1
|
Li T, Chiang JYL. Bile Acid Signaling in Metabolic and Inflammatory Diseases and Drug Development. Pharmacol Rev 2024; 76:1221-1253. [PMID: 38977324 PMCID: PMC11549937 DOI: 10.1124/pharmrev.124.000978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024] Open
Abstract
Bile acids are the end products of cholesterol catabolism. Hepatic bile acid synthesis accounts for a major fraction of daily cholesterol turnover in humans. Biliary secretion of bile acids generates bile flow and facilitates biliary secretion of lipids, endogenous metabolites, and xenobiotics. In intestine, bile acids facilitate the digestion and absorption of dietary lipids and fat-soluble vitamins. Through activation of nuclear receptors and G protein-coupled receptors and interaction with gut microbiome, bile acids critically regulate host metabolism and innate and adaptive immunity and are involved in the pathogenesis of cholestasis, metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, type-2 diabetes, and inflammatory bowel diseases. Bile acids and their derivatives have been developed as potential therapeutic agents for treating chronic metabolic and inflammatory liver diseases and gastrointestinal disorders. SIGNIFICANCE STATEMENT: Bile acids facilitate biliary cholesterol solubilization and dietary lipid absorption, regulate host metabolism and immunity, and modulate gut microbiome. Targeting bile acid metabolism and signaling holds promise for treating metabolic and inflammatory diseases.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| | - John Y L Chiang
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| |
Collapse
|
2
|
Zheng Z, Zhao Z, Li S, Lu X, Jiang M, Lin J, An Y, Xie Y, Xu M, Shen W, Guo GL, Huang Y, Li S, Zhang X, Xie W. Altenusin, a Nonsteroidal Microbial Metabolite, Attenuates Nonalcoholic Fatty Liver Disease by Activating the Farnesoid X Receptor. Mol Pharmacol 2017; 92:425-436. [PMID: 28739572 PMCID: PMC5588546 DOI: 10.1124/mol.117.108829] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/07/2017] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disease. The incidence of NAFLD has increased steadily due to its close association with the global epidemic of obesity and type 2 diabetes. However, there is no effective pharmacological therapy approved for NAFLD. Farnesoid X receptor (FXR), a member of the nuclear receptor subfamily, plays important roles in maintaining the homeostasis of bile acids, glucose, and lipids. FXR agonists have shown promise for the treatment of NAFLD. In this study, we report altenusin (2076A), a natural nonsteroidal fungal metabolite, as a novel selective agonist of FXR with an EC50 value of 3.2 ± 0.2 μM. Administration of 2076A protected mice from high-fat diet (HFD)-induced obesity by reducing the body weight and fat mass by 22.9% and 50.0%, respectively. Administration of 2076A also decreased the blood glucose level from 178.3 ± 12.4 mg/dl to 116.2 ± 4.1 mg/dl and the serum insulin level from 1.4 ± 0.6 ng/dl to 0.4 ± 0.1 ng/dl. Moreover, 2076A treatment nearly reversed HFD-induced hepatic lipid droplet accumulation and macrovesicular steatosis. These metabolic effects were abolished in FXR knockout mice. Mechanistically, the metabolic benefits of 2076A might have been accounted for by the increased insulin sensitivity and suppression of genes that are involved in hepatic gluconeogenesis and lipogenesis. In summary, we have uncovered a new class of nonsteroidal FXR agonist that shows promise in treating NAFLD and the associated metabolic syndrome.
Collapse
Affiliation(s)
- Zhihui Zheng
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Zanmei Zhao
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Shuqiang Li
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Xinhua Lu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Mengxi Jiang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Jie Lin
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Yunqi An
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Yang Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Wenbin Shen
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Grace L Guo
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Yixian Huang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Song Li
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Xuexia Zhang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| |
Collapse
|
3
|
Zhang L, Cheng Y, Du X, Chen S, Feng X, Gao Y, Li S, Liu L, Yang M, Chen L, Peng Z, Yang Y, Luo W, Wang R, Chen W, Chai J. Swertianlarin, an Herbal Agent Derived from Swertia mussotii Franch, Attenuates Liver Injury, Inflammation, and Cholestasis in Common Bile Duct-Ligated Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2015; 2015:948376. [PMID: 26273316 PMCID: PMC4530240 DOI: 10.1155/2015/948376] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/23/2014] [Accepted: 12/24/2014] [Indexed: 12/24/2022]
Abstract
Swertianlarin is an herbal agent abundantly distributed in Swertia mussotii Franch, a Chinese traditional herb used for treatment of jaundice. To study the therapeutic effect of swertianlarin on cholestasis, liver injury, serum proinflammatory cytokines, and bile salt concentrations were measured by comparing rats treated with swertianlarin 100 mg/kg/d or saline for 3, 7, or 14 days after bile duct ligation (BDL). Serum alanine aminotransferase (ATL) and aspartate aminotransferase (AST) levels were significantly decreased in BDL rats treated with swertianlarin for 14 days (P < 0.05). The reduced liver injury in BDL rats by swertianlarin treatment for 14 days was further confirmed by liver histopathology. Levels of serum tumor necrosis factor alpha (TNFα) were decreased by swertianlarin in BDL rats for 3 and 7 days (P < 0.05). Moreover, reductions in serum interleukins IL-1β and IL-6 levels were also observed in BDL rats treated with swertianlarin (P < 0.05). In addition, most of serum toxic bile salt concentrations (e.g., chenodeoxycholic acid (CDCA) and deoxycholic acid (DCA)) in cholestatic rats were decreased by swertianlarin (P < 0.05). In conclusion, the data suggest that swertianlarin derived from Swertia mussotii Franch attenuates liver injury, inflammation, and cholestasis in bile duct-ligated rats.
