Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marc-Emmanuel Dumas is active.

Publication


Featured researches published by Marc-Emmanuel Dumas.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice

Marc-Emmanuel Dumas; Richard H. Barton; Ayo Toye; Olivier Cloarec; Christine Blancher; Alice R. Rothwell; Jane Fearnside; Roger Tatoud; Veronique Blanc; John C. Lindon; Steve Chappell Mitchell; Elaine Holmes; Mark McCarthy; James Scott; Dominique Gauguier; Jeremy K. Nicholson

Here, we study the intricate relationship between gut microbiota and host cometabolic phenotypes associated with dietary-induced impaired glucose homeostasis and nonalcoholic fatty liver disease (NAFLD) in a mouse strain (129S6) known to be susceptible to these disease traits, using plasma and urine metabotyping, achieved by 1H NMR spectroscopy. Multivariate statistical modeling of the spectra shows that the genetic predisposition of the 129S6 mouse to impaired glucose homeostasis and NAFLD is associated with disruptions of choline metabolism, i.e., low circulating levels of plasma phosphatidylcholine and high urinary excretion of methylamines (dimethylamine, trimethylamine, and trimethylamine-N-oxide), coprocessed by symbiotic gut microbiota and mammalian enzyme systems. Conversion of choline into methylamines by microbiota in strain 129S6 on a high-fat diet reduces the bioavailability of choline and mimics the effect of choline-deficient diets, causing NAFLD. These data also indicate that gut microbiota may play an active role in the development of insulin resistance.


Molecular Systems Biology | 2007

A top‐down systems biology view of microbiome‐mammalian metabolic interactions in a mouse model

François-Pierre Martin; Marc-Emmanuel Dumas; Yulan Wang; Cristina Legido-Quigley; Ivan K. S. Yap; Huiru Tang; Severine Zirah; Gerard M. Murphy; Olivier Cloarec; John C. Lindon; Norbert Sprenger; Laurent B. Fay; Sunil Kochhar; Peter J. van Bladeren; Elaine Holmes; Jeremy K. Nicholson

Symbiotic gut microorganisms (microbiome) interact closely with the mammalian hosts metabolism and are important determinants of human health. Here, we decipher the complex metabolic effects of microbial manipulation, by comparing germfree mice colonized by a human baby flora (HBF) or a normal flora to conventional mice. We perform parallel microbiological profiling, metabolic profiling by 1H nuclear magnetic resonance of liver, plasma, urine and ileal flushes, and targeted profiling of bile acids by ultra performance liquid chromatography–mass spectrometry and short‐chain fatty acids in cecum by GC‐FID. Top‐down multivariate analysis of metabolic profiles reveals a significant association of specific metabotypes with the resident microbiome. We derive a transgenomic graph model showing that HBF flora has a remarkably simple microbiome/metabolome correlation network, impacting directly on the hosts ability to metabolize lipids: HBF mice present higher ileal concentrations of tauro‐conjugated bile acids, reduced plasma levels of lipoproteins but higher hepatic triglyceride content associated with depletion of glutathione. These data indicate that the microbiome modulates absorption, storage and the energy harvest from the diet at the systems level.


Gut | 2016

Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology

Maria Carlota Dao; Amandine Everard; Judith Aron-Wisnewsky; Nataliya Sokolovska; Edi Prifti; Eric O Verger; Brandon D. Kayser; Florence Levenez; Julien Chilloux; Lesley Hoyles; Marc-Emmanuel Dumas; Salwa Rizkalla; Joël Doré; Patrice D. Cani; Karine Clément

Objective Individuals with obesity and type 2 diabetes differ from lean and healthy individuals in their abundance of certain gut microbial species and microbial gene richness. Abundance of Akkermansia muciniphila, a mucin-degrading bacterium, has been inversely associated with body fat mass and glucose intolerance in mice, but more evidence is needed in humans. The impact of diet and weight loss on this bacterial species is unknown. Our objective was to evaluate the association between faecal A. muciniphila abundance, faecal microbiome gene richness, diet, host characteristics, and their changes after calorie restriction (CR). Design The intervention consisted of a 6-week CR period followed by a 6-week weight stabilisation diet in overweight and obese adults (N=49, including 41 women). Faecal A. muciniphila abundance, faecal microbial gene richness, diet and bioclinical parameters were measured at baseline and after CR and weight stabilisation. Results At baseline A. muciniphila was inversely related to fasting glucose, waist-to-hip ratio and subcutaneous adipocyte diameter. Subjects with higher gene richness and A. muciniphila abundance exhibited the healthiest metabolic status, particularly in fasting plasma glucose, plasma triglycerides and body fat distribution. Individuals with higher baseline A. muciniphila displayed greater improvement in insulin sensitivity markers and other clinical parameters after CR. These participants also experienced a reduction in A. muciniphila abundance, but it remained significantly higher than in individuals with lower baseline abundance. A. muciniphila was associated with microbial species known to be related to health. Conclusions A. muciniphila is associated with a healthier metabolic status and better clinical outcomes after CR in overweight/obese adults. The interaction between gut microbiota ecology and A. muciniphila warrants further investigation. Trial registration number NCT01314690.


