Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Antioxidants (Basel) ; 13(2)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38397759

ABSTRACT

Obesity is a risk factor for highly prevalent age-related neurodegenerative diseases, the pathogenesis of whichinvolves mitochondrial dysfunction and protein oxidative damage. Lipoxidation, driven by high levels of peroxidizable unsaturated fatty acids and low antioxidant protection of the brain, stands out as a significant risk factor. To gain information on the relationship between obesity and brain molecular damage, in a porcine model of obesity we evaluated (1) the level of mitochondrial respiratory chain complexes, as the main source of free radical generation, by Western blot; (2) the fatty acid profile by gas chromatography; and (3) the oxidative modification of proteins by mass spectrometry. The results demonstrate a selectively higher amount of the lipoxidation-derived biomarker malondialdehyde-lysine (MDAL) (34% increase) in the frontal cortex, and positive correlations between MDAL and LDL levels and body weight. No changes were observed in brain fatty acid profile by the high-fat diet, and the increased lipid peroxidative modification was associated with increased levels of mitochondrial complex I (NDUFS3 and NDUFA9 subunits) and complex II (flavoprotein). Interestingly, introducing n3 fatty acids and a probiotic in the high-fat diet prevented the observed changes, suggesting that dietary components can modulate protein oxidative modification at the cerebral level and opening new possibilities in neurodegenerative diseases' prevention.

2.
Eur J Nutr ; 62(6): 2463-2473, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37148357

ABSTRACT

PURPOSE: Obesity during childhood has become a pandemic disease, mainly caused by a diet rich in sugars and fatty acids. Among other negative effects, these diets can induce cognitive impairment and reduce neuroplasticity. It is well known that omega-3 and probiotics have a beneficial impact on health and cognition, and we have hypothesized that a diet enriched with Bifidobacterium breve and omega-3 could potentiate neuroplasticity in prepubertal pigs on a high-fat diet. METHODS: Young female piglets were fed during 10 weeks with: standard diet (T1), high-fat (HF) diet (T2), HF diet including B. breve CECT8242 (T3) and HF diet including the probiotic and omega-3 fatty acids (T4). Using hippocampal sections, we analyzed by immunocytochemistry the levels of doublecortin (DCX) to study neurogenesis, and activity-regulated cytoskeleton-associated protein (Arc) as a synaptic plasticity related protein. RESULTS: No effect of T2 or T3 was observed, whereas T4 increased both DCX+ cells and Arc expression. Therefore, a diet enriched with supplements of B. breve and omega-3 increases neurogenesis and synaptic plasticity in prepubertal females on a HF diet from nine weeks of age to sexual maturity. Furthermore, the analysis of serum cholesterol and HDL indicate that neurogenesis was related to lipidic demand in piglets fed with control or HF diets, but the neurogenic effect induced by the T4 diet was exerted by mechanisms independent of this lipidic demand. CONCLUSION: Our results show that the T4 dietary treatment is effective in potentiating neural plasticity in the dorsal hippocampus of prepubertal females on a HF diet.


Subject(s)
Bifidobacterium breve , Fatty Acids, Omega-3 , Animals , Female , Swine , Fatty Acids, Omega-3/pharmacology , Hippocampus/metabolism , Diet, High-Fat/adverse effects , Neurogenesis
3.
J Nutr Biochem ; 111: 109184, 2023 01.
Article in English | MEDLINE | ID: mdl-36265688

ABSTRACT

The aim of this study was to assess the effects of a mixture of four dietary fibers on obese rats. Four groups of male Wistar rats were fed with either standard chow (STD) or cafeteria diet (CAF) and were orally supplemented with either fibre mixture (2 g kg-1 of body weight) (STD+F or CAF+F groups) or vehicle (STD+VH or CAF+VH groups). We studied a wide number of biometric, biochemical, transcriptomic, metagenomic and metabolomic variables and applied an integrative multivariate approach based on multiple factor analysis and Pearson's correlation analysis. A significant reduction in body weight, adiposity, HbA1c and HDL-cholesterol serum levels, and colon MPO activity was observed, whereas cecal weight and small intestine length:weight ratio were significantly increased in F-treated groups compared to control animals. CAF+F rats displayed a significant enhancement in energy expenditure, fat oxidation and fresh stool weight, and a significant reduction in adiponectin and LPS serum levels, compared to control group. Animals in STD+F group showed reduced serum LDL-cholesterol levels and a significant reduction in total cholesterol levels in the liver compared to STF+VH group. The intervention effect was reflected at the metabolomic (i.e., production of short-chain fatty acids, phenolic acids, and amino acids), metagenomic (i.e., modulation of Ruminococcus and Lactobacillus genus) and transcriptomic (i.e., expression of tight junctions and proteolysis) levels. Altogether, our integrative multi-omics approach highlights the potential of supplementation with a mixture of fibers to ameliorate the impairments triggered by obesity in terms of adiposity, metabolic profile, and intestinal health.


