Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Discov ; 9(1): 114, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37968259

RESUMEN

CD8+ T cell activation via immune checkpoint blockade (ICB) is successful in microsatellite instable (MSI) colorectal cancer (CRC) patients. By comparison, the success of immunotherapy against microsatellite stable (MSS) CRC is limited. Little is known about the most critical features of CRC CD8+ T cells that together determine the diverse immune landscapes and contrasting ICB responses. Hence, we pursued a deep single cell mapping of CRC CD8+ T cells on transcriptomic and T cell receptor (TCR) repertoire levels in a diverse patient cohort, with additional surface proteome validation. This revealed that CRC CD8+ T cell dynamics are underscored by complex interactions between interferon-γ signaling, tumor reactivity, TCR repertoire, (predicted) TCR antigen-specificities, and environmental cues like gut microbiome or colon tissue-specific 'self-like' features. MSI CRC CD8+ T cells showed tumor-specific activation reminiscent of canonical 'T cell hot' tumors, whereas the MSS CRC CD8+ T cells exhibited tumor unspecific or bystander-like features. This was accompanied by inflammation reminiscent of 'pseudo-T cell hot' tumors. Consequently, MSI and MSS CRC CD8+ T cells showed overlapping phenotypic features that differed dramatically in their TCR antigen-specificities. Given their high discriminating potential for CD8+ T cell features/specificities, we used the single cell tumor-reactive signaling modules in CD8+ T cells to build a bulk tumor transcriptome classification for CRC patients. This "Immune Subtype Classification" (ISC) successfully distinguished various tumoral immune landscapes that showed prognostic value and predicted immunotherapy responses in CRC patients. Thus, we deliver a unique map of CRC CD8+ T cells that drives a novel tumor immune landscape classification, with relevance for immunotherapy decision-making.

2.
Int J Mol Sci ; 24(3)2023 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-36768196

RESUMEN

Environmental factors, including westernised diets and alterations to the gut microbiota, are considered risk factors for inflammatory bowel diseases (IBD). The mechanisms underpinning diet-microbiota-host interactions are poorly understood in IBD. We present evidence that feeding a lard-based high-fat (HF) diet can protect mice from developing DSS-induced acute and chronic colitis and colitis-associated cancer (CAC) by significantly reducing tumour burden/incidence, immune cell infiltration, cytokine profile, and cell proliferation. We show that HF protection was associated with increased gut microbial diversity and a significant reduction in Proteobacteria and an increase in Firmicutes and Clostridium cluster XIVa abundance. Microbial functionality was modulated in terms of signalling fatty acids and bile acids (BA). Faecal secondary BAs were significantly induced to include moieties that can activate the vitamin D receptor (VDR), a nuclear receptor richly represented in the intestine and colon. Indeed, colonic VDR downstream target genes were upregulated in HF-fed mice and in combinatorial lipid-BAs-treated intestinal HT29 epithelial cells. Collectively, our data indicate that HF diet protects against colitis and CAC risk through gut microbiota and BA metabolites modulating vitamin D targeting pathways. Our data highlights the complex relationship between dietary fat-induced alterations of microbiota-host interactions in IBD/CAC pathophysiology.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Neoplasias , Ratones , Animales , Vitamina D/metabolismo , Inflamación/metabolismo , Colitis/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Colon/patología , Dieta Alta en Grasa/efectos adversos , Bacterias , Ácidos y Sales Biliares/metabolismo , Ratones Endogámicos C57BL , Sulfato de Dextran/efectos adversos , Neoplasias/metabolismo
3.
Neuropsychobiology ; 80(3): 253-263, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33075780

RESUMEN

INTRODUCTION: Butyrate is a short-chain fatty acid metabolite produced by microbiota in the colon. With its antioxidant properties, butyrate has also been shown to alter the neurological functions in affective disorder models, suggesting it as a key mediator in gut-brain interactions. OBJECTIVE: Here, we evaluated the negative effect of oxidative stress on the transport of the serotonin precursor tryptophan as present in affective disorders. Butyrate was hypothesized to be able to rescue these deficits due to its antioxidative capacities and its effect on transmembrane transport of tryptophan. Human skin-derived fibroblasts were used as cellular models to address these objectives. METHODS: Human fibroblasts were treated with hydrogen peroxide to induce oxidative stress. Stressed as well as control cells were treated with different concentrations of butyrate. Tryptophan (3H) was used as a tracer to measure the transport of tryptophan across the cell membranes (n = 6). Furthermore, gene expression profiles of different amino acid transporters were analyzed (n = 2). RESULTS: As hypothesized,oxidative stress significantly decreased the uptake of tryptophan in fibroblast cells, while butyrate counteracted this effect. Oxidative stress did not alter the gene expression profile of amino acid transporters. However, treatment of stressed and control cells with different concentrations of butyrate differentially regulated the gene expression of large amino acid transporters 1 and 2, which are the major transporters of tryptophan. CONCLUSIONS: Gut microbiota-derived butyrate may have therapeutic potential in affective disorders characterized by either aberrant serotonergic activity or neuroinflammation due to its role in rescuing the oxidative stress-induced perturbations of tryptophan transport.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Encéfalo/metabolismo , Butiratos/metabolismo , Fibroblastos/metabolismo , Microbioma Gastrointestinal/fisiología , Expresión Génica/fisiología , Trastornos del Humor/metabolismo , Estrés Oxidativo/fisiología , Triptófano/metabolismo , Sistemas de Transporte de Aminoácidos/efectos de los fármacos , Butiratos/farmacología , Expresión Génica/efectos de los fármacos , Humanos , Trastornos del Humor/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos
4.
Nutrients ; 12(9)2020 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-32867153

