Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nutr Metab Cardiovasc Dis ; 28(12): 1296-1303, 2018 12.
Article in English | MEDLINE | ID: mdl-30459055

ABSTRACT

BACKGROUND & AIMS: Serum lipids and lipoproteins are established biomarkers of cardiovascular disease risk that could be influenced by impaired gut barrier function via effects on the absorption of dietary and biliary cholesterol. The aim of this study was to examine the potential relationship between gut barrier function (gut permeability) and concentration of serum lipids and lipoproteins, in an ancillary analysis of serum samples taken from a previous study. METHODS AND RESULTS: Serum lipids, lipoproteins and functional gut permeability, as assessed by the percentage of the urinary recovery of 51Cr-labelled EDTA absorbed within 24 h, were measured in a group of 30 healthy men. Serum lipopolysaccharide, high sensitivity C-reactive protein and interleukin-6 were also measured as markers of low-grade inflammation. The group expressed a 5-fold variation in total gut permeability (1.11-5.03%). Gut permeability was unrelated to the concentration of both serum total and low density lipoprotein (LDL)-cholesterol, but was positively associated with serum high density lipoprotein (HDL)-cholesterol (r = 0.434, P = 0.015). Serum HDL-cholesterol was also positively associated with serum endotoxaemia (r = 0.415, P = 0.023). CONCLUSION: The significant association between increased gut permeability and elevated serum HDL-cholesterol is consistent with the role of HDL as an acute phase reactant, and in this situation, potentially dysfunctional lipoprotein. This finding may have negative implications for the putative role of HDL as a cardio-protective lipoprotein.


Subject(s)
Cholesterol, HDL/blood , Dyslipidemias/blood , Inflammation Mediators/blood , Inflammation/blood , Intestinal Absorption , Intestines/physiopathology , Adult , Aged , Biomarkers/blood , C-Reactive Protein/metabolism , Cross-Sectional Studies , Dyslipidemias/diagnosis , Dyslipidemias/physiopathology , Humans , Inflammation/diagnosis , Inflammation/physiopathology , Interleukin-6/blood , Lipopolysaccharides/blood , Male , Middle Aged , Permeability , Up-Regulation
2.
Int J Obes Suppl ; 6(Suppl 1): S28-S31, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28685027

ABSTRACT

Gut microbes are now considered as key partners involved in human physiology. Data have shown that microbes contribute to regulate energy, lipid, and glucose homeostasis through several mechanisms. Among them, the role of pathogen-associated molecular pattern and bacterial metabolites has been proposed (for example, metabolic endotoxemia and bioactive lipids). This short review, briefly discusses the role of the gut barrier as well as the impact of both the innate immune system and bioactive molecules (for example, endocannabinoids, cytochrome P450 derived arachidonic acids compounds) in the framework of gut microbes and cardiometabolic disorders.

3.
Clin Nutr ; 34(3): 341-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25459400

ABSTRACT

Increasing evidence shows that gut microbiota composition is related to changes of gut barrier function including gut permeability and immune function. Gut microbiota is different in obese compared to lean subjects, suggesting that gut microbes are also involved in energy metabolism and subsequent nutritional state. While research on gut microbiota and gut barrier has presently mostly focused on intestinal inflammatory bowel diseases and more recently on obesity and type 2 diabetes, this review aims at summarizing the present knowledge regarding the impact, in vivo, of depleted nutritional states on structure and function of the gut epithelium, the gut-associated lymphoid tissue (GALT), the gut microbiota and the enteric nervous system. It highlights the complex interactions between the components of gut barrier in depleted states due to food deprivation, food restriction and protein energy wasting and shows that these interactions are multidirectional, implying the existence of feedbacks.


Subject(s)
Cachexia/microbiology , Food Deprivation , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Protein-Energy Malnutrition/microbiology , Animals , Cachexia/pathology , Diabetes Mellitus, Type 2/microbiology , Disease Models, Animal , Energy Metabolism , Gastrointestinal Tract/metabolism , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Nutritional Status , Obesity/microbiology
4.
Diabetes Metab ; 40(4): 246-57, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24631413

ABSTRACT

The gut microbiota is now considered as a key factor in the regulation of numerous metabolic pathways. Growing evidence suggests that cross-talk between gut bacteria and host is achieved through specific metabolites (such as short-chain fatty acids) and molecular patterns of microbial membranes (lipopolysaccharides) that activate host cell receptors (such as toll-like receptors and G-protein-coupled receptors). The endocannabinoid (eCB) system is an important target in the context of obesity, type 2 diabetes (T2D) and inflammation. It has been demonstrated that eCB system activity is involved in the control of glucose and energy metabolism, and can be tuned up or down by specific gut microbes (for example, Akkermansia muciniphila). Numerous studies have also shown that the composition of the gut microbiota differs between obese and/or T2D individuals and those who are lean and non-diabetic. Although some shared taxa are often cited, there is still no clear consensus on the precise microbial composition that triggers metabolic disorders, and causality between specific microbes and the development of such diseases is yet to be proven in humans. Nevertheless, gastric bypass is most likely the most efficient procedure for reducing body weight and treating T2D. Interestingly, several reports have shown that the gut microbiota is profoundly affected by the procedure. It has been suggested that the consistent postoperative increase in certain bacterial groups such as Proteobacteria, Bacteroidetes and Verrucomicrobia (A. muciniphila) may explain its beneficial impact in gnotobiotic mice. Taken together, these data suggest that specific gut microbes modulate important host biological systems that contribute to the control of energy homoeostasis, glucose metabolism and inflammation in obesity and T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Endocannabinoids/metabolism , Glucose/metabolism , Microbiota/physiology , Obesity/metabolism , Animals , Humans , Mice
5.
Benef Microbes ; 5(1): 3-17, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23886976

ABSTRACT

Crosstalk between organs is crucial for controlling numerous homeostatic systems (e.g. energy balance, glucose metabolism and immunity). Several pathological conditions, such as obesity and type 2 diabetes, are characterised by a loss of or excessive inter-organ communication that contributes to the development of disease. Recently, we and others have identified several mechanisms linking the gut microbiota with the development of obesity and associated disorders (e.g. insulin resistance, type 2 diabetes, hepatic steatosis). Among these, we described the concept of metabolic endotoxaemia (increase in plasma lipopolysaccharide levels) as one of the triggering factors leading to the development of metabolic inflammation and insulin resistance. Growing evidence suggests that gut microbes contribute to the onset of low-grade inflammation characterising these metabolic disorders via mechanisms associated with gut barrier dysfunctions. We have demonstrated that enteroendocrine cells (producing glucagon-like peptide-1, peptide YY and glucagon-like peptide-2) and the endocannabinoid system control gut permeability and metabolic endotoxaemia. Recently, we hypothesised that specific metabolic dysregulations occurring at the level of numerous organs (e.g. gut, adipose tissue, muscles, liver and brain) rely from gut microbiota modifications. In this review, we discuss the mechanisms linking gut permeability, adipose tissue metabolism, and glucose homeostasis, and recent findings that show interactions between the gut microbiota, the endocannabinoid system and the apelinergic system. These specific systems are discussed in the context of the gut-to-peripheral organ axis (intestine, adipose tissue and brain) and impacts on metabolic regulation. In the present review, we also briefly describe the impact of a variety of non-digestible nutrients (i.e. inulin-type fructans, arabinoxylans, chitin glucans and polyphenols). Their effects on the composition of the gut microbiota and activity are discussed in the context of obesity and type 2 diabetes.


Subject(s)
Adipose Tissue/growth & development , Gastrointestinal Tract/microbiology , Glucose/metabolism , Obesity/microbiology , Prebiotics , Adipose Tissue/metabolism , Adipose Tissue/microbiology , Animals , Diabetes Mellitus, Type 2/microbiology , Endocannabinoids/metabolism , Endotoxemia/microbiology , Fatty Liver/microbiology , Humans , Inflammation/immunology , Inflammation/microbiology , Insulin Resistance , Lipopolysaccharides/blood , Liver/pathology , Mice , Microbiota
6.
Obes Rev ; 14(9): 721-35, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23663746

ABSTRACT

Obesity is associated with numerous metabolic comorbidities. Weight loss is an effective measure for alleviating many of these metabolic abnormalities. However, considering the limited success of most medical weight-management approaches in producing a sustained weight loss, approaches that improve obesity-related metabolic abnormalities independent of weight loss would be extremely attractive and of practical benefit. Metabolically healthy obesity supports the notion that a better metabolic profile is possible despite obesity. Moreover, adequate expansion of adipose tissue appears to confer protection from obesity-induced metabolic comorbidities. To this end, the 10th Stock conference examined new approaches to improve metabolic comorbidities independent of weight loss. In particular, human adenovirus 36 (Ad36) and specific gut microbes were examined for their potential to influence lipid and glucose homeostasis in animals and humans. While these microbes possess some undesirable properties, research has identified attributes of adenovirus Ad36 and gut microbes that may be selectively harnessed to improve metabolic profile without the obligatory weight loss. Furthermore, identifying the host signalling pathways that these microbes recruit to improve the metabolic profile may offer new templates and targets, which may facilitate the development of novel treatment strategies for obesity-related metabolic conditions.


Subject(s)
Adipogenesis , Adipose Tissue/microbiology , Gastrointestinal Tract/microbiology , Glucose/metabolism , Lipids/blood , Obesity/therapy , Adipose Tissue/metabolism , Comorbidity , Gastrointestinal Tract/metabolism , Homeostasis , Humans , Metabolic Diseases/metabolism , Metabolic Diseases/microbiology , Microbiota , Obesity/microbiology , Weight Loss
7.
Nutr Diabetes ; 2: e28, 2012 Jan 23.
Article in English | MEDLINE | ID: mdl-23154683

ABSTRACT

BACKGROUND: Alterations in the composition of gut microbiota -known as dysbiosis- have been proposed to contribute to the development of obesity, thereby supporting the potential interest of nutrients acting on the gut microbes to produce beneficial effect on host energetic metabolism. Non-digestible fermentable carbohydrates present in cereals may be interesting nutrients able to influence the gut microbiota composition. OBJECTIVE AND DESIGN: The aim of the present study was to test the prebiotic potency of arabinoxylan oligosaccharides (AXOS) prepared from wheat bran in a nutritional model of obesity, associated with a low-grade chronic systemic inflammation. Mice were fed either a control diet or a high fat (HF) diet, or a HF diet supplemented with AXOS during 8 weeks. RESULTS: AXOS supplementation induced caecal and colon enlargement associated with an important bifidogenic effect. It increased the level of circulating satietogenic peptides produced by the colon (peptide YY and glucagon-like peptide-1), and coherently counteracted HF-induced body weight gain and fat mass development. HF-induced hyperinsulinemia and the Homeostasis Model Assessment of insulin resistance were decreased upon AXOS feeding. In addition, AXOS reduced HF-induced metabolic endotoxemia, macrophage infiltration (mRNA of F4/80) in the adipose tissue and interleukin 6 (IL6) in the plasma. The tight junction proteins (zonula occludens 1 and claudin 3) altered upon HF feeding were upregulated by AXOS treatment suggesting that the lower inflammatory tone was associated with the improvement of gut barrier function. CONCLUSION: Together, these findings suggest that specific non-digestible carbohydrates produced from cereals such as AXOS constitute a promising prebiotic nutrient in the control of obesity and related metabolic disorders.

8.
Br J Cancer ; 107(8): 1337-44, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-22976799

ABSTRACT

BACKGROUND: Metabolites released by the gut microbiota may influence host metabolism and immunity. We have tested the hypothesis that inulin-type fructans (ITF), by promoting microbial production of short-chain fatty acids (SCFA), influence cancer cell proliferation outside the gut. METHODS: Mice transplanted with Bcr-Abl-transfected BaF3 cells, received ITF in their drinking water. Gut microbiota was analysed by 16S rDNA polymerase chain reaction (PCR)-denaturing gradient gel electrophoresis (DGGE) and qPCR. Serum Short-chain fatty acids were quantified by UHPLC-MS. Cell proliferation was evaluated in vivo, by molecular biology and histology, and in vitro. RESULTS: Inulin-type fructans treatment reduces hepatic BaF3 cell infiltration, lessens inflammation and increases portal propionate concentration. In vitro, propionate reduces BaF3 cell growth through a cAMP level-dependent pathway. Furthermore, the activation of free fatty acid receptor 2 (FFA2), a Gi/Gq-protein-coupled receptor also known as GPR43 and that binds propionate, lessens the proliferation of BaF3 and other human cancer cell lines. CONCLUSION: We show for the first time that the fermentation of nutrients such as ITF into propionate can counteract malignant cell proliferation in the liver tissue. Our results support the interest of FFA2 activation as a new strategy for cancer therapeutics. This study highlights the importance of research focusing on gut microbes-host interactions for managing systemic and severe diseases such as leukaemia.


Subject(s)
Fructans/administration & dosage , Intestines/microbiology , Leukemia/metabolism , Liver/pathology , Metagenome/immunology , Propionates/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Proliferation , Diet , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Female , Fructans/metabolism , Fructans/pharmacology , Metagenome/drug effects , Mice , Mice, Inbred BALB C , Prebiotics
9.
Clin Microbiol Infect ; 18 Suppl 4: 50-3, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22647050

ABSTRACT

Obesity is associated with type 2 diabetes, insulin resistance and low grade inflammation. The gut microbiota is now considered as one of the most important environmental factors impacting on host physiology and metabolism. We have recently pointed out the role of this 'organ' on the onset of insulin resistance and the low grade inflammatory tone characterizing obesity. Among the mechanisms, we have introduced the novel concept of metabolic endotoxaemia as factor triggering low grade inflammation and associated disorders. More recently, two novel mechanisms involved in the development of gut permeability and adipose tissue plasticity have been identified. Specific attention has been paid to the role of the glucagon-like peptide 2 and the endocannabinoid system. This review briefly discusses the role of prebiotics as a key tool to modulate the gut microbiota, the gut barrier function, inflammation and the insulin resistance associated with obesity.


Subject(s)
Adipose Tissue/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Metagenome , Obesity/complications , Receptors, Cannabinoid/metabolism , Adipose Tissue/immunology , Humans , Inflammation/etiology , Insulin Resistance , Prebiotics/microbiology
10.
Acta Gastroenterol Belg ; 73(2): 267-9, 2010.
Article in English | MEDLINE | ID: mdl-20690567

ABSTRACT

Nowadays, the literature provides evidence that obesity, type 2 diabetes and insulin resistance are characterized by a low grade inflammation. Among the environmental factors involved in such diseases, the gut microbiota has been proposed as a key player. This neglected "organ" has been found to be different between healthy and or obese and type 2 diabetic patients. For example, recent data have proposed that dysbiosis of gut microbiota (at phyla, genus, or species level) affects host metabolism and energy storage. Among the mechanisms, metabolic endotoxemia (higher plasma LPS levels), gut permeability and the modulation of gut peptides (GLP-1 and GLP-2) have been proposed as putative targets. Here we discuss 1 degrees the specific modulation of the gut microbiota composition by using prebiotics and 2 degrees the novel findings that may explain how gut microbiota can be involved in the development or in the control of obesity and associated low-grade inflammation.


Subject(s)
Inflammation/microbiology , Intestines/microbiology , Metagenome , Obesity/pathology , Humans
11.
Gut ; 58(8): 1091-103, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19240062

ABSTRACT

BACKGROUND AND AIMS: Obese and diabetic mice display enhanced intestinal permeability and metabolic endotoxaemia that participate in the occurrence of metabolic disorders. Our recent data support the idea that a selective increase of Bifidobacterium spp. reduces the impact of high-fat diet-induced metabolic endotoxaemia and inflammatory disorders. Here, we hypothesised that prebiotic modulation of gut microbiota lowers intestinal permeability, by a mechanism involving glucagon-like peptide-2 (GLP-2) thereby improving inflammation and metabolic disorders during obesity and diabetes. METHODS: Study 1: ob/ob mice (Ob-CT) were treated with either prebiotic (Ob-Pre) or non-prebiotic carbohydrates as control (Ob-Cell). Study 2: Ob-CT and Ob-Pre mice were treated with GLP-2 antagonist or saline. Study 3: Ob-CT mice were treated with a GLP-2 agonist or saline. We assessed changes in the gut microbiota, intestinal permeability, gut peptides, intestinal epithelial tight-junction proteins ZO-1 and occludin (qPCR and immunohistochemistry), hepatic and systemic inflammation. RESULTS: Prebiotic-treated mice exhibited a lower plasma lipopolysaccharide (LPS) and cytokines, and a decreased hepatic expression of inflammatory and oxidative stress markers. This decreased inflammatory tone was associated with a lower intestinal permeability and improved tight-junction integrity compared to controls. Prebiotic increased the endogenous intestinotrophic proglucagon-derived peptide (GLP-2) production whereas the GLP-2 antagonist abolished most of the prebiotic effects. Finally, pharmacological GLP-2 treatment decreased gut permeability, systemic and hepatic inflammatory phenotype associated with obesity to a similar extent as that observed following prebiotic-induced changes in gut microbiota. CONCLUSION: We found that a selective gut microbiota change controls and increases endogenous GLP-2 production, and consequently improves gut barrier functions by a GLP-2-dependent mechanism, contributing to the improvement of gut barrier functions during obesity and diabetes.


Subject(s)
Cecum/microbiology , Glucagon-Like Peptide 2/physiology , Inflammation/prevention & control , Obesity/complications , Probiotics/therapeutic use , Adiposity/drug effects , Adiposity/physiology , Animals , Bacteria/isolation & purification , Cecum/physiopathology , Endotoxemia/etiology , Endotoxemia/prevention & control , Glucagon-Like Peptide 2/agonists , Glucagon-Like Peptide 2/antagonists & inhibitors , Hepatitis/etiology , Hepatitis/prevention & control , Inflammation/etiology , Inflammation/microbiology , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Membrane Proteins/metabolism , Mice , Mice, Obese , Obesity/microbiology , Obesity/physiopathology , Occludin , Oxidative Stress/drug effects , Oxidative Stress/physiology , Permeability , Phosphoproteins/metabolism , Proglucagon/genetics , RNA, Messenger/genetics , Tight Junctions/metabolism , Zonula Occludens-1 Protein
12.
BMC Physiol ; 8: 21, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19046413

ABSTRACT

BACKGROUND: There are only few data relating the metabolic consequences of feeding diets very low in n-3 fatty acids. This experiment carried out in mice aims at studying the impact of dietary n-3 polyunsaturated fatty acids (PUFA) depletion on hepatic metabolism. RESULTS: n-3 PUFA depletion leads to a significant decrease in body weight despite a similar caloric intake or adipose tissue weight. n-3 PUFA depleted mice exhibit hypercholesterolemia (total, HDL, and LDL cholesterol) as well as an increase in hepatic cholesteryl ester and triglycerides content. Fatty acid pattern is profoundly modified in hepatic phospholipids and triglycerides. The decrease in tissue n-3/n-6 PUFA ratio correlates with steatosis. Hepatic mRNA content of key factors involved in lipid metabolism suggest a decreased lipogenesis (SREBP-1c, FAS, PPAR gamma), and an increased beta-oxidation (CPT1, PPAR alpha and PGC1 alpha) without modification of fatty acid esterification (DGAT2, GPAT1), secretion (MTTP) or intracellular transport (L-FABP). Histological analysis reveals alterations of liver morphology, which can not be explained by inflammatory or oxidative stress. However, several proteins involved in the unfolded protein response are decreased in depleted mice. CONCLUSION: n-3 PUFA depletion leads to important metabolic alterations in murine liver. Steatosis occurs through a mechanism independent of the shift between beta-oxidation and lipogenesis. Moreover, long term n-3 PUFA depletion decreases the expression of factors involved in the unfolded protein response, suggesting a lower protection against endoplasmic reticulum stress in hepatocytes upon n-3 PUFA deficiency.


Subject(s)
Fatty Acids, Omega-3/metabolism , Fatty Liver/metabolism , Animals , Dietary Fats/administration & dosage , Dietary Fats/metabolism , Fatty Acids, Omega-3/blood , Fatty Liver/blood , Fatty Liver/etiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Tissue Distribution/physiology
13.
Diabetes Metab ; 34 Suppl 2: S49-55, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18640586

ABSTRACT

A major, yet poorly understood, feature of type 2 diabetes is the excessive hepatic glucose production and the corresponding insulin resistance leading to fasting hyperglycaemia. The tremendous amount of work done to provide the physiological and molecular mechanisms explaining this impairment has led to the emergence of several consensual hypotheses. Among these, is the increased daily and unregulated plasma glucagon concentration in type 2 diabetic patients. Therefore, studies aiming to understand the physiological regulation of glucagon secretion and the corresponding impairment during diabetes are directly relevant to the treatment of type 2 diabetes. Glucagon secretion by alpha-cells is an immediate response to glucopenia. Abnormal secretion of glucagon and other counterregulatory hormones is a hallmark of type 1 and type 2 diabetes and a major limitation to the use of strong hypoglycaemia agents. A few molecular mechanisms of glucose detection triggering counterregulation and in particular inducing glucagon secretion or suppressing it during hyperglycaemic episodes, have been identified. Such mechanisms are related to those of the insulin secreted beta-cell. The glucose transporter GLUT2 and the K-ATP dependent channel, as well as regulatory mechanisms, involved the central nervous system and the gut-brain hormone GLP-1. Over the last years, glucoincretins have provided promising results for the normalization of plasma glucagon concentration of type 2 diabetic patients, which could partly explain the therapeutic benefits of incretin-related therapy. The underlined mechanisms of GLP-1 regulated glucagon secretion are most likely related to the action of the hormone on the activation of the portal and brain glucose sensors. Certainly, strategies aiming to restore glucose-regulated glucagon secretion are important milestones for the treatment of diabetic patient and the prevention of iatrogenic hypoglycaemia.


Subject(s)
Diabetes Mellitus/physiopathology , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Hyperglycemia/physiopathology , Humans , Hyperglycemia/blood
14.
Pathol Biol (Paris) ; 56(5): 305-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18178333

ABSTRACT

A recent growing number of evidences shows that the increased prevalence of obesity and type 2 diabetes cannot be solely attributed to changes in the human genome, nutritional habits, or reduction of physical activity in our daily lives. Gut microflora may play an even more important role in maintaining human health. Recent data suggests that gut microbiota affects host nutritional metabolism with consequences on energy storage. Several mechanisms are proposed, linking events occurring in the colon and the regulation of energy metabolism. The present review discusses new findings that may explain how gut microbiota can be involved in the development of obesity and insulin resistance. Recently, studies have highlighted some key aspects of the mammalian host-gut microbial relationship. Gut microbiota could now be considered as a "microbial organ" localized within the host. Therefore, specific strategies aiming to regulate gut microbiota could be useful means to reduce the impact of high-fat feeding on the occurrence of metabolic diseases.


Subject(s)
Bacterial Physiological Phenomena , Dietary Fats/adverse effects , Insulin Resistance , Intestines/microbiology , Obesity/etiology , Animals , Bacteria/drug effects , Bacteria/metabolism , Bifidobacterium/physiology , Body Weight , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/physiopathology , Dietary Carbohydrates/pharmacology , Dietary Fats/pharmacology , Dietary Fiber/therapeutic use , Energy Metabolism , Humans , Inflammation/etiology , Inflammation/microbiology , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/physiology , Mice , Mice, Knockout , Mice, Obese , Obesity/epidemiology , Obesity/microbiology , Obesity/physiopathology , Polysaccharides/pharmacology , Prevalence
15.
Diabetologia ; 50(11): 2374-83, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17823788

ABSTRACT

AIMS/HYPOTHESIS: Recent evidence suggests that a particular gut microbial community may favour occurrence of the metabolic diseases. Recently, we reported that high-fat (HF) feeding was associated with higher endotoxaemia and lower Bifidobacterium species (spp.) caecal content in mice. We therefore tested whether restoration of the quantity of caecal Bifidobacterium spp. could modulate metabolic endotoxaemia, the inflammatory tone and the development of diabetes. METHODS: Since bifidobacteria have been reported to reduce intestinal endotoxin levels and improve mucosal barrier function, we specifically increased the gut bifidobacterial content of HF-diet-fed mice through the use of a prebiotic (oligofructose [OFS]). RESULTS: Compared with normal chow-fed control mice, HF feeding significantly reduced intestinal Gram-negative and Gram-positive bacteria including levels of bifidobacteria, a dominant member of the intestinal microbiota, which is seen as physiologically positive. As expected, HF-OFS-fed mice had totally restored quantities of bifidobacteria. HF-feeding significantly increased endotoxaemia, which was normalised to control levels in HF-OFS-treated mice. Multiple-correlation analyses showed that endotoxaemia significantly and negatively correlated with Bifidobacterium spp., but no relationship was seen between endotoxaemia and any other bacterial group. Finally, in HF-OFS-treated-mice, Bifidobacterium spp. significantly and positively correlated with improved glucose tolerance, glucose-induced insulin secretion and normalised inflammatory tone (decreased endotoxaemia, plasma and adipose tissue proinflammatory cytokines). CONCLUSIONS/INTERPRETATION: Together, these findings suggest that the gut microbiota contribute towards the pathophysiological regulation of endotoxaemia and set the tone of inflammation for occurrence of diabetes and/or obesity. Thus, it would be useful to develop specific strategies for modifying gut microbiota in favour of bifidobacteria to prevent the deleterious effect of HF-diet-induced metabolic diseases.


Subject(s)
Bifidobacterium/physiology , Diabetes Mellitus/epidemiology , Dietary Fats/adverse effects , Adipose Tissue/anatomy & histology , Animals , Body Weight , Diabetes Mellitus/prevention & control , Endotoxemia/physiopathology , Endotoxins/analysis , Energy Intake , Male , Mice , Mice, Inbred C57BL
16.
Diabetologia ; 49(3): 552-61, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16404553

ABSTRACT

AIMS/HYPOTHESIS: Hepatic insulin resistance is thought to be a critical component in the pathogenesis of type 2 diabetes but the role of intrinsic insulin signalling pathways in the regulation of hepatic metabolism remains controversial. Global gene targeting in mice and in vitro studies have suggested that IRS2 mediates the physiological effects of insulin in the liver. Reduced hepatic production of IRS2 is found in many cases of insulin resistance. To investigate the role of IRS2 in regulating liver function in vivo, we generated mice that specifically lack Irs2 in the liver (LivIrs2KO). MATERIALS AND METHODS: Hepatic insulin signalling events were examined in LivIrs2KO mice by western blotting. Glucose homeostasis and insulin sensitivity were assessed by glucose tolerance tests and hyperinsulinaemic-euglycaemic clamp studies. The effects of high-fat feeding upon glucose homeostasis were also determined. Liver function tests were performed and expression of key metabolic genes in the liver was determined by RT-PCR. RESULTS: Proximal insulin signalling events and forkhead box O1 and A2 function were normal in the liver of LivIrs2KO mice, which displayed minimal abnormalities in glucose and lipid homeostasis, hepatic gene expression and liver function. In addition, hepatic lipid homeostasis and the metabolic response to a high-fat diet did not differ between LivIrs2KO and control mice. CONCLUSIONS/INTERPRETATION: Our findings suggest that liver IRS2 signalling, surprisingly, is not required for the long-term maintenance of glucose and lipid homeostasis, and that extra-hepatic IRS2-dependent mechanisms are involved in the regulation of these processes.


Subject(s)
Gene Deletion , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism , Liver/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Animal Feed , Animals , Gene Expression Regulation , Glucose/metabolism , Homeostasis , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/deficiency , Mice , Mice, Knockout , Phosphoproteins/deficiency , Signal Transduction
17.
Eur J Clin Nutr ; 60(5): 567-72, 2006 May.
Article in English | MEDLINE | ID: mdl-16340949

ABSTRACT

OBJECTIVE: The administration of a fermentable dietary fibre (oligofructose) in rats increases satietogenic gut peptides and lowered spontaneous energy intake. The aim of the study was to assess the relevance of those effects of oligofructose on satiety and energy intake in humans. DESIGN: Single-blinded, crossover, placebo-controlled design, pilot study. SUBJECTS: Volunteers included five men and five women aged 21-39 years, BMI ranging from 18.5 to 27.4 kg/m(2), were randomly assigned as described below. INTERVENTIONS: Subjects were included in two 2-week experimental phases during which they received either fibre (oligofructose (OFS)) or placebo (dextrine maltose (DM)); a 2-week washout period was included between crossover phases. In total, 8 g OFS or 8 g DM were ingested twice daily (16 g/day in total). Energy intake, hunger, satiety, fullness and prospective food consumption were assessed with analogue scales at the end of each experimental phase. RESULTS: During breakfast, OFS significantly increases the satiety (P=0.04) without any difference on other sensations as compared to DM treatment periods. After lunch, no significant differences are observed between treatment period. At dinner, OFS significantly increases satiety (P=0.04), reduces hunger (P=0.04) and prospective food consumption (P=0.05). The energy intake at breakfast and lunch are significantly lower (P=0.01, 0.03, respectively) after OFS treatment than after DM treatment. Total energy intake per day is 5% lower during OFS than in DM period. CONCLUSION: Oligofructose treatment increases satiety following breakfast and dinner, reduces hunger and prospective food consumption following dinner. This pilot study presents a rationale to propose oligofructose supplements in the management of food intake in overweight and obese patients.


Subject(s)
Dietary Fiber/administration & dosage , Energy Intake/drug effects , Obesity/prevention & control , Oligosaccharides/administration & dosage , Satiation/drug effects , Adult , Appetite/drug effects , Appetite/physiology , Cross-Over Studies , Dietary Carbohydrates/administration & dosage , Eating/drug effects , Eating/physiology , Energy Intake/physiology , Female , Fermentation , Humans , Male , Pilot Projects , Satiation/physiology , Single-Blind Method , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...