Subject(s)
Dietary Fiber , Obesity , Animals , Male , Rats , Adiposity , Cholesterol , Dietary Fiber/pharmacology , Dietary Fiber/therapeutic use , Metabolome , Obesity/diet therapy , Obesity/metabolism , Rats, Wistar
4.
Eur Stroke J ; 7(3): 259-266, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36082245

ABSTRACT

Introduction: Acute ischemic stroke therapy has improved in recent decades, decreasing the rates of disability and death among stroke patients. Unfortunately, all health care systems have geographical disparities in infrastructure for stroke patients. A centralized telestroke network might be a low-cost strategy to reduce differences in terms of geographical barriers, equitable access, and quality monitoring across different hospitals. Aims: We aimed to quantify changes in stroke patients' geographic access to specialized evaluation by neurologists and to intravenous acute stroke reperfusion treatments following the rapid implementation of a centralized telestroke network in the large region of Andalusia (8.5 million inhabitants). Methods: We conducted an observational study using spatial and analytical methods to examine how a centralized telestroke network influences the quality and accessibility of stroke care for a large region. Results: In the pre-implementation period, 5,005,477 (59.72% of the Andalusian population) had access to specialized stroke care in less than 30 min. After the 5-month process of implementing the telestroke network, 7,832,988 (93.5%) inhabitants had an access time of less than 30 min, bridging the gap in acute stroke care in rural hospitals. Conclusions: A centralized telestroke network may be an efficient tool to reduce the differences in stroke care access and quality monitoring across different hospitals, especially in large regions with low population density.

5.
Int J Mol Sci ; 23(18)2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36142723

ABSTRACT

Previous works have described the activity of Bifidobacterium longum subsp. infantis CECT 7210 (also commercially named B. infantis IM-1®) against rotavirus in mice and intestinal pathogens in piglets, as well as its diarrhea-reducing effect on healthy term infants. In the present work, we focused on the intestinal immunomodulatory effects of B. infantis IM-1® and for this purpose we used the epithelial cell line isolated from colorectal adenocarcinoma Caco-2 and a co-culture system of human dendritic cells (DCs) from peripheral blood together with Caco-2 cells. Single Caco-2 cultures and Caco-2: DC co-cultures were incubated with B. infantis IM-1® or its supernatant either in the presence or absence of Escherichia coli CECT 515. The B. infantis IM-1® supernatant exerted a protective effect against the cytotoxicity caused by Escherichia coli CECT 515 on single cultures of Caco-2 cells as viability reached the values of untreated cells. B. infantis IM-1® and its supernatant also decreased the secretion of pro-inflammatory cytokines by Caco-2 cells and the co-cultures incubated in the presence of E. coli CECT 515, with the response being more modest in the latter, which suggests that DCs modulate the activity of Caco-2 cells. Overall, the results obtained point to the immunomodulatory activity of this probiotic strain, which might underlie its previously reported beneficial effects.


Subject(s)
Escherichia coli Infections , Probiotics , Animals , Bifidobacterium/physiology , Bifidobacterium longum subspecies infantis/metabolism , Caco-2 Cells , Cytokines/metabolism , Escherichia coli/metabolism , Humans , Infant , Mice , Probiotics/pharmacology , Swine
6.
Nutr Hosp ; 39(Spec No3): 65-68, 2022 Sep 01.
Article in Spanish | MEDLINE | ID: mdl-36039996

ABSTRACT

Introduction: Diarrhea caused by severe acute gastroenteritis is one of the main causes of infant mortality in children under 5 years of age. Therefore, it is interesting to perform a preclinical and clinical validation of the efficacy of B. longum subsp. infantis IM-1® against various gastrointestinal pathogens. B. infantis IM-1® was evaluated against different gastrointestinal pathogens that cause diarrhea in infants, using in vitro models, animal models, and clinical studies. B. infantis IM-1® is able in an in vitro model of MA-104 and HT-29 cells to inhibit rotavirus replication (up to 36.05%) as well as to protect cells from infection due to rotavirus (up to 48.50 %). An 11-amino acid peptide (MHQPHQPLPPT) with a molecular mass of 1,282 KDa produced by this probiotic with antirotaviral capacity has been identified. In a murine model, the IM-1® strain has been shown to provide in vivo protection against rotavirus infection. In adhesion experiments with HT29, IM-1® was able to displace some pathogens from the enterocyte, especially Cronobacter sakazakii and Salmonella enterica, and prevent the adhesion of C. sakazakii and Shigella sonnei. In a clinical study with 190 babies under 3 months of age, IM-1® reduced episodes of diarrhea, being safe, well tolerated and associated with a lower prevalence of constipation. B. infantis IM-1® is a safe, well tolerated and effective probiotic in reducing episodes of diarrhea caused by the main gastrointestinal pathogens in infants.


