Network


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

Hotspot


Dive into the research topics where Matthias Van Hul is active.

Publication


Featured researches published by Matthias Van Hul.


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.


Nature Communications | 2014

Intestinal epithelial MyD88 is a sensor switching host metabolism towards obesity according to nutritional status

Amandine Everard; Lucie Geurts; Robert Caesar; Matthias Van Hul; Sébastien Matamoros; Thibaut Duparc; Raphaël G P Denis; Perrine M. Cochez; Florian Pierard; Julien Castel; Laure B. Bindels; Hubert Plovier; Sylvie Robine; Giulio G. Muccioli; Jean-Christophe Renauld; Laure Dumoutier; Nathalie M. Delzenne; Serge Luquet; Fredrik Bäckhed; Patrice D. Cani

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.


Nature Communications | 2015

Adipose tissue NAPE-PLD controls fat mass development by altering the browning process and gut microbiota

Lucie Geurts; Amandine Everard; Matthias Van Hul; Ahmed Essaghir; Thibaut Duparc; Sébastien Matamoros; Hubert Plovier; Julien Castel; Raphaël G P Denis; Marie Bergiers; Céline Druart; Mireille Alhouayek; Nathalie M. Delzenne; Giulio G. Muccioli; Jean-Baptiste Demoulin; Serge Luquet; Patrice D. Cani

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.


Current Opinion in Biotechnology | 2015

Novel opportunities for next-generation probiotics targeting metabolic syndrome

Patrice D. Cani; Matthias Van Hul

Various studies have described the beneficial effects of specific bacteria on the characteristics of metabolic syndrome. Intestinal microbiota might therefore represent a modifiable trait for translational intervention to improve the metabolic profiles of obese and type 2 diabetic patients. However, identifying potential probiotic strains that can effectively colonize the gastrointestinal tract and significantly affect host metabolism has been challenging. This review aims to summarize the notable advances and contributions in the field that may prove useful for identifying next-generation probiotics that target metabolic syndrome and its related disorders.


Molecular Nutrition & Food Research | 2017

Rhubarb extract prevents hepatic inflammation induced by acute alcohol intake, an effect related to the modulation of the gut microbiota.

Audrey M. Neyrinck; Usune Etxeberria; Bernard Taminiau; Georges Daube; Matthias Van Hul; Amandine Everard; Patrice D. Cani; Laure B. Bindels; Nathalie M. Delzenne

SCOPE Binge consumption of alcohol is an alarming global health problem. Acute ethanol intoxication is characterized by hepatic inflammation and oxidative stress, which could be promoted by gut barrier function alterations. In this study, we have tested the hypothesis of the hepatoprotective effect of rhubarb extract in a mouse model of binge drinking and we explored the contribution of the gut microbiota in the related metabolic effects. METHODS AND RESULTS Mice were fed a control diet supplemented with or without 0.3% rhubarb extract for 17 days and were necropsied 6 h after an alcohol challenge. Supplementation with rhubarb extract changed the microbial ecosystem (assessed by 16S rDNA pyrosequencing) in favor of Akkermansia muciniphila and Parabacteroides goldsteinii. Furthermore, it improved alcohol-induced hepatic injury, downregulated key markers of both inflammatory and oxidative stresses in the liver tissue, without affecting significantly steatosis. In the gut, rhubarb supplementation increased crypt depth, tissue weight, and the expression of antimicrobial peptides. CONCLUSIONS These findings suggest that some bacterial genders involved in gut barrier function, are promoted by phytochemicals present in rhubarb extract, and could therefore be involved in the modulation of the susceptibility to hepatic diseases linked to acute alcohol consumption.


Gut | 2017

Hepatocyte MyD88 affects bile acids, gut microbiota and metabolome contributing to regulate glucose and lipid metabolism

Thibaut Duparc; Hubert Plovier; Vannina G. Marrachelli; Matthias Van Hul; Ahmed Essaghir; Marcus Ståhlman; Sébastien Matamoros; Lucie Geurts; Mercedes M. Pardo-Tendero; Céline Druart; Nathalie M. Delzenne; Jean-Baptiste Demoulin; Schalk Van der Merwe; Jos van Pelt; Fredrik Bäckhed; Daniel Monleón; Amandine Everard; Patrice D. Cani

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.


American Journal of Physiology-endocrinology and Metabolism | 2018

Reduced obesity, diabetes and steatosis upon cinnamon and grape pomace are associated with changes in gut microbiota and markers of gut barrier.

Matthias Van Hul; Lucie Geurts; Hubert Plovier; Céline Druart; Amandine Everard; Marcus Ståhlman; Moez Rhimi; Kleopatra Chira; Pierre-Louis Teissedre; Nathalie M. Delzenne; Emmanuelle Maguin; Angèle Guilbot; Amandine Brochot; Philippe Gérard; Fredrik Bäckhed; Patrice D. Cani

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.


Journal of Nutritional Biochemistry | 2017

A polyphenolic extract from green tea leaves activates fat browning in high-fat-diet-induced obese mice

Audrey M. Neyrinck; Laure B. Bindels; Lucie Geurts; Matthias Van Hul; Patrice D. Cani; Nathalie M. Delzenne

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.


Oncotarget | 2018

Increased gut permeability in cancer cachexia: mechanisms and clinical relevance

Laure B. Bindels; Audrey M. Neyrinck; Audrey Loumaye; Emilie Catry; Hannah Walgrave; Claire Cherbuy; Sophie Leclercq; Matthias Van Hul; Hubert Plovier; Barbara D. Pachikian; Luis G. Bermúdez-Humarán; Philippe Langella; Patrice D. Cani; Jean-Paul Thissen; Nathalie M. Delzenne

Intestinal disorders often occur in cancer patients, in association with body weight loss, and this alteration is commonly attributed to the chemotherapy. Here, using a mouse model of cancer cachexia induced by ectopic transplantation of C26 cancer cells, we discovered a profound alteration in the gut functions (gut permeability, epithelial turnover, gut immunity, microbial dysbiosis) independently of any chemotherapy. These alterations occurred independently of anorexia and were driven by interleukin 6. Gut dysfunction was found to be resistant to treatments with an anti-inflammatory bacterium (Faecalibacterium prausnitzii) or with gut peptides involved in intestinal cell renewal (teduglutide, a glucagon-like peptide 2 analogue). The translational value of our findings was evaluated in 152 colorectal and lung cancer patients with or without cachexia. The serum level of the lipopolysaccharide-binding protein, often presented as a reflection of the bacterial antigen load, was not only increased in cachectic mice and cancer patients, but also strongly correlated with the serum IL-6 level and predictive of death and cachexia occurrence in these patients. Altogether, our data highlight profound alterations of the intestinal homeostasis in cancer cachexia occurring independently of any chemotherapy and food intake reduction, with potential relevance in humans. In addition, we point out the lipopolysaccharide-binding protein as a new biomarker of cancer cachexia related to gut dysbiosis.


NMR in Biomedicine | 2017

Combined endogenous MR biomarkers to predict basal tumor oxygenation and response to hyperoxic challenge

Thanh-Trang Cao-Pham; Nicolas Joudiou; Matthias Van Hul; Caroline Bouzin; Patrice D. Cani; Bernard Gallez; Bénédicte F. Jordan

Hypoxia is a common feature of solid tumors, which translates into increased angiogenesis, malignant phenotype cell selection, change in gene expression and greater resistance to radiotherapy and chemotherapy. Therefore, there is a need for markers of hypoxia to stratify patients, in order to personalize treatment to improve therapeutic outcome. However, no modality has yet been validated for the screening of hypoxia in routine clinical practice. Magnetic resonance imaging (MRI) R1 and R2* relaxation parameters are sensitive to tissue oxygenation: R1 is sensitive to dissolved oxygen and R2* is sensitive to intravascular deoxyhemoglobin content. Two rat tumor models with distinct levels of hypoxia, 9L–glioma and rhabdomyosarcoma, were imaged for R1 and R2* under air and carbogen (95% O2 and 5% CO2) breathing conditions. It was observed that the basal tumor oxygenation level had an impact on the amplitude of response to carbogen in the vascular compartment (R2*), but not in the tissue compartment (R1). In addition, the change in tissue oxygenation estimated by ΔR1 correlated with the change in vascular oxygenation estimated by ΔR2*, which is consistent with an increase in oxygen supply generating an elevated tumor pO2. At the intra‐tumoral level, we identified four types of voxel to which a hypoxic feature was attributed (mild hypoxia, severe hypoxia, normoxia and vascular steal), depending on the carbogen‐induced change in R1 and R2* values for each voxel. The results showed that 9L–gliomas present more normoxic fractions, whereas rhabdomyosarcomas present more hypoxic fractions, which is in accordance with a previous study using 18F–fluoroazomycin arabinoside (18F–FAZA) and electron paramagnetic resonance (EPR) oximetry. The response of the combined endogenous MRI contrasts to carbogen challenge could be a useful tool to predict different tumor hypoxic fractions.

Collaboration


Dive into the Matthias Van Hul's collaboration.

Top Co-Authors

Avatar

Patrice D. Cani

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Nathalie M. Delzenne

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Amandine Everard

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Hubert Plovier

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Lucie Geurts

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Céline Druart

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Audrey M. Neyrinck

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Laure B. Bindels

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Sébastien Matamoros

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Thibaut Duparc

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge