffold for infection imaging agents encompassing better pharmacokinetic properties and in vivo stability than other maltodextrins (e.g. maltohexose).


Subject(s)
Fluorescent Dyes/administration & dosage , Molecular Imaging/methods , Myositis/diagnostic imaging , Surgical Wound Infection/diagnostic imaging , Trisaccharides/administration & dosage , Animals , Carbocyanines/administration & dosage , Carbocyanines/chemistry , Disease Models, Animal , Drug Stability , Escherichia coli/isolation & purification , Escherichia coli/metabolism , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Humans , Injections, Intravenous , Luminescent Measurements/methods , Mice , Microscopy, Fluorescence/methods , Molecular Probes/administration & dosage , Molecular Probes/chemistry , Molecular Probes/metabolism , Myositis/microbiology , Photoacoustic Techniques/methods , Rats , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/metabolism , Surgical Wound Infection/microbiology , Trisaccharides/chemistry , Trisaccharides/metabolism
16.
J Dairy Sci ; 103(4): 3816-3827, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32089300

ABSTRACT

Oligosaccharides are the third most abundant component in human milk. It is widely accepted that they play several important protective, physiological, and biological roles, including selective growth stimulation of beneficial gut microbiota, inhibition of pathogen adhesion, and immune modulation. However, until recently, very few commercial products on the market have capitalized on these functions. This is mainly because the quantities of human milk oligosaccharides required for clinical trials have been unavailable. Recently, clinical studies have tested the potential beneficial effects of feeding infants formula containing 2'-fucosyllactose, which is the most abundant oligosaccharide in human milk. These studies have opened this field for further well-designed studies, which are required to fully understand the role of human milk oligosaccharides. However, one of the most striking features of human milk is its diversity of oligosaccharides, with over 200 identified to date. It may be that a mixture of oligosaccharides is even more beneficial to infants than a single structure. For this reason, the milk of domestic animals has become a focal point in recent years as an alternative source of complex oligosaccharides with associated biological activity. This review will focus specifically on free oligosaccharides found in bovine and caprine milk and the biological roles associated with such structures. These dairy streams are ideal sources of oligosaccharides, given their wide availability and use in so many regularly consumed dairy products. The aim of this review was to provide an overview of research into the functional role of bovine and caprine milk oligosaccharides in host-microbial interactions in the gut and provide current knowledge related to the isolation of oligosaccharides as ingredients for incorporation in functional or medical foods.


Subject(s)
Milk, Human/chemistry , Milk/chemistry , Oligosaccharides/metabolism , Animals , Cattle , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/metabolism , Goats , Humans , Infant , Oligosaccharides/administration & dosage , Trisaccharides/administration & dosage
17.
Clin Transl Gastroenterol ; 11(12): e00276, 2020 12.
Article in English | MEDLINE | ID: mdl-33512807

ABSTRACT

INTRODUCTION: Treatment options for irritable bowel syndrome (IBS) are limited, causing many patients to remain symptomatic. This study assessed the potential of human milk oligosaccharides (HMOs) to normalize bowel habits. Secondary outcomes included IBS severity and health-related quality of life. METHODS: This multicenter, open-label trial recruited patients with IBS from 17 sites across the United States. Patients received daily orally administrated 5-g intervention of the HMOs 2'-fucosyllactose and lacto-N-neotetraose in a 4:1 mix. Bowel habits, IBS symptoms, and quality of life were assessed at baseline and every 4 weeks during the 12-week intervention. RESULTS: A total of 317 patients (70.7% women; mean age of 44.0 years, range 18-93 years) received the trial product, and 245 patients completed the trial according to protocol. Patients had a significant improvement from baseline to 12 weeks in total percentage of bowel movements with abnormal stool consistency (mean and [95% confidence interval]: 90.7 [88.9-92.9] vs 57.2% [53.9-60.5], P < 0.0001), overall IBS Symptom Severity Score (323 [314-332] vs 144 [133-155], P < 0.0001) and health-rela,ted quality of life (50.4 [48.0-52.8] vs 74.6 [72.3-76.9], P < 0.0001). Improvement was similar across IBS subtypes. Symptoms improved most in the first 4 weeks of intervention. The most common side effects were mild gastrointestinal symptoms such as flatulence, abdominal pain and discomfort, and distension. DISCUSSION: Supplementation with 2 selected HMOs improves IBS symptoms and quality of life without substantial side effects. These promising results suggest that this novel approach to IBS should be confirmed in a randomized, placebo-controlled trial.


Subject(s)
Irritable Bowel Syndrome/drug therapy , Milk, Human/chemistry , Oligosaccharides/administration & dosage , Trisaccharides/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Defecation/drug effects , Defecation/physiology , Female , Humans , Irritable Bowel Syndrome/diagnosis , Irritable Bowel Syndrome/physiopathology , Irritable Bowel Syndrome/psychology , Male , Middle Aged , Oligosaccharides/adverse effects , Quality of Life , Severity of Illness Index , Treatment Outcome , Trisaccharides/adverse effects , Young Adult
18.
Anaerobe ; 61: 102076, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31326442

ABSTRACT

Prebiotics are widely used to shape a balanced microbiota in humans and animals. 1-Kestose (kestose) is one of the major components in commercialized short-chain fructooligosaccharide and is a promising prebiotic for infants. We herein studied the impact of kestose on the healthy adult microbiota in an in vitro fecal batch culture model. Stool samples obtained from seven healthy adults were diluted, inoculated into broth supplemented with or without 0.5% (w/v) kestose (kestose group and control group, respectively), and cultured under anaerobic conditions. Microbiota in the groups and stool samples were analyzed using 16S rRNA gene sequencing. At the phylum level, the kestose group showed increases in Bacteroidetes, whereas the control group showed increases in Proteobacteria. At the species level, Bifidobacterium longum was the only species showing significantly higher levels in the kestose group than in the control group and stool samples. On the other hand, levels of Escherichia coli were significantly higher in the control group than in stool samples, while the levels were not significantly different between the kestose group and stool samples. Quantitative PCR assays also revealed significantly higher levels of B. longum and lower tendency of E. coli in the kestose group than in the control group. These results suggest that supplementation with kestose increased the levels of beneficial microorganism and prevented the growth of risk-associated microorganisms related to disease development. Further interventional studies are needed to understand the health benefits of kestose in adult humans.