Introducción: La diarrea causada por gastroenteritis agudas graves es una de las principales causas de mortalidad infantil en niños menores de 5 años. Por ello es interesante realizar una validación preclínica y clínica de la eficacia de B. longum subsp. infantis IM-1® frente a diversos patógenos gastrointestinales. El B. infantis IM-1® fue evaluado frente a diferentes patógenos gastrointestinales causantes de diarrea en bebés utilizando modelos in vitro, modelos animales y estudios clínicos. B. infantis IM-1® es capaz en un modelo in vitro de células MA-104 y HT-29 de inhibir la replicación de rotavirus (hasta un 36,05 %), así como de proteger las células de la infección por rotavirus (hasta un 48,50 %). Se ha identificado un péptido de 11 aminoácidos (MHQPHQPLPPT) con una masa molecular de 1282 KDa producido por este probiótico con capacidad antirotaviral. En un modelo murino, la cepa IM-1® ha demostrado proporcionar protección in vivo contra la infección por rotavirus. En experimentos de adhesión con HT29, IM-1® fue capaz de desplazar algunos patógenos del enterocito, especialmente C. sakazakii y Salmonella entérica, e impedir la adhesión de C. sakazakii y Shigella sonnei. En un estudio clínico con 190 bebés de menos de 3 meses de edad IM-1® redujo los episodios de diarrea. Fue seguro, se toleró bien y se asoció con una menor prevalencia de estreñimiento. B. infantis IM-1® es un probiótico seguro, que se tolera bien y es eficaz en la reducción de los episodios de diarrea causados por los principales patógenos gastrointestinales en bebés.


Subject(s)
Bifidobacterium longum subspecies infantis , Probiotics , Animals , Diarrhea/drug therapy , Feces/microbiology , Humans , Intestines , Mice , Probiotics/therapeutic use
7.
Clin Nutr ; 41(8): 1697-1711, 2022 08.
Article in English | MEDLINE | ID: mdl-35777109

ABSTRACT

BACKGROUND & AIMS: The critical window of concurrent developmental paths of the nervous system and gut microbiota in infancy provides an opportunity for nutritional interventions with potential health benefits later in life. METHODS: We compared the dynamics of gut microbiota maturation and explored its association with neurodevelopment at 12 months and 4 years of age in 170 full-term healthy infants fed a standard formula (SF) or a new formula (EF) based on standard formula supplemented with synbiotics, long chain polyunsaturated fatty acids (LC-PUFA) and bovine milk fat globule membranes (MFGM), including a breastfed reference group (BF). RESULTS: Using Dirichlet Multinomial Modelling, we characterized three microbial enterotypes (Mixed, anaerobic and aerobic profile; Bact, Bacteroides-dominant; Firm, Firmicutes-enriched) and identified a new enterotype dominated by an unidentified genus within Lachnospiraceae (U_Lach). Enterotypes were associated with age (Mixed with baseline, U_Lach with month 6, Bact and Firm with months 12 and 18). Trajectories or timely enterotype shifts in each infant were not random but strongly associated with type of feeding. Trajectories in SF shifted from initial Mixed to U_Lach, Bact or Firm at month. Microbiota maturation in EF split into a fast trajectory as in SF, and a slow trajectory with Mixed to U_Lach, Bact or Firm transitions at months 12 or 18, as in BF. EF infants with slow trajectories were more often in-home reared and born by vaginal delivery to mothers with pre-pregnancy lean BMI. At 12 months of age, language and expressive language scores were significantly higher in EF infants with fast trajectories than in BF. Neurodevelopmental outcomes were similar between EF infants with slow trajectories and BF at 12 months and 4 years of age. CONCLUSIONS: Feeding a synbiotics, LC-PUFA and MFGM supplemented formula in a specific infant environment promoted probiotic growth and retarded gut microbiota maturation with similar neurodevelopment outcomes to breastfed infants. CLINICAL TRIAL REGISTRY NUMBER: NCT02094547.


