Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Brain Behav Immun ; 119: 363-380, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38608741

ABSTRACT

The gut microbiota is altered in epilepsy and is emerging as a potential target for new therapies. We studied the effects of rifaximin, a gastrointestinal tract-specific antibiotic, on seizures and neuropathology and on alterations in the gut and its microbiota in a mouse model of temporal lobe epilepsy (TLE). Epilepsy was induced by intra-amygdala kainate injection causing status epilepticus (SE) in C57Bl6 adult male mice. Sham mice were injected with vehicle. Two cohorts of SE mice were fed a rifaximin-supplemented diet for 21 days, starting either at 24 h post-SE (early disease stage) or at day 51 post-SE (chronic disease stage). Corresponding groups of SE mice (one each disease stage) were fed a standard (control) diet. Cortical ECoG recording was done at each disease stage (24/7) for 21 days in all SE mice to measure the number and duration of spontaneous seizures during either rifaximin treatment or control diet. Then, epileptic mice ± rifaximin and respective sham mice were sacrificed and brain, gut and feces collected. Biospecimens were used for: (i) quantitative histological analysis of the gut structural and cellular components; (ii) markers of gut inflammation and intestinal barrier integrity by RTqPCR; (iii) 16S rRNA metagenomics analysis in feces. Hippocampal neuronal cell loss was assessed in epileptic mice killed in the early disease phase. Rifaximin administered for 21 days post-SE (early disease stage) reduced seizure duration (p < 0.01) and prevented hilar mossy cells loss in the hippocampus compared to epileptic mice fed a control diet. Epileptic mice fed a control diet showed a reduction of both villus height and villus height/crypt depth ratio (p < 0.01) and a decreased number of goblet cells (p < 0.01) in the duodenum, as well as increased macrophage (Iba1)-immunostaining in the jejunum (p < 0.05), compared to respective sham mice. Rifaximin's effect on seizures was associated with a reversal of gut structural and cellular changes, except for goblet cells which remained reduced. Seizure duration in epileptic mice was negatively correlated with the number of mossy cells (p < 0.01) and with villus height/crypt depth ratio (p < 0.05). Rifaximin-treated epileptic mice also showed increased tight junctions (occludin and ZO-1, p < 0.01) and decreased TNF mRNA expression (p < 0.01) in the duodenum compared to epileptic mice fed a control diet. Rifaximin administered for 21 days in chronic epileptic mice (chronic disease stage) did not change the number or duration of seizures compared to epileptic mice fed a control diet. Chronic epileptic mice fed a control diet showed an increased crypt depth (p < 0.05) and reduced villus height/crypt depth ratio (p < 0.01) compared to respective sham mice. Rifaximin treatment did not affect these intestinal changes. At both disease stages, rifaximin modified α- and ß-diversity in epileptic and sham mice compared to respective mice fed a control diet. The microbiota composition in epileptic mice, as well as the effects of rifaximin at the phylum, family and genus levels, depended on the stage of the disease. During the early disease phase, the abundance of specific taxa was positively correlated with seizure duration in epileptic mice. In conclusion, gut-related alterations reflecting a dysfunctional state, occur during epilepsy development in a TLE mouse model. A short-term treatment with rifaximin during the early phase of the disease, reduced seizure duration and neuropathology, and reversed some intestinal changes, strengthening the therapeutic effects of gut-based therapies in epilepsy.


Subject(s)
Disease Models, Animal , Epilepsy, Temporal Lobe , Gastrointestinal Microbiome , Mice, Inbred C57BL , Rifaximin , Seizures , Animals , Rifaximin/therapeutic use , Rifaximin/pharmacology , Mice , Male , Gastrointestinal Microbiome/drug effects , Seizures/drug therapy , Epilepsy, Temporal Lobe/drug therapy , Status Epilepticus/drug therapy , Brain/drug effects , Brain/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Epilepsy/drug therapy
2.
Nutrients ; 16(7)2024 Mar 24.
Article in English | MEDLINE | ID: mdl-38612971

