Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Compr Rev Food Sci Food Saf ; 23(3): e13356, 2024 May.
Article in English | MEDLINE | ID: mdl-38767859

ABSTRACT

Recently, the role of the gut microbiota in metabolic health, immunity, behavioral balance, longevity, and intestine comfort has been the object of several studies from scientific communities. They were encouraged by a growing interest from food industries and consumers toward novel fermented ingredients and formulations with powerful biological effects, such as pre, pro, and postbiotic products. Depending on the selected strains, the operating conditions, the addition of suitable reagents or enzymes, the equipment, and the reactor configurations, functional compounds with high bioactivity, such as short-chain fatty acids, gamma-aminobutyric acid, bioactive peptides, and serotonin, can be enhanced and/or produced through fermentation of several vegetable matrices. Otherwise, their formation can also be promoted directly in the gut after the dietary intake of fermented foods: In this case, fermentation will aim to increase the content of precursor substances, such as indigestible fibers, polyphenols, some amino acids, and resistant starch, which can be potentially metabolized by endogenous gut microorganisms and converted in healthy molecules. This review provides an overview of the main functional components currently investigated in literature and the associated gut health benefits. The current state of the art about fermentation technology as a promising functionalization tool to promote the direct or indirect formation of gut-health-enhancing components was deepened, highlighting the importance of optimizing microorganism selection, system setups, and process conditions according to the target compound of interest. The collected data suggested the possibility of gaining novel functional food ingredients or products rich in functional molecules through fermentation without performing additional extraction and purification stages, which are needed when conventional culture broths are used.


Subject(s)
Fermentation , Gastrointestinal Microbiome , Gastrointestinal Microbiome/physiology , Humans , Fermented Foods/microbiology , Dietary Fiber
2.
Front Nutr ; 10: 1104617, 2023.
Article in English | MEDLINE | ID: mdl-36819696

ABSTRACT

Reformulating packaged foods has the potential to improve the nutrient density of the global diet. The present perspective illustrates The Kraft Heinz Company's approach to product (re)formulation to develop healthier product lines that are lower in saturated fats, total sugars, and sodium, and contain health promoting components. Here we present the rationale for The Kraft Heinz Company's global nutrition targets used for the global innovation and renovation of foods and beverages. The global nutrition targets use a category specific approach to set maximum levels for the main nutrients of public health concern: saturated fat, total sugars and sodium, taking into account product characteristics (typical portion size, eating occasion, role in the diet, etc.) as well as regulatory, technological, sensory and safety constraints. Benchmarking examples illustrate how the nutrition targets are positioned within the United States, France, and Australia. These global nutrition targets serve as part of The Kraft Heinz Company's environmental, social and governance nutrition commitments and demonstrates how the food industry is improving the nutritional value of packaged foods and beverages both now and into the future.

3.
Int J Mol Sci ; 23(7)2022 Mar 26.
Article in English | MEDLINE | ID: mdl-35409015

ABSTRACT

Celiac disease (CD) is an autoimmune disease characterized by an altered immune response stimulated by gliadin peptides that are not digested and cause damage to the intestinal mucosa. The aim of this study was to investigate whether the postbiotic Lactobacillus paracasei (LP) could prevent the action of gliadin peptides on mTOR, autophagy, and the inflammatory response. Most of the experiments performed were conducted on intestinal epithelial cells Caco-2 treated with a peptic-tryptic digest of gliadin (PTG) and P31-43. Furthermore, we pretreated the Caco-2 with the postbiotic LP before treatment with the previously described stimuli. In both cases, we evaluated the levels of pmTOR, p70S6k, and p4EBP-1 for the mTOR pathway, pNFkß, and pERK for inflammation and LC 3 and p62 for autophagy. For autophagy, we also used immunofluorescence analysis. Using intestinal organoids derivate from celiac (CD) patients, we analyzed the effect of gliadin after postbiotic pretreatment with LP on inflammation marker NFkß. Through these experiments, we showed that gliadin peptides are able to induce the increase of the inflammatory response in a more complex model of intestinal epithelial cells. LP postbiotic was able to induce autophagy in Caco-2 cells and prevent gliadin effects. In conclusion, postbiotic pretreatment with LP could be considered for in vivo clinical trials.