Subject(s)
Microbiota , Synbiotics , Breast Feeding , Fatty Acids , Fatty Acids, Unsaturated , Female , Glycolipids , Glycoproteins , Humans , Infant , Infant Formula , Lipid Droplets
8.
Nutr. hosp ; 39(Esp. 3): 65-68, 2022.
Article in Spanish | IBECS | ID: ibc-212543

ABSTRACT

La diarrea causada por gastroenteritis agudas graves es una de las principales causas de mortalidad infantil en niños menores de 5 años. Por ello es interesante realizar una validación preclínica y clínica de la eficacia de B. longum subsp. infantis IM-1® frente a diversos patógenos gastrointestinales. El B. infantis IM-1® fue evaluado frente a diferentes patógenos gastrointestinales causantes de diarrea en bebés utilizando modelos in vitro, modelos animales y estudios clínicos.B. infantis IM-1® es capaz en un modelo in vitro de células MA-104 y HT-29 de inhibir la replicación de rotavirus (hasta un 36,05 %), así como de proteger las células de la infección por rotavirus (hasta un 48,50 %). Se ha identificado un péptido de 11 aminoácidos (MHQPHQPLPPT) con una masa molecular de 1282 KDa producido por este probiótico con capacidad antirotaviral. En un modelo murino, la cepa IM-1® ha demostrado proporcionar protección in vivo contra la infección por rotavirus. En experimentos de adhesión con HT29, IM-1® fue capaz de desplazar algunos patógenos del enterocito, especialmente C. sakazakii y Salmonella entérica, e impedir la adhesión de C. sakazakii y Shigella sonnei. En un estudio clínico con 190 bebés de menos de 3 meses de edad IM-1® redujo los episodios de diarrea. Fue seguro, se toleró bien y se asoció con una menor prevalencia de estreñimiento.B. infantis IM-1® es un probiótico seguro, que se tolera bien y es eficaz en la reducción de los episodios de diarrea causados por los principales patógenos gastrointestinales en bebés. (AU)


Diarrhea caused by severe acute gastroenteritis is one of the main causes of infant mortality in children under 5 years of age. Therefore, it is interesting to perform a preclinical and clinical validation of the efficacy of B. longum subsp. infantis IM-1R against various gastrointestinal pathogens. B. infantis IM-1R was evaluated against different gastrointestinal pathogens that cause diarrhea in infants, using in vitro models, animal models, and clinical studies.B. infantis IM-1R is able in an in vitro model of MA-104 and HT-29 cells to inhibit rotavirus replication (up to 36.05%) as well as to protect cells from infection due to rotavirus (up to 48.50 %). An 11-amino acid peptide (MHQPHQPLPPT) with a molecular mass of 1,282 KDa produced by this probiotic with antirotaviral capacity has been identified. In a murine model, the IM-1R strain has been shown to provide in vivo protection against rotavirus infection. In adhesion experiments with HT29, IM-1R was able to displace some pathogens from the enterocyte, especially Cronobacter sakazakii and Salmonella enterica, and prevent the adhesion of C. sakazakii and Shigella sonnei. In a clinical study with 190 babies under 3 months of age, IM-1R reduced episodes of diarrhea, being safe, well tolerated and associated with a lower prevalence of constipation.B. infantis IM-1R is a safe, well tolerated and effective probiotic in reducing episodes of diarrhea caused by the main gastrointestinal pathogens in infants. (AU)


Subject(s)
Humans , Male , Female , Infant, Newborn , Infant , Child, Preschool , Bifidobacterium longum subspecies infantis , Noxae , Intestines , Gastroenteritis , Microbiota , Probiotics , Diarrhea
9.
Front Microbiol ; 12: 642549, 2021.
Article in English | MEDLINE | ID: mdl-33935999

ABSTRACT

We evaluated the potential of multi-strain probiotic (Bifidobacterium longum subsp. infantis CECT 7210 and Lactobacillus rhamnosus HN001) with or without galacto-oligosaccharides against enterotoxigenic Escherichia coli (ETEC) F4 infection in post-weaning pigs. Ninety-six piglets were distributed into 32 pens assigned to five treatments: one non-challenged (CTR+) and four challenged: control diet (CTR-), with probiotics (>3 × 1010 CFU/kg body weight each, PRO), prebiotic (5%, PRE), or their combination (SYN). After 1 week, animals were orally inoculated with ETEC F4. Feed intake, weight, and clinical signs were recorded. On days 4 and 8 post-inoculation (PI), one animal per pen was euthanized and samples from blood, digesta, and tissues collected. Microbiological counts, ETEC F4 real-time PCR (qPCR) quantification, fermentation products, serum biomarkers, ileal histomorphometry, and genotype for mucin 4 (MUC4) polymorphism were determined. Animals in the PRO group had similar enterobacteria and coliform numbers to the CTR+ group, and the ETEC F4 prevalence, the number of mitotic cells at day 4 PI, and villus height at day 8 PI were between that observed in the CTR+ and CTR- groups. The PRO group exhibited reduced pig major acute-phase protein (Pig-MAP) levels on day 4 PI. The PRE diet group presented similar reductions in ETEC F4 and Pig-MAP, but there was no effect on microbial groups. The SYN group showed reduced fecal enterobacteria and coliform counts after the adaptation week but, after the inoculation, the SYN group showed lower performance and more animals with high ETEC F4 counts at day 8 PI. SYN treatment modified the colonic fermentation differently depending on the MUC4 polymorphism. These results confirm the potential of the probiotic strains and the prebiotic to fight ETEC F4, but do not show any synergy when administered together, at least in this animal model.