Collapse
Affiliation(s)
- Liangjun Zhang
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ying Cheng
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiaohuang Du
- Department of Traditional Chinese Medicine, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Sheng Chen
- Department of Pediatrics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xinchan Feng
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yu Gao
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Shaoxue Li
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Li Liu
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Mei Yang
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lei Chen
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Zhihong Peng
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yong Yang
- Chongqing Academy of Chinese Material Medicine, Chongqing 400065, China
| | - Weizao Luo
- Chongqing Academy of Chinese Material Medicine, Chongqing 400065, China
| | - Rongquan Wang
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wensheng Chen
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jin Chai
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
4
|
Pan X, Lee YK, Jeong H. Farnesoid X Receptor Agonist Represses Cytochrome P450 2D6 Expression by Upregulating Small Heterodimer Partner. Drug Metab Dispos 2015; 43:1002-7. [PMID: 25926433 PMCID: PMC4468439 DOI: 10.1124/dmd.115.064758] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 04/29/2015] [Indexed: 01/05/2023] Open
Abstract
Cytochrome P450 2D6 (CYP2D6) is a major drug-metabolizing enzyme responsible for eliminating approximately 20% of marketed drugs. Studies have shown that differential transcriptional regulation of CYP2D6 may contribute to large interindividual variability in CYP2D6-mediated drug metabolism. However, the factors governing CYP2D6 transcription are largely unknown. We previously demonstrated small heterodimer partner (SHP) as a novel transcriptional repressor of CYP2D6 expression. SHP is a representative target gene of the farnesoid X receptor (FXR). The objective of this study is to investigate whether an agonist of FXR, 3-(2,6-dichlorophenyl)-4-(3'-carboxy-2-chlorostilben-4-yl)oxymethyl-5-isopropylisoxazole (GW4064), alters CYP2D6 expression and activity. In CYP2D6-humanized transgenic mice, GW4064 decreased hepatic CYP2D6 expression and activity (by 2-fold) while increasing SHP expression (by 2-fold) and SHP recruitment to the CYP2D6 promoter. CYP2D6 repression by GW4064 was abrogated in Shp(-/-);CYP2D6 mice, indicating a critical role of SHP in CYP2D6 regulation by GW4064. Also, GW4064 decreased CYP2D6 expression (by 2-fold) in primary human hepatocytes, suggesting that the results obtained in CYP2D6-humanized transgenic mice can be translated to humans. This proof of concept study provides evidence for CYP2D6 regulation by an inducer of SHP expression, namely, the FXR agonist GW4064.
Collapse
Affiliation(s)
- Xian Pan
- Departments of Pharmacy Practice (H.J.) and Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois; and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (Y.K.L.)
| | - Yoon-Kwang Lee
- Departments of Pharmacy Practice (H.J.) and Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois; and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (Y.K.L.)
| | - Hyunyoung Jeong
- Departments of Pharmacy Practice (H.J.) and Biopharmaceutical Sciences (X.P., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois; and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (Y.K.L.)
| |
Collapse
|
5
|
Tan Y, Bi W, Zhang GX. Role and molecular mechanism of farnesoid X receptor in obstructive jaundice. Shijie Huaren Xiaohua Zazhi 2015; 23:2574-2581. [DOI: 10.11569/wcjd.v23.i16.2574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Obstructive jaundice is a common and frequently occurring disease, which can result in multiple organ dysfunction syndrome in serious conditions due to the abnormal accumulation of bile acids in blood. Farnesoid X receptor (FXR), a nuclear receptor for bile acid, plays a significant role in bile acid metabolism. Recent research demonstrates that FXR also participates in the regulation of the pathological and physiological processes during obstructive jaundice. In this article, we review the latest research about the role and molecular mechanism of FXR in obstructive jaundice, in order to explore new methods and strategies for curing the disease.
