Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Gut Microbes ; 13(1): 1973835, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34553672

RESUMEN

Certain existing prebiotics meant to facilitate the growth of beneficial bacteria in the intestine also promote the growth of other prominent bacteria. Therefore, the growth-promoting effects of ß-galactosides on intestinal bacteria were analyzed. Galactosyl-ß1,4-l-rhamnose (Gal-ß1,4-Rha) selectively promoted the growth of Bifidobacterium. Bifidobacterium longum subsp. longum 105-A (JCM 31944) has multiple solute-binding proteins belonging to ATP-binding cassette transporters for sugars. Each strain in the library of 11 B. longum subsp. longum mutants, in which each gene of the solute-binding protein was disrupted, was cultured in a medium containing Gal-ß1,4-Rha as the sole carbon source, and only the BL105A_0502 gene-disruption mutant showed delayed and reduced growth compared to the wild-type strain. BL105A_0502 homolog is highly conserved in bifidobacteria. In a Gal-ß1,4-Rha-containing medium, Bifidobacterium longum subsp. infantis JCM 1222T, which possesses BLIJ_2090, a homologous protein to BL105A_0502, suppressed the growth of enteric pathogen Clostridioides difficile, whereas the BLIJ_2090 gene-disrupted mutant did not. In vivo, administration of B. infantis and Gal-ß1,4-Rha alleviated C. difficile infection-related weight loss in mice. We have successfully screened Gal-ß1,4-Rha as a next-generation prebiotic candidate that specifically promotes the growth of beneficial bacteria without promoting the growth of prominent bacteria and pathogens.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Bifidobacterium/crecimiento & desarrollo , Clostridioides difficile/crecimiento & desarrollo , Disacáridos/farmacología , Prebióticos/análisis , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Bifidobacterium/genética , Bifidobacterium longum subspecies infantis/genética , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL
2.
Front Immunol ; 12: 694344, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34211480

RESUMEN

Immunodeficiency is a very common condition in suboptimal health status and during the development or treatment of many diseases. Recently, probiotics have become an important means for immune regulation. The present study aimed to investigate the mechanism of the immunomodulatory effect of a combination of live Bifidobacterium, Lactobacillus, Enterococcus, and Bacillus (CBLEB), which is a drug used by approximately 10 million patients every year, on cyclophosphamide-immunosuppressed rats. Cyclophosphamide (40 mg/kg) was intraperitoneally injected to induce immunosuppression in a rat model on days 1, 2, 3, and 10. Starting from day 4, the rats were continuously gavaged with CBLEB solution for 15 days. The samples were collected to determine routine blood test parameters, liver and kidney functions, serum cytokine levels, gut microbiota, fecal and serum metabolomes, transcriptomes, and histopathological features. The results indicated that CBLEB treatment reduced cyclophosphamide-induced death, weight loss, and damage to the gut, liver, spleen, and lungs and eliminated a cyclophosphamide-induced increase in the mean hemoglobin content and GGT, M-CSF, and MIP-3α levels and a decrease in the red blood cell distribution width and total protein and creatinine levels in the blood. Additionally, CBLEB corrected cyclophosphamide-induced dysbiosis of the gut microbiota and eliminated all cyclophosphamide-induced alterations at the phylum level in rat feces, including the enrichment in Proteobacteria, Fusobacteriota, and Actinobacteriota and depletion of Spirochaetota and Cyanobacteria. Furthermore, CBLEB treatment alleviated cyclophosphamide-induced alterations in the whole fecal metabolome profile, including enrichment in 1-heptadecanol, succinic acid, hexadecane-1,2-diol, nonadecanoic acid, and pentadecanoic acid and depletion of benzenepropanoic acid and hexane. CBLEB treatment also alleviated cyclophosphamide-induced enrichment in serum D-lyxose and depletion of serum succinic acid, D-galactose, L-5-oxoproline, L-alanine, and malic acid. The results of transcriptome analysis indicated that the mechanism of the effect of CBLEB was related to the induction of recovery of cyclophosphamide-altered carbohydrate metabolism and signal transduction. In conclusion, the present study provides an experimental basis and comprehensive analysis of application of CBLEB for the treatment of immunodeficiency.


