Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Clin Nutr ; 38(3): 1373-1381, 2019 06.
Article in English | MEDLINE | ID: mdl-29960810

ABSTRACT

BACKGROUND & AIMS: Celiac disease (CD) is an immune-mediated systemic disease, caused by ingestion of gluten in genetically predisposed individuals. Gut microbiota dysbiosis might play a significant role in pathogenesis of chronic enteropathies and its modulation can be used as an intervention strategy in CD as well. In this study, we aimed to identify correlations between fecal microbiota, serum tumor necrosis factor alpha (TNF-α) and fecal short-chain fatty acids (SCFAs) in healthy children and children with CD after administration of probiotic Bifidobacterium breve BR03 and B632. METHODS: A double-blind placebo-controlled study enrolled 40 children with CD (CD) and 16 healthy children (HC). CD children were randomly allocated into two groups, of which 20 belonged to the placebo (PL) group and 20 to the Probiotic (PR) group. The PR group received a probiotic formulation containing a mixture of 2 strains, B. breve BR03 (DSM 16604) and B. breve B632 (DSM 24706) in 1:1 ratio for 3 months. Subsequently, for statistical analysis, blood and fecal samples from CD children (on enrolment - T0 and after 3 months, at the end of intervention with probiotic/placebo - T1) and HC children were used. The HC group was sampled only once (T0). RESULTS: Verrucomicrobia, Parcubacteria and some yet unknown phyla of Bacteria and Archaea may be involved in the disease, indicated by a strong correlation to TNF-α. Likewise, Proteobacteria strongly correlated with fecal SCFAs concentration. The effect of probiotic administration has disclosed a negative correlation between Verrucomicrobia, some unknown phyla of Bacteria, Synergistetes, Euryarchaeota and some SCFAs, turning them into an important target in microbiome restoration process. Synergistetes and Euryarchaeota may have a role in the anti-inflammatory process in healthy human gut. CONCLUSIONS: Our results highlight new phyla, which may have an important relation to disease-related parameters, CD itself and health.


Subject(s)
Bifidobacterium breve , Celiac Disease/drug therapy , Celiac Disease/metabolism , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome , Probiotics/therapeutic use , Tumor Necrosis Factor-alpha/blood , Celiac Disease/microbiology , Child , Double-Blind Method , Feces/microbiology , Female , Humans , Male
2.
Front Nutr ; 5: 39, 2018.
Article in English | MEDLINE | ID: mdl-29888226

ABSTRACT

Infantile functional gastrointestinal disorders are common in the first months of life. Their pathogenesis remains unknown although evidences suggest multiple independent causes, including gut microbiota modifications. Feeding type, influencing the composition of intestinal microbiota, could play a significant role in the pathogenesis. Previous studies supported probiotic supplementation success against colics, however mainly Lactobacillus spp. were tested. The aim of this study was to evaluate the effectiveness against functional gastrointestinal disorders of a Bifidobacterium breve based probiotic formulation including in the study both breast-fed and bottle-fed subjects. Two hundred and sixty-eight newborns were enrolled within 15 days from birth. One hundred and fifty-five of them effectively entered the study and were randomized in probiotic and placebo group, receiving the formulation for 90 days. The probiotic formulation consists of a 1:1 mixture of 2 strains of B. breve prepared in an oily suspension and administered in a daily dosage of 5 drops containing 108 CFU of each strain. Absolute quantification of selected microbial groups in the faeces was performed using qPCR. Anthropometric data, daily diary minutes of crying, number of regurgitations, vomits and evacuations, and colour and consistency of stools were evaluated before and after treatment. The study confirmed the positive role of breast milk in influencing the counts of target microbial groups, in particular the bifidobacteria community. No adverse events upon probiotic administration were reported, suggesting the safety of the product in this regimen. B. breve counts increased significantly in all administered newborns (p < 0.02). The study demonstrates that a 3 months treatment with B. breve strains in healthy breast-fed newborns helps to prevent functional gastrointestinal disorders, in particular reducing 56% of daily vomit frequency (p < 0.03), decreasing 46.5% of daily evacuation over time (p < 0.03), and improving the stool consistency (type 6 at the Bristol Stool chart instead of type 5) in those at term (p < 0.0001). Moreover, a significant reduction (8.65 vs. 7.98 LogCFU/g of feces, p < 0.03) of B. fragilis in the bottle-fed group receiving the probiotic formulation was observed.

