Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JHEP Rep ; 3(4): 100323, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34355155

RESUMEN

BACKGROUND & AIMS: Retrospective cross-sectional studies linked sarcopenia and myosteatosis with metabolic dysfunction-associated fatty liver disease (MAFLD). Here, we wanted to clarify the dynamic relationship between sarcopenia, myosteatosis, and MAFLD. METHODS: A cohort of 48 obese patients was randomised for a dietary intervention consisting of 16 g/day of inulin (prebiotic) or maltodextrin (placebo) supplementation. Before and after the intervention, we evaluated liver steatosis and stiffness with transient elastography (TE); we assessed skeletal muscle index (SMI) and skeletal muscle fat index (SMFI) (a surrogate for absolute fat content in muscle) using computed tomography (CT) and bioelectrical impedance analysis (BIA). RESULTS: At baseline, sarcopenia was uncommon in patients with MAFLD (4/48, 8.3%). SMFI was higher in patients with high liver stiffness than in those with low liver stiffness (640.6 ± 114.3 cm2/ Hounsfield unit [HU] vs. 507.9 ± 103.0 cm2/HU, p = 0.001). In multivariate analysis, SMFI was robustly associated with liver stiffness even when adjusted for multiple confounders (binary logistic regression, p <0.05). After intervention, patients with inulin supplementation lost weight, but this was not associated with a decrease in liver stiffness. Remarkably, upon intervention (being inulin or maltodextrin), patients who lowered their SMFI, but not those who increased SMI, had a 12.7% decrease in liver stiffness (before = 6.36 ± 2.15 vs. after = 5.55 ± 1.97 kPa, p = 0.04). CONCLUSIONS: Myosteatosis, but not sarcopenia, is strongly and independently associated with liver stiffness in obese patients with MAFLD. After intervention, patients in which the degree of myosteatosis decreased reduced their liver stiffness, irrespective of body weight loss or prebiotic treatment. The potential contribution of myosteatosis to liver disease progression should be investigated. CLINICAL TRIALS REGISTRATION NUMBER: NCT03852069. LAY SUMMARY: The fat content in skeletal muscles (or myosteatosis) is strongly associated with liver stiffness in obese patients with MAFLD. After a dietary intervention, patients in which the degree of myosteatosis decreased also reduced their liver stiffness. The potential contribution of myosteatosis to liver disease progression should be investigated.

2.
Nutrients ; 13(5)2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33925432

RESUMEN

A saffron extract has been found to be effective in the context of depression and anxiety, but its effect on sleep quality has not been investigating yet using objective approaches. For this purpose, a randomized double-blind controlled study was conducted in subjects presenting mild to moderate sleep disorder associated with anxiety. Sixty-six subjects were randomized and supplemented with a placebo (maltodextrin) or a saffron extract (15.5 mg per day) for 6 weeks. Actigraphy was used to collect objective data related to sleep quality at baseline, at the middle and at the end of the intervention. Sleep quality was also assessed by completion of the LSEQ and PSQI questionnaires and quality of life by completion of the SF-36 questionnaire. Six weeks of saffron supplementation led to an increased time in bed assessed by actigraphy, to an improved ease of getting to sleep evaluated by the LSEQ questionnaire and to an improved sleep quality, sleep latency, sleep duration, and global scores evaluated by the PSQI questionnaire, whereas those parameters were not modified by the placebo. In conclusion, those results suggest that a saffron extract could be a natural and safe nutritional strategy to improve sleep duration and quality.


Asunto(s)
Crocus , Extractos Vegetales/farmacología , Sueño/efectos de los fármacos , Adulto , Anciano , Bélgica , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Encuestas y Cuestionarios
3.
Clin Nutr ; 39(12): 3618-3628, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32340903

RESUMEN

BACKGROUND: The gut microbiota is altered in obesity and is strongly influenced by nutrients and xenobiotics. We have tested the impact of native inulin as prebiotic present in vegetables and added as a supplement on gut microbiota-related outcomes in obese patients. Metformin treatment was analyzed as a potential modulator of the response. METHODS: A randomized, single-blinded, multicentric, placebo-controlled trial was conducted in 150 obese patients who received 16 g/d native inulin versus maltodextrin, coupled to dietary advice to consume inulin-rich versus -poor vegetables for 3 months, respectively, in addition to dietary caloric restriction. Anthropometry, diagnostic imaging (abdominal CT-scan, fibroscan), food-behavior questionnaires, serum biology and fecal microbiome (primary outcome; 16S rDNA sequencing) were analyzed before and after the intervention. RESULTS: Both placebo and prebiotic interventions lowered energy intake, BMI, systolic blood pressure, and serum γ-GT. The prebiotic induced greater weight loss and additionally decreased diastolic blood pressure, AST and insulinemia. Metformin treatment compromised most of the gut microbiota changes and metabolic improvements linked to prebiotic intervention. The prebiotic modulated specific bacteria, associated with the improvement of anthropometry (i.e. a decrease in Desulfovibrio and Clostridium sensu stricto). A large increase in Bifidobacterium appears as a signature of inulin intake rather than a driver of prebiotic-linked biological outcomes. CONCLUSIONS: Inulin-enriched diet is able to promote weight loss in obese patients, the treatment efficiency being related to gut microbiota characteristics. This treatment is more efficacious in patients who did not receive metformin as anti-diabetic drugs prior the intervention, supporting that both drug treatment and microbiota might be taken into account in personalized nutrition interventions. Registered under ClinicalTrials.gov Identifier no NCT03852069.


Asunto(s)
Restricción Calórica/métodos , Microbioma Gastrointestinal/fisiología , Inulina/administración & dosificación , Obesidad/dietoterapia , Prebióticos/administración & dosificación , Adolescente , Adulto , Anciano , Antropometría , Presión Sanguínea , Índice de Masa Corporal , Ingestión de Energía , Heces/microbiología , Conducta Alimentaria , Femenino , Humanos , Masculino , Metformina/administración & dosificación , Persona de Mediana Edad , Obesidad/microbiología , Polisacáridos/administración & dosificación , Método Simple Ciego , Resultado del Tratamiento , Verduras , Pérdida de Peso , Adulto Joven
4.
Gut ; 69(11): 1975-1987, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32041744

RESUMEN

OBJECTIVE: The gut microbiota has been proposed as an interesting therapeutic target for metabolic disorders. Inulin as a prebiotic has been shown to lessen obesity and related diseases. The aim of the current study was to investigate whether preintervention gut microbiota characteristics determine the physiological response to inulin. DESIGN: The stools from four obese donors differing by microbial diversity and composition were sampled before the dietary intervention and inoculated to antibiotic-pretreated mice (hum-ob mice; humanised obese mice). Hum-ob mice were fed with a high-fat diet and treated with inulin. Metabolic and microbiota changes on inulin treatment in hum-ob mice were compared with those obtained in a cohort of obese individuals supplemented with inulin for 3 months. RESULTS: We show that hum-ob mice colonised with the faecal microbiota from different obese individuals differentially respond to inulin supplementation on a high-fat diet. Among several bacterial genera, Barnesiella, Bilophila, Butyricimonas, Victivallis, Clostridium XIVa, Akkermansia, Raoultella and Blautia correlated with the observed metabolic outcomes (decrease in adiposity and hepatic steatosis) in hum-ob mice. In addition, in obese individuals, the preintervention levels of Anaerostipes, Akkermansia and Butyricicoccus drive the decrease of body mass index in response to inulin. CONCLUSION: These findings support that characterising the gut microbiota prior to nutritional intervention with prebiotics is important to increase the positive outcome in the context of obesity and metabolic disorders.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal/efectos de los fármacos , Inulina/uso terapéutico , Obesidad/microbiología , Obesidad/terapia , Prebióticos , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Método Simple Ciego
5.
Am J Clin Nutr ; 109(6): 1683-1695, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31108510

RESUMEN

BACKGROUND: Inulin-type fructans (ITFs) are a type of fermentable dietary fiber that can confer beneficial health effects through changes in the gut microbiota. However, their effect on gut sensitivity and nutritional behavior is a matter of debate. OBJECTIVE: We evaluated the impact of consuming ITF-rich vegetables daily on gut microbiota, gastro-intestinal symptoms, and food-related behavior in healthy individuals. METHODS: A single group-design trial was conducted in 26 healthy individuals. During 2 wk, the participants were instructed to adhere to a controlled diet based on ITF-rich vegetables (providing a mean intake of 15 g ITF/d). Three test days were organized: before and after the nutritional intervention and 3 wk after returning to their usual diet. We assessed nutrient intake, food-related behavior, fecal microbiota composition, microbial fermentation, and gastrointestinal symptoms. RESULTS: The major microbial modifications during the intervention were an increased proportion of the Bifidobacterium genus, a decreased level of unclassified Clostridiales, and a tendency to decrease Oxalobacteraceae. These changes were reversed 3 wk after the intervention. The volunteers showed greater satiety, a reduced desire to eat sweet, salty, and fatty food, and a trend to increase hedonic attitudes towards some inulin-rich vegetables. Only flatulence episodes were reported during the dietary intervention, whereas intestinal discomfort, inversely associated with Clostridium cluster IV and Ruminococcus callidus, was improved at the end of the intervention. CONCLUSIONS: A higher consumption of ITF-rich vegetables allows a substantial increase in well-tolerated dietary fiber, which may in turn improve food-related behavior. Moreover, it leads to beneficial modifications of the gut microbiota composition and function. This trial is registered at clinicaltrial.gov as NCT03540550.


Asunto(s)
Conducta Alimentaria , Microbioma Gastrointestinal , Inulina/metabolismo , Verduras/metabolismo , Adolescente , Adulto , Anciano , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Dieta , Fibras de la Dieta/análisis , Fibras de la Dieta/metabolismo , Heces/microbiología , Femenino , Voluntarios Sanos , Humanos , Inulina/análisis , Masculino , Persona de Mediana Edad , Prebióticos/análisis , Verduras/química , Adulto Joven
6.
Nutrients ; 10(5)2018 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-29693598

RESUMEN

Postprandial hyperlipidemia is an important risk factor for cardiovascular diseases in the context of obesity. Inulin is a non-digestible carbohydrate, known for its beneficial properties in metabolic disorders. We investigated the impact of inulin on postprandial hypertriglyceridemia and on lipid metabolism in a mouse model of diet-induced obesity. Mice received a control or a western diet for 4 weeks and were further supplemented or not with inulin for 2 weeks (0.2 g/day per mouse). We performed a lipid tolerance test, measured mRNA expression of genes involved in postprandial lipid metabolism, assessed post-heparin plasma and muscle lipoprotein lipase activity and measured lipid accumulation in the enterocytes and fecal lipid excretion. Inulin supplementation in western diet-fed mice decreases postprandial serum triglycerides concentration, decreases the mRNA expression levels of Cd36 (fatty acid receptor involved in lipid uptake and sensing) and apolipoprotein C3 (Apoc3, inhibitor of lipoprotein lipase) in the jejunum and increases fecal lipid excretion. In conclusion, inulin improves postprandial hypertriglyceridemia by targeting intestinal lipid metabolism. This work confirms the interest of using inulin supplementation in the management of dyslipidemia linked to obesity and cardiometabolic risk.


Asunto(s)
Hipertrigliceridemia/tratamiento farmacológico , Hipolipemiantes/farmacología , Intestino Delgado/efectos de los fármacos , Inulina/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Obesidad/tratamiento farmacológico , Periodo Posprandial , Triglicéridos/sangre , Animales , Apolipoproteína C-III/genética , Apolipoproteína C-III/metabolismo , Biomarcadores/sangre , Antígenos CD36/genética , Antígenos CD36/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hipertrigliceridemia/sangre , Hipertrigliceridemia/genética , Intestino Delgado/metabolismo , Metabolismo de los Lípidos/genética , Masculino , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo
7.
PLoS One ; 13(2): e0192447, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29389988

RESUMEN

SCOPE: Conjugated linoleic acids are linoleic acid isomers found in the diet that can also be produced through bacterial metabolism of polyunsaturated fatty acids. Our objective was to evaluate the contribution of fatty acid metabolites produced from polyunsaturated fatty acids by the gut microbiota in vivo to regulation of hepatic lipid metabolism and steatosis. METHODS AND RESULTS: In mice with depleted n-3 polyunsaturated fatty acids, we observed an accumulation of trans-11,trans-13 CLA and cis-9,cis-11 conjugated linoleic acids in the liver tissue that were associated with an increased triglyceride content and expression of lipogenic genes. We used an in vitro model to evaluate the impact of these two conjugated linoleic acids on hepatic lipid metabolism. In HepG2 cells, we observed that only trans-11,trans-13 conjugated linoleic acids recapitulated triglyceride accumulation and increased lipogenic gene expression, which is a phenomenon that may implicate the nuclear factors sterol regulatory element binding protein 1c (SREBP-1c) and carbohydrate-responsive element-binding protein (ChREBP). CONCLUSION: The trans-11,trans-13 conjugated linoleic acids can stimulate hepatic lipogenesis, which supports the conclusion that gut microbiota and related metabolites should be considered in the treatment of non-alcoholic liver disease.


