Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 30(2): 351-366.e7, 2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31940481

RESUMO

Human rhinoviruses cause the common cold and exacerbate chronic respiratory diseases. Although infection elicits neutralizing antibodies, these do not persist or cross-protect across multiple rhinovirus strains. To analyze rhinovirus-specific B cell responses in humans, we developed techniques using intact RV-A16 and RV-A39 for high-throughput high-dimensional single-cell analysis, with parallel assessment of antibody isotypes in an experimental infection model. Our approach identified T-bet+ B cells binding both viruses that account for ∼5% of CXCR5- memory B cells. These B cells infiltrate nasal tissue and expand in the blood after infection. Their rapid secretion of heterotypic immunoglobulin G (IgG) in vitro, but not IgA, matches the nasal antibody profile post-infection. By contrast, CXCR5+ memory B cells binding a single virus are clonally distinct, absent in nasal tissue, and secrete homotypic IgG and IgA, mirroring the systemic response. Temporal and spatial functions of dichotomous memory B cells might explain the ability to resolve infection while rendering the host susceptible to re-infection.


Assuntos
Linfócitos B/imunologia , Reações Cruzadas/imunologia , Imunoglobulina G/imunologia , Memória Imunológica/imunologia , Rhinovirus/imunologia , Humanos
2.
Sci Rep ; 8(1): 11411, 2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-30061588

RESUMO

The role of nasal and fecal microbiota in viral respiratory infections has not been established. We collected nasal swabs and washes, and fecal samples in a clinical study assessing the effect of probiotic Bifidobacterium animalis subsp. lactis Bl-04 on experimental rhinovirus infection. The nasal and fecal microbiota were characterized by 16S rRNA gene sequencing. The resulting data were compared with nasal inflammatory marker concentrations, viral load, and clinical symptoms. By using unsupervised clustering, the nasal microbiota divided into six clusters. The clusters predominant of Staphylococcus, Corynebacterium/Alloiococcus, Moraxella, and Pseudomonadaceae/Mixed had characteristic inflammatory marker and viral load profiles in nasal washes. The nasal microbiota clusters of subjects before the infection associated with the severity of clinical cold symptoms during rhinovirus infection. Rhinovirus infection and probiotic intervention did not significantly alter the composition of nasal or fecal microbiota. Our results suggest that nasal microbiota may influence the virus load, host innate immune response, and clinical symptoms during rhinovirus infection, however, further studies are needed.


Assuntos
Inflamação/patologia , Microbiota , Nariz/microbiologia , Nariz/virologia , Rhinovirus/fisiologia , Carga Viral , Bactérias/classificação , Biodiversidade , Biomarcadores/metabolismo , Análise por Conglomerados , Fezes/microbiologia , Humanos , Infecções por Picornaviridae/microbiologia , Infecções por Picornaviridae/virologia , Adulto Jovem
3.
J Infect Dis ; 217(3): 381-392, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29309618

RESUMO

Background: Little is known about T cells that respond to human rhinovirus in vivo, due to timing of infection, viral diversity, and complex T-cell specificities. We tracked circulating CD4+ T cells with identical epitope specificities that responded to intranasal challenge with rhinovirus (RV)-A39, and we assessed T-cell signatures in the nose. Methods: Cells were monitored using a mixture of 2 capsid-specific major histocompatibility complex II tetramers over a 7-week period, before and after RV-A39 challenge, in 16 human leukocyte antigen-DR4+ subjects who participated in a trial of Bifidobacterium lactis (Bl-04) supplementation. Results: Pre-existing tetramer+ T cells were linked to delayed viral shedding, enriched for activated CCR5+ Th1 effectors, and included a minor interleukin-21+ T follicular helper cell subset. After RV challenge, expansion and activation of virus-specific CCR5+ Th1 effectors was restricted to subjects who had a rise in neutralizing antibodies, and tetramer-negative CCR5+ effector memory types were comodulated. In the nose, CXCR3-CCR5+ T cells present during acute infection were activated effector memory type, whereas CXCR3+ cells were central memory type, and cognate chemokine ligands were elevated over baseline. Probiotic had no T-cell effects. Conclusions: We conclude that virus-specific CCR5+ effector memory CD4+ T cells primed by previous exposure to related viruses contribute to the control of rhinovirus.


Assuntos
Infecções por Enterovirus/imunologia , Enterovirus/imunologia , Memória Imunológica , Células Th1/imunologia , Adolescente , Adulto , Sangue/imunologia , Rastreamento de Células , Infecções por Enterovirus/virologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mucosa Nasal/imunologia , Receptores CCR5/análise , Adulto Jovem
4.
Data Brief ; 14: 118-122, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28861445

RESUMO

The present study describes the growth of the very well-known probiotic bacterium Lactobacillus acidophilus NCFM on different carbohydrates. Furthermore, recombinant production of putative moonlighting proteins elongation factor G and pyruvate kinase from this bacterium is described. For further and detailed interpretation of the data presented here, please see the research article "Mucin- and carbohydrate-stimulated adhesion and subproteome changes of the probiotic bacterium Lactobacillus acidophilus NCFM" (Celebioglu et al., 2017) [1].