Asunto(s)
Bacillus cereus/crecimiento & desarrollo , Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Enterococcus faecalis/crecimiento & desarrollo , Microbioma Gastrointestinal , Huésped Inmunocomprometido , Síndromes de Inmunodeficiencia/terapia , Lactobacillus acidophilus/crecimiento & desarrollo , Probióticos , Animales , Bacillus cereus/inmunología , Bacillus cereus/metabolismo , Bifidobacterium longum subspecies infantis/inmunología , Bifidobacterium longum subspecies infantis/metabolismo , Ciclofosfamida , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Disbiosis , Metabolismo Energético , Enterococcus faecalis/inmunología , Enterococcus faecalis/metabolismo , Síndromes de Inmunodeficiencia/inducido químicamente , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/microbiología , Lactobacillus acidophilus/inmunología , Lactobacillus acidophilus/metabolismo , Masculino , Metaboloma , Ratas Sprague-Dawley , Transducción de Señal , Transcriptoma
3.
Int J Mol Sci ; 22(5)2021 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-33673498

RESUMEN

For bioactive milk peptides to be relevant to infant health, they must be released by gastrointestinal proteolysis and resist further proteolysis until they reach their site of activity. The intestinal tract is the likeliest site for most bioactivities, but it is currently unknown whether bioactive milk peptides are present therein. The purpose of the present study was to identify antimicrobial and bifidogenic peptides in the infant intestinal tract. Milk peptides were extracted from infant intestinal samples, and the activities of the bulk peptide extracts were determined by measuring growth of Escherichia coli, Staphylococcus aureus, and Bifidobacterium longum spp. infantis after incubation with serial dilutions. The peptide profiles of active and inactive samples were determined by peptidomics analysis and compared to identify candidate peptides for bioactivity testing. We extracted peptides from 29 intestinal samples collected from 16 infants. Five samples had antimicrobial activity against S. aureus and six samples had bifidogenic activity for B. infantis. We narrowed down a list of 6645 milk peptides to 11 candidate peptides for synthesis, of which 6 fully inhibited E. coli and S. aureus growth at concentrations of 2500 and 3000 µg/mL. This study provides evidence for the potential bioactivity of milk peptides in the infant intestinal tract.


Asunto(s)
Bifidobacterium longum subspecies infantis/efectos de los fármacos , Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Escherichia coli/efectos de los fármacos , Intestinos/microbiología , Péptidos/farmacología , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/farmacología , Humanos , Lactante , Intestinos/química , Leche Humana/química
4.
Benef Microbes ; 12(1): 69-83, 2021 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-33191780

RESUMEN

The establishment of the gut microbiota immediately after birth is a dynamic process that may impact lifelong health. At this important developmental stage in early life, human milk oligosaccharides (HMOs) serve as specific substrates to shape the gut microbiota of the nursling. The well-orchestrated transition is important as an aberrant microbial composition and bacterial-derived metabolites are associated with colicky symptoms and atopic diseases in infants. Here, we study the trophic interactions between an HMO-degrader, Bifidobacterium infantis and the butyrogenic Anaerostipes caccae using carbohydrate substrates that are relevant in the early life period including lactose and total human milk carbohydrates. Mono- and co-cultures of these bacterial species were grown at pH 6.5 in anaerobic bioreactors supplemented with lactose or total human milk carbohydrates. A. caccae was not able to grow on these substrates except when grown in co-culture with B. infantis, leading to growth and concomitant butyrate production. Two levels of cross-feeding were observed, in which A. caccae utilised the liberated monosaccharides as well as lactate and acetate produced by B. infantis. This microbial cross-feeding points towards the key ecological role of bifidobacteria in providing substrates for other important species that will colonise the infant gut. The progressive shift of the gut microbiota composition that contributes to the gradual production of butyrate could be important for host-microbial crosstalk and gut maturation.