3.
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: S68-S70, 2018.
Article in English | MEDLINE | ID: mdl-29782468

ABSTRACT

BACKGROUND: Recent preclinical studies suggest that dysfunction of gastrointestinal tract may play a role in amyotrophic lateral sclerosis (ALS) pathogenesis through a modification of the gut microbiota brain axis. Our study is the first focused on microbiota analysis in ALS patients. AIM: Our aim was to study the main human gut microbial groups and the overall microbial diversity in ALS and healthy subjects. Moreover we have examined the influence of a treatment with a specific bacteriotherapy composed of Lactobacillus strains (Lactobacillus fermentum, Lactobacillus delbrueckii, Lactobacillus plantarum, Lactobacillus salivarius) acting on the gastrointestinal barrier. METHODS: We enrolled 50 ALS patients and 50 healthy controls, matched for sex, age, and origin. Fecal samples were used for total genomic DNA extraction. Enterobacteria, Bifidobacterium spp., Lactobacillus spp., Clostridium sensu stricto, Escherichia coli and yeast were quantified using quantitative polymerase chain reaction approach. Denaturing gradient gel electrophoresis analyses were performed to investigate total eubacteria and yeasts populations. Patients were randomized to double-blind treatment either with microorganisms or placebo for 6 months and monitored for clinical progression and microbiota composition. RESULTS: The comparison between ALS subjects and healthy group revealed a variation in the intestinal microbial composition with a higher abundance of E. coli and enterobacteria and a low abundance of total yeast in patients. Polymerase chain reaction denaturing gradient gel electrophoresis analysis showed a cluster distinction between the bacterial profiles of ALS patients and the healthy subjects. The complexity of the profiles in both cases may indicate that a real dysbiosis status is not evident in the ALS patients although differences between healthy and patients exist. The effects of the progression of the disease and of the bacteriotherapy on the bacterial and yeast populations are currently in progress. CONCLUSIONS: Our preliminary results confirm that there is a difference in the microbiota profile in ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/microbiology , Gastrointestinal Microbiome , Probiotics/administration & dosage , Adult , Amyotrophic Lateral Sclerosis/therapy , Bifidobacterium/growth & development , Colony Count, Microbial , Double-Blind Method , Enterobacteriaceae/growth & development , Escherichia coli/growth & development , Feces/microbiology , Female , Gastrointestinal Tract/microbiology , Humans , Lactobacillus , Male , Phenotype , Yeasts/growth & development
4.
Dalton Trans ; 47(9): 3155-3165, 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29350728

ABSTRACT

Nanocomposites based on poly(butylene succinate) (PBS) and hydrotalcite-type anionic clays (HTs) organo-modified with biomolecules characterized by antibacterial and/or antioxidant activities, such as l-ascorbic acid (ASA), phloretic acid (HPP), l-tyrosine (TYR) and l-tryptophan (TRP), have been prepared by in situ polymerization. From XRD analysis and rheology experiments in a molten polymer state, intercalated HT hybrid platelets acting here as a hybrid filler are found to be well dispersed into polymers while providing a chain extension effect on PBS. Moreover, the molecules, when hosted within a HT interlayer gap, do preserve their pristine antibacterial activity, both in HT and in the resulting PBS composites. In particular, under the experimental conditions tested, HT/ASA and HT/TYR present the best combination of both properties (chain extension effect and antibacterial), especially versus E. coli as high as 90 and 97% of inhibition, respectively, using 2.5 wt% hybrid filler only. These findings open future applications for PBS associated with the hybrid HT filler as multifunctional materials in active packaging applications.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Butylene Glycols/chemistry , Hydroxides/chemistry , Polymerization , Polymers/chemistry , Aluminum Hydroxide/chemistry , Escherichia coli/drug effects , Magnesium Hydroxide/chemistry , Nanocomposites/chemistry , Staphylococcus aureus/drug effects
5.
Nutrients ; 8(10)2016 Oct 22.
Article in English | MEDLINE | ID: mdl-27782071

