1
|
Sudo K, Delmas-Eliason A, Soucy S, Barrack KE, Liu J, Balasubramanian A, Shu CJ, James MJ, Hegner CL, Dionne HD, Rodriguez-Palacios A, Krause HM, O'Toole GA, Karpen SJ, Dawson PA, Schultz D, Sundrud MS. Quantifying Forms and Functions of Enterohepatic Bile Acid Pools in Mice. Cell Mol Gastroenterol Hepatol 2024; 18:101392. [PMID: 39179177 PMCID: PMC11490680 DOI: 10.1016/j.jcmgh.2024.101392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUNDS & AIMS Bile acids (BAs) are core gastrointestinal metabolites with dual functions in lipid absorption and cell signaling. BAs circulate between the liver and distal small intestine (i.e., ileum), yet the dynamics through which complex BA pools are absorbed in the ileum and interact with host intestinal cells in vivo remain poorly understood. Because ileal absorption is rate-limiting in determining which BAs in the intestinal lumen gain access to host intestinal cells and receptors, and at what concentrations, we hypothesized that defining the rates and routes of ileal BA absorption in vivo would yield novel insights into the physiological forms and functions of mouse enterohepatic BA pools. METHODS Using ex vivo mass spectrometry, we quantified 88 BA species and metabolites in the intestinal lumen and superior mesenteric vein of individual wild-type mice, and cage-mates lacking the ileal BA transporter, Asbt/Slc10a2. RESULTS Using these data, we calculated that the pool of BAs circulating through ileal tissue (i.e., the ileal BA pool) in fasting C57BL/6J female mice is ∼0.3 μmol/g. Asbt-mediated transport accounted for ∼80% of this pool and amplified size. Passive permeability explained the remaining ∼20% and generated diversity. Compared with wild-type mice, the ileal BA pool in Asbt-deficient mice was ∼5-fold smaller, enriched in secondary BA species and metabolites normally found in the colon, and elicited unique transcriptional responses on addition to exvivo-cultured ileal explants. CONCLUSIONS This study defines quantitative traits of the mouse enterohepatic BA pool and reveals how aberrant BA metabolism can impinge directly on host intestinal physiology.
Collapse
Affiliation(s)
- Koichi Sudo
- Center for Digestive Health, Dartmouth Health, Lebanon, New Hampshire
| | - Amber Delmas-Eliason
- Department of Immunology and Microbiology, Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida
| | - Shannon Soucy
- Department of Biomedical Data Science, Geisel School of Medicine, Hanover, New Hampshire
| | - Kaitlyn E Barrack
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - Jiabao Liu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Akshaya Balasubramanian
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | | | | | - Courtney L Hegner
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, Florida
| | - Henry D Dionne
- Center for Digestive Health, Dartmouth Health, Lebanon, New Hampshire
| | - Alex Rodriguez-Palacios
- Division of Gastroenterology and Liver Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio; Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio; University Hospitals Research and Education Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Henry M Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - George A O'Toole
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - Saul J Karpen
- Division of Pediatric Gastroenterology, Department of Pediatrics, Hepatology, and Nutrition, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, Georgia; Stravitz-Sanyal Liver Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University, Richmond, Virginia
| | - Paul A Dawson
- Division of Pediatric Gastroenterology, Department of Pediatrics, Hepatology, and Nutrition, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, Georgia
| | - Daniel Schultz
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - Mark S Sundrud
- Center for Digestive Health, Dartmouth Health, Lebanon, New Hampshire; Department of Immunology and Microbiology, Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida; Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire; The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, Florida; Department of Medicine, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire; Dartmouth Cancer Center, Lebanon, New Hampshire.
| |
Collapse
|
2
|
Gallucci GM, Hayes CM, Boyer JL, Barbier O, Assis DN, Ghonem NS. PPAR-Mediated Bile Acid Glucuronidation: Therapeutic Targets for the Treatment of Cholestatic Liver Diseases. Cells 2024; 13:1296. [PMID: 39120326 PMCID: PMC11312002 DOI: 10.3390/cells13151296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Cholestatic liver diseases, including primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC), result from an impairment of bile flow that leads to the hepatic retention of bile acids, causing liver injury. Until recently, the only approved treatments for PBC were ursodeoxycholic acid (UDCA) and obeticholic acid (OCA). While these therapies slow the progression of PBC in the early stage of the disease, approximately 40% of patients respond incompletely to UDCA, and advanced cases do not respond. UDCA does not improve survival in patients with PSC, and patients often have dose-limiting pruritus reactions to OCA. Left untreated, these diseases can progress to fibrosis and cirrhosis, resulting in liver failure and the need for transplantation. These shortcomings emphasize the urgent need for alternative treatment strategies. Recently, nuclear hormone receptors have been explored as pharmacological targets for adjunct therapy because they regulate enzymes involved in bile acid metabolism and detoxification. In particular, the peroxisome proliferator-activated receptor (PPAR) has emerged as a therapeutic target for patients with PBC or PSC who experience an incomplete response to UDCA. PPARα is predominantly expressed in the liver, and it plays an essential role in the regulation of cytochrome P450 (CYP) and uridine 5'-diphospho-glucuronosyltransferase (UGT) enzymes, both of which are critical enzyme families involved in the regulation of bile acid metabolism and glucuronidation, respectively. Importantly, PPARα agonists, e.g., fenofibrate, have shown therapeutic benefits in reducing elevated markers of cholestasis in patients with PBC and PSC, and elafibranor, the first PPAR (dual α, β/δ) agonist, has been FDA-approved for the second-line treatment of PBC. Additionally, newer PPAR agonists that target various PPAR isoforms (β/δ, γ) are under development as an adjunct therapy for PBC or PSC, although their impact on glucuronidation pathways are less characterized. This review will focus on PPAR-mediated bile acid glucuronidation as a therapeutic pathway to improve outcomes for patients with PBC and PSC.
Collapse
Affiliation(s)
- Gina M. Gallucci
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Colleen M. Hayes
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - James L. Boyer
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, USA
| | - Olivier Barbier
- Faculty of Pharmacy, Laval University, Québec, QC G1V 0A6, Canada
| | - David N. Assis
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, USA
| | - Nisanne S. Ghonem
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
3
|
Kang L, Li D, Jiang X, Zhang Y, Pan M, Hu Y, Si L, Zhang Y, Huang J. Hepatotoxicity of the Major Anthraquinones Derived From Polygoni Multiflori Radix Based on Bile Acid Homeostasis. Front Pharmacol 2022; 13:878817. [PMID: 35662717 PMCID: PMC9157432 DOI: 10.3389/fphar.2022.878817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/15/2022] [Indexed: 01/22/2023] Open
Abstract
Polygoni Multiflori Radix (PMR), the dried root of Polygonum Multiflorum Thunb., has been widely used as traditional Chinese medicines in clinical practice for centuries. However, the frequently reported hepatotoxic adverse effects hindered its safe use in clinical practice. This study aims to explore the hepatotoxic effect of PMR extract and the major PMR derived anthraquinones including emodin, chrysophanol, and physcion in mice and the underlying mechanisms based on bile acid homeostasis. After consecutively treating the ICR mice with PMR extract or individual anthraquinones for 14 or 28 days, the liver function was evaluated by measuring serum enzymes levels and liver histological examination. The compositions of bile acids (BAs) in the bile, liver, and plasma were measured by LC-MS/MS, followed by Principal Component Analysis (PCA) and Partial Least Squares Discriminate Analysis (PLS-DA). Additionally, gene and protein expressions of BA efflux transporters, bile salt export pump (Bsep) and multidrug resistance-associated protein 2 (Mrp2), were examined to investigate the underlying mechanisms. After 14-day administration, mild inflammatory cell infiltration in the liver was observed in the physcion- and PMR-treated groups, while it was found in all the treated groups after 28-day treatment. Physcion and PMR extract induced hepatic BA accumulation after 14-day treatment, but such accumulation was attenuated after 28-day treatment. Based on the PLS-DA results, physcion- and PMR-treated groups were partially overlapping and both groups showed a clear separation with the control group in the mouse liver. The expression of Bsep and Mrp2 in the physcion- and PMR-treated mouse liver was decreased after 14-day treatment, while the downregulation was abrogated after 28-day treatment. Our study, for the first time, demonstrated that both PMR extract and tested anthraquinones could alter the disposition of either the total or individual BAs in the mouse bile, liver, and plasma via regulating the BA efflux transporters and induce liver injury, which provide a theoretical basis for the quality control and safe use of PMR in practice.
Collapse
Affiliation(s)
- Li Kang
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan, China.,School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan, China
| | - Dan Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Pharmacy, Shenzhen University General Hospital, Shenzhen, China
| | - Xin Jiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Zhang
- College of Pharmacy, Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi, China
| | - Minhong Pan
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen, China
| | - Yixin Hu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Luqin Si
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongjun Zhang
- The Third Affiliated Hospital of School of Medicine, Shihezi University, Shihezi, China
| | - Jiangeng Huang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Gallucci GM, Trottier J, Hemme C, Assis DN, Boyer JL, Barbier O, Ghonem NS. Adjunct Fenofibrate Up-regulates Bile Acid Glucuronidation and Improves Treatment Response For Patients With Cholestasis. Hepatol Commun 2021; 5:2035-2051. [PMID: 34558841 PMCID: PMC8631103 DOI: 10.1002/hep4.1787] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/10/2021] [Accepted: 06/25/2021] [Indexed: 12/26/2022] Open
Abstract
Accumulation of cytotoxic bile acids (BAs) during cholestasis can result in liver failure. Glucuronidation, a phase II metabolism pathway responsible for BA detoxification, is regulated by peroxisome proliferator-activated receptor alpha (PPARα). This study investigates the efficacy of adjunct fenofibrate therapy to up-regulate BA-glucuronidation and reduce serum BA toxicity during cholestasis. Adult patients with primary biliary cholangitis (PBC, n = 32) and primary sclerosing cholangitis (PSC, n = 23), who experienced an incomplete response while receiving ursodiol monotherapy (13-15 mg/kg/day), defined as serum alkaline phosphatase (ALP) ≥ 1.5 times the upper limit of normal, received additional fenofibrate (145-160 mg/day) as standard of care. Serum BA and BA-glucuronide concentrations were measured by liquid chromatography-mass spectrometry. Combination therapy with fenofibrate significantly decreased elevated serum ALP (-76%, P < 0.001), aspartate transaminase, alanine aminotransferase, bilirubin, total serum BAs (-54%), and increased serum BA-glucuronides (+2.1-fold, P < 0.01) versus ursodiol monotherapy. The major serum BA-glucuronides that were favorably altered following adjunct fenofibrate include hyodeoxycholic acid-6G (+3.7-fold, P < 0.01), hyocholic acid-6G (+2.6-fold, P < 0.05), chenodeoxycholic acid (CDCA)-3G (-36%), and lithocholic acid (LCA)-3G (-42%) versus ursodiol monotherapy. Fenofibrate also up-regulated the expression of uridine 5'-diphospho-glucuronosyltransferases and multidrug resistance-associated protein 3 messenger RNA in primary human hepatocytes. Pearson's correlation coefficients identified strong associations between serum ALP and metabolic ratios of CDCA-3G (r2 = 0.62, P < 0.0001), deoxycholic acid (DCA)-3G (r2 = 0.48, P < 0.0001), and LCA-3G (r2 = 0.40, P < 0.001), in ursodiol monotherapy versus control. Receiver operating characteristic analysis identified serum BA-glucuronides as measures of response to therapy. Conclusion: Fenofibrate favorably alters major serum BA-glucuronides, which correlate with reduced serum ALP levels and improved outcomes. A PPARα-mediated anti-cholestatic mechanism is involved in detoxifying serum BAs in patients with PBC and PSC who have an incomplete response on ursodiol monotherapy and receive adjunct fenofibrate. Serum BA-glucuronides may serve as a noninvasive measure of treatment response in PBC and PSC.