5.
J Proteomics ; 163: 102-110, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28533178

RESUMO

Adhesion to intestinal mucosa is a crucial property for probiotic bacteria. Adhesion is thought to increase host-bacterial interactions, thus potentially enabling health benefits to the host. Molecular events connected with adhesion and surface proteome changes were investigated for the probiotic Lactobacillus acidophilus NCFM cultured with established or emerging prebiotic carbohydrates as carbon source and in the presence of mucin, the glycoprotein of the epithelial mucus layer. Variation in adhesion to HT29-cells and mucin was associated with carbon source and mucin-induced subproteome abundancy differences. Specifically, while growth on fructooligosaccharides (FOS) only stimulated adhesion to intestinal HT-29 cells, cellobiose and polydextrose in addition increased adhesion to mucin. Adhesion to HT-29 cells increased by about 2-fold for bacteria grown on mucin-supplemented glucose. Comparative 2DE-MS surface proteome analysis showed different proteins in energy metabolism appearing on the surface, suggesting they exert moonlighting functions. Mucin-supplemented bacteria had relative abundance of pyruvate kinase and fructose-bisphosphate aldolase increased by about 2-fold while six spots with 3.2-2.1 fold reduced relative abundance comprised elongation factor G, phosphoglycerate kinase, BipAEFTU family GTP-binding protein, ribonucleoside triphosphate reductase, adenylosuccinate synthetase, 30S ribosomal protein S1, and manganese-dependent inorganic pyrophosphatase. Surface proteome of cellobiose- compared to glucose-grown L. acidophilus NCFM had phosphate starvation inducible protein stress-related, thermostable pullulanase, and elongation factor G increasing 4.4-2.4 fold, while GAPDH, elongation factor Ts, and pyruvate kinase were reduced by 2.0-1.5 fold in relative abundance. Addition of recombinant L. acidophilus NCFM elongation factor G and pyruvate kinase to a coated mucin layer significantly suppressed subsequent adhesion of the bacterium. BIOLOGICAL SIGNIFICANCE: Human diet is important for intestinal health and food components, especially non-digestible carbohydrates can beneficially modify the microbiota. In the present study, effects of emerging and established prebiotic carbohydrates on the probiotic potential of Lactobacillus acidophilus NCFM were investigated by testing adhesion to a mucin layer and intestinal cells, and comparing this with changes in abundancy of surface proteins thought to be important for host interactions. Increased adhesion was observed following culturing of the bacterium with fructooligosaccharides, cellobiose or polydextrose, as well as mucin-supplemented glucose as carbon source. Enhanced adhesion ability can prolong bacterial residence in GIT yielding positive health effects. Higher relative abundance of certain surface proteins under various conditions (i.e. grown on cellobiose or mucin-supplemented glucose) suggested involvement of these proteins in adhesion, as confirmed by competition in case of two recombinantly produced moonlighting proteins. Combination of Lactobacillus acidophilus NCFM with different carbohydrates revealed potential bacterial determinants of synbiotic interactions, including stimulation of adhesion.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Lactobacillus acidophilus/química , Proteoma/metabolismo , Proteínas de Bactérias/análise , Carboidratos/farmacologia , Células HT29 , Humanos , Lactobacillus acidophilus/crescimento & desenvolvimento , Mucinas/farmacologia , Fator G para Elongação de Peptídeos/metabolismo , Probióticos , Piruvato Quinase/metabolismo
7.
Proteomics ; 16(9): 1361-75, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26959526

RESUMO

Whole cell and surface proteomes were analyzed together with adhesive properties of the probiotic bacterium Lactobacillus acidophilus NCFM (NCFM) grown on the emerging prebiotic raffinose, exemplifying a synbiotic. Adhesion of NCFM to mucin and intestinal HT-29 cells increased three-fold after culture with raffinose versus glucose, as also visualized by scanning electron microscopy. Comparative proteomics using 2D-DIGE showed 43 unique proteins to change in relative abundance in whole cell lysates from NCFM grown on raffinose compared to glucose. Furthermore, 14 unique proteins in 18 spots of the surface subproteome underwent changes identified by differential 2DE, including elongation factor G, thermostable pullulanase, and phosphate starvation inducible stress-related protein increasing in a range of +2.1 - +4.7 fold. By contrast five known moonlighting proteins decreased in relative abundance by up to -2.4 fold. Enzymes involved in raffinose catabolism were elevated in the whole cell proteome; α-galactosidase (+13.9 fold); sucrose phosphorylase (+5.4 fold) together with metabolic enzymes from the Leloir pathway for galactose utilization and the glycolysis; ß-galactosidase (+5.7 fold); galactose (+2.9/+3.1 fold) and fructose (+2.8 fold) kinases. The insights at the molecular and cellular levels contributed to the understanding of the interplay of a synbiotic composed of NCFM and raffinose with the host.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Lactobacillus acidophilus/efeitos dos fármacos , Probióticos/metabolismo , Proteoma/genética , Rafinose/farmacologia , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Galactose/metabolismo , Ontologia Genética , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Células HT29 , Humanos , Lactobacillus acidophilus/genética , Lactobacillus acidophilus/crescimento & desenvolvimento , Lactobacillus acidophilus/metabolismo , Anotação de Sequência Molecular , Fator G para Elongação de Peptídeos/genética , Fator G para Elongação de Peptídeos/metabolismo , Prebióticos , Proteoma/metabolismo , Coloração e Rotulagem , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo
8.
Biomed Res Int ; 2015: 347216, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25961012

RESUMO

Probiotics, prebiotics, and combinations thereof, that is, synbiotics, are known to exert beneficial health effects in humans; however interactions between pro- and prebiotics remain poorly understood at the molecular level. The present study describes changes in abundance of different proteins of the probiotic bacterium Lactobacillus acidophilus NCFM (NCFM) when grown on the potential prebiotic cellobiose as compared to glucose. Cytosolic cell extract proteomes after harvest at late exponential phase of NCFM grown on cellobiose or glucose were analyzed by two dimensional difference gel electrophoresis (2D-DIGE) in the acidic (pH 4-7) and the alkaline (pH 6-11) regions showing a total of 136 spots to change in abundance. Proteins were identified by MS or MS/MS from 81 of these spots representing 49 unique proteins and either increasing 1.5-13.9-fold or decreasing 1.5-7.8-fold in relative abundance. Many of these proteins were associated with energy metabolism, including the cellobiose related glycoside hydrolases phospho-ß-glucosidase (LBA0881) and phospho-ß-galactosidase II (LBA0726). The data provide insight into the utilization of the candidate prebiotic cellobiose by the probiotic bacterium NCFM. Several of the upregulated or downregulated identified proteins associated with utilization of cellobiose indicate the presence of carbon catabolite repression and regulation of enzymes involved in carbohydrate metabolism.


Assuntos
Glicosídeo Hidrolases/biossíntese , Lactobacillus acidophilus/enzimologia , Proteoma/genética , beta-Galactosidase/biossíntese , Proteínas de Bactérias/biossíntese , Celobiose/biossíntese , Regulação Bacteriana da Expressão Gênica , Glicosídeo Hidrolases/isolamento & purificação , Humanos , Lactobacillus acidophilus/genética , Prebióticos/microbiologia , Probióticos/metabolismo , Eletroforese em Gel Diferencial Bidimensional , beta-Galactosidase/isolamento & purificação
9.
FEMS Microbiol Ecol ; 90(1): 225-36, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25098489

RESUMO

Probiotics, prebiotics, and combinations thereof, that is synbiotics, have been reported to modulate gut microbiota of humans. In this study, effects of a novel synbiotic on the composition and metabolic activity of human gut microbiota were investigated. Healthy volunteers (n = 18) were enrolled in a double-blinded, randomized, and placebo-controlled cross-over study and received synbiotic [Lactobacillus acidophilus NCFM (10(9) CFU) and cellobiose (5 g)] or placebo daily for 3 weeks. Fecal samples were collected and lactobacilli numbers were quantified by qPCR. Furthermore, 454 tag-encoded amplicon pyrosequencing was used to monitor the effect of synbiotic on the composition of the microbiota. The synbiotic increased levels of Lactobacillus spp. and relative abundances of the genera Bifidobacterium, Collinsella, and Eubacterium while the genus Dialister was decreased (P < 0.05). No other effects were found on microbiota composition. Remarkably, however, the synbiotic increased concentrations of branched-chain fatty acids, measured by gas chromatography, while short-chain fatty acids were not affected.


