Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nutr Res Rev ; 36(2): 471-483, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36156184

ABSTRACT

As we continue to elucidate the mechanisms underlying age-related brain diseases, the reductionist strategy in nutrition­brain function research has focused on establishing the impact of individual foods. However, the biological processes connecting diet and cognition are complex. Therefore, consideration of a combination of nutritional compounds may be most efficacious. One barrier to establishing the efficacy of multi-nutrient interventions is that the area lacks an established set of evidence-based guidelines for studying their effect on brain health. This review is an output of the International Life Sciences Institute (ILSI) Europe. A multi-disciplinary expert group was assembled with the aim of developing a set of considerations to guide research into the effects of multi-nutrient combinations on brain functions. Consensus recommendations converged on six key issues that should be considered to advance research in this area: (1) establish working mechanisms of the combination and contributions of each individual compound; (2) validate the relevance of the mechanisms for the targeted human condition; (3) include current nutrient status, intake or dietary pattern as inclusion/exclusion criteria in the study design; (4) select a participant population that is clinically and biologically appropriate for all nutritional components of the combination; (5) consider a range of cognitive outcomes; (6) consider the limits of reductionism and the 'gold standard' randomised controlled trial. These guiding principles will enhance our understanding of the interactive/complementary activities of dietary components, thereby strengthening the evidence base for recommendations aimed at delaying cognitive decline.


Subject(s)
Cognitive Aging , Nutrients , Humans , Food , Brain , Cognition , Randomized Controlled Trials as Topic
2.
Lancet Planet Health ; 6(9): e749-e759, 2022 09.
Article in English | MEDLINE | ID: mdl-36087605

ABSTRACT

The EAT-Lancet Commission devised a sustainable reference diet with the aim of reducing the incidence of non-communicable diseases and mortality globally while improving food system sustainability. The extent to which the reference diet supports cognitive function across the life course, however, has not yet been evaluated. This Review assesses the evidence for diet supporting cognitive function from childhood into old age. A comprehensive but non-exhaustive literature search was done, synthesising studies that investigated the effect of whole foods on cognition in healthy, community-dwelling human participants. We found that the current evidence base is weak with mixed conclusions and multiple methodological caveats, which precludes strong conclusions pertaining to the suitability of dietary recommendations for each food group per age group. Long-term intervention and prospective cohort studies are needed to reduce this knowledge deficit. Revising dietary recommendations with the aim of maintaining an adequate nutrient intake to sustain healthy cognitive function across the life course could be worthwhile. This Review outlines recommendations for future work to help improve the current knowledge deficit regarding dietary intake and cognitive function across the life course and its implications for dietary guidelines such as the EAT-Lancet Commission.


Subject(s)
Diet , Life Change Events , Child , Cognition , Humans , Nutrition Policy , Prospective Studies
3.
Cell Mol Life Sci ; 79(2): 80, 2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35044528

ABSTRACT

The gut and brain link via various metabolic and signalling pathways, each with the potential to influence mental, brain and cognitive health. Over the past decade, the involvement of the gut microbiota in gut-brain communication has become the focus of increased scientific interest, establishing the microbiota-gut-brain axis as a field of research. There is a growing number of association studies exploring the gut microbiota's possible role in memory, learning, anxiety, stress, neurodevelopmental and neurodegenerative disorders. Consequently, attention is now turning to how the microbiota can become the target of nutritional and therapeutic strategies for improved brain health and well-being. However, while such strategies that target the gut microbiota to influence brain health and function are currently under development with varying levels of success, still very little is yet known about the triggers and mechanisms underlying the gut microbiota's apparent influence on cognitive or brain function and most evidence comes from pre-clinical studies rather than well controlled clinical trials/investigations. Filling the knowledge gaps requires establishing a standardised methodology for human studies, including strong guidance for specific focus areas of the microbiota-gut-brain axis, the need for more extensive biological sample analyses, and identification of relevant biomarkers. Other urgent requirements are new advanced models for in vitro and in vivo studies of relevant mechanisms, and a greater focus on omics technologies with supporting bioinformatics resources (training, tools) to efficiently translate study findings, as well as the identification of relevant targets in study populations. The key to building a validated evidence base rely on increasing knowledge sharing and multi-disciplinary collaborations, along with continued public-private funding support. This will allow microbiota-gut-brain axis research to move to its next phase so we can identify realistic opportunities to modulate the microbiota for better brain health.


Subject(s)
Brain-Gut Axis , Brain/physiology , Gastrointestinal Microbiome , Animals , Brain/physiopathology , Cognition , Humans , Metabolic Networks and Pathways , Signal Transduction
4.
Obes Rev ; 22(8): e13234, 2021 08.
Article in English | MEDLINE | ID: mdl-33754456

ABSTRACT

This systematic review with meta-analyses aimed to identify the sensory and physical characteristics of foods/beverages which increase satiation and/or decrease/delay subsequent consumption without affecting acceptability. Systematic searches were first undertaken to identify review articles investigating the effects of any sensory and physical food characteristic on food intake. These articles provided some evidence that various textural parameters (aeration, hardness, homogeneity, viscosity, physical form, added water) can impact food intake. Individual studies investigating these effects while also investigating acceptability were then assessed. Thirty-seven individual studies investigated a textural manipulation and provided results on food intake and acceptability, 13 studies (27 comparisons, 898 participants) investigated effects on satiation, and 29 studies (54 comparisons, 916 participants) investigated effects on subsequent intake. Meta-analyses of within-subjects comparisons (random-effects models) demonstrated greater satiation (less weight consumed) from food products that were harder, chunkier, more viscous, voluminous, and/or solid, while demonstrating no effects on acceptability. Textural parameters had limited effects on subsequent consumption. Between-subjects studies and sensitivity analyses confirmed these results. These findings provide some evidence that textural parameters can increase satiation without affecting acceptability. The development of harder, chunkier, more viscous, voluminous, and/or solid food/beverage products may be of value in reducing overconsumption.


Subject(s)
Eating , Energy Intake , Food , Food Preferences , Humans , Satiation
5.
Ageing Res Rev ; 62: 101079, 2020 09.
Article in English | MEDLINE | ID: mdl-32461136

ABSTRACT

The global increases in life expectancy and population have resulted in a growing ageing population and with it a growing number of people living with age-related neurodegenerative conditions and dementia, shifting focus towards methods of prevention, with lifestyle approaches such as nutrition representing a promising avenue for further development. This overview summarises the main themes discussed during the 3rd Symposium on "Nutrition for the Ageing Brain: Moving Towards Clinical Applications" held in Madrid in August 2018, enlarged with the current state of knowledge on how nutrition influences healthy ageing and gives recommendations regarding how the critical field of nutrition and neurodegeneration research should move forward into the future. Specific nutrients are discussed as well as the impact of multi-nutrient and whole diet approaches, showing particular promise to combatting the growing burden of age-related cognitive decline. The emergence of new avenues for exploring the role of diet in healthy ageing, such as the impact of the gut microbiome and development of new techniques (imaging measures of brain metabolism, metabolomics, biomarkers) are enabling researchers to approach finding answers to these questions. But the translation of these findings into clinical and public health contexts remains an obstacle due to significant shortcomings in nutrition research or pressure on the scientific community to communicate recommendations to the general public in a convincing and accessible way. Some promising programs exist but further investigation to improve our understanding of the mechanisms by which nutrition can improve brain health across the human lifespan is still required.


Subject(s)
Healthy Aging , Nutritional Status , Aging , Brain , Diet , Humans
6.
Article in English | MEDLINE | ID: mdl-31550212

ABSTRACT

Some of the chemicals in materials used for packaging food may leak into the food, resulting in human exposure. These include so-called Non-intentionally Added Substances (NIAS), many of them being unidentified and toxicologically uncharacterized. This raises the question of how to address their safety. An approach consisting of identification and toxicologically testing all of them appears neither feasible nor necessary. Instead, it has been proposed to use the threshold of toxicological concern (TTC) Cramer class III to prioritise unknown NIAS on which further safety investigations should focus. Use of the Cramer class III TTC for this purpose would be appropriate if amongst others sufficient evidence were available that the unknown chemicals were not acetylcholinesterase inhibitors or direct DNA-reactive mutagens. While knowledge of the material and analytical chemistry may efficiently address the first concern, the second could not be addressed in this way. An alternative would be use of a bioassay capable of detecting DNA-reactive mutagens at very low levels. No fully satisfactory bioassay was identified. The Ames test appeared the most suitable since it specifically detects DNA-reactive mutagens and the limit of biological detection of highly potent genotoxic carcinogens is low. It is proposed that for a specific migrate, the evidence for absence of mutagenicity based on the Ames test, together with analytical chemistry and information on packaging manufacture could allow application of the Cramer class III TTC to prioritise unknown NIAS. Recommendations, as well as research proposals, have been developed on sample preparation and bioassay improvement with the ultimate aim of improving limits of biological detection of mutagens. Although research is still necessary, the proposed approach should bring significant benefits over the current practices used for safety evaluation of food contact materials.