Collapse
Affiliation(s)
- Gina M. Gallucci
- College of Pharmacy, Biomedical and Pharmaceutical SciencesUniversity of Rhode IslandKingstonRIUSA
| | - Jocelyn Trottier
- Laboratory of Molecular PharmacologyEndocrinology and Nephrology AxisCHU de Québec Research CenterLavalQuébecCanada
| | - Christopher Hemme
- College of Pharmacy, Biomedical and Pharmaceutical SciencesUniversity of Rhode IslandKingstonRIUSA
- RI‐INBRE Bioinformatics CoreKingstonRIUSA
| | | | | | - Olivier Barbier
- Laboratory of Molecular PharmacologyEndocrinology and Nephrology AxisCHU de Québec Research CenterLavalQuébecCanada
- Faculty of PharmacyLaval UniversityLavalQuébecCanada
| | - Nisanne S. Ghonem
- College of Pharmacy, Biomedical and Pharmaceutical SciencesUniversity of Rhode IslandKingstonRIUSA
| |
Collapse
|
5
|
Thérien A, Cieślak A, Verreault M, Perreault M, Trottier J, Gobeil S, Vohl MC, Barbier O. Omega-3 Polyunsaturated Fatty Acid: A Pharmaco-Nutraceutical Approach to Improve the Responsiveness to Ursodeoxycholic Acid. Nutrients 2021; 13:nu13082617. [PMID: 34444777 PMCID: PMC8400581 DOI: 10.3390/nu13082617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Ursodeoxycholic acid (UDCA) is the first line therapy for the treatment of cholestatic and autoimmune liver diseases. Its clinical use is currently limited by a significant proportion of non-responder patients. Polyunsaturated fatty acids (n-3 PUFAs) possess important anti-inflammatory properties and protect liver cells against bile acid (BA)-induced toxicity. The present study was designed to rapidly evaluate whether combining n-3 PUFAs (i.e., eicosapentaenoic [EPA] and docosahexaenoic [DHA] acids) to UDCA would provide additional benefits when compared to the drug alone. The parameters evaluated were (i) the expression of genes governing BA synthesis, transport, and metabolism; (ii) the prevention of BA-induced apoptosis and endoplasmic reticulum (ER)-stress; and (iii) the control of BA- and LPS-dependent inflammation. In the absence of n-3 PUFAs, most of the parameters investigated were unaffected by UDCA or were only altered by the higher dose (500 µM) of the drug. By contrast, in the presence of EPA/DHA (50/50 µM), all parameters showed a strongly improved response and the lowest UDCA dosage (50 µM) provided equal or better benefits than the highest dose used alone. For example, the combination EPA/DHA + UDCA 50 µM caused comparable down-regulation of the CYP7A1 gene expression and of the BA-induced caspase 3 activity as observed with UDCA 500 µM. In conclusion, these results suggest that the addition of n-3 PUFAs to UDCA may improve the response to the drug, and that such a pharmaco-nutraceutical approach could be used in clinic to open the narrow therapeutic dose of UDCA in cholestatic liver diseases.
Collapse
Affiliation(s)
- Ariane Thérien
- CHU de Québec Research Center, Québec, QC G1V 4G2, Canada; (A.T.); (A.C.); (M.V.); (M.P.); (J.T.); (S.G.)
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada
| | - Anna Cieślak
- CHU de Québec Research Center, Québec, QC G1V 4G2, Canada; (A.T.); (A.C.); (M.V.); (M.P.); (J.T.); (S.G.)
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada
| | - Mélanie Verreault
- CHU de Québec Research Center, Québec, QC G1V 4G2, Canada; (A.T.); (A.C.); (M.V.); (M.P.); (J.T.); (S.G.)
| | - Martin Perreault
- CHU de Québec Research Center, Québec, QC G1V 4G2, Canada; (A.T.); (A.C.); (M.V.); (M.P.); (J.T.); (S.G.)
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Jocelyn Trottier
- CHU de Québec Research Center, Québec, QC G1V 4G2, Canada; (A.T.); (A.C.); (M.V.); (M.P.); (J.T.); (S.G.)
| | - Stéphane Gobeil
- CHU de Québec Research Center, Québec, QC G1V 4G2, Canada; (A.T.); (A.C.); (M.V.); (M.P.); (J.T.); (S.G.)
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Marie-Claude Vohl
- Centre Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada;
| | - Olivier Barbier
- CHU de Québec Research Center, Québec, QC G1V 4G2, Canada; (A.T.); (A.C.); (M.V.); (M.P.); (J.T.); (S.G.)
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada
- Centre Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC G1V 0A6, Canada;
- Correspondence:
| |
Collapse
|
6
|
Gui L, Wu Q, Hu Y, Zeng W, Tan X, Zhu P, Li X, Yang L, Jia W, Liu C, Lan K. Compensatory Transition of Bile Acid Metabolism from Fecal Disposition of Secondary Bile Acids to Urinary Excretion of Primary Bile Acids Underlies Rifampicin-Induced Cholestasis in Beagle Dogs. ACS Pharmacol Transl Sci 2021; 4:1001-1013. [PMID: 33860216 DOI: 10.1021/acsptsci.1c00052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Indexed: 12/12/2022]
Abstract
Drug induced cholestasis (DIC) is complexly associated with dysbiosis of the host-gut microbial cometabolism of bile acids (BAs). Murine animals are not suitable for transitional studies because the murine BA metabolism is quite different from human metabolism. In this work, the rifampicin (RFP) induced cholestasis was established in beagle dogs that have a humanlike BA profile to disclose how RFP affects the host-gut microbial cometabolism of BAs. The daily excretion of BA metabolites in urine and feces was extensively analyzed during cholestasis by quantitative BA profiling along the primary-secondary-tertiary axis. Oral midazolam clearance was also acquired to monitor the RFP-induced enterohepatic CYP3A activities because CYP3A is exclusively responsible for the tertiary oxidation of hydrophobic secondary BAs. RFP treatments caused a compensatory transition of the BA metabolism from the fecal disposition of secondary BAs to the urinary excretion of primary BAs in dogs, resulting in an infantile BA metabolism pattern recently disclosed in newborns. However, the tertiary BAs consistently constituted limitedly in the daily BA excretion, indicating that the detoxification role of the CYP3A catalyzed tertiary BA metabolism was not as strong as expected in this model. Multiple host-gut microbial factors might have contributed to the transition of the BA metabolism, such as inhibition of BA transporters, induction of liver-kidney interplaying detoxification mechanisms, and elimination of gut bacteria responsible for secondary BA production. Transitional studies involving more cholestatic drugs in preclinical animals with a humanlike BA profile and DIC patients may pave the way for understanding the complex mechanism of DIC in the era of metagenomics.
Collapse
Affiliation(s)
- LanLan Gui
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17 People's South Road, Chengdu 610041, China
| | - QingLiang Wu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17 People's South Road, Chengdu 610041, China
| | - YiTing Hu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17 People's South Road, Chengdu 610041, China
| | - WuShuang Zeng
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17 People's South Road, Chengdu 610041, China
| | - XianWen Tan
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17 People's South Road, Chengdu 610041, China
| | - PingPing Zhu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17 People's South Road, Chengdu 610041, China
| | - XueJing Li
- Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu 610000, China
| | - Lian Yang
- Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu 610000, China.,WestChina-Frontier PharmaTech Co., Ltd., Chengdu 610041, China
| | - Wei Jia
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - ChangXiao Liu
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin 300193, China
| | - Ke Lan
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, No. 17 People's South Road, Chengdu 610041, China.,Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu 610000, China
| |
Collapse
|
7
|
Grant SM, DeMorrow S. Bile Acid Signaling in Neurodegenerative and Neurological Disorders. Int J Mol Sci 2020; 21:E5982. [PMID: 32825239 PMCID: PMC7503576 DOI: 10.3390/ijms21175982] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Bile acids are commonly known as digestive agents for lipids. The mechanisms of bile acids in the gastrointestinal track during normal physiological conditions as well as hepatic and cholestatic diseases have been well studied. Bile acids additionally serve as ligands for signaling molecules such as nuclear receptor Farnesoid X receptor and membrane-bound receptors, Takeda G-protein-coupled bile acid receptor and sphingosine-1-phosphate receptor 2. Recent studies have shown that bile acid signaling may also have a prevalent role in the central nervous system. Some bile acids, such as tauroursodeoxycholic acid and ursodeoxycholic acid, have shown neuroprotective potential in experimental animal models and clinical studies of many neurological conditions. Alterations in bile acid metabolism have been discovered as potential biomarkers for prognosis tools as well as the expression of various bile acid receptors in multiple neurological ailments. This review explores the findings of recent studies highlighting bile acid-mediated therapies and bile acid-mediated signaling and the roles they play in neurodegenerative and neurological diseases.
Collapse
Affiliation(s)
- Stephanie M. Grant
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA;
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Sharon DeMorrow
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA;
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
- Research Division, Central Texas Veterans Healthcare System, Austin, TX 78712, USA
| |
Collapse
|
8
|
Dosedělová V, Itterheimová P, Kubáň P. Analysis of bile acids in human biological samples by microcolumn separation techniques: A review. Electrophoresis 2020; 42:68-85. [PMID: 32645223 DOI: 10.1002/elps.202000139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/13/2022]
Abstract
Bile acids are a group of compounds essential for lipid digestion and absorption with a steroid skeleton and a carboxylate side chain usually conjugated to glycine or taurine. Bile acids are regulatory molecules for a number of metabolic processes and can be used as biomarkers of various disorders. Since the middle of the twentieth century, the detection of bile acids has evolved from simple qualitative analysis to accurate quantification in complicated mixtures. Advanced methods are required to characterize and quantify individual bile acids in these mixtures. This article overviews the literature from the last two decades (2000-2020) and focuses on bile acid analysis in various human biological samples. The methods for sample preparation, including the sample treatment of conventional (blood plasma, blood serum, and urine) and unconventional samples (bile, saliva, duodenal/gastric juice, feces, etc.) are shortly discussed. Eventually, the focus is on novel analytical approaches and methods for each particular biological sample, providing an overview of the microcolumn separation techniques, such as high-performance liquid chromatography, gas chromatography, and capillary electrophoresis, used in their analysis. This is followed by a discussion on selected clinical applications.
Collapse
Affiliation(s)
- Věra Dosedělová
- Department of Bioanalytical Instrumentation, CEITEC Masaryk University, Brno, Czech Republic
| | - Petra Itterheimová
- Department of Bioanalytical Instrumentation, CEITEC Masaryk University, Brno, Czech Republic
| | - Petr Kubáň
- Department of Bioanalytical Instrumentation, Institute of Analytical Chemistry, Academy of Sciences of the Czech Republic, Brno, Czech Republic
| |
Collapse
|
9
|
Beaudoin JJ, Bezençon J, Sjöstedt N, Fallon JK, Brouwer KLR. Role of Organic Solute Transporter Alpha/Beta in Hepatotoxic Bile Acid Transport and Drug Interactions. Toxicol Sci 2020; 176:34-35. [PMID: 32294204 PMCID: PMC7357176 DOI: 10.1093/toxsci/kfaa052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Organic solute transporter (OST) α/β is a key bile acid transporter expressed in various organs, including the liver under cholestatic conditions. However, little is known about the involvement of OSTα/β in bile acid-mediated drug-induced liver injury (DILI), a major safety concern in drug development. This study investigated whether OSTα/β preferentially transports more hepatotoxic, conjugated, primary bile acids and to what extent xenobiotics inhibit this transport. Kinetic studies with OSTα/β-overexpressing cells revealed that OSTα/β preferentially transported bile acids in the following order: taurochenodeoxycholate > glycochenodeoxycholate > taurocholate > glycocholate. The apparent half-maximal inhibitory concentrations for OSTα/β-mediated bile acid (5 µM) transport inhibition by fidaxomicin, troglitazone sulfate, and ethinyl estradiol were: 210, 334, and 1050 µM, respectively, for taurochenodeoxycholate; 97.6, 333, and 337 µM, respectively, for glycochenodeoxycholate; 140, 265, and 527 µM, respectively, for taurocholate; 59.8, 102, and 117 µM, respectively, for glycocholate. The potential role of OSTα/β in hepatocellular glycine-conjugated bile acid accumulation and cholestatic DILI was evaluated using sandwich-cultured human hepatocytes (SCHH). Treatment of SCHH with the farnesoid X receptor agonist chenodeoxycholate (100 µM) resulted in substantial OSTα/β induction, among other proteomic alterations, reducing glycochenodeoxycholate and glycocholate accumulation in cells+bile 4.0- and 4.5-fold, respectively. Treatment of SCHH with troglitazone and fidaxomicin together under cholestatic conditions resulted in increased hepatocellular toxicity compared with either compound alone, suggesting that OSTα/β inhibition may accentuate DILI. In conclusion, this study provides insights into the role of OSTα/β in preferential disposition of bile acids associated with hepatotoxicity, the impact of xenobiotics on OSTα/β-mediated bile acid transport, and the role of this transporter in SCHH and cholestatic DILI.
Collapse
Affiliation(s)
| | | | - Noora Sjöstedt
- Division of Pharmacotherapy and Experimental Therapeutics
| | - John K Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599
| | | |
Collapse
|
10
|
Zhang J, Gao LZ, Chen YJ, Zhu PP, Yin SS, Su MM, Ni Y, Miao J, Wu WL, Chen H, Brouwer KLR, Liu CX, Xu L, Jia W, Lan K. Continuum of Host-Gut Microbial Co-metabolism: Host CYP3A4/3A7 are Responsible for Tertiary Oxidations of Deoxycholate Species. Drug Metab Dispos 2019; 47:283-294. [PMID: 30606729 PMCID: PMC6378331 DOI: 10.1124/dmd.118.085670] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/31/2018] [Indexed: 02/05/2023] Open
Abstract
The gut microbiota modifies endogenous primary bile acids (BAs) to produce exogenous secondary BAs, which may be further metabolized by cytochrome P450 enzymes (P450s). Our primary aim was to examine how the host adapts to the stress of microbe-derived secondary BAs by P450-mediated oxidative modifications on the steroid nucleus. Five unconjugated tri-hydroxyl BAs that were structurally and/or biologically associated with deoxycholate (DCA) were determined in human biologic samples by liquid chromatography-tandem mass spectrometry in combination with enzyme-digestion techniques. They were identified as DCA-19-ol, DCA-6β-ol, DCA-5β-ol, DCA-6α-ol, DCA-1β-ol, and DCA-4β-ol based on matching in-laboratory synthesized standards. Metabolic inhibition assays in human liver microsomes and recombinant P450 assays revealed that CYP3A4 and CYP3A7 were responsible for the regioselective oxidations of both DCA and its conjugated forms, glycodeoxycholate (GDCA) and taurodeoxycholate (TDCA). The modification of secondary BAs to tertiary BAs defines a host liver (primary BAs)-gut microbiota (secondary BAs)-host liver (tertiary BAs) axis. The regioselective oxidations of DCA, GDCA, and TDCA by CYP3A4 and CYP3A7 may help eliminate host-toxic DCA species. The 19- and 4β-hydroxylation of DCA species demonstrated outstanding CYP3A7 selectivity and may be useful as indicators of CYP3A7 activity.
Collapse
Affiliation(s)
- Jian Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Ling-Zhi Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Yu-Jie Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Ping-Ping Zhu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Shan-Shan Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Ming-Ming Su
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Yan Ni
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Jia Miao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Wen-Lin Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Hong Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Kim L R Brouwer
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Chang-Xiao Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Liang Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Wei Jia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| | - Ke Lan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China (J.Z., L.Z.G., Y.J.C., P.P.Z., S.S.Y., L.X., K.L.); Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii (M.M.S., Y.N., W.J.); Institute of Clinical Pharmacology, West China Hospital, Sichuan University, Chengdu, China (J.M.); Chengdu Institutes for Food and Drug Control, Chengdu, China (W.L.W., H.C.); UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina (K.L.R.B.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.X.L.); and Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (S.S.Y.)
| |
Collapse
|
11
|
Fernández-Murga ML, Petrov PD, Conde I, Castell JV, Goméz-Lechón MJ, Jover R. Advances in drug-induced cholestasis: Clinical perspectives, potential mechanisms and in vitro systems. Food Chem Toxicol 2018; 120:196-212. [PMID: 29990576 DOI: 10.1016/j.fct.2018.07.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 12/12/2022]
Abstract
Despite growing research, drug-induced liver injury (DILI) remains a serious issue of increasing importance to the medical community that challenges health systems, pharmaceutical industries and drug regulatory agencies. Drug-induced cholestasis (DIC) represents a frequent manifestation of DILI in humans, which is characterised by an impaired canalicular bile flow resulting in a detrimental accumulation of bile constituents in blood and tissues. From a clinical point of view, cholestatic DILI generates a wide spectrum of presentations and can be a diagnostic challenge. The drug classes mostly associated with DIC are anti-infectious, anti-diabetic, anti-inflammatory, psychotropic and cardiovascular agents, steroids, and other miscellaneous drugs. The molecular mechanisms of DIC have been investigated since the 1980s but they remain debatable. It is recognised that altered expression and/or function of hepatobiliary membrane transporters underlies some forms of cholestasis, and this and other concomitant mechanisms are very likely in DIC. Deciphering these processes may pave the ways for diagnosis, prognosis and prevention, for which currently major gaps and caveats exist. In this review, we summarise recent advances in the field of DIC, including clinical aspects, the potential mechanisms postulated so far and the in vitro systems that can be useful to investigate and identify new cholestatic drugs.
Collapse
Affiliation(s)
- M Leonor Fernández-Murga
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Petar D Petrov
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Isabel Conde
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Jose V Castell
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Spain
| | - M José Goméz-Lechón
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain.
| | - Ramiro Jover
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Spain.
| |
Collapse
|
12
|
Zhu P, Zhang J, Chen Y, Yin S, Su M, Xie G, Brouwer KLR, Liu C, Lan K, Jia W. Analysis of human C24 bile acids metabolome in serum and urine based on enzyme digestion of conjugated bile acids and LC-MS determination of unconjugated bile acids. Anal Bioanal Chem 2018; 410:5287-5300. [PMID: 29907951 DOI: 10.1007/s00216-018-1183-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/20/2018] [Accepted: 06/04/2018] [Indexed: 01/01/2023]
Abstract
Host-gut microbiota metabolic interactions are closely associated with health and disease. A manifestation of such co-metabolism is the vast structural diversity of bile acids (BAs) involving both oxidative stereochemistry and conjugation. Herein, we describe the development and validation of a LC-MS-based method for the analysis of human C24 BA metabolome in serum and urine. The method has high throughput covering the discrimination of oxidative stereochemistry of unconjugated species in a 15-min analytical cycle. The validated quantitative performance provided an indirect way to ascertain the conjugation patterns of BAs via enzyme-digestion protocols that incorporated the enzymes, sulfatase, β-glucuronidase, and choloylglycine hydrolase. Application of the method has led to the detection of at least 70 unconjugated BAs including 27 known species and 43 newly found species in the post-prandial serum and urine samples from 7 nonalcoholic steatohepatitis patients and 13 healthy volunteers. Newly identified unconjugated BAs included 3α, 12β-dihydroxy-5β-cholan-24-oic acid, 12α-hydroxy-3-oxo-5β-cholan-24-oic acid, and 3α, 7α, 12β-trihydroxy-5β-cholan-24-oic acid. High-definition negative fragment spectra of the other major unknown species were acquired to facilitate future identification endeavors. An extensive conjugation pattern is the major reason for the "invisibility" of the newly found BAs to other common analytical methods. Metabolomic analysis of the total unconjugated BA profile in combination with analysis of their conjugation patterns and urinary excretion tendencies have provided substantial insights into the interconnected roles of host and gut microbiota in maintaining BA homeostasis. It was proposed that the urinary total BA profile may serve as an ideal footprint for the functional status of the host-gut microbial BA co-metabolism. In summary, this work provided a powerful tool for human C24 BA metabolome analysis that bridges the gap between GC-MS techniques in the past age and LC-MS techniques currently prevailing in biomedical researches. Further applications of the present method in clinical, translational research, and other biomedical explorations will continue to boost the construction of a host-gut microbial co-metabolism network of BAs and thus facilitate the decryption of BA-mediated host-gut microbiota crosstalk in health and diseases. Graphical abstract ᅟ.
Collapse
Affiliation(s)
- Pingping Zhu
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jian Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yujie Chen
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Shanshan Yin
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Mingming Su
- Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, HI, 96801, USA
| | - Guoxiang Xie
- Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, HI, 96801, USA
| | - Kim L R Brouwer
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Changxiao Liu
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, 300193, China
| | - Ke Lan
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China. .,Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, 610000, China.
| | - Wei Jia
- Metabolomics Shared Resource, University of Hawaii Cancer Center, Honolulu, HI, 96801, USA.
| |
Collapse
|
13
|
Mostarda S, Passeri D, Carotti A, Cerra B, Colliva C, Benicchi T, Macchiarulo A, Pellicciari R, Gioiello A. Synthesis, physicochemical properties, and biological activity of bile acids 3-glucuronides: Novel insights into bile acid signalling and detoxification. Eur J Med Chem 2017; 144:349-358. [PMID: 29275233 DOI: 10.1016/j.ejmech.2017.12.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/29/2017] [Accepted: 12/11/2017] [Indexed: 12/21/2022]
Abstract
Glucuronidation is considered an important detoxification pathway of bile acids especially in cholestatic conditions. Glucuronides are less toxic than the parent free forms and are more easily excreted in urine. However, the pathophysiological significance of bile acid glucuronidation is still controversial and debated among the scientific community. Progress in this field has been strongly limited by the lack of appropriate methods for the preparation of pure glucuronides in the amount needed for biological and pharmacological studies. In this work, we have developed a new synthesis of bile acid C3-glucuronides enabling the convenient preparation of gram-scale quantities. The synthesized compounds have been characterized in terms of physicochemical properties and abilities to modulate key nuclear receptors including the farnesoid X receptor (FXR). In particular, we found that C3-glucuronides of chenodeoxycholic acid and lithocholic acid, respectively the most abundant and potentially cytotoxic species formed in patients affected by cholestasis, behave as FXR agonists and positively regulate the gene expression of transporter proteins, the function of which is critical in human conditions related to imbalances of bile acid homeostasis.
Collapse
Affiliation(s)
- Serena Mostarda
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1, 06123 Perugia, Italy
| | | | - Andrea Carotti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1, 06123 Perugia, Italy
| | - Bruno Cerra
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1, 06123 Perugia, Italy
| | | | | | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1, 06123 Perugia, Italy
| | | | - Antimo Gioiello
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1, 06123 Perugia, Italy.
| |
Collapse
|
14
|
Cao L, Che Y, Meng T, Deng S, Zhang J, Zhao M, Xu W, Wang D, Pu Z, Wang G, Hao H. Repression of intestinal transporters and FXR-FGF15 signaling explains bile acids dysregulation in experimental colitis-associated colon cancer. Oncotarget 2017; 8:63665-63679. [PMID: 28969019 PMCID: PMC5609951 DOI: 10.18632/oncotarget.18885] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 06/02/2017] [Indexed: 12/11/2022] Open
Abstract
Bile acids (BAs) are important endogenous signaling molecules that play vital roles in the pathological development of various diseases including colitis-associated cancer (CAC). BAs were previously found dysregulated under conditions of CAC; however, the exact patterns and underlying molecular mechanisms remain largely elusive. Based on the development of a method for comprehensive analysis of BAs, this study aims to elucidate the dysregulation patterns and involved mechanisms in a typical CAC model induced by azoxymethane (AOM)/dextran sodium sulfate (DSS). CAC mice showed decreased BAs transformation in gut and glucuronidation in colon, leading to accumulation of primary BAs but reduction of secondary BAs in colon. CAC mice were characterized by an accumulation of BAs in various compartments except ileum, which is in line with repressed ileal FXR-FGF15 feedback signaling and the increased expression of hepatic CYP7A1. The compromised ileal FXR-FGF15 signaling was caused in part by the reduced absorption of FXR ligands including free and tauro-conjungated BAs due to the downregulation of various transporters of BAs in the ileum of CAC mice.
Collapse
Affiliation(s)
- Lijuan Cao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan Che
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Tuo Meng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Shanshan Deng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Min Zhao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Wanfeng Xu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Dandan Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Zhichen Pu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
15
|
Saraswathi V, Perriotte-Olson C, Ganesan M, Desouza CV, Alnouti Y, Duryee MJ, Thiele GM, Nordgren TM, Clemens DL. A combination of dietary N-3 fatty acids and a cyclooxygenase-1 inhibitor attenuates nonalcoholic fatty liver disease in mice. J Nutr Biochem 2017; 42:149-159. [PMID: 28187366 DOI: 10.1016/j.jnutbio.2017.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/13/2016] [Accepted: 01/19/2017] [Indexed: 12/12/2022]
Abstract
We sought to determine whether a combination of purified n-3 fatty acids (n-3) and SC-560 (SC), a cyclooxygenase-1-specific inhibitor, is effective in ameliorating nonalcoholic fatty liver disease in obesity. Female wild-type mice were fed a high-fat and high-cholesterol diet (HF) supplemented with n-3 in the presence or absence of SC. Mice treated with SC alone exhibited no change in liver lipids, whereas n-3-fed mice tended to have lower hepatic lipids. Mice given n-3+SC had significantly lower liver lipids compared with HF controls indicating enhanced lipid clearance. Total and sulfated bile acids were significantly higher only in n-3+SC-treated mice compared with chow diet (CD) controls. Regarding mechanisms, the level of pregnane X receptor (PXR), a nuclear receptor regulating drug/bile detoxification, was significantly higher in mice given n-3 or n-3+SC. Studies in precision-cut liver slices and in cultured hepatoma cells showed that n-3+SC enhanced not only the expression/activation of PXR and its target genes but also the expression of farnesoid X receptor (FXR), another regulator of bile synthesis/clearance, indicating that n-3+SC can induce both PXR and FXR. The mRNA level of FGFR4 which inhibits bile formation showed a significant reduction in Huh 7 cells upon n-3 and n-3+SC treatment. PXR overexpression in hepatoma cells confirmed that n-3 or SC each induced the expression of PXR target genes and in combination had an enhanced effect. Our findings suggest that combining SC with n-3 potentiates its lipid-lowering effect, in part, by enhanced PXR and/or altered FXR/FGFR4 signaling.
Collapse
Affiliation(s)
- Viswanathan Saraswathi
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Curtis Perriotte-Olson
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA
| | - Murali Ganesan
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cyrus V Desouza
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Yazen Alnouti
- Department of Pharmaceutical Science, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael J Duryee
- Department of Internal Medicine, Division of Rheumatology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pharmaceutical Science, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Tara M Nordgren
- Department of Internal Medicine, Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dahn L Clemens
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
16
|
Naito H, Jia X, Yetti H, Yanagiba Y, Tamada H, Kitamori K, Hayashi Y, Wang D, Kato M, Ishii A, Nakajima T. Importance of detoxifying enzymes in differentiating fibrotic development between SHRSP5/Dmcr and SHRSP rats. Environ Health Prev Med 2016; 21:368-381. [PMID: 27209494 DOI: 10.1007/s12199-016-0539-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 05/09/2016] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES High-fat and -cholesterol diet (HFC) induced fibrotic steatohepatitis in stroke-prone spontaneously hypertensive rat (SHRSP) 5/Dmcr, the fifth substrain from SHRSP, by dysregulating bile acid (BA) kinetics. This study aimed to clarify the histopathological and BA kinetic differences in HFC-induced fibrosis between SHRSP5/Dmcr and SHRSP. METHODS Ten-week-old male SHRSP5/Dmcr and SHRSP were randomly allocated to groups and fed with either control diet or HFC for 2 and 8 weeks. The liver histopathology, biochemical features, and molecular signaling involved in BA kinetics were measured. RESULTS HFC caused more severe hepatocyte ballooning, macrovesicular steatosis and fibrosis in SHRSP5/Dmcr than in SHRSP. It was noted that fibrosis was disproportionately formed in retroperitoneal side of both strains. As for BA kinetics, HFC greatly increased the level of Cyp7a1 and Cyp7b1 to the same degree in both strains at 8 weeks, while multidrug resistance-associated protein 3 was greater in SHRSP5/Dmcr than SHRSP. The diet decreased the level of bile salt export pump by the same degree in both strains, while constitutive androstane receptor, pregnane X receptor, and UDP-glucuronosyltransferase activity more prominent in SHRSP5/Dmcr than SHRSP at 8 weeks. In the fibrosis-related genes, only expression of collagen, type I, alpha 1 mRNA was greater in SHRSP5/Dmcr than SHRSP. CONCLUSIONS The greater progression of fibrosis in SHRSP5/Dmcr induced by HFC may be due to greater suppression of UDP-glucuronosyltransferase activity detoxifying toxicants, such as hydrophobic BAs.
Collapse
Affiliation(s)
- Hisao Naito
- Department of Public Health, Fujita Health University School of Medicine, Dengakugakubo 1-98, Kutsukake-cho, Toyoake, 470-1192, Japan. .,Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Xiaofang Jia
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Husna Yetti
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukie Yanagiba
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hazuki Tamada
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan.,College of Human Life and Environment, Kinjo Gakuin University, Nagoya, Japan
| | - Kazuya Kitamori
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan.,College of Human Life and Environment, Kinjo Gakuin University, Nagoya, Japan
| | - Yumi Hayashi
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Dong Wang
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Ishii
- Department of Legal Medicine and Bioethics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tamie Nakajima
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan.,College of Life and Health Sciences, Chubu University, Kasugai, Japan
| |
Collapse
|
17
|
In search of the active metabolites of an anticancer piperazinedione, TW01003, in rats. BIOMED RESEARCH INTERNATIONAL 2014; 2014:793504. [PMID: 24864259 PMCID: PMC4016869 DOI: 10.1155/2014/793504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 03/02/2014] [Indexed: 11/25/2022]
Abstract
TW01003, a piperazinedione derivative designed as an antimitotic agent, exhibited potent anticancer and antiangiogenesis activities in mice. However, oral administration of this compound in rats led to poor systemic bioavailability which suggested that in vivo efficacy might come from its metabolites. This report describes the identification of TW01003 metabolites in pig and Wistar rats. Following intravenous administration of TW01003, pig urine samples were subjected to sulfatase and glucuronidase treatment to monitor the biotransformation products. Rats were given TW01003 both intravenously and orally, and blood samples were collected and then analyzed by HPLC to quantitatively determine the metabolic transformation of TW01003 to its metabolite. A sulfate conjugate, TW01003 sulfate, was identified as the major metabolite for TW01003 after intravenous injection in both pig and rats. However, in rats, the glucuronide conjugate became major metabolite 30 min after TW01003 oral dosing. Pharmacokinetic analysis after intravenous administration of TW01003 indicated that TW01003 sulfate had a systemic bioavailability 2.5 times higher, volume of distribution three times higher, residence time seven times longer, and clearance rate 2.3 times lower compared to TW01003. Our results indicate that the potent anticancer and antiangiogenesis activities of TW01003 might not come from TW01003 per se but from its metabolites TW01003 sulfate.
Collapse
|
18
|
Marine and semi-synthetic hydroxysteroids as new scaffolds for pregnane X receptor modulation. Mar Drugs 2014; 12:3091-115. [PMID: 24871460 PMCID: PMC4071567 DOI: 10.3390/md12063091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 04/29/2014] [Accepted: 04/30/2014] [Indexed: 12/16/2022] Open
Abstract
In recent years many sterols with unusual structures and promising biological profiles have been identified from marine sources. Here we report the isolation of a series of 24-alkylated-hydroxysteroids from the soft coral Sinularia kavarattiensis, acting as pregnane X receptor (PXR) modulators. Starting from this scaffold a number of derivatives were prepared and evaluated for their ability to activate the PXR by assessing transactivation and quantifying gene expression. Our study reveals that ergost-5-en-3β-ol (4) induces PXR transactivation in HepG2 cells and stimulates the expression of the PXR target gene CYP3A4. To shed light on the molecular basis of the interaction between these ligands and PXR, we investigated, through docking simulations, the binding mechanism of the most potent compound of the series, 4, to the PXR. Our findings provide useful functional and structural information to guide further investigations and drug design.
Collapse
|
19
|
Perreault M, Białek A, Trottier J, Verreault M, Caron P, Milkiewicz P, Barbier O. Role of glucuronidation for hepatic detoxification and urinary elimination of toxic bile acids during biliary obstruction. PLoS One 2013; 8:e80994. [PMID: 24244729 PMCID: PMC3828276 DOI: 10.1371/journal.pone.0080994] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/08/2013] [Indexed: 12/15/2022] Open
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.
Collapse
Affiliation(s)
- Martin Perreault
- Laboratory of molecular pharmacology, CHU-Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Andrzej Białek
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Jocelyn Trottier
- Laboratory of molecular pharmacology, CHU-Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Mélanie Verreault
- Laboratory of molecular pharmacology, CHU-Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Patrick Caron
- Laboratory of molecular pharmacology, CHU-Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Piotr Milkiewicz
- Liver Research Laboratories, Pomeranian Medical University, Szczecin, Poland
- Liver Unit, Department of Surgery and Liver Transplantation, Warsaw Medical University, Warsaw, Poland
| | - Olivier Barbier
- Laboratory of molecular pharmacology, CHU-Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec, Canada
- * E-mail:
| |
Collapse
|
20
|
Quintero P, Arrese M. Regulation of bile acid metabolism: new insights from inside. Hepatology 2013; 58:1850-3. [PMID: 23775943 DOI: 10.1002/hep.26569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 05/07/2013] [Accepted: 05/26/2013] [Indexed: 01/17/2023]
Affiliation(s)
- Pablo Quintero
- Departamento de Gastroenterología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | |
Collapse
|
21
|
Bigo C, Caron S, Dallaire-Théroux A, Barbier O. Nuclear receptors and endobiotics glucuronidation: the good, the bad, and the UGT. Drug Metab Rev 2013; 45:34-47. [PMID: 23330540 DOI: 10.3109/03602532.2012.751992] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The recent progresses in molecular biology and pharmacology approaches allowed the characterization of a series of nuclear receptors (NRs) as efficient regulators of uridine diphosphate glucuronosyltransferase (UGT) genes activity. These regulatory processes ensure an optimized UGT expression in response to specific endo- and/or exogenous stimuli. Many of these NRs are activated by endobiotics that also are substrates for UGTs. Thus, by activating their receptors, these endogenous substances control their own conjugation, leading to the concept that glucuronidation is an important part of feed-forward/feedback mechanisms by which bioactive molecules control their own concentrations. On the other hand, numerous studies have established the pharmacological relevance of NR-UGT regulatory pathways in the response to therapeutic ligands. The present review article aims at providing a comprehensive view of the physiological and pharmacological importance of the NR regulation of the expression and activity of endobiotics-conjugating UGT enzymes. Selected examples will illustrate how the organism profits from the feed-forward/feedback mechanisms involving NR-UGT pathways, but also how such regulatory processes are involved in the initiation and/or progression of several pathological situations. Finally, we will discuss how the present pharmacopeia involves NR-dependent regulation of endobiotics glucuronidation, and whether the unexploited NR-UGT axes could serve as pharmacological targets for novel therapeutics to restore endobiotics homeostasis.
Collapse
Affiliation(s)
- Cyril Bigo
- Laboratory of Molecular Pharmacology, CHUQ Research Center and the Faculty of Pharmacy, Laval University, Québec City, Québec, Canada
| | | | | | | |
Collapse
|
22
|
Trottier J, Perreault M, Rudkowska I, Levy C, Dallaire-Theroux A, Verreault M, Caron P, Staels B, Vohl MC, Straka RJ, Barbier O. Profiling serum bile acid glucuronides in humans: gender divergences, genetic determinants, and response to fenofibrate. Clin Pharmacol Ther 2013; 94:533-43. [PMID: 23756370 PMCID: PMC4844538 DOI: 10.1038/clpt.2013.122] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 05/31/2013] [Indexed: 12/12/2022]
Abstract
Glucuronidation, catalyzed by UDP-glucuronosyltransferase (UGT) enzymes detoxifies cholestatic bile acids (BAs). We aimed at i) characterizing the circulating BA-glucuronide (-G) pool composition in humans, ii) evaluating how sex and UGT polymorphisms influence this composition, and iii) analyzing the effects of lipid-lowering drug fenofibrate on the circulating BA-G profile in 300 volunteers and 5 cholestatic patients. Eleven BA-Gs were determined in pre- and post-fenofibrate samples. Men exhibited higher BA-G concentrations, and various genotype/BA-G associations were discovered in relevant UGT genes. The chenodeoxycholic acid-3G concentration was associated with the UGT2B7 802C>T polymorphism. Glucuronidation assays confirmed the predominant role of UGT2B7 and UGT1A4 in CDCA-3G formation. Fenofibrate exposure increased the serum levels of 5 BA-G species, including CDCA-3G, and up-regulated expression of UGT1A4, but not UGT2B7, in hepatic cells. This study demonstrates that fenofibrate stimulates BA glucuronidation in humans, and thus reduces bile acid toxicity in the liver.
Collapse
Affiliation(s)
- J Trottier
- Laboratory of Molecular Pharmacology, Endocrinology, and Nephrology, CHU-Québec Research Centre and the Faculty of Pharmacy, Laval University, Quebec City, Québec, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Salvador JAR, Carvalho JFS, Neves MAC, Silvestre SM, Leitão AJ, Silva MMC, Sá e Melo ML. Anticancer steroids: linking natural and semi-synthetic compounds. Nat Prod Rep 2013; 30:324-74. [PMID: 23151898 DOI: 10.1039/c2np20082a] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Steroids, a widespread class of natural organic compounds occurring in animals, plants and fungi, have shown great therapeutic value for a broad array of pathologies. The present overview is focused on the anticancer activity of steroids, which is very representative of a rich structural molecular diversity and ability to interact with various biological targets and pathways. This review encompasses the most relevant discoveries on steroid anticancer drugs and leads through the last decade and comprises 668 references.
Collapse
Affiliation(s)
- Jorge A R Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, 3000-508, Coimbra, Portugal.
| | | | | | | | | | | | | |
Collapse
|
24
|
Jia X, Naito H, Yetti H, Tamada H, Kitamori K, Hayashi Y, Wang D, Yanagiba Y, Wang J, Ikeda K, Yamori Y, Nakajima T. Dysregulated bile acid synthesis, metabolism and excretion in a high fat-cholesterol diet-induced fibrotic steatohepatitis in rats. Dig Dis Sci 2013; 58:2212-22. [PMID: 23824403 PMCID: PMC3731517 DOI: 10.1007/s10620-013-2747-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 06/05/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Cholesterol over-intake is involved in the onset of nonalcoholic steatohepatitis (NASH), and hepatocellular bile acid (BA) accumulation correlates with liver injuries. However, how dietary cholesterol influences cholesterol and BA kinetics in NASH liver remains ambiguous and needs to be clarified. METHODS Molecular markers involved in cholesterol and BA kinetics were investigated at protein and mRNA levels in an already-established stroke-prone spontaneously hypertensive 5/Dmcr rat model with fibrotic steatohepatitis, by feeding a high fat-cholesterol (HFC) diet. RESULTS Unlike the control diet, the HFC diet deposited cholesterol greatly in rat livers, where 3-hydroxy-3-methylglutaryl CoA reductase, low-density lipoprotein (LDL) receptor and LDL receptor-related protein-1 were expectedly downregulated, especially at 8 and 14 weeks, suggesting that cholesterol synthesis and uptake in response to cholesterol accumulation may not be disorganized. The HFC diet did not upregulate liver X receptor-α, conversely, it enhanced classic BA synthesis by upregulating cholesterol 7α-hydroxylase but downregulating sterol 12α-hydroxylase, and influenced alternative synthesis by downregulating sterol 27-hydroxylase but upregulating oxysterol 7α-hydroxylase, mainly at 8 and 14 weeks, indicating that there were different productions of primary BA species. Unexpectedly, no feedback inhibition of BA synthesis by farnesoid X receptor occurred. Additionally, the HFC diet impaired BA detoxification by UDP-glucuronosyltransferase and sulfotransferase 2A1, and decreased excretion by bile salt export pump at 8 and 14 weeks, although it induced compensatory export by multidrug resistance-associated protein-3. The disturbed BA detoxification may correlate with suppressed pregnane X receptor and constitutive androstane receptor. CONCLUSIONS The HFC diet may accumulate BA in rat livers, which influences fibrotic steatohepatitis progression.
Collapse
Affiliation(s)
- Xiaofang Jia
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Hisao Naito
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Husna Yetti
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Hazuki Tamada
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan ,College of Human Life and Environment, Kinjo Gakuin University, Nagoya, 463-8521 Japan
| | - Kazuya Kitamori
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan ,College of Human Life and Environment, Kinjo Gakuin University, Nagoya, 463-8521 Japan
| | - Yumi Hayashi
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Dong Wang
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Yukie Yanagiba
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Juncai Wang
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Katsumi Ikeda
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, Nishinomiya, 663-8179 Japan
| | - Yukio Yamori
- Institute for World Health Development, Mukogawa Women’s University, Nishinomiya, 663-8143 Japan
| | - Tamie Nakajima
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan ,College of Life and Health Sciences, Chubu University, Kasugai, 487-8501 Japan
| |
Collapse
|
25
|
Hoekstra R, Nibourg GAA, van der Hoeven TV, Plomer G, Seppen J, Ackermans MT, Camus S, Kulik W, van Gulik TM, Elferink RPO, Chamuleau RAFM. Phase 1 and phase 2 drug metabolism and bile acid production of HepaRG cells in a bioartificial liver in absence of dimethyl sulfoxide. Drug Metab Dispos 2012; 41:562-7. [PMID: 23238784 DOI: 10.1124/dmd.112.049098] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The human liver cell line HepaRG has been recognized as a promising source for in vitro testing of metabolism and toxicity of compounds. However, currently the hepatic differentiation of these cells relies on exposure to dimethylsulfoxide (DMSO), which, as a side effect, has a cytotoxic effect and represses an all-round hepatic functionality. The AMC-bioartificial liver (AMC-BAL) is a three-dimensional bioreactor that has previously been shown to upregulate various liver functions of cultured cells. We therefore cultured HepaRG cells in the AMC-BAL without DMSO and characterized the drug metabolism. Within 14 days of culture, the HepaRG-AMC-BALs contained highly polarized viable liver-like tissue with heterogeneous expression of CYP3A4. We found a substantial metabolism of the tested substrates, ranging from 26% (UDP-glucuronosyltransferase 1A1), 47% (CYP3A4), to 240% (CYP2C9) of primary human hepatocytes. The CYP3A4 activity could be induced 2-fold by rifampicin, whereas CYP2C9 activity remained equally high. The HepaRG-AMC-BAL secreted bile acids at 43% the rate of primary human hepatocytes and demonstrated hydroxylation, conjugation, and transport of bile salts. Concluding, culturing HepaRG cells in the AMC-BAL yields substantial phase 1 and phase 2 drug metabolism, while maintaining high viability, rendering DMSO addition superfluous for the promotion of drug metabolism. Therefore, AMC-BAL culturing makes the HepaRG cells more suitable for testing metabolism and toxicity of drugs.
Collapse
Affiliation(s)
- Ruurdtje Hoekstra
- Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wang YG, Zhou JM, Ma ZC, Li H, Liang QD, Tan HL, Xiao CR, Zhang BL, Gao Y. Pregnane X receptor mediated-transcription regulation of CYP3A by glycyrrhizin: A possible mechanism for its hepatoprotective property against lithocholic acid-induced injury. Chem Biol Interact 2012; 200:11-20. [DOI: 10.1016/j.cbi.2012.08.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 08/17/2012] [Accepted: 08/27/2012] [Indexed: 10/27/2022]
|
27
|
Zhang Y, Hong JY, Rockwell CE, Copple BL, Jaeschke H, Klaassen CD. Effect of bile duct ligation on bile acid composition in mouse serum and liver. Liver Int 2012; 32:58-69. [PMID: 22098667 PMCID: PMC3263524 DOI: 10.1111/j.1478-3231.2011.02662.x] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 09/10/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cholestatic liver diseases can be caused by genetic defects, drug toxicities, hepatobiliary malignancies or obstruction of the biliary tract. Cholestasis leads to accumulation of bile acids (BAs) in hepatocytes. Direct toxicity of BAs is currently the most accepted hypothesis for cholestatic liver injury. However, information on which bile acids are actually accumulating during cholestasis is limited. AIM To assess the BA composition in liver and serum after bile duct ligation (BDL) in male C57Bl/6 mice between 6 h and 14 days and evaluate toxicity of the most abundant BAs. RESULTS Bile acid concentrations increased in liver (27-fold) and serum (1400-fold) within 6 h after surgery and remained elevated up to 14 days. BAs in livers of BDL mice became more hydrophilic than sham controls, mainly because of increased 6β-hydroxylation and taurine conjugation. Among the eight unconjugated and 16 conjugated BAs identified in serum and liver, only taurocholic acid (TCA), β-muricholic acid (βMCA) and TβMCA were substantially elevated representing >95% of these BAs over the entire time course. Although glycochenodeoxycholic acid and other conjugated BAs increased in BDL animals, the changes were several orders of magnitude lower compared with TCA, βMCA and TβMCA. A mixture of these BAs did not cause apoptosis or necrosis, but induced inflammatory gene expression in cultured murine hepatocytes. CONCLUSION The concentrations of cytotoxic BAs are insufficient to cause hepatocellular injury. In contrast, TCA, βMCA and TβMCA are able to induce pro-inflammatory mediators in hepatocytes. Thus, BAs act as inflammagens and not as cytotoxic mediators after BDL in mice.
Collapse
Affiliation(s)
- Youcai Zhang
- Department of Pharmacology, Toxicology and Therapeutics; University of Kansas Medical Center; Kansas City; KS; USA
| | - Ji-Young Hong
- Department of Pharmacology, Toxicology and Therapeutics; University of Kansas Medical Center; Kansas City; KS; USA
| | - Cheryl E. Rockwell
- Department of Pharmacology, Toxicology and Therapeutics; University of Kansas Medical Center; Kansas City; KS; USA
| | - Bryan L. Copple
- Department of Pharmacology, Toxicology and Therapeutics; University of Kansas Medical Center; Kansas City; KS; USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics; University of Kansas Medical Center; Kansas City; KS; USA
| | - Curtis D. Klaassen
- Department of Pharmacology, Toxicology and Therapeutics; University of Kansas Medical Center; Kansas City; KS; USA
| |
Collapse
|
28
|
Trottier J, Białek A, Caron P, Straka RJ, Milkiewicz P, Barbier O. Profiling circulating and urinary bile acids in patients with biliary obstruction before and after biliary stenting. PLoS One 2011; 6:e22094. [PMID: 21760958 PMCID: PMC3132779 DOI: 10.1371/journal.pone.0022094] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 06/15/2011] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Bile acids are considered as extremely toxic at the high concentrations reached during bile duct obstruction, but each acid displays variable cytotoxic properties. This study investigates how biliary obstruction and restoration of bile flow interferes with urinary and circulating levels of 17 common bile acids. Bile acids (conjugated and unconjugated) were quantified by liquid chromatography coupled with tandem mass spectrometry in serum and urine samples from 17 patients (8 men and 9 women) with biliary obstruction, before and after biliary stenting. Results were compared with serum concentrations measured in 40 age- and sex-paired control donors (20 men and 20 women). The total circulating bile acid concentration increases from 2.7 µM in control donors to 156.9 µM in untreated patients with biliary stenosis. Serum taurocholic and glycocholic acids exhibit 304- and 241-fold accumulations in patients with biliary obstruction compared to controls. The enrichment in chenodeoxycholic acid species reached a maximum of only 39-fold, while all secondary and 6α-hydroxylated species--except taurolithocholic acids--were either unchanged or significantly reduced. Stenting was efficient in restoring an almost normal circulating profile and in reducing urinary bile acids. CONCLUSION These results demonstrate that biliary obstruction affects differentially the circulating and/or urinary levels of the various bile acids. The observation that the most drastically affected acids correspond to the less toxic species supports the activation of self-protecting mechanisms aimed at limiting the inherent toxicity of bile acids in face of biliary obstruction.
Collapse
Affiliation(s)
- Jocelyn Trottier
- Laboratory of Molecular Pharmacology, Centre Hospitalier Universitaire de Québec (CHUQ) Research Center and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Andrzej Białek
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Patrick Caron
- Laboratory of Molecular Pharmacology, Centre Hospitalier Universitaire de Québec (CHUQ) Research Center and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Robert J. Straka
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Piotr Milkiewicz
- Liver Unit and Liver Research Laboratories, Pomeranian Medical University, Szczecin, Poland
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, Centre Hospitalier Universitaire de Québec (CHUQ) Research Center and the Faculty of Pharmacy, Laval University, Québec, Canada
| |
Collapse
|
29
|
Nuclear receptor PXR, transcriptional circuits and metabolic relevance. Biochim Biophys Acta Mol Basis Dis 2011; 1812:956-63. [PMID: 21295138 DOI: 10.1016/j.bbadis.2011.01.014] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 01/25/2011] [Accepted: 01/26/2011] [Indexed: 12/14/2022]
Abstract
The pregnane X receptor (PXR, NR1I2) is a ligand activated transcription factor that belongs to the nuclear hormone receptor (NR) superfamily. PXR is highly expressed in the liver and intestine, but low levels of expression have also been found in many other tissues. PXR plays an integral role in xenobiotic and endobiotic metabolism by regulating the expression of drug-metabolizing enzymes and transporters, as well as genes implicated in the metabolism of endobiotics. PXR exerts its transcriptional regulation by binding to its DNA response elements as a heterodimer with the retinoid X receptor (RXR) and recruitment of a host of coactivators. The biological and physiological implications of PXR activation are broad, ranging from drug metabolism and drug-drug interactions to the homeostasis of numerous endobiotics, such as glucose, lipids, steroids, bile acids, bilirubin, retinoic acid, and bone minerals. The purpose of this article is to provide an overview on the transcriptional circuits and metabolic relevance controlled by PXR. This article is part of a Special Issue entitled: Translating Nuclear Receptors from Health to Disease.
Collapse
|
30
|
Roberts RE, Glicksman C, Alaghband-Zadeh J, Sherwood RA, Akuji N, le Roux CW. The relationship between postprandial bile acid concentration, GLP-1, PYY and ghrelin. Clin Endocrinol (Oxf) 2011; 74:67-72. [PMID: 21039722 DOI: 10.1111/j.1365-2265.2010.03886.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Gut hormones peptide YY (PYY) and glucagon-like peptide-1 (GLP-1) play an integral role in appetite control and energy homeostasis. Entero-endocrine L-cells can be stimulated by nutrients and or bile acids to co-secrete PYY and GLP-1. The aim of this study was to determine the response of bile acids, PYY, GLP-1 and ghrelin after a test meal. DESIGN Twelve subjects with a BMI of 22·8 (0·52) kg/m² [mean (SEM)] received a 400 kcal test meal after which blood samples were taken every 30 min from 0 to 180 min. PYY, GLP-1 and ghrelin were measured by radioimmunoassays. Fractionated bile acids were measured by HPLC-MSMS. RESULTS PYY positively correlated with glycochenodeoxycholic acid (GCDCA) (rs = 0·23, P = 0·03) and taurochenodeoxycholic acid (TCDCA) (rs = 0·26, P = 0·02). GLP-1 positively correlated with GCDCA (rs = 0·22, P = 0·047) and glycodeoxycholic acid (GDCA) (rs = 0·3, P = 0·005). Ghrelin negatively correlated with GDCA (rs = -0·45, P≤ 0·0001), TCDCA (rs = -0·23, P = 0·034) and taurodeoxycholic acid (TDCA) (rs = -0·44, P≤ 0·0001). CONCLUSION PYY and GLP-1 responses correlated with chenodeoxycholic acid (CDCA) counterparts, whereas ghrelin negatively correlated with deoxycholic acid (DCA) counterparts. Specific bile acids may thus differentially affect entero-endocrine cells.
Collapse
Affiliation(s)
- R E Roberts
- King's College London School of Medicine, UK.
| | | | | | | | | | | |
Collapse
|
31
|
Regulation of drug-metabolizing enzymes by xenobiotic receptors: PXR and CAR. Adv Drug Deliv Rev 2010; 62:1238-49. [PMID: 20727377 DOI: 10.1016/j.addr.2010.08.006] [Citation(s) in RCA: 272] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 08/09/2010] [Accepted: 08/10/2010] [Indexed: 12/24/2022]
Abstract
Drug-metabolizing enzymes (DMEs) and transporters play pivotal roles in the disposition and detoxification of numerous foreign and endogenous chemicals. To accommodate chemical challenges, the expression of many DMEs and transporters is up-regulated by a group of ligand-activated transcription factors namely nuclear receptors (NRs). The importance of NRs in xenobiotic metabolism and clearance is best exemplified by the most promiscuous xenobiotic receptors: pregnane X receptor (PXR, NR1I2) and constitutive androstane/activated receptor (CAR, NR1I3). Together, these two receptors govern the inductive expression of a largely overlapping array of target genes encoding phase I and II DMEs, and drug transporters. Moreover, PXR and CAR also represent two distinctive mechanisms of NR activation, whereby CAR demonstrates both constitutive and ligand-independent activation. In this review, recent advances in our understanding of PXR and CAR as xenosensors are discussed with emphasis placed on the differences rather than similarities of these two xenobiotic receptors in ligand recognition and target gene regulation.
Collapse
|
32
|
Verreault M, Kaeding J, Caron P, Trottier J, Grosse L, Houssin E, Pâquet S, Perreault M, Barbier O. Regulation of endobiotics glucuronidation by ligand-activated transcription factors: physiological function and therapeutic potential. Drug Metab Rev 2010; 42:110-22. [PMID: 19831728 DOI: 10.3109/03602530903219220] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
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.
Collapse
Affiliation(s)
- Mélanie Verreault
- Laboratory of Molecular Pharmacology, CHUQ Research Center and Faculty of Pharmacy, Laval University, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lefebvre P, Cariou B, Lien F, Kuipers F, Staels B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev 2009; 89:147-91. [PMID: 19126757 DOI: 10.1152/physrev.00010.2008] [Citation(s) in RCA: 1177] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The incidence of the metabolic syndrome has taken epidemic proportions in the past decades, contributing to an increased risk of cardiovascular disease and diabetes. The metabolic syndrome can be defined as a cluster of cardiovascular disease risk factors including visceral obesity, insulin resistance, dyslipidemia, increased blood pressure, and hypercoagulability. The farnesoid X receptor (FXR) belongs to the superfamily of ligand-activated nuclear receptor transcription factors. FXR is activated by bile acids, and FXR-deficient (FXR(-/-)) mice display elevated serum levels of triglycerides and high-density lipoprotein cholesterol, demonstrating a critical role of FXR in lipid metabolism. In an opposite manner, activation of FXR by bile acids (BAs) or nonsteroidal synthetic FXR agonists lowers plasma triglycerides by a mechanism that may involve the repression of hepatic SREBP-1c expression and/or the modulation of glucose-induced lipogenic genes. A cross-talk between BA and glucose metabolism was recently identified, implicating both FXR-dependent and FXR-independent pathways. The first indication for a potential role of FXR in diabetes came from the observation that hepatic FXR expression is reduced in animal models of diabetes. While FXR(-/-) mice display both impaired glucose tolerance and decreased insulin sensitivity, activation of FXR improves hyperglycemia and dyslipidemia in vivo in diabetic mice. Finally, a recent report also indicates that BA may regulate energy expenditure in a FXR-independent manner in mice, via activation of the G protein-coupled receptor TGR5. Taken together, these findings suggest that modulation of FXR activity and BA metabolism may open new attractive pharmacological approaches for the treatment of the metabolic syndrome and type 2 diabetes.
Collapse
Affiliation(s)
- Philippe Lefebvre
- Institut National de la Sante et de la Recherche Medicale, Lille, France
| | | | | | | | | |
Collapse
|
34
|
Alnouti Y. Bile Acid sulfation: a pathway of bile acid elimination and detoxification. Toxicol Sci 2009; 108:225-46. [PMID: 19131563 DOI: 10.1093/toxsci/kfn268] [Citation(s) in RCA: 289] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sulfotransferase-2A1 catalyzes the formation of bile acid-sulfates (BA-sulfates). Sulfation of BAs increases their solubility, decreases their intestinal absorption, and enhances their fecal and urinary excretion. BA-sulfates are also less toxic than their unsulfated counterparts. Therefore, sulfation is an important detoxification pathway of BAs. Major species differences in BA sulfation exist. In humans, only a small proportion of BAs in bile and serum are sulfated, whereas more than 70% of BAs in urine are sulfated, indicating their efficient elimination in urine. The formation of BA-sulfates increases during cholestatic diseases. Therefore, sulfation may play an important role in maintaining BA homeostasis under pathologic conditions. Farnesoid X receptor, pregnane X receptor, constitutive androstane receptor, and vitamin D receptor are potential nuclear receptors that may be involved in the regulation of BA sulfation. This review highlights current knowledge about the enzymes and transporters involved in the formation and elimination of BA-sulfates, the effect of sulfation on the pharmacologic and toxicologic properties of BAs, the role of BA sulfation in cholestatic diseases, and the regulation of BA sulfation.
Collapse
Affiliation(s)
- Yazen Alnouti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| |
Collapse
|
35
|
Lipid-activated transcription factors control bile acid glucuronidation. Mol Cell Biochem 2009; 326:3-8. [PMID: 19130183 DOI: 10.1007/s11010-008-0001-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Accepted: 06/06/2008] [Indexed: 10/21/2022]
Abstract
Bile acids subserve important physiological functions in the control of cholesterol homeostasis. Indeed, hepatic bile acid synthesis and biliary excretion constitute the main route for cholesterol removal from the human body. On the other hand, bile acids serve as natural detergents for the intestinal absorption of dietary cholesterol. However, due to their detergent properties, bile acids are inherently cytotoxic, and their cellular level may be tightly controlled to avoid pathological situations such as cholestasis. Recent investigations have illustrated the crucial roles that a series of ligand-activated transcription factors has in the control of hepatic bile acids synthesis, transport and metabolism. Thus, the lipid-activated nuclear receptors, farnesoid X-receptor (FXR), liver X-receptor (LXR), pregnane X-receptor (PXR) and peroxisome proliferator-activated receptor alpha (PPAR alpha), modulate the expression and activity of genes controlling bile acid homeostasis in the liver. Several members of the UDP-glucuronosyltransferase (UGT) enzymes family are among the bile acid metabolizing enzymes regulated by these receptors. UGTs catalyze glucuronidation, a major phase II metabolic reaction, which converts hydrophobic bile acids into polar and urinary excretable metabolites. This article summarizes our recent observations on the regulation of bile acid conjugating UGTs upon pharmacological activation of lipid-activated receptors, with a particular interest for the role of PPAR alpha and LXRalpha in controlling human UGT1A3 expression.
Collapse
|
36
|
Dixon PH, Williamson C. The molecular genetics of intrahepatic cholestasis of pregnancy. Obstet Med 2008; 1:65-71. [PMID: 27582788 DOI: 10.1258/om.2008.080010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2008] [Indexed: 12/21/2022] Open
Abstract
Intrahepatic cholestasis of pregnancy (ICP), also known as obstetric cholestasis, causes maternal pruritus and liver impairment, and may be complicated by spontaneous preterm labour, fetal asphyxial events and intrauterine death. Our understanding of the aetiology of this disease has expanded significantly in the last decade due to a better understanding of the role played by genetic factors. In particular, advances in our knowledge of bile homeostasis has led to the identification of genes that play a considerable role in susceptibility to ICP. In this review we consider these advances and discuss the disease in the context of bile synthesis and metabolism, focusing on the genetic discoveries that have shed light on the molecular aetiology and pathophysiology of the condition.
Collapse
Affiliation(s)
- P H Dixon
- Maternal and Fetal Disease Group , Institute of Reproductive and Developmental Biology, Imperial College London , Du Cane Road, London W12 0NN , UK
| | - C Williamson
- Maternal and Fetal Disease Group , Institute of Reproductive and Developmental Biology, Imperial College London , Du Cane Road, London W12 0NN , UK
| |
Collapse
|
37
|
Abstract
Transcriptional control of cellular energy metabolic pathways is achieved by the coordinated action of numerous transcription factors and associated coregulators. Several members of the nuclear receptor superfamily have been shown to play important roles in this process because they can translate hormonal, nutrient, and metabolite signals into specific gene expression networks to satisfy energy demands in response to distinct physiological cues. Estrogen-related receptor (ERR) alpha, ERRbeta, and ERRgamma are nuclear receptors that have yet to be associated with a natural ligand and are thus considered as orphan receptors. However, the transcriptional activity of the ERRs is exquisitely sensitive to the presence of coregulatory proteins known to be essential for the control of energy homeostasis, and for all intents and purposes, these coregulators function as protein ligands for the ERRs. In particular, functional genomics and biochemical studies have shown that ERRalpha and ERRgamma operate as the primary conduits for the activity of members of the family of PGC-1 coactivators. As transcription factors, the ERRs control vast gene networks involved in all aspects of energy homeostasis, including fat and glucose metabolism as well as mitochondrial biogenesis and function. Phenotypic analyses of knockout mouse models have shown that all three ERRs are indispensable for proper development and/or survival of the organism when subjected to a variety of physiological challenges. The focus of this review is on the recent and rapid advances in understanding the functions of the ERRs in regulating bioenergetic pathways, with an emphasis on their roles in the specification of energetic properties required for cell- and tissue-specific functions.
Collapse
Affiliation(s)
- Vincent Giguère
- The Rosalind and Morris Goodman Cancer Centre, Cancer Pavilion, 1160 Pine Avenue West, Montreal, Quebec, Canada H3A 1A3.
| |
Collapse
|
38
|
Hepatocellular transport in acquired cholestasis: new insights into functional, regulatory and therapeutic aspects. Clin Sci (Lond) 2008; 114:567-88. [PMID: 18377365 DOI: 10.1042/cs20070227] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The recent overwhelming advances in molecular and cell biology have added enormously to our understanding of the physiological processes involved in bile formation and, by extension, to our comprehension of the consequences of their alteration in cholestatic hepatopathies. The present review addresses in detail this new information by summarizing a number of recent experimental findings on the structural, functional and regulatory aspects of hepatocellular transporter function in acquired cholestasis. This comprises (i) a short overview of the physiological mechanisms of bile secretion, including the nature of the transporters involved and their role in bile formation; (ii) the changes induced by nuclear receptors and hepatocyte-enriched transcription factors in the constitutive expression of hepatocellular transporters in cholestasis, either explaining the primary biliary failure or resulting from a secondary adaptive response; (iii) the post-transcriptional changes in transporter function and localization in cholestasis, including a description of the subcellular structures putatively engaged in the endocytic internalization of canalicular transporters and the involvement of signalling cascades in this effect; and (iv) a discussion on how this new information has contributed to the understanding of the mechanism by which anticholestatic agents exert their beneficial effects, or the manner in which it has helped the design of new successful therapeutic approaches to cholestatic liver diseases.
Collapse
|
39
|
Farrell EK, Merkler DJ. Biosynthesis, degradation and pharmacological importance of the fatty acid amides. Drug Discov Today 2008; 13:558-68. [PMID: 18598910 DOI: 10.1016/j.drudis.2008.02.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 01/29/2008] [Accepted: 02/18/2008] [Indexed: 01/08/2023]
Abstract
The identification of two biologically active fatty acid amides, N-arachidonoylethanolamine (anandamide) and oleamide, has generated a great deal of excitement and stimulated considerable research. However, anandamide and oleamide are merely the best-known and best-understood members of a much larger family of biologically occurring fatty acid amides. In this review, we will outline which fatty acid amides have been isolated from mammalian sources, detail what is known about how these molecules are made and degraded in vivo, and highlight their potential for the development of novel therapeutics.
Collapse
Affiliation(s)
- Emma K Farrell
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA
| | | |
Collapse
|
40
|
Activators of the farnesoid X receptor negatively regulate androgen glucuronidation in human prostate cancer LNCAP cells. Biochem J 2008; 410:245-53. [PMID: 17988216 DOI: 10.1042/bj20071136] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Androgens are major regulators of prostate cell growth and physiology. In the human prostate, androgens are inactivated in the form of hydrophilic glucuronide conjugates. These metabolites are formed by the two human UGT2B15 [UGT (UDP-glucuronosyltransferase) 2B15] and UGT2B17 enzymes. The FXR (farnesoid X receptor) is a bile acid sensor controlling hepatic and/or intestinal cholesterol, lipid and glucose metabolism. In the present study, we report the expression of FXR in normal and cancer prostate epithelial cells, and we demonstrate that its activation by chenodeoxycholic acid or GW4064 negatively interferes with the levels of UGT2B15 and UGT2B17 mRNA and protein in prostate cancer LNCaP cells. FXR activation also causes a drastic reduction of androgen glucuronidation in these cells. These results point out activators of FXR as negative regulators of androgen-conjugating UGT expression in the prostate. Finally, the androgen metabolite androsterone, which is also an activator of FXR, dose-dependently reduces the glucuronidation of androgens catalysed by UGT2B15 and UGT2B17 in an FXR-dependent manner in LNCaP cells. In conclusion, the present study identifies for the first time the activators of FXR as important regulators of androgen metabolism in human prostate cancer cells.
Collapse
|
41
|
Marschall HU, Wagner M, Zollner G, Trauner M. Clinical Hepatotoxicity. Regulation and Treatment with Inducers of Transport and Cofactors. Mol Pharm 2007; 4:895-910. [DOI: 10.1021/mp060133c] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hanns-Ulrich Marschall
- Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden, and Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Martin Wagner
- Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden, and Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Gernot Zollner
- Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden, and Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Michael Trauner
- Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden, and Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| |
Collapse
|
42
|
Trottier J, Verreault M, Grepper S, Monté D, Bélanger J, Kaeding J, Caron P, Inaba TT, Barbier O. Human UDP-glucuronosyltransferase (UGT)1A3 enzyme conjugates chenodeoxycholic acid in the liver. Hepatology 2006; 44:1158-70. [PMID: 17058234 DOI: 10.1002/hep.21362] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
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.
Collapse
Affiliation(s)
- Jocelyn Trottier
- Molecular Endocrinology and Oncology Research Center, CHUL Research Center and the faculty of pharmacy, Laval University, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Verreault M, Senekeo-Effenberger K, Trottier J, Bonzo JA, Bélanger J, Kaeding J, Staels B, Caron P, Tukey RH, Barbier O. The liver X-receptor alpha controls hepatic expression of the human bile acid-glucuronidating UGT1A3 enzyme in human cells and transgenic mice. Hepatology 2006; 44:368-78. [PMID: 16871576 DOI: 10.1002/hep.21259] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Glucuronidation, an important bile acid detoxification pathway, is catalyzed by enzymes belonging to the UDP-glucuronosyltransferase (UGT) family. Among UGT enzymes, UGT1A3 is considered the major human enzyme for the hepatic C24-glucuronidation of the primary chenodeoxycholic (CDCA) and secondary lithocholic (LCA) bile acids. We identify UGT1A3 as a positively regulated target gene of the oxysterol-activated nuclear receptor liver X-receptor alpha (LXRalpha). In human hepatic cells and human UGT1A transgenic mice, LXRalpha activators induce UGT1A3 mRNA levels and the formation of CDCA-24glucuronide (24G) and LCA-24G. Furthermore, a functional LXR response element (LXRE) was identified in the UGT1A3 promoter by site-directed mutagenesis, electrophoretic mobility shift assays and chromatin immunoprecipitation experiment. In addition, LXRalpha is found to interact with the SRC-1alpha and NCoR cofactors to regulate the UGT1A3 gene, but not with PGC-1beta. In conclusion, these observations establish LXRalpha as a crucial regulator of bile acid glucuronidation in humans and suggest that accumulation of oxysterols in hepatocytes during cholestasis favors bile acid detoxification as glucuronide conjugates. LXR agonists may be useful for stimulating both bile acid detoxification and cholesterol removal in cholestatic or hypercholesterolemic patients, respectively.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cells, Cultured
- Chromatin Immunoprecipitation
- DNA-Binding Proteins/drug effects
- DNA-Binding Proteins/metabolism
- Gene Expression
- Glucuronosyltransferase/drug effects
- Glucuronosyltransferase/genetics
- Glucuronosyltransferase/metabolism
- Hepatocytes/cytology
- Hepatocytes/metabolism
- Humans
- Hydrocarbons, Fluorinated
- In Vitro Techniques
- Liver X Receptors
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Orphan Nuclear Receptors
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- Receptors, Cytoplasmic and Nuclear/drug effects
- Receptors, Cytoplasmic and Nuclear/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sulfonamides/pharmacology
Collapse
Affiliation(s)
- Mélanie Verreault
- Molecular Endocrinology and Oncology Research Center, CHUL Research Center and the Faculty of Pharmacy, Laval University, Québec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|