16 research outputs found

    Obesity promotes fumonisin B1 hepatotoxicity

    Get PDF
    Obesity, which is a worldwide public health issue, is associated with chronic inflammation that contribute to long-term complications, including insulin resistance, type 2 diabetes and non-alcoholic fatty liver disease. We hypothesized that obesity may also influence the sensitivity to food contaminants, such as fumonisin B1 (FB1), a mycotoxin produced mainly by the Fusarium verticillioides. FB1, a common contaminant of corn, is the most abundant and best characterized member of the fumonisins family. We investigated whether diet-induced obesity could modulate the sensitivity to oral FB1 exposure, with emphasis on gut health and hepatotoxicity. Thus, metabolic effects of FB1 were assessed in obese and non-obese male C57BL/6J mice. Mice received a high-fat diet (HFD) or normal chow diet (CHOW) for 15 weeks. Then, during the last three weeks, mice were exposed to these diets in combination or not with FB1 (10 mg/kg body weight/day) through drinking water. As expected, HFD feeding induced significant body weight gain, increased fasting glycemia, and hepatic steatosis. Combined exposure to HFD and FB1 resulted in body weight loss and a decrease in fasting blood glucose level. This co-exposition also induces gut dysbiosis, an increase in plasma FB1 level, a decrease in liver weight and hepatic steatosis. Moreover, plasma transaminase levels were significantly increased and associated with liver inflammation in HFD/FB1-treated mice. Liver gene expression analysis revealed that the combined exposure to HFD and FB1 was associated with reduced expression of genes involved in lipogenesis and increased expression of immune response and cell cycle-associated genes. These results suggest that, in the context of obesity, FB1 exposure promotes gut dysbiosis and severe liver inflammation. To our knowledge, this study provides the first example of obesity-induced hepatitis in response to a food contaminant.L.D. PhD was supported by the INRAE Animal Health department. This work was also supported by grants from the French National Research Agency (ANR) Fumolip (ANR-16-CE21-0003) and the Hepatomics FEDER program of Région Occitanie. We thank Prof Wentzel C. Gelderblom for generously providing the FB1 and for his interest and support in our project. B.C. laboratory is supported by a Starting Grant from the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation program (grant agreement No. ERC-2018-StG- 804135), a Chaire d'Excellence from IdEx Université de Paris - ANR-18-IDEX-0001, an Innovator Award from the Kenneth Rainin Foundation, an ANR grant EMULBIONT ANR-21-CE15-0042-01 and the national program “Microbiote” from INSERM. We thank Anexplo (Genotoul, Toulouse) for their excellent work on plasma biochemistry. Neutral Lipids MS and NMR experiments were performed with instruments in the Metatoul-AXIOM platform. Sphingolipid MS analysis were performed with instruments in the RUBAM platform. The FB1 plasma levels were determined using an UPLC-MS/MS instrument part of the Ghent University MSsmall expertise centre for advanced mass spectrometry analysis of small organic molecules. We thank Elodie Rousseau-Bacquié and all members of the EZOP staff for their assistance in the animal facility. We are very grateful to Talal al Saati for histology analyses and review, and we thank all members of the US006/CREFRE staff at the histology facility and the Genom'IC platforms (INSERM U1016, Paris, France) for their expertise.Peer reviewe

    Rôle dimorphique du récepteur nucléaire CAR dans la régulation de l'homéostasie énergétique et des perturbations métaboliques induites par un mélange de pesticides