Assuntos
Celobiose/farmacologia , Ácidos Graxos/biossíntese , Intestinos/microbiologia , Lactobacillus acidophilus , Microbiota , Simbióticos , Adulto , Bifidobacterium/genética , Bifidobacterium/isolamento & purificação , Biodiversidade , Estudos Cross-Over , Método Duplo-Cego , Ácidos Graxos Voláteis/biossíntese , Feminino , Humanos , Lactobacillus/genética , Lactobacillus/isolamento & purificação , Lactobacillus acidophilus/genética , Lactobacillus acidophilus/isolamento & purificação , Masculino , Adulto Jovem
10.
Clin Nutr ; 33(4): 581-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24268677

RESUMO

BACKGROUND & AIMS: To examine the effect of supplementation with probiotics on respiratory and gastrointestinal illness in healthy active men and women. METHODS: A randomised double-blind placebo-controlled trial was conducted. Four hundred and sixty five participants (241 males; age 35 ± 12 y (mean ± SD) and 224 females; age 36 ± 12 y) were assigned to one of three groups: Group 1 - Bifidobacterium animalis subsp. lactis Bl-04 (Bl-04) 2.0 × 10(9)colony forming units per day, CFU per day, Group 2 - Lactobacillus acidophilus NCFM and Bifidobacterium animalis subsp. lactis Bi-07 (NCFM & Bi-07) 5 × 10(9) CFU each per day) or Group 3 - placebo mixed in a drink. RESULTS: The risk of an upper respiratory illness episode was significantly lower in the Bl-04 group (hazard ratio 0.73; 95% confidence interval 0.55-0.95; P = 0.022) compared to placebo. There was no significant difference in illness risk between the NCFM & Bi-07 group (hazard ratio 0.81; 0.62-1.08; P = 0.15) and the placebo group. There was a 0.7 and 0.9 month delay in the median time to an illness episode in the Bl-04 and NCFM & Bi-07 groups respectively compared to placebo (placebo 2.5 months; Bl-04 3.2 months; NCFM & Bi-07 3.4 months). There were insufficient GI illness episodes for analysis. The NCFM & Bi-07 group but not the Bl-04 group undertook significantly more physical activity (8.5%; 6.7%-10%; P < 0.003) than the placebo group. CONCLUSION: The probiotic Bl-04 appears to be a useful nutritional supplement in reducing the risk of URTI in healthy physically-active adults. TRIAL REGISTRATION: Australia New Zealand Clinical Trials Registry: Number ACTRN12611000130965.


Assuntos
Suplementos Nutricionais , Gastroenteropatias/terapia , Probióticos/administração & dosagem , Adulto , Bifidobacterium , Índice de Massa Corporal , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Lactobacillus acidophilus , Masculino , Pessoa de Meia-Idade , Atividade Motora , Nova Zelândia , Adulto Jovem
11.
BMC Genomics ; 14: 312, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23663691

RESUMO

BACKGROUND: Probiotic bifidobacteria in combination with prebiotic carbohydrates have documented positive effects on human health regarding gastrointestinal disorders and improved immunity, however the selective routes of uptake remain unknown for most candidate prebiotics. The differential transcriptomes of Bifidobacterium animalis subsp. lactis Bl-04, induced by 11 potential prebiotic oligosaccharides were analyzed to identify the genetic loci involved in the uptake and catabolism of α- and ß-linked hexoses, and ß-xylosides. RESULTS: The overall transcriptome was modulated dependent on the type of glycoside (galactosides, glucosides or xylosides) utilized. Carbohydrate transporters of the major facilitator superfamily (induced by gentiobiose and ß-galacto-oligosaccharides (GOS)) and ATP-binding cassette (ABC) transporters (upregulated by cellobiose, GOS, isomaltose, maltotriose, melibiose, panose, raffinose, stachyose, xylobiose and ß-xylo-oligosaccharides) were differentially upregulated, together with glycoside hydrolases from families 1, 2, 13, 36, 42, 43 and 77. Sequence analysis of the identified solute-binding proteins that determine the specificity of ABC transporters revealed similarities in the breadth and selectivity of prebiotic utilization by bifidobacteria. CONCLUSION: This study identified the differential gene expression for utilization of potential prebiotics highlighting the extensive capabilities of Bifidobacterium lactis Bl-04 to utilize oligosaccharides. Results provide insights into the ability of this probiotic microbe to utilize indigestible carbohydrates in the human gastrointestinal tract.


Assuntos
Bifidobacterium/efeitos dos fármacos , Bifidobacterium/genética , Perfilação da Expressão Gênica , Oligossacarídeos/farmacologia , Prebióticos , Transcrição Gênica/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Mineração de Dados , Variação Genética/genética , Genômica , Família Multigênica/genética , Regulação para Cima/efeitos dos fármacos
12.
Food Funct ; 4(5): 784-93, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23580006

