16 results on '"Barbier, Olivier"'
Search Results
2. High-Fat Diet Modulates Hepatic Amyloid β and Cerebrosterol Metabolism in the Triple Transgenic Mouse Model of Alzheimer's Disease.
- Author
-
Bosoi CR, Vandal M, Tournissac M, Leclerc M, Fanet H, Mitchell PL, Verreault M, Trottier J, Virgili J, Tremblay C, Lippman HR, Bajaj JS, Barbier O, Marette A, and Calon F
- Subjects
- Alzheimer Disease etiology, Animals, Brain metabolism, Brain-Gut Axis physiology, Disease Models, Animal, Lipogenesis physiology, Mice, Mice, Obese, Mice, Transgenic, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Diet, High-Fat adverse effects, Hydroxycholesterols metabolism, Liver metabolism
- Abstract
Obesity and diabetes are strongly associated not only with fatty liver but also cognitive dysfunction. Moreover, their presence, particularly in midlife, is recognized as a risk factor for Alzheimer's disease (AD). AD, the most common cause of dementia, is increasingly considered as a metabolic disease, although underlying pathogenic mechanisms remain unclear. The liver plays a major role in maintaining glucose and lipid homeostasis, as well as in clearing the AD neuropathogenic factor amyloid-β (Aβ) and in metabolizing cerebrosterol, a cerebral-derived oxysterol proposed as an AD biomarker. We hypothesized that liver impairment induced by obesity contributes to AD pathogenesis. We show that the AD triple transgenic mouse model (3xTg-AD) fed a chow diet presents a hepatic phenotype similar to nontransgenic controls (NTg) at 15 months of age. A high-fat diet (HFD), started at the age of 6 months and continued for 9 months, until sacrifice, induced hepatic steatosis in NTg, but not in 3xTg-AD mice, whereas HFD did not induce changes in hepatic fatty acid oxidation, de novo lipogenesis, and gluconeogenesis. HFD-induced obesity was associated with a reduction of insulin-degrading enzyme, one of the main hepatic enzymes responsible for Aβ clearance. The hepatic rate of cerebrosterol glucuronidation was lower in obese 3xTg-AD than in nonobese controls ( P < 0.05) and higher compared with obese NTg ( P < 0.05), although circulating levels remained unchanged. Conclusion: Modulation of hepatic lipids, Aβ, and cerebrosterol metabolism in obese 3xTg-AD mice differs from control mice. This study sheds light on the liver-brain axis, showing that the chronic presence of NAFLD and changes in liver function affect peripheral AD features and should be considered during development of biomarkers or AD therapeutic targets., (© 2020 The Authors. Hepatology Communications published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases.)
- Published
- 2020
- Full Text
- View/download PDF
3. Effect of S-adenosyl-L-methionine on liver biochemistry and quality of life in patients with primary biliary cholangitis treated with ursodeoxycholic acid. A prospective, open label pilot study.
- Author
-
Wunsch E, Raszeja-Wyszomirska J, Barbier O, Milkiewicz M, Krawczyk M, and Milkiewicz P
- Subjects
- Adult, Aged, Alkaline Phosphatase blood, Biomarkers blood, Cholagogues and Choleretics adverse effects, Cholesterol blood, Fatigue etiology, Fatigue prevention & control, Female, Humans, Liver metabolism, Liver Cirrhosis, Biliary blood, Liver Cirrhosis, Biliary complications, Liver Cirrhosis, Biliary diagnosis, Middle Aged, Pilot Projects, Poland, Proof of Concept Study, Prospective Studies, Pruritus etiology, Pruritus prevention & control, S-Adenosylmethionine adverse effects, Surveys and Questionnaires, Time Factors, Treatment Outcome, Ursodeoxycholic Acid adverse effects, gamma-Glutamyltransferase blood, Cholagogues and Choleretics therapeutic use, Liver drug effects, Liver Cirrhosis, Biliary drug therapy, Quality of Life, S-Adenosylmethionine therapeutic use, Ursodeoxycholic Acid therapeutic use
- Abstract
Background and Aims: Chronic liver disease induces an acquired deficiency of S-adenosyl-L-methionine (SAMe) leading to impairment of detoxifying processes in the liver. Ursodeoxycholic acid (UDCA) represents the standard treatment in primary biliary cholangitis (PBC). As both compounds exert their hepatoprotective effects by different mechanisms, it is conceivable that when used together their effect might be additive. The aim of this study was to analyse the effect of SAMe supplementation on liver biochemistry and health-related quality of life (HRQoL) in patients with PBC, treated with UDCA., Methods: In this prospective pilot, proof of the principle, non-randomized and open label study we enrolled 24 patients with PBC treated with UDCA for at least 6 months. They had received both UDCA in a standard dose of 13-15 mg/kg b.w. and SAMe in the dose of 1200 mg daily over a period of 6 months. A group of 24 patients with PBC treated with UDCA served as control for liver biochemistry (Study registered on the platform ClinicalTrials.gov under ID: NCT02557360)., Results: We observed a significant decrease of ALP, GGT and total cholesterol in non-cirrhotic patients treated with SAMe. There was also a significant improvement of fatigue and pruritus in PBC-40 questionnaire and amelioration of anxiety in STAI 2 questionnaire in the SAMe group. Treatment with SAMe neither increased sulfation capacity of the liver nor had an effect on fibroblast growth factor-19 serum levels., Conclusions: Our pilot study demonstrates a positive effect of adding SAMe to UDCA in non-cirrhotic patients with PBC.