10.
Nutrients ; 13(4)2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33924514

ABSTRACT

The present study aims to evaluate the effects of an infant formula supplemented with a mixture of prebiotic short and long chain inulin-type oligosaccharides on health outcomes, safety and tolerance, as well as on fecal microbiota composition during the first year of life. In a prospective, multicenter, randomized, double-blind study, n = 160 healthy term infants under 4 months of age were randomized to receive either an infant formula enriched with 0.8 g/dL of Orafti®Synergy1 or an unsupplemented control formula until the age of 12 months. Growth, fever (>38 °C) and infections were regularly followed up by a pediatrician. Digestive symptoms, stool consistency as well as crying and sleeping patterns were recorded during one week each study month. Fecal microbiota and immunological biomarkers were determined from a subgroup of infants after 2, 6 and 12 months of life. The intention to treat (ITT) population consisted of n = 149 infants. Both formulae were well tolerated. Mean duration of infections was significantly lower in the prebiotic fed infants (p < 0.05). The prebiotic group showed higher Bifidobacterium counts at month 6 (p = 0.006), and higher proportions of Bifidobacterium in relation to total bacteria at month 2 and 6 (p = 0.042 and p = 0.013, respectively). Stools of infants receiving the prebiotic formula were softer (p < 0.05). Orafti®Synergy1 tended to beneficially impact total daily amount of crying (p = 0.0594). Supplementation with inulin-type prebiotic oligosaccharides during the first year of life beneficially modulates the infant gut microbiota towards higher Bifidobacterium levels at the first 6 months of life, and is associated with reduced duration of infections.


Subject(s)
Bottle Feeding/adverse effects , Infant Formula/adverse effects , Infections/epidemiology , Inulin/adverse effects , Prebiotics/adverse effects , Bifidobacterium/isolation & purification , Biomarkers/analysis , Bottle Feeding/methods , Double-Blind Method , Feces/chemistry , Feces/microbiology , Female , Gastrointestinal Microbiome/immunology , Humans , Incidence , Infant , Infant Formula/chemistry , Infant, Newborn , Infections/immunology , Intention to Treat Analysis , Inulin/administration & dosage , Inulin/analogs & derivatives , Male , Prebiotics/administration & dosage , Prospective Studies , Time Factors , Treatment Outcome
11.
Nutrients ; 12(11)2020 Oct 24.
Article in English | MEDLINE | ID: mdl-33114404

ABSTRACT

Certain non-digestible oligosaccharides (NDO) are specifically fermented by bifidobacteria along the human gastrointestinal tract, selectively favoring their growth and the production of health-promoting metabolites. In the present study, the ability of the probiotic strain Bifidobacterium longum subsp. infantis CECT7210 (herein referred to as B. infantis IM-1®) to utilize a large range of oligosaccharides, or a mixture of oligosaccharides, was investigated. The strain was able to utilize all prebiotics screened. However, galactooligosaccharides (GOS), and GOS-containing mixtures, effectively increased its growth to a higher extent than the other prebiotics. The best synbiotic combination was used to examine the antimicrobial activity against Escherichia coli, Cronobacter sakazakii, Listeria monocytogenes and Clostridium difficile in co-culture experiments. C. difficile was inhibited by the synbiotic, but it failed to inhibit E. coli. Moreover, Cr. sakazakii growth decreased during co-culture with B. infantis IM-1®. Furthermore, adhesion experiments using the intestinal cell line HT29 showed that the strain IM-1® was able to displace some pathogens from the enterocyte layer, especially Cr. sakazakii and Salmonella enterica, and prevented the adhesion of Cr. sakazakii and Shigella sonnei. In conclusion, a new synbiotic (probiotic strain B. infantis IM-1® and GOS) appears to be a potential effective supplement for maintaining infant health. However, further studies are needed to go more deeply into the mechanisms that allow B.infantis IM-1® to compete with enteropathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bifidobacterium longum subspecies infantis , Intestines/microbiology , Oligosaccharides/pharmacology , Probiotics/pharmacology , Clostridioides difficile/drug effects , Coculture Techniques , Complex Mixtures , Cronobacter sakazakii/drug effects , Escherichia coli/drug effects , Female , Gastrointestinal Tract/microbiology , HT29 Cells , Humans , Infant , Infant, Newborn , Listeria monocytogenes/drug effects , Male , Prebiotics/microbiology , Salmonella enterica/drug effects , Synbiotics
12.
Front Microbiol ; 11: 2012, 2020.
Article in English | MEDLINE | ID: mdl-32973728

ABSTRACT

Salmonella is a common causative agent of enteric disease and is developing mechanisms of resistance to antimicrobials. Probiotics, such as bifidobacteria and lactobacilli, and prebiotic fibers are a potential alternative to counteract this pathogen as they have demonstrated effectiveness in preventing its adhesion, reducing intestinal damage, and enhancing the host immune system. Furthermore, the benefits are expected to be potentiated when these compounds are administered together. A trial was performed to evaluate the efficacy of two probiotic strains (Bifidobacterium longum subsp. infantis CECT 7210 (Laboratorios Ordesa S.L.) and Lactobacillus rhamnosus HN001, combined or not with a prebiotic containing oligofructose-enriched inulin, against Salmonella Typhimurium. Ninety-six piglets (28 days old) were distributed into 32 pens assigned to 5 treatments: one non-challenged (control diet, CTR+) and four challenged: control diet (CTR-) or supplemented with probiotics (>3 × 1010 cfu/kg each strain, PRO), prebiotic (5%, PRE), or their combination (SYN). After 1 week of adaptation, animals were orally challenged with Salmonella Typhimurium. Feed intake, weight, and clinical signs were recorded. On days 4 and 8 post-inoculation (PI), one animal per pen was euthanized, and samples from blood, digestive content, and ileal tissues were collected to determine Salmonella counts, fermentation products, ileal histomorphology, and serum TNF-α and Pig-MAP concentrations. The effect of the oral challenge was evidenced by animal performance, fecal consistency, and intestinal architecture. Regarding the experimental treatments, animals belonging to the PRO group experienced a faster clearance of the pathogen, with more pigs being negative to its excretion at the end of the study and recovering the impaired ileal villi/crypt ratio more rapidly. Animals receiving the PRE diet showed a lower intestinal colonization by Salmonella, with no countable levels (<3 cfu/g) in any of the analyzed samples, and an augmented immune response suggested by serum Pig-MAP concentrations. Treatments including the prebiotic (PRE and SYN) showed similar changes in the fermentation pattern, with an increase in the molar percentage of valeric acid concentration in the colon. The SYN group, however, did not show any of the outcomes registered for PRO and PRE in Salmonella colonization or in immunity markers, suggesting the lack of synbiotic action in this animal model. Further research is needed to better understand the complex mechanisms behind these effects.

13.
Pediatr Res ; 83(6): 1120-1128, 2018 06.
Article in English | MEDLINE | ID: mdl-29538368

ABSTRACT

BackgroundIntestinal microbiota of breast-fed infants is plenty of beneficial bifidobacteria. We aimed to determine whether an infant formula supplemented with probiotic Bifidobacterium longum subsp. infantis CECT7210 (B. infantis IM1) is effective at reducing diarrhea incidence in healthy term infants.MethodsDouble-blinded, randomized, multicenter, controlled clinical trial, where formula-fed infants (<3 months) received an infant formula supplemented (Probiotic) or not (Control) with 107 cfu/g of B. infantis IM1 over 12 weeks. Diarrheas, growth, digestive symptoms, stool bifidobacteria, and microbiota were assessed.ResultsIn all, 97 (Control) and 93 (Probiotic) infants were randomized, and 78 (Control) and 73 (Probiotic) completed the 12 week-follow-up. In the overall study period, a median of 0.29±1.07 and 0.05±0.28 diarrhea events/infant was observed in the Control and Probiotic groups, respectively (P=0.059). This trend to less diarrhea episodes in the Probiotic group reached statistical significance at 8 weeks (0.12±0.47 vs. 0.0±0.0 events/infant, P=0.047). Constipation incidence was higher (odds ratio (OR) 2.67 (1.09-6.50)) and stool frequency lower (2.0±1.0 vs. 2.6±1.3 stools/day, P=0.038) in the Control group after 4 weeks. No differences were found at other time points nor in other digestive symptoms, growth, or formula intake.ConclusionA B. infantis IM1-supplemented infant formula may reduce diarrhea episodes, being safe, well tolerated, and associated with lower constipation prevalence.


