Network


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

Hotspot


Dive into the research topics where Alain Paris is active.

Publication


Featured researches published by Alain Paris.


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.


Hepatology | 2012

Low doses of bisphenol a induce gene expression related to lipid synthesis and trigger triglyceride accumulation in adult mouse liver

Alice Marmugi; Simon Ducheix; Frédéric Lasserre; Arnaud Polizzi; Alain Paris; Nathalie Priymenko; Justine Bertrand-Michel; Thierry Pineau; Hervé Guillou; Pascal G.P. Martin; Laila Mselli-Lakhal

Changes in lifestyle are suspected to have strongly influenced the current obesity epidemic. Based on recent experimental, clinical, and epidemiological work, it has been proposed that some food contaminants may exert damaging effects on endocrine and metabolic functions, thereby promoting obesity and associated metabolic diseases such as nonalcoholic fatty liver disease (NAFLD). In this work, we investigated the effect of one suspicious food contaminant, bisphenol A (BPA), in vivo. We used a transcriptomic approach in male CD1 mice exposed for 28 days to different doses of BPA (0, 5, 50, 500, and 5,000 μg/kg/day) through food contamination. Data analysis revealed a specific impact of low doses of BPA on the hepatic transcriptome, more particularly on genes involved in lipid synthesis. Strikingly, the effect of BPA on the expression of de novo lipogenesis followed a nonmonotonic dose‐response curve, with more important effects at lower doses than at the higher dose. In addition to lipogenic enzymes (Acc, Fasn, Scd1), the expression of transcription factors such as liver X Receptor, the sterol regulatory element binding protein‐1c, and the carbohydrate responsive element binding protein that govern the expression of lipogenic genes also followed a nonmonotonic dose‐response curve in response to BPA. Consistent with an increased fatty acid biosynthesis, determination of fat in the liver showed an accumulation of cholesteryl esters and of triglycerides. Conclusion: Our work suggests that exposure to low BPA doses may influence de novo fatty acid synthesis through increased expression of lipogenic genes, thereby contributing to hepatic steatosis. Exposure to such contaminants should be carefully examined in the etiology of metabolic diseases such as NAFLD and nonalcoholic steatohepatitis. (Hepatology 2012)


Atherosclerosis | 2009

1H NMR metabonomics can differentiate the early atherogenic effect of dairy products in hyperlipidemic hamsters

Jean-Charles Martin; Cécile Canlet; Bernadette Delplanque; Genevieve Agnani; Denis Lairon; Gaëlle Gottardi; Karima Bencharif; Daniel Gripois; Anissa Thaminy; Alain Paris

Diet is an important environmental factor modulating the onset of atherosclerosis. The aim of this study was to evaluate the effects of different dairy-based food products on early atherogenesis using both conventional and metabonomic approaches in hyperlipidemic hamsters. The hamsters received up to 200 g/kg of fat as anhydrous butter or cheese made from various milk fats or canola-based oil (CV), in addition to a non-atherogenic low-fat diet. Aortic cholesteryl ester loading was considered to be an early atherogenic point, and metabolic changes linked to atherogenesis were measured using plasma (1)H NMR-based metabonomics. The lowest atherogenicity was obtained with the plant-oil cheese diet, followed by the dairy fat cheese diet, while the greatest atherogenicity was observed with the butter diet (P<0.05). Disease outcome was correlated with conventional plasma biomarkers (total cholesterol, triglycerides, LDL cholesterol, R(2)=0.42-0.60). NMR plasma metabonomics selectively captured part of the diet-induced metabotypes correlated with aortic cholesteryl esters (R(2)=0.63). In these metabotypes, VLDL lipids, cholesterol, and N-acetylglycoproteins (R(2) range: 0.45-0.51) were the most positively correlated metabolites, whereas a multimetabolite response at 3.75 ppm, albumin lysyl residues, and trimethylamine-N-oxide were the most negatively correlated metabolites (R(2) range: 0.43-0.63) of the aortic cholesteryl esters. Collectively, these metabolites predicted 89% of atherogenic variability compared to the 60% predicted by total plasma cholesterol alone. In conclusion, we show that the food environment can modulate the atherogenic effect of dairy fat. This proof-of-principle study demonstrates the first use of plasma metabonomics for improving the prognosis of diet-induced atherogenesis, revealing novel potential disease biomarkers.