ABSTRACT

Crohn's and ulcerative colitis are common conditions associated with inflammatory bowel disease as well as intestinal flora and epithelial barrier dysfunction. A novel fermented Lactobacillus brevis (AL0035) herein assayed in a trinitro benzene sulfonic acid (TNBS)-induced colitis mice model after oral administration significantly counteracted the body weight loss and improves the disease activity index and histological injury scores. AL0035 significantly decreased the mRNA and protein expression of different pro-inflammatory cytokines (TNFalpha, IL-1beta, IL-6, IL-12, IFN-gamma) and enhanced the expression of IL-10. In addition, the probiotic promoted the expression of tight junction proteins, such as ZO-1, keeping the intestinal mucosal barrier function to attenuate colitis symptoms in mice. Markers of inflammation cascade such as myeloperoxidase (MPO) and PPAR-gamma measured in the colon were also modified by AL0035 treatment. AL0035 was also able to reduce different lymphocyte markers' infiltration in the colon (GATA-3, T-Bet, NK1.1) and monocyte chemoattractant protein-1 (MCP-1/CCL2), a key chemokine involved in the migration and infiltration of monocytes/macrophages in the immunological surveillance of tissues and inflammation. In colonic microbiota profile analysis through 16S rRNA sequencing, AL0035 increased the microbial diversity depleted by TNBS administration and the relative abundance of the Lactobacillaceae and Lachnospiraceae families, whereas it decreased the abundance of Proteobacteria. Altogether, these data indicated that AL0035 could lower the severity of colitis induced by TNBS by regulating inflammatory cytokines, increasing the expression of tight junction proteins and modulating intestinal microbiota, thus preventing tissue damage induced by colitis.


Subject(s)
Colitis , Gastrointestinal Microbiome , Levilactobacillus brevis , Humans , Animals , Mice , Vegetables , RNA, Ribosomal, 16S , Colitis/chemically induced , Inflammation , Cytokines , Tight Junction Proteins/genetics
3.
NPJ Parkinsons Dis ; 9(1): 140, 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37783672

ABSTRACT

In patients with Parkinson's disease (PD), constipation is common, and it appears in a prodromal stage before the hallmark motor symptoms. The present study aimed to investigate whether Velusetrag, a selective 5­HT4 receptor agonist, may be a suitable candidate to improve intestinal motility in a mouse model of PD. Five months old PrP human A53T alpha-synuclein transgenic (Tg) mice, which display severe constipation along with decreased colonic cholinergic transmission already at 3 months, were treated daily with the drug for 4 weeks. Velusetrag treatment reduced constipation by significantly stimulating both the longitudinal and circular-driven contractions and improved inflammation by reducing the level of serum and colonic IL1ß and TNF-α and by decreasing the number of GFAP-positive glia cells in the colon of treated mice. No significant downregulation of the 5-HT4 receptor was observed but instead Velusetrag seemed to improve axonal degeneration in Tgs as shown by an increase in NF-H and VAChT staining. Ultimately, Velusetrag restored a well-balanced intestinal microbial composition comparable to non-Tg mice. Based on these promising data, we are confident that Velusetrag is potentially eligible for clinical studies to treat constipation in PD patients.

4.
Gels ; 9(8)2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37623062

ABSTRACT

An innovative and stable probiotic-containing mucoadhesive gel (AL0020), integrated with botanical extracts, has been developed to rebalance the dysbiosis associated with periodontal diseases. Tau-Marin gel, prepared with anhydrous ingredients to prevent the replication of bacteria and ensure good stability over time, was tested against some pathogenic bacteria, belonging to the so-called "red complex", recognized as the most important pathogens in plaque specimens, adherent to the epithelial lining of periodontal pockets. This lipogel was tested in vitro, in a physiological solution (PS) and in a simulated saliva (SS), for up to 8 h, to monitor its ability to release probiotics over time. Probiotics were enumerated through two different techniques, Lacto-Counter Assay (LCA) and Colony Forming Unit (CFU). A detailed physico-chemical profile of AL0020 and its in vitro efficacy in protecting activity against pathogenic bacteria as well as soothing or irritative effect on gingival epithelium were reported. Moreover, a clinical-dermatological trial on 20 volunteers using the product once a day for 30 days was also performed, where the efficacy of the gel in the control of gum disorders was observed.