Asunto(s)
Hígado Graso/inducido químicamente , Ácidos Linoleicos Conjugados/farmacología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Factores de Transcripción/fisiología
8.
Gut ; 67(2): 271-283, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28377388

RESUMEN

OBJECTIVE: To investigate the beneficial role of prebiotics on endothelial dysfunction, an early key marker of cardiovascular diseases, in an original mouse model linking steatosis and endothelial dysfunction. DESIGN: We examined the contribution of the gut microbiota to vascular dysfunction observed in apolipoprotein E knockout (Apoe-/-) mice fed an n-3 polyunsaturated fatty acid (PUFA)-depleted diet for 12 weeks with or without inulin-type fructans (ITFs) supplementation for the last 15 days. Mesenteric and carotid arteries were isolated to evaluate endothelium-dependent relaxation ex vivo. Caecal microbiota composition (Illumina Sequencing of the 16S rRNA gene) and key pathways/mediators involved in the control of vascular function, including bile acid (BA) profiling, gut and liver key gene expression, nitric oxide and gut hormones production were also assessed. RESULTS: ITF supplementation totally reverses endothelial dysfunction in mesenteric and carotid arteries of n-3 PUFA-depleted Apoe-/- mice via activation of the nitric oxide (NO) synthase/NO pathway. Gut microbiota changes induced by prebiotic treatment consist in increased NO-producing bacteria, replenishment of abundance in Akkermansia and decreased abundance in bacterial taxa involved in secondary BA synthesis. Changes in gut and liver gene expression also occur upon ITFs suggesting increased glucagon-like peptide 1 production and BA turnover as drivers of endothelium function preservation. CONCLUSIONS: We demonstrate for the first time that ITF improve endothelial dysfunction, implicating a short-term adaptation of both gut microbiota and key gut peptides. If confirmed in humans, prebiotics could be proposed as a novel approach in the prevention of metabolic disorders-related cardiovascular diseases.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Fructanos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Prebióticos , Aminopeptidasas/genética , Animales , Péptidos Catiónicos Antimicrobianos/genética , Bacterias/efectos de los fármacos , Ácidos y Sales Biliares/biosíntesis , Ácidos y Sales Biliares/sangre , Arterias Carótidas/fisiología , Ciego/microbiología , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ácidos Grasos Omega-3/administración & dosificación , Ácidos Grasos Omega-3/deficiencia , Expresión Génica/efectos de los fármacos , Péptido 1 Similar al Glucagón/biosíntesis , Masculino , Arterias Mesentéricas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Neurotensina/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Proglucagón/genética , Simportadores/genética , Vasodilatación
9.
Mol Nutr Food Res ; 60(10): 2198-2207, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27136390

RESUMEN

SCOPE: Western diets are characterized by low intake of n-3 PUFA compensated by constant amounts of n-6 PUFA. Reduced intake of n-3 PUFA is associated with increased cardiovascular risk, as observed in nonalcoholic fatty liver disease patients. The study aimed to evaluating the impact of dietary n-3 PUFA depletion on endothelial function, an early key event of cardiovascular diseases. METHODS AND RESULTS: C57Bl/6J or apolipoprotein E knock-out (apoE-/- ) were fed control (CT) or n-3 PUFA-depleted diets (DEF) for 12 wks. Mice fed n-3 DEF diet developed a hepatic steatosis, linked to changes in hepatic expression of genes controlled by Sterol Regulatory Element Binding Protein-1 and -2. Vascular function was assessed on second- and third-order mesenteric arteries and n-3 PUFA-depleted apoE-/- mice presented endothelial dysfunction characterized by decreased vasorelaxation in response of acetylcholine. The presence of a nitric oxide synthase (NOS) inhibitor blunted the relaxation in each groups and heme-nitrosylated hemoglobin blood (Hb-NO) level was significantly lower in n-3 PUFA-depleted apoE-/- mice. CONCLUSION: Twelve weeks of n-3 DEF diet promote steatosis and accelerate the process of endothelial dysfunction in apoE-/- mice by a mechanism involving the NOS/NO pathway. We propose n-3 PUFA-depleted apoE-/- mice as a new model to study endothelial dysfunction related to hepatic steatosis independently of obesity.