ABSTRACT

Coeliac disease (CD) is associated with alterations of the intestinal microbiota. Although several Bifidobacterium strains showed anti-inflammatory activity and prevention of toxic gliadin peptides generation in vitro, few data are available on their efficacy when administered to CD subjects. This study evaluated the effect of administration for three months of a food supplement based on two Bifidobacterium breve strains (B632 and BR03) to restore the gut microbial balance in coeliac children on a gluten free diet (GFD). Microbial DNA was extracted from faeces of 40 coeliac children before and after probiotic or placebo administration and 16 healthy children (Control group). Sequencing of the amplified V3-V4 hypervariable region of 16S rRNA gene as well as qPCR of Bidobacterium spp., Lactobacillus spp., Bacteroides fragilis group Clostridiumsensu stricto and enterobacteria were performed. The comparison between CD subjects and Control group revealed an alteration in the intestinal microbial composition of coeliacs mainly characterized by a reduction of the Firmicutes/Bacteroidetes ratio, of Actinobacteria and Euryarchaeota. Regarding the effects of the probiotic, an increase of Actinobacteria was found as well as a re-establishment of the physiological Firmicutes/Bacteroidetes ratio. Therefore, a three-month administration of B. breve strains helps in restoring the healthy percentage of main microbial components.


Subject(s)
Bifidobacterium breve , Celiac Disease/therapy , Dietary Supplements , Gastrointestinal Microbiome/physiology , Probiotics/administration & dosage , Actinobacteria , Adolescent , Bacteroidetes , Celiac Disease/microbiology , Child , Child, Preschool , Combined Modality Therapy , DNA, Bacterial/analysis , Diet, Gluten-Free , Double-Blind Method , Feces/microbiology , Female , Firmicutes , Humans , Infant , Male , Pilot Projects , RNA, Ribosomal, 16S/analysis , Young Adult
6.
Int J Food Microbiol ; 235: 53-9, 2016 Oct 17.
Article in English | MEDLINE | ID: mdl-27400453

ABSTRACT

In meat fermented foods, Clostridium spp. growth is kept under control by the addition of nitrite. The growing request of consumers for safer products has led to consider alternative bio-based approaches, the use of protective cultures being one of them. This work is aimed at checking the possibility of using two Lactobacillus spp. strains as protective cultures against Clostridium spp. in pork ground meat for fermented salami preparation. Both Lactobacillus strains displayed anti-clostridia activity in vitro using the spot agar test and after co-culturing them in liquid medium with each Clostridium strain. Only one of them, however, namely L. plantarum PCS20, was capable of effectively surviving in ground meat and of performing anti-microbial activity in carnis in a challenge test where meat was inoculated with the Clostridium strain. Therefore, this work pointed out that protective cultures can be a feasible approach for nitrite reduction in fermented meat products.


Subject(s)
Anti-Bacterial Agents/metabolism , Clostridium/growth & development , Food Microbiology , Food Preservation/methods , Lactobacillus/metabolism , Meat Products/microbiology , Red Meat/microbiology , Animals , Bioreactors , Clostridium/drug effects , Fermentation , Food Safety/methods , Nitrites/pharmacology , Swine
7.
Appl Microbiol Biotechnol ; 100(12): 5537-46, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26971496

ABSTRACT

Different factors are known to influence the early gut colonization in newborns, among them the perinatal use of antibiotics. On the other hand, the effect on the baby of the administration of antibiotics to the mother during labor, referred to as intrapartum antibiotic prophylaxis (IAP), has received less attention, although routinely used in group B Streptococcus positive women to prevent the infection in newborns. In this work, the fecal microbiota of neonates born to mothers receiving IAP and of control subjects were compared taking advantage for the first time of high-throughput DNA sequencing technology. Seven different 16S rDNA hypervariable regions (V2, V3, V4, V6 + V7, V8, and V9) were amplified and sequenced using the Ion Torrent Personal Genome Machine. The results obtained showed significant differences in the microbial composition of newborns born to mothers who had received IAP, with a lower abundance of Actinobacteria and Bacteroidetes as well as an overrepresentation of Proteobacteria. Considering that the seven hypervariable regions showed different discriminant ability in the taxonomic identification, further analyses were performed on the V4 region evidencing in IAP infants a reduced microbial richness and biodiversity, as well as a lower number of bacterial families with a predominance of Enterobacteriaceae members. In addition, this analysis pointed out a significant reduction in Bifidobacterium spp. strains. The reduced abundance of these beneficial microorganisms, together with the increased amount of potentially pathogenic bacteria, may suggest that IAP infants are more exposed to gastrointestinal or generally health disorders later in age.