RESUMO

The present study aimed at examining oligosaccharides (OS) for potential stimulation of probiotic bacteria. Nineteen structurally well-defined candidate OS covering groups of ß-glucosides, α-glucosides and α-galactosides with degree of polymerization 2-4 were prepared in >100 mg amounts by chemoenzymatic synthesis (i.e. reverse phosphorolysis or transglycosylation). Fourteen of the OS are not naturally occurring and five (ß-D-glucosyl-fructose, ß-D-glucosyl-xylitol, α-glucosyl-(1,4)-D-mannose, α-glucosyl-(1,4)-D-xylose; α-glucosyl-(1,4)-L-fucose) have recently been synthesized for the first time. These OS have not been previously tested for effects of bacterial growth and here the ability of all 19 OS to support growth of four gastrointestinal bacteria: three probiotic bacteria Bifidobacterium lactis, Bifidobacterium longum, and Lactobacillus acidophilus, and one commensal bacterium, Bacteroides vulgatus has been evaluated in monocultures. The disaccharides ß-D-glucosyl-xylitol and ß-D-glucosyl-(1,4)-xylose noticeably stimulated growth yields of L. acidophilus NCFM, and additionally, ß-D-glucosyl-(1,4)-xylose stimulated B. longum Bl-05. α-Glucosyl-(1,4)-glucosamine and α-glucosyl-(1,4)-N-acetyl-glucosamine enhanced the growth rate of B. animalis subsp. lactis and B. longum Bl-05, whereas L. acidophilus NCFM and Bac. vulgatus did not grow on these OS. α-Galactosyl-(1,6)-α-galactosyl-(1,6)-glucose advanced the growth rate of B. animalis subsp. lactis and L. acidophilus NCFM. Thus several of the structurally well-defined OS supported growth of beneficial gut bacteria. This reflects a broad specificity of their sugar transporters for OS, including specificity for non-naturally occurring OS, hence showing promise for design of novel prebiotics.


Assuntos
Bacteroides/crescimento & desenvolvimento , Bifidobacterium/crescimento & desenvolvimento , Trato Gastrointestinal/microbiologia , Lactobacillus acidophilus/crescimento & desenvolvimento , Oligossacarídeos/química , Bacteroides/isolamento & purificação , Bifidobacterium/isolamento & purificação , Dissacarídeos/metabolismo , Humanos , Lactobacillus acidophilus/isolamento & purificação , Manose/metabolismo , Prebióticos/análise , Probióticos , Xilose/metabolismo
13.
Clin Nutr ; 32(6): 935-40, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23510724

RESUMO

BACKGROUND & AIMS: This study is a part of the clinical trials with probiotic bacterium Lactobacillus salivarius Ls-33 conducted in obese adolescents. Previously reported clinical studies showed no effect of Ls-33 consumption on the metabolic syndrome in the subject group. The aim of the study was to investigate the impact of L. salivarius Ls-33 on fecal microbiota in obese adolescents. METHODS: The study was a double-blinded intervention with 50 subjects randomized to intake of L. salivarius Ls-33 or placebo for 12 weeks. The fecal microbiota was assessed by real-time quantitative PCR before and after intervention. Concentrations of fecal short chain fatty acids were determined using gas chromatography. RESULTS: Ratios of Bacteroides-Prevotella-Porphyromonas group to Firmicutes belonging bacteria, including Clostridium cluster XIV, Blautia coccoides_Eubacteria rectale group and Roseburia intestinalis, were significantly increased (p ≤ 0.05) after administration of Ls-33. The cell numbers of fecal bacteria, including the groups above as well as Clostridium cluster I, Clostridium cluster IV, Faecalibacterium prausnitzii, Enterobacteriaceae, Enterococcus, the Lactobacillus group and Bifidobacterium were not significantly altered by intervention. Similarly, short chain fatty acids remained unaffected. CONCLUSION: L. salivarius Ls-33 might modify the fecal microbiota in obese adolescents in a way not related to metabolic syndrome. CLINICAL TRIAL NUMBER: NCT 01020617.


Assuntos
Fezes/microbiologia , Lactobacillus , Microbiota , Probióticos/administração & dosagem , Adolescente , Criança , DNA Bacteriano/isolamento & purificação , Método Duplo-Cego , Ácidos Graxos Voláteis/análise , Bactérias Gram-Positivas , Humanos , Síndrome Metabólica/terapia , Obesidade/terapia
14.
PLoS One ; 7(9): e44409, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028535

RESUMO

The human gastrointestinal tract can be positively modulated by dietary supplementation of probiotic bacteria in combination with prebiotic carbohydrates. Here differential transcriptomics and functional genomics were used to identify genes in Lactobacillus acidophilus NCFM involved in the uptake and catabolism of 11 potential prebiotic compounds consisting of α- and ß-linked galactosides and glucosides. These oligosaccharides induced genes encoding phosphoenolpyruvate-dependent sugar phosphotransferase systems (PTS), galactoside pentose hexuronide (GPH) permease, and ATP-binding cassette (ABC) transporters. PTS systems were upregulated primarily by di- and tri-saccharides such as cellobiose, isomaltose, isomaltulose, panose and gentiobiose, while ABC transporters were upregulated by raffinose, Polydextrose, and stachyose. A single GPH transporter was induced by lactitol and galactooligosaccharides (GOS). The various transporters were associated with a number of glycoside hydrolases from families 1, 2, 4, 13, 32, 36, 42, and 65, involved in the catabolism of various α- and ß-linked glucosides and galactosides. Further subfamily specialization was also observed for different PTS-associated GH1 6-phospho-ß-glucosidases implicated in the catabolism of gentiobiose and cellobiose. These findings highlight the broad oligosaccharide metabolic repertoire of L. acidophilus NCFM and establish a platform for selection and screening of both probiotic bacteria and prebiotic compounds that may positively influence the gastrointestinal microbiota.