Asunto(s)
Bifidobacterium longum subspecies infantis/metabolismo , Clostridiales/metabolismo , Lactosa/metabolismo , Leche Humana/metabolismo , Oligosacáridos/metabolismo , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Reactores Biológicos/microbiología , Clostridiales/genética , Clostridiales/crecimiento & desarrollo , Técnicas de Cocultivo , Medios de Cultivo/metabolismo , Humanos
5.
Benef Microbes ; 11(6): 527-534, 2020 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-33032471

RESUMEN

Bifidobacterium infantis NLS super strain (B. infantis NLS-SS) was previously shown to alleviate gastrointestinal symptoms in newly diagnosed coeliac disease (CD) patients consuming gluten. A high proportion of patients following a gluten-free diet experiences symptoms despite dietary compliance. The role of B. infantis in persistently symptomatic CD patients has not been explored. The aim of the study was to evaluate the effect of B. infantis NLS-SS on persistent gastrointestinal symptoms in patients with CD following a long-term GFD. We conducted a randomised, cross-over, double-blind, placebo-controlled trial in symptomatic adult CD patients on a GFD for at least two years. After one-week run-in, patients were randomised to B. infantis NLS-SS or placebo for 3 weeks with cross-over after a 2-week wash-out period. We estimated changes (Δ) in celiac symptom index (CSI) before and after treatment. Stool samples were collected for faecal microbiota analysis (16S rRNA sequencing). Gluten immunogenic peptide (GIP) excretion in stool and urine samples was measured at each study period. Eighteen patients were enrolled; six patients were excluded due violations in protocol. For patients with the highest clinical burden, CD symptoms were lower in probiotic than in placebo treatment (P=0.046). B. infantis and placebo treated groups had different microbiota profiles as assessed by beta diversity clustering. In probiotic treated groups, we observed an increase in abundance of B. infantis. Treatment with B. infantis was associated with decreased abundance of Ruminococcus sp. and Bifidobacterium adolescentis. GIP excretion in stools and urine was similar at each treatment period. There were no differences in adverse effects between the two groups. B. infantis NLS-SS improves specific CD symptoms in a subset of highly symptomatic treated patients (GFD). This is associated with a shift in stool microbiota profile. Larger studies are needed to confirm these findings. ClinicalTrials.gov: NCT03271138.


Asunto(s)
Bifidobacterium longum subspecies infantis , Enfermedad Celíaca/terapia , Dieta Sin Gluten , Microbioma Gastrointestinal , Probióticos/uso terapéutico , Adulto , Carga Bacteriana , Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Enfermedad Celíaca/dietoterapia , Enfermedad Celíaca/microbiología , Estudios Cruzados , Método Doble Ciego , Heces/química , Heces/microbiología , Femenino , Glútenes/análisis , Glútenes/orina , Humanos , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/orina , Ruminococcus/crecimiento & desarrollo
6.
Colloids Surf B Biointerfaces ; 188: 110815, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31986332

RESUMEN

It is important to develop efficient therapeutic methods to maintain a healthy balance among gut microbiota by increasing the beneficial bacteria and decreasing the harmful bacteria. In this work, a cationic polythiophene derivative poly(3-(3'-N,N,N-triethylamino-1'-propyloxy)-4-methyl-2,5-thiophene hydrochloride) (PMNT) with quaternary ammonium groups as side chains has been used for efficiently promoting the initial adhesion and biofilm formation of beneficial bacteria in gut microbiota. Upon addition of PMNT, three species of gut microbiota have an increased biofilm formation ability (216.5 % for Escherichia coli (E. coli), 130.7 % for Bifidobacterium infantis (B. infants) and 47.6 % for Enterococcus faecalis (E. faecalis)). As the initial adhesion of bacteria to a surface is an essential step during biofilm formation, PMNT can promote the attachment of bacteria by forming bacteria /PMNT aggregates which possess more cell-to-cell interactions. RNA sequencing results of bacteria within biofilm indicate that the utilization of carbohydrate and glycan is accelerated in the presence of PMNT, leading to enhanced quorum sensing and biofilm formation of E. coli. After forming biofilm, beneficial bacteria have an enhanced resistance to adverse environmental conditions which is significant for maintaining the balance of gut microbiota. Conjugated polymers exhibit a good potential application in modulating the balance of gut microbiota and development of new probiotics drugs.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Polímeros/farmacología , Compuestos de Amonio Cuaternario/farmacología , Tiofenos/farmacología , Antibacterianos/química , Adhesión Bacteriana/efectos de los fármacos , Bifidobacterium longum subspecies infantis/efectos de los fármacos , Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Biopelículas/crecimiento & desarrollo , Cationes/química , Cationes/farmacología , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/crecimiento & desarrollo , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Tamaño de la Partícula , Polímeros/química , Compuestos de Amonio Cuaternario/química , Propiedades de Superficie , Tiofenos/química
7.
Gut Microbes ; 12(1): 1690364, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-31760871