Collapse
|
6
|
Abstract
Bile acids are the end products of cholesterol catabolism. Hepatic bile acid synthesis accounts for a major fraction of daily cholesterol turnover in humans. Biliary secretion of bile acids generates bile flow and facilitates hepatobiliary secretion of lipids, lipophilic metabolites, and xenobiotics. In the intestine, bile acids are essential for the absorption, transport, and metabolism of dietary fats and lipid-soluble vitamins. Extensive research in the last 2 decades has unveiled new functions of bile acids as signaling molecules and metabolic integrators. The bile acid-activated nuclear receptors farnesoid X receptor, pregnane X receptor, constitutive androstane receptor, vitamin D receptor, and G protein-coupled bile acid receptor play critical roles in the regulation of lipid, glucose, and energy metabolism, inflammation, and drug metabolism and detoxification. Bile acid synthesis exhibits a strong diurnal rhythm, which is entrained by fasting and refeeding as well as nutrient status and plays an important role for maintaining metabolic homeostasis. Recent research revealed an interaction of liver bile acids and gut microbiota in the regulation of liver metabolism. Circadian disturbance and altered gut microbiota contribute to the pathogenesis of liver diseases, inflammatory bowel diseases, nonalcoholic fatty liver disease, diabetes, and obesity. Bile acids and their derivatives are potential therapeutic agents for treating metabolic diseases of the liver.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| | - John Y L Chiang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| |
Collapse
|
7
|
Chen P, Zeng H, Wang Y, Fan X, Xu C, Deng R, Zhou X, Bi H, Huang M. Low dose of oleanolic acid protects against lithocholic acid-induced cholestasis in mice: potential involvement of nuclear factor-E2-related factor 2-mediated upregulation of multidrug resistance-associated proteins. Drug Metab Dispos 2014; 42:844-52. [PMID: 24510383 DOI: 10.1124/dmd.113.056549] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oleanolic acid (OA) is a natural triterpenoid and has been demonstrated to protect against varieties of hepatotoxicants. Recently, however, OA at high doses was reported to produce apparent cholestasis in mice. In this study, we characterized the protective effect of OA at low doses against lithocholic acid (LCA)-induced cholestasis in mice and explored further mechanisms. OA cotreatment (5, 10, and 20 mg/kg, i.p.) significantly improved mouse survival rate, attenuated liver necrosis, and decreased serum alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase; more importantly, serum total bile acids and bilirubin, as well as hepatic total bile acids were also remarkably reduced. Gene and protein expression analysis showed that hepatic expression of multidrug resistance-associated protein 2 (Mrp2), Mrp3, and Mrp4 was significantly increased by OA cotreatment, whereas other bile acid metabolism- and transport-related genes, including Na+/taurocholate cotransporter, organic anion transporter 1b2, bile salt export pump, multidrug resistance protein 3, Cyp3a11, Cyp2b10, Sulfotransferase 2a1 (Sult2a1), and UDP-glucuronosyltransferase 1a1 (Ugt1a1), were only slightly changed. OA also caused increased nuclear factor-E2-related factor (Nrf2) mRNA expression and nuclear protein accumulation, whereas nuclear receptors farnesoid X receptor (FXR), pregnane X receptor (PXR), and constitutive androstane receptor were not significantly influenced by OA. Luciferase (Luc) assays performed in HepG2 cells illustrated that OA was a strong Nrf2 agonist with moderate PXR and weak FXR agonism. Finally, in mouse primary cultured hepatocytes, OA dose- and time-dependently induced expression of Mrp2, Mrp3, and Mrp4; however, this upregulation was abrogated when Nrf2 was silenced. In conclusion, OA produces a protective effect against LCA-induced hepatotoxicity and cholestasis, possibly due to Nrf2-mediated upregulation of Mrp2, Mrp3, and Mrp4.
Collapse
Affiliation(s)
- Pan Chen
- School of Pharmaceutical Sciences (P.C., H.Z., Y.W., X.F., R.D., X.Z., H.B., M.H.) and The First Affiliated Hospital (C.X.), Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Cuperus FJC, Claudel T, Gautherot J, Halilbasic E, Trauner M. The role of canalicular ABC transporters in cholestasis. Drug Metab Dispos 2014; 42:546-60. [PMID: 24474736 DOI: 10.1124/dmd.113.056358] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cholestasis, a hallmark feature of hepatobiliary disease, is characterized by the retention of biliary constituents. Some of these constituents, such as bile acids, inflict damage to hepatocytes and bile duct cells. This damage may lead to inflammation, fibrosis, cirrhosis, and eventually carcinogenesis, sequelae that aggravate the underlying disease and deteriorate clinical outcome. Canalicular ATP-binding cassette (ABC) transporters, which mediate the excretion of individual bile constituents, play a key role in bile formation and cholestasis. The study of these transporters and their regulatory nuclear receptors has revolutionized our understanding of cholestatic disease. This knowledge has served as a template to develop novel treatment strategies, some of which are currently already undergoing phase III clinical trials. In this review we aim to provide an overview of the structure, function, and regulation of canalicular ABC transporters. In addition, we will focus on the role of these transporters in the pathogenesis and treatment of cholestatic bile duct and liver diseases.
Collapse
Affiliation(s)
- Frans J C Cuperus
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | | | | | | |
Collapse
|
9
|
Molecular Signatures of Recurrent Hepatocellular Carcinoma Secondary to Hepatitis C Virus following Liver Transplantation. J Transplant 2013; 2013:878297. [PMID: 24377043 PMCID: PMC3860124 DOI: 10.1155/2013/878297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/25/2013] [Indexed: 01/12/2023] Open
Abstract
Chronic hepatitis C virus (HCV) induced hepatocellular carcinoma (HCC) is a primary indication for liver transplantation (LT). In western countries, the estimated rate of HCC recurrence following LT is between 15% and 20% and is a major cause of mortality. Currently, there is no standard method to treat patients who are at high risk for HCC recurrence. The aim of this study was to investigate the molecular signatures underlying HCC recurrence that may lead to future studies on gene regulation contributing to new therapeutic options. Two groups of patients were selected, one including patients with HCV who developed HCC recurrence (HCC-R) ≤3 years from LT and the second group including patients with HCV who did not have recurrent HCC (HCC-NR). Microarray analysis containing more than 29,000 known genes was performed on formalin-fixed-paraffin-embedded (FFPE) liver tissue from explanted livers. Gene expression profiling revealed 194 differentially regulated genes between the two groups. These genes belonged to cellular networks including cell cycle G1/S checkpoint regulators, RAN signaling, chronic myeloid leukemia signaling, molecular mechanisms of cancer, FXR/RXR activation and hepatic cholestasis. A subset of molecular signatures associated with HCC recurrence was found. The expression levels of these genes were validated by quantitative PCR analysis.
Collapse
|
10
|
Woolbright BL, Jaeschke H. Novel insight into mechanisms of cholestatic liver injury. World J Gastroenterol 2012; 18:4985-93. [PMID: 23049206 PMCID: PMC3460324 DOI: 10.3748/wjg.v18.i36.4985] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 04/12/2012] [Accepted: 04/20/2012] [Indexed: 02/06/2023] Open
Abstract
Cholestasis results in a buildup of bile acids in serum and in hepatocytes. Early studies into the mechanisms of cholestatic liver injury strongly implicated bile acid-induced apoptosis as the major cause of hepatocellular injury. Recent work has focused both on the role of bile acids in cell signaling as well as the role of sterile inflammation in the pathophysiology. Advances in modern analytical methodology have allowed for more accurate measuring of bile acid concentrations in serum, liver, and bile to very low levels of detection. Interestingly, toxic bile acid levels are seemingly far lower than previously hypothesized. The initial hypothesis has been based largely upon the exposure of μmol/L concentrations of toxic bile acids and bile salts to primary hepatocytes in cell culture, the possibility that in vivo bile acid concentrations may be far lower than the observed in vitro toxicity has far reaching implications in the mechanism of injury. This review will focus on both how different bile acids and different bile acid concentrations can affect hepatocytes during cholestasis, and additionally provide insight into how these data support recent hypotheses that cholestatic liver injury may not occur through direct bile acid-induced apoptosis, but may involve largely inflammatory cell-mediated liver cell necrosis.
Collapse
|
11
|
Kaimal R, Song X, Yan B, King R, Deng R. Differential modulation of farnesoid X receptor signaling pathway by the thiazolidinediones. J Pharmacol Exp Ther 2009; 330:125-34. [PMID: 19369578 PMCID: PMC2700164 DOI: 10.1124/jpet.109.151233] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 04/13/2009] [Indexed: 01/24/2023] Open
Abstract
Thiazolidinediones (TZD), including troglitazone, rosiglitazone, and pioglitazone, are agonists of peroxisome proliferator-activated receptor (PPAR)-gamma and belong to a class of insulin-sensitizing drugs for type 2 diabetes mellitus. However, member-specific, PPARgamma-independent activities and toxicity have been reported, especially for troglitazone. Currently, the underlying mechanisms are not fully understood. In this study, we demonstrated that troglitazone but not rosiglitazone or pioglitazone modulated expression of farnesoid X receptor (FXR) target genes bile salt export pump (BSEP) and small heterodimer partner (SHP) in Huh-7 cells. More specifically, troglitazone acted as a partial agonist of FXR to weakly increase BSEP and SHP expression but functioned as a potent antagonist to significantly suppress bile acid-induced expression. Consistent with the finding, troglitazone partially induced but markedly antagonized bile acid-mediated BSEP promoter transactivation. However, such modulating effects were not detected with rosiglitazone or pioglitazone. Using the crystal structure of ligand-bound FXR ligand binding domain (LBD), molecular docking predicted that troglitazone, but not rosiglitazone or pioglitazone, could form a stable complex with FXR LBD. The specific alpha-tocopherol side chain of troglitazone significantly contributed to the formation of such a stable complex through extensive interactions with FXR LBD. The docking model was further validated by functional analyses of a series of docking-guided FXR mutants. In summary, the data demonstrated that troglitazone, but not rosiglitazone or pioglitazone, was an FXR modulator and potently antagonized bile acid-induced expression of FXR target genes. Such differential modulation of FXR signaling pathway by TZDs may represent one of the mechanisms for member-specific, PPARgamma-independent activities and toxicity.
Collapse
Affiliation(s)
- Rajani Kaimal
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, College of Pharmacy, University of Rhode Island, 41 Lower College Rd., Kingston, RI 02881, USA
| | | | | | | | | |
Collapse
|
12
|
Ogura M, Nishida S, Ishizawa M, Sakurai K, Shimizu M, Matsuo S, Amano S, Uno S, Makishima M. Vitamin D3 modulates the expression of bile acid regulatory genes and represses inflammation in bile duct-ligated mice. J Pharmacol Exp Ther 2009; 328:564-70. [PMID: 18988769 DOI: 10.1124/jpet.108.145987] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Vitamin D receptor (VDR), a nuclear receptor that regulates calcium homeostasis, has been found to function as a receptor for secondary bile acids. Because the in vivo role of VDR in bile acid metabolism remains unknown, we investigated the effect of VDR activation in a mouse model of cholestasis. We treated mice with 1alpha-hydroxyvitamin D(3) [1alpha(OH)D(3)] after bile duct ligation (BDL) and examined mRNA expression and cytokine levels. 1alpha(OH)D(3) treatment altered the expression of genes involved in bile acid synthesis and transport in the liver, kidney, and intestine but did not decrease bile acid levels in the plasma and liver of BDL mice. 1alpha(OH)D(3) treatment suppressed mRNA expression of proinflammatory cytokines in the liver and strongly decreased the plasma levels of proinflammatory cytokines in BDL mice. These findings indicate that 1alpha(OH)D(3) regulates a network of bile acid metabolic genes and represses proinflammatory cytokine expression in BDL mice. VDR ligands have the potential to prevent the cholestasis-induced inflammatory response.
Collapse
Affiliation(s)
- Michitaka Ogura
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Kong B, Luyendyk JP, Tawfik O, Guo GL. Farnesoid X receptor deficiency induces nonalcoholic steatohepatitis in low-density lipoprotein receptor-knockout mice fed a high-fat diet. J Pharmacol Exp Ther 2009; 328:116-22. [PMID: 18948497 PMCID: PMC2685903 DOI: 10.1124/jpet.108.144600] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 10/22/2008] [Indexed: 12/23/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) comprises dysregulation of lipid metabolism and inflammation. Identification of the various genetic and environmental susceptibility factors for NASH may provide novel treatments to limit inflammation and fibrosis in patients. This study utilized a mouse model of hypercholesterolemia, low-density lipoprotein receptor knockout (LDLr(-/-)) mice fed a high-fat diet for 5 months, to test the hypothesis that farnesoid X receptor (FXR) deficiency contributed to NASH development. Either the high-fat diet or FXR deficiency increased serum alanine aminotransferase activity, whereas only FXR deficiency increased bile acid and alkaline phosphatase levels. FXR deficiency and high-fat feeding increased serum cholesterol and triglycerides. Although high fat led to macrosteatosis and hepatocyte ballooning in livers of mice regardless of genotype, no inflammatory infiltrate was observed in the livers of LDLr(-/-) mice. In contrast, in the livers of LDLr(-/-)/FXR(-/-) mice, foci of inflammatory cells were observed occasionally when fed the control diet and were greatly increased when fed the high-fat diet. Consistent with enhanced inflammatory cells, hepatic levels of tumor necrosis factor alpha and intercellular adhesion molecule-1 mRNA were increased by the high-fat diet in LDLr(-/-)/FXR(-/-) mice. In agreement with elevated levels of procollagen 1 alpha 1 and TGF-beta mRNA, type 1 collagen protein levels were increased in livers of LDLr(-/-)/FXR(-/-) mice fed a high-fat diet. In conclusion, FXR deficiency induces pathologic manifestations required for NASH diagnosis in a mouse model of hypercholesterolemia, including macrosteatosis, hepatocyte ballooning, and inflammation, which suggest a combination of FXR deficiency and high-fat diet is a risk factor for NASH development, and activation of FXR may be a therapeutic intervention in the treatment of NASH.
