1
|
Jung SM, Zhu HJ. Regulation of Human Hydrolases and Its Implications in Pharmacokinetics and Pharmacodynamics. Drug Metab Dispos 2024; 52:1139-1151. [PMID: 38777597 PMCID: PMC11495669 DOI: 10.1124/dmd.123.001609] [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: 12/15/2023] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Hydrolases represent an essential class of enzymes indispensable for the metabolism of various clinically essential medications. Individuals exhibit marked differences in the expression and activation of hydrolases, resulting in significant variability in the pharmacokinetics (PK) and pharmacodynamics (PD) of drugs metabolized by these enzymes. The regulation of hydrolase expression and activity involves both genetic polymorphisms and nongenetic factors. This review examines the current understanding of genetic and nongenetic regulators of six clinically significant hydrolases, including carboxylesterase (CES)-1 CES2, arylacetamide deacetylase (AADAC), paraoxonase (PON)-1 PON3, and cathepsin A (CTSA). We explore genetic variants linked to the expression and activity of the hydrolases and their effects on the PK and PD of their substrate drugs. Regarding nongenetic regulators, we focus on the inhibitors and inducers of these enzymes. Additionally, we examine the developmental expression patterns and gender differences in the hydrolases when pertinent information was available. Many genetic and nongenetic regulators were found to be associated with the expression and activity of the hydrolases and PK and PD. However, hydrolases remain generally understudied compared with other drug-metabolizing enzymes, such as cytochrome P450s. The clinical significance of genetic and nongenetic regulators has not yet been firmly established for the majority of hydrolases. Comprehending the mechanisms that underpin the regulation of these enzymes holds the potential to refine therapeutic regimens, thereby enhancing the efficacy and safety of drugs metabolized by the hydrolases. SIGNIFICANCE STATEMENT: Hydrolases play a crucial role in the metabolism of numerous clinically important medications. Genetic polymorphisms and nongenetic regulators can affect hydrolases' expression and activity, consequently influencing the exposure and clinical outcomes of hydrolase substrate drugs. A comprehensive understanding of hydrolase regulation can refine therapeutic regimens, ultimately enhancing the efficacy and safety of drugs metabolized by the enzymes.
Collapse
Affiliation(s)
- Sun Min Jung
- Departments of Pharmaceutical Sciences (S.M.J.) and Clinical Pharmacy (H.-J.Z.), University of Michigan, Ann Arbor, Michigan
| | - Hao-Jie Zhu
- Departments of Pharmaceutical Sciences (S.M.J.) and Clinical Pharmacy (H.-J.Z.), University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
2
|
Dornas W, Silva M. Modulation of the antioxidant enzyme paraoxonase-1 for protection against cardiovascular diseases. Nutr Metab Cardiovasc Dis 2024:S0939-4753(24)00154-6. [PMID: 39277536 DOI: 10.1016/j.numecd.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/12/2024] [Accepted: 04/04/2024] [Indexed: 09/17/2024]
Abstract
AIM The enzyme paraoxonase 1 (PON1) bound to high-density lipoprotein has received special attention for its protective role against stress-mediated damage and use as a potential regulatory target in atherosclerosis and related vascular diseases. DATA SYNTHESIS We present an overview of the literature on PON1 activity and mRNA levels by investigating its modulation for clinical translations. Specifically, the expression of PON1 and its regulated activity can be modified in different ways with natural substances, drugs, and lifestyle factors thar affect the development of atherosclerosis. CONCLUSIONS The endothelial contribution of PON1 to overcome differences considering an individual's disease development risk is supported by polymorphism interaction data and the susceptibility to modify PON1 responses in chronic events composed by biological and environmental factors.
Collapse
Affiliation(s)
- Waleska Dornas
- Course Superior of Technology in Radiology, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Maisa Silva
- Department of Basic Life Sciences, Universidade Federal de Juiz de Fora, Governador Valadares, MG, Brazil
| |
Collapse
|
3
|
The Role of Fibroblast Growth Factor 19 in Hepatocellular Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1180-1192. [PMID: 34000282 DOI: 10.1016/j.ajpath.2021.04.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/09/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common type of cancer and the third leading cause of cancer-related deaths worldwide. Liver resection or liver transplantation is the most effective therapy for HCC because drugs approved by the US Food and Drug Administration to treat patients with unresectable HCC have an unfavorable overall survival rate. Therefore, the development of biomarkers for early diagnosis and effective therapy strategies are still necessary to improve patient outcomes. Fibroblast growth factor (FGF) 19 was amplified in patients with HCC from various studies, including patients from The Cancer Genome Atlas. FGF19 plays a syngeneic function with other signaling pathways in primary liver cancer development, such as epidermal growth factor receptor, Wnt/β-catenin, the endoplasmic reticulum-related signaling pathway, STAT3/IL-6, RAS, and extracellular signal-regulated protein kinase, among others. The current review presents a comprehensive description of the FGF19 signaling pathway involved in liver cancer development. The use of big data and bioinformatic analysis can provide useful clues for further studies of the FGF19 pathway in HCC, including its application as a biomarker, targeted therapy, and combination therapy strategies.
Collapse
|
4
|
Perino A, Demagny H, Velazquez-Villegas L, Schoonjans K. Molecular Physiology of Bile Acid Signaling in Health, Disease, and Aging. Physiol Rev 2020; 101:683-731. [PMID: 32790577 DOI: 10.1152/physrev.00049.2019] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Over the past two decades, bile acids (BAs) have become established as important signaling molecules that enable fine-tuned inter-tissue communication from the liver, their site of production, over the intestine, where they are modified by the gut microbiota, to virtually any organ, where they exert their pleiotropic physiological effects. The chemical variety of BAs, to a large extent determined by the gut microbiome, also allows for a complex fine-tuning of adaptive responses in our body. This review provides an overview of the mechanisms by which BA receptors coordinate several aspects of physiology and highlights new therapeutic strategies for diseases underlying pathological BA signaling.