Mbio | 2011

Colonization-Induced Host-Gut Microbial Metabolic Interaction

Sandrine P. Claus; S.L. Ellero; Bernard Berger; Lutz Krause; Anne Bruttin; J. Molina; Alain Paris; Elizabeth J. Want; I. de Waziers; Olivier Cloarec; Selena E. Richards; Yulan Wang; Marc-Emmanuel Dumas; Alastair B. Ross; Serge Rezzi; Sunil Kochhar; P.J. van Bladeren; John C. Lindon; Edward C. Holmes; Jeremy K. Nicholson

ABSTRACT The gut microbiota enhances the host’s metabolic capacity for processing nutrients and drugs and modulate the activities of multiple pathways in a variety of organ systems. We have probed the systemic metabolic adaptation to gut colonization for 20 days following exposure of axenic mice (n = 35) to a typical environmental microbial background using high-resolution 1H nuclear magnetic resonance (NMR) spectroscopy to analyze urine, plasma, liver, kidney, and colon (5 time points) metabolic profiles. Acquisition of the gut microbiota was associated with rapid increase in body weight (4%) over the first 5 days of colonization with parallel changes in multiple pathways in all compartments analyzed. The colonization process stimulated glycogenesis in the liver prior to triggering increases in hepatic triglyceride synthesis. These changes were associated with modifications of hepatic Cyp8b1 expression and the subsequent alteration of bile acid metabolites, including taurocholate and tauromuricholate, which are essential regulators of lipid absorption. Expression and activity of major drug-metabolizing enzymes (Cyp3a11 and Cyp2c29) were also significantly stimulated. Remarkably, statistical modeling of the interactions between hepatic metabolic profiles and microbial composition analyzed by 16S rRNA gene pyrosequencing revealed strong associations of the Coriobacteriaceae family with both the hepatic triglyceride, glucose, and glycogen levels and the metabolism of xenobiotics. These data demonstrate the importance of microbial activity in metabolic phenotype development, indicating that microbiota manipulation is a useful tool for beneficially modulating xenobiotic metabolism and pharmacokinetics in personalized health care. IMPORTANCE Gut bacteria have been associated with various essential biological functions in humans such as energy harvest and regulation of blood pressure. Furthermore, gut microbial colonization occurs after birth in parallel with other critical processes such as immune and cognitive development. Thus, it is essential to understand the bidirectional interaction between the host metabolism and its symbionts. Here, we describe the first evidence of an in vivo association between a family of bacteria and hepatic lipid metabolism. These results provide new insights into the fundamental mechanisms that regulate host-gut microbiota interactions and are thus of wide interest to microbiological, nutrition, metabolic, systems biology, and pharmaceutical research communities. This work will also contribute to developing novel strategies in the alteration of host-gut microbiota relationships which can in turn beneficially modulate the host metabolism. Gut bacteria have been associated with various essential biological functions in humans such as energy harvest and regulation of blood pressure. Furthermore, gut microbial colonization occurs after birth in parallel with other critical processes such as immune and cognitive development. Thus, it is essential to understand the bidirectional interaction between the host metabolism and its symbionts. Here, we describe the first evidence of an in vivo association between a family of bacteria and hepatic lipid metabolism. These results provide new insights into the fundamental mechanisms that regulate host-gut microbiota interactions and are thus of wide interest to microbiological, nutrition, metabolic, systems biology, and pharmaceutical research communities. This work will also contribute to developing novel strategies in the alteration of host-gut microbiota relationships which can in turn beneficially modulate the host metabolism.


Nature Medicine | 2017

A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice

Hubert Plovier; Amandine Everard; Céline Druart; Clara Depommier; Matthias Van Hul; Lucie Geurts; Julien Chilloux; Noora Ottman; Thibaut Duparc; Laeticia Lichtenstein; Antonis Myridakis; Nathalie M. Delzenne; Judith Klievink; Arnab Bhattacharjee; Kees C. H. van der Ark; Steven Aalvink; Laurent O. Martinez; Marc-Emmanuel Dumas; Dominique Maiter; Audrey Loumaye; Michel Hermans; Jean-Paul Thissen; Clara Belzer; Willem M. de Vos; Patrice D. Cani

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.


Genome Medicine | 2016

Impact of the gut microbiota on inflammation, obesity, and metabolic disease

Claire L. Boulangé; Ana Luísa Neves; Julien Chilloux; Jeremy K. Nicholson; Marc-Emmanuel Dumas

The human gut harbors more than 100 trillion microbial cells, which have an essential role in human metabolic regulation via their symbiotic interactions with the host. Altered gut microbial ecosystems have been associated with increased metabolic and immune disorders in animals and humans. Molecular interactions linking the gut microbiota with host energy metabolism, lipid accumulation, and immunity have also been identified. However, the exact mechanisms that link specific variations in the composition of the gut microbiota with the development of obesity and metabolic diseases in humans remain obscure owing to the complex etiology of these pathologies. In this review, we discuss current knowledge about the mechanistic interactions between the gut microbiota, host energy metabolism, and the host immune system in the context of obesity and metabolic disease, with a focus on the importance of the axis that links gut microbes and host metabolic inflammation. Finally, we discuss therapeutic approaches aimed at reshaping the gut microbial ecosystem to regulate obesity and related pathologies, as well as the challenges that remain in this area.


The American Journal of Clinical Nutrition | 2010

Metabolic profiling strategy for discovery of nutritional biomarkers: proline betaine as a marker of citrus consumption

Silke S. Heinzmann; Ian J. Brown; Queenie Chan; Magda Bictash; Marc-Emmanuel Dumas; Sunil Kochhar; Jeremiah Stamler; Elaine Holmes; Paul Elliott; Jeremy K. Nicholson

BACKGROUND New food biomarkers are needed to objectively evaluate the effect of diet on health and to check adherence to dietary recommendations and healthy eating patterns. OBJECTIVE We developed a strategy for food biomarker discovery, which combined nutritional intervention with metabolic phenotyping and biomarker validation in a large-scale epidemiologic study. DESIGN We administered a standardized diet to 8 individuals and established a putative urinary biomarker of fruit consumption by using (1)H nuclear magnetic resonance (NMR) spectroscopic profiling. The origin of the biomarker was confirmed by using targeted NMR spectroscopy of various fruit. Excretion kinetics of the biomarker were measured. The biomarker was validated by using urinary NMR spectra from UK participants of the INTERMAP (International Collaborative Study of Macronutrients, Micronutrients, and Blood Pressure) (n = 499) in which citrus consumption was ascertained from four 24-h dietary recalls per person. Finally, dietary patterns of citrus consumers (n = 787) and nonconsumers (n = 1211) were compared. RESULTS We identified proline betaine as a putative biomarker of citrus consumption. High concentrations were observed only in citrus fruit. Most proline betaine was excreted < or =14 h after a first-order excretion profile. Biomarker validation in the epidemiologic data showed a sensitivity of 86.3% for elevated proline betaine excretion in participants who reported citrus consumption and a specificity of 90.6% (P < 0.0001). In comparison with noncitrus consumers, citrus consumers had lower intakes of fats, lower urinary sodium-potassium ratios, and higher intakes of vegetable protein, fiber, and most micronutrients. CONCLUSION The biomarker identification and validation strategy has the potential to identify biomarkers for healthier eating patterns associated with a reduced risk of major chronic diseases. The trials were registered at clinicaltrials.gov as NCT01102049 and NCT01102062.


Nature Genetics | 2007

Direct quantitative trait locus mapping of mammalian metabolic phenotypes in diabetic and normoglycemic rat models

Marc-Emmanuel Dumas; Steven P. Wilder; Marie-Thérèse Bihoreau; Richard H. Barton; Jane Fearnside; Karène Argoud; Lisa D'Amato; Robert H. Wallis; Christine Blancher; Hector C. Keun; Dorrit Baunsgaard; James Scott; Ulla G. Sidelmann; Jeremy K. Nicholson; Dominique Gauguier