Subject(s)
Celiac Disease , Lacticaseibacillus paracasei , Autophagy , Caco-2 Cells , Gliadin/chemistry , Humans , Inflammation/metabolism , Intestinal Mucosa/metabolism , Lacticaseibacillus paracasei/metabolism , Peptide Fragments/metabolism , Peptides/pharmacology , TOR Serine-Threonine Kinases/metabolism
4.
Heliyon ; 6(9): e04920, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32984612

ABSTRACT

Studies of the ability of probiotics to ferment cereal flours are necessary to obtain products with enhanced nutritional value. In this study, Lactobacillus paracasei CBA-L74 was used to ferment cereal aqueous mixtures containing both oat (7.5% w/v) and rice flours (7.5% w/v), with and without glucose, to understand whether glucose addition could have any effect on growth and metabolism. Viability, pH, metabolites production during fermentation (24 h, 37 °C) and substrates reduction were analysed. The strain showed good growth in the cereal aqueous mixture both with and without glucose addition, but suspensions prepared with glucose showed the best results. A bacterial concentration of 7 log CFU mL-1, a pH value of 4.70 and lactic acid production of 1250 mg L-1 were achieved when fermentation was performed without glucose addition, while in the presence of glucose, a t24 bacterial growth of 8 log CFU mL-1 was reached, with a pH value of 3.11 and lactic acid production of 6050 mg L-1.

5.
Nat Commun ; 11(1): 2703, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32483147

ABSTRACT

Mother's milk is the best choice for infants nutrition, however when it is not available or insufficient to satisfy the needs of the infant, formula is proposed as an effective substitute. Here, we report the results of a randomized controlled clinical trial (NCT03637894) designed to evaluate the effects of two different dietary regimens (standard formula and Lactobacillus paracasei CBA L74-fermented formula) versus breastfeeding (reference group) on immune defense mechanisms (primary endpoint: secretory IgA, antimicrobial peptides), the microbiota and its metabolome (secondary outcomes), in healthy full term infants according to the type of delivery (n = 13/group). We show that the fermented formula, safe and well tolerated, induces an increase in secretory IgA (but not in antimicrobial peptides) and reduces the diversity of the microbiota, similarly, but not as much as, breastmilk. Metabolome analysis allowed us to distinguish subjects based on their dietary regimen and mode of delivery. Together, these results suggest that a fermented formula favors the maturation of the immune system, microbiota and metabolome.


Subject(s)
Diet , Immune System/physiology , Metabolome/physiology , Microbiota/physiology , Antimicrobial Cationic Peptides/metabolism , Breast Feeding , Double-Blind Method , Feces/microbiology , Female , Fermentation , Humans , Immunoglobulin A, Secretory/metabolism , Infant Formula , Infant, Newborn , Lacticaseibacillus paracasei/metabolism , Male , Milk, Human , beta-Defensins/metabolism , Cathelicidins
6.
Cell Microbiol ; 21(8): e13035, 2019 08.
Article in English | MEDLINE | ID: mdl-31042331

ABSTRACT

We previously identified a Neisseria flavescens strain in the duodenum of celiac disease (CD) patients that induced immune inflammation in ex vivo duodenal mucosal explants and in CaCo-2 cells. We also found that vesicular trafficking was delayed after the CD-immunogenic P31-43 gliadin peptide-entered CaCo-2 cells and that Lactobacillus paracasei CBA L74 (L. paracasei-CBA) supernatant reduced peptide entry. In this study, we evaluated if metabolism and trafficking was altered in CD-N. flavescens-infected CaCo-2 cells and if any alteration could be mitigated by pretreating cells with L. paracasei-CBA supernatant, despite the presence of P31-43. We measured CaCo-2 bioenergetics by an extracellular flux analyser, N. flavescens and P31-43 intracellular trafficking by immunofluorescence, cellular stress by TBARS assay, and ATP by bioluminescence. We found that CD-N. flavescens colocalised more than control N. flavescens with early endocytic vesicles and more escaped autophagy thereby surviving longer in infected cells. P31-43 increased colocalisation of N. flavescens with early vesicles. Mitochondrial respiration was lower (P < .05) in CD-N. flavescens-infected cells versus not-treated CaCo-2 cells, whereas pretreatment with L. paracasei-CBA reduced CD-N. flavescens viability and improved cell bioenergetics and trafficking. In conclusion, CD-N. flavescens induces metabolic imbalance in CaCo-2 cells, and the L. paracasei-CBA probiotic could be used to correct CD-associated dysbiosis.


Subject(s)
Lacticaseibacillus paracasei/chemistry , Mitochondria/drug effects , Neisseria/drug effects , Oxidative Phosphorylation/drug effects , Probiotics/pharmacology , Adenosine Triphosphate/agonists , Adenosine Triphosphate/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/microbiology , Autophagy/drug effects , Autophagy/genetics , Caco-2 Cells , Celiac Disease/metabolism , Celiac Disease/microbiology , Celiac Disease/therapy , Culture Media, Conditioned/pharmacology , Dysbiosis/metabolism , Dysbiosis/microbiology , Dysbiosis/therapy , Gene Expression , Gliadin/antagonists & inhibitors , Gliadin/pharmacology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Lacticaseibacillus paracasei/physiology , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Neisseria/genetics , Neisseria/growth & development , Neisseria/pathogenicity , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Thiobarbituric Acid Reactive Substances/metabolism , Transport Vesicles/drug effects , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
7.
Int J Food Sci Nutr ; 70(8): 950-958, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30969137

ABSTRACT

Coeliac disease is an increasingly recognised pathology, induced by the ingestion of gluten in genetically predisposed patients. Undigested gliadin peptide can induce adaptive and innate immune response that unleash the typical intestinal mucosal alterations. A growing attention is paid to alternative therapeutic approaches to the gluten-free diet: one of these approaches is the use of probiotics and/or postbiotics. We performed lactic fermentation of rice flour with and without pH control, using Lactobacillus paracasei CBA L74 as fermenting strain. We evaluated bacterial growth, lactic acid production during fermentation and gliadin peptide P31-43 entrance in CaCo-2 cells with and without pH control. When pH control was applied no differences were observed in terms of bacterial growth; on the contrary, lactic acid production was greater, as expected. Both samples could inhibit the P31-43 entrance in CaCo-2 cells but the effect was significantly greater for samples obtained when the pH control was applied.


Subject(s)
Epithelial Cells/metabolism , Fermentation , Gliadin/metabolism , Hydrogen-Ion Concentration , Oryza/microbiology , Peptide Fragments/metabolism , Caco-2 Cells , Celiac Disease/drug therapy , Celiac Disease/prevention & control , Diet, Gluten-Free , Food Hypersensitivity/prevention & control , Functional Food , Gliadin/antagonists & inhibitors , Glutens , Humans , Lactic Acid/metabolism , Lacticaseibacillus paracasei/metabolism , Oryza/metabolism , Peptide Fragments/antagonists & inhibitors
8.
J Pediatr ; 194: 116-122.e2, 2018 03.
Article in English | MEDLINE | ID: mdl-29478494

ABSTRACT

OBJECTIVE: To evaluate the long-term validity and safety of pure oats in the treatment of children with celiac disease. STUDY DESIGN: This noninferiority clinical trial used a double-blind, placebo-controlled, crossover design extended over 15 months. Three hundred six children with a biopsy-proven diagnosis of celiac disease on a gluten-free diet for ≥2 years were randomly assigned to eat specifically prepared gluten-free food containing an age-dependent amount (15-40 g) of either placebo or purified nonreactive varieties of oats for 2 consecutive 6-month periods separated by washout standard gluten-free diet for 3 months. Clinical (body mass index, Gastrointestinal Symptoms Rating Scale score), serologic (IgA antitransglutaminase antibodies, and IgA anti-avenin antibodies), and intestinal permeability data were measured at baseline, and after 6, 9, and 15 months. Direct treatment effect was evaluated by a nonparametric approach using medians (95% CI) as summary statistic. RESULTS: After the exclusion of 129 patients who dropped out, the cohort included 177 children (79 in the oats-placebo and 98 in the placebo-oats group; median, 0.004; 95% CI, -0.0002 to 0.0089). Direct treatment effect was not statistically significant for clinical, serologic, and intestinal permeability variables (body mass index: median, -0.5; 95% CI, -0.12 to 0.00; Gastrointestinal Symptoms Rating Scale score: median, 0; 95% CI, -2.5 to 0.00; IgA antitransglutaminase antibodies: median, -0.02; 95% CI, -0.25 to 0.23; IgA anti-avenin antibodies: median, -0.0002; 95% CI, -0.0007 to 0.0003; intestinal permeability test: median, 0.004; 95% CI, -0.0002 to 0.0089). CONCLUSIONS: Pure nonreactive oat products are a safe dietary choice in the treatment of children with celiac disease. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00808301.


Subject(s)
Avena/adverse effects , Celiac Disease/diet therapy , Celiac Disease/immunology , Child , Cross-Over Studies , Diet, Gluten-Free , Double-Blind Method , Female , Humans , Intestinal Mucosa/immunology , Male
9.
PLoS One ; 12(12): e0189807, 2017.
Article in English | MEDLINE | ID: mdl-29261742

ABSTRACT

BACKGROUND: In vivo assays cannot always be conducted because of ethical reasons, technical constraints or costs, but a better understanding of the digestive process, especially in infants, could be of great help in preventing food-related pathologies and in developing new formulas with health benefits. In this context, in vitro dynamic systems to simulate human digestion and, in particular, infant digestion could become increasingly valuable. OBJECTIVE: To simulate the digestive process through the use of a dynamic model of the infant gastroenteric apparatus to study the digestibility of starch-based infant foods. DESIGN: Using M.I.D.A (Model of an Infant Digestive Apparatus), the oral, gastric and intestinal digestibility of two starch-based products were measured: 1) rice starch mixed with distilled water and treated using two different sterilization methods (the classical method with a holding temperature of 121°C for 37 min and the HTST method with a holding temperature of 137°C for 70 sec) and 2) a rice cream with (premium product) or without (basic product) an aliquot of rice flour fermented by Lactobacillus paracasei CBA L74. After the digestion the foods were analyzed for the starch concentration, the amount of D-glucose released and the percentage of hydrolyzed starch. RESULTS: An in vitro dynamic system, which was referred to as M.I.D.A., was obtained. Using this system, the starch digestion occurred only during the oral and intestinal phase, as expected. The D-glucose released during the intestinal phase was different between the classical and HTST methods (0.795 grams for the HTST versus 0.512 for the classical product). The same analysis was performed for the basic and premium products. In this case, the premium product had a significant difference in terms of the starch hydrolysis percentage during the entire process. CONCLUSIONS: The M.I.D.A. system was able to digest simple starches and a more complex food in the correct compartments. In this study, better digestibility of the premium product was revealed.


Subject(s)
Gastrointestinal Tract/metabolism , Models, Biological , Body Fluids , Digestion , Electrolytes/chemistry , Fermentation , Glucose/metabolism , Humans , Hydrolysis , Infant , Oryza/chemistry , Starch/metabolism , Sterilization
10.
Clin Nutr ; 36(1): 238-245, 2017 02.
Article in English | MEDLINE | ID: mdl-26718667

ABSTRACT

BACKGROUND & AIMS: Safety and growth adequacy of infant formulae enriched by functional ingredients need stringent evaluation by means of randomized controlled trials (RCTs), therefore we performed a double-blind RCT to evaluate an infant formula enriched with galacto-oligosaccharides, beta-palmitate, and acidified milk vs. a standard infant formula. METHODS: Weight, length, head circumference and fecal bacteria (Bifidobacteria, BIF/Clostridia, CLO) were measured in healthy full term infants, at baseline - as before 21 days of life - at 60 and 135 days thereafter. A group of 51 neonates received the enriched formula (ENR), 59 the standard one (ST). Parents were trained to daily register gastrointestinal diseases. RESULTS: All the infants grew homogeneously increasing the anthropometric parameters and complying with WHO and Italian standards: the mean (SD) difference in daily weight between ENR and ST groups was -0.74 (1.13) g/day, corresponding to a 90% CI of -2.62 to 1.13 g/day, well within the postulated interval of equivalence of -3.9 to +3.9 g/day. A statistical improvement in BIF concentration in the microbiota of infants fed by ENR was recorded. There was no between-group change in log10CLO, but log10BIF increase was higher at T2 vs. T0 in ENR (treatment × time interaction = 0.71, 95% CI 0.08-1.34, p = 0.028) than in ST neonates. This corresponds to estimated mean (95% CI) values of 8.37 (8.04-8.69) log10-units for ENR vs. 8.08 (7.77-8.39) log10-units for ST neonates. Gastrointestinal effects were mild and similar, with no statistical difference between two groups. CONCLUSION: Safety and growth ability of the enriched formula has been confirmed. A positive effect on neonatal gut microbiota, consisting of increased fecal BIF counts at T2 vs. baseline has been shown too. Nonetheless, larger RCTs are needed to estimate with greater precision the effective potential attributable to the enriched formula on neonatal microbiota, with particular reference to the mode of delivery.


Subject(s)
Food Safety , Functional Food/analysis , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Infant Formula/chemistry , Bifidobacterium , Clostridium , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Dietary Fiber/administration & dosage , Dietary Proteins/administration & dosage , Double-Blind Method , Humans , Infant , Infant Formula/microbiology , Micronutrients/administration & dosage , Treatment Outcome
11.
J Proteome Res ; 15(10): 3712-3723, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27650928

ABSTRACT

NMR-based metabolomics was used to compare the metabolic urinary profiles of exclusively breast-fed term infants (n = 11) with those of a double-blinded controlled trial with 49 formula-fed term newborns randomized to receive either an infant formula enriched by functional ingredients (n = 24) or a standard formula (n = 25). Anthropometric measurements and urine samples were taken at enrollment (within the first month of life), at around 60 days of life, and at the end of study period (average age of 130 days). The metabolic profiles were examined in relation to time and diet strategy. A common age-dependent modification of the urine metabolome was observed for the three types of nutrition, mainly characterized by similar temporal trends of choline, betaine, myoinositol, taurine, and citrate. Contrariwise, differences in the metabolic profiles were identified according to the type of diet (human versus formula milk), while no significant difference was observed between the two formulas. These modifications are discussed mainly in terms of the different milk compositions. Despite the low number of enrolled infants (n = 60), these findings pointed out the potential of the metabolomics approach for neonatal nutritional science, in particular to provide important contributions to the optimization of formula milk.


Subject(s)
Breast Feeding , Infant Formula , Metabolome , Nutrition Assessment , Urine/chemistry , Humans , Infant , Infant, Newborn , Magnetic Resonance Spectroscopy , Time Factors
12.
Int J Food Sci Nutr ; 66(7): 811-4, 2015.
Article in English | MEDLINE | ID: mdl-26313813

ABSTRACT

Within an observational open study on the effects of a scheduled dosage of biscuits with iron, children with juvenile idiopathic arthritis were either supplemented with biscuits supplying iron fumarate (median 3.6 mg per day) or left to their customary dietary habits. After 4 months, supplemented children showed a more favourable percentage change of blood haemoglobin, while ferritin levels (markers of inflammation) remained stable. We conclude that the supply of iron with available dietary products may contribute to an adequate iron status in children with chronic inflammatory disorders in a stable situation.


Subject(s)
Arthritis, Juvenile/pathology , Bread , Food, Fortified , Inflammation/blood , Iron, Dietary/therapeutic use , Iron/therapeutic use , Nutritional Status , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/prevention & control , Arthritis, Juvenile/blood , Child , Child, Preschool , Dietary Supplements , Female , Ferritins/blood , Hemoglobins/metabolism , Humans , Inflammation/complications , Iron/blood , Iron/pharmacology , Iron, Dietary/blood , Iron, Dietary/pharmacology , Male
13.
Int J Food Sci Nutr ; 66(4): 409-15, 2015.
Article in English | MEDLINE | ID: mdl-26017322

ABSTRACT

Human milk is a highly valuable food for newborns and infants. Its protein fraction plays an important role for the development of the newborn. In the present study, an in vitro digestive model, developed for resembling closely the digestive system of an infant, was applied to human milk in order to identify and characterize the peptide profile. The peptide profile obtained after digestion was analyzed by µLC-LTQ-Orbitrap-MS. A total of 149 peptides from ß-casein, 30 peptides from α-lactalbumin, 26 peptides from αs1-casein, 24 peptides from κ-casein, 28 peptides from osteopontin, and 29 from lactoferrin was recovered. The identified peptide profile of partially hydrolyzed proteins, such as caseins, α-lactalbumin, and osteopontin, was different from that previously reported demonstrating a different performance of the developed neonatal digestive system with respect to other previously applied. These results would be useful as a starting point to investigate the physiological function of breast milk peptides.


Subject(s)
Milk Proteins/analysis , Milk Proteins/chemistry , Milk, Human/chemistry , Milk, Human/metabolism , Peptides/analysis , Peptides/chemistry , Adult , Caseins/analysis , Caseins/chemistry , Chromatography, Liquid , Digestion , Female , Humans , In Vitro Techniques , Lactalbumin/analysis , Lactalbumin/chemistry , Lactoferrin/analysis , Lactoferrin/chemistry , Mass Spectrometry , Middle Aged , Osteopontin/analysis , Osteopontin/chemistry
15.
Int J Food Sci Nutr ; 65(8): 953-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25030417

ABSTRACT

Several recent reports describe a role of probiotics as a therapeutic approach for celiac disease (CD). Two undigested A-gliadin peptides, P31-43 and P57-68, are central to CD pathogenesis, inducing an innate and an adaptive immune response, respectively. They enter enterocytes and localize to vesicular compartment to induce their toxic/immunogenics effects. In this article, we tested the effect of probiotic Lactobacillus paracasei (LP) CBA L74 (International Depository Accession Number LMG P-24778), its supernatant and LP-fermented cereals on gliadin peptides, P31-43 and P57-68, entrance in Caco-2 cells. Both LP CBA L74 and its supernatant inhibit P31-43 (intensity of fluorescence; FI: 75%) and P57-68 (FI: 50%) entrance in Caco2 cells, indicating that this biological effect is due to some product included in LP CBA L74 supernatant. This effect was present also after fermentation of cereals. This study describes a novel effect of probiotics in the prevention of undigested gliadin peptides toxic effects.


Subject(s)
Biological Products/pharmacology , Celiac Disease/metabolism , Gliadin/metabolism , Intestinal Mucosa/metabolism , Lactobacillus , Peptides/metabolism , Probiotics , Biological Products/therapeutic use , Caco-2 Cells , Celiac Disease/drug therapy , Cells, Cultured , Colon/drug effects , Colon/metabolism , Edible Grain/microbiology , Enterocytes/drug effects , Enterocytes/metabolism , Fermentation , Humans , Intestinal Mucosa/drug effects , Probiotics/therapeutic use
16.
PLoS One ; 9(2): e87615, 2014.
Article in English | MEDLINE | ID: mdl-24520333

ABSTRACT

The rapid expansion of commercially available fermented food products raises important safety issues particularly when infant food is concerned. In many cases, the activity of the microorganisms used for fermentation as well as what will be the immunological outcome of fermented food intake is not known. In this manuscript we used complex in vitro, ex-vivo and in vivo systems to study the immunomodulatory properties of probiotic-fermented products (culture supernatant and fermented milk without live bacteria to be used in infant formula). We found in vitro and ex-vivo that fermented products of Lactobacillus paracasei CBA L74 act via the inhibition of proinflammatory cytokine release leaving anti-inflammatory cytokines either unaffected or even increased in response to Salmonella typhimurium. These activities are not dependent on the inactivated bacteria but to metabolic products released during the fermentation process. We also show that our in vitro systems are predictive of an in vivo efficacy by the fermented products. Indeed CBA L74 fermented products (both culture medium and fermented milk) could protect against colitis and against an enteric pathogen infection (Salmonella typhimurium). Hence we found that fermented products can act via the inhibition of immune cell inflammation and can protect the host from pathobionts and enteric pathogens. These results open new perspectives in infant nutrition and suggest that L. paracasei CBA L74 fermented formula can provide immune benefits to formula-fed infants, without carrying live bacteria that may be potentially dangerous to an immature infant immune system.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/prevention & control , Dendritic Cells/metabolism , Fermentation/drug effects , Infant Formula/pharmacology , Lactobacillus/metabolism , Milk/metabolism , Salmonella typhimurium/drug effects , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Colitis/drug therapy , Colitis/microbiology , Dendritic Cells/drug effects , Humans , Infant , Infant Formula/administration & dosage , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Protective Agents/administration & dosage , Protective Agents/pharmacology , Protective Agents/therapeutic use , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/prevention & control , Salmonella typhimurium/physiology
17.
Eur J Nutr ; 53(5): 1177-86, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24240659

ABSTRACT

PURPOSE: Celiac disease (CD) is an autoimmune enteropathy, triggered by dietary gluten. The only treatment is a strict gluten-free diet. Oats are included in the list of gluten-free ingredients by European Regulation, but the safety of oats in CD is still a matter of debate. The present study examined the capability of different oat cultivars of activating the gliadin-induced transglutaminase-2 (TG2)-dependent events in some in vitro models of CD. In addition, we compared this capability with the electrophoresis pattern of peptic-tryptic digests of the proteins of the oat cultivars. METHODS: K562(S) cells agglutination, transepithelial electrical resistance of T84-cell monolayers, intracellular levels of TG2 and phosphorylated form of protein 42-44 in T84 cells were the early gliadin-dependent events studied. RESULTS: The results showed that the Nave oat cultivar elicited these events, whereas Irina and Potenza varieties did not. The ability of a cultivar to activate the above-described events was associated with the electrophoretic pattern of oat proteins and their reactivity to anti-gliadin antibodies. CONCLUSION: We found significant differences among oat cultivars in eliciting the TG2-mediated events of CD inflammation. Therefore, the safety of an oat cultivar in CD might be screened in vitro by means of biochemical and biological assays, before starting a clinical trial to definitely assess its safety.


Subject(s)
Avena/adverse effects , Avena/classification , Celiac Disease/immunology , Gliadin/chemistry , Avena/chemistry , Cell Line, Tumor , Child , Diet, Gluten-Free , Duodenum/pathology , Electrophoresis, Polyacrylamide Gel , Female , GTP-Binding Proteins/immunology , GTP-Binding Proteins/metabolism , Humans , K562 Cells , Male , Phosphorylation , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/immunology , Transglutaminases/metabolism
18.
Nutrients ; 5(11): 4653-64, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24264227

ABSTRACT

A gluten-free diet (GFD) is currently the only available treatment for patients with celiac disease (CD). Several clinical trials have demonstrated that most celiac patients can tolerate a medium-high quantity of oats without any negative clinical effects; however, the inclusion of oats in GFD is still a matter of debate. In this study, Italian children with CD were enrolled in a 15-month, randomized, double-blind, placebo-controlled multicenter trial. Participants were randomized in two groups following either A-B treatment (6 months of diet "A", 3 months of standard GFD, 6 months of diet "B"), or B-A treatment (6 months of diet "B", 3 months of standard GFD, 6 months of diet "A"). A and B diets included gluten-free (GF) products (flour, pasta, biscuits, cakes and crisp toasts) with either purified oats or placebo. Clinical data (Gastrointestinal Symptoms Rate Scale [GSRS] score) and intestinal permeability tests (IPT), were measured through the study period. Although the study is still blinded, no significant differences were found in GSRS score or the urinary lactulose/mannitol (L/M) ratio between the two groups after 6 months of treatment. These preliminary results suggest that the addition of non-contaminated oats from selected varieties in the treatment of children with CD does not determine changes in intestinal permeability and gastrointestinal symptoms.


Subject(s)
Avena , Celiac Disease , Diet, Gluten-Free , Intestinal Mucosa/pathology , Adolescent , Celiac Disease/complications , Celiac Disease/diet therapy , Celiac Disease/urine , Child , Child, Preschool , Double-Blind Method , Female , Gastrointestinal Diseases/etiology , Humans , Italy , Lactulose/urine , Male , Mannitol/urine , Permeability , Seeds
SELECTION OF CITATIONS
SEARCH DETAIL
...