Subject(s)
Dietary Supplements , Feces/microbiology , Gastrointestinal Microbiome , Trisaccharides/administration & dosage , Adult , Age Factors , Female , Fermentation , Healthy Volunteers , Humans , Male , Metabolomics/methods , Metagenome , Metagenomics/methods , Young Adult
19.
J Vet Med Sci ; 82(1): 1-8, 2020 Jan 10.
Article in English | MEDLINE | ID: mdl-31761826

ABSTRACT

Kestose, a fructooligosaccharide (FOS) with one fructose monomer linked to sucrose, is a key component of the prebiotic activity of FOS. This study aimed to evaluate the prebiotic potential of Kestose in terms of the impact on population change in the intestinal microbiota and fecal short-chain fatty acid (SCFA) concentration in dogs. Kestose 2 g per dog was administered daily with conventional diet to 6 healthy, adult beagle dogs for 8 weeks followed by 4 weeks of follow-up period without Kestose supplementation. Fresh fecal samples were obtained before and every 4 weeks until the end of the follow-up period. Genomic DNA extracted from the fecal samples was subjected to 16S rRNA gene analysis using next generation sequencer and to quantitative polymerase chain reaction (qPCR). Fecal acetate, propionate, butyrate, lactate and ethanol concentrations were measured by high-performance liquid chromatography. 16S rRNA gene analysis and qPCR showed increasing trend of genus Bifidobacterium after Kestose supplementation while genera Bacteroides and Sutterella decreased. Clostridium perfringens decreased below the detection limit within first 4 weeks after starting Kestose supplementation. Fecal butyrate concentration was significantly increased at week 8 and returned to the base level after 4 weeks of the washing period. To the best of our knowledge, this is the first study to reveal effect of Kestose on the populational changes in fecal microbiota and fecal butyrate concentration in dogs.


Subject(s)
Animal Feed , Butyrates/metabolism , Gastrointestinal Microbiome , Trisaccharides/administration & dosage , Animals , Bifidobacterium/genetics , Dogs , Feces/chemistry , Feces/microbiology , Female , Male , Prebiotics/administration & dosage , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
20.
Nutrients ; 11(7)2019 Jun 26.
Article in English | MEDLINE | ID: mdl-31248026

ABSTRACT

BACKGROUND: We sought to determine whether an extensively hydrolyzed formula (EHF) supplemented with two human milk oligosaccharides (HMO) was tolerated by infants with cow's milk protein allergy (CMPA). METHODS: A whey-based EHF (Test formula) containing 2'fucosyl-lactose (2'FL) and lacto-N-neotetraose (LNnT) was assessed for clinical hypoallergenicity and safety. The Control formula was a currently marketed EHF without HMO. Children with CMPA, aged 2 months to 4 years, were assessed by double-blind, placebo-controlled food challenges (DBPCFC) to both formulas, in randomized order. If both DBPCFC were negative, subjects participated in a one-week, open food challenge (OFC) with the Test formula. Symptoms and adverse events were recorded. Hypoallergenicity was accepted if at least 90% (with 95% confidence intervals) of subjects tolerated the Test formula. RESULTS: Of the 82 children with CMPA that were screened, 67 (intention-to-treat [ITT] cohort-mean age 24.5 ± 13.6 months; range 2-57; 45 [67.2%] male) were randomized to receive either the Test or the Control formula during the first DBPCFC. Of these, 64 children completed at least one DBPCFC (modified intention-to-treat [mITT] cohort). Three children were excluded due to protocol deviations (per protocol [PP] cohort; n = 61). There was one allergic reaction to the Test, and one to the Control formula. On the mITT analysis, 63 out of 64 (98.4%; 95% CI lower bound 92.8%), and on the PP analysis 60 out of 61 (98.4%; 95% CI lower bound 92.5%) participants tolerated the Test formula, confirming hypoallergenicity. CONCLUSION: The whey-based EHF supplemented with 2'FL and LNnT met the clinical hypoallergenicity criteria and can be recommended for the management of CMPA in infants and young children.


Subject(s)
Infant Formula , Milk Hypersensitivity/therapy , Oligosaccharides/administration & dosage , Protein Hydrolysates/administration & dosage , Trisaccharides/administration & dosage , Whey Proteins/administration & dosage , Child, Preschool , Cross-Over Studies , Double-Blind Method , Female , Humans , Infant , Infant Formula/adverse effects , Male , Milk Hypersensitivity/diagnosis , Milk Hypersensitivity/immunology , Nutritive Value , Oligosaccharides/adverse effects , Oligosaccharides/immunology , Protein Hydrolysates/adverse effects , Protein Hydrolysates/immunology , Time Factors , Treatment Outcome , Trisaccharides/adverse effects , Trisaccharides/immunology , Whey Proteins/adverse effects , Whey Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...