- Published
- 2018
- Full Text
- View/download PDF
4. Arctic berry extracts target the gut-liver axis to alleviate metabolic endotoxaemia, insulin resistance and hepatic steatosis in diet-induced obese mice.
- Author
-
Anhê FF, Varin TV, Le Barz M, Pilon G, Dudonné S, Trottier J, St-Pierre P, Harris CS, Lucas M, Lemire M, Dewailly É, Barbier O, Desjardins Y, Roy D, and Marette A
- Subjects
- Animals, C-Peptide blood, Diet, High-Fat, Endotoxemia metabolism, Fruit chemistry, Glucose metabolism, Homeostasis, Insulin blood, Insulin metabolism, Liver metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Obese, Obesity metabolism, RNA, Ribosomal, 16S genetics, Time Factors, Fatty Liver drug therapy, Fatty Liver metabolism, Insulin Resistance, Intestines drug effects, Liver drug effects, Plant Extracts pharmacology
- Abstract
Aims/hypothesis: There is growing evidence that fruit polyphenols exert beneficial effects on the metabolic syndrome, but the underlying mechanisms remain poorly understood. In the present study, we aimed to analyse the effects of polyphenolic extracts from five types of Arctic berries in a model of diet-induced obesity., Methods: Male C57BL/6 J mice were fed a high-fat/high-sucrose (HFHS) diet and orally treated with extracts of bog blueberry (BBE), cloudberry (CLE), crowberry (CRE), alpine bearberry (ABE), lingonberry (LGE) or vehicle (HFHS) for 8 weeks. An additional group of standard-chow-fed, vehicle-treated mice was included as a reference control for diet-induced obesity. OGTTs and insulin tolerance tests were conducted, and both plasma insulin and C-peptide were assessed throughout the OGTT. Quantitative PCR, western blot analysis and ELISAs were used to assess enterohepatic immunometabolic features. Faecal DNA was extracted and 16S rRNA gene-based analysis was used to profile the gut microbiota., Results: Treatment with CLE, ABE and LGE, but not with BBE or CRE, prevented both fasting hyperinsulinaemia (mean ± SEM [pmol/l]: chow 67.2 ± 12.3, HFHS 153.9 ± 19.3, BBE 114.4 ± 14.3, CLE 82.5 ± 13.0, CRE 152.3 ± 24.4, ABE 90.6 ± 18.0, LGE 95.4 ± 10.5) and postprandial hyperinsulinaemia (mean ± SEM AUC [pmol/l × min]: chow 14.3 ± 1.4, HFHS 31.4 ± 3.1, BBE 27.2 ± 4.0, CLE 17.7 ± 2.2, CRE 32.6 ± 6.3, ABE 22.7 ± 18.0, LGE 23.9 ± 2.5). None of the berry extracts affected C-peptide levels or body weight gain. Levels of hepatic serine phosphorylated Akt were 1.6-, 1.5- and 1.2-fold higher with CLE, ABE and LGE treatment, respectively, and hepatic carcinoembryonic antigen-related cell adhesion molecule (CEACAM)-1 tyrosine phosphorylation was 0.6-, 0.7- and 0.9-fold increased in these mice vs vehicle-treated, HFHS-fed mice. These changes were associated with reduced liver triacylglycerol deposition, lower circulating endotoxins, alleviated hepatic and intestinal inflammation, and major gut microbial alterations (e.g. bloom of Akkermansia muciniphila, Turicibacter and Oscillibacter) in CLE-, ABE- and LGE-treated mice., Conclusions/interpretation: Our findings reveal novel mechanisms by which polyphenolic extracts from ABE, LGE and especially CLE target the gut-liver axis to protect diet-induced obese mice against metabolic endotoxaemia, insulin resistance and hepatic steatosis, which importantly improves hepatic insulin clearance. These results support the potential benefits of these Arctic berries and their integration into health programmes to help attenuate obesity-related chronic inflammation and metabolic disorders., Data Availability: All raw sequences have been deposited in the public European Nucleotide Archive server under accession number PRJEB19783 ( https://www.ebi.ac.uk/ena/data/view/PRJEB19783 ).
