Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Cancer ; 117(9): 1336-1340, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-28873082

RESUMEN

BACKGROUND: Activation of free fatty acid receptor 2 (FFAR2) by microbiota-derived metabolites (e.g., propionate) reduces leukaemic cell proliferation in vitro. This study aims to test whether Ffar2 expression per se also influences leukaemia cell growth in vivo. METHODS: Bcr-Abl-expressing BaF cells were used as a leukaemia model and the role of Ffar2 was evaluated in Balb/c mice after lentiviral shRNA transduction. RESULTS: Our data formally establish that reduced leukaemic cell proliferation is associated with increased Ffar2 expression in vivo and in vitro. Going beyond association, we point out that decreasing Ffar2 expression fosters cancer cell growth in vitro and in vivo. CONCLUSIONS: Our data demonstrate the role of Ffar2 in the control of leukaemic cell proliferation in vivo and indicate that a modulation of Ffar2 expression through nutritional tools or pharmacological agents may constitute an attractive therapeutic approach to tackle leukaemia progression in humans.


Asunto(s)
Proliferación Celular , Leucemia Experimental/patología , Receptores Acoplados a Proteínas G/fisiología , Animales , Apoptosis , Biomarcadores de Tumor/metabolismo , Femenino , Leucemia Experimental/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Tumorales Cultivadas
2.
Clin Nutr ; 34(3): 501-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24969566

RESUMEN

BACKGROUND & AIMS: Inulin-type fructans (ITF) prebiotics promote changes in the composition and activity of the gut microbiota. The aim of this study was to determine variations on fecal short chain fatty acids (SCFA) concentration in obese women treated with ITF and to explore associations between Bifidobacterium species, SCFA and host biological markers of metabolism. METHODS: Samples were obtained in a randomized, double blind, parallel, placebo-controlled trial, with 30 obese women randomly assigned to groups that received either 16 g/day ITF (n = 15) or maltodextrin (n = 15) for 3 months. The qualitative and quantitative analysis of Bifidobacterium spp. was performed in feces by PCR-DGGE and q-PCR, and SCFA profile was analyzed by gas chromatography. Spearman correlation analysis was performed between the different variables analyzed. RESULTS: The species Bifidobacterium longum, Bifidobacterium pseudocatenulatum and Bifidobacterium adolescentis were significantly increased at the end of the treatment in the prebiotic group (p < 0.01) with being B. longum negatively correlated with serum lipopolysaccharide (LPS) endotoxin (p < 0.01). Total SCFA, acetate and propionate, that positively correlated with BMI, fasting insulinemia and homeostasis model assessment (HOMA) (p < 0.05), were significantly lower in prebiotic than in placebo group after the treatment period. CONCLUSIONS: ITF consumption selectively modulates Bifidobacterium spp. and decreases fecal SCFA concentration in obese women. ITF could lessen metabolic risk factors associated with higher fecal SCFA concentration in obese individuals.


Asunto(s)
Bifidobacterium/aislamiento & purificación , Ácidos Grasos Volátiles/análisis , Intestinos/microbiología , Inulina/administración & dosificación , Obesidad/tratamiento farmacológico , Adolescente , Adulto , Anciano , Biomarcadores/sangre , Índice de Masa Corporal , ADN Bacteriano/genética , Método Doble Ciego , Heces/química , Heces/microbiología , Femenino , Humanos , Intestinos/efectos de los fármacos , Persona de Mediana Edad , Prebióticos/administración & dosificación , Adulto Joven
3.
Obesity (Silver Spring) ; 22(7): 1653-61, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24585705

RESUMEN

OBJECTIVES: To investigate whether inulin-type fructan (ITF) prebiotics could counteract the thiazolidinedione (TZD, PPARγ activator) induced-fat mass gain, without affecting its beneficial effect on glucose homeostasis, in high-fat (HF) diet fed mice. METHODS: Male C57bl6/J mice were fed a HF diet alone or supplemented with ITF prebiotics (0.2 g/day × mouse) or TZD (30 mg pioglitazone (PIO)/kg body weight × day) or both during 4 weeks. An insulin tolerance test was performed after 3 weeks of treatment. RESULTS: As expected, PIO improved glucose homeostasis and increased adiponectinaemia. Furthermore, it induced an over-expression of several PPARγ target genes in white adipose tissues. ITF prebiotics modulated the PIO-induced PPARγ activation in a tissue-dependent manner. The co-treatment with ITF prebiotics and PIO maintained the beneficial impact of TZD on glucose homeostasis and adiponectinaemia. Moreover, the combination of both treatments reduced fat mass accumulation, circulating lipids and hepatic triglyceride content, suggesting an overall improvement of metabolism. Finally, the co-treatment favored induction of white-to-brown fat conversion in subcutaneous adipose tissue, thereby leading to the development of brite adipocytes that could increase the oxidative capacity of the tissue. CONCLUSIONS: ITF prebiotics decrease adiposity and improve the metabolic response in HF fed mice treated with TZD.


Asunto(s)
Adiposidad/efectos de los fármacos , Glucemia/metabolismo , Dieta Alta en Grasa , Homeostasis/efectos de los fármacos , Prebióticos , Tiazolidinedionas/farmacología , Adipocitos/efectos de los fármacos , Adiponectina/sangre , Animales , Insulina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , PPAR gamma/sangre , Pioglitazona , Grasa Subcutánea/efectos de los fármacos
4.
Lipids ; 49(4): 397-402, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24473752

RESUMEN

The aim of this human study was to assess the influence of prebiotic-induced gut microbiota modulation on PUFA-derived bacterial metabolites production. Therefore, we analyzed the circulating fatty acid profile including CLA/CLnA in obese women treated during 3 months with inulin-type fructan prebiotics. In these patients, we had already determined gut microbiota composition by phylogenetic microarray and qPCR analysis of 16S rDNA. Some PUFA-derived bacterial metabolites were detected in the serum of obese patients. Despite the prebiotic-induced modulation of gut microbiota, including changes in CLA/CLnA-producing bacteria, the treatment did not impact significantly on the circulating level of these metabolites. However, some PUFA-derived bacterial metabolites were positively correlated with specific fecal bacteria (Bifidobacterium spp., Eubacterium ventriosum and Lactobacillus spp.) and inversely correlated with serum cholesterol (total, LDL, HDL). These correlations suggest a potential beneficial effect of some of these metabolites but this remains to be confirmed by further investigation.


Asunto(s)
Ácidos Grasos Insaturados/sangre , Síndrome Metabólico/sangre , Microbiota , Obesidad/sangre , Adulto , Anciano , Bifidobacterium/metabolismo , Ácidos Grasos Insaturados/aislamiento & purificación , Heces/microbiología , Femenino , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Humanos , Síndrome Metabólico/microbiología , Síndrome Metabólico/patología , Metagenoma , Obesidad/microbiología , Obesidad/patología , Filogenia , Prebióticos , ARN Ribosómico 16S/genética
5.
Br J Nutr ; 110(6): 998-1011, 2013 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23507010

RESUMEN

In vitro experiments have shown that isolated human gut bacteria are able to metabolise PUFA into conjugated PUFA like conjugated linoleic acids (CLA). The hypothesis of the present paper was that high-fat (HF) diet feeding and supplementation with fermentable carbohydrates that have prebiotic properties modulate the in vivo production of CLA by the mouse gut microbiota. Mice were treated for 4 weeks as follows: control (CT) groups were fed a standard diet; HF groups were fed a HF diet rich in linoleic acid (18 : 2n-6); the third groups were fed with the HF diet supplemented with either inulin-type fructans (HF-ITF) or arabinoxylans (HF-Ax). HF diet feeding increased rumenic acid (cis-9,trans-11-18 : 2 CLA) content both in the caecal and liver tissues compared with the CT groups. ITF supplementation had no major effect compared with the HF diet whereas Ax supplementation increased further rumenic acid (cis-9,trans-11-18 : 2 CLA) in the caecal tissue. These differences between both prebiotics may be linked to the high fat-binding capacity of Ax that provides more substrates for bacterial metabolism and to differential modulation of the gut microbiota (specific increase in Roseburia spp. in HF-Ax v. HF). In conclusion, these experiments supply the proof of concept that the mouse gut microbiota produces CLA in vivo, with consequences on the level of CLA in the caecal and liver tissues. We postulate that the CLA-producing bacteria could be a mediator to consider in the metabolic effects of both HF diet feeding and prebiotic supplementation.


Asunto(s)
Bacterias/efectos de los fármacos , Carbohidratos/química , Grasas de la Dieta/farmacología , Intestinos/microbiología , Ácidos Linoleicos Conjugados/metabolismo , Animales , Metabolismo de los Hidratos de Carbono , Grasas de la Dieta/administración & dosificación , Ácidos Grasos Omega-6/metabolismo , Fermentación , Regulación Enzimológica de la Expresión Génica , Intestinos/enzimología , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Prebióticos , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo
6.
Trends Pharmacol Sci ; 34(4): 226-32, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23489932

RESUMEN

Research interest in free fatty acid-binding receptors has been growing during the past decade, with an aim to better understand the modulation of host physiology in response to nutrition. G-protein-coupled receptor 43 (GPR43), also called free fatty acid receptor 2 (FFA2/FFAR2), binds short-chain fatty acids (SCFAs) produced by the microbial fermentation of carbohydrates and has shown promising therapeutic potential. This review presents current knowledge regarding the pharmacological properties of GPR43 and addresses its functions in selected organs (adipose tissue, intestine and immune cells). Furthermore, the demonstration of GPR43 involvement in several pathological conditions such as obesity, inflammatory disease, and cancer suggests new fields of interest related to this receptor. Finally, GPR43 could be a key player in gut microbes-host crosstalk, although further research is needed to clearly evaluate its role in the management of host health by nutrients or treatments targeting the gut microbiota.


Asunto(s)
Receptores Acoplados a Proteínas G/fisiología , Animales , Humanos , Terapia Molecular Dirigida , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética
7.
Nutr Metab (Lond) ; 10(1): 11, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23327542

RESUMEN

BACKGROUND: GPR43 is a G-protein-coupled receptor that participates in adipocyte differentiation in mice and is over-expressed in adipose tissue of obese mice. The aim of this study was to investigate the implication of GPR43 in adipogenesis in humans and to determine the influence of obesity on its expression in human adipose tissue. FINDINGS: Preadipocytes were isolated from human omental adipose tissue and cultured during 13 days. One PPARγ agonist (troglitazone) and three GPR43 agonists (two physiological and one synthetic) were tested for their ability to induce differentiation. After 13 days, the three GPR43 agonists had no impact on aP2 expression, a marker of adipocyte differentiation, whereas troglitazone led to a huge over-expression of aP2 in these cells but tended to decrease GPR43 expression (p=0.06).GPR43 and inflammatory markers expression was also quantified in omental adipose tissue from lean and obese individuals. GPR43 expression in total adipose tissue was similar between obese patients and lean subjects and did not correlate with aP2 expression. In contrast, GPR43 expression positively correlated with TNFα mRNA. CONCLUSIONS: Our results suggest the absence of relationship between GPR43 and adipocyte differentiation in humans, unlike what was observed in mice. Furthermore, GPR43 expression is not increased in adipose tissue from obese subjects but could be related to TNFα-related inflammatory processes.

8.
Mol Nutr Food Res ; 57(2): 347-59, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23203768

RESUMEN

SCOPE: Recent data suggest that gut microbiota contributes to the regulation of host lipid metabolism. We report how fermentable dietary fructo-oligosaccharides (FOS) control hepatic steatosis induced by n-3 PUFA depletion, which leads to hepatic alterations similar to those observed in non-alcoholic fatty liver disease patients. METHODS AND RESULTS: C57Bl/6J mice fed an n-3 PUFA-depleted diet for 3 months were supplemented with FOS during the last 10 days of treatment. FOS-treated mice exhibited higher caecal Bifidobacterium spp. and lower Roseburia spp. content. Microarray analysis of hepatic mRNA revealed that FOS supplementation reduced hepatic triglyceride accumulation through a proliferator-activated receptor α-stimulation of fatty acid oxidation and lessened cholesterol accumulation by inhibiting sterol regulatory element binding protein 2-dependent cholesterol synthesis. Cultured precision-cut liver slices confirmed the inhibition of fatty acid oxidation. FOS effects were related to a decreased hepatic micro-RNA33 expression and to an increased colonic glucagon-like peptide 1 production. CONCLUSIONS: The changes in gut microbiota composition by n-3 PUFA-depletion and prebiotics modulate hepatic steatosis by changing gene expression in the liver, a phenomenon that could implicate micro-RNA and gut-derived hormones. Our data underline the advantage of targeting the gut microbiota by colonic nutrients in the management of liver disease.


Asunto(s)
Colesterol/biosíntesis , Suplementos Dietéticos , Ácidos Grasos Omega-3/metabolismo , Hígado Graso/patología , Prebióticos , Animales , Bifidobacterium/crecimiento & desarrollo , Ingestión de Energía , Hígado Graso/metabolismo , Tracto Gastrointestinal/microbiología , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Metagenoma/fisiología , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , Oligosacáridos/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , PPAR alfa/genética , PPAR alfa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Gut ; 62(8): 1112-21, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23135760

RESUMEN

OBJECTIVE: To highlight the contribution of the gut microbiota to the modulation of host metabolism by dietary inulin-type fructans (ITF prebiotics) in obese women. METHODS: A double blind, placebo controlled, intervention study was performed with 30 obese women treated with ITF prebiotics (inulin/oligofructose 50/50 mix; n=15) or placebo (maltodextrin; n=15) for 3 months (16 g/day). Blood, faeces and urine sampling, oral glucose tolerance test, homeostasis model assessment and impedancemetry were performed before and after treatment. The gut microbial composition in faeces was analysed by phylogenetic microarray and qPCR analysis of 16S rDNA. Plasma and urine metabolic profiles were analysed by 1H-NMR spectroscopy. RESULTS: Treatment with ITF prebiotics, but not the placebo, led to an increase in Bifidobacterium and Faecalibacterium prausnitzii; both bacteria negatively correlated with serum lipopolysaccharide levels. ITF prebiotics also decreased Bacteroides intestinalis, Bacteroides vulgatus and Propionibacterium, an effect associated with a slight decrease in fat mass and with plasma lactate and phosphatidylcholine levels. No clear treatment clustering could be detected for gut microbial analysis or plasma and urine metabolomic profile analyses. However, ITF prebiotics led to subtle changes in the gut microbiota that may importantly impact on several key metabolites implicated in obesity and/or diabetes. CONCLUSIONS: ITF prebiotics selectively changed the gut microbiota composition in obese women, leading to modest changes in host metabolism, as suggested by the correlation between some bacterial species and metabolic endotoxaemia or metabolomic signatures.


Asunto(s)
Inulina/farmacología , Obesidad/microbiología , Prebióticos , Adolescente , Adulto , Antropometría , Bacterias/clasificación , Bacterias/efectos de los fármacos , Bacterias/aislamiento & purificación , Técnicas de Tipificación Bacteriana , Índice de Masa Corporal , Método Doble Ciego , Heces/microbiología , Femenino , Humanos , Metaboloma/efectos de los fármacos , Metagenoma/efectos de los fármacos , Persona de Mediana Edad , Obesidad/metabolismo , Obesidad/fisiopatología , Adulto Joven
10.
PLoS One ; 7(6): e37971, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761662

RESUMEN

The gut microbiota has recently been proposed as a novel component in the regulation of host homeostasis and immunity. We have assessed for the first time the role of the gut microbiota in a mouse model of leukemia (transplantation of BaF3 cells containing ectopic expression of Bcr-Abl), characterized at the final stage by a loss of fat mass, muscle atrophy, anorexia and inflammation. The gut microbial 16S rDNA analysis, using PCR-Denaturating Gradient Gel Electrophoresis and quantitative PCR, reveals a dysbiosis and a selective modulation of Lactobacillus spp. (decrease of L. reuteri and L. johnsonii/gasseri in favor of L. murinus/animalis) in the BaF3 mice compared to the controls. The restoration of Lactobacillus species by oral supplementation with L. reuteri 100-23 and L. gasseri 311476 reduced the expression of atrophy markers (Atrogin-1, MuRF1, LC3, Cathepsin L) in the gastrocnemius and in the tibialis, a phenomenon correlated with a decrease of inflammatory cytokines (interleukin-6, monocyte chemoattractant protein-1, interleukin-4, granulocyte colony-stimulating factor, quantified by multiplex immuno-assay). These positive effects are strain- and/or species-specific since L. acidophilus NCFM supplementation does not impact on muscle atrophy markers and systemic inflammation. Altogether, these results suggest that the gut microbiota could constitute a novel therapeutic target in the management of leukemia-associated inflammation and related disorders in the muscle.


Asunto(s)
Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Inflamación/prevención & control , Lactobacillus/fisiología , Leucemia Experimental/complicaciones , Atrofia Muscular/prevención & control , Enfermedad Aguda , Animales , Células Cultivadas , Suplementos Dietéticos , Femenino , Proteínas de Fusión bcr-abl/genética , Tracto Gastrointestinal/microbiología , Inflamación/etiología , Leucemia Experimental/genética , Leucemia Experimental/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/microbiología , Neoplasias Hepáticas/patología , Metagenoma , Ratones , Ratones Endogámicos BALB C , Atrofia Muscular/etiología , Células Precursoras de Linfocitos B/trasplante , Neoplasias del Bazo/metabolismo , Neoplasias del Bazo/microbiología , Neoplasias del Bazo/patología
11.
PLoS One ; 6(8): e23365, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21853118

RESUMEN

Patients with non-alcoholic fatty liver disease are characterised by a decreased n-3/n-6 polyunsaturated fatty acid (PUFA) ratio in hepatic phospholipids. The metabolic consequences of n-3 PUFA depletion in the liver are poorly understood. We have reproduced a drastic drop in n-3 PUFA among hepatic phospholipids by feeding C57Bl/6J mice for 3 months with an n-3 PUFA depleted diet (DEF) versus a control diet (CT), which only differed in the PUFA content. DEF mice exhibited hepatic insulin resistance (assessed by euglycemic-hyperinsulinemic clamp) and steatosis that was associated with a decrease in fatty acid oxidation and occurred despite a higher capacity for triglyceride secretion. Microarray and qPCR analysis of the liver tissue revealed higher expression of all the enzymes involved in lipogenesis in DEF mice compared to CT mice, as well as increased expression and activation of sterol regulatory element binding protein-1c (SREBP-1c). Our data suggest that the activation of the liver X receptor pathway is involved in the overexpression of SREBP-1c, and this phenomenon cannot be attributed to insulin or to endoplasmic reticulum stress responses. In conclusion, n-3 PUFA depletion in liver phospholipids leads to activation of SREBP-1c and lipogenesis, which contributes to hepatic steatosis.


Asunto(s)
Ácidos Grasos Omega-3/metabolismo , Hígado Graso/genética , Genoma/genética , Resistencia a la Insulina/genética , Hígado/metabolismo , Animales , Moduladores de Receptores de Cannabinoides/metabolismo , Colesterol/biosíntesis , Dieta , Estrés del Retículo Endoplásmico/genética , Hígado Graso/patología , Conducta Alimentaria , Regulación de la Expresión Génica , Metabolismo de los Lípidos/genética , Hígado/patología , Receptores X del Hígado , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Nucleares Huérfanos/metabolismo , Oxidación-Reducción , Fosfolípidos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo
12.
J Nutr Biochem ; 22(8): 712-22, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21115338

RESUMEN

Inulin-type fructans (ITF) are nondigestible/fermentable carbohydrates which are able - through the modification of the gut microbiota - to counteract high-fat (HF) diet-induced obesity, endotoxemia and related-metabolic alterations. However, their influence on adipose tissue metabolism has been poorly studied until now. The aim of this study was to assess the influence of ITF supplementation on adipose tissue metabolism, by focusing on a G protein-coupled receptor (GPR), GPR43, as a potential link between gut fermentation processes and white adipose tissue development. Male C57bl6/J mice were fed a standard diet or an HF diet without or with ITF (0.2 g/day per mouse) during 4 weeks. The HF diet induced an accumulation of large adipocytes, promoted peroxisome proliferator activated receptor gamma (PPARγ)-activated differentiation factors and led to a huge increase in GPR43 expression in the subcutaneous adipose tissue. All those effects were blunted by ITF treatment, which modulated the gut microbiota in favor of bifidobacteria at the expense of Roseburia spp. and of Clostridium cluster XIVa. The dietary modulation of GPR43 expression seems independent of endotoxemia, in view of data obtained in vivo (acute and chronic lipopolysaccharides treatment). In conclusion, ITF, which promote gut fermentation, paradoxically counteract GPR43 overexpression induced in the adipose tissue by an HF diet, a phenomenon that correlates with a beneficial effect on adiposity and with potential decrease in PPARγ-activated processes.


