Littérature scientifique sur le sujet « Α-Galactooligosaccharides (α-GOS) »

Créez une référence correcte selon les styles APA, MLA, Chicago, Harvard et plusieurs autres

Choisissez une source :

Consultez les listes thématiques d’articles de revues, de livres, de thèses, de rapports de conférences et d’autres sources académiques sur le sujet « Α-Galactooligosaccharides (α-GOS) ».

À côté de chaque source dans la liste de références il y a un bouton « Ajouter à la bibliographie ». Cliquez sur ce bouton, et nous générerons automatiquement la référence bibliographique pour la source choisie selon votre style de citation préféré : APA, MLA, Harvard, Vancouver, Chicago, etc.

Vous pouvez aussi télécharger le texte intégral de la publication scolaire au format pdf et consulter son résumé en ligne lorsque ces informations sont inclues dans les métadonnées.

Articles de revues sur le sujet "Α-Galactooligosaccharides (α-GOS)"

1

Marín-Manzano, María, Oswaldo Hernandez-Hernandez, Marina Diez-Municio, Cristina Delgado-Andrade, Francisco Moreno et Alfonso Clemente. « Prebiotic Properties of Non-Fructosylated α-Galactooligosaccharides from PEA (Pisum sativum L.) Using Infant Fecal Slurries ». Foods 9, no 7 (13 juillet 2020) : 921. http://dx.doi.org/10.3390/foods9070921.

Texte intégral
Résumé :
The interest for naturally-occurring oligosaccharides from plant origin having prebiotic properties is growing, with special focus being paid to supplemented products for infants. Currently, non-fructosylated α-galactooligosaccharides (α-GOS) from peas have peaked interest as a result of their prebiotic activity in adults and their mitigated side-effects on gas production from colonic bacterial fermentation. In this study, commercially available non-fructosylated α-GOS from peas and β-galactooligosaccharides (β-GOS) derived from lactose were fermented using fecal slurries from children aged 11 to 24 months old during 6 and 24 h. The modulatory effect of both GOS on different bacterial groups and bifidobacteria species was assessed; non-fructosylated α-GOS consumption was monitored throughout the fermentation process and the amounts of lactic acid and short-chain fatty acids (SCFA) generated were analyzed. Non-fructosylated α-GOS, composed mainly of manninotriose and verbascotetraose and small amounts of melibiose, were fully metabolized and presented remarkable bifidogenic activity, similar to that obtained with β-GOS. Furthermore, non-fructosylated α-GOS selectively caused an increase on the population of Bifidobacterium longum subsp. longum and Bifidobacterium catenulatum/pseudo-catenulatum. In conclusion, non-fructosylated α-GOS could be used as potential ingredient in infant formula supplemented with prebiotic oligosaccharides.
Styles APA, Harvard, Vancouver, ISO, etc.
2

Sun, Congcong, Bifang Hao, Daorui Pang, Qian Li, Erna Li, Qiong Yang, Yuxiao Zou, Sentai Liao et Fan Liu. « Diverse Galactooligosaccharides Differentially Reduce LPS-Induced Inflammation in Macrophages ». Foods 11, no 24 (8 décembre 2022) : 3973. http://dx.doi.org/10.3390/foods11243973.

Texte intégral
Résumé :
The effects of natural and synthetic galactooligosaccharides (GOS) on inflammation were explored by investigating the structure-activity relationship between the degree of GOS polymerization and in vitro anti-inflammatory activity, together with the potential underlying mechanism of their anti-inflammatory effects. The results demonstrated that GOS had strong anti-inflammatory effects in lipopolysaccharide (LPS)-induced RAW264.7 macrophages, including the inhibition of nitric oxide production and the reduced expression of pro-inflammatory mediators (interleukin-1β, interleukin-6, and tumor necrosis factor α), induced nitric oxide synthase (iNOS), cyclooxygenase 2 (COX-2), and proteins related to the Toll-like receptor 4 (TLR4)/nuclear factor (NF)-κB signaling pathway. GOS4, which has the highest degree of polymerization, exerted the strongest anti-inflammatory activity among the GOS examined. More importantly, our findings confirmed the anti-inflammatory effects of GOS on RAW264.7 macrophages via the TLR4/NF-κB pathway. Our experimental results could provide further support for the exploration of GOS in human nutrition and health.
Styles APA, Harvard, Vancouver, ISO, etc.
3

Solarte, Daniela Alejandra, Ana Isabel Ruiz-Matute, Diana M. Chito-Trujillo, Maite Rada-Mendoza et María Luz Sanz. « Microwave Assisted Extraction of Bioactive Carbohydrates from Different Morphological Parts of Alfalfa (Medicago sativa L.) ». Foods 10, no 2 (6 février 2021) : 346. http://dx.doi.org/10.3390/foods10020346.

Texte intégral
Résumé :
Despite the nutritional properties of alfalfa, its production is mainly for animal feed and it is undervalued as a food source. In this study, the valorization of alfalfa as a potential source of bioactive carbohydrates [inositols, α-galactooligosaccharides (α-GOS)] is presented. A Box–Behnken experimental design was used to optimize the extraction of these carbohydrates from leaves, stems, and seeds of alfalfa by solid–liquid extraction (SLE) and microwave-assisted extraction (MAE). Optimal extraction temperatures were similar for both treatments (40 °C leaves, 80 °C seeds); however, SLE required longer times (32.5 and 60 min vs. 5 min). In general, under similar extraction conditions, MAE provided higher yields of inositols (up to twice) and α-GOS (up to 7 times); hence, MAE was selected for their extraction from 13 alfalfa samples. Pinitol was the most abundant inositol of leaves and stems (24.2–31.0 mg·g−1 and 15.5–22.5 mg·g−1, respectively) while seed extracts were rich in α-GOS, mainly in stachyose (48.8–84.7 mg·g−1). In addition, inositols and α-GOS concentrations of lyophilized MAE extracts were stable for up to 26 days at 50 °C. These findings demonstrate that alfalfa is a valuable source of bioactive carbohydrates and MAE a promising alternative technique to obtain functional extracts.
Styles APA, Harvard, Vancouver, ISO, etc.
4

YAMASHITA, Akiko, Hiroyuki HASHIMOTO, Koki FUJITA, Masamichi OKADA, Shigeharu MORI et Sumio KITAHATA. « Reverse Reaction ofAspergillus nigerAPC-9319 α-Galactosidase in a Supersaturated Substrate Solution : Production of α-Linked Galactooligosaccharide (α-GOS) ». Bioscience, Biotechnology, and Biochemistry 69, no 7 (janvier 2005) : 1381–88. http://dx.doi.org/10.1271/bbb.69.1381.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
5

Williams, Neil C., Michael A. Johnson, Dominick E. Shaw, Ian Spendlove, Jelena Vulevic, Graham R. Sharpe et Kirsty A. Hunter. « A prebiotic galactooligosaccharide mixture reduces severity of hyperpnoea-induced bronchoconstriction and markers of airway inflammation ». British Journal of Nutrition 116, no 5 (3 août 2016) : 798–804. http://dx.doi.org/10.1017/s0007114516002762.

Texte intégral
Résumé :
AbstractGut microbes have a substantial influence on systemic immune function and allergic sensitisation. Manipulation of the gut microbiome through prebiotics may provide a potential strategy to influence the immunopathology of asthma. This study investigated the effects of prebiotic Bimuno-galactooligosaccharide (B-GOS) supplementation on hyperpnoea-induced bronchoconstriction (HIB), a surrogate for exercise-induced bronchoconstriction, and airway inflammation. A total of ten adults with asthma and HIB and eight controls without asthma were randomised to receive 5·5 g/d of either B-GOS or placebo for 3 weeks separated by a 2-week washout period. The peak fall in forced expiratory volume in 1 s (FEV1) following eucapnic voluntary hyperpnoea (EVH) defined HIB severity. Markers of airway inflammation were measured at baseline and after EVH. Pulmonary function remained unchanged in the control group. In the HIB group, the peak post-EVH fall in FEV1 at day 0 (−880 (sd 480) ml) was unchanged after placebo, but was attenuated by 40 % (−940 (sd 460) v. −570 (sd 310) ml, P=0·004) after B-GOS. In the HIB group, B-GOS reduced baseline chemokine CC ligand 17 (399 (sd 140) v. 323 (sd 144) pg/ml, P=0·005) and TNF-α (2·68 (sd 0·98) v. 2·18 (sd 0·59) pg/ml, P=0·040) and abolished the EVH-induced 29 % increase in TNF-α. Baseline C-reactive protein was reduced following B-GOS in HIB (2·46 (sd 1·14) v. 1·44 (sd 0·41) mg/l, P=0·015) and control (2·16 (sd 1·02) v. 1·47 (sd 0·33) mg/l, P=0·050) groups. Chemokine CC ligand 11 and fraction of exhaled nitric oxide remained unchanged. B-GOS supplementation attenuated airway hyper-responsiveness with concomitant reductions in markers of airway inflammation associated with HIB.
Styles APA, Harvard, Vancouver, ISO, etc.
6

