Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
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 Bioeng Biotechnol ; 9: 660691, 2021.
Article in English | MEDLINE | ID: mdl-34124020

ABSTRACT

The intestinal microbiota is a real ecosystem composed of several bacterial species and a very huge amount of strains that through their metabolic activities play a crucial role in the development and performance of the immune system and other functions. Microbiota modulation by probiotics establishes a new era into the pharmaceutical and healthcare market. Probiotics play, in fact, an important role in helping and sustaining human health, but in order to produce benefits, their viability must be preserved throughout the production process up to consumption, and in addition, their bioactivity required to be safeguarded while passing through the gastrointestinal tract. In this frame, encouraging results come from encapsulation strategies that have proven to be very promising in protecting bacteria and their viability. However, specific effort has to be dedicated to the design optimization of the encapsulation process and, in particular, to the processing parameters that affect capsules microstructure. Herein, focusing on calcium alginate microspheres, after a preliminary selection of their processing conditions based on size distribution, we implemented a micro-rheological analysis, by using the multiple-particle tracking technique, to correlate the inner microstructure to the selected process conditions and to the viability of the Lactobacillus paracasei CBA L74. It was assessed that the explored levels of cross-linking, although changing the microorganism constriction, did not affect its viability. The obtained results confirm how this technology is a promising and a valid strategy to protect the microorganism viability and ensure its stability during the production process.

3.
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.

4.
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
5.
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
6.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...