    Get PDF
    The incidence of metabolic diseases has steadily increased in recent decades reaching epidemic proportions. It is conventionally accepted that their main cause is related to a diet rich in fats and sugar and/or a sedentary lifestyle that can be aggravated by certain genetic polymorphisms. Chemical contaminants in our environment are also suspected to contribute to the development of these metabolic disorders by disrupting the energy balance of organisms. Several studies report the role of nuclear receptors as mediators of these metabolic effects induced by environmental contaminants. As part of this PhD work, we investigated the role of the CAR nuclear receptor in the regulation of energy homeostasis and as a mediator of the metabolic effects induced by exposure to a mixture of pesticides. CAR is a key nuclear receptor for the detoxification system of compounds, whether exogenous or endogenous. Its role in energy metabolism has been studied mainly in male mice, but metabolic and detoxification functions are highly dependent on sex. The first objective of this work was to evaluate the consequences on the energy homeostasis of the deletion of the CAR nuclear receptor in male and female mice. These animals were followed over a period of more than one year and their phenotype was compared to that of non-invalidated mice for this receptor. The results show that the absence of CAR is very deleterious in males that develop obesity, diabetes and hepatic steatosis. CAR-/- females mice are protected from these disorders and even have better glucose tolerance. This protection is lifted by ovariectomy of these females suggesting a role of female sex hormones in their protection. Transcriptomic, metabolomic and lipidomic analysis are in agreement with this phenotypic change. The second objective of this work was to evaluate the in vivo metabolic consequences of chronic exposure to a mixture of pesticides present in the diet at presumed non-toxic doses. After one year of exposure to this mixture, male mice developed an overweight with an increase in their fat masses. This overweight was accompanied by glucose intolerance and hepatic steatosis. On the other hand, female mice showed fasting hyperglycemia, hepatic oxidative stress and a disturbance of urinary microbiota related to the intestinal microbiota. These results show for the first time an obesogenic and diabetogenic sex-dependent effect of exposure to a mixture of pesticides. We have also demonstrated a role of the CAR nuclear receptor in the sexual dimorphism observed following this exposure. All of this work provides causal links in favor of a relationship between environmental contaminants and sex-dependent health and a role of the nuclear receptor CAR in the effects observed. This raises the issue of gender and mixture in the risk assessment linked to exposure to environmental contaminants.L'incidence des pathologies métaboliques n'a de cesse d'augmenter au cours des dernières décennies atteignant des proportions épidémiques. Les facteurs génétiques, l'alimentation et/ou la sédentarité n'expliquent qu'en partie ce phénomène. Les contaminants environnementaux sont également suspectés être impliqués dans l'étiologie de ces perturbations. Plusieurs études rapportent l'implication des récepteurs nucléaires comme médiateur de ces désordres métaboliques. Dans le cadre de ces travaux nous nous sommes intéressés aux rôles du récepteur nucléaire CAR dans la régulation de l'homéostasie énergétique et comme médiateur des perturbations métaboliques induites suite à une exposition à un mélange de pesticides. CAR est un récepteur nucléaire clé du système de détoxification de composés, qu'ils soient de nature exogène ou endogène. Son rôle sur le métabolisme énergétique a été étudié principalement chez les souris mâles, or certaines fonctions métaboliques et de détoxification sont fortement dépendantes du sexe. Le premier objectif de ces travaux a consisté à évaluer les conséquences sur l'homéostasie énergétique de la délétion du récepteur nucléaire CAR (CAR-/-) chez des souris mâles et femelles au cours du vieillissement. Les résultats montrent que l'absence de CAR est délétère chez les mâles qui développent une obésité, un diabète et une stéatose hépatique. Les souris femelles CAR-/- sont protégées de ces troubles et présentent une amélioration de leur tolérance au glucose. Cependant la suppression de leurs hormones sexuelles grâce à la réalisation d'ovariectomies enlève totalement cette protection, suggérant un rôle majeur de ces hormones dans cette protection. Les analyses du transcriptome, lipidome et métabolome hépatique sont en accord avec ces données phénotypiques. Le deuxième objectif de ces travaux a consisté à évaluer les conséquences métaboliques d'une exposition chronique in vivo à un mélange de pesticides présent dans l'alimentation à des doses supposées non toxiques. Après un an d'exposition à ce mélange, les souris mâles développent un surpoids avec une augmentation de leurs masses adipeuses. Ce surpoids s'accompagne d'une intolérance au glucose et d'une stéatose hépatique. Les souris femelles présentent, en revanche une hyperglycémie à jeun, du stress oxydatif au niveau hépatique et une perturbation de métabolites urinaires liées au microbiote intestinal. Ces résultats montrent pour la première fois un effet obésogène et diabétogène dépendant du sexe d'une exposition à un mélange de pesticides. Nous avons également mis en évidence un rôle du récepteur nucléaire CAR dans le dimorphisme sexuel observé suite à cette exposition. L'ensemble de ces travaux apporte des liens de causalité en faveur d'une relation contaminants environnementaux et santé dépendant du sexe et un rôle du récepteur nucléaire CAR dans les effets observés. Cela soulève la question de la prise en considération du sexe et de l'effet mélange dans l'évaluation des risques pour la santé liée à l'exposition à des contaminants environnementaux