Subject(s)
Bifidobacterium longum , Diarrhea/prevention & control , Infant Formula , Probiotics/therapeutic use , Anthropometry , Constipation/prevention & control , Double-Blind Method , Feces/microbiology , Female , Flatulence , Humans , Immune System , Infant , Infant, Newborn , Male , Microbiota , Milk, Human/microbiology , Patient Safety
14.
Nutr Hosp ; 34(Suppl 4): 8-12, 2017 10 15.
Article in Spanish | MEDLINE | ID: mdl-29156925

ABSTRACT

There is an increasing body of evidence about the effect that early nutrition and lifestyle could have on the programming of later health and disease. Infant formula must cover all the nutritional needs to promote adequate infant growth and development. Currently, important research efforts have been made to supplement infant formulae with new bioactive ingredients with health benefits for the infant. The milk fat globule membrane is one of the new ingredients, which provides phospholipids and gangliosides, as well as bioactive proteins. In addition, ingredients such as probiotics and prebiotics modulate intestinal microbiota and contribute to improve gastrointestinal health. Other ingredients relevant in infant formulas are long chain polyunsaturated fatty acids, α-lactalbumin and nucleotides. Recent research studies have demonstrated that adding these ingredients to infant formulas improves cognitive development and decreases the number of infections and allergies in infants. However, more studies are needed to find if these effects are long lasting and can be seen in childhood and adulthood.


Existe cada vez mayor evidencia científica de que la nutrición temprana y el estilo de vida tienen un efecto de programación sobre la salud y el riesgo de enfermedad futura. Las fórmulas infantiles deben cubrir los requerimientos nutricionales y promover un crecimiento y desarrollo correcto de los lactantes. Actualmente, un ámbito de innovación importante consiste en el aporte de componentes bioactivos capaces de aportar beneficios funcionales al lactante. Uno de los nuevos ingredientes es la membrana del glóbulo graso lácteo, que aporta componentes como fosfolípidos y gangliósidos, así como proteínas bioactivas. Además, ingredientes como probióticos y prebióticos actúan como moduladores de la microbiota intestinal y contribuyen a mejorar la salud gastrointestinal. Otros componentes relevantes en fórmulas infantiles son los ácidos grasos poliinsaturados de cadena larga, la α-lactoalbúmina y los nucleótidos. Estudios recientes demuestran que la adición de estos ingredientes a las fórmulas infantiles mejora el desarrollo cognitivo y reduce la incidencia de infecciones y alergias en los lactantes. Nuevos estudios de seguimiento a largo plazo son necesarios para valorar si los efectos observados son duraderos y se mantienen en etapas posteriores de la vida.


Subject(s)
Infant Formula , Nutritive Value , Child Development , Functional Food , Humans , Infant , Infant, Newborn
15.
PLoS One ; 12(10): e0186781, 2017.
Article in English | MEDLINE | ID: mdl-29073182

ABSTRACT

BACKGROUND: The weaning pig is used as an experimental model to assess the impact of diet on intestinal health. Blood parameters (BP) are considered a useful tool in humans, but there is very scarce information of such indicators in the weaning pig. The objective of the present study is to evaluate the use of different BP as indicators in an experimental model of salmonellosis. METHODOLOGY: Seventy-two 28-day-old piglets were divided into four groups in a 2x2 factorial arrangement, with animals receiving or not a probiotic combination based on B. infantis IM1® and B. lactis BPL6 (109 colony forming units (cfu)/d) and orally challenged or not a week later with Salmonella Typhimurium (5x108 cfu). Blood samples of one animal per pen (N = 24) were taken four days post-inoculation for the evaluation of different BP using an I-stat® System and of plasmatic concentrations of zinc, iron and copper. PRINCIPAL FINDINGS: Results reported marginal deficiencies of zinc in piglets at weaning. Moreover, plasmatic zinc, copper and iron presented good correlations with weight gain (r 0.57, r -0.67, r 0.54 respectively; P < 0.01). Blood electrolytes (Na+, Cl- and K+) decreased (P < 0.01) only when the performance of the animals was seriously compromised and clinical symptoms were more apparent. Acid-base balance parameters such as HCO3-, TCO2 and BEecf significantly correlated with weight gain, but only in the challenged animals (r -0.54, r -0.55, and r -0.51, respectively; P < 0.05), suggesting metabolic acidosis depending on Salmonella infection. Glucose was affected by the challenge (P = 0.040), while Htc and Hgb increased with the challenge and decreased with the probiotic (P < 0.05). Furthermore, correlations of Glu, Htc and Hgb with weight gain were observed (P < 0.05). Overall, BP could be regarded as simple, useful indexes to assess performance and health of weaning piglets.