RESUMEN

BACKGROUND: Gastrointestinal (GI) health is an important aspect of general health. Gastrointestinal symptoms are of specific importance for the elderly, an increasing group globally. Hence, promoting the elderly's health and especially gastrointestinal health is important. Gut microbiota can influence gastrointestinal health by modulation of the immune system and the gut-brain axis. Diverse gut microbiota have been shown to be beneficial; however, for the elderly, the gut microbiota is often less diverse. Nutrition and physical activity, in particular, are two components that have been suggested to influence composition or diversity. MATERIALS AND METHODS: In this study, we compared gut microbiota between two groups of elderly individuals: community-dwelling older adults and physically active senior orienteering athletes, where the latter group has less gastrointestinal symptoms and a reported better well-being. With this approach, we explored if certain gut microbiota were related to healthy ageing. The participant data and faecal samples were collected from these two groups and the microbiota was whole-genome sequenced and taxonomically classified with MetaPhlAn. RESULTS: The physically active senior orienteers had a more homogeneous microbiota within the group and a higher abundance of Faecalibacterium prausnitzii compared to the community-dwelling older adults. Faecalibacterium prausnitzii has previously shown to have beneficial properties. Senior orienteers also had a lower abundance of Parasutterella excrementihominis and Bilophila unclassified, which have been associated with impaired GI health. We could not observe any difference between the groups in terms of Shannon diversity index. Interestingly, a subgroup of community-dwelling older adults showed an atypical microbiota profile as well as the parameters for gastrointestinal symptoms and well-being closer to senior orienteers. CONCLUSIONS: Our results suggest specific composition characteristics of healthy microbiota in the elderly, and show that certain components of nutrition as well as psychological distress are not as tightly connected with composition or diversity variation in faecal microbiota samples.


Asunto(s)
Atletas/estadística & datos numéricos , Microbioma Gastrointestinal , Evaluación Geriátrica/métodos , Evaluación Geriátrica/estadística & datos numéricos , Anciano , Heces/microbiología , Femenino , Tracto Gastrointestinal/microbiología , Humanos , Vida Independiente , Masculino
5.
Biomolecules ; 10(5)2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32422994

RESUMEN

Intact intestinal barrier function is essential for maintaining intestinal homeostasis. A dysfunctional intestinal barrier can lead to local and systemic inflammation through translocation of luminal antigens and has been associated with a range of health disorders. Butyrate, a short-chain fatty acid derived from microbial fermentation of dietary fibers in the colon, has been described as an intestinal barrier-strengthening agent, although mainly by using in vitro and animal models. This study aimed to investigate butyrate's ability to prevent intestinal hyperpermeability, induced by the mast cell degranulator Compound 48/80 (C48/80), in human colonic tissues. Colonic biopsies were collected from 16 healthy subjects and intestinal permeability was assessed by Ussing chamber experiments. Furthermore, the expression levels of tight junction-related proteins were determined by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Pre-treatment with 5 mM butyrate or 25 mM butyrate did not protect the colonic tissue against induced paracellular or transcellular hyperpermeability, measured by FITC-dextran and horseradish peroxidase passage, respectively. Biopsies treated with 25 mM butyrate prior to stimulation with C48/80 showed a reduced expression of claudin 1. In conclusion, this translational ex vivo study did not demonstrate an acute protective effect of butyrate against a chemical insult to the intestinal barrier in healthy humans.