Asunto(s)
Apolipoproteínas E/genética , Endotelio Vascular/fisiopatología , Ácidos Grasos Omega-3/farmacología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Acetilcolina/farmacología , Animales , Enfermedades Cardiovasculares/etiología , Suplementos Dietéticos , Modelos Animales de Enfermedad , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacocinética , Enfermedad del Hígado Graso no Alcohólico/etiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Vasodilatación/efectos de los fármacos
10.
Life Sci ; 132: 77-84, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25916803

RESUMEN

AIMS: Hypolipidemic drugs are prescribed in the most of cases for the treatment of cardiovascular diseases. Several studies have showed that the gut microbiota is able to regulate the host cholesterol metabolism. This study aimed to investigate the potential impact of hypolipidemic drugs on the gut microbiota in mice, and to correlate it to the regulation of cholesterol metabolism. MAIN METHODS: Male C57Bl/6J mice were divided into four groups fed either a control diet alone (CT), or supplemented with simvastatin (0.1% w/w, Zocor®, MSD), or ezetimibe (0.021% w/w, Ezetrol®, MSD) or a combination of simvastatin and ezetimibe (0.1% and 0.021%, respectively) for one week. KEY FINDINGS: The combination of ezetimibe and simvastatin is required to observe a drop in cholesterolemia, linked to a huge activation of hepatic SREBP-2 and the consequent increased expression of genes involved in LDL cholesterol uptake and cholesterol synthesis. The gut microbiota analysis revealed no change in total bacteria, and in major Gram positive and Gram negative bacteria, but a selective significant increase in Lactobacillus spp. in mice treated with the ezetimibe and a decrease by the combination. The changes in lactobacilli level observed in ezetimibe or combination treated-mice are negatively correlated to expression of genes related to cholesterol metabolism. SIGNIFICANCE: The present study showed that ezetimibe taken alone is able to modify the composition of gut microbiota in favor of Lactobacillus spp. These results suggest that members of the genus Lactobacillus play an important role in cholesterol metabolism, even in normocholesterolemic mouse model.


Asunto(s)
Azetidinas/farmacología , Colesterol/metabolismo , Tracto Gastrointestinal/microbiología , Regulación de la Expresión Génica/efectos de los fármacos , Redes y Vías Metabólicas/genética , Microbiota/efectos de los fármacos , Simvastatina/farmacología , Análisis de Varianza , Animales , Western Blotting , Colesterol/biosíntesis , Cartilla de ADN/genética , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Ezetimiba , Regulación de la Expresión Génica/genética , Lactobacillus/efectos de los fármacos , Lactobacillus/crecimiento & desarrollo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
11.
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
12.
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
13.
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
14.
Nutr Metab (Lond) ; 8(1): 44, 2011 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-21707971

RESUMEN

BACKGROUND: Western diet is characterized by an insufficient n-3 polyunsaturated fatty acid (PUFA) consumption which is known to promote the pathogenesis of several diseases. We have previously observed that mice fed with a diet poor in n-3 PUFA for two generations exhibit hepatic steatosis together with a decrease in body weight. The gut microbiota contributes to the regulation of host energy metabolism, due to symbiotic relationship with fermentable nutrients provided in the diet. In this study, we have tested the hypothesis that perturbations of the gut microbiota contribute to the metabolic alterations occurring in mice fed a diet poor in n-3 PUFA for two generations (n-3/- mice). METHODS: C57Bl/6J mice fed with a control or an n-3 PUFA depleted diet for two generations were supplemented with prebiotic (inulin-type Fructooligosaccharides, FOS, 0.20 g/day/mice) during 24 days. RESULTS: n-3/-mice exhibited a marked drop in caecum weight, a decrease in lactobacilli and an increase in bifidobacteria in the caecal content as compared to control mice (n-3/+ mice). Dietary supplementation with FOS for 24 days was sufficient to increase caecal weight and bifidobacteria count in both n-3/+ and n-3/-mice. Moreover, FOS increased lactobacilli content in n-3/-mice, whereas it decreased their level in n-3/+ mice. Interestingly, FOS treatment promoted body weight gain in n-3/-mice by increasing energy efficiency. In addition, FOS treatment decreased fasting glycemia and lowered the higher expression of key factors involved in the fatty acid catabolism observed in the liver of n-3/-mice, without lessening steatosis. CONCLUSIONS: the changes in the gut microbiota composition induced by FOS are different depending on the type of diet. We show that FOS may promote lactobacilli and counteract the catabolic status induced by n-3 PUFA depletion in mice, thereby contributing to restore efficient fat storage.