Collapse
Affiliation(s)
- Bo Kong
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center. 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | | | | | | |
Collapse
|
14
|
Suzuki T, Tamehiro N, Sato Y, Kobayashi T, Ishii-Watabe A, Shinozaki Y, Nishimaki-Mogami T, Hashimoto T, Asakawa Y, Inoue K, Ohno Y, Yamaguchi T, Kawanishi T. The Novel Compounds That Activate Farnesoid X Receptor: the Diversity of Their Effects on Gene Expression. J Pharmacol Sci 2008; 107:285-94. [DOI: 10.1254/jphs.08006fp] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
15
|
Sanyal S, Båvner A, Haroniti A, Nilsson LM, Lundåsen T, Rehnmark S, Witt MR, Einarsson C, Talianidis I, Gustafsson JÅ, Treuter E. Involvement of corepressor complex subunit GPS2 in transcriptional pathways governing human bile acid biosynthesis. Proc Natl Acad Sci U S A 2007; 104:15665-70. [PMID: 17895379 PMCID: PMC2000397 DOI: 10.1073/pnas.0706736104] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Coordinated regulation of bile acid biosynthesis, the predominant pathway for hepatic cholesterol catabolism, is mediated by few key nuclear receptors including the orphan receptors liver receptor homolog 1 (LRH-1), hepatocyte nuclear factor 4alpha (HNF4alpha), small heterodimer partner (SHP), and the bile acid receptor FXR (farnesoid X receptor). Activation of FXR initiates a feedback regulatory loop via induction of SHP, which suppresses LRH-1- and HNF4alpha-dependent expression of cholesterol 7alpha hydroxylase (CYP7A1) and sterol 12alpha hydroxylase (CYP8B1), the two major pathway enzymes. Here we dissect the transcriptional network governing bile acid biosynthesis in human liver by identifying GPS2, a stoichiometric subunit of a conserved corepressor complex, as a differential coregulator of CYP7A1 and CYP8B1 expression. Direct interactions of GPS2 with SHP, LRH-1, HNF4alpha, and FXR indicate alternative coregulator recruitment strategies to cause differential transcriptional outcomes. In addition, species-specific differences in the regulation of bile acid biosynthesis were uncovered by identifying human CYP8B1 as a direct FXR target gene, which has implications for therapeutic approaches in bile acid-related human disorders.
Collapse
Affiliation(s)
- Sabyasachi Sanyal
- *Department of Biosciences and Nutrition, Karolinska Institutet
- To whom correspondence may be addressed. E-mail: or
| | | | - Anna Haroniti
- Biomedical Sciences Research Center, Alexander Fleming, 16672 Vari, Athens, Greece
| | | | - Thomas Lundåsen
- Department of Endocrinology, Metabolism, and Diabetes, Karolinska University Hospital, S-14157 Huddinge, Sweden; and
| | | | | | | | - Iannis Talianidis
- Biomedical Sciences Research Center, Alexander Fleming, 16672 Vari, Athens, Greece
| | | | - Eckardt Treuter
- *Department of Biosciences and Nutrition, Karolinska Institutet
- To whom correspondence may be addressed. E-mail: or
| |
Collapse
|
16
|
Moore DD, Kato S, Xie W, Mangelsdorf DJ, Schmidt DR, Xiao R, Kliewer SA. International Union of Pharmacology. LXII. The NR1H and NR1I receptors: constitutive androstane receptor, pregnene X receptor, farnesoid X receptor alpha, farnesoid X receptor beta, liver X receptor alpha, liver X receptor beta, and vitamin D receptor. Pharmacol Rev 2006; 58:742-59. [PMID: 17132852 DOI: 10.1124/pr.58.4.6] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The nuclear receptors of the NR1H and NR1I subgroups include the constitutive androstane receptor, pregnane X receptor, farnesoid X receptors, liver X receptors, and vitamin D receptor. The newly emerging functions of these related receptors are under the control of metabolic pathways, including metabolism of xenobiotics, bile acids, cholesterol, and calcium. This review summarizes results of structural, pharmacologic, and genetic studies of these receptors.
Collapse
Affiliation(s)
- David D Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Stedman C, Liddle C, Coulter S, Sonoda J, Alvarez JG, Evans RM, Downes M. Benefit of farnesoid X receptor inhibition in obstructive cholestasis. Proc Natl Acad Sci U S A 2006; 103:11323-8. [PMID: 16844773 PMCID: PMC1544085 DOI: 10.1073/pnas.0604772103] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The nuclear hormone receptors farnesoid X receptor (FXR) and pregnane X receptor have been implicated in regulating bile acid, lipid, carbohydrate, and xenobiotic metabolism. Bile duct ligation was used to increase endogenous bile acids and evaluate the roles of these receptors in modulating cholestatic liver injury. FXR knockout (KO) mice were found to be protected from obstructive cholestasis. Concurrent deletion of FXR also could ameliorate an increase in liver injury that is seen usually in pregnane X receptor KO mice with cholestasis. Mechanisms proposed for this protection include the lowering of bile acid concentrations and altered expression of the hepatic transporters Mdr1, Mdr2, BSEP, and Mrp4. FXR KO mice also exhibit a biphasic lipid profile after bile duct ligation, with an increase in high-density lipoprotein cholesterol and triglycerides by day 6. The expression of apolipoprotein AV was reduced in these mice, implicating FXR in triglyceride regulation. We show that FXR modulates cholestasis by controlling bile acids within the hepatocyte and is involved in bile acid synthesis, bile excretion via BSEP, and serum export via Mrp4. This study strongly suggests a potential clinical role for FXR antagonists in the treatment of obstructive cholestatic liver disorders.
Collapse
Affiliation(s)
- Catherine Stedman
- *Department of Clinical Pharmacology, Molecular Pharmacology Laboratory, Westmead Millennium Institute and Institute of Clinical Pathology and Medical Research, Westmead Hospital, University of Sydney, Sydney NSW 2145, Australia; and
| | - Christopher Liddle
- *Department of Clinical Pharmacology, Molecular Pharmacology Laboratory, Westmead Millennium Institute and Institute of Clinical Pathology and Medical Research, Westmead Hospital, University of Sydney, Sydney NSW 2145, Australia; and
| | - Sally Coulter
- *Department of Clinical Pharmacology, Molecular Pharmacology Laboratory, Westmead Millennium Institute and Institute of Clinical Pathology and Medical Research, Westmead Hospital, University of Sydney, Sydney NSW 2145, Australia; and
| | - Junichiro Sonoda
- Howard Hughes Medical Institute, Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 Torrey Pines Road, La Jolla, CA 92037
| | - Jacqueline G. Alvarez
- Howard Hughes Medical Institute, Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 Torrey Pines Road, La Jolla, CA 92037
| | - Ronald M. Evans
- Howard Hughes Medical Institute, Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 Torrey Pines Road, La Jolla, CA 92037
- To whom correspondence may be addressed. E-mail:
or
| | - Michael Downes
- Howard Hughes Medical Institute, Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 Torrey Pines Road, La Jolla, CA 92037
- To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
18
|
Abstract
Liver X receptors (LXRs) and farnesoid X receptor (FXR) are nuclear receptors that function as intracellular sensors for sterols and bile acids, respectively. In response to their ligands, these receptors induce transcriptional responses that maintain a balanced, finely tuned regulation of cholesterol and bile acid metabolism. LXRs also permit the efficient storage of carbohydrate- and fat-derived energy, whereas FXR activation results in an overall decrease in triglyceride levels and modulation of glucose metabolism. The elegant, dual interplay between these two receptor systems suggests that they coevolved to constitute a highly sensitive and efficient system for the maintenance of total body fat and cholesterol homeostasis. Emerging evidence suggests that the tissue-specific action of these receptors is also crucial for the proper function of the cardiovascular, immune, reproductive, endocrine pancreas, renal, and central nervous systems. Together, LXRs and FXR represent potential therapeutic targets for the treatment and prevention of numerous metabolic and lipid-related diseases.
Collapse
Affiliation(s)
- Nada Y Kalaany
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.
| | | |
Collapse
|
19
|
Fiorucci S, Rizzo G, Antonelli E, Renga B, Mencarelli A, Riccardi L, Orlandi S, Pruzanski M, Morelli A, Pellicciari R. A farnesoid x receptor-small heterodimer partner regulatory cascade modulates tissue metalloproteinase inhibitor-1 and matrix metalloprotease expression in hepatic stellate cells and promotes resolution of liver fibrosis. J Pharmacol Exp Ther 2005; 314:584-95. [PMID: 15860571 DOI: 10.1124/jpet.105.084905] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The farnesoid X receptor (FXR) is expressed by and regulates hepatic stellate cells (HSCs). In the present study, we investigated whether 6-ethyl chenodeoxycholic acid (6-ECDCA or INT-747), a semisynthetic derivative of chenodeoxycholic acid (CDCA), modulates tissue metalloproteinase inhibitor (TIMP)-1 and matrix metalloprotease (MMP)-2 expression/activity in HSCs and in the liver of rats rendered cirrhotic by 4-week administration of CCl(4). Exposure of HSCs to FXR ligands increases small heterodimer partner (SHP) mRNA by 3-fold and reduces basal and thrombin-stimulated expression of alpha1(I)collagen, alpha-smooth muscle actin (alpha-SMA), TIMP-1, and TIMP-2 by approximately 60 to 70%, whereas it increased matrix metalloprotease (MMP)-2 activity by 2-fold. In coimmunoprecipitation, electromobility shift, and transactivation experiments, FXR activation/overexpression caused a SHP-dependent inhibition of JunD binding to its consensus element in the TIMP-1 promoter. Inhibition of TIMP-1 expression by SHP overexpression enhanced the sensitivity of HSCs to proapoptogenic stimuli. Administration of 3 mg/kg 6-ECDCA, but not 15 mg/kg ursodeoxycholic acid, resulted in early (3-5-day) induction of SHP and prevention of early up-regulation of TIMP-1 mRNA induced by CCl(4). In the prevention protocol, 4-week administration of 6-ECDCA reduced alpha1(I)collagen, alpha-SMA, and TIMP-1 mRNA by 60 to 80%, whereas it increased MMP-2 activity by 5-fold. In the resolution protocol, administration of 3 mg/kg 6-ECDCA promoted liver fibrosis resolution and increased the apoptosis of nonparenchyma liver cells. By demonstrating that a FXR-SHP regulatory cascade promotes the development of a quiescent phenotype and increases apoptosis of HSCs, this study establishes that FXR ligands may be beneficial in treatment of liver fibrosis.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dept. of Clinical and Experimental Medicine, University of Perugia, Policlinico Monteluce, Via E. Dal Pozzo, 06122 Perugia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Fiorucci S, Clerici C, Antonelli E, Orlandi S, Goodwin B, Sadeghpour BM, Sabatino G, Russo G, Castellani D, Willson TM, Pruzanski M, Pellicciari R, Morelli A. Protective effects of 6-ethyl chenodeoxycholic acid, a farnesoid X receptor ligand, in estrogen-induced cholestasis. J Pharmacol Exp Ther 2005; 313:604-12. [PMID: 15644430 DOI: 10.1124/jpet.104.079665] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The farnesoid X receptor (FXR), an endogenous sensor for bile acids, regulates a program of genes involved in bile acid biosynthesis, conjugation, and transport. Cholestatic liver diseases are a group of immunologically and genetically mediated disorders in which accumulation of endogenous bile acids plays a role in the disease progression and symptoms. Here, we describe the effect of 6-ethyl chenodeoxycholic acid (6-ECDCA or INT-747), a semisynthetic bile acid derivative and potent FXR ligand, in a model of cholestasis induced by 5-day administration of 17alpha-ethynylestradiol (E(2)17alpha) to rats. The exposure of rat hepatocytes to 1 microM 6-ECDCA caused a 3- to 5-fold induction of small heterodimer partner (Shp) and bile salt export pump (bsep) mRNA and 70 to 80% reduction of cholesterol 7alpha-hydroxylase (cyp7a1), oxysterol 12beta-hydroxylase (cyp8b1), and Na(+)/taurocholate cotransporting peptide (ntcp). In vivo administration of 6-ECDCA protects against cholestasis induced by E(2)17alpha. Thus, 6-ECDCA reverted bile flow impairment induced by E(2)17alpha, reduced secretion of cholic acid and deoxycholic acid, but increased muricholic acid and chenodeoxycholic acid secretion. In vivo administration of 6-ECDCA increased liver expression of Shp, bsep, multidrug resistance-associated protein-2, and multidrug resistance protein-2, whereas it reduced cyp7a1 and cyp8b1 and ntcp mRNA. These changes were reproduced by GW4064, a synthetic FXR ligand. In conclusion, by demonstrating that 6-ECDCA protects against E(2)17alpha cholestasis, our data support the notion that development of potent FXR ligands might represent a new approach for the treatment of cholestatic disorders.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Gastroenterologia ed Epatologia, Policlinico Monteluce, Perugia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Poupon R, Chignard N, Rosmorduc O, Barbu V, Housset C. La fonction biliaire et sa régulation. Med Sci (Paris) 2004; 20:1096-9. [PMID: 15581462 DOI: 10.1051/medsci/200420121096] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Biliary function is essential for intestinal absorption of fat, homeostasis of cholesterol and elimination of diverse metabolic end-products. Bile is elaborated in hepatocyte canaliculi and modified by cholangiocytes through both secretion and absorption processes. The main determinant of bile formation is an osmotic filtration process resulting from active transport of bile acids and other osmotic solutes. Most of the membrane transporters ensuring bile formation have now been identified. The expression of these membrane transporters is regulated in particular through transcriptional mechanisms under the control of nuclear receptors activated by ligands, such as bile acids, which act as endogenous steroids synthesized from cholesterol in hepatocytes. Monogenic cholestatic diseases illustrate the key role of membrane transporters in biliary function. Bile acids are potent modulators of transporters and thus trigger an adaptative response to cholestasis. The extent of this adaptative response could explain the compelling phenotypic variability of cholestatic diseases in childhood and adults. The firstline medical treatment is currently ursodeoxycholic acid. In case of failure of this medical treatment, liver transplantation is required. Recent progress in the molecular pathogenesis of bile formation and cholestatic liver diseases is expected to provide the design for drugs targeted to the molecular abnormalities responsible of cholestatic diseases.
Collapse
Affiliation(s)
- Raoul Poupon
- Inserm U.402 et Département d'Hépato-gastroentérologie, Faculté de Médecine Saint-Antoine, Université Pierre et Marie Curie, Paris 6, 27, rue Chaligny, 75571 Paris Cedex 12, France.
| | | | | | | | | |
Collapse
|