Characterizing the relationships between genomic and phenotypic variation is essential to understanding disease etiology. Information-dense data sets derived from pathophysiological, proteomic and transcriptomic profiling have been applied to map quantitative trait loci (QTLs). Metabolic traits, already used in QTL studies in plants, are essential phenotypes in mammalian genetics to define disease biomarkers. Using a complex mammalian system, here we show chromosomal mapping of untargeted plasma metabolic fingerprints derived from NMR spectroscopic analysis in a cross between diabetic and control rats. We propose candidate metabolites for the most significant QTLs. Metabolite profiling in congenic strains provided evidence of QTL replication. Linkage to a gut microbial metabolite (benzoate) can be explained by deletion of a uridine diphosphate glucuronosyltransferase. Mapping metabotypic QTLs provides a practical approach to understanding genome-phenotype relationships in mammals and may uncover deeper biological complexity, as extended genome (microbiome) perturbations that affect disease processes through transgenomic effects may influence QTL detection.


Molecular Systems Biology | 2014

Human metabolic profiles are stably controlled by genetic and environmental variation.

George Nicholson; Mattias Rantalainen; Anthony D. Maher; Jia V. Li; Daniel Malmodin; Kourosh R. Ahmadi; Johan H. Faber; Ingileif B. Hallgrímsdóttir; Amy Barrett; Henrik Toft; Maria Krestyaninova; Juris Viksna; Sudeshna Guha Neogi; Marc-Emmanuel Dumas; Ugis Sarkans; Bernard W. Silverman; Peter Donnelly; Jeremy K. Nicholson; Maxine Allen; Krina T. Zondervan; John C. Lindon; Tim D. Spector; Mark McCarthy; Elaine Holmes; Dorrit Baunsgaard; Christopher Holmes

1H Nuclear Magnetic Resonance spectroscopy (1H NMR) is increasingly used to measure metabolite concentrations in sets of biological samples for top‐down systems biology and molecular epidemiology. For such purposes, knowledge of the sources of human variation in metabolite concentrations is valuable, but currently sparse. We conducted and analysed a study to create such a resource. In our unique design, identical and non‐identical twin pairs donated plasma and urine samples longitudinally. We acquired 1H NMR spectra on the samples, and statistically decomposed variation in metabolite concentration into familial (genetic and common‐environmental), individual‐environmental, and longitudinally unstable components. We estimate that stable variation, comprising familial and individual‐environmental factors, accounts on average for 60% (plasma) and 47% (urine) of biological variation in 1H NMR‐detectable metabolite concentrations. Clinically predictive metabolic variation is likely nested within this stable component, so our results have implications for the effective design of biomarker‐discovery studies. We provide a power‐calculation method which reveals that sample sizes of a few thousand should offer sufficient statistical precision to detect 1H NMR‐based biomarkers quantifying predisposition to disease.


Journal of Chemometrics | 2010

The evolution of partial least squares models and related chemometric approaches in metabonomics and metabolic phenotyping

Judith M. Fonville; Selena E. Richards; Richard H. Barton; Claire L. Boulangé; Timothy M. D. Ebbels; Jeremy K. Nicholson; Elaine Holmes; Marc-Emmanuel Dumas

Metabonomics is a key element in systems biology, and with current analytical methods, generates vast amounts of quantitative or qualitative metabolic data. Understanding of the global function of the living organism can be achieved by integration of ‘omics’ approaches including metabonomics, genomics, transcriptomics and proteomics, increasing the complexity of the full data sets. Multivariate statistical approaches are well suited to extract the characterizing metabolic information associated with each level of dynamic process. In this review, we discuss techniques that have evolved from principal component analysis and partial least squares (PLS) methods with a focus on improved interpretation and modeling with respect to biomarker recovery and data visualization in the context of metabonomic applications. Visualization is of paramount importance to investigate complex metabolic signatures, the power and potential of which is illustrated with key papers. Recent improvements based on the removal of orthogonal variation are discussed in terms of interpretation enhancement, and are supported by relevant applications. Flexibility of PLS methods in general and of O‐PLS in particular allows implementation of derivative methods such as O2‐PLS, O‐PLS‐variance components, nonlinear methods, and batch modeling to improve analysis of complex data sets, which facilitates extraction of information related to subtle biological processes. These approaches can be used to address issues present in complex multi‐factorial data sets. Thus, we highlight the key advantages and limitations of the different latent variable applications for top‐down systems biology and assess the differences between the methods available. Copyright

Collaboration


Dive into the Marc-Emmanuel Dumas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jane Fearnside

Wellcome Trust Centre for Human Genetics

View shared research outputs
Researchain Logo
Decentralizing Knowledge