Subject(s)
Salmonella Infections/blood , Salmonella , Swine Diseases/blood , Acid-Base Equilibrium , Animals , Biomarkers/blood , Blood Glucose/metabolism , Disease Models, Animal , Metals/blood , Swine
16.
Nutr. hosp ; 34(supl.4): 8-12, 2017. tab
Article in Spanish | IBECS | ID: ibc-168820

ABSTRACT

Existe cada vez mayor evidencia científica de que la nutrición temprana y el estilo de vida tienen un efecto de programación sobre la salud y el riesgo de enfermedad futura. Las fórmulas infantiles deben cubrir los requerimientos nutricionales y promover un crecimiento y desarrollo correcto de los lactantes. Actualmente, un ámbito de innovación importante consiste en el aporte de componentes bioactivos capaces de aportar beneficios funcionales al lactante. Uno de los nuevos ingredientes es la membrana del glóbulo graso lácteo, que aporta componentes como fosfolípidos y gangliósidos, así como proteínas bioactivas. Además, ingredientes como probióticos y prebióticos actúan como moduladores de la microbiota intestinal y contribuyen a mejorar la salud gastrointestinal. Otros componentes relevantes en fórmulas infantiles son los ácidos grasos poliinsaturados de cadena larga, la α-lactoalbúmina y los nucleótidos. Estudios recientes demuestran que la adición de estos ingredientes a las fórmulas infantiles mejora el desarrollo cognitivo y reduce la incidencia de infecciones y alergias en los lactantes. Nuevos estudios de seguimiento a largo plazo son necesarios para valorar si los efectos observados son duraderos y se mantienen en etapas posteriores de la vida (AU)


There is an increasing body of evidence about the effect that early nutrition and lifestyle could have on the programming of later health and disease. Infant formula must cover all the nutritional needs to promote adequate infant growth and development. Currently, important research efforts have been made to supplement infant formulae with new bioactive ingredients with health benefits for the infant. The milk fat globule membrane is one of the new ingredients, which provides phospholipids and gangliosides, as well as bioactive proteins. In addition, ingredients such as probiotics and prebiotics modulate intestinal microbiota and contribute to improve gastrointestinal health. Other ingredients relevant in infant formulas are long chain polyunsaturated fatty acids, α-lactalbumin and nucleotides. Recent research studies have demonstrated that adding these ingredients to infant formulas improves cognitive development and decreases the number of infections and allergies in infants. However, more studies are needed to find if these effects are long lasting and can be seen in childhood and adulthood (AU)


Subject(s)
Humans , Infant , Infant Formula , Immune System/growth & development , Life Style , Gastrointestinal Microbiome/physiology , Infant Nutrition , Quality Improvement/trends , Probiotics/therapeutic use , Prebiotics
17.
Genome Announc ; 3(2)2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25838473

ABSTRACT

Bifidobacterium longum subsp. infantis CECT 7210 is a probiotic strain able to inhibit rotavirus in vitro and protect against viral infection in both cell cultures and mice. Here, we report its complete genome sequence, as deciphered by PacBio single-molecule real-time (SMRT) technology. An analysis of the sequence may provide insights into its functional activity.

18.
Int J Food Microbiol ; 120(1-2): 2-12, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17629975

ABSTRACT

The availability of complete bacterial genome sequences has significantly furthered our understanding of the genetics, physiology and biochemistry of the microorganisms in question, particularly those that have commercially important applications. Bifidobacteria are among such microorganisms, as they constitute mammalian commensals of biotechnological significance due to their perceived role in maintaining a balanced gastrointestinal (GIT) microflora. Bifidobacteria are therefore frequently used as health-promoting or probiotic components in functional food products. A fundamental understanding of the metabolic activities employed by these commensal bacteria, in particular their capability to utilize a wide range of complex oligosaccharides, can reveal ways to provide in vivo growth advantages relative to other competing gut bacteria or pathogens. Furthermore, an in depth analysis of adaptive responses to nutritional or environmental stresses may provide methodologies to retain viability and improve functionality during commercial preparation, storage and delivery of the probiotic organism.


Subject(s)
Adaptation, Physiological/genetics , Bifidobacterium , Food Microbiology , Genome, Bacterial , Phylogeny , Bifidobacterium/classification , Bifidobacterium/genetics , Bifidobacterium/physiology , Genomics , Humans , Probiotics , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...