Assuntos
Regulação Bacteriana da Expressão Gênica , Lactobacillus acidophilus/metabolismo , Prebióticos , Transportadores de Cassetes de Ligação de ATP/metabolismo , Celobiose/farmacologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Glucanos/farmacologia , Isomaltose/análogos & derivados , Isomaltose/farmacologia , Lactobacillus acidophilus/efeitos dos fármacos , Lactobacillus acidophilus/enzimologia , Oligossacarídeos/farmacologia , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/metabolismo , Rafinose/farmacologia , Álcoois Açúcares/farmacologia
15.
PLoS One ; 7(10): e47212, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23082149

RESUMO

BACKGROUND: Prebiotics, probiotics and synbiotics can be used to modulate both the composition and activity of the gut microbiota and thereby potentially affecting host health beneficially. The aim of this study was to investigate the effects of eight synbiotic combinations on the composition and activity of human fecal microbiota using a four-stage semicontinuous model system of the human colon. METHODS AND FINDINGS: Carbohydrates were selected by their ability to enhance growth of the probiotic bacteria Lactobacillus acidophilus NCFM (NCFM) and Bifidobacterium animalis subsp. lactis Bl-04 (Bl-04) under laboratory conditions. The most effective carbohydrates for each probiotic were further investigated, using the colonic model, for the ability to support growth of the probiotic bacteria, influence the composition of the microbiota and stimulate formation of short-chain fatty acids (SCFA).The following combinations were studied: NCFM with isomaltulose, cellobiose, raffinose and an oat ß-glucan hydrolysate (OBGH) and Bl-04 with melibiose, xylobiose, raffinose and maltotriose. All carbohydrates showed capable of increasing levels of NCFM and Bl-04 during fermentations in the colonic model by 10(3)-10(4) fold and 10-10(2) fold, respectively. Also the synbiotic combinations decreased the modified ratio of Bacteroidetes/Firmicutes (calculated using qPCR results for Bacteroides-Prevotella-Porphyromonas group, Clostridium perfringens cluster I, Clostridium coccoides - Eubacterium rectale group and Clostridial cluster XIV) as well as significantly increasing SCFA levels, especially acetic and butyric acid, by three to eight fold, as compared to the controls. The decreases in the modified ratio of Bacteroidetes/Firmicutes were found to be correlated to increases in acetic and butyric acid (p=0.04 and p=0.03, respectively). CONCLUSIONS: The results of this study show that all synbiotic combinations investigated are able to shift the predominant bacteria and the production of SCFA of fecal microbiota in a model system of the human colon, thereby potentially being able to manipulate the microbiota in a way connected to human health.


Assuntos
Bactérias/efeitos dos fármacos , Carboidratos/farmacologia , Colo/metabolismo , Colo/microbiologia , Ácidos Graxos Voláteis/biossíntese , Modelos Biológicos , Simbióticos , Bacteroides/efeitos dos fármacos , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/crescimento & desenvolvimento , Cromatografia Gasosa , Colo/efeitos dos fármacos , Fermentação/efeitos dos fármacos , Humanos , Lactobacillus/efeitos dos fármacos , Lactobacillus/crescimento & desenvolvimento , Metagenoma/efeitos dos fármacos , Metagenoma/genética , Reação em Cadeia da Polimerase , Prebióticos
16.
World J Gastroenterol ; 18(32): 4404-11, 2012 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-22969206