5.
Int J Mol Sci ; 24(10)2023 May 15.
Article in English | MEDLINE | ID: mdl-37240150

ABSTRACT

Pelvic radiation disease (PRD), a frequent side effect in patients with abdominal/pelvic cancers treated with radiotherapy, remains an unmet medical need. Currently available preclinical models have limited applications for the investigation of PRD pathogenesis and possible therapeutic strategies. In order to select the most effective irradiation protocol for PRD induction in mice, we evaluated the efficacy of three different locally and fractionated X-ray exposures. Using the selected protocol (10 Gy/day × 4 days), we assessed PRD through tissue (number and length of colon crypts) and molecular (expression of genes involved in oxidative stress, cell damage, inflammation, and stem cell markers) analyses at short (3 h or 3 days after X-ray) and long (38 days after X-rays) post-irradiation times. The results show that a primary damage response in term of apoptosis, inflammation, and surrogate markers of oxidative stress was found, thus determining a consequent impairment of cell crypts differentiation and proliferation as well as a local inflammation and a bacterial translocation to mesenteric lymph nodes after several weeks post-irradiation. Changes were also found in microbiota composition, particularly in the relative abundance of dominant phyla, related families, and in alpha diversity indices, as an indication of dysbiotic conditions induced by irradiation. Fecal markers of intestinal inflammation, measured during the experimental timeline, identified lactoferrin, along with elastase, as useful non-invasive tools to monitor disease progression. Thus, our preclinical model may be useful to develop new therapeutic strategies for PRD treatment.


Subject(s)
Radiation Injuries , Mice , Animals , X-Rays , Disease Models, Animal , Apoptosis/radiation effects , Inflammation
6.
Int J Dent ; 2022: 5007930, 2022.
Article in English | MEDLINE | ID: mdl-35265133

ABSTRACT

Objective: Periodontitis is a common disorder that leads to the loss of both tooth and personal well-being, contributing to worsen the risk for metabolic and cardiovascular diseases. Recently, probiotics, characterized by rapid oral dispersion, have been topically used. Here, we present data of a mucoadhesive gel containing probiotics, capable of ensuring a slow release of bacteria to prevent and treat periodontitis. Methods: An original mucoadhesive gel (AL0005) that is anhydrous and of food grade, loaded with the blend of lactobacilli and plants' dry extracts, has been assayed. Results: The release kinetics of the bacterial mixture in different experimental models in vitro, including simulated saliva or physiological solutions, showed a significant and stable release for 5-8 hours. In one in vivo study of a mouse model of periodontitis, a locally applied mucoadhesive gel enriched with probiotic strains improved significantly the tissue pathology when compared with vehicle-exposed mice. Conclusions: Together, the results suggest that this mucoadhesive gel can be useful in the normalization of the gum bacterial flora and improvement of the tissue pathology of gum disorders.

7.
Neurobiol Stress ; 10: 100138, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30937345

ABSTRACT

Stress affects the immune system and intestinal microbiota composition and can lead to imbalance between pro- and anti-inflammatory cytokines or to uncontrolled production of cytokines. The effect of emotional stress on secretory IgA levels also indicates that stress decreases mucosal integrity. Our aim was to evaluate whether a probiotic product (Lactoflorene® Plus) can prevent alterations in the immune response associated with self-reported stress and microbiota composition. Healthy adult volunteers who self-reported psychological stress were enrolled and randomised into a placebo and a probiotic group. Salivary stress markers (α-amylase, cortisol, chromogranin A) and immunological parameters (sIgA, NK cell activity, IL-8, IL-10, TNF-α) in feces and the composition of intestinal microbiota were evaluated. Administration of the product did not exert a direct effect on the salivary stress markers or NK cell activity but did reduce abdominal pain and increase faecal IgA and IL-10 levels. The probiotic product induced a moderate increase in Bifidobacterium and Lactobacillus spp., as expected, and in Faecalibacterium spp., and decreased the size of the Dialister spp. and Escherichia and Shigella populations. Administration of the product helped protect the mucosal barrier by supporting the number of short-chain fatty acid producers and decreasing the load of potentially harmful bacteria, thus reducing intestinal inflammation and abdominal discomfort. CLINICALTRIALSGOV: NCT03234452.