- Published
- 2018
- Full Text
- View/download PDF
5. Treatment with a novel agent combining docosahexaenoate and metformin increases protectin DX and IL-6 production in skeletal muscle and reduces insulin resistance in obese diabetic db/db mice.
- Author
-
Mitchell PL, Nachbar R, Lachance D, St-Pierre P, Trottier J, Barbier O, and Marette A
- Subjects
- Animals, Blood Glucose metabolism, Disease Models, Animal, Drug Combinations, Glucose metabolism, Glucose Clamp Technique, Liver metabolism, Mice, Mice, Obese, Muscle, Skeletal metabolism, Blood Glucose drug effects, Diabetes Mellitus, Type 2 metabolism, Docosahexaenoic Acids metabolism, Docosahexaenoic Acids pharmacology, Glutamates pharmacology, Hypoglycemic Agents pharmacology, Insulin Resistance, Interleukin-6 metabolism, Liver drug effects, Metformin pharmacology, Muscle, Skeletal drug effects, Obesity metabolism
- Abstract
Aims: To compare the therapeutic potential of TP-113, a unique molecular entity linking DHA with metformin, for alleviating insulin resistance in obese diabetic mice through the PDX/IL-6 pathway., Material and Methods: We utilized the generically obese diabetic db/db mouse model for all experiments. Initial studies investigated both a dose and time course response. These results were then utilized to design a long-term (5 week) treatment protocol. Mice were gavaged twice daily with 1 of 3 treatments: 200 mg/kg BW TP113, an equivalent dose of metformin alone (70 mg/kg BW) or water. Whole-body insulin sensitivity was measured using the hyperinsulinaemic-isoglycaemic clamp procedure in awake unrestrained mice., Results: We first confirmed that acute TP-113 treatment raises PDX and IL-6 levels in skeletal muscle. We next tested the long-term glucoregulatory effect of oral TP-113 in obese diabetic db/db mice and compared its effect to an equivalent dose of metformin. A 5-week oral treatment with TP-113 reduced insulin resistance compared to both vehicle treatment and metformin alone, revealed by the determination of whole-body insulin sensitivity for glucose disposal using the clamp technique. This insulin-sensitizing effect was explained primarily by improvement of insulin action to suppress hepatic glucose production in TP-113-treated mice. These effects of TP-113 were greater than that of an equivalent dose of metformin, indicating that TP-113 increases metformin efficacy for reducing insulin resistance., Conclusion: We conclude that TP-113 improves insulin sensitivity in obese diabetic mice through activation of the PDX/IL-6 signaling axis in skeletal muscle and improved glucoregulatory action in the liver., (© 2016 John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
6. Long noncoding RNA MEG3 induces cholestatic liver injury by interaction with PTBP1 to facilitate shp mRNA decay.
- Author
-
Zhang L, Yang Z, Trottier J, Barbier O, and Wang L
- Subjects
- Animals, Binding Sites, Cells, Cultured, Cholestasis pathology, Disease Models, Animal, Down-Regulation, Hep G2 Cells, Hepatocytes cytology, Hepatocytes metabolism, Humans, Liver pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Promoter Regions, Genetic, Random Allocation, Sensitivity and Specificity, Cholestasis metabolism, Heterogeneous-Nuclear Ribonucleoproteins genetics, Liver injuries, Polypyrimidine Tract-Binding Protein genetics, RNA Stability genetics, RNA, Long Noncoding genetics
- Abstract
Bile acids (BAs) play critical physiological functions in cholesterol homeostasis, and deregulation of BA metabolism causes cholestatic liver injury. The long noncoding RNA maternally expressed gene 3 (MEG3) was recently shown as a potential tumor suppressor; however, its basic hepatic function remains elusive. Using RNA pull-down with biotin-labeled sense or anti-sense MEG 3RNA followed by mass spectrometry, we identified RNA-binding protein polypyrimidine tract-binding protein 1 (PTBP1) as a MEG3 interacting protein and validated their interaction by RNA immunoprecipitation (RIP). Bioinformatics analysis revealed putative binding sites for PTBP1 within the coding region (CDS) of small heterodimer partner (SHP), a key repressor of BA biosynthesis. Forced expression of MEG3 in hepatocellular carcinoma cells guided and facilitated PTBP1 binding to the Shp CDS, resulting in Shp mRNA decay. Transient overexpression of MEG3 RNA in vivo in mouse liver caused rapid Shp mRNA degradation and cholestatic liver injury, which was accompanied by the disruption of BA homeostasis, elevation of liver enzymes, as well as dysregulation of BA synthetic enzymes and metabolic genes. Interestingly, RNA sequencing coupled with quantitative PCR (qPCR) revealed a drastic induction of MEG3 RNA in Shp
-/- liver. SHP inhibited MEG3 gene transcription by repressing cAMP response element-binding protein (CREB) transactivation of the MEG3 promoter. In addition, the expression of MEG3 and PTBP1 was activated in human fibrotic and cirrhotic livers., Conclusion: MEG3 causes cholestasis by serving as a guide RNA scaffold to recruit PTBP1 to destabilize Shp mRNA. SHP in turn represses CREB-mediated activation of MEG3 expression in a feedback-regulatory fashion. (Hepatology 2017;65:604-615)., Competing Interests: No conflicts of interests exist for all authors., (© 2016 by the American Association for the Study of Liver Diseases.)- Published
- 2017
- Full Text
- View/download PDF
7. Liver Expression of Sulphotransferase 2A1 Enzyme Is Impaired in Patients with Primary Sclerosing Cholangitis: Lack of the Response to Enhanced Expression of PXR.