RESUMEN

Triclocarban (TCC) is a widely used antimicrobial ingredient in consumer products and is a ubiquitous contaminant in the environment. In 2016, the FDA removed TCC from over-the-counter handwashing products, but this compound is still approved for use in many other personal care products. A better understanding of its impact on human health could lead to significant impact for public health and regulatory policies. Here we show that exposure to low-dose TCC exaggerated the severity of colitis and exacerbated the development of colitis-associated colon tumorigenesis, via gut microbiota-dependent mechanisms. Exposure to TCC increased dextran sodium sulfate (DSS)- and interleukin 10 (IL-10) knockout-induced colitis, and exaggerated azoxymethane (AOM)/DSS-induced colon tumorigenesis in mice. Regarding the mechanisms, TCC exposure reduced the diversity and altered the composition of gut microbiota and failed to promote DSS-induced colitis in mice lacking the microbiota, supporting that the presence of the microbiota is critical for the pro-colitis effects of TCC. Together, these results support TCC could be a novel risk factor for colitis and colitis-associated colon cancer, and further regulatory policies on this compound could be needed.


Asunto(s)
Antiinfecciosos Locales/efectos adversos , Carbanilidas/efectos adversos , Transformación Celular Neoplásica/efectos de los fármacos , Colitis/inducido químicamente , Neoplasias del Colon/inducido químicamente , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Antiinfecciosos Locales/farmacología , Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Carbanilidas/farmacología , Colitis/microbiología , Colitis/patología , Neoplasias del Colon/patología , Sulfato de Dextran , Humanos , Inflamación/inducido químicamente , Interleucina-10/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contaminantes Químicos del Agua/efectos adversos
8.
Pediatr Res ; 86(6): 749-757, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31443102

RESUMEN

BACKGROUND: Infant gut dysbiosis, often associated with low abundance of bifidobacteria, is linked to impaired immune development and inflammation-a risk factor for increased incidence of several childhood diseases. We investigated the impact of B. infantis EVC001 colonization on enteric inflammation in a subset of exclusively breastfed term infants from a larger clinical study. METHODS: Stool samples (n = 120) were collected from infants randomly selected to receive either 1.8 × 1010 CFU B. infantis EVC001 daily for 21 days (EVC001) or breast milk alone (controls), starting at day 7 postnatal. The fecal microbiome was analyzed using 16S ribosomal RNA, proinflammatory cytokines using multiplexed immunoassay, and fecal calprotectin using ELISA at three time points: days 6 (Baseline), 40, and 60 postnatal. RESULTS: Fecal calprotectin concentration negatively correlated with Bifidobacterium abundance (P < 0.0001; ρ = -0.72), and proinflammatory cytokines correlated with Clostridiaceae and Enterobacteriaceae, yet negatively correlated with Bifidobacteriaceae abundance. Proinflammatory cytokines were significantly lower in EVC001-fed infants on days 40 and 60 postnatally compared to baseline and compared to control infants. CONCLUSION: Our findings indicate that gut dysbiosis (absence of B. infantis) is associated with increased intestinal inflammation. Early addition of EVC001 to diet represents a novel strategy to prevent enteric inflammation during a critical developmental phase.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Lactancia Materna , Enteritis/prevención & control , Citocinas/metabolismo , Enteritis/metabolismo , Enteritis/microbiología , Heces/química , Heces/microbiología , Femenino , Microbioma Gastrointestinal , Humanos , Recién Nacido , Mediadores de Inflamación/metabolismo , Complejo de Antígeno L1 de Leucocito/análisis , Masculino , Estudios Prospectivos
9.
Syst Appl Microbiol ; 42(4): 506-516, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31128887