General Pharmacology-the Vascular System | 1998

Glucuronidation : A dual control

Françoise Guéraud; Alain Paris

1. Glucuronidation is a major detoxication process catalyzed by uridine diphosphate glucuronosyltransferases. 2. The amount of enzyme can be modulated by numerous foreign compounds, such as common chemical inducers already implicated in the induction of other detoxication enzymes. 3. Hormones such as thyroid hormones or growth hormone also are implicated in the control of glucuronidation. 4. Because glucuronidation enzymes (isozymes) are anchored in the endoplasmic reticulum membrane, with their active site likely being located on the lumenal side of the membrane, the membrane environment of these enzymes was shown to modulate their functional state as evaluated by the conjugating activity per enzymatic molecular unit. 5. In accord with a first, previously proposed model, it seems that this modulation can be attributed to different conformational states of the enzymes, depending on the physicochemical state of the membrane. 6. In accord with a second model, the membrane may act as a barrier between the enzymes and the cosubstrate UDP-glucuronic acid, which is a polar and charged molecule synthesized in the cytosol. This would imply a transporting process for this molecule through the reticulum membrane, which has been characterized in vitro and could be of importance in vivo. 7. Glucuronidation is under the control of a dual regulation, by means of a specific isozyme expression level and by the modulation of their functional state.


Hepatology | 2010

Loss of hepatocyte nuclear factor 1α function in human hepatocellular adenomas leads to aberrant activation of signaling pathways involved in tumorigenesis

Laura Pelletier; Sandra Rebouissou; Alain Paris; Estelle Rathahao-Paris; Elisabeth Perdu; Paulette Bioulac-Sage; Sandrine Imbeaud; Jessica Zucman-Rossi

Hepatocellular adenomas (HCAs) are benign liver tumors that usually develop in women who are taking oral contraceptives. Among these tumors, biallelic inactivating mutations of the hepatocyte nuclear factor 1α (HNF1A) transcription factor have been frequently identified and in rare cases of hepatocellular carcinomas developed in noncirrhotic liver. Because HNF1A meets the genetic criteria of a tumor suppressor gene, we aimed to elucidate the tumorigenic mechanisms related to HNF1α inactivation in hepatocytes. We searched for signaling pathways aberrantly activated in human HNF1A‐mutated HCA (H‐HCA) using a genome‐wide transcriptome analysis comparing five H‐HCA with four normal livers. We validated the main pathways by quantitative reverse transcription polymerase chain reaction (RT‐PCR) and western blotting in a large series of samples. Then, we assessed the role of HNF1α in the observed deregulations in hepatocellular cell models (HepG2 and Hep3B) by silencing its endogenous expression using small interfering RNA. Along with the previously described induction of glycolysis and lipogenesis, H‐HCA also displayed overexpression of several genes encoding growth factor receptors, components of the translation machinery, cell cycle, and angiogenesis regulators, with, in particular, activation of the mammalian target of rapamycin (mTOR) pathway. Moreover, estradiol detoxification activities were shut down, suggesting a hypersensitivity of H‐HCA to estrogenic stimulation. In the cell model, inhibition of HNF1α recapitulated most of these identified transcriptional deregulations, demonstrating that they were related to HNF1α inhibition. Conclusion: H‐HCA showed a combination of alterations related to HNF1α inactivation that may cooperate to promote tumor development. Interestingly, mTOR appears as a potential new attractive therapeutic target for treatment of this group of HCAs. (HEPATOLOGY 2009.)


PLOS ONE | 2011

Immunological and Metabolomic Impacts of Administration of Cry1Ab Protein and MON 810 Maize in Mouse

Karine Adel-Patient; Valeria Guimarães; Alain Paris; Marie-Françoise Drumare; Sandrine Ah-Leung; Patricia Lamourette; Marie-Claire Nevers; Cécile Canlet; Jérôme Molina; Hervé Bernard; Christophe Créminon; Jean-Michel Wal

We have investigated the immunological and metabolomic impacts of Cry1Ab administration to mice, either as a purified protein or as the Cry1Ab-expressing genetically modified (GM) MON810 maize. Humoral and cellular specific immune responses induced in BALB/cJ mice after intra-gastric (i.g.) or intra-peritoneal (i.p.) administration of purified Cry1Ab were analyzed and compared with those induced by proteins of various immunogenic and allergic potencies. Possible unintended effects of the genetic modification on the pattern of expression of maize natural allergens were studied using IgE-immunoblot and sera from maize-allergic patients. Mice were experimentally sensitized (i.g. or i.p. route) with protein extracts from GM or non-GM maize, and then anti-maize proteins and anti-Cry1Ab–induced immune responses were analyzed. In parallel, longitudinal metabolomic studies were performed on the urine of mice treated via the i.g. route. Weak immune responses were observed after i.g. administration of the different proteins. Using the i.p. route, a clear Th2 response was observed with the known allergenic proteins, whereas a mixed Th1/Th2 immune response was observed with immunogenic protein not known to be allergenic and with Cry1Ab. This then reflects protein immunogenicity in the BALB/c Th2-biased mouse strain rather than allergenicity. No difference in natural maize allergen profiles was evidenced between MON810 and its non-GM comparator. Immune responses against maize proteins were quantitatively equivalent in mice treated with MON810 vs the non-GM counterpart and no anti-Cry1Ab–specific immune response was detected in mice that received MON810. Metabolomic studies showed a slight “cultivar” effect, which represented less than 1% of the initial metabolic information. Our results confirm the immunogenicity of purified Cry1Ab without evidence of allergenic potential. Immunological and metabolomic studies revealed slight differences in mouse metabolic profiles after i.g. administration of MON810 vs its non-GM counterpart, but no significant unintended effect of the genetic modification on immune responses was seen.


Toxicology and Applied Pharmacology | 2009

Identification of potential mechanisms of toxicity after di-(2-ethylhexyl)-phthalate (DEHP) adult exposure in the liver using a systems biology approach

Alexandre Eveillard; F. Lasserre; Marie de Tayrac; Arnaud Polizzi; Sandrine P. Claus; Cécile Canlet; Laila Mselli-Lakhal; Gaëlle Gotardi; Alain Paris; Hervé Guillou; Pascal Martin; Thierry Pineau

Phthalates are industrial additives widely used as plasticizers. In addition to deleterious effects on male genital development, population studies have documented correlations between phthalates exposure and impacts on reproductive tract development and on the metabolic syndrome in male adults. In this work we investigated potential mechanisms underlying the impact of DEHP on adult mouse liver in vivo. A parallel analysis of hepatic transcript and metabolic profiles from adult mice exposed to varying DEHP doses was performed. Hepatic genes modulated by DEHP are predominantly PPARalpha targets. However, the induction of prototypic cytochrome P450 genes strongly supports the activation of additional NR pathways, including Constitutive Androstane Receptor (CAR). Integration of transcriptomic and metabonomic profiles revealed a correlation between the impacts of DEHP on genes and metabolites related to heme synthesis and to the Rev-erbalpha pathway that senses endogenous heme level. We further confirmed the combined impact of DEHP on the hepatic expression of Alas1, a critical enzyme in heme synthesis and on the expression of Rev-erbalpha target genes involved in the cellular clock and in energy metabolism. This work shows that DEHP interferes with hepatic CAR and Rev-erbalpha pathways which are both involved in the control of metabolism. The identification of these new hepatic pathways targeted by DEHP could contribute to metabolic and endocrine disruption associated with phthalate exposure. Gene expression profiles performed on microdissected testis territories displayed a differential responsiveness to DEHP. Altogether, this suggests that impacts of DEHP on adult organs, including testis, could be documented and deserve further investigations.


Apmis | 2001

Assessment of estradiol and its metabolites in meat

Daniel Maume; Yoann Deceuninck; Karinne Pouponneau; Alain Paris; Bruno Le Bizec; François Andre

Most studies related to research on steroids in main edible tissues (muscle, liver or kidney) have focused on measurement of parent or major metabolite residues. In order to evaluate the estradiol content in bovine edible tissues, a multi‐step extraction procedure was developed in conjunction with parallel metabolism studies of [14C]‐17β‐estradiol in cattle (1–2). Various classes of free estradiol and conjugates were separated: estradiol −17β and −17α, estradiol‐ 17‐fatty acid esters, estradiol 17‐glycoside, estradiol 3‐glucuronide, estradiol‐17‐glycoside and 3‐ glucuronide (diconjugates) were separated. No sulphates conjugated forms have been found at the detection level of the method. The quantification was realized by calibration with deuterated 17β −estradiol −d3 standard and was validated at the ng · kg−1(ppt) level. Muscle, liver, kidney and fat samples from control or Revalor S® single (licensed implantation) or multi‐implanted steers have been assayed.


Journal of Animal Science | 2012

Phenotypic prediction based on metabolomic data for growing pigs from three main European breeds.

Florian Rohart; Alain Paris; B. Laurent; Cécile Canlet; Jérôme Molina; Marie-José Mercat; Thierry Tribout; Nelly Muller; Nathalie Iannuccelli; Laurence Liaubet; Denis Milan; M. San Cristobal

Predicting phenotypes is a statistical and biotechnical challenge, both in medicine (predicting an illness) and animal breeding (predicting the carcass economical value on a young living animal). High-throughput fine phenotyping is possible using metabolomics, which describes the global metabolic status of an individual, and is the closest to the terminal phenotype. The purpose of this work was to quantify the prediction power of metabolomic profiles for commonly used production phenotypes from a single blood sample from growing pigs. Several statistical approaches were investigated and compared on the basis of cross validation: raw data vs. signal preprocessing (wavelet transformation), with a single-feature selection method. The best results in terms of prediction accuracy were obtained when data were preprocessed using wavelet transformations on the Daubechies basis. The phenotypes related to meat quality were not well predicted because the blood sample was taken some time before slaughter, and slaughter is known to have a strong influence on these traits. By contrast, phenotypes of potential economic interest (e.g., lean meat percentage and ADFI) were well predicted (R(2) = 0.7; P < 0.0001) using metabolomic data.


Steroids | 2005

In vitro aromatic bioactivation of the weak estrogen E2α and genesis of DNA adducts

Virginie Rizzati; Estelle Rathahao; Laurence Gamet-Payrastre; Georges Delous; Isabelle Jouanin; Françoise Guéraud; Alain Paris

Abstract Specific A-ring hydroxylated metabolites of 17β-estrogens are known to be endogenous pro-carcinogens, more particularly the 4-hydroxylated forms of estrogens produced by cytochrome P4501B1. In this study, we investigated whether estradiol-17α, the main hepatic residue of estradiol-17β in cattle treated for anabolic purposes with estradiol containing implants, could be significantly metabolized by human cells, and whether its aromatic metabolites could induce the formation of DNA adducts as estradiol-17β and estrone do. First, using a human kidney adenocarcinoma cell line, which expresses specifically the cytochrome P4501B1, we showed that estradiol-17α is bioactivated into a mixture of 2- and 4-catechol estrogens leading to the corresponding methoxyestrogens unambiguously identified by LC–APCI–MS/MS. We then demonstrated that the 2- and 4-hydroxylated derivatives of estradiol-17α incubated under oxidative conditions with calf thymus DNA gave stable DNA adducts and abasic sites, respectively. From these results, we can consider that human cells expressing CYP1B1-dependent hydroxylation activities metabolize estradiol-17α at the same magnitude as estradiol-17β and estrone, and that in oxidative conditions, the resulting aromatic metabolites can lead to the formation of both stable and unstable DNA adducts.

Collaboration


Dive into the Alain Paris's collaboration.

Top Co-Authors

Avatar

Cécile Canlet

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Laurent Debrauwer

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Estelle Rathahao

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laurence Liaubet

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Jérôme Molina

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Pascal Martin

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

François Andre

École Normale Supérieure

View shared research outputs
Top Co-Authors

Avatar

Isabelle Jouanin

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Nathalie Iannuccelli

Institut national de la recherche agronomique

View shared research outputs
Researchain Logo
Decentralizing Knowledge