- Author
-
Wunsch E, Klak M, Wasik U, Milkiewicz M, Blatkiewicz M, Urasinska E, Barbier O, Bielicki D, Bogdanos DP, Elias E, and Milkiewicz P
- Subjects
- Adolescent, Adult, Aged, Base Sequence, Cholangitis, Sclerosing diagnosis, Female, Gene Expression Regulation, Humans, Intestine, Small metabolism, Liver pathology, Liver Cirrhosis, Biliary diagnosis, Liver Cirrhosis, Biliary genetics, Liver Cirrhosis, Biliary metabolism, Male, MicroRNAs genetics, Middle Aged, Molecular Sequence Data, Polymorphism, Single Nucleotide, Pregnane X Receptor, Promoter Regions, Genetic, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Steroid metabolism, Young Adult, Cholangitis, Sclerosing genetics, Cholangitis, Sclerosing metabolism, Liver metabolism, Receptors, Steroid genetics, Sulfotransferases genetics, Sulfotransferases metabolism
- Abstract
Background/aim: Sulphotransferase 2A1 (SULT2A1) exerts hepatoprotective effects. Transcription of SULT2A1 gene is induced by pregnane-X-receptor (PXR) and can be repressed by miR-378a-5p. We studied the PXR/SULT2A1 axis in chronic cholestatic conditions: primary sclerosing cholangitis (PSC) and primary biliary cirrhosis (PBC)., Materials/methods: Western-blot/PCRs for SULT2A1/PXR were performed in PSC (n = 11), PBC (n = 19), and control liver tissues (n = 19). PXR and SULT2A1 mRNA was analyzed in intestinal tissues from 22 PSC patients. Genomic DNA was isolated from blood of PSC patients (n = 120) and an equal number of healthy volunteers. Liver miRNA expression was evaluated using Affymetrix-Gene-Chip miRNA4.0., Results: Increased PXR protein was observed in both PSC and PBC compared to controls and was accompanied by a significant increase of SULT2A1 in PBC but not in PSC. Decreased expression of SULT2A1 mRNA was also seen in ileum of patients with PSC. Unlike PBC, miRNA analysis in PSC has shown a substantial increase in liver miR-378a-5p., Conclusions: PSC is characterized by disease-specific impairment of SULT2A1 expression following PXR activation, a phenomenon which is not noted in PBC, and may account for the impaired hepatoprotection in PSC. miRNA analysis suggests that SULT2A1 expression in PSC may be regulated by miR-378a-5p, connoting its pathogenic role.
- Published
- 2015
- Full Text
- View/download PDF
8. Role of glucuronidation for hepatic detoxification and urinary elimination of toxic bile acids during biliary obstruction.
- Author
-
Perreault M, Białek A, Trottier J, Verreault M, Caron P, Milkiewicz P, and Barbier O
- Subjects
- Apoptosis drug effects, Chenodeoxycholic Acid toxicity, Chenodeoxycholic Acid urine, Deoxycholic Acid toxicity, Deoxycholic Acid urine, Female, Hep G2 Cells, Humans, Lithocholic Acid toxicity, Lithocholic Acid urine, Male, Bile Acids and Salts toxicity, Bile Acids and Salts urine, Cholestasis metabolism, Cholestasis urine, Liver metabolism
- Abstract
Biliary obstruction, a severe cholestatic condition, results in a huge accumulation of toxic bile acids (BA) in the liver. Glucuronidation, a conjugation reaction, is thought to protect the liver by both reducing hepatic BA toxicity and increasing their urinary elimination. The present study evaluates the contribution of each process in the overall BA detoxification by glucuronidation. Glucuronide (G), glycine, taurine conjugates, and unconjugated BAs were quantified in pre- and post-biliary stenting urine samples from 12 patients with biliary obstruction, using liquid chromatography-tandem mass spectrometry (LC-MS/MS). The same LC-MS/MS procedure was used to quantify intra- and extracellular BA-G in Hepatoma HepG2 cells. Bile acid-induced toxicity in HepG2 cells was evaluated using MTS reduction, caspase-3 and flow cytometry assays. When compared to post-treatment samples, pre-stenting urines were enriched in glucuronide-, taurine- and glycine-conjugated BAs. Biliary stenting increased the relative BA-G abundance in the urinary BA pool, and reduced the proportion of taurine- and glycine-conjugates. Lithocholic, deoxycholic and chenodeoxycholic acids were the most cytotoxic and pro-apoptotic/necrotic BAs for HepG2 cells. Other species, such as the cholic, hyocholic and hyodeoxycholic acids were nontoxic. All BA-G assayed were less toxic and displayed lower pro-apoptotic/necrotic effects than their unconjugated precursors, even if they were able to penetrate into HepG2 cells. Under severe cholestatic conditions, urinary excretion favors the elimination of amidated BAs, while glucuronidation allows the conversion of cytotoxic BAs into nontoxic derivatives.
- Published
- 2013
- Full Text
- View/download PDF
9. Enantiomer selective glucuronidation of the non-steroidal pure anti-androgen bicalutamide by human liver and kidney: role of the human UDP-glucuronosyltransferase (UGT)1A9 enzyme.
- Author
-
Grosse L, Campeau AS, Caron S, Morin FA, Meunier K, Trottier J, Caron P, Verreault M, and Barbier O
- Subjects
- Chromatography, Liquid, Humans, Kidney drug effects, Liver drug effects, Male, Microsomes enzymology, Prostatic Neoplasms drug therapy, Stereoisomerism, Tandem Mass Spectrometry, UDP-Glucuronosyltransferase 1A9, Androgen Antagonists pharmacology, Anilides pharmacology, Glucuronosyltransferase metabolism, Kidney enzymology, Liver enzymology, Nitriles pharmacology, Tosyl Compounds pharmacology
- Abstract
Bicalutamide (Casodex(®) ) is a non-steroidal pure anti-androgen used in the treatment of localized prostate cancer. It is a racemate drug, and its activity resides in the (R)-enantiomer, with little in the (S)-enantiomer. A major metabolic pathway for bicalutamide is glucuronidation catalysed by UDP-glucuronosyltransferase (UGT) enzymes. While (S)bicalutamide is directly glucuronidated, (R)bicalutamide requires hydroxylation prior to glucuronidation. The contribution of human tissues and UGT isoforms in the metabolism of these enantiomers has not been extensively investigated. In this study, both (R) and/or (S)bicalutamide were converted into glucuronide (-G) derivatives after incubation of pure and racemic solutions with microsomal extracts from human liver and kidney. Intestinal microsomes exhibited only low reactivity with these substrates. Km values of liver and kidney samples for (S)bicalutamide glucuronidation were similar, and lower than values obtained with the (R)-enantiomer. Among the 16 human UGTs tested, UGT1A8 and UGT1A9 were able to form both (S) and (R)bicalutamide-G from pure or racemic substrates. UGT2B7 was also able to form (R)bicalutamide-G. Kinetic parameters of the recombinant UGT2B7, UGT1A8 and UGT1A9 enzymes support a predominant role of the UGT1A9 isoform in bicalutamide metabolism. Accordingly, (S)bicalutamide inhibited the ability of human liver and kidney microsomes to glucuronidate the UGT1A9 probe substrate, propofol. In conclusion, the present study provides the first comprehensive analysis of in vitro bicalutamide glucuronidation by human tissues and UGTs and identifies UGT1A9 as a major contributor for (R) and (S) glucuronidation in the human liver and kidney., (© 2013 Nordic Pharmacological Society. Published by John Wiley & Sons Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
10. Regulation of endobiotics glucuronidation by ligand-activated transcription factors: physiological function and therapeutic potential.
- Author
-
Verreault M, Kaeding J, Caron P, Trottier J, Grosse L, Houssin E, Pâquet S, Perreault M, and Barbier O
- Subjects
- Activating Transcription Factors metabolism, Cells, Cultured, Glucuronides metabolism, Glucuronosyltransferase metabolism, Promoter Regions, Genetic, Signal Transduction physiology, Activating Transcription Factors physiology, Bile Acids and Salts metabolism, Bilirubin blood, Liver metabolism, Receptors, Cytoplasmic and Nuclear physiology
- Abstract
Recent progresses in molecular pharmacology approaches have allowed the identification and characterization of a series of nuclear receptors (NR) which efficiently control the level UDP-glucuronosyltransferase (UGT) genes expression. These regulatory processes ensure optimized UGT expression in response to specific endogenous and/or exogenous stimuli. Interestingly, numerous endogenous activators of these NRs are conjugated by the UGT enzymes they regulate. In such a case, the NR-dependent regulation of UGT genes corresponds to a feedforward/feedback mechanism by which a bioactive molecule controls its own concentrations. In the present review, we will discuss i) how bilirubin reduces its circulating levels by activating AhR in the liver; ii) how bile acids modulate their hepatic glucuronidation via PXR- and FXR-dependent processes in enterohepatic tissues; and iii) how androgens inhibit their cellular metabolism in prostate cancer cells through an AR-dependent mechanism. Subsequently, with further discussion of the same examples (bilirubin and bile acids), we will illustrate how NR-dependent regulation of UGT enzymes may contribute to the beneficial effects of pharmacological activators of nuclear receptors, such as CAR and PPARa.
- Published
- 2010
- Full Text
- View/download PDF
11. Human UDP-glucuronosyltransferase (UGT)1A3 enzyme conjugates chenodeoxycholic acid in the liver.
- Author
-
Trottier J, Verreault M, Grepper S, Monté D, Bélanger J, Kaeding J, Caron P, Inaba TT, and Barbier O
- Subjects
- Adult, Cell Line, Clofibric Acid pharmacology, DNA-Binding Proteins metabolism, Female, Hepatocytes metabolism, Humans, Male, Microbodies, Microsomes, Liver metabolism, Middle Aged, PPAR alpha metabolism, Pyrimidines metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Transcription Factors metabolism, Chenodeoxycholic Acid metabolism, Glucuronosyltransferase metabolism, Liver enzymology
- Abstract
Chenodeoxycholic acid (CDCA) is a liver-formed detergent and plays an important role in the control of cholesterol homeostasis. During cholestasis, toxic bile acids (BA) accumulate in hepatocytes causing damage and consequent impairment of their function. Glucuronidation, a conjugation reaction catalyzed by UDP-glucuronosyltransferase (UGT) enzymes, is considered an important metabolic pathway for hepatic BA. This study identifies the human UGT1A3 enzyme as the major enzyme responsible for the hepatic formation of the acyl CDCA-24glucuronide (CDCA-24G). Kinetic analyses revealed that human liver and UGT1A3 catalyze the formation of CDCA-24G with similar K(m) values of 10.6 to 18.6 mumol/L, respectively. In addition, electrophoretic mobility shift assays and transient transfection experiments revealed that glucuronidation reduces the ability of CDCA to act as an activator of the nuclear farnesoid X-receptor (FXR). Finally, we observed that treatment of human hepatocytes with fibrates increases the expression and activity of UGT1A3, whereas CDCA has no effect. In conclusion, UGT1A3 is the main UGT enzyme for the hepatic formation of CDCA-24G and glucuronidation inhibits the ability of CDCA to act as an FXR activator. In vitro data also suggest that fibrates may favor the formation of bile acid glucuronides in cholestatic patients.
- Published
- 2006
- Full Text
- View/download PDF
12. Hepatic expression of the UGT1A9 gene is governed by hepatocyte nuclear factor 4alpha.
- Author
-
Barbier O, Girard H, Inoue Y, Duez H, Villeneuve L, Kamiya A, Fruchart JC, Guillemette C, Gonzalez FJ, and Staels B
- Subjects
- Carcinoma, Hepatocellular, Cell Line, Tumor, Cloning, Molecular, Hepatocyte Nuclear Factor 4, Humans, Mutagenesis, Site-Directed, Plasmids, Promoter Regions, Genetic, Recombinant Proteins metabolism, Restriction Mapping, Reverse Transcriptase Polymerase Chain Reaction, UDP-Glucuronosyltransferase 1A9, DNA-Binding Proteins physiology, Gene Expression Regulation, Enzymologic physiology, Glucuronosyltransferase genetics, Liver enzymology, Phosphoproteins physiology, Transcription Factors physiology
- Abstract
UDP-glucuronosyltransferase (UGT) enzymes catalyze the glucuronidation reaction, which is a major pathway in the catabolism and elimination of numerous endo- and xenobiotics. Among the UGT enzyme family members, the UGT1A7, UGT1A8, UGT1A9, and UGT1A10 isoforms are issued from a single gene through differential splicing. However, these enzymes display distinct tissue-specific expression patterns. Indeed, UGT1A7, UGT1A8, and UGT1A10 are exclusively expressed in extrahepatic tissues, whereas UGT1A9 transcripts are found at high concentrations in liver. In the present study, we report that the liver-enriched hepatocyte nuclear factor 4 (HNF4)-alpha controls the hepatic expression of the UGT1A9 enzyme. Liver-specific disruption of the HNF4alpha gene in mice drastically decreases liver UGT1A9 mRNA levels. Furthermore, an HNF4alpha response element (HNF4alpha RE) was identified in the promoter of human UGT1A9 at position -372 to -360 base pairs by transient transfection, electrophoretic mobility shift assays, and chromatin immunoprecipitation experiments. It is interesting that this response element is absent in the proximal UGT1A7, UGT1A8, and UGT1A10 gene promoters. In conclusion, the present study identifies HNF4alpha as a major factor for the control of UGT1A9 hepatic expression and suggests that the absence of UGT1A7, UGT1A8, and UGT1A10 expression in the liver is caused by, at least in part, a few base pair changes in their promoter sequences in the region corresponding to the HNF4alpha RE of the UGT1A9 gene.
- Published
- 2005
- Full Text
- View/download PDF
13. Glucose regulates the expression of the farnesoid X receptor in liver.
- Author
-
Duran-Sandoval D, Mautino G, Martin G, Percevault F, Barbier O, Fruchart JC, Kuipers F, and Staels B
- Subjects
- Animals, Base Sequence, DNA Primers, Diabetes Mellitus, Experimental genetics, Hepatocytes drug effects, Insulin pharmacology, Kinetics, Liver drug effects, Male, Polymerase Chain Reaction methods, RNA, Messenger drug effects, RNA, Messenger genetics, Rats, Rats, Wistar, Receptors, Cytoplasmic and Nuclear, Transcription, Genetic drug effects, DNA-Binding Proteins genetics, Diabetes Mellitus, Experimental physiopathology, Gene Expression Regulation drug effects, Glucose pharmacology, Hepatocytes physiology, Liver physiology, Transcription Factors genetics
- Abstract
An increased prevalence of hypertriglyceridemia and gallbladder disease occurs in patients with diabetes or insulin resistance. Hypertriglyceridemia is positively associated to gall bladder disease risk. The farnesoid X receptor (FXR) is a bile acid-activated nuclear receptor that plays a key role in bile acid and triglyceride homeostasis. The mechanisms controlling FXR gene expression are poorly understood. This study evaluated whether FXR gene expression is regulated by alterations in glucose homeostasis. FXR expression was decreased in livers of streptozotocin-induced diabetic rats and normalized upon insulin supplementation. Concomitantly with diabetes progression, FXR expression also decreased in aging diabetic Zucker rats. In primary rat hepatocytes, D-glucose increased FXR mRNA in a dose- and time-dependent manner, whereas insulin counteracted this effect. Addition of xylitol, a precursor of xylulose-5-phosphate, to primary rat hepatocytes increased FXR expression to a comparable level as D-glucose. Finally, expression of the FXR target genes, SHP and apolipoprotein C-III, were additively regulated by D-glucose and FXR ligands. This study demonstrates that FXR is decreased in animal models of diabetes. In addition, FXR is regulated by glucose likely via the pentose phosphate pathway. Dysregulation of FXR expression may contribute to alterations in lipid and bile acid metabolism in patients with diabetes or insulin resistance.
- Published
- 2004
- Full Text
- View/download PDF
14. Peroxisome proliferator-activated receptor alpha induces hepatic expression of the human bile acid glucuronidating UDP-glucuronosyltransferase 2B4 enzyme.