RESUMEN

The infant gut harbors a diverse microbial community consisting of several taxa whose persistence depends on adaptation to the ecosystem. In healthy breast-fed infants, the gut microbiota is dominated by Bifidobacterium spp.. Cutibacterium avidum is among the initial colonizers, however, the phylogenetic relationship of infant fecal isolates to isolates from other body sites, and C. avidum carbon utilization related to the infant gut ecosystem have been little investigated. In this study, we investigated the phylogenetic and phenotypic diversity of 28 C. avidum strains, including 16 strains isolated from feces of healthy infants. We investigated the in vitro capacity of C. avidum infant isolates to degrade and consume carbon sources present in the infant gut, and metabolic interactions of C. avidum with infant associated Bifidobacterium longum subsp. infantis and Bifidobacterium bifidum. Isolates of C. avidum showed genetic heterogeneity. C. avidum consumed d- and l-lactate, glycerol, glucose, galactose, N-acetyl-d-glucosamine and maltodextrins. Alpha-galactosidase- and ß-glucuronidase activity were a trait of a group of non-hemolytic strains, which were mostly isolated from infant feces. Beta-glucuronidase activity correlated with the ability to ferment glucuronic acid. Co-cultivation with B. infantis and B. bifidum enhanced C. avidum growth and production of propionate, confirming metabolic cross-feeding. This study highlights the phylogenetic and functional diversity of C. avidum, their role as secondary glycan degraders and propionate producers, and suggests adaptation of a subpopulation to the infant gut.


Asunto(s)
Adaptación Fisiológica , Microbioma Gastrointestinal , Propionibacteriaceae/genética , Propionibacteriaceae/metabolismo , Bifidobacterium bifidum/crecimiento & desarrollo , Bifidobacterium bifidum/metabolismo , Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Bifidobacterium longum subspecies infantis/metabolismo , Heces/microbiología , Microbioma Gastrointestinal/genética , Genes Bacterianos/genética , Variación Genética , Genoma Bacteriano/genética , Humanos , Lactante , Interacciones Microbianas , Leche Humana/metabolismo , Filogenia , Polisacáridos/metabolismo , Propionatos/metabolismo , Propionibacteriaceae/clasificación , Propionibacteriaceae/crecimiento & desarrollo , Análisis de Secuencia de ADN
10.
Pharmazie ; 74(6): 326-333, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31138368

RESUMEN

Objective: Formulations containing probiotics are promoted due to health benefits. During lyophilization and subsequent storage in the gastrointestinal tract, bacteria are exposed to stress conditions that can lead to impairment and loss of viability. Methods: The suitability of various excipients for enhancing the stability and functionality of Bifidobacterium longum subsp. infantis during storage as freeze-dried powder and through exposure to acid and bile was investigated. Cells were lyophilized in the presence of sucrose, trehalose, lactose, cellobiose and fructooligosaccharide (FOS) and stored at 4 °C or 25 °C. The effect of diverse protectants on the persistence after exposure to acid and bile environment was examined through determination of the colony forming units, the ß-glucosidase and ß-galactosidase activity and the membrane integrity changes. Results: Cells freeze-dried in the presence of cryoprotectants had comparable survivability during storage at 4 °C whereas the survival rate at 25 °C of cells protected by cellobiose and FOS was higher than for those protected with sucrose and trehalose. Furthermore, the respective excipients used as cryoprotectants enhanced the stability of cells exposed to simulated gastric and small intestinal medium. Stabilization may be achieved through different mechanism of action such as protecting the membrane integrity and as metabolizable substrates. Overall, prebiotic and thus metabolizable protectants including cellobiose and FOS were superior to other protectants used. Conclusion: In symbiotic formulas with B. infantis, these sugars might serve as prebiotics and stabilizers of this probiotic strain during lyophilization, storage and in gastrointestinal conditions simultaneously, potentially increasing its health-promoting effects.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Excipientes , Prebióticos/microbiología , Tracto Gastrointestinal , Probióticos
11.
Benef Microbes ; 10(2): 111-120, 2019 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-30525951

RESUMEN

Individuals in a community who developed irritable bowel syndrome (IBS) after major floods have significant mental health impairment. We aimed to determine if Bifidobacterium infantis M-63 was effective in improving symptoms, psychology and quality of life measures in flood-affected individuals with IBS and if the improvement was mediated by gut microbiota changes. Design was non-randomised, open-label, controlled before-and-after. Of 53 participants, 20 with IBS were given B. infantis M-63 (1×109 cfu/sachet/day) for three months and 33 were controls. IBS symptom severity scale, hospital anxiety and depression scale, SF-36 Questionnaire, hydrogen breath testing for small intestinal bacterial overgrowth and stools for 16S rRNA metagenomic analysis were performed before and after intervention. 11 of 20 who were given probiotics (M-63) and 20 of 33 controls completed study as per-protocol. Mental well-being was improved with M-63 vs controls for full analysis (P=0.03) and per-protocol (P=0.01) populations. Within-group differences were observed for anxiety and bodily pain (both P=0.04) in the M-63 per-protocol population. Lower ratio of Firmicutes/Bacteroidetes was observed with M-63 vs controls (P=0.01) and the lower ratio was correlated with higher post-intervention mental score (P=0.04). B. infantis M-63 is probably effective in improving mental health of victims who developed IBS after floods and this is maybe due to restoration of microbial balance and the gut-brain axis. However, our conclusion must be interpreted within the context of limited sample size. The study was retrospectively registered on 12 October 2017 and the Trial Registration Number (TRN) was NCT03318614.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Inundaciones , Síndrome del Colon Irritable/complicaciones , Trastornos Mentales/terapia , Probióticos/administración & dosificación , Análisis por Conglomerados , Estudios Controlados Antes y Después , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Ribosómico/química , ADN Ribosómico/genética , Femenino , Microbioma Gastrointestinal , Humanos , Masculino , Salud Mental , Persona de Mediana Edad , Filogenia , Estudios Prospectivos , Calidad de Vida , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Resultado del Tratamiento
12.
Benef Microbes ; 9(6): 927-935, 2018 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-30099889

RESUMEN

The ban on the use of antibiotics as feed additives for animal growth promotion in the European Union and United States and the expectation of this trend to further expand to other countries in the short term have prompted a surge in probiotic research. Multi-species probiotics including safe and compatible strains with the ability to bind different nutritional lectins with detrimental effects on poultry nutrition could replace antibiotics as feed additives. Lactobacillus salivarius LET201, Lactobacillus reuteri LET210, Enterococcus faecium LET301, Propionibacterium acidipropionici LET103 and Bifidobacterium infantis CRL1395 have proved to be compatible as evaluated through three different approaches: the production and excretion of antimicrobial compounds, growth inhibition by competition for essential nutrients and physical contact, and a combination of both. The safety of P. acidipropionici LET103 was confirmed, since no expression of virulence factors or antibiotic resistance was detected. The innocuity of E. faecium LET301 should be further evaluated, since the presence of genes coding for certain virulence factors (gelE, efaAfm and efaAfs) was observed, albeit no expression of gelE was previously detected for this strain and there are no reports of involvement of efaAfm in animal pathogenicity. Finally, a combination of the five strains effectively protected intestinal epithelial cells of broilers from the cytotoxicity of mixtures of soybean agglutinin, wheat germ agglutinin and concanavalin A. To our knowledge, this is the first time that a combination of strains is evaluated for their protection against lectins that might be simultaneously present in poultry feeds.


Asunto(s)
Antiinfecciosos/metabolismo , Bifidobacterium longum subspecies infantis/metabolismo , Enterococcus faecium/metabolismo , Lactobacillus/metabolismo , Enfermedades de las Aves de Corral/prevención & control , Probióticos/farmacología , Propionibacterium/metabolismo , Animales , Antibiosis , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Bifidobacterium longum subspecies infantis/patogenicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Concanavalina A/toxicidad , Farmacorresistencia Bacteriana , Enterococcus faecium/genética , Enterococcus faecium/crecimiento & desarrollo , Enterococcus faecium/patogenicidad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Lactobacillus/genética , Lactobacillus/crecimiento & desarrollo , Lactobacillus/patogenicidad , Lectinas/metabolismo , Modelos Teóricos , Lectinas de Plantas/toxicidad , Probióticos/efectos adversos , Propionibacterium/genética , Propionibacterium/crecimiento & desarrollo , Propionibacterium/patogenicidad , Unión Proteica , Proteínas de Soja/toxicidad , Virulencia , Factores de Virulencia/genética , Aglutininas del Germen de Trigo/toxicidad
13.
J Dairy Sci ; 101(8): 6730-6741, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29803426

RESUMEN

Bovine milk glycomacropeptide (GMP) is derived from κ-casein, with exclusively o-linked glycosylation. Glycomacropeptide promoted the growth of Bifidobacterium longum ssp. infantis in a concentration-dependent manner, and this activity was lost following periodate treatment of the GMP (GMP-P), which disables biological recognition of the conjugated oligosaccharides. Transcriptional analysis of B. longum ssp. infantis following exposure to GMP revealed a substantial response to GMP relative to bacteria treated with GMP-P, with a greater number of differentially expressed transcripts and larger fold changes versus the control. Therefore, stimulation of B. longum ssp. infantis growth by GMP is intrinsically linked to the peptide's O-linked glycosylation. The pool of differentially expressed transcripts included 2 glycoside hydrolase (family 25) genes, which were substantially upregulated following exposure to GMP, but not GMP-P. These GH25 genes were present in duplicated genomic islands that also contained genes encoding fibronectin type III binding domain proteins and numerous phage-related proteins, all of which were also upregulated. Homologs of this genomic arrangement were present in other Bifidobacterium species, which suggest it may be a conserved domain for the utilization of glycosylated peptides. This study provides insights into the molecular basis for the prebiotic effect of bovine milk GMP on B. longum ssp. infantis.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Bifidobacterium longum subspecies infantis/genética , Caseínas/farmacología , Regulación Bacteriana de la Expresión Génica , Fragmentos de Péptidos/farmacología , Animales , Bovinos , Oligosacáridos
14.
J Appl Microbiol ; 124(2): 572-584, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29236340

RESUMEN

AIMS: Classical microbiology techniques are the gold standard for probiotic enumeration. However, these techniques are limited by parameters of time, specificity and incapacity to detect viable but nonculturable (VBNC) micro-organisms and nonviable cells. The aim of the study was to evaluate flow cytometry as a novel method for the specific quantification of viable and nonviable probiotics in multistrain products. METHODS AND RESULTS: Custom polyclonal antibodies were produced against five probiotic strains from different species (Bifidobacterium bifidum R0071, Bifidobacterium longum ssp. infantis R0033, Bifidobacterium longum ssp. longum R0175, Lactobacillus helveticus R0052 and Lactobacillus rhamnosus R0011). Evaluation of specificity confirmed that all antibodies were specific at least at the subspecies level. A flow cytometry method combining specific antibodies and viability assessment with SYTO® 24 and propidium iodide was applied to quantify these strains in three commercial products. Analyses were conducted on two flow cytometry instruments by two operators and compared with classical microbiology using selective media. Results indicated that flow cytometry provides higher cell counts than classical microbiology (P < 0·05) in 73% of cases highlighting the possible presence of VBNC. Equivalent performances (repeatability and reproducibility) were obtained for both methods. CONCLUSIONS: This study showed that flow cytometry methods can be applied to probiotic enumeration and viability assessment. Combination with polyclonal antibodies can achieve sufficient specificity to differentiate closely related strains. SIGNIFICANCE AND IMPACT OF THE STUDY: Flow cytometry provides absolute and specific quantification of viable and nonviable probiotic strains in a very short time (<2 h) compared with classical techniques (>48 h), bringing efficient tools for research and development and quality control.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Citometría de Flujo/métodos , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Lactobacillus helveticus/crecimiento & desarrollo , Probióticos/química , Bifidobacterium longum subspecies infantis/química , Bifidobacterium longum subspecies infantis/aislamiento & purificación , Lactobacillus helveticus/química , Lactobacillus helveticus/aislamiento & purificación , Lacticaseibacillus rhamnosus/química , Lacticaseibacillus rhamnosus/aislamiento & purificación , Viabilidad Microbiana , Reproducibilidad de los Resultados
15.
J Clin Gastroenterol ; 51(9): 814-817, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27636409

RESUMEN

BACKGROUND: We have previously shown a reduction of gastrointestinal symptoms after the oral administration of Bifidobacterium infantis Natren Life Start super strain (NLS-SS) in untreated celiac disease (CD) patients. The symptomatic improvement was not associated with changes in intestinal permeability or serum levels of cytokines, chemokines, or growth factors. Therefore, we hypothesized that the beneficial symptomatic effect observed previously in patients with CD treated with B. infantis may be related to the modulation of innate immunity. GOALS: To investigate the potential mechanisms of a probiotic B. infantis Natren Life Start super strain on the mucosal expression of innate immune markers in adult patients with active untreated CD compared with those treated with B. infantis×6 weeks and after 1 year of gluten-free diet (GFD). METHODS: Numbers of macrophages and Paneth cells and α-defensin-5 expression were assessed by immunohistochemistry in duodenal biopsies. RESULTS: We showed that GFD decreases duodenal macrophage counts in CD patients more effectively than B. infantis. In contrast, B. infantis decreases Paneth cell counts and expression of α-defensin-5 in CD (P<0.001). CONCLUSIONS: The results identify differential innate immune effects of treatment with B. infantis compared with 1 year of GFD. Further studies are needed to investigate synergistic effects of GFD and B. infantis supplementation in CD.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Enfermedad Celíaca/terapia , Dieta Sin Gluten , Duodeno/metabolismo , Inmunidad Innata , Inmunidad Mucosa , Mucosa Intestinal/metabolismo , Probióticos/uso terapéutico , alfa-Defensinas/metabolismo , Adulto , Biomarcadores/metabolismo , Biopsia , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/microbiología , Regulación hacia Abajo , Duodeno/inmunología , Duodeno/microbiología , Femenino , Microbioma Gastrointestinal , Humanos , Inmunohistoquímica , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Persona de Mediana Edad , Células de Paneth/inmunología , Células de Paneth/metabolismo , Células de Paneth/microbiología , Probióticos/efectos adversos , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
16.
Food Chem ; 216: 97-105, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27596397

RESUMEN

Mucilage (M) and soluble protein (SP) extracted from chia seed and flaxseed were used as encapsulating material for two probiotic bacteria: Bifidobacterium infantis and Lactobacillus plantarum by spray drying. Probiotic survival and viability after spray drying and during storage were evaluated. B. infantis and L. plantarum displayed high survival (⩾98%) after encapsulation with mixtures of maltodextrin (MD) combined with M and SP from flaxseed (MD:FM:FSP - 7.5:0.2:7.5%, w/w/w) and chia seed (MD:CM:CSP - 7.5:0.6:7.5%, w/w/w), respectively. These ternary blends protected the probiotics and enhanced their resistance to simulated gastric juice and bile solution. Probiotics encapsulated with the ternary blends incorporated in instant juice powder exhibited high viability (>9Log10CFU/g) after 45days refrigerated storage. Encapsulation with the ternary blends reduced particle size of the probiotic powders thereby offering additional functional benefits. Our results reveal that chia seed and flaxseed are excellent sources of probiotic encapsulating agents.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Lino/química , Lactobacillus plantarum/crecimiento & desarrollo , Exudados de Plantas/química , Polvos/química , Probióticos/administración & dosificación , Salvia/química , Semillas/química , Bifidobacterium longum subspecies infantis/efectos de los fármacos , Desecación , Liofilización , Jugo Gástrico/efectos de los fármacos , Jugo Gástrico/microbiología , Lactobacillus plantarum/efectos de los fármacos
17.
Trials ; 17(1): 340, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27449926

RESUMEN

BACKGROUND: The development of probiotics as therapies to cure or prevent disease lags far behind that of other investigational medications. Rigorously designed phase I clinical trials are nearly non-existent in the field of probiotic research, which is a contributing factor to this disparity. As a consequence, how to appropriately dose probiotics to study their efficacy is unknown. Herein we propose a novel phase I ascending dose trial of Bifidobacterium longum subsp. infantis (B. infantis) to identify the dose required to produce predominant gut colonisation in healthy breastfed infants at 6 weeks of age. METHODS/DESIGN: This is a parallel-group, placebo-controlled, randomised, double-blind ascending dose phase I clinical trial of dietary supplementation with B. infantis in healthy breastfed infants. The objective is to determine the pharmacologically effective dose (ED) of B. infantis required to produce predominant (>50 %) gut colonisation in breastfed infants at 6 weeks of age. Successively enrolled infant groups will be randomised to receive two doses of either B. infantis or placebo on days 7 and 14 of life. Stool samples will be used to characterise the gut microbiota at increasing doses of B. infantis. DISCUSSION: Probiotic supplementation has shown promising results for the treatment of a variety of ailments, but evidence-based dosing regimes are currently lacking. The ultimate goal of this trial is to establish a recommended starting dose of B. infantis for further efficacy-testing phase II trials designed to evaluate B. infantis for the prevention of atopic dermatitis and food allergies in at-risk children. TRIAL REGISTRATION: Clinicaltrials.gov # NCT02286999 , date of trial registration 23 October 2014.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Lactancia Materna , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Probióticos/uso terapéutico , Factores de Edad , California , Protocolos Clínicos , Método Doble Ciego , Heces/microbiología , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Probióticos/efectos adversos , Proyectos de Investigación , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...