Collapse
Affiliation(s)
- Alessia Perino
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| | - Hadrien Demagny
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| | - Laura Velazquez-Villegas
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| | - Kristina Schoonjans
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne (EPFL), Switzerland
| |
Collapse
|
5
|
Hu J, Liu Z, Tong Y, Mei Z, Xu A, Zhou P, Chen X, Tang W, Zhou Z, Xiao Y. Fibroblast Growth Factor 19 Levels Predict Subclinical Atherosclerosis in Men With Type 2 Diabetes. Front Endocrinol (Lausanne) 2020; 11:282. [PMID: 32528406 PMCID: PMC7258879 DOI: 10.3389/fendo.2020.00282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 04/15/2020] [Indexed: 12/11/2022] Open
Abstract
Objective: Fibroblast growth factor 19 (FGF19) plays an indispensable role in regulating bile acid, glucose, and lipid metabolism, and alterations of its circulating concentration is associated with the development of type 2 diabetes (T2D). Atherosclerosis is directly related to the death-deriving diabetic macroangiopathy in T2D, yet relationships between FGF19 and atherosclerosis in T2D remain unclear. The aim of this study was to investigate the association of circulating FGF19 levels with the development of subclinical atherosclerosis (subAS) in patients with T2D in a 3-year prospective study. Methods: In the present study, 153 newly diagnosed T2D patients without subAS were recruited at baseline, and 137 of them completed a 3-year follow-up. FGF19 levels were measured in fasting serum samples collected at baseline and the third-year visits. Carotid, femoral, and iliac intima-media thickness (IMT) were detected by high-resolution B-mode ultrasound to determine the presence of subAS. Logistic regression analysis was applied to assess the relationship between serum FGF19 and subAS in patients with T2D. Results: At baseline, serum FGF19 levels were positively correlated with carotid IMT and iliac IMT in men (r = 0.239, P = 0.036; r = 0.309, P = 0.006). At the 3-year follow-up, 25 out of 153 patients developed subAS, and FGF19 levels in men were higher in the subAS group than in the non-subAS group [202.7 (177.9-373.6) vs. 133.4 (85.6-171.3) pg/ml, P = 0.028]. Furthermore, in men, higher baseline levels of FGF19 were independently associated with a greater risk of subAS at year 3 in patients with T2D with an odds ratio (OR) of 4.798 per 1 standard deviation (SD) of the FGF19 concentration [OR = 4.798 (95% CI, 1.680-13.706), P = 0.003]. Baseline FGF19 levels yielded an area under the receiver operating characteristic curve of 0.769 to predict the development of subAS at year 3 in men with T2D. Conclusions: Serum FGF19 levels could help in predicting the development of atherosclerosis in men with T2D.
Collapse
Affiliation(s)
- Jingyi Hu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhiwen Liu
- Department of Endocrinology, Xuhui District Central Hospital, Shanghai, China
| | - Yue Tong
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zubing Mei
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Anorectal Disease Institute of Shuguang Hospital, Shanghai, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
- Research Center of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China
| | - Pengcheng Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
- Research Center of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China
| | - Xiaoyan Chen
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Weili Tang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhiguang Zhou
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Yang Xiao
| |
Collapse
|
6
|
Somm E, Jornayvaz FR. Fibroblast Growth Factor 15/19: From Basic Functions to Therapeutic Perspectives. Endocr Rev 2018; 39:960-989. [PMID: 30124818 DOI: 10.1210/er.2018-00134] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Discovered 20 years ago, fibroblast growth factor (FGF)19, and its mouse ortholog FGF15, were the first members of a new subfamily of FGFs able to act as hormones. During fetal life, FGF15/19 is involved in organogenesis, affecting the development of the ear, eye, heart, and brain. At adulthood, FGF15/19 is mainly produced by the ileum, acting on the liver to repress hepatic bile acid synthesis and promote postprandial nutrient partitioning. In rodents, pharmacologic doses of FGF19 induce the same antiobesity and antidiabetic actions as FGF21, with these metabolic effects being partly mediated by the brain. However, activation of hepatocyte proliferation by FGF19 has long been a challenge to its therapeutic use. Recently, genetic reengineering of the molecule has resolved this issue. Despite a global overlap in expression pattern and function, murine FGF15 and human FGF19 exhibit several differences in terms of regulation, molecular structure, signaling, and biological properties. As most of the knowledge originates from the use of FGF19 in murine models, differences between mice and humans in the biology of FGF15/19 have to be considered for a successful translation from bench to bedside. This review summarizes the basic knowledge concerning FGF15/19 in mice and humans, with a special focus on regulation of production, morphogenic properties, hepatocyte growth, bile acid homeostasis, as well as actions on glucose, lipid, and energy homeostasis. Moreover, implications and therapeutic perspectives concerning FGF19 in human diseases (including obesity, type 2 diabetes, hepatic steatosis, biliary disorders, and cancer) are also discussed.
Collapse
Affiliation(s)
- Emmanuel Somm
- Service of Endocrinology, Diabetes, Hypertension, and Nutrition, Geneva University Hospitals, University of Geneva Medical School, Geneva, Switzerland
| | - François R Jornayvaz
- Service of Endocrinology, Diabetes, Hypertension, and Nutrition, Geneva University Hospitals, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
7
|
Jiao N, Baker SS, Chapa-Rodriguez A, Liu W, Nugent CA, Tsompana M, Mastrandrea L, Buck MJ, Baker RD, Genco RJ, Zhu R, Zhu L. Suppressed hepatic bile acid signalling despite elevated production of primary and secondary bile acids in NAFLD. Gut 2018; 67:1881-1891. [PMID: 28774887 DOI: 10.1136/gutjnl-2017-314307] [Citation(s) in RCA: 440] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Bile acids are regulators of lipid and glucose metabolism, and modulate inflammation in the liver and other tissues. Primary bile acids such as cholic acid and chenodeoxycholic acid (CDCA) are produced in the liver, and converted into secondary bile acids such as deoxycholic acid (DCA) and lithocholic acid by gut microbiota. Here we investigated the possible roles of bile acids in non-alcoholic fatty liver disease (NAFLD) pathogenesis and the impact of the gut microbiome on bile acid signalling in NAFLD. DESIGN Serum bile acid levels and fibroblast growth factor 19 (FGF19), liver gene expression profiles and gut microbiome compositions were determined in patients with NAFLD, high-fat diet-fed rats and their controls. RESULTS Serum concentrations of primary and secondary bile acids were increased in patients with NAFLD. In per cent, the farnesoid X receptor (FXR) antagonistic DCA was increased, while the agonistic CDCA was decreased in NAFLD. Increased mRNA expression for cytochrome P450 7A1, Na+-taurocholate cotransporting polypeptide and paraoxonase 1, no change in mRNA expression for small heterodimer partner and bile salt export pump, and reduced serum FGF19 were evidence of impaired FXR and fibroblast growth factor receptor 4 (FGFR4)-mediated signalling in NAFLD. Taurine and glycine metabolising bacteria were increased in the gut of patients with NAFLD, reflecting increased secondary bile acid production. Similar changes in liver gene expression and the gut microbiome were observed in high-fat diet-fed rats. CONCLUSIONS The serum bile acid profile, the hepatic gene expression pattern and the gut microbiome composition consistently support an elevated bile acid production in NAFLD. The increased proportion of FXR antagonistic bile acid explains, at least in part, the suppression of hepatic FXR-mediated and FGFR4-mediated signalling. Our study suggests that future NAFLD intervention may target the components of FXR signalling, including the bile acid converting gut microbiome.
Collapse
Affiliation(s)
- Na Jiao
- Department of Bioinformatics, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Susan S Baker
- Department of Pediatrics, Digestive Diseases and Nutrition Center, The State University of New York at Buffalo, Buffalo, New York, USA
- Genome, Environment and Microbiome Community of Excellence, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Adrian Chapa-Rodriguez
- Department of Pediatrics, Digestive Diseases and Nutrition Center, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Wensheng Liu
- Department of Pediatrics, Digestive Diseases and Nutrition Center, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Colleen A Nugent
- Department of Pediatrics, Digestive Diseases and Nutrition Center, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Maria Tsompana
- Department of Biochemistry and Center of Excellence in Bioinformatics and Life Sciences, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Lucy Mastrandrea
- Division of Endocrinology, Department of Pediatrics, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Michael J Buck
- Genome, Environment and Microbiome Community of Excellence, The State University of New York at Buffalo, Buffalo, New York, USA
- Department of Biochemistry and Center of Excellence in Bioinformatics and Life Sciences, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Robert D Baker
- Department of Pediatrics, Digestive Diseases and Nutrition Center, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Robert J Genco
- Genome, Environment and Microbiome Community of Excellence, The State University of New York at Buffalo, Buffalo, New York, USA
- Departments of Oral Biology, Microbiology and Immunology, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Ruixin Zhu
- Department of Bioinformatics, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lixin Zhu
- Department of Pediatrics, Digestive Diseases and Nutrition Center, The State University of New York at Buffalo, Buffalo, New York, USA
- Genome, Environment and Microbiome Community of Excellence, The State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
8
|
Medina-Díaz IM, Ponce-Ruiz N, Ramírez-Chávez B, Rojas-García AE, Barrón-Vivanco BS, Elizondo G, Bernal-Hernández YY. Downregulation of human paraoxonase 1 (PON1) by organophosphate pesticides in HepG2 cells. ENVIRONMENTAL TOXICOLOGY 2017; 32:490-500. [PMID: 26948828 DOI: 10.1002/tox.22253] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/09/2016] [Accepted: 02/14/2016] [Indexed: 06/05/2023]
Abstract
Paraoxonase 1 (PON1) is a calcium-dependent esterase synthesized primarily in the liver and secreted into the plasma where it is associated with high-density lipoproteins (HDL). PON1 hydrolyzes and detoxifies some toxic metabolites of organophosphorus compounds (OPs) such as methyl parathion and chlorpyrifos. Thus, PON1 activity and expression levels are important for determining susceptibility against OPs poisoning. Some studies have demonstrated that OPs can modulate gene expression through interactions with nuclear receptors. In this study, we evaluated the effects of methyl parathion and chlorpyrifos on the modulation of PON1 in Human Hepatocellular Carcinoma (HepG2) cells by real-time PCR, PON1 activity assay, and western blot. The results showed that the treatments with methyl parathion and chlorpyrifos decreased PON1 mRNA and immunoreactive protein and increased inflammatory cytokines in HepG2 cells. The effects of methyl parathion and chlorpyrifos on the downregulation of PON1 gene expression in HepG2 cells may provide evidence of OPs cytotoxicity related to oxidative stress and an inflammatory response. A decrease in the expression of the PON1 gene may increase the susceptibility to OPs intoxication and the risk of diseases related to inflammation and oxidative stress. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 490-500, 2017.
Collapse
Affiliation(s)
- Irma Martha Medina-Díaz
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado
| | - Néstor Ponce-Ruiz
- Posgrado en Ciencias Biológico Agropecuarias, Universidad Autónoma de Nayarit
| | | | - Aurora Elizabeth Rojas-García
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado
| | - Briscia S Barrón-Vivanco
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado
| | - Guillermo Elizondo
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, México, D.F., México
| | - Yael Y Bernal-Hernández
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado
| |
Collapse
|
9
|
Role of farnesoid X receptor in establishment of ontogeny of phase-I drug metabolizing enzyme genes in mouse liver. Acta Pharm Sin B 2016; 6:453-459. [PMID: 27709014 PMCID: PMC5045554 DOI: 10.1016/j.apsb.2016.07.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/08/2016] [Accepted: 06/21/2016] [Indexed: 12/12/2022] Open
Abstract
The expression of phase-I drug metabolizing enzymes in liver changes dramatically during postnatal liver maturation. Farnesoid X receptor (FXR) is critical for bile acid and lipid homeostasis in liver. However, the role of FXR in regulating ontogeny of phase-I drug metabolizing genes is not clear. Hence, we applied RNA-sequencing to quantify the developmental expression of phase-I genes in both Fxr-null and control (C57BL/6) mouse livers during development. Liver samples of male C57BL/6 and Fxr-null mice at 6 different ages from prenatal to adult were used. The Fxr-null showed an overall effect to diminish the “day-1 surge” of phase-I gene expression, including cytochrome P450s at neonatal ages. Among the 185 phase-I genes from 12 different families, 136 were expressed, and differential expression during development occurred in genes from all 12 phase-I families, including hydrolysis: carboxylesterase (Ces), paraoxonase (Pon), and epoxide hydrolase (Ephx); reduction: aldoketo reductase (Akr), quinone oxidoreductase (Nqo), and dihydropyrimidine dehydrogenase (Dpyd); and oxidation: alcohol dehydrogenase (Adh), aldehyde dehydrogenase (Aldh), flavin monooxygenases (Fmo), molybdenum hydroxylase (Aox and Xdh), cytochrome P450 (P450), and cytochrome P450 oxidoreductase (Por). The data also suggested new phase-I genes potentially targeted by FXR. These results revealed an important role of FXR in regulation of ontogeny of phase-I genes.