- Author
-
Barbier O, Duran-Sandoval D, Pineda-Torra I, Kosykh V, Fruchart JC, and Staels B
- Subjects
- Animals, Bile Acids and Salts metabolism, Blotting, Northern, Blotting, Western, Cell Line, Cells, Cultured, Cholesterol metabolism, DNA, Complementary metabolism, Dose-Response Relationship, Drug, Gene Expression Regulation, Hepatocytes metabolism, Homozygote, Humans, Lipid Metabolism, Luciferases metabolism, Mice, Microsomes, Liver metabolism, Models, Biological, Mutagenesis, Site-Directed, Peroxisome Proliferators pharmacology, Plasmids metabolism, Promoter Regions, Genetic, Pyrimidines pharmacology, RNA metabolism, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Transfection, Tumor Cells, Cultured, Glucuronosyltransferase metabolism, Liver enzymology, Liver metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Transcription Factors metabolism
- Abstract
Glucuronidation, a major metabolic pathway for a large variety of endobiotics and xenobiotics, is catalyzed by enzymes belonging to the UDP-glucuronosyltransferase (UGT) family. Among UGT enzymes, UGT2B4 conjugates a large variety of endogenous and exogenous molecules and is considered to be the major bile acid conjugating UGT enzyme in human liver. In the present study, we identify UGT2B4 as a novel target gene of the nuclear receptor peroxisome proliferator-activated receptor alpha (PPAR alpha), which mediates the hypolipidemic action of fibrates. Incubation of human hepatocytes or hepatoblastoma HepG2 and Huh7 cells with synthetic PPAR alpha agonists, fenofibric acid, or Wy 14643 resulted in an increase of UGT2B4 mRNA levels. Furthermore, treatment of HepG2 cells with Wy 14643 induced the glucuronidation of hyodeoxycholic acid, a specific bile acid UGT2B4 substrate. Analysis of UGT2B mRNA and protein levels in PPAR alpha wild type and null mice revealed that PPAR alpha regulates both basal and fibrate-induced expression of these enzymes in rodents also. Finally, a PPAR response element was identified in the UGT2B4 promoter by site-directed mutagenesis and electromobility shift assays. These results demonstrate that PPAR alpha agonists may control the catabolism of cytotoxic bile acids and reinforce recent data indicating that PPAR alpha, which has been largely implicated in the control of lipid and cholesterol metabolism, is also an important modulator of the metabolism of endobiotics and xenobiotics in human hepatocytes.
- Published
- 2003
- Full Text
- View/download PDF
15. Farnesoid X receptor agonists suppress hepatic apolipoprotein CIII expression.
- Author
-
Claudel T, Inoue Y, Barbier O, Duran-Sandoval D, Kosykh V, Fruchart J, Fruchart JC, Gonzalez FJ, and Staels B
- Subjects
- Animals, Apolipoprotein C-III, Apolipoproteins C metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors, Bile Acids and Salts pharmacology, Cells, Cultured, Cholesterol 7-alpha-Hydroxylase physiology, Cholestyramine Resin pharmacology, Dimerization, Hepatocyte Nuclear Factor 4, Hepatocytes, Humans, Isoxazoles pharmacology, Male, Mice, Mice, Inbred C57BL, Phosphoproteins deficiency, Phosphoproteins metabolism, Promoter Regions, Genetic, RNA, Messenger analysis, Receptors, Cytoplasmic and Nuclear agonists, Receptors, Retinoic Acid physiology, Response Elements, Retinoid X Receptors, Transcription Factors deficiency, Transcription Factors metabolism, Transcription Factors physiology, Triglycerides blood, Apolipoproteins C genetics, DNA-Binding Proteins, Gene Expression Regulation, Liver metabolism, Receptors, Cytoplasmic and Nuclear physiology
- Abstract
Background & Aims: Increased serum triglyceride levels constitute a risk factor for coronary heart disease. Apolipoprotein CIII (Apo CIII) is a determinant of serum triglyceride metabolism. In this study, we investigated whether activators of the nuclear farnesoid X receptor (FXR) modulate Apo CIII gene expression., Methods: The influence of bile acids and synthetic FXR activators on Apo CIII and triglyceride metabolism was studied in vivo by using FXR wild-type and FXR-deficient mice and in vitro by using human primary hepatocytes and HepG2 cells., Results: In mice, treatment with the FXR agonist taurocholic acid strongly decreased serum triglyceride levels, an effect associated with reduced Apo CIII serum and liver messenger RNA levels. By contrast, no change was observed in FXR-deficient mice. Incubation of human primary hepatocytes and HepG2 cells with bile acids or the nonsteroidal synthetic FXR agonist GW4064 resulted in a dose-dependent down-regulation of Apo CIII gene expression. Promoter transfection experiments and mutation analysis showed that bile acid-activated FXR decrease human Apo CIII promoter activity via a negative FXR response element located in the I(4) footprint between nucleotides -739 and -704. Chromatin immunoprecipitation experiments showed that bile acid treatment led to binding of FXR/retinoid X receptor heterodimers to and displacement of HNF4alpha from this site. Bile acid treatment still repressed liver Apo CIII gene expression in hepatic HNF4alpha-deficient mice, suggesting an active rather than a competitive mechanism of Apo CIII repression by the FXR., Conclusions: We identified bile acid and synthetic activators of the nuclear FXR as negative regulators of Apo CIII expression, an effect that may contribute to the triglyceride-decreasing action of FXR agonists.
- Published
- 2003
- Full Text
- View/download PDF
16. Intestinal glucuronidation protects against chemotherapy-induced toxicity by irinotecan (CPT-11)
- Author
-
Chen, Shujuan, Yueh, Mei-Fei, Bigo, Cyril, Barbier, Olivier, Wang, Kepeng, Karin, Michael, Nguyen, Nghia, and Tukey, Robert H.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.