Subject(s)
Antibiotic Prophylaxis , Gastrointestinal Microbiome/drug effects , Actinobacteria/genetics , Actinobacteria/physiology , Adult , Antibiotic Prophylaxis/adverse effects , Bacteria/genetics , Bifidobacterium/genetics , Bifidobacterium/physiology , Biodiversity , DNA, Ribosomal , Enterobacteriaceae/genetics , Enterobacteriaceae/physiology , Feces/microbiology , Female , Genetic Variation , High-Throughput Nucleotide Sequencing/methods , Humans , Infant , Infant, Newborn , Labor, Obstetric , Pregnancy , RNA, Ribosomal, 16S , Streptococcal Infections/drug therapy , Streptococcal Infections/prevention & control , Streptococcus agalactiae/genetics , Streptococcus agalactiae/physiology
8.
J Pediatr Gastroenterol Nutr ; 62(2): 304-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26237371

ABSTRACT

OBJECTIVES: The effect of intrapartum antibiotic prophylaxis (IAP) for group B Streptococcus (GBS) on bacterial colonization of the infant's gut has not been investigated extensively. We aimed to evaluate the effect of IAP on gut microbiota in healthy term infants, also exploring the influence of type of feeding. METHODS: Healthy term infants, whose mothers had been screened for GBS in late gestation, were divided into 2 groups: infants born to GBS-positive mothers who had received IAP versus controls. Neonatal fecal samples were collected at 7 and 30 days of life; DNA was extracted, and quantification of selected microbial groups (Lactobacillus spp, Bifidobacterium spp, and Bacteroides fragilis group) was performed by real-time PCR. RESULTS: A total of 84 infant-mother pairs were recruited. Bifidobacteria count was significantly lower in the IAP group at 7 days of life (median [interquartile range] 6.01 Log colony-forming unit per gram [5.51-6.98] vs 7.80 [6.61-8.26], P = 0.000). No differences in Bifidobacteria count at 30 days or in Lactobacilli and B fragilis counts at any time point were documented. Furthermore, at 7 days of life, infants who had not received IAP and were exclusively human milk-fed had higher counts of Bifidobacteria. Regardless of IAP treatment, infants fed exclusively human milk had higher Lactobacillus spp counts both at 7 and 30 days of life. CONCLUSIONS: IAP alters gut microflora by reducing the count of Bifidobacteria, which is further affected in infants receiving formula feeding. Whether these alterations could have long-term consequences on health and disease requires further investigation.


Subject(s)
Anti-Bacterial Agents/adverse effects , Bifidobacterium/drug effects , Gastrointestinal Microbiome/drug effects , Infectious Disease Transmission, Vertical/prevention & control , Lactobacillus/drug effects , Streptococcal Infections/prevention & control , Streptococcus agalactiae , Adult , Anti-Bacterial Agents/pharmacology , Antibiotic Prophylaxis , Bacteroides/drug effects , Bacteroides/growth & development , Bifidobacterium/growth & development , DNA, Bacterial/analysis , Feces/microbiology , Feeding Behavior , Female , Gastrointestinal Tract/microbiology , Humans , Infant , Infant Formula , Lactobacillus/growth & development , Male , Milk, Human/microbiology , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Real-Time Polymerase Chain Reaction , Retrospective Studies , Streptococcal Infections/microbiology , Streptococcal Infections/transmission , Streptococcus agalactiae/growth & development
9.
Appl Microbiol Biotechnol ; 98(13): 6051-60, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24687755