Collapse
Key Words
- ADH, alcohol dehydrogenase
- AKR, aldoketo reductase
- ALDH, aldehyde dehydrogenase
- CES, carboxylesterase (Ces)
- DPYD, dihydropyrimidine dehydrogenase
- Drug metabolizing enzymes
- EPHX, epoxide hydrolase
- FMO, flavin monooxygenases, Farnesoid X receptor (FXR)
- Farnesoid X receptor
- Fxr-null mouse
- Gene expression
- Liver
- NQO, quinone oxidoreductase
- Ontogeny
- P450, cytochrome P450
- PON, paraoxonase
- POR, cytochrome P450 oxidoreductase
Collapse
|
10
|
Zhang X, Osaka T, Tsuneda S. Bacterial metabolites directly modulate farnesoid X receptor activity. Nutr Metab (Lond) 2015; 12:48. [PMID: 26604978 PMCID: PMC4657204 DOI: 10.1186/s12986-015-0045-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/18/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The farnesoid X receptor (FXR), a ligand-activated transcription factor belonging to the adopted orphan receptor, plays an important role in maintaining health of the liver and intestine. In this study, we identified individual bacterial strains that directly modulated the activation of intestinal FXR. METHODS The FXR stimulatory potential of 38 bacterial strains was determined using a stable FXR reporter system derived from intestinal epithelial cells (IEC). The induction of FXR target genes by screened FXR stimulatory bacteria was determined by real-time PCR. In addition, a high fat diet (HFD)-induced obese mouse model was used to evaluate in vivo FXR stimulatory potential of bacterial metabolites screened in this study. RESULTS A luciferase assay with the FXR reporter cell line demonstrated that the FXR-stimulatory activity of most bacterial cell samples was less than 2-fold. The culture supernatants of Bacteroides dorei and Eubacterium limosum induced FXR activity and selectively regulated FXR target expression in the FXR reporter system. Treatment with B. dorei-derived metabolites strongly induced ileal bile acid binding protein (IBABP) (8.4-fold) and organic solute transporter (OST) α (3.1-fold) compared with E. limosum-derived metabolites. Furthermore, administration of B. dorei derived metabolites showed significant reduction in body weight gain, and both two bacterial metabolites reduced liver weight in obese mice compared to PBS-treated controls. Administration of each bacterial metabolites improved in serum levels of obesity-related metabolic biochemical markers such as ALT, AST, total cholesterol, and triglyceride. Furthermore, two bacterial metabolites enhanced the Fxr gene expression in the intestine and liver, and ileal Shp gene expression tended to be increased by treatment with the metabolites derived from B. dorei. CONCLUSIONS B. dorei and E. limosum secreted the bioactive substances that directly stimulate FXR in the intestinal epithelial cells. Administration of these bacterial FXR-stimulatory metabolites improves the obesity phenotype including body weight gain, liver damage, lipid metabolism in DIO mice.
Collapse
Affiliation(s)
- Xianqin Zhang
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480 Japan
| | - Toshifumi Osaka
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480 Japan.,Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, 162-8666 Japan
| | - Satoshi Tsuneda
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, 162-8480 Japan
| |
Collapse
|
11
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by the aberrant accumulation of triglycerides in hepatocytes in the absence of significant alcohol consumption, viral infection or other specific causes of liver disease. NAFLD has become a global health problem, but its pathogenesis remains poorly understood and no efficient pharmaceutical treatments have yet been established. The farnesoid X receptor (FXR) is a member of nuclear receptors of intracellular ligand-activated transcription factors and plays an important role in metabolism of bile acids, lipid and glucose. In addition, it has been recently reported that FXR participates in regulating insulin resistance and lipid metabolic disorder, inhibiting the activation of hepatic stellate cells and penetration of inflammatory cells, and promoting the enterohepatic circulation and regeneration of liver cells to defer liver fibrosis, which is significant for NAFLD. Several FXR agonists have been identified and proved to be optimistic in preventing and treating NAFLD both experimentally and clinically, indicating that FXR may be a therapeutic target for NAFLD. The use of FXR in NAFLD remains controversial currently.
Collapse
|
12
|
Ding L, Yang L, Wang Z, Huang W. Bile acid nuclear receptor FXR and digestive system diseases. Acta Pharm Sin B 2015; 5:135-44. [PMID: 26579439 PMCID: PMC4629217 DOI: 10.1016/j.apsb.2015.01.004] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 12/31/2014] [Accepted: 01/05/2015] [Indexed: 12/14/2022] Open
Abstract
Bile acids (BAs) are not only digestive surfactants but also important cell signaling molecules, which stimulate several signaling pathways to regulate some important biological processes. The bile-acid-activated nuclear receptor, farnesoid X receptor (FXR), plays a pivotal role in regulating bile acid, lipid and glucose homeostasis as well as in regulating the inflammatory responses, barrier function and prevention of bacterial translocation in the intestinal tract. As expected, FXR is involved in the pathophysiology of a wide range of diseases of gastrointestinal tract, including inflammatory bowel disease, colorectal cancer and type 2 diabetes. In this review, we discuss current knowledge of the roles of FXR in physiology of the digestive system and the related diseases. Better understanding of the roles of FXR in digestive system will accelerate the development of FXR ligands/modulators for the treatment of digestive system diseases.
Collapse
Key Words
- 6-ECDCA, 6α-ethyl-chenodeoxycholic acid
- AF2, activation domain
- ANGTPL3, angiopoietin-like protein 3
- AOM, azoxymethane
- AP-1, activator protein-1
- ASBT, apical sodium-dependent bile salt transporter
- Apo, apolipoprotein
- BAAT, bile acid-CoA amino acid N-acetyltransferase
- BACS, bile acid-CoA synthetase
- BAs, bile acids
- BMI, body mass index
- BSEP, bile salt export pump
- Bile acids
- CA, cholic acid
- CD, Crohn׳s disease
- CDCA, chenodeoxycholic acid
- CREB, cAMP regulatory element-binding protein
- CYP7A1, cholesterol 7α-hydroxylase
- Colorectal cancer
- DBD, DNA binding domain
- DCA, deoxycholic acid
- DSS, dextrane sodium sulfate
- ERK, extracellular signal-regulated kinase
- FABP6, fatty acid-binding protein subclass 6
- FFAs, free fatty acids
- FGF19, fibroblast growth factor 19
- FGFR4, fibroblast growth factor receptor 4
- FXR, farnesoid X receptor
- FXRE, farnesoid X receptor response element
- Farnesoid X receptor
- G6Pase, glucose-6-phosphatase
- GLP-1, glucagon-like peptide 1
- GLUT2, glucose transporter type 2
- GPBAR, G protein-coupled BA receptor
- GPCRs, G protein-coupled receptors
- GSK3, glycogen synthase kinase 3
- Gastrointestinal tract
- HDL-C, high density lipoprotein cholesterol
- HNF4α, hepatic nuclear factor 4α
- I-BABP, intestinal bile acid-binding protein
- IBD, inflammatory bowel disease
- IL-1, interleukin 1
- Inflammatory bowel disease
- KLF11, Krüppel-like factor 11
- KRAS, Kirsten rat sarcoma viral oncogene homolog
- LBD, ligand binding domain
- LCA, lithocholic acid
- LPL, lipoprotein lipase
- LRH-1, liver receptor homolog-1
- MCA, muricholicacid
- MRP2, multidrug resistance-associated protein 2
- NF-κB, nuclear factor-kappa B
- NOD, non-obese diabetic
- NRs, nuclear receptors
- OSTα, organic solute transporter alpha
- OSTβ, organic solute transporter beta
- PEPCK, phosphoenol pyruvate carboxykinase
- PGC-1α, peroxisome proliferators-activated receptor γ coactivator protein-1α
- SHP, small heterodimer partner
- SREBP-1c, sterol regulatory element-binding protein 1c
- STAT3, signal transducers and activators of transcription 3
- T2D, type 2 diabetes
- TLCA, taurolithocholic acid
- TNBS, trinitrobenzensulfonic acid
- TNFα, tumor necrosis factors α
- Type 2 diabetes
- UC, ulcerative colitis
- UDCA, ursodeoxycholic acid
- VSG, vertical sleeve gastrectomy
- db/db, diabetic mice
Collapse
|
13
|
Dail MB, Pickin R, Crow JA, Chambers JE. The Effect ofIn VitroDieldrin Exposure on the Rat Paraoxonase 1 (Pon1) Promoter. J Biochem Mol Toxicol 2014; 28:224-31. [DOI: 10.1002/jbt.21557] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/14/2014] [Accepted: 01/24/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Mary Beth Dail
- Center for Environmental Health Sciences and Department of Basic Sciences; College of Veterinary Medicine; Mississippi State University; Mississippi State MS USA
| | - Rebecca Pickin
- Center for Environmental Health Sciences and Department of Basic Sciences; College of Veterinary Medicine; Mississippi State University; Mississippi State MS USA
| | - John Allen Crow
- Center for Environmental Health Sciences and Department of Basic Sciences; College of Veterinary Medicine; Mississippi State University; Mississippi State MS USA
| | - Janice E. Chambers
- Center for Environmental Health Sciences and Department of Basic Sciences; College of Veterinary Medicine; Mississippi State University; Mississippi State MS USA
| |
Collapse
|
14
|
Litvinov D, Mahini H, Garelnabi M. Antioxidant and anti-inflammatory role of paraoxonase 1: implication in arteriosclerosis diseases. NORTH AMERICAN JOURNAL OF MEDICAL SCIENCES 2012. [PMID: 23181222 PMCID: PMC3503369 DOI: 10.4103/1947-2714.103310] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Paraoxonase 1 (PON1) is a hydrolytic enzyme with wide range of substrates, and capability to protect against lipid oxidation. Despite of the large number of compounds that can be hydrolyzed by paraoxonase, the biologically relevant substrates are still not clearly determined. There is a massive in vitro and in vivo data to demonstrate the beneficial effects of PON1 in several atherosclerosis-related processes. The enzyme is primarily expressed in liver; however, it is also localized in other tissues. PON1 attracted significant interest as a protein that is responsible for the most of antioxidant properties of high-density lipoprotein (HDL). Several bioactive molecules such as dietary polyphenols, aspirin and its hydrolysis product salicylate, are known to stimulate PON1 transcription activation in mouse liver and HepG2 cell line. Studies on the activity, function, and genetic makeup have revealed a protective role of PON1. Some striking data were obtained in PON1 gene knockout and PON1 transgenic mouse models and in human studies. The goal of this review is to assess the current understanding of PON1 expression, enzymatic and antioxidant activity, and its atheroprotective effects. Results from in vivo and in vitro basic studies; and from human studies on the association of PON1 with coronary artery disease (CAD) and ischemic stroke will be discussed.