Massot-Cladera, Malén, María del Mar Rigo-Adrover, Laura Herrero, Àngels Franch, Margarida Castell, Jelena Vulevic, Francisco J. Pérez-Cano et María J. Rodríguez Lagunas. « A Galactooligosaccharide Product Decreases the Rotavirus Infection in Suckling Rats ». Cells 11, no 10 (18 mai 2022) : 1669. http://dx.doi.org/10.3390/cells11101669.

Texte intégral
Résumé :
The leading cause of gastroenteritis among young children worldwide is the Group A rotaviruses (RV), which produce a wide range of symptoms, from a limited diarrhea to severe dehydration and even death. After an RV infection, immunity is not complete and less severe re-infections usually occur. These infections could be ameliorated by nutritional interventions with bioactive compounds, such as prebiotics. The aim of this research was to study the impact of a particular galactooligosaccharide (B-GOS) on the RV symptomatology and immune response during two consecutive infections. Lewis neonatal rats were inoculated with SA11 (first RV infection) on day 6 of life and with EDIM (second RV infection) on day 17 of life. B-GOS group was administered by oral gavage with a daily dose of B-GOS between days three to nine of life. Clinical and immunological variables were assessed during both infective processes. In the first infection, after the prebiotic intervention with B-GOS, a lower incidence, duration, and overall severity of the diarrhea (p < 0.05) was observed. In addition, it improved another severity indicator, the fecal weight output, during the diarrhea period (p < 0.05). The second RV infection failed in provoking diarrhea in the groups studied. The immune response during first infection with SA11 was not affected by B-GOS administration and had no impact on second infection, but the prebiotic intervention significantly increased IFN-γ and TNF-α intestinal production after the second infection (p < 0.05). In summary, B-GOS supplementation is able to reduce the incidence and severity of the RV-associated diarrhea and to influence the immune response against RV infections.
Styles APA, Harvard, Vancouver, ISO, etc.
7

Lee, Dong Hyeon, Hyunbin Seong, Daniel Chang, Vinod K. Gupta, Jiseung Kim, Seongwon Cheon, Geonhee Kim, Jaeyun Sung et Nam Soo Han. « Evaluating the prebiotic effect of oligosaccharides on gut microbiome wellness using in vitro fecal fermentation ». npj Science of Food 7, no 1 (9 mai 2023). http://dx.doi.org/10.1038/s41538-023-00195-1.

Texte intégral
Résumé :
AbstractWe previously proposed the Gut Microbiome Wellness Index (GMWI), a predictor of disease presence based on a gut microbiome taxonomic profile. As an application of this index for food science research, we applied GMWI as a quantitative tool for measuring the prebiotic effect of oligosaccharides. Mainly, in an in vitro anaerobic batch fermentation system, fructooligosaccharides (FOS), galactooligosaccharides (GOS), xylooligosaccharides (XOS), inulin (IN), and 2’-fucosyllactose (2FL), were mixed separately with fecal samples obtained from healthy adult volunteers. To find out how 24 h prebiotic fermentation influenced the GMWI values in their respective microbial communities, changes in species-level relative abundances were analyzed in the five prebiotics groups, as well as in two control groups (no substrate addition at 0 h and for 24 h). The GMWI of fecal microbiomes treated with any of the five prebiotics (IN (0.48 ± 0.06) > FOS (0.47 ± 0.03) > XOS (0.33 ± 0.02) > GOS (0.26 ± 0.02) > 2FL (0.16 ± 0.06)) were positive, which indicates an increase of relative abundances of microbial species previously found to be associated with a healthy, disease-free state. In contrast, the GMWI of samples without substrate addition for 24 h (–0.60 ± 0.05) reflected a non-healthy, disease-harboring microbiome state. Compared to the original prebiotic index (PI) and α-diversity metrics, GMWI provides a more data-driven, evidence-based indexing system for evaluating the prebiotic effect of food components. This study demonstrates how GMWI can be applied as a novel PI in dietary intervention studies, with wider implications for designing personalized diets based on their impact on gut microbiome wellness.
Styles APA, Harvard, Vancouver, ISO, etc.
8

Pham, Hai-Ha-Thi, Do-Hyung Kim et Thanh Luan Nguyen. « Wide-genome selection of lactic acid bacteria harboring genes that promote the elimination of antinutritional factors ». Frontiers in Plant Science 14 (26 avril 2023). http://dx.doi.org/10.3389/fpls.2023.1145041.