RESUMO

AIM: To compare quantities of predominant and pathogenic bacteria in mucosal and faecal samples. METHODS: Twenty patients undergoing diagnostic colonoscopy with endoscopically and histologically normal mucosa were recruited to the study, 14 subjects of which also supplied faecal (F) samples between 15 d to 105 d post colonoscopy. Mucosal biopsies were taken from each subject from the midportion of the ascending colon (right side samples, RM) and the sigmoid (left side samples, LM). Predominant intestinal and mucosal bacteria including clostridial 16S rRNA gene clusters IV and XIVab, Bacteroidetes, Enterobacteriaceae, Bifidobacterium spp., Akkermansia muciniphila (A. muciniphila), Veillonella spp., Collinsella spp., Faecalibacterium prausnitzii (F. prausnitzii) and putative pathogens such as Escherichia coli (E. coli), Clostridium difficile (C. difficile), Helicobacter pylori (H. pylori) and Staphylococcus aureus (S. aureus) were analysed by quantitative polymerase chain reaction (qPCR). Host DNA was quantified from the mucosal samples with human glyceraldehyde 3-phosphate dehydrogenase gene targeting qPCR. Paired t tests and the Pearson correlation were applied for statistical analysis. RESULTS: The most prominent bacterial groups were clostridial groups IV and XIVa+b and Bacteroidetes and bacterial species F. prausnitzii in both sample types. H. pylori and S. aureus were not detected and C. difficile was detected in only one mucosal sample and three faecal samples. E. coli was detected in less than half of the mucosal samples at both sites, but was present in all faecal samples. All detected bacteria, except Enterobacteriaceae, were present at higher levels in the faeces than in the mucosa, but the different locations in the colon presented comparable quantities (RM, LM and F followed by P(1) for RM vs F, P(2) for LM vs F and P(3) for RM vs LM: 4.17 ± 0.60 log(10)/g, 4.16 ± 0.56 log(10)/g, 5.88 ± 1.92 log(10)/g, P(1) = 0.011, P(2) = 0.0069, P(3) = 0.9778 for A. muciniphila; 6.25 ± 1.3 log(10)/g, 6.09 ± 0.81 log(10)/g, 8.84 ± 1.38 log(10)/g, P(1) < 0.0001, P(2) = 0.0002, P(3) = 0.6893 for Bacteroidetes; 5.27 ± 1.68 log(10)/g, 5.38 ± 2.06 log(10)/g, 8.20 ± 1.14 log(10)/g, P(1) < 0.0001, P(2) ≤ 0.0001, P(3) = 0.7535 for Bifidobacterium spp.; 6.44 ± 1.15 log(10)/g, 6.07 ±1.45 log(10)/g, 9.74 ±1.13 log(10)/g, P(1) < 0.0001, P(2) ≤ 0.0001, P(3) = 0.637 for Clostridium cluster IV; 6.65 ± 1.23 log(10)/g, 6.57 ± 1.52 log(10)/g, 9.13 ± 0.96 log(10)/g, P(1) < 0.0001, P(2) ≤ 0.0001, P(3) = 0.9317 for Clostridium cluster XIVa; 4.57 ± 1.44 log(10)/g, 4.63 ± 1.34 log(10)/g, 7.05 ± 2.48 log(10)/g, P(1) = 0.012, P(2) = 0.0357, P(3) = 0.7973 for Collinsella spp.; 7.66 ± 1.50 log(10)/g, 7.60 ± 1.05 log(10)/g, 10.02 ± 2.02 log(10)/g, P(1) ≤ 0.0001, P(2) = 0.0013, P(3) = 0.9919 for F. prausnitzsii; 6.17 ± 1.3 log(10)/g, 5.85 ± 0.93 log(10)/g, 7.25 ± 1.01 log(10)/g, P(1) = 0.0243, P(2) = 0.0319, P(3) = 0.6982 for Veillonella spp.; 4.68 ± 1.21 log(10)/g, 4.71 ± 0.83 log(10)/g, 5.70 ± 2.00 log(10)/g, P(1) = 0.1927, P(2) = 0.0605, P(3) = 0.6476 for Enterobacteriaceae). The Bifidobacterium spp. counts correlated significantly between mucosal sites and mucosal and faecal samples (Pearson correlation coefficients 0.62, P = 0.040 and 0.81, P = 0.005 between the right mucosal sample and faeces and the left mucosal sample and faeces, respectively). CONCLUSION: Non-invasive faecal samples do not reflect bacterial counts on the mucosa at the individual level, except for bifidobacteria often analysed in probiotic intervention studies.


Assuntos
Bactérias/isolamento & purificação , Bifidobacterium/isolamento & purificação , Colo/microbiologia , Fezes/microbiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Colo/metabolismo , Colonoscopia , Contagem de Colônia Microbiana , DNA Bacteriano/metabolismo , Feminino , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Pessoa de Meia-Idade
17.
Gastroenterol Res Pract ; 2012: 615051, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22848210

RESUMO

Gastrointestinal (GI) adverse effects such as erosion and increased permeability are common during the use of nonsteroidal anti-inflammatory drugs (NSAIDs). Our objective was to assess whether Bifidobacterium animalis ssp. lactis 420 protects against NSAID-induced GI side effects in a rat model. A total of 120 male Wistar rats were allocated into groups designated as control, NSAID, and probiotic. The NSAID and probiotic groups were challenged with indomethacin (10 mg/kg(-1); single dose). The probiotic group was also supplemented daily with 10(10) CFU of B. lactis 420 for seven days prior to the indomethacin administration. The control group rats received no indomethacin or probiotic. The permeability of the rat intestine was analysed using carbohydrate probes and the visual damage of the rat stomach mucosa was graded according to severity. B. lactis 420 significantly reduced the indomethacin-induced increase in stomach permeability. However, the protective effect on the visual mucosal damage was not significant. The incidence of severe NSAID-induced lesions was, nevertheless, reduced from 50% to 33% with the probiotic treatment. To conclude, the B. lactis 420 supplementation protected the rats from an NSAID-induced increase in stomach permeability and may reduce the formation of more serious GI mucosal damage and/or enhance the recovery rate of the stomach mucosa.

18.
Biosci Biotechnol Biochem ; 76(6): 1135-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22790935

RESUMO

Different ways of treating bran by baking enzymes prior to dough making and the baking process were used to increase the amount of water-soluble dietary fiber (DF) in wheat bread with added bran. Soluble DF was extracted from the bread with water and separated from the digestible material with gastrointestinal tract enzymes and by solvent precipitation. The baking enzyme mixtures tested (xylanase and glucanase/cellulase, with and without lipase) increased the amounts of soluble arabinoxylan and protein resistant to digestion. The isolated fiber was used as a growth substrate for 11 probiotic and intestinal Bifidobacterium strains, for commensal strains of Bacteroides fragilis and Escherichia coli, and for potential intestinal pathogenic strains of E. coli O157:H7, Salmonella typhimurium, and Clostridium perfringens. Fermentation analyses indicated that the tested strains had varying capacity to grow in the presence of the extracted fiber. Of the tested probiotic strains B. longum species generally showed the highest ability to utilize the fiber extracts, although the potential pathogens tested also showed an ability to grow on these fiber extracts. In sum, the enzymes used to improve the baking process for high-fiber bread can also be used to produce in situ soluble fiber material, which in turn can exert prebiotic effects on certain potentially beneficial microbes.


Assuntos
Pão/microbiologia , Fibras na Dieta/metabolismo , Probióticos/metabolismo , Bacteroides fragilis/crescimento & desenvolvimento , Bacteroides fragilis/metabolismo , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Celulases/metabolismo , Clostridium perfringens/crescimento & desenvolvimento , Clostridium perfringens/metabolismo , Endo-1,4-beta-Xilanases/metabolismo , Escherichia coli O157/crescimento & desenvolvimento , Escherichia coli O157/metabolismo , Fermentação , Microbiologia de Alimentos , Tecnologia de Alimentos , Lipase/metabolismo , Proteínas de Plantas/biossíntese , Xilanos/biossíntese
19.
BMC Microbiol ; 12: 94, 2012 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-22672382

RESUMO

BACKGROUND: The mucus layer covering the human intestinal epithelium forms a dynamic surface for host-microbial interactions. In addition to the environmental factors affecting the intestinal equilibrium, such as diet, it is well established that the microbiota composition is individually driven, but the host factors determining the composition have remained unresolved. RESULTS: In this study, we show that ABO blood group is involved in differences in relative proportion and overall profiles of intestinal microbiota. Specifically, the microbiota from the individuals harbouring the B antigen (secretor B and AB) differed from the non-B antigen groups and also showed higher diversity of the Eubacterium rectale-Clostridium coccoides (EREC) and Clostridium leptum (CLEPT) -groups in comparison with other blood groups. CONCLUSIONS: Our novel finding indicates that the ABO blood group is one of the genetically determined host factors modulating the composition of the human intestinal microbiota, thus enabling new applications in the field of personalized nutrition and medicine.


Assuntos
Sistema ABO de Grupos Sanguíneos , Biota , Trato Gastrointestinal/microbiologia , Metagenoma , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
20.
Proteomics ; 12(7): 1006-14, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22522807

RESUMO

Lactobacillus acidophilus NCFM (NCFM) is a well-documented probiotic bacterium isolated from human gut. Detailed 2D gel-based NCFM proteomics addressed the so-called alkaline range, i.e., pH 6-11. Proteins were identified in 150 of the 202 spots picked from the Coomassie Brilliant Blue stained 2D gel using MALDI-TOF-MS. The 102 unique gene products among the 150 protein identifications were assigned to different functional categories, and evaluated by considering a calculated distribution of abundance as well as grand average of hydrophobicity values. None of the very few available lactic acid bacteria proteome reference maps included the range of pI >7.0. The present report of such data on the proteome of NCFM fundamentally complements current knowledge on protein profiles limited to the acid and neutral pH range.


Assuntos
Proteínas de Bactérias/análise , Eletroforese em Gel Bidimensional/métodos , Lactobacillus acidophilus/química , Proteoma/análise , Proteômica/métodos , Proteínas de Bactérias/química , Proteínas de Bactérias/classificação , Concentração de Íons de Hidrogênio , Proteoma/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...