Asunto(s)
Butiratos/farmacología , Colon/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Proteínas de Uniones Estrechas/metabolismo , Adulto , Colon/citología , Colon/metabolismo , Impedancia Eléctrica , Femenino , Humanos , Absorción Intestinal , Mucosa Intestinal/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de Uniones Estrechas/genética , Transcitosis
6.
BMC Syst Biol ; 12(1): 144, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30558589

RESUMEN

BACKGROUND: Gut microbiota interacts with the human gut in multiple ways. Microbiota composition is altered in inflamed gut conditions. Likewise, certain microbial fermentation products as well as the lipopolysaccharides of the outer membrane are examples of microbial products with opposing influences on gut epithelium inflammation status. This system of intricate interactions is known to play a core role in human gut inflammatory diseases. Here, we present and analyse a simplified model of bidirectional interaction between the microbiota and the host: in focus is butyrate as an example for a bacterial fermentation product with anti-inflammatory properties. RESULTS: We build a dynamical model based on an existing model of inflammatory regulation in gut epithelial cells. Our model introduces both butyrate as a bacterial product which counteracts inflammation, as well as bacterial LPS as a pro-inflammatory bacterial product. Moreover, we propose an extension of this model that also includes a feedback interaction towards bacterial composition. The analysis of these dynamical models shows robust bi-stability driven by butyrate concentrations in the gut. The extended model hints towards a further possible enforcement of the observed bi-stability via alteration of gut bacterial composition. A theoretical perspective on the stability of the described switch-like character is discussed. CONCLUSIONS: Interpreting the results of this qualitative model allows formulating hypotheses about the switch-like character of inflammatory regulation in the gut epithelium, involving bacterial products as constitutive parts of the system. We also speculate about possible explanations for observed bimodal distributions in bacterial compositions in the human gut. The switch-like behaviour of the system proved to be mostly independent of parameter choices. Further implications of the qualitative character of our modeling approach for the robustness of the proposed hypotheses are discussed, as well as the pronounced role of butyrate compared to other inflammatory regulators, especially LPS, NF- κB and cytokines.


Asunto(s)
Butiratos/metabolismo , Microbioma Gastrointestinal , Modelos Biológicos , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Distribución Normal
7.
Sci Rep ; 7: 43300, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28265110

RESUMEN

This study focused on the mechanisms that fatty acid conjugating strains - Bifidobacterium breve NCIMB 702258 and Bifidobacterium breve DPC 6330 - influence lipid metabolism when ingested with α-linolenic acid (ALA) enriched diet. Four groups of BALB/c mice received ALA enriched diet (3% (w/w)) either alone or in combination with B. breve NCIMB 702258 or B. breve DPC 6330 (109 CFU/day) or unsupplemented control diet for six weeks. The overall n-3 PUFA score was increased in all groups receiving the ALA enriched diet. Hepatic peroxisomal beta oxidation increased following supplementation of the ALA enriched diet with B. breve (P < 0.05) and so the ability of the strains to produce c9t11 conjugated linoleic acid (CLA) was identified in adipose tissue. Furthermore, a strain specific effect of B. breve NCIMB 702258 was found on the endocannabinoid system (ECS). Liver triglycerides (TAG) were reduced following ALA supplementation, compared with unsupplemented controls (P < 0.01) while intervention with B. breve further reduced liver TAG (P < 0.01), compared with the ALA enriched control. These data indicate that the interactions of the gut microbiota with fatty acid metabolism directly affect host health by modulating n-3 PUFA score and the ECS.


Asunto(s)
Bifidobacterium breve/metabolismo , Dieta/métodos , Metabolismo de los Lípidos , Probióticos/administración & dosificación , Ácido alfa-Linolénico/administración & dosificación , Animales , Ratones Endogámicos BALB C
8.
Br J Nutr ; 113(5): 728-38, 2015 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-25697178

RESUMEN

The main aim of the present study was to investigate the effects of dietary trans-10, cis-12-conjugated linoleic acid (t10c12-CLA) on intestinal microbiota composition and SCFA production. C57BL/6 mice (n 8 per group) were fed a standard diet either supplemented with t10c12-CLA (0·5 %, w/w) (intervention) or with no supplementation (control), daily for 8 weeks. Metabolic markers (serum glucose, leptin, insulin and TAG, and liver TAG) were assessed by ELISA commercial kits, tissue long-chain fatty acids and caecal SCFA by GC, and microbial composition by 16S rRNA pyrosequencing. Dietary t10c12-CLA significantly decreased visceral fat mass (P< 0·001), but did not affect body weight (intervention), when compared with no supplementation (control). Additionally, lipid mass and composition were affected by t10c12-CLA intake. Caecal acetate, propionate and isobutyrate concentrations were higher (P< 0·05) in the t10c12-CLA-supplemented group than in the control group. The analysis of the microbiota composition following 8 weeks of t10c12-CLA supplementation revealed lower proportions of Firmicutes (P= 0·003) and higher proportions of Bacteroidetes (P= 0·027) compared with no supplementation. Furthermore, t10c12-CLA supplementation for 8 weeks significantly altered the gut microbiota composition, harbouring higher proportions of Bacteroidetes, including Porphyromonadaceae bacteria previously linked with negative effects on lipid metabolism and induction of hepatic steatosis. These results indicate that the mechanism of dietary t10c12-CLA on lipid metabolism in mice may be, at least, partially mediated by alterations in gut microbiota composition and functionality.


Asunto(s)
Fármacos Antiobesidad/efectos adversos , Suplementos Dietéticos/efectos adversos , Ácidos Grasos Volátiles/metabolismo , Mucosa Intestinal/microbiología , Intestinos/microbiología , Ácidos Linoleicos Conjugados/efectos adversos , Microbiota , Adiposidad , Animales , Bacteroidetes/clasificación , Bacteroidetes/crecimiento & desarrollo , Bacteroidetes/aislamiento & purificación , Bacteroidetes/metabolismo , Biomarcadores/análisis , Biomarcadores/sangre , Biomarcadores/metabolismo , Ciego , Ácidos Grasos Volátiles/análisis , Contenido Digestivo/química , Contenido Digestivo/microbiología , Mucosa Intestinal/metabolismo , Grasa Intraabdominal/patología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Tipificación Molecular , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/patología , Tamaño de los Órganos
9.
J Nutr ; 144(12): 1956-62, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25320181

RESUMEN

BACKGROUND: Probiotic bacteria have been associated with a reduction in cardiovascular disease risk, a leading cause of death and disability. OBJECTIVES: The aim of this study was to assess the impact of dietary administration of exopolysaccharide-producing probiotic Lactobacillus cultures on lipid metabolism and gut microbiota in apolipoprotein E (apoE)-deficient mice. METHODS: First, we examined lipid metabolism in response to dietary supplementation with recombinant ß-glucan-producing Lactobacillus paracasei National Food Biotechnology Centre (NFBC) 338 expressing the glycosyltransferase (Gtf) gene from Pediococcus parvulus 2.6 (GTF), and naturally exopolysaccharide-producing Lactobacillus mucosae Dairy Product Culture Collection (DPC) 6426 (DPC 6426) compared with the non-ß-glucan-producing isogenic control strain Lactobacillus paracasei NFBC 338 (PNZ) and placebo (15% wt:vol trehalose). Second, we examined the effects on the gut microbiota of dietary administration of DPC 6426 compared with placebo. Probiotic Lactobacillus strains at 1 × 10(9) colony-forming units/d per animal were administered to apoE(-/-) mice fed a high-fat (60% fat)/high-cholesterol (2% wt:wt) diet for 12 wk. At the end of the study, aortic plaque development and serum, liver, and fecal variables involved in lipid metabolism were analyzed, and culture-independent microbial analyses of cecal content were performed. RESULTS: Total cholesterol was reduced in serum (P < 0.001; ∼33-50%) and liver (P < 0.05; ∼30%) and serum triglyceride concentrations were reduced (P < 0.05; ∼15-25%) in mice supplemented with GTF or DPC 6426 compared with the PNZ or placebo group, respectively. In addition, dietary intervention with GTF led to increased amounts of fecal cholesterol excretion (P < 0.05) compared with all other groups. Compositional sequencing of the gut microbiota revealed a greater prevalence of Porphyromonadaceae (P = 0.001) and Prevotellaceae (P = 0.001) in the DPC 6426 group and lower proportions of Clostridiaceae (P < 0.05), Peptococcaceae (P < 0.001), and Staphylococcaceae (P < 0.01) compared with the placebo group. CONCLUSION: Ingestion of exopolysaccharide-producing lactobacilli resulted in seemingly favorable improvements in lipid metabolism, which were associated with changes in the gut microbiota of mice.


Asunto(s)
Colesterol/sangre , Glicosiltransferasas/metabolismo , Lactobacillus/metabolismo , Metabolismo de los Lípidos , Microbiota , Probióticos/administración & dosificación , Animales , Apolipoproteínas E/genética , Aterosclerosis/prevención & control , Dieta , Suplementos Dietéticos , Modelos Animales de Enfermedad , Heces/microbiología , Tracto Gastrointestinal/microbiología , Regulación Enzimológica de la Expresión Génica , Glicosiltransferasas/genética , Lactobacillus/genética , Hígado/metabolismo , Ratones , Ratones Noqueados , Pediococcus/enzimología , Triglicéridos/sangre , Molécula 1 de Adhesión Celular Vascular/sangre , beta-Glucanos/sangre
10.
Adv Exp Med Biol ; 817: 221-39, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24997036

RESUMEN

We recently coined the phrase 'psychobiotics' to describe an emerging class of probiotics of relevance to psychiatry [Dinan et al., Biol Psychiatry 2013;74(10):720-726]. Such "mind-altering" probiotics may act via their ability to produce various biologically active compounds, such as peptides and mediators normally associated with mammalian neurotransmission. Several molecules with neuroactive functions such as gamma-aminobutyric acid (GABA), serotonin, catecholamines and acetylcholine have been reported to be microbially-derived, many of which have been isolated from bacteria within the human gut. Secreted neurotransmitters from bacteria in the intestinal lumen may induce epithelial cells to release molecules that in turn modulate neural signalling within the enteric nervous system and consequently signal brain function and behaviour of the host. Consequently, neurochemical containing/producing probiotic bacteria may be viewed as delivery vehicles for neuroactive compounds and as such, probiotic bacteria may possibly have the potential as a therapeutic strategy in the prevention and/or treatment of certain neurological and neurophysiological conditions.


Asunto(s)
Bacterias/metabolismo , Neurotransmisores/biosíntesis , Probióticos/metabolismo , Acetilcolina/biosíntesis , Animales , Catecolaminas/biosíntesis , Tracto Gastrointestinal/microbiología , Humanos , Serotonina/biosíntesis , Ácido gamma-Aminobutírico/biosíntesis
11.
Br J Nutr ; 111(11): 1905-17, 2014 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-24555449

RESUMEN

Different dietary fat and energy subtypes have an impact on both the metabolic health and the intestinal microbiota population of the host. The present study assessed the impact of dietary fat quality, with a focus on dietary fatty acid compositions of varying saturation, on the metabolic health status and the intestinal microbiota composition of the host. C57BL/6J mice (n 9-10 mice per group) were fed high-fat (HF) diets containing either (1) palm oil, (2) olive oil, (3) safflower oil or (4) flaxseed/fish oil for 16 weeks and compared with mice fed low-fat (LF) diets supplemented with either high maize starch or high sucrose. Tissue fatty acid compositions were assessed by GLC, and the impact of the diet on host intestinal microbiota populations was investigated using high-throughput 16S rRNA sequencing. Compositional sequencing analysis revealed that dietary palm oil supplementation resulted in significantly lower populations of Bacteroidetes at the phylum level compared with dietary olive oil supplementation (P< 0·05). Dietary supplementation with olive oil was associated with an increase in the population of the family Bacteroidaceae compared with dietary supplementation of palm oil, flaxseed/fish oil and high sucrose (P< 0·05). Ingestion of the HF-flaxseed/fish oil diet for 16 weeks led to significantly increased tissue concentrations of EPA, docosapentaenoic acid and DHA compared with ingestion of all the other diets (P< 0·05); furthermore, the diet significantly increased the intestinal population of Bifidobacterium at the genus level compared with the LF-high-maize starch diet (P< 0·05). These data indicate that both the quantity and quality of fat have an impact on host physiology with further downstream alterations to the intestinal microbiota population, with a HF diet supplemented with flaxseed/fish oil positively shaping the host microbial ecosystem.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Ácidos Grasos/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Animales , Bacteroidetes/efectos de los fármacos , Bacteroidetes/aislamiento & purificación , Dieta Alta en Grasa , Ácido Eicosapentaenoico/análisis , Ácidos Grasos Insaturados/análisis , Aceites de Pescado/administración & dosificación , Intestinos/microbiología , Aceite de Linaza/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Aceite de Oliva/administración & dosificación , Aceite de Palma , Aceites de Plantas/administración & dosificación , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN
12.
Antimicrob Agents Chemother ; 56(11): 5811-20, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22948872

RESUMEN

The infant gut microbiota undergoes dramatic changes during the first 2 years of life. The acquisition and development of this population can be influenced by numerous factors, and antibiotic treatment has been suggested as one of the most significant. Despite this, however, there have been relatively few studies which have investigated the short-term recovery of the infant gut microbiota following antibiotic treatment. The aim of this study was to use high-throughput sequencing (employing both 16S rRNA and rpoB-specific primers) and quantitative PCR to compare the gut microbiota of nine infants who underwent parenteral antibiotic treatment with ampicillin and gentamicin (within 48 h of birth), 4 and 8 weeks after the conclusion of treatment, relative to that of nine matched healthy controls. The investigation revealed that the gut microbiota of the antibiotic-treated infants had significantly higher proportions of Proteobacteria (P = 0.0049) and significantly lower proportions of Actinobacteria (P = 0.00001) (and the associated genus Bifidobacterium [P = 0.0132]) as well as the genus Lactobacillus (P = 0.0182) than the untreated controls 4 weeks after the cessation of treatment. By week 8, the Proteobacteria levels remained significantly higher in the treated infants (P = 0.0049), but the Actinobacteria, Bifidobacterium, and Lactobacillus levels had recovered and were similar to those in the control samples. Despite this recovery of total Bifidobacterium numbers, rpoB-targeted pyrosequencing revealed that the number of different Bifidobacterium species present in the antibiotic-treated infants was reduced. It is thus apparent that the combined use of ampicillin and gentamicin in early life can have significant effects on the evolution of the infant gut microbiota, the long-term health implications of which remain unknown.


Asunto(s)
Actinobacteria/efectos de los fármacos , Bifidobacterium/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Lactobacillus/efectos de los fármacos , Metagenoma/efectos de los fármacos , Proteobacteria/efectos de los fármacos , ARN Ribosómico 16S/genética , Actinobacteria/genética , Ampicilina/efectos adversos , Antibacterianos/efectos adversos , Bifidobacterium/genética , Recuento de Colonia Microbiana , Cartilla de ADN , Heces/microbiología , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Gentamicinas/efectos adversos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Infusiones Parenterales , Lactobacillus/genética , Masculino , Reacción en Cadena de la Polimerasa , Proteobacteria/genética , Recuperación de la Función/fisiología
13.
Am J Clin Nutr ; 95(5): 1278-87, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22492373

RESUMEN

BACKGROUND: We previously showed that microbial metabolism in the gut influences the composition of bioactive fatty acids in host adipose tissue. OBJECTIVE: This study compared the effect of dietary supplementation for 8 wk with human-derived Bifidobacterium breve strains on fat distribution and composition and the composition of the gut microbiota in mice. METHODS: C57BL/6 mice (n = 8 per group) received B. breve DPC 6330 or B. breve NCIMB 702258 (10(9) microorganisms) daily for 8 wk or no supplement (controls). Tissue fatty acid composition was assessed by gas-liquid chromatography while 16S rRNA pyrosequencing was used to investigate microbiota composition. RESULTS: Visceral fat mass and brain stearic acid, arachidonic acid, and DHA were higher in mice supplemented with B. breve NCIMB 702258 than in mice in the other 2 groups (P < 0.05). In addition, both B. breve DPC 6330 and B. breve NCIMB 702258 supplementation resulted in higher propionate concentrations in the cecum than did no supplementation (P < 0.05). Compositional sequencing of the gut microbiota showed a tendency for greater proportions of Clostridiaceae (25%, 12%, and 18%; P = 0.08) and lower proportions of Eubacteriaceae (3%, 12%, and 13%; P = 0.06) in mice supplemented with B. breve DPC 6330 than in mice supplemented with B. breve NCIMB 702258 and unsupplemented controls, respectively. CONCLUSION: The response of fatty acid metabolism to administration of bifidobacteria is strain-dependent, and strain-strain differences are important factors that influence modulation of the gut microbial community by ingested microorganisms.


Asunto(s)
Bifidobacterium/clasificación , Encéfalo/metabolismo , Suplementos Dietéticos , Ácidos Grasos/química , Tracto Gastrointestinal/microbiología , Metagenoma , Administración Oral , Animales , Cromatografía de Gases , Ácidos Grasos/análisis , Heces/microbiología , Tracto Gastrointestinal/metabolismo , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Probióticos/administración & dosificación
14.
Microbiology (Reading) ; 157(Pt 2): 609-615, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21178166

RESUMEN

We have previously demonstrated that oral administration of a metabolically active Bifidobacterium breve strain, with ability to form cis-9, trans-11 conjugated linoleic acid (CLA), resulted in modulation of the fatty acid composition of the host, including significantly elevated concentrations of c9, t11 CLA and omega-3 (n-3) fatty acids in liver and adipose tissue. In this study, we investigated whether a recombinant lactobacillus expressing linoleic acid isomerase (responsible for production of t10, c12 CLA) from Propionibacterium acnes (PAI) could influence the fatty acid composition of different tissues in a mouse model. Linoleic-acid-supplemented diets (2 %, w/w) were fed in combination with either a recombinant t10, c12 CLA-producing Lactobacillus paracasei NFBC 338 (Lb338), or an isogenic (vector-containing) control strain, to BALB/c mice for 8 weeks. A third group of mice received linoleic acid alone (2 %, w/w). Tissue fatty acid composition was assessed by GLC at the end of the trial. Ingestion of the strain expressing linoleic acid isomerase was associated with a 4-fold increase (P<0.001) in t10, c12 CLA in adipose tissues of the mice when compared with mice that received the isogenic non-CLA-producing strain. The livers of the mice that received the recombinant CLA-producing Lb338 also contained a 2.5-fold (albeit not significantly) higher concentration of t10, c12 CLA, compared to the control group. These data demonstrate that a single gene (encoding linoleic acid isomerase) expressed in an intestinal microbe can influence the fatty acid composition of host fat.


Asunto(s)
Tejido Adiposo/química , Bifidobacterium/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Ácido Linoleico/administración & dosificación , Propionibacterium acnes/enzimología , Animales , Bifidobacterium/genética , Dieta , Ácidos Grasos/análisis , Heces/microbiología , Tránsito Gastrointestinal , Oxidorreductasas Intramoleculares/genética , Lactobacillus/genética , Lactobacillus/metabolismo , Ácidos Linoleicos Conjugados/metabolismo , Hígado/química , Masculino , Ratones , Ratones Endogámicos BALB C , Probióticos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
15.
Int J Microbiol ; 20112011.
Artículo en Inglés | MEDLINE | ID: mdl-20811542

RESUMEN

We investigated the impact of parenteral antibiotic treatment in the early neonatal period on the evolution of bifidobacteria in the newborn. Nine babies treated with intravenous ampicillin/gentamicin in the first week of life and nine controls (no antibiotic treatment) were studied. Denaturing gradient gel electrophoresis was used to investigate the composition of Bifidobacterium in stool samples taken at four and eight weeks. Bifidobacteria were detected in all control infants at both four and eight weeks, while only six of nine antibiotic-treated infants had detectable bifidobacteria at four weeks and eight of nine at eight weeks. Moreover, stool samples of controls showed greater diversity of Bifidobacterium spp. compared with antibiotic-treated infants. In conclusion, short-term parenteral antibiotic treatment of neonates causes a disturbance in the expected colonization pattern of bifidobacteria in the first months of life. Further studies are required to probiotic determine if supplementation is necessary in this patient group.

16.
Curr Opin Biotechnol ; 21(2): 149-56, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20434324

RESUMEN

The neonatal period is crucial for intestinal colonisation, and the composition of this ecosystem in early life is influenced by such factors as mode of birth, environment, diet and antibiotics. The intestinal microbiota contributes to protection against pathogens, maturation of the immune system and metabolic welfare of the host, but under some circumstances can contribute to the pathogenesis of certain diseases. Because colonisation with non-pathogenic microbiota is important for infant health and may affect health in later life, it is important to understand how the composition of this microbial organ is established and by which dietary means (e.g. supplementation with prebiotics/probiotics/food ingredients) it can be programmed in order to achieve an ecosystem that is valuable for the host.


Asunto(s)
Dieta , Ecosistema , Microbiología de Alimentos , Intestinos/microbiología , Humanos , Recién Nacido
17.
Nutr Rev ; 68(5): 280-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20500789

RESUMEN

Omega-6 (n-6) and omega-3 (n-3) polyunsaturated fatty acids (PUFA) are precursors of potent lipid mediators, termed eicosanoids, which play an important role in the regulation of inflammation. Eicosanoids derived from n-6 PUFAs (e.g., arachidonic acid) have proinflammatory and immunoactive functions, whereas eicosanoids derived from n-3 PUFAs [e.g., eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)] have anti-inflammatory properties, traditionally attributed to their ability to inhibit the formation of n-6 PUFA-derived eicosanoids. While the typical Western diet has a much greater ratio of n-6 PUFAs compared with n-3 PUFAs, research has shown that by increasing the ratio of n-3 to n-6 fatty acids in the diet, and consequently favoring the production of EPA in the body, or by increasing the dietary intake of EPA and DHA through consumption of fatty fish or fish-oil supplements, reductions may be achieved in the incidence of many chronic diseases that involve inflammatory processes; most notably, these include cardiovascular diseases, inflammatory bowel disease (IBD), cancer, and rheumatoid arthritis, but psychiatric and neurodegenerative illnesses are other examples.


Asunto(s)
Ácidos Grasos Omega-3/administración & dosificación , Peces , Inflamación/prevención & control , Alimentos Marinos/análisis , Animales , Enfermedad Crónica/prevención & control , Ácidos Grasos Omega-3/análisis , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/administración & dosificación , Ácidos Grasos Omega-6/análisis , Ácidos Grasos Omega-6/metabolismo , Humanos , Inflamación/metabolismo , Valor Nutritivo
18.
Lipids ; 45(5): 429-36, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20405232

RESUMEN

Recently, we reported that administration of Bifidobacteria resulted in increased concentrations of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in murine adipose tissue [1]. The objective of this study was to assess the impact of co-administration of Bifidobacterium breve NCIMB 702258 and the substrate for EPA, alpha-linolenic acid, on host fatty acid composition. alpha-Linolenic acid-supplemented diets (1%, wt/wt) were fed to mice (n = 8), with or without B. breve NCIMB 702258 (daily dose of 10(9) microorganisms) for 8 weeks. Two further groups received either supplement of B. breve alone or unsupplemented diet. Tissue fatty acid composition was assessed by gas liquid chromatography. Dietary supplementation of alpha-linolenic acid resulted in higher (P < 0.05) alpha-linolenic acid and EPA concentrations in liver and adipose tissue and lower (P < 0.05) arachidonic acid in liver, adipose tissue and brain compared with mice that did not receive alpha-linolenic acid. Supplementation with B. breve NCIMB 702258 in combination with alpha-linolenic acid resulted in elevated (P < 0.05) liver EPA concentrations compared with alpha-linolenic acid supplementation alone. Furthermore, the former group had higher (P < 0.05) DHA in brain compared with the latter group. These results suggest a role for interactions between fatty acids and commensals in the gastrointestinal tract. This interaction between administered microbes and fatty acids could result in a highly effective nutritional approach to the therapy of a variety of inflammatory and neurodegenerative conditions.


Asunto(s)
Bifidobacterium , Ácidos Grasos/química , Ácido alfa-Linolénico/química , Administración Oral , Animales , Química Encefálica , Heces/microbiología , Femenino , Hígado/química , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Ácido alfa-Linolénico/administración & dosificación , Ácido alfa-Linolénico/metabolismo
19.
Am J Clin Nutr ; 89(5): 1393-401, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19357220

RESUMEN

BACKGROUND: Recent reports suggest that the metabolic activity of the gut microbiota may contribute to the pathogenesis of obesity and hepatic steatosis. OBJECTIVE: The objective was to determine whether the fat composition of host tissues might be influenced by oral administration of commensal bifidobacteria previously shown by us to produce bioactive isomers of conjugated linoleic acid (CLA). DESIGN: Murine trials were conducted in which linoleic acid-supplemented diets were fed with or without Bifidobacterium breve NCIMB 702258 (daily dose of 10(9) microorganisms) to healthy BALB/c mice and to severe combined immunodeficient mice for 8-10 wk. To ensure that the observations were not peculiar to mice, a similar trial was conducted in weanling pigs over 21 d. Tissue fatty acid composition was assessed by gas-liquid chromatography. RESULTS: In comparison with controls, there was an increase in cis-9, trans-11 CLA in the livers of the mice and pigs after feeding with linoleic acid in combination with B. breve NCIMB 702258 (P < 0.05). In addition, an altered profile of polyunsaturated fatty acid composition was observed, including higher concentrations of the omega-3 (n-3) fatty acids eicosapentaenoic acid and docosahexaenoic acid in adipose tissue (P < 0.05). These changes were associated with reductions in the proinflammatory cytokines tumor necrosis factor-alpha and interferon-gamma (P < 0.05). CONCLUSIONS: These results are consistent with the concept that the metabolome is a composite of host and microbe metabolic activity and that the influence of the microbiota on host fatty acid composition can be manipulated by oral administration of CLA-producing microorganisms.


Asunto(s)
Tejido Adiposo/metabolismo , Tejido Adiposo/microbiología , Bifidobacterium/metabolismo , Ácidos Grasos/metabolismo , Hígado/metabolismo , Hígado/microbiología , Alimentación Animal , Animales , Heces/microbiología , Linfocitos/inmunología , Linfocitos/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Porcinos
20.
ISME J ; 2(1): 83-91, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18059489

RESUMEN

The overall purpose of this study was to examine the lactobacilli and bifidobacteria microbiota in the human ileum at a very early stage of life. Ileostomy effluents from two infants, taken at different time points, were plated on Lactobacillus selective agar and cys-MRS containing mupirocin to select for bifidobacteria. In one case, a stool sample following ileostomy reversal was subsequently analyzed microbiologically. Pulse-field gel electrophoresis and 16S rRNA sequencing was used to investigate the cultivable population of bifidobacteria and lactobacilli and denaturing gradient gel electrophoresis (DGGE) to examine the non-cultivable population. The probiotic strain, Lactobacillus paracasei NFBC 338, was recovered at both time points from one of the infants and dominated in the small intestine for a period of over 3 weeks. Moreover, the probiotic strain, B. animalis subsp. lactis Bb12, was obtained from the other infant. This study shows the presence of two known probiotic strains in the upper intestinal tract at an early stage of human life and thus provides some evidence for their ability to colonize the infant small intestine.


Asunto(s)
Bifidobacterium/aislamiento & purificación , Íleon/microbiología , Lactobacillus/aislamiento & purificación , Adhesión Bacteriana , Bifidobacterium/genética , Ciego , Dermatoglifia del ADN , ADN Bacteriano/genética , ADN Ribosómico/genética , Electroforesis en Gel de Campo Pulsado , Heces/microbiología , Humanos , Ileostomía , Lactante , Recién Nacido , Lactobacillus/genética , ARN Ribosómico 16S/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...