8.
J Nutr ; 148(8): 1300-1308, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29982534

ABSTRACT

Background: Inulin-type fructans used in formula have been shown to promote microbiota composition and stool consistency closer to those of breastfed infants and to have beneficial effects on fever occurrence, diarrhea, and incidence of infections requiring antibiotic treatment in infants. Objectives: The primary study aim was to explore whether prophylactic supplementation with prebiotic fructans is able to influence the frequency of infectious diseases in kindergarten children during a winter period. A secondary objective was to ascertain the effect on the intestinal microbiota. Methods: 142 boys and 128 girls aged 3-6 y were randomly allocated to consume 6 g/d fructans or maltodextrin for 24 wk. At baseline, stool samples were collected for microbiota analysis and anthropometric measurements were made. During the intervention period diagnoses were recorded by physicians, whereas disease symptoms, kindergarten absenteeism, dietary habits, and stool consistency were recorded by parents. Baseline measurements were repeated at wk 24. Results: In total 219 children finished the study. Both the relative abundance of Bifidobacterium (P < 0.001) and that of Lactobacillus (P = 0.014) were 19.9% and 7.8% higher, respectively, post data normalization, in stool samples of children receiving fructans as compared with those of controls at wk 24. This was accompanied by significantly softer stools within the normal range in the prebiotic group from wk 12 onwards. The incidence of febrile episodes requiring medical attention [0.65 ± 1.09 compared with 0.9 ± 1.11 infections/(24 wk × child), P = 0.04] and that of sinusitis (0.01 ± 0.1 compared with 0.06 ± 0.25, P = 0.03) were significantly lower in the prebiotic group. The number of infectious episodes and their duration reported by parents did not differ significantly between the 2 intervention groups. Conclusions: Prebiotic supplementation modified the composition of the intestinal microbiota and resulted in softer stools in kindergarten-aged children. The reduction in febrile episodes requiring medical attention supports the concept of further studies on prebiotics in young children. This trial was registered at clinicaltrials.gov as NCT03241355.


Subject(s)
Bifidobacterium/growth & development , Feces/microbiology , Fructans/therapeutic use , Infections , Inulin/therapeutic use , Prebiotics , Severity of Illness Index , Child , Child, Preschool , Colon/microbiology , Female , Fever/etiology , Fructans/pharmacology , Gastrointestinal Microbiome , Humans , Incidence , Infections/complications , Inulin/pharmacology , Lactobacillus/growth & development , Male , Sinusitis/prevention & control
9.
J Clin Gastroenterol ; 52 Suppl 1, Proceedings from the 9th Probiotics, Prebiotics and New Foods, Nutraceuticals and Botanicals for Nutrition & Human and Microbiota Health Meeting, held in Rome, Italy from September 10 to 12, 2017: S66-S67, 2018.
Article in English | MEDLINE | ID: mdl-29863587
10.
Clin Exp Gastroenterol ; 8: 309-25, 2015.
Article in English | MEDLINE | ID: mdl-26673000

ABSTRACT

Rifaximin, with its low systemic absorption, may represent a treatment of choice for irritable bowel syndrome (IBS), mainly due to its ability to act on IBS pathogenesis, through the influence on gut microbiota. The aim of the present study was to assess, by biomolecular tools, the rifaximin active modulation exerted on gut microbiota of non-constipated IBS patients. Fifteen non-constipated IBS subjects were treated with 550 mg rifaximin three times a day for 14 days. Stool samples were collected before starting the treatment, at the end of it, and after a 6-week washout period. Real-time polymerase chain reaction, denaturing gradient gel electrophoresis, and next-generation sequencing were applied to all the samples to verify and quantify possible microbial fluctuations. Rifaximin treatment did not affect the overall composition of the microbiota of the treated subjects, inducing fluctuations in few bacterial groups, balanced by the replacement of homologs or complementary bacterial groups. Rifaximin appeared to influence mainly potentially detrimental bacteria, such as Clostridium, but increasing the presence of some species, such as Faecalibacterium prausnitzii. A decrease in the Firmicutes/Bacteroidetes ratio after 14 days of treatment and bacterial profiles with higher biodiversity were observed during the follow-up compared to baseline. Rifaximin treatment, although effective on IBS symptom relief and normalization of lactulose breath test, did not induce dramatic shifts in the microbiota composition of the subjects, stimulating microbial reorganization in some populations toward a more diverse composition. It was not possible to speculate on differences of fecal microbiota modification between responders vs nonresponders and to correlate the quali-/quantitative modification of upper gastrointestinal microbiota and clinical response.

11.
J Am Coll Nutr ; 33(5): 385-93, 2014.
Article in English | MEDLINE | ID: mdl-25302927

ABSTRACT

OBJECTIVE: The objective of the study was to investigate the effects of a galacto-oligosaccharides (GOS)-supplemented formula on the intestinal microbiota in healthy term infants, with a specific consideration for gastrointestinal symptoms as colic, stool frequency and consistency, regurgitation. METHODS: This was a randomized, double-blind, controlled, parallel-group clinical trial performed simultaneously by 6 centers in Italy. Three groups were considered: breastfed, formula-fed, and GOS-supplemented formula-fed infants. Formula-fed infants were randomized to receive either the control or the study formula and consume the assigned formula exclusively until the introduction of complementary feeding. The nutritional composition of the 2 formulas were identical, apart from the supplemented GOS (0.4 g/100 mL) in the study formula. Four different types of bacteria were evaluated in order to assess the efficacy of GOS-supplemented formula on infants: Bifidobacterium, Lactobacillus, and Clostridium, Escherichia coli. RESULTS: A total of 199 breastfed infants and 163 formula-fed infants were recruited. When considering stool frequency and consistency, GOS-supplemented formula presented normal and soft stools in the majority of episodes (89%). In the supplemented group the incidence of colic was lower with respect to the control group. A significantly lower count of Clostridium and a higher count of Bifidobacterium were found when comparing study formula and control formula in infants with colic. In children with colic the ratio between Clostridium count and Bifidobacterium and Lactobacillus count was in favor of the latter two when considering the GOS-supplemented formula group with respect to the control one. CONCLUSIONS: The prebiotic-supplemented formula mimicked the effect of human milk in promoting Bifidobacterium and Lactobacillus growth and in inhibiting Clostridium growth, resulting in a significantly lower presence of colic.


Subject(s)
Bacteria/drug effects , Colic/prevention & control , Defecation/drug effects , Infant Formula , Intestines/drug effects , Oligosaccharides/pharmacology , Prebiotics , Bacteria/growth & development , Breast Feeding , Colic/microbiology , Dietary Supplements , Double-Blind Method , Feces/microbiology , Female , Galactose/pharmacology , Humans , Infant , Infant Nutritional Physiological Phenomena , Intestines/microbiology , Italy , Male , Milk, Human
12.
J Pediatr Gastroenterol Nutr ; 53(1): 80-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21478759

ABSTRACT

OBJECTIVES: The aim of this study was to identify a link between the total amount of breast milk oligosaccharides and faecal microbiota composition of newborns at the end of the first month of life, with special attention paid to bifidobacteria, and establish the role, if any, of the different oligosaccharides in determining the gut microbiota composition. SUBJECTS AND METHODS: Milk oligosaccharide groups were identified by high-performance anion exchange chromatography analysis. DPCRNA from newborns' faecal samples at 30 days of life was isolated and processed by polymerase chain reaction analyses that allow the identification of 6 species of bifidobacteria (adolescentis, bifidum, breve, catenulatum, longum, infantis) and Ruminococcus spp; denaturing gradient gel electrophoresis analysis was also performed. RESULTS: No substantial differences in bifidobacteria species composition within milk groups 1, 2, and 3 were observed; however, infants fed with group 4 milk show a microbiota characterised by a greater frequency of Bifidobacteria adolescentis and the absence of Bifidobacteria catenulatum. For the first time, a high percentage of the Ruminococcus genus in infants fed with all milk groups was found. CONCLUSIONS: Our data show that milk groups 1, 2, and 3, containing an amount of oligosaccharides ranging within 10 to 15 g/L, share a substantially identical composition of the intestinal microbiota in breast-fed infants, despite quali-quantitative difference in oligosaccharides content. Newborns taking milk with only 5 g/L of oligosaccharides (group 4) harbour a different intestinal microbiota.


Subject(s)
Bifidobacterium/metabolism , Breast Feeding , Feces/microbiology , Milk, Human/metabolism , Oligosaccharides/metabolism , Polysaccharides, Bacterial/metabolism , Ruminococcus/metabolism , Adult , Anion Exchange Resins , Bifidobacterium/classification , Bifidobacterium/isolation & purification , Chromatography, High Pressure Liquid , DNA, Bacterial/isolation & purification , DNA, Bacterial/metabolism , Denaturing Gradient Gel Electrophoresis , Humans , Infant, Newborn , Italy , Lewis Blood Group Antigens/metabolism , Male , Molecular Typing , Oligosaccharides/chemistry , Pilot Projects , Polymerase Chain Reaction , Polysaccharides, Bacterial/chemistry , Ruminococcus/classification , Ruminococcus/isolation & purification
13.
J Clin Gastroenterol ; 42 Suppl 3 Pt 2: S174-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18685505

ABSTRACT

GOAL: To assess the effects of the consumption of psyllium seed husk on fecal bifidobacteria in healthy women and the ability of fecal bifidobacteria to metabolize psyllium seed husk in vitro. BACKGROUND: Poor microbiologic evidences are nowadays available concerning the ability of psyllium seed husk to promote the growth of bifidobacteria in human gut. STUDY: Eleven healthy women consumed 7.0 g/d of psyllium seed husk for 1 month. Viability of bifidobacteria in feces was assessed at different time points. RESULTS: In vivo results showed that the average fecal content of viable bifidobacteria was not significantly affected even if fecal counts were found to increase significantly after treatment in 6 out of 11 women having low initial concentration. In vitro trials conducted on bifidobacteria strains isolated from treated women failed to confirm the prebiotic potential of undigested psyllium seed husk, whereas treatment with simulated gastric and pancreatic juices and mimicking physical and chemical alterations during human gut transit allowed fecal Bifidobacterium isolates to metabolize psyllium seed husk as carbon source in a growth medium deprived of sugar. CONCLUSIONS: Psyllium seed husk can be metabolized by bifidobacteria only after partial hydrolysis. Bifidogenic potential can be detected in healthy women only in case of low level of fecal bifidobacteria before treatment.


Subject(s)
Bifidobacterium/metabolism , Dietary Fiber , Psyllium , Adult , Bifidobacterium/drug effects , Bifidobacterium/growth & development , Bifidobacterium/isolation & purification , Colony Count, Microbial , Dietary Fiber/administration & dosage , Dietary Fiber/pharmacology , Feces/microbiology , Female , Humans , Middle Aged , Psyllium/administration & dosage , Psyllium/pharmacology , Seeds , Time Factors
14.
Appl Environ Microbiol ; 72(7): 5113-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16820518

ABSTRACT

Whether Lactobacillus delbrueckii subsp. bulgaricus and Streptococcus thermophilus can be recovered after passage through the human gut was tested by feeding 20 healthy volunteers commercial yogurt. Yogurt bacteria were found in human feces, suggesting that they can survive transit in the gastrointestinal tract.


Subject(s)
Gastrointestinal Tract/microbiology , Lactobacillus delbrueckii/growth & development , Probiotics , Streptococcus thermophilus/growth & development , Yogurt/microbiology , Adult , Colony Count, Microbial , Electrophoresis, Gel, Pulsed-Field , Feces/microbiology , Female , Humans , Lactobacillus delbrueckii/genetics , Lactobacillus delbrueckii/isolation & purification , Male , Polymerase Chain Reaction/methods , Streptococcus thermophilus/genetics , Streptococcus thermophilus/isolation & purification
15.
J Clin Gastroenterol ; 38(6 Suppl): S80-3, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15220665

ABSTRACT

The development of intestinal microflora in newborns is strictly related to the kind of feeding. Breast-fed infants, unlike the bottle-fed ones, have an intestinal ecosystem characterized by a strong prevalence of bifidobacteria and lactobacilli. Data available so far in the literature show that, among the numerous substances present in human milk, oligosaccharides have a clear prebiotic effect. They are quantitatively one of the main components of human milk and are only partially digested in the small intestine, so they reach the colon, where they stimulate selectively the development of bifidogenic flora. Such results have been recently proved both by characterization of oligosaccharides in breast-fed infant feces and by the study of intestinal microflora using new techniques of molecular analysis, confirming that human milk oligosaccharides represent the first prebiotics in humans.


Subject(s)
Intestines/microbiology , Milk, Human/chemistry , Oligosaccharides/pharmacology , Bifidobacterium/growth & development , Breast Feeding , Humans , Infant, Newborn
SELECTION OF CITATIONS
SEARCH DETAIL
...