Collapse
Affiliation(s)
- Dmitry Litvinov
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | | | | |
Collapse
|
15
|
Abstract
Pomegranate is an ancient fruit that is still part of the diet in the Mediterranean area, the Middle East, and India. Health-promoting effects have long been attributed to this fruit. Modern research corroborates the use of pomegranate as a folk remedy for diabetes and metabolic syndrome, and is responsible for a new evaluation of nutritional and pharmaceutical aspects of pomegranate in the general public. In the last decade, industry and agricultural production have been adapted to meet higher market demands for pomegranate. In vivo and in vitro studies have demonstrated that pomegranate exerts hypoglycaemic effects, including increased insulin sensitivity, inhibition of α-glucosidase, and impact on glucose transporter type 4 function, but is also responsible for a reduction of total cholesterol, and the improvement of blood lipid profiles, as well as anti-inflammatory effects through the modulation of peroxisome proliferator-activated receptor pathways. These effects may also explain how pomegranate-derived compounds function in the amelioration of adverse health effects caused by metabolic syndrome. Pomegranate contains polyphenols such as ellagitannins and anthocyanins, as well as phenolic acids, fatty acids and a variety of volatile compounds. Ellagitannins are some of the most prevalent compounds present in pomegranate, and may be responsible for certain benevolent characteristics associated with pomegranate. A brief overview of rising health problems due to obesity will be provided, followed by characterisation of the biological activity, bioavailability, and safety of pomegranate and pomegranate-derived compounds. Although the fruit is consumed in many countries, epidemiological and clinical studies are unavailable. Additional research is necessary to corroborate the promise of current in vivo and in vitro findings.
Collapse
Affiliation(s)
- Svjetlana Medjakovic
- Department of Biotechnology, Christian-Doppler-Laboratory of Receptor Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria
| | | |
Collapse
|
16
|
She ZG, Chen HZ, Yan Y, Li H, Liu DP. The human paraoxonase gene cluster as a target in the treatment of atherosclerosis. Antioxid Redox Signal 2012; 16:597-632. [PMID: 21867409 PMCID: PMC3270057 DOI: 10.1089/ars.2010.3774] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The paraoxonase (PON) gene cluster contains three adjacent gene members, PON1, PON2, and PON3. Originating from the same fungus lactonase precursor, all of the three PON genes share high sequence identity and a similar β propeller protein structure. PON1 and PON3 are primarily expressed in the liver and secreted into the serum upon expression, whereas PON2 is ubiquitously expressed and remains inside the cell. Each PON member has high catalytic activity toward corresponding artificial organophosphate, and all exhibit activities to lactones. Therefore, all three members of the family are regarded as lactonases. Under physiological conditions, they act to degrade metabolites of polyunsaturated fatty acids and homocysteine (Hcy) thiolactone, among other compounds. By detoxifying both oxidized low-density lipoprotein and Hcy thiolactone, PONs protect against atherosclerosis and coronary artery diseases, as has been illustrated by many types of in vitro and in vivo experimental evidence. Clinical observations focusing on gene polymorphisms also indicate that PON1, PON2, and PON3 are protective against coronary artery disease. Many other conditions, such as diabetes, metabolic syndrome, and aging, have been shown to relate to PONs. The abundance and/or activity of PONs can be regulated by lipoproteins and their metabolites, biological macromolecules, pharmacological treatments, dietary factors, and lifestyle. In conclusion, both previous results and ongoing studies provide evidence, making the PON cluster a prospective target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Zhi-Gang She
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | | | | | | | | |
Collapse
|
17
|
Non-alcoholic Fatty liver disease: the bile Acid-activated farnesoid x receptor as an emerging treatment target. J Lipids 2011; 2012:934396. [PMID: 22187656 PMCID: PMC3236512 DOI: 10.1155/2012/934396] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 09/18/2011] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently evolving as the most common liver disease worldwide. It may progress to liver cirrhosis and liver cancer and is poised to represent the most common indication for liver transplantation in the near future. The pathogenesis of NAFLD is multifactorial and not fully understood, but it represents an insulin resistance state characterized by a cluster of cardiovascular risk factors including obesity, dyslipidemia, hyperglycemia, and hypertension. Importantly, NAFLD also has evolved as independent risk factor for cardiovascular disease. Unfortunately thus far no established treatment does exist for NAFLD. The bile acid-activated nuclear farnesoid X receptor (FXR) has been shown to play a role not only in bile acid but also in lipid and glucose homeostasis. Specific targeting of FXR may be an elegant and very effective way to readjust dysregulated nuclear receptor-mediated metabolic pathways. This review discusses the body's complex response to the activation of FXR with its beneficial actions but also potential undesirable side effects.
Collapse
|
18
|
Wang H, Venkatesh M, Li H, Goetz R, Mukherjee S, Biswas A, Zhu L, Kaubisch A, Wang L, Pullman J, Whitney K, Kuro-o M, Roig AI, Shay JW, Mohammadi M, Mani S. Pregnane X receptor activation induces FGF19-dependent tumor aggressiveness in humans and mice. J Clin Invest 2011; 121:3220-32. [PMID: 21747170 DOI: 10.1172/jci41514] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 05/18/2011] [Indexed: 01/10/2023] Open
Abstract
The nuclear receptor pregnane X receptor (PXR) is activated by a range of xenochemicals, including chemotherapeutic drugs, and has been suggested to play a role in the development of tumor cell resistance to anticancer drugs. PXR also has been implicated as a regulator of the growth and apoptosis of colon tumors. Here, we have used a xenograft model of colon cancer to define a molecular mechanism that might underlie PXR-driven colon tumor growth and malignancy. Activation of PXR was found to be sufficient to enhance the neoplastic characteristics, including cell growth, invasion, and metastasis, of both human colon tumor cell lines and primary human colon cancer tissue xenografted into immunodeficient mice. Furthermore, we were able to show that this PXR-mediated phenotype required FGF19 signaling. PXR bound to the FGF19 promoter in both human colon tumor cells and "normal" intestinal crypt cells. However, while both cell types proliferated in response to PXR ligands, the FGF19 promoter was activated by PXR only in cancer cells. Taken together, these data indicate that colon cancer growth in the presence of a specific PXR ligand results from tumor-specific induction of FGF19. These observations may lead to improved therapeutic regimens for colon carcinomas.