Subject(s)
Biological Assay , Food Analysis , Food Contamination/analysis , Food Packaging , Humans
7.
Nat Commun ; 10(1): 457, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30692526

ABSTRACT

Variations in N-acylethanolamines (NAE) levels are associated with obesity and metabolic comorbidities. Their role in the gut remains unclear. Therefore, we generated a mouse model of inducible intestinal epithelial cell (IEC)-specific deletion of N-acylphosphatidylethanolamine phospholipase D (NAPE-PLD), a key enzyme involved in NAE biosynthesis (Napepld∆IEC). We discovered that Napepld∆IEC mice are hyperphagic upon first high-fat diet (HFD) exposure, and develop exacerbated obesity and steatosis. These mice display hypothalamic Pomc neurons dysfunctions and alterations in intestinal and plasma NAE and 2-acylglycerols. After long-term HFD, Napepld∆IEC mice present reduced energy expenditure. The increased steatosis is associated with higher gut and liver lipid absorption. Napepld∆IEC mice display altered gut microbiota. Akkermansia muciniphila administration partly counteracts the IEC NAPE-PLD deletion effects. In conclusion, intestinal NAPE-PLD is a key sensor in nutritional adaptation to fat intake, gut-to-brain axis and energy homeostasis and thereby constitutes a novel target to tackle obesity and related disorders.


Subject(s)
Dietary Fats/metabolism , Fatty Liver/metabolism , Intestinal Mucosa/metabolism , Obesity/metabolism , Phosphatidylethanolamines/metabolism , Phospholipase D/metabolism , Adaptation, Physiological , Animals , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Gastrointestinal Microbiome/physiology , Homeostasis , Intestinal Mucosa/microbiology , Lipid Metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Obesity/etiology
8.
Am J Physiol Endocrinol Metab ; 314(4): E334-E352, 2018 04 01.
Article in English | MEDLINE | ID: mdl-28874357

ABSTRACT

Increasing evidence suggests that polyphenols have a significant potential in the prevention and treatment of risk factors associated with metabolic syndrome. The objective of this study was to assess the metabolic outcomes of two polyphenol-containing extracts from cinnamon bark (CBE) and grape pomace (GPE) on C57BL/6J mice fed a high-fat diet (HFD) for 8 wk. Both CBE and GPE were able to decrease fat mass gain and adipose tissue inflammation in mice fed a HFD without reducing food intake. This was associated with reduced liver steatosis and lower plasma nonesterified fatty acid levels. We also observed a beneficial effect on glucose homeostasis, as evidenced by an improved glucose tolerance and a lower insulin resistance index. These ameliorations of the overall metabolic profile were associated with a significant impact on the microbial composition, which was more profound for the GPE than for the CBE. At the genus level, Peptococcus were decreased in the CBE group. In the GPE-treated group, several key genera that have been previously found to be linked with HFD, metabolic effects, and gut barrier integrity were affected: we observed a decrease of Desulfovibrio, Lactococcus, whereas Allobaculum and Roseburia were increased. In addition, the expression of several antimicrobial peptides and tight junction proteins was increased in response to both CBE and GPE supplementation, indicating an improvement of the gut barrier function. Collectively, these data suggest that CBE and GPE can ameliorate the overall metabolic profile of mice on a high-fat diet, partly by acting on the gut microbiota.


Subject(s)
Cinnamomum zeylanicum/chemistry , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/drug effects , Metabolic Diseases/prevention & control , Plant Extracts/pharmacology , Vitis/chemistry , Animals , Biomarkers/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Experimental/prevention & control , Diet, High-Fat/adverse effects , Fatty Liver/metabolism , Fatty Liver/microbiology , Fatty Liver/prevention & control , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Metabolic Diseases/microbiology , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/microbiology , Obesity/prevention & control , Permeability , Plant Extracts/therapeutic use
9.
Ageing Res Rev ; 42: 40-55, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29248758

ABSTRACT

BACKGROUND: Ageing is a highly complex process marked by a temporal cascade of events, which promote alterations in the normal functioning of an individual organism. The triggers of normal brain ageing are not well understood, even less so the factors which initiate and steer the neuronal degeneration, which underpin disorders such as dementia. A wealth of data on how nutrients and diets may support cognitive function and preserve brain health are available, yet the molecular mechanisms underlying their biological action in both normal ageing, age-related cognitive decline, and in the development of neurodegenerative disorders have not been clearly elucidated. OBJECTIVES: This review aims to summarise the current state of knowledge of vulnerabilities that predispose towards dysfunctional brain ageing, highlight potential protective mechanisms, and discuss dietary interventions that may be used as therapies. A special focus of this paper is on the impact of nutrition on neuroprotection and the underlying molecular mechanisms, and this focus reflects the discussions held during the 2nd workshop 'Nutrition for the Ageing Brain: Functional Aspects and Mechanisms' in Copenhagen in June 2016. The present review is the most recent in a series produced by the Nutrition and Mental Performance Task Force under the auspice of the International Life Sciences Institute Europe (ILSI Europe). CONCLUSION: Coupling studies of cognitive ageing with studies investigating the effect of nutrition and dietary interventions as strategies targeting specific mechanisms, such as neurogenesis, protein clearance, inflammation, and non-coding and microRNAs is of high value. Future research on the impact of nutrition on cognitive ageing will need to adopt a longitudinal approach and multimodal nutritional interventions will likely need to be imposed in early-life to observe significant impact in older age.


Subject(s)
Cognitive Aging/physiology , Cognitive Aging/psychology , Diet Therapy/methods , Nutritional Status/physiology , Aged , Aged, 80 and over , Aging/metabolism , Animals , Brain/metabolism , Cognition/physiology , Cognition Disorders/diet therapy , Cognition Disorders/metabolism , Cognition Disorders/psychology , Diet Therapy/trends , Humans , Nutrients/administration & dosage , Nutrients/metabolism , Obesity/diet therapy , Obesity/metabolism , Obesity/psychology
10.
J Nutr Biochem ; 49: 15-21, 2017 11.
Article in English | MEDLINE | ID: mdl-28863365

ABSTRACT

Fat browning has emerged as an attractive target for the treatment of obesity and related metabolic disorders. Its activation leads to increased energy expenditure and reduced adiposity, thus contributing to a better energy homeostasis. Green tea extracts (GTEs) were shown to attenuate obesity and low-grade inflammation and to induce the lipolytic pathway in the white adipose tissue (WAT) of mice fed a high-fat diet. The aim of the present study was to determine whether the antiobesity effect of an extract from green tea leaves was associated with the activation of browning in the WAT and/or the inhibition of whitening in the brown adipose tissue (BAT) in HF-diet induced obese mice. Mice were fed a control diet or an HF diet supplemented with or without 0.5% polyphenolic GTE for 8 weeks. GTE supplementation significantly reduced HF-induced adiposity (WAT and BAT) and HF-induced inflammation in WAT. Histological analysis revealed that GTE reduced the adipocyte size in the WAT and the lipid droplet size in the BAT. Markers of browning were induced in the WAT upon GTE treatment, whereas markers of HF-induced whitening were reduced in the BAT. These results suggest that browning activation in the WAT and whitening reduction in the BAT by the GTE could participate to the improvement of metabolic and inflammatory disorders mediated by GTE upon HF diet. Our study emphasizes the importance of using GTE as a nutritional tool to activate browning and to decrease fat storage in all adipose tissues, which attenuate obesity.


Subject(s)
Adipose Tissue, Brown/pathology , Anti-Obesity Agents/therapeutic use , Camellia sinensis/chemistry , Dietary Supplements , Obesity/prevention & control , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Adipogenesis , Adipose Tissue, Beige/immunology , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity , Animals , Biomarkers/metabolism , Cell Size , Diet, High-Fat/adverse effects , Food Handling , Lipid Droplets/immunology , Lipid Droplets/metabolism , Lipid Droplets/pathology , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Oxidation-Reduction , Polyphenols/therapeutic use , Random Allocation , Specific Pathogen-Free Organisms
11.
Gut ; 66(4): 620-632, 2017 04.
Article in English | MEDLINE | ID: mdl-27196572

ABSTRACT

OBJECTIVE: To examine the role of hepatocyte myeloid differentiation primary-response gene 88 (MyD88) on glucose and lipid metabolism. DESIGN: To study the impact of the innate immune system at the level of the hepatocyte and metabolism, we generated mice harbouring hepatocyte-specific deletion of MyD88. We investigated the impact of the deletion on metabolism by feeding mice with a normal control diet or a high-fat diet for 8 weeks. We evaluated body weight, fat mass gain (using time-domain nuclear magnetic resonance), glucose metabolism and energy homeostasis (using metabolic chambers). We performed microarrays and quantitative PCRs in the liver. In addition, we investigated the gut microbiota composition, bile acid profile and both liver and plasma metabolome. We analysed the expression pattern of genes in the liver of obese humans developing non-alcoholic steatohepatitis (NASH). RESULTS: Hepatocyte-specific deletion of MyD88 predisposes to glucose intolerance, inflammation and hepatic insulin resistance independently of body weight and adiposity. These phenotypic differences were partially attributed to differences in gene expression, transcriptional factor activity (ie, peroxisome proliferator activator receptor-α, farnesoid X receptor (FXR), liver X receptors and STAT3) and bile acid profiles involved in glucose, lipid metabolism and inflammation. In addition to these alterations, the genetic deletion of MyD88 in hepatocytes changes the gut microbiota composition and their metabolomes, resembling those observed during diet-induced obesity. Finally, obese humans with NASH displayed a decreased expression of different cytochromes P450 involved in bioactive lipid synthesis. CONCLUSIONS: Our study identifies a new link between innate immunity and hepatic synthesis of bile acids and bioactive lipids. This dialogue appears to be involved in the susceptibility to alterations associated with obesity such as type 2 diabetes and NASH, both in mice and humans.


Subject(s)
Bile Acids and Salts/metabolism , Gastrointestinal Microbiome/genetics , Glucose/metabolism , Hepatocytes/metabolism , Lipid Metabolism/genetics , Metabolome/genetics , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Adiposity , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Body Weight , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Diet, High-Fat , Gene Expression , Humans , Immunity, Innate/genetics , Insulin Resistance/genetics , Liver/metabolism , Liver X Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/genetics , Obesity/metabolism , PPAR alpha/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , STAT3 Transcription Factor/metabolism
12.
Nat Med ; 23(1): 107-113, 2017 01.
Article in English | MEDLINE | ID: mdl-27892954

ABSTRACT

Obesity and type 2 diabetes are associated with low-grade inflammation and specific changes in gut microbiota composition. We previously demonstrated that administration of Akkermansia muciniphila to mice prevents the development of obesity and associated complications. However, the underlying mechanisms of this protective effect remain unclear. Moreover, the sensitivity of A. muciniphila to oxygen and the presence of animal-derived compounds in its growth medium currently limit the development of translational approaches for human medicine. We have addressed these issues here by showing that A. muciniphila retains its efficacy when grown on a synthetic medium compatible with human administration. Unexpectedly, we discovered that pasteurization of A. muciniphila enhanced its capacity to reduce fat mass development, insulin resistance and dyslipidemia in mice. These improvements were notably associated with a modulation of the host urinary metabolomics profile and intestinal energy absorption. We demonstrated that Amuc_1100, a specific protein isolated from the outer membrane of A. muciniphila, interacts with Toll-like receptor 2, is stable at temperatures used for pasteurization, improves the gut barrier and partly recapitulates the beneficial effects of the bacterium. Finally, we showed that administration of live or pasteurized A. muciniphila grown on the synthetic medium is safe in humans. These findings provide support for the use of different preparations of A. muciniphila as therapeutic options to target human obesity and associated disorders.


Subject(s)
Adipose Tissue/drug effects , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/metabolism , Dyslipidemias/metabolism , Membrane Proteins/pharmacology , Obesity/metabolism , Toll-Like Receptor 2/drug effects , Verrucomicrobia , Adult , Animals , Blood Glucose/metabolism , Blotting, Western , Chromatography, Liquid , Disease Models, Animal , Female , Humans , Insulin Resistance , Intestinal Mucosa/metabolism , Intestines/drug effects , Male , Metabolic Syndrome/metabolism , Mice, Obese , Middle Aged , Toll-Like Receptor 2/metabolism
13.
Nat Rev Endocrinol ; 12(3): 133-43, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26678807

ABSTRACT

Various metabolic disorders are associated with changes in inflammatory tone. Among the latest advances in the metabolism field, the discovery that gut microorganisms have a major role in host metabolism has revealed the possibility of a plethora of associations between gut bacteria and numerous diseases. However, to date, few mechanisms have been clearly established. Accumulating evidence indicates that the endocannabinoid system and related bioactive lipids strongly contribute to several physiological processes and are a characteristic of obesity, type 2 diabetes mellitus and inflammation. In this Review, we briefly define the gut microbiota as well as the endocannabinoid system and associated bioactive lipids. We discuss existing literature regarding interactions between gut microorganisms and the endocannabinoid system, focusing specifically on the triad of adipose tissue, gut bacteria and the endocannabinoid system in the context of obesity and the development of fat mass. We highlight gut-barrier function by discussing the role of specific factors considered to be putative 'gate keepers' or 'gate openers', and their role in the gut microbiota-endocannabinoid system axis. Finally, we briefly discuss data related to the different pharmacological strategies currently used to target the endocannabinoid system, in the context of cardiometabolic disorders and intestinal inflammation.


Subject(s)
Endocannabinoids/metabolism , Endocannabinoids/physiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Animals , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/drug effects , Humans , Receptors, Cannabinoid/drug effects
14.
Nat Commun ; 6: 6495, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25757720

ABSTRACT

Obesity is a pandemic disease associated with many metabolic alterations and involves several organs and systems. The endocannabinoid system (ECS) appears to be a key regulator of energy homeostasis and metabolism. Here we show that specific deletion of the ECS synthesizing enzyme, NAPE-PLD, in adipocytes induces obesity, glucose intolerance, adipose tissue inflammation and altered lipid metabolism. We report that Napepld-deleted mice present an altered browning programme and are less responsive to cold-induced browning, highlighting the essential role of NAPE-PLD in regulating energy homeostasis and metabolism in the physiological state. Our results indicate that these alterations are mediated by a shift in gut microbiota composition that can partially transfer the phenotype to germ-free mice. Together, our findings uncover a role of adipose tissue NAPE-PLD on whole-body metabolism and provide support for targeting NAPE-PLD-derived bioactive lipids to treat obesity and related metabolic disorders.


Subject(s)
Adipose Tissue, Brown/metabolism , Gastrointestinal Microbiome/physiology , Glucose Intolerance/metabolism , Obesity/metabolism , Phospholipase D/genetics , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Body Fat Distribution , Cold Temperature , Endocannabinoids/metabolism , Energy Metabolism/physiology , Gene Expression , Glucose Intolerance/genetics , Glucose Intolerance/microbiology , Glucose Intolerance/pathology , Inflammation , Male , Mice , Mice, Knockout , Obesity/genetics , Obesity/microbiology , Obesity/pathology , Phospholipase D/deficiency
15.
Nat Commun ; 5: 5648, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25476696

ABSTRACT

Obesity is associated with a cluster of metabolic disorders, low-grade inflammation and altered gut microbiota. Whether host metabolism is controlled by intestinal innate immune system and the gut microbiota is unknown. Here we report that inducible intestinal epithelial cell-specific deletion of MyD88 partially protects against diet-induced obesity, diabetes and inflammation. This is associated with increased energy expenditure, an improved glucose homeostasis, reduced hepatic steatosis, fat mass and inflammation. Protection is transferred following gut microbiota transplantation to germ-free recipients. We also demonstrate that intestinal epithelial MyD88 deletion increases anti-inflammatory endocannabinoids, restores antimicrobial peptides production and increases intestinal regulatory T cells during diet-induced obesity. Targeting MyD88 after the onset of obesity reduces fat mass and inflammation. Our work thus identifies intestinal epithelial MyD88 as a sensor changing host metabolism according to the nutritional status and we show that targeting intestinal epithelial MyD88 constitutes a putative therapeutic target for obesity and related disorders.


Subject(s)
Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Myeloid Differentiation Factor 88/metabolism , Obesity/metabolism , Animals , Energy Metabolism , Female , Gene Deletion , Glucose/metabolism , Humans , Intestines/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Nutritional Status , Obesity/genetics , Obesity/prevention & control
16.
mBio ; 5(3): e01011-14, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24917595

ABSTRACT

UNLABELLED: Growing evidence shows that gut microbes are key factors involved in the regulation of energy homeostasis, metabolic inflammation, lipid metabolism, and glucose metabolism. Therefore, gut microbiota modulations caused by selectively fermented oligosaccharides or probiotic bacteria constitute an interesting target in the physiopathology of obesity. However, to date, no probiotic yeast has been investigated in this context. Therefore, our study aimed to evaluate the impact of the most-studied probiotic yeast (i.e., Saccharomyces boulardii Biocodex) on obesity and associated metabolic features, such as fat mass development, hepatic steatosis, and low-grade inflammation, in obese mice. S. boulardii was administered daily by oral gavage to leptin-resistant obese and type 2 diabetic mice (db/db) for 4 weeks. We found that S. boulardii-treated mice exhibited reduced body weight, fat mass, hepatic steatosis, and inflammatory tone. Interestingly, these effects of S. boulardii on host metabolism were associated with local effects in the intestine. S. boulardii increased cecum weight and cecum tissue weight but also induced dramatic changes in the gut microbial composition at the phylum, family, and genus levels. These gut microbiota changes in response to S. boulardii may also be correlated with the host metabolism response. In conclusion, this study demonstrates for the first time that S. boulardii may act as a beneficial probiotic treatment in the context of obesity and type 2 diabetes. IMPORTANCE: To date, no probiotic yeast have been investigated in the context of obesity and type 2 diabetes. Here we found that type 2 diabetic and obese mice (db/db) treated with Saccharomyces boulardii exhibited reduced body weight, fat mass, hepatic steatosis, and inflammatory tone. These effects on host metabolism were associated with local effects in the intestine. Importantly, by using pyrosequencing, we found that S. boulardii treatment induces changes of the gut microbiota composition at the phylum, family, and genus levels. Moreover, we found that gut microbiota changes in response to S. boulardii were correlated with several host metabolism responses.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Fatty Liver/etiology , Microbiota , Obesity/drug therapy , Probiotics/administration & dosage , Saccharomyces/physiology , Animals , Body Weight/drug effects , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/microbiology , Fats/metabolism , Fatty Liver/metabolism , Female , Gastrointestinal Tract/microbiology , Humans , Male , Mice , Mice, Obese , Microbiota/drug effects , Obesity/complications , Obesity/immunology , Obesity/microbiology
17.
Antioxid Redox Signal ; 20(4): 557-73, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23879244

ABSTRACT

AIMS: We have previously demonstrated that central apelin is implicated in the control of peripheral glycemia, and its action depends on nutritional (fast versus fed) and physiological (normal versus diabetic) states. An intracerebroventricular (icv) injection of a high dose of apelin, similar to that observed in obese/diabetic mice, increase fasted glycemia, suggesting (i) that apelin contributes to the establishment of a diabetic state, and (ii) the existence of a hypothalamic to liver axis. Using pharmacological, genetic, and nutritional approaches, we aim at unraveling this system of regulation by identifying the hypothalamic molecular actors that trigger the apelin effect on liver glucose metabolism and glycemia. RESULTS: We show that icv apelin injection stimulates liver glycogenolysis and gluconeogenesis via an over-activation of the sympathetic nervous system (SNS), leading to fasted hyperglycemia. The effect of central apelin on liver function is dependent of an increased production of hypothalamic reactive oxygen species (ROS). These data are strengthened by experiments using lentiviral vector-mediated over-expression of apelin in hypothalamus of mice that present over-activation of SNS associated to an increase in hepatic glucose production. Finally, we report that mice fed a high-fat diet present major alterations of hypothalamic apelin/ROS signaling, leading to activation of glycogenolysis. INNOVATION/CONCLUSION: These data bring compelling evidence that hypothalamic apelin is one master switch that participates in the onset of diabetes by directly acting on liver function. Our data support the idea that hypothalamic apelin is a new potential therapeutic target to treat diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Liver/metabolism , Reactive Oxygen Species/metabolism , Adipokines , Animals , Apelin , Autonomic Nervous System , Blood Glucose , Gluconeogenesis , Glycogenolysis , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Signal Transduction
18.
Proc Natl Acad Sci U S A ; 110(22): 9066-71, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23671105

ABSTRACT

Obesity and type 2 diabetes are characterized by altered gut microbiota, inflammation, and gut barrier disruption. Microbial composition and the mechanisms of interaction with the host that affect gut barrier function during obesity and type 2 diabetes have not been elucidated. We recently isolated Akkermansia muciniphila, which is a mucin-degrading bacterium that resides in the mucus layer. The presence of this bacterium inversely correlates with body weight in rodents and humans. However, the precise physiological roles played by this bacterium during obesity and metabolic disorders are unknown. This study demonstrated that the abundance of A. muciniphila decreased in obese and type 2 diabetic mice. We also observed that prebiotic feeding normalized A. muciniphila abundance, which correlated with an improved metabolic profile. In addition, we demonstrated that A. muciniphila treatment reversed high-fat diet-induced metabolic disorders, including fat-mass gain, metabolic endotoxemia, adipose tissue inflammation, and insulin resistance. A. muciniphila administration increased the intestinal levels of endocannabinoids that control inflammation, the gut barrier, and gut peptide secretion. Finally, we demonstrated that all these effects required viable A. muciniphila because treatment with heat-killed cells did not improve the metabolic profile or the mucus layer thickness. In summary, this study provides substantial insight into the intricate mechanisms of bacterial (i.e., A. muciniphila) regulation of the cross-talk between the host and gut microbiota. These results also provide a rationale for the development of a treatment that uses this human mucus colonizer for the prevention or treatment of obesity and its associated metabolic disorders.


Subject(s)
Diabetes Mellitus, Type 2/microbiology , Endocannabinoids/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Obesity/microbiology , Verrucomicrobia/metabolism , Adipose Tissue/metabolism , Analysis of Variance , Animals , Antimicrobial Cationic Peptides/metabolism , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Homeostasis/physiology , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Microarray Analysis , Obesity/therapy , Oligosaccharides , Prebiotics , Real-Time Polymerase Chain Reaction
19.
PLoS One ; 8(2): e55963, 2013.
Article in English | MEDLINE | ID: mdl-23393605

ABSTRACT

AIMS: Obesity and type 2 diabetes are characterised by low-grade inflammation, metabolic endotoxaemia (i.e., increased plasma lipopolysaccharides [LPS] levels) and altered endocannabinoid (eCB)-system tone. The aim of this study was to decipher the specific role of eCB-system stimulation or metabolic endotoxaemia in the onset of glucose intolerance, metabolic inflammation and altered lipid metabolism. METHODS: Mice were treated with either a cannabinoid (CB) receptor agonist (HU210) or low-dose LPS using subcutaneous mini-pumps for 6 weeks. After 3 weeks of the treatment under control (CT) diet, one-half of each group of mice were challenged with a high fat (HF) diet for the following 3-week period. RESULTS: Under basal conditions (control diet), chronic CB receptor agonist treatment (i.e., 6 weeks) induced glucose intolerance, stimulated metabolic endotoxaemia, and increased macrophage infiltration (CD11c and F4/80 expression) in the muscles; this phenomenon was associated with an altered lipid metabolism (increased PGC-1α expression and decreased CPT-1b expression) in this tissue. Chronic LPS treatment tended to increase the body weight and fat mass, with minor effects on the other metabolic parameters. Challenging mice with an HF diet following pre-treatment with the CB agonist exacerbated the HF diet-induced glucose intolerance, the muscle macrophage infiltration and the muscle's lipid content without affecting the body weight or the fat mass. CONCLUSION: Chronic CB receptor stimulation under basal conditions induces glucose intolerance, stimulates metabolic inflammation and alters lipid metabolism in the muscles. These effects worsen following the concomitant ingestion of an HF diet. Here, we highlight the central roles played by the eCB system and LPS in the pathophysiology of several hallmarks of obesity and type 2 diabetes.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Endocannabinoids/therapeutic use , Endotoxemia/metabolism , Macrophages/drug effects , Animals , Diabetes Mellitus, Type 2 , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Male , Mice , Mice, Inbred C57BL
20.
Gut Microbes ; 3(4): 279-88, 2012.
Article in English | MEDLINE | ID: mdl-22572877

ABSTRACT

Obesity is associated with metabolic alterations related to glucose homeostasis and cardiovascular risk factors. These metabolic alterations are associated with low-grade inflammation that contributes to the onset of these diseases. We and others have provided evidence that gut microbiota participates in whole-body metabolism by affecting energy balance, glucose metabolism, and low-grade inflammation associated with obesity and related metabolic disorders. Recently, we defined gut microbiota-derived lipopolysaccharide (LPS) (and metabolic endotoxemia) as a factor involved in the onset and progression of inflammation and metabolic diseases. In this review, we discuss mechanisms involved in the development of metabolic endotoxemia such as the gut permeability. We also discuss our latest discoveries demonstrating a link between the gut microbiota, endocannabinoid system tone, leptin resistance, gut peptides (glucagon-like peptide-1 and -2), and metabolic features. Finally, we will introduce the role of the gut microbiota in specific dietary treatments (prebiotics and probiotics) and surgical interventions (gastric bypass).


Subject(s)
Diabetes Mellitus, Type 2/etiology , Gastrointestinal Tract/microbiology , Inflammation/etiology , Metagenome , Obesity/complications , Obesity/pathology , Endotoxins/toxicity , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...