Asunto(s)
Adipogénesis , Inulina/farmacología , PPAR gamma/genética , Prebióticos/análisis , Receptores Acoplados a Proteínas G/genética , Tejido Adiposo/metabolismo , Animales , Peso Corporal , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/administración & dosificación , Electroforesis en Gel de Poliacrilamida , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Fructanos/farmacología , Regulación de la Expresión Génica , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/antagonistas & inhibidores
13.
J Nutr ; 140(3): 509-14, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20089787

RESUMEN

Magnesium (Mg) deficiency is a common nutritional disorder that is linked to an inflammatory state characterized by increased plasma acute phase protein and proinflammatory cytokine concentrations. Recent studies have shown that changes in the composition of gut microbiota composition participate in systemic inflammation. In this study, therefore, we assessed the potential role of gut microbiota in intestinal and systemic inflammation associated with Mg deficiency in mice. For this purpose, mice were fed a control or Mg-deficient diet (500 mg vs. 70 mg Mg/kg) for 4 or 21 d. Compared with the mice fed the control diet, mice fed the Mg-deficient diet for 4 d had a lower gut bifidobacteria content (-1.5 log), a 36-50% lower mRNA content of factors controlling gut barrier function in the ileum (zonula occludens-1, occludin, proglucagon), and a higher mRNA content (by approximately 2-fold) in the liver and/or intestine of tumor necrosis factor-alpha, interleukin-6, CCAAT/enhancer binding protein homologous protein, and activating transcription factor 4, reflecting inflammatory and cellular stress. In contrast, mice fed the Mg-deficient diet for 21 d had a higher cecal bifidobacteria content compared with the control group, a phenomenon accompanied by restoration of the intestinal barrier and the absence of inflammation. In conclusion, we show that Mg deficiency, independently of any other changes in nutrient intake, modulates the concentration of bifidobacteria in the gut, a phenomenon that may time-dependently affect inflammation and metabolic disorders in mice.


Asunto(s)
Bifidobacterium/fisiología , Colon/microbiología , Inflamación/metabolismo , Magnesio/metabolismo , Animales , Peso Corporal , Colon/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos Nutricionales
14.
Am J Clin Nutr ; 90(5): 1236-43, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19776140

RESUMEN

BACKGROUND: We have previously shown that gut microbial fermentation of prebiotics promotes satiety and lowers hunger and energy intake in humans. In rodents, these effects are associated with an increase in plasma gut peptide concentrations, which are involved in appetite regulation and glucose homeostasis. OBJECTIVE: Our aim was to examine the effects of prebiotic supplementation on satiety and related hormones during a test meal for human volunteers by using a noninvasive micromethod for blood sampling to measure plasma gut peptide concentrations. DESIGN: This study was a randomized, double-blind, parallel, placebo-controlled trial. A total of 10 healthy adults (5 men and 5 women) were randomly assigned to groups that received either 16 g prebiotics/d or 16 g dextrin maltose/d for 2 wk. Meal tolerance tests were performed in the morning to measure the following: hydrogen breath test, satiety, glucose homeostasis, and related hormone response. RESULTS: We show that the prebiotic treatment increased breath-hydrogen excretion (a marker of gut microbiota fermentation) by approximately 3-fold and lowered hunger rates. Prebiotics increased plasma glucagon-like peptide 1 and peptide YY concentrations, whereas postprandial plasma glucose responses decreased after the standardized meal. The areas under the curve for plasma glucagon-like peptide 1 and breath-hydrogen excretion measured after the meal (0-60 min) were significantly correlated (r = 0.85, P = 0.007). The glucose response was inversely correlated with the breath-hydrogen excretion areas under the curve (0-180 min; r = -0.73, P = 0.02). CONCLUSION: Prebiotic supplementation was associated with an increase in plasma gut peptide concentrations (glucagon-like peptide 1 and peptide YY), which may contribute in part to changes in appetite sensation and glucose excursion responses after a meal in healthy subjects.