    Constitutive Androstane Receptor: A Peripheral and a Neurovascular Stress or Environmental Sensor

    No full text
    International audienceXenobiotic nuclear receptors (NR) are intracellular players involved in an increasing number of physiological processes. Examined and characterized in peripheral organs where they govern metabolic, transport and detoxification mechanisms, accumulating data suggest a functional expression of specific NR at the neurovascular unit (NVU). Here, we focus on the Constitutive Androstane Receptor (CAR), expressed in detoxifying organs such as the liver, intestines and kidneys. By direct and indirect activation, CAR is implicated in hepatic detoxification of xenobiotics, environmental contaminants, and endogenous molecules (bilirubin, bile acids). Importantly, CAR participates in physiological stress adaptation responses, hormonal and energy homeostasis due to glucose and lipid sensing. We next analyze the emerging evidence supporting a role of CAR in NVU cells including the blood-brain barrier (BBB), a key vascular interface regulating communications between the brain and the periphery. We address the emerging concept of how CAR may regulate specific P450 cytochromes at the NVU and the associated relevance to brain diseases. A clear understanding of how CAR engages during pathological conditions could enable new mechanistic, and perhaps pharmacological, entry-points within a peripheral-brain axis

    Regulation of hepatokine gene expression in response to fasting and feeding: Influence of PPARa and insulin-dependent signaling in hepatocytes

    No full text
    International audienceAim.-In hepatocyte, PPARa and insulin receptor (IR) are critical for the transcriptional responses to fasting and feeding, respectively. Here we analyzed the effects of the nutritional status (fasting vs feeding) on the expression of a large panel of hepatokines in hepatocyte-specific PPARa (Ppara hep−/−) and IR (IR hep−/−) null mice. Methods.-Ppara hep−/− , and IR hep−/− mice and their wild-type littermate were subjected to fasting or feeding metabolic challenges, and then analyzed for hepatokine gene expression. Experiments were conducted in mice of both sexes

    Activation of the Constitutive Androstane Receptor induces hepatic lipogenesis and regulates Pnpla3 gene expression in a LXR-independent way

    No full text
    International audienceThe Constitutive Androstane Receptor (CAR, NR1I3) has been newly described as a regulator of energy metabolism. A relevant number of studies using animal models of obesity suggest that CAR activation could be beneficial on the metabolic balance. However, this remains controversial and the underlying mechanisms are still unknown. This work aimed to investigate the effect of CAR activation on hepatic energy metabolism during physiological conditions, i.e. in mouse models not subjected to metabolic/nutritional stress. Gene expression profiling in the liver of CAR knockout and control mice on chow diet and treated with a CAR agonist highlighted CAR-mediated up-regulations of lipogenic genes, concomitant with neutral lipid accumulation. A strong CAR-mediated up-regulation of the patatin-like phospholipase domain-containing protein 3 (Pnpla3) was demonstrated. Pnpla3 is a gene whose polymorphism is associated with the pathogenesis of nonalcoholic fatty liver disease (NAFLD) development. This observation was confirmed in human hepatocytes treated with the antiepileptic drug and CAR activator, phenobarbital and in immortalized human hepatocytes treated with CITCO. Studying the molecular mechanisms controlling Pnpla3 gene expression, we demonstrated that CAR does not act by a direct regulation of Pnpla3 transcription or via the Liver X Receptor but may rather involve the transcription factor Carbohydrate Responsive Element-binding protein. These data provide new insights into the regulation by CAR of glycolytic and lipogenic genes and on pathogenesis of steatosis. This also raises the question concerning the impact of drugs and environmental contaminants in lipid-associated metabolic diseases

    Insights into the role of hepatocyte PPARα activity in response to fasting

    No full text
    The liver plays a central role in the regulation of fatty acid metabolism. Hepatocytes are highly sensitive to nutrients and hormones that drive extensive transcriptional responses. Nuclear hormone receptors are key transcription factors involved in this process. Among these factors, PPARα is a critical regulator of hepatic lipid catabolism during fasting.This study aimed to analyse the wide array of hepatic PPARα-dependent transcriptional responses during fasting. We compared gene expression in male mice with a hepatocyte specific deletion of PPARα and their wild-type littermates in the fed (ad libitum) and 24-h fasted states. Liver samples were acquired, and transcriptome and lipidome analyses were performed.Our data extended and confirmed the critical role of hepatocyte PPARα as a central for regulator of gene expression during starvation. Interestingly, we identified novel PPARα-sensitive genes, including Cxcl-10, Rab30, and Krt23. We also found that liver phospholipid remodelling was a novel fasting-sensitive pathway regulated by PPARα. These results may contribute to investigations on transcriptional control in hepatic physiology and underscore the clinical relevance of drugs that target PPARα in liver pathologies, such as non-alcoholic fatty liver disease

    Gene expression profiling reveals that PXR activation inhibits Hhepatic PPAR alpha activity and decreases FGF21 secretion in male C57Bl6/J mice

    No full text
    The pregnane X receptor (PXR) is the main nuclear receptor regulating the expression of xenobiotic-metabolizing enzymes and is highly expressed in the liver and intestine. Recent studies have highlighted its additional role in lipid homeostasis, however, the mechanisms of these regulations are not fully elucidated. We investigated the transcriptomic signature of PXR activation in the liver of adult wild-type vs. Pxr(-/-) C57Bl6/J male mice treated with the rodent specific ligand pregnenolone 16 alpha-carbonitrile (PCN). PXR activation increased liver triglyceride accumulation and significantly regulated the expression of 1215 genes, mostly xenobiotic-metabolizing enzymes. Among the down-regulated genes, we identified a strong peroxisome proliferator-activated receptor alpha (PPAR alpha) signature. Comparison of this signature with a list of fasting-induced PPAR alpha target genes confirmed that PXR activation decreased the expression of more than 25 PPAR alpha target genes, among which was the hepatokine fibroblast growth factor 21 (Fgf21). PXR activation abolished plasmatic levels of FGF21. We provide a comprehensive signature of PXR activation in the liver and identify new PXR target genes that might be involved in the steatogenic effect of PXR. Moreover, we show that PXR activation down-regulates hepatic PPAR alpha activity and FGF21 circulation, which could participate in the pleiotropic role of PXR in energy homeostasis

    Metabolic effects of a chronic dietary exposure to a low-dose oesticide cocktail in mice: Sexual dimorphism and role of the constitutive androstane receptor

    No full text
    International audienceEpidemiological evidence suggests a link between pesticide exposure and the development of metabolic diseases. However, most experimental studies have evaluated the metabolic effects of pesticides using individual molecules, often at nonrelevant doses or in combination with other risk factors such as high-fat diets. We aimed to evaluate, in mice, the metabolic consequences of chronic dietary exposure to a pesticide mixture at nontoxic doses, relevant to consumers' risk assessment. A mixture of six pesticides commonly used in France, i.e., boscalid, captan, chlorpyrifos, thiofanate, thiacloprid, and ziram, was incorporated in a standard chow at doses exposing mice to the tolerable daily intake (TDI) of each pesticide. Wild-type (WT) and constitutive androstane receptor-deficient (CAR-/-) male and female mice were exposed for 52 wk. We assessed metabolic parameters [body weight (BW), food and water consumption, glucose tolerance, urinary metabolome] throughout the experiment. At the end of the experiment, we evaluated liver metabolism (histology, transcriptomics, metabolomics, lipidomics) and pesticide detoxification using liquid chromatography-mass spectrometry (LC-MS). Compared to those fed control chow, WT male mice fed pesticide chow had greater BW gain and more adiposity. Moreover, these WT males fed pesticide chow exhibited characteristics of hepatic steatosis and glucose intolerance, which were not observed in those fed control chow. WT exposed female mice exhibited fasting hyperglycemia, higher reduced glutathione (GSH):oxidized glutathione (GSSG) liver ratio and perturbations of gut microbiota-related urinary metabolites compared to WT mice fed control chow. When we performed these experiments on CAR-/- mice, pesticide-exposed CAR-/- males did not exhibit BW gain or changes in glucose metabolism compared to the CAR-/- males fed control chow. Moreover, CAR-/- females fed pesticide chow exhibited pesticide toxicity with higher BWs and mortality rate compared to the CAR-/- females fed control chow. To our knowledge, we are the first to demonstrate a sexually dimorphic obesogenic and diabetogenic effect of chronic dietary exposure to a common mixture of pesticides at TDI levels, and to provide evidence for a partial role for CAR in an in vivo mouse model. This raises questions about the relevance of TDI for individual pesticides when present in a mixture
    corecore