15.
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
16.
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
17.
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
18.
Biochem Pharmacol ; 78(11): 1391-400, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19632207

RESUMEN

BACKGROUND: Diabetes and obesity are metabolic disorders induced by an excessive dietary intake of fat, usually related to inflammation and oxidative stress. AIMS: The aim of the study is to investigate the effect of the antioxidant coenzyme Q10 (CoQ10) on hepatic metabolic and inflammatory disorders associated with diet-induced obesity and glucose intolerance. METHODS: C57bl6/j mice were fed for 8 weeks, either a control diet (CT) or a high-fat diet plus 21% fructose in the drinking water (HFF). CoQ10 supplementation was performed in this later condition (HFFQ). RESULTS: HFF mice exhibit increased energy consumption, fat mass development, fasting glycaemia and insulinemia and impaired glucose tolerance. HFF treatment promoted the expression of genes involved in reactive oxygen species production (NADPH oxidase), inflammation (CRP, STAMP2) and metabolism (CPT1alpha) in the liver. CoQ10 supplementation decreased the global hepatic mRNA expression of inflammatory and metabolic stresses markers without changing obesity and tissue lipid peroxides compared to HFF mice. HFF diets paradoxically decreased TBARS (reflecting lipid peroxides) levels in liver, muscle and adipose tissue versus CT group, an effect related to vitamin E content of the diet. CONCLUSION: In conclusion, HFF model promotes glucose intolerance and obesity by a mechanism independent on the level of tissue peroxides. CoQ10 tends to decrease hepatic stress gene expression, independently of any modulation of lipid peroxidation, which is classically considered as its most relevant effect.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Hígado/efectos de los fármacos , Obesidad/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Ubiquinona/análogos & derivados , Animales , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Fructosa/administración & dosificación , Glucosa/metabolismo , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/metabolismo , Homeostasis , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Peróxidos Lipídicos/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , ARN Mensajero/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/farmacología , Ubiquinona/uso terapéutico
19.
Int Immunopharmacol ; 9(6): 767-73, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19286482

RESUMEN

Recent data reported that chitosan reduces high-fat (HF) diet-induced obesity in mice without describing the metabolic consequences of such an effect. The aim of this study was to investigate the capacity of chitosan derived from edible mushrooms to modify adipocytokine levels and to assess the relevance of this effect on the development of fat mass, and on glucose and lipid metabolism in obese mice. Mice were fed a HF diet or a HF diet supplemented with 5% fungal chitosan for ten weeks. HF-induced hypertriglyceridaemia, fasting hyperinsulinaemia and fat accumulation in liver, muscle and white adipose tissue (WAT) were reduced after chitosan treatment. The higher lipid content in the caecum following treatment with chitosan suggested that this dietary fiber reduced lipid absorption. We postulated that the lower triglyceridaemia observed upon chitosan treatment could also be the result of the lower FIAF (fasting-induced adipose factor) expression observed in visceral adipose tissue. IL-6, resistin and leptin levels decreased in the serum after chitosan supplementation. We conclude that fungal chitosan counteracts some inflammatory disorders and metabolic alterations occurring in diet-induced obese mice since it decreases feed efficiency, fat mass, adipocytokine secretion and ectopic fat deposition in the liver and the muscle.


Asunto(s)
Adipoquinas/antagonistas & inhibidores , Agaricales/química , Anticolesterolemiantes/uso terapéutico , Quitosano/administración & dosificación , Suplementos Dietéticos , Obesidad/dietoterapia , Adipoquinas/metabolismo , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/antagonistas & inhibidores , Angiopoyetinas/metabolismo , Animales , Anticolesterolemiantes/farmacología , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/metabolismo , Quitosano/química , Dieta , Insulina/sangre , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Leptina/antagonistas & inhibidores , Leptina/metabolismo , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Músculos/efectos de los fármacos , Músculos/metabolismo , Músculos/patología , Obesidad/patología , Resistina/antagonistas & inhibidores , Resistina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...