Asunto(s)
Apetito/fisiología , Fibras de la Dieta/farmacología , Suplementos Dietéticos , Ingestión de Alimentos/fisiología , Incretinas/biosíntesis , Respuesta de Saciedad/fisiología , Adulto , Apetito/efectos de los fármacos , Glucemia/metabolismo , Pruebas Respiratorias , Método Doble Ciego , Femenino , Péptido 1 Similar al Glucagón/sangre , Humanos , Hidrógeno/análisis , Masculino , Polipéptido Pancreático/sangre , Péptido YY/sangre
15.
Biochem Biophys Res Commun ; 385(3): 351-6, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19463788

RESUMEN

The aim of this study was to investigate the role of Kupffer cell in glucose metabolism and hepatic insulin sensitivity in mice. Both phagocytic activity and secretory capacity of Kupffer cells were blunted 24h after GdCl3 administration. Glucose tolerance--evaluated following an oral glucose tolerance test (OGTT)--was higher in GdCl3-treated mice whereas fasting insulinemia and HOMA-IR index decreased. The improvement of glucose tolerance and hepatic insulin signalling pathway after inhibition of Kupffer cells was supported by a lower hepatic gluconeogenic enzyme expression and a higher phosphorylation of Akt upon insulin challenge. Moreover, fasting hyperglycemia, insulin resistance and impaired glucose tolerance--induced by high fat (HF) diet--were improved through chronic administration of GdCl3. Interestingly, the inhibition of Kupffer cell exerted antiobesity effects in HF-fed mice, and lowered hepatic steatosis. Therefore, strategies targeting Kupffer cell functions could be a promising approach to counteract obesity and related metabolic disorders.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Macrófagos del Hígado/fisiología , Hígado/metabolismo , Obesidad/metabolismo , Animales , Grasas de la Dieta/administración & dosificación , Gadolinio/farmacología , Gluconeogénesis , Prueba de Tolerancia a la Glucosa , Insulina/farmacología , Resistencia a la Insulina , Macrófagos del Hígado/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo
16.
Br J Nutr ; 102(3): 462-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19161640

RESUMEN

The mechanism, by which a high-fat (HF) diet could impair glucose metabolism, is not completely understood but could be related to inflammation, lipotoxicity and oxidative stress. Lipid peroxides have been proposed as key mediators of intracellular metabolic response. The purpose of the present study was to analyse, in mice fed with a HF diet, the possible association between obesity and glucose tolerance on the one hand, and between oxidative stress and lipid peroxidation on the other hand. The present results show that a HF diet (70 % energy as fat), v. a high-carbohydrate chow diet (control), increases body weight and fat mass development, and impairs glycaemia and insulinaemia within 4 weeks. It also promotes the expression of NADPH oxidase in the liver--signing both oxidative and inflammatory stress--but decreases thiobarbituric acid-reactive substances content in the liver as well as in epididymal, subcutaneous and visceral adipose tissues. HF diet, with elevated vitamin E content, induces high concentration of alpha-tocopherol in liver and adipose tissues, which contributes to the protection against lipid peroxidation. Thus, lipid peroxidation in key organs is not necessarily related to the development of metabolic disorders associated with diabetes and obesity.


Asunto(s)
Diabetes Mellitus/metabolismo , Grasas de la Dieta/efectos adversos , Peroxidación de Lípido/fisiología , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Animales , Antioxidantes/análisis , Biomarcadores/análisis , Diabetes Mellitus/inmunología , Suplementos Dietéticos , Ácidos Grasos/análisis , Inflamación , Resistencia a la Insulina , Lípidos/análisis , Hígado/química , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/análisis , NADPH Oxidasas/genética , Estrés Oxidativo , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sustancias Reactivas al Ácido Tiobarbitúrico/análisis , Vitamina E/administración & dosificación , Vitaminas/administración & dosificación , alfa-Tocoferol/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...