Texte intégral
Résumé :
Anti-nutritional factors (ANFs) substances in plant products, such as indigestible non-starchy polysaccharides (α-galactooligosaccharides, α-GOS), phytate, tannins, and alkaloids can impede the absorption of many critical nutrients and cause major physiological disorders. To enhance silage quality and its tolerance threshold for humans as well as other animals, ANFs must be reduced. This study aims to identify and compare the bacterial species/strains that are potential use for industrial fermentation and ANFs reduction. A pan-genome study of 351 bacterial genomes was performed, and binary data was processed to quantify the number of genes involved in the removal of ANFs. Among four pan-genomes analysis, all 37 tested Bacillus subtilis genomes had one phytate degradation gene, while 91 out of 150 Enterobacteriacae genomes harbor at least one genes (maximum three). Although, no gene encoding phytase detected in genomes of Lactobacillus and Pediococcus species, they have genes involving indirectly in metabolism of phytate-derivatives to produce Myo-inositol, an important compound in animal cells physiology. In contrast, genes related to production of lectin, tannase and saponin degrading enzyme did not include in genomes of B. subtilis and Pediococcus species. Our findings suggest a combination of bacterial species and/or unique strains in fermentation, for examples, two Lactobacillus strains (DSM 21115 and ATCC 14869) with B. subtilis SRCM103689, would maximize the efficiency in reducing the ANFs concentration. In conclusion, this study provides insights into bacterial genomes analysis for maximizing nutritional value in plant-based food. Further investigations of gene numbers and repertories correlated to metabolism of different ANFs will help clarifying the efficiency of time consuming and food qualities.
Styles APA, Harvard, Vancouver, ISO, etc.
9

Rattanaprasert, Monchaya, Jan-Peter van Pijkeren, Amanda E. Ramer-Tait, Maria Quintero, Car Reen Kok, Jens Walter et Robert W. Hutkins. « Genes Involved in Galactooligosaccharide Metabolism in Lactobacillus reuteri and Their Ecological Role in the Gastrointestinal Tract ». Applied and Environmental Microbiology 85, no 22 (13 septembre 2019). http://dx.doi.org/10.1128/aem.01788-19.

Texte intégral
Résumé :
ABSTRACT Strains of Lactobacillus reuteri are commonly used as probiotics due to their demonstrated therapeutic properties. Many strains of L. reuteri also utilize the prebiotic galactooligosaccharide (GOS), providing a basis for formulating synergistic synbiotics that could enhance growth or persistence of this organism in vivo. In this study, in-frame deletion mutants were constructed to characterize the molecular basis of GOS utilization in L. reuteri ATCC PTA-6475. Results suggested that GOS transport relies on a permease encoded by lacS, while a second unidentified protein may function as a galactoside transporter. Two β-galactosidases, encoded by lacA and lacLM, sequentially degrade GOS oligosaccharides and GOS disaccharides, respectively. Inactivation of lacL and lacM resulted in impaired growth in the presence of GOS and lactose. In vitro competition experiments between the wild-type and ΔlacS ΔlacM strains revealed that the GOS-utilizing genes conferred a selective advantage in media with GOS but not glucose. GOS also provided an advantage to the wild-type strain in experiments in gnotobiotic mice but only on a purified, no sucrose diet. Differences in cell numbers between GOS-fed mice and mice that did not receive GOS were small, suggesting that carbohydrates other than GOS were sufficient to support growth. On a complex diet, the ΔlacS ΔlacM strain was outcompeted by the wild-type strain in gnotobiotic mice, suggesting that lacL and lacM are involved in the utilization of alternative dietary carbohydrates. Indeed, the growth of the mutants was impaired in raffinose and stachyose, which are common in plants, demonstrating that α-galactosides may constitute alternate substrates of the GOS pathway. IMPORTANCE This study shows that lac genes in Lactobacillus reuteri encode hydrolases and transporters that are necessary for the metabolism of GOS, as well as α-galactoside substrates. Coculture experiments with the wild-type strain and a gos mutant clearly demonstrated that GOS utilization confers a growth advantage in medium containing GOS as the sole carbohydrate source. However, the wild-type strain also outcompeted the mutant in germfree mice, suggesting that GOS genes in L. reuteri also provide a basis for utilization of other carbohydrates, including α-galactosides, ordinarily present in the diets of humans and other animals. Collectively, our work provides information on the metabolism of L. reuteri in its natural niche in the gut and may provide a basis for the development of synbiotic strategies.
Styles APA, Harvard, Vancouver, ISO, etc.