Collapse
Affiliation(s)
- Hongwei Wang
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, New York 10461, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Wu AL, Coulter S, Liddle C, Wong A, Eastham-Anderson J, French DM, Peterson AS, Sonoda J. FGF19 regulates cell proliferation, glucose and bile acid metabolism via FGFR4-dependent and independent pathways. PLoS One 2011; 6:e17868. [PMID: 21437243 PMCID: PMC3060878 DOI: 10.1371/journal.pone.0017868] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 02/11/2011] [Indexed: 12/22/2022] Open
Abstract
Fibroblast growth factor 19 (FGF19) is a hormone-like protein that regulates carbohydrate, lipid and bile acid metabolism. At supra-physiological doses, FGF19 also increases hepatocyte proliferation and induces hepatocellular carcinogenesis in mice. Much of FGF19 activity is attributed to the activation of the liver enriched FGF Receptor 4 (FGFR4), although FGF19 can activate other FGFRs in vitro in the presence of the coreceptor βKlotho (KLB). In this report, we investigate the role of FGFR4 in mediating FGF19 activity by using Fgfr4 deficient mice as well as a variant of FGF19 protein (FGF19v) which is specifically impaired in activating FGFR4. Our results demonstrate that FGFR4 activation mediates the induction of hepatocyte proliferation and the suppression of bile acid biosynthesis by FGF19, but is not essential for FGF19 to improve glucose and lipid metabolism in high fat diet fed mice as well as in leptin-deficient ob/ob mice. Thus, FGF19 acts through multiple receptor pathways to elicit pleiotropic effects in regulating nutrient metabolism and cell proliferation.
Collapse
Affiliation(s)
- Ai-Luen Wu
- Department of Molecular Biology, Genentech, Inc., South San Francisco, California, United States of America
| | - Sally Coulter
- Storr Liver Unit, Westmead Millennium Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Christopher Liddle
- Storr Liver Unit, Westmead Millennium Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Anne Wong
- Department of Assay and Automation Technology, Genentech, Inc., South San Francisco, California, United States of America
| | - Jeffrey Eastham-Anderson
- Department of Pathology, Genentech, Inc., South San Francisco, California, United States of America
| | - Dorothy M. French
- Department of Pathology, Genentech, Inc., South San Francisco, California, United States of America
| | - Andrew S. Peterson
- Department of Molecular Biology, Genentech, Inc., South San Francisco, California, United States of America
| | - Junichiro Sonoda
- Department of Molecular Biology, Genentech, Inc., South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
20
|
Vacca M, Degirolamo C, Mariani-Costantini R, Palasciano G, Moschetta A. Lipid-sensing nuclear receptors in the pathophysiology and treatment of the metabolic syndrome. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2011; 3:562-87. [PMID: 21755605 DOI: 10.1002/wsbm.137] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metabolic syndrome (MS) is a cluster of different diseases, namely central obesity, hypertension, hyperglycemia, and dyslipidemia, together with a pro-thrombotic and pro-inflammatory state. These metabolic abnormalities are often associated with an increased risk for cardiovascular disease (CVD) and cancer. Dietary and lifestyle modifications are currently believed more effective than pharmacological therapies in the management of MS patients. Nevertheless, the relatively low grade of compliance of patients to these recommendations, as well as the failure of current therapies, highlights the need for the discovery of new pharmacological and nutraceutic approaches. A deeper knowledge of the patho-physiological events that initiate and support the MS is mandatory. Lipid-sensing nuclear receptors (NRs) are the master transcriptional regulators of lipid and carbohydrate metabolism and inflammatory responses, thus standing as suitable targets. This review focuses on the physiological relevance of the NRs (peroxisome proliferator-activated receptors, liver X receptors, and farnesoid X receptor) in the control of whole-body homeostasis, with a special emphasis on lipid and glucose metabolism, and on the relationships between metabolic unbalances, systemic inflammation, and the onset of CVD. Future perspectives and possible clinical applications are also presented.
Collapse
Affiliation(s)
- Michele Vacca
- Clinica Medica Augusto Murri, Aldo Moro University of Bari, and Department of Translational Pharmacology, Consorzio Mario Negri Sud, Santa Maria Imbaro (CH), Italy
| | | | | | | | | |
Collapse
|
21
|
Modica S, Gadaleta RM, Moschetta A. Deciphering the nuclear bile acid receptor FXR paradigm. NUCLEAR RECEPTOR SIGNALING 2010; 8:e005. [PMID: 21383957 PMCID: PMC3049226 DOI: 10.1621/nrs.08005] [Citation(s) in RCA: 207] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 10/14/2010] [Indexed: 12/12/2022]
Abstract
Originally called retinoid X receptor interacting protein 14 (RIP14), the farnesoid X receptor (FXR) was renamed after the ability of its rat form to bind supra-physiological concentrations of farnesol. In 1999 FXR was de-orphanized since primary bile acids were identified as natural ligands. Strongly expressed in the liver and intestine, FXR has been shown to be the master transcriptional regulator of several entero-hepatic metabolic pathways with relevance to the pathophysiology of conditions such as cholestasis, fatty liver disease, cholesterol gallstone disease, intestinal inflammation and tumors. Furthermore, given the importance of FXR in the gut-liver axis feedbacks regulating lipid and glucose homeostasis, FXR modulation appears to have great input in diseases such as metabolic syndrome and diabetes. Exciting results from several cellular and animal models have provided the impetus to develop synthetic FXR ligands as novel pharmacological agents. Fourteen years from its discovery, FXR has gone from bench to bedside; a novel nuclear receptor ligand is going into clinical use.