ABSTRACT

Several factors are known to influence the early colonization of the gut in newborns. Among them, the use of antibiotics on the mother during labor, referred to as intrapartum antibiotic prophylaxis (IAP), has scarcely been investigated, although this practice is routinely used in group B Streptococcus (GBS)-positive women. This work is therefore aimed at verifying whether IAP can influence the main microbial groups of the newborn gut microbiota at an early stage of microbial establishment. Fifty-two newborns were recruited: 26 born by mothers negative to GBS (control group) and 26 by mothers positive to GBS and subjected to IAP with ampicillin (IAP group). Selected microbial groups (Lactobacillus spp., Bidobacterium spp., Bacteroides fragilis, Clostridium difficile, and Escherichia coli) were quantified with real-time PCR on DNA extracted from newborn feces. Further analysis was performed within the Bidobacterium genus by using DGGE after amplification with genus-specific primers. Results obtained showed a significant decrease of the bifidobacteria counts after antibiotic treatment of the mother. Bifidobacteria were found to be affected by IAP not only quantitatively but also qualitatively. In fact, IAP determined a decrement in the frequency of Bidobacterium breve, Bidobacterium bifidum, and Bidobacterium dentium with respect to the control group. Moreover, this study has preliminarily evaluated that some bifidobacterial strains, previously selected for use in infants, have antibacterial properties against GBS and are therefore potential candidates for being applied as probiotics for the prevention of GBS infections.


Subject(s)
Antibiotic Prophylaxis/methods , Bifidobacterium/growth & development , Biota , Gastrointestinal Tract/microbiology , Infectious Disease Transmission, Vertical/prevention & control , Pregnancy Complications, Infectious/drug therapy , Streptococcal Infections/prevention & control , Antibiosis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Denaturing Gradient Gel Electrophoresis , Female , Humans , Infant, Newborn , Molecular Sequence Data , Polymerase Chain Reaction , Pregnancy , Sequence Analysis, DNA , Streptococcus agalactiae/growth & development
10.
Appl Microbiol Biotechnol ; 98(2): 563-77, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24287935

ABSTRACT

This review is aimed at describing the most recent advances in the gut microbiota composition of newborns and infants with a particular emphasis on bifidobacteria. The newborn gut microbiota is quite unstable, whereas after weaning, it becomes more stable and gets closer to the typical adult microbiota. The newborn and infant gut microbiota composition is impaired in several enteric and non-enteric pathologies. The core of this review is the description of the most recent documented applications of bifidobacteria to newborns and infants for their prevention and treatment. Acute diarrhea is the most studied disease for which bifidobacteria are applied with great success, Bifidobacterium longum and Bifidobacterium breve being the most applied species. Moreover, the most recent updates in the use of bifidobacteria for the prevention and treatment of pathologies typical of newborns, such as necrotizing enterocolitis, colics, and streptococcal infections, are presented. In addition, a number of not strictly enteric pathologies have in recent years evidenced a strict correlation with an aberrant gut microbiota in infants, in particular showing a reduced level of bifidobacteria. These diseases represent new potential opportunities for probiotic applications. Among them, allergic diseases, celiac disease, obesity, and neurologic diseases are described in this review. The preliminary use of bifidobacteria in in vitro systems and animal models is summarized as well as preliminary in vivo studies. Only after validation of the results via human clinical trials will the potentiality of bifidobacteria in the prevention and cure of these pathologies be definitely assessed.


Subject(s)
Bifidobacterium/physiology , Biota , Diarrhea/prevention & control , Diarrhea/therapy , Gastrointestinal Tract/microbiology , Probiotics/administration & dosage , Animals , Bifidobacterium/growth & development , Celiac Disease/therapy , Disease Models, Animal , Humans , Hypersensitivity/therapy , Infant , Infant, Newborn , Nervous System Diseases/therapy , Obesity/therapy
11.
Appl Microbiol Biotechnol ; 96(6): 1561-76, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22588500

ABSTRACT

Several studies support the use of probiotics for the treatment of minor gastrointestinal problems in infants. Positive effects on newborn colics have been evidenced after administration of Lactobacillus strains, whereas no studies have been reported regarding the use of bifidobacteria for this purpose. This work was therefore aimed at the characterization of Bifidobacterium strains capable of inhibiting the growth of pathogens typical of the infant gastrointestinal tract and of coliforms isolated from colic newborns. Among the 46 Bifidobacterium strains considered, 16 showed high antimicrobial activity against potential pathogens; these strains were further characterized from a taxonomic point of view, for the presence and transferability of antibiotic resistances, for citotoxic effects and adhesion to nontumorigenic gut epithelium cell lines. Moreover, their ability to stimulate gut health by increasing the metabolic activity and the immune response of epithelial cells was also studied. The examination of all these features allowed to identify three Bifidobacterium breve strains and a Bifidobacterium longum subsp. longum strain as potential probiotics for the treatments of enteric disorders in newborns such as infantile colics. A validation clinical trial involving the selected strains is being planned.


Subject(s)
Bifidobacterium/physiology , Colic/therapy , Gastrointestinal Diseases/therapy , Infant, Newborn, Diseases/therapy , Probiotics/administration & dosage , Bifidobacterium/genetics , Bifidobacterium/isolation & purification , Colic/microbiology , Feces/microbiology , Female , Gastrointestinal Diseases/microbiology , Gastrointestinal Tract/microbiology , Humans , Infant, Newborn , Infant, Newborn, Diseases/microbiology , Male
12.
J Sci Food Agric ; 92(10): 2133-40, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22298124

ABSTRACT

BACKGROUND: Wheat grains are a rich source of dietary fibres, particularly in the western human diet. Many of the health effects attributed to dietary fibres are believed to be related to their microbial fermentation in the gut. This study evaluated the ability of two potentially probiotic strains, Lactobacillus plantarum L12 and Bifidobacterium pseudocatenulatum B7003, to ferment soluble dietary fibres (SDFs) from modern and ancient durum-type wheat grains. RESULTS: Fibre microbial utilisation was highly variable and dependent on the strain. SDFs from the varieties Svevo and Solex supported the growth of L. plantarum L12 the best, whereas those from the varieties Anco Marzio, Solex and Kamut(®) Khorasan were good carbohydrate substrates for B. pseudocatenulatum B7003. The highest prebiotic activity scores (describing the extent to which prebiotics support selective growth of probiotics) for B7003 were obtained with SDFs from the varieties Solex (0.57), Kamut(®) Khorasan (0.56) and Iride (0.55), whereas for L12 the highest scores were achieved with the varieties Orobel (0.63), Kamut(®) Khorasan (0.56) and Solex (0.53). CONCLUSION: The present study has identified some SDFs from durum-type wheat grains as suitable prebiotic substrates for the selective proliferation of B. pseudocatenulatum B7003 and L. plantarum L12 in vitro. The results provide the basis for the potential utilisation of wheat-based prebiotics as a component of synbiotic formulations.


Subject(s)
Bifidobacterium/metabolism , Dietary Fiber/metabolism , Lactobacillus/metabolism , Prebiotics , Probiotics , Synbiotics , Triticum/chemistry , Fermentation , Humans , Seeds/chemistry , Species Specificity , Triticum/classification
13.
J Sep Sci ; 30(4): 604-11, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17444230

ABSTRACT

In this work, a new CE method with diode array detection (DAD) was developed for the monitoring and quantitation of flavonoids in different beans treated and untreated with UV-B radiation. Flavonoid concentration was monitored in UV-B-treated and untreated sprouts of three common beans (Zolfino ecotype, cv. Verdone, cv. Lingua di Fuoco) and one soybean (cv. Pacific). After acid hydrolysis of extracts, the CE-DAD method provides reproducible quantitative determinations of daidzein, glycitein, genistein, and kaempferol at ppm level in these natural matrices within a relatively short time (less than 16 min). Total flavonoid content determined by CE-DAD was 159 +/- 8, 26 +/- 2, 13 +/- 1, and 1.3 +/- 0.3 microg/g fresh weight for untreated sprouts of Pacific soybean, Verdone bean, Zolfino bean, and Lingua di Fuoco bean, respectively. UV-B treatment caused no significant quantitative effect on Pacific soybean sprouts, whereas it enhanced the total isoflavone content by 1.5, 1.8, and 3.2-fold in Verdone, Zolfino, and Lingua di Fuoco beans, respectively. The proposed method shows (i) the potentialities of bean sprouts as a natural source of bioactive compounds (antioxidants); (ii) the technological role of UV-B treatment for sprout isoflavone enrichment; and (iii) the good capabilities of CE-DAD to monitor this process.


Subject(s)
Electrophoresis, Capillary/methods , Fabaceae/metabolism , Fabaceae/radiation effects , Microarray Analysis/methods , Seedlings/metabolism , Seedlings/radiation effects , Ultraviolet Rays , Flavonoids/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...