Thèses sur le sujet "Α-Galactooligosaccharides (α-GOS)"

1

Chartrel, Valentine. « Fonctionnalisation d’une matrice végétale à base de pois protéagineux (Pisum sativum) par voie microbienne ». Electronic Thesis or Diss., université Paris-Saclay, 2020. http://www.theses.fr/2020UPASB029.

Texte intégral
Résumé :
ROQUETTE transforme et valorise le pois (Pisum sativum) pour produire des protéines, des fibres et des amidons. Au cours du procédé, diffé-rentes fractions secondaires sont générées dont le soluble de pois, désigné LAB 4960 après atomisation. Ce coproduit riche en fibres est inapte pour la nutrition humaine car il peut causer des désordres diges-tifs, engendrés par la forte teneur en α-GOS, des α-galacto-oligosaccharides formés de 1 à 3 unités de galactose liées par des liaisons α-(1-6). Parmi les α-GOS, on retrouve le raffinose, le stachyose et le verbascose qui sont non digérés par l’homme mais fermentés par le microbiote intestinal. L’objectif de ce projet de thèse est donc de ré-duire la teneur en α-GOS du LAB 4960 par voie microbienne afin d’en améliorer sa digestibilité. Pour atteindre cet objectif, une stratégie a été mise en place en deux temps. Dans une première partie, une collec-tion microbienne très diverse en termes d’espèce et d’origine (végétale vs animale) a été constituée à partir de graines de pois avec la caracté-risation de la diversité microbienne du pois de différents terroirs et à partir des collections internes de l’INRAE et de la société ROQUETTE. Dans une deuxième partie, la collection a été testée pour son aptitude à hydrolyser les α-GOS du LAB 4960. Le criblage de souches a été réparti sur trois phases, impliquant différents critères de sélection. La phase 1 a permis de sélectionner les souches capables de croître sur le LAB 4960 gélosé dans deux conditions d’oxygénation (aérobie et anaérobie) et de pH (acide et neutre). La phase 2 a permis d’identifier les sucres par Chromatographie sur Couche Mince après 72 h de culture sur le LAB 4960 liquide. Les souches ayant réduit les α-GOS ont été retenues en phase 3 pour le dosage des sucres par Chromatographie liquide haute performance couplée à la spectrométrie de masse. Dans la première partie, l’étude par métagénétique de la diversité de la surface de pois de différents terroirs après trempage, a montré une forte dominance d’espèces bactériennes appartenant aux Proteobacteria (57%) et Firmicutes (28%) et d’espèces fongiques appartenant aux Ascomyco-ta (89%) et Basidiomycota (11%). La structure de la communauté épiphyte associée à la graine de pois a été fortement impactée par son origine (coopératives et pays). A partir du jus de trempage des graines de pois, 102 souches ont été isolées et assignées à 52 es-pèces. Les 52 souches du pois représentatives de chaque espèce identifiée ont été ajoutées aux 157 souches représentatives de 82 espèces microbiennes des collections internes. Dans la seconde partie, le criblage de la collection a montré que 89% des souches testées ont pu croître sur le LAB 4960 gélosé. A peu près 20% des souches ont dégradé uniquement le saccharose. L’apparition des sucres mélibiose, manninotriose et manninotétraose traduisant une défructosylation a suggéré que 19% de souches ont hydrolysé les α-GOS via une β-fructosyltransférase dont 4% provenaient du pois. Enfin, 4% des souches ont hydrolysé les α-GOS via une α-galactosidase dont 1% provenait de pois. Sur les 49 (23%) souches hydrolysant les α-GOS, deux souches se sont démarquées par leur forte activité hydrolytique : Candida pseudoglaebosa CBS 6715T et Serratia liquefaciens GBM09. Une étude sur milieu minimum, milieu LAB 4960 et en bioréacteur sur LAB 4960 de concentrations diffé-rentes a montré que, dans des conditions optimales de croissance, la bactérie GBM09 est capable d’hydrolyser les α-GOS par ordre de degré de polymérisation croissant à pH neutre et à 20°C alors que la levure CBS 6715T hydrolyse l’ensemble des α-GOS simultanément à pH acide et à 28°C. Ces essais préliminaires ont permis de valider une première preuve de concept d’un aliment fonctionnel fermenté et laissent espérer favorablement leur développement à l’échelle industrielle en ouvrant la voie à de nombreuses innovations
ROQUETTE transforms and valorizes peas (Pisum sativum) to produce proteins, fibers and starches. During this process, various secondary fractions are generated, including the pea soluble, designated LAB 4960 after atomization. This fiber-rich co-product is unfit for human consumption in its current form as it can cause digestive disorders, caused by the high content of α-GOS, α-galactooligosaccharides formed from 1 to 3 galactose units linked by α-(1-6) bonds. Among the α-GOS are raffinose, stachyose and verbascose which are not digested by humans, but fermented by the intestinal microbiota. The aim of this thesis project is therefore to reduce the α-GOS content of LAB 4960 by microbial fermentation in order to improve its digestibility. To achieve this objective, a twofold strategy has been implemented. In a first part, a microbial collection that is very diverse in terms of species and ori-gins (plant vs. animal) was built up from pea seeds with the characteri-zation of the microbial diversity of peas from different terroirs and from the private collections of the INRAE Laboratory and the ROQUETTE Company. In a second part, the constituted collection was tested for its ability to hydrolyze the α-GOS from LAB 4960. The screening of the strains was split into three steps, involving different selection criteria. Step 1 allowed the selection of strains capable of growing on LAB 4960 agar under two conditions of oxygenation (aerobic and anaerobic) and pH (acidic and neutral). Step 2 allowed the identification of sugars by Thin-layer chromatography after 72 hours of culture on the liquid LAB 4960. The strains that reduced the α-GOS were selected in step 3 for the quantification of sugars by High performance liquid chromatog-raphy coupled to mass spectrometry. In the first part, the metagenetic study of pea surface diversity according to different terroirs after soak-ing, showed a strong dominance of bacterial species belonging to Proteobacteria (57%) and Firmicutes (28%) and fungal species be-longing to Ascomycota (89%) and Basidiomycota (11%). The structure of the epiphytic community associated with the pea seed was strong-ly influenced by its origin (storage cooperatives and countries). From the pea seed soaking juice, 102 strains were isolated and assigned to 52 species. The 52 pea strains representative of each identified spe-cies were added to the 157 strains representative of 82 microbial species in the internal collections. Screening of the collection showed that 89% of the strains tested were capable of growing on LAB 4960 agar. About 20% of the strains degraded only sucrose. The occurrence of sugars as melibiose, manninotriose and manninotetraose, known to be the product of defructosylation, suggested that 19% of the strains hydrolyzed α-GOS by a β-fructosyltransferase of which 4% came from peas. Finally, 4% of the strains hydrolyzed α-GOS by an α-galactosidase, of which 1% came from peas. Among the 23% strains hydrolyzing α-GOS, two strains stood out for their strong hydrolytic activity: Candida pseudoglaebosa CBS 6715T and Serratia liquefa-ciens GBM09. A study on minimum medium, LAB 4960 medium and in a bioreactor on LAB 4960 of different concentrations showed that, under optimal growth conditions, the GBM09 bacterium is capable of hydrolyzing the α-GOS in increasing order of degree of polymeriza-tion at neutral pH and at 20°C whereas the yeast CBS 6715T hydro-lyzes all the α-GOS simultaneously at acid pH and at 28°C.These preliminary trials have made it possible to validate a proof of con-cept for a fermented functional food and hold out promise of their development on an industrial scale, paving the way for many innova-tions
Styles APA, Harvard, Vancouver, ISO, etc.
Nous offrons des réductions sur tous les plans premium pour les auteurs dont les œuvres sont incluses dans des sélections littéraires thématiques. Contactez-nous pour obtenir un code promo unique!

Vers la bibliographie