Collapse
Affiliation(s)
- Salvatore Modica
- Laboratory of Lipid Metabolism and Cancer, Consorzio Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy
| | | | | |
Collapse
|
22
|
Costa LG, Giordano G, Furlong CE. Pharmacological and dietary modulators of paraoxonase 1 (PON1) activity and expression: the hunt goes on. Biochem Pharmacol 2010; 81:337-44. [PMID: 21093416 DOI: 10.1016/j.bcp.2010.11.008] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 11/07/2010] [Accepted: 11/08/2010] [Indexed: 12/15/2022]
Abstract
Paraoxonase 1 (PON1) is a high density lipoprotein (HDL)-associated enzyme displaying esterase and lactonase activity. PON1 hydrolyzes several organophosphorus (OP) insecticides and nerve agents, a number of exogenous and endogenous lactones, and metabolizes toxic oxidized lipids of low density lipoproteins (LDL) and HDL. As such, PON1 plays a relevant role in determining susceptibility to OP toxicity, cardiovascular diseases and several other diseases. Serum PON1 activity in a given population can vary by at least 40-fold. Most of this variation can be accounted for by genetic polymorphisms in the coding region (Q192R, L55M) and in the promoter region (T-108C). However, exogenous factors may also modulate PON1 activity and/or level of expression. This paper examines various factors that have been found to positively modulate PON1. Certain drugs (e.g. hypolipemic and anti-diabetic compounds), dietary factors (antioxidants, polyphenols), and life-style factors (moderate alcohol consumption) appear to increase PON1 activity. Given the relevance of PON1 in protecting from certain environmental exposure and from cardiovascular and other diseases, there is a need for further mechanistic, animal, and clinical research in this area, and for consideration of possible alternative strategies for increasing the levels and activity of PON1.
Collapse
Affiliation(s)
- Lucio G Costa
- Dept. of Environmental and Occupational Health Sciences, University of Washington, 4225 Roosevelt Way NE, Seattle, WA 98105, USA.
| | | | | |
Collapse
|
23
|
Karami-Mohajeri S, Abdollahi M. Toxic influence of organophosphate, carbamate, and organochlorine pesticides on cellular metabolism of lipids, proteins, and carbohydrates: a systematic review. Hum Exp Toxicol 2010; 30:1119-40. [PMID: 21071550 DOI: 10.1177/0960327110388959] [Citation(s) in RCA: 258] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pesticides, including organophosphate (OP), organochlorine (OC), and carbamate (CB) compounds, are widely used in agricultural and indoor purposes. OP and CB act as acetyl cholinesterase (AChE) inhibitors that affect lots of organs such as peripheral and central nervous systems, muscles, liver, pancreas, and brain, whereas OC are neurotoxic involved in alteration of ion channels. There are several reports about metabolic disorders, hyperglycemia, and also oxidative stress in acute and chronic exposures to pesticides that are linked with diabetes and other metabolic disorders. In this respect, there are several in vitro and in vivo but few clinical studies about mechanism underlying these effects. Bibliographic databases were searched for the years 1963-2010 and resulted in 1652 articles. After elimination of duplicates or irrelevant papers, 204 papers were included and reviewed. Results indicated that OP and CB impair the enzymatic pathways involved in metabolism of carbohydrates, fats and protein within cytoplasm, mitochondria, and proxisomes. It is believed that OP and CB show this effect through inhibition of AChE or affecting target organs directly. OC mostly affect lipid metabolism in the adipose tissues and change glucose pathway in other cells. As a shared mechanism, all OP, CB and OC induce cellular oxidative stress via affecting mitochondrial function and therefore disrupt neuronal and hormonal status of the body. Establishing proper epidemiological studies to explore exact relationships between exposure levels to these pesticides and rate of resulted metabolic disorders in human will be helpful.
Collapse
Affiliation(s)
- Somayyeh Karami-Mohajeri
- Faculty of Pharmacy, and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
24
|
Miyata M, Takamatsu Y, Kuribayashi H, Yamazoe Y. Administration of ampicillin elevates hepatic primary bile acid synthesis through suppression of ileal fibroblast growth factor 15 expression. J Pharmacol Exp Ther 2009; 331:1079-85. [PMID: 19767447 DOI: 10.1124/jpet.109.160093] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Administration of the antibacterial drug ampicillin (ABPC) significantly increased hepatic bile acid concentrations. In the present study, we investigated the mechanisms for the elevation of bile acid levels in ABPC-treated mice. Hepatic microsomal cholesterol 7alpha-hydroxylation and CYP7A1 mRNA level were increased 2.0-fold in ABPC-treated mice despite higher bile acid levels in the liver and small intestinal lumen. A significant change in hepatic small heterodimer partner (SHP) mRNA level was not observed in ABPC-treated mice, whereas a marked decrease in ileal fibroblast growth factor 15 (FGF15) mRNA level was observed (3% of vehicle-treated mice). These phenomena were also observed in mice cotreated with bacitracin/streptomycin/neomycin, which are barely absorbed from the intestine. Primary bile acid contents in the small intestinal lumen were increased in ABPC-treated mice, whereas secondary bile acid, deoxycholic acid (DCA), contents were reduced to below detection limits (<0.01 micromol). In ABPC-treated mice, cotreatment with tauroDCA reversed reductions in ileal FGF15 mRNA level. Ileal SHP mRNA level was, however, not decreased in ABPC-treated mice. ABPC administration to farnesoid X receptor (Fxr)-null mice also decreased ileal FGF15 mRNA levels and secondary bile acid content in the small intestinal lumen. These results suggest that ABPC administration elevates hepatic primary bile acid synthesis, at least in part, through suppression of ileal FGF15 expression.
Collapse
Affiliation(s)
- Masaaki Miyata
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan.
| | | | | | | |
Collapse
|
25
|
Xing X, Burgermeister E, Geisler F, Einwächter H, Fan L, Hiber M, Rauser S, Walch A, Röcken C, Ebeling M, Wright MB, Schmid RM, Ebert MPA. Hematopoietically expressed homeobox is a target gene of farnesoid X receptor in chenodeoxycholic acid-induced liver hypertrophy. Hepatology 2009; 49:979-88. [PMID: 19072826 DOI: 10.1002/hep.22712] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Farnesoid X receptor (FXR/Fxr) is a bile acid-regulated nuclear receptor that promotes hepatic bile acid metabolism, detoxification, and liver regeneration. However, the adaptive pathways under conditions of bile acid stress are not fully elucidated. We found that wild-type but not Fxr knockout mice on diets enriched with chenodeoxycholic acid (CDCA) increase their liver/body weight ratios by 50% due to hepatocellular hypertrophy. Microarray analysis identified Hex (Hematopoietically expressed homeobox), a central transcription factor in vertebrate embryogenesis and liver development, as a novel CDCA- and Fxr-regulated gene. HEX/Hex was also regulated by FXR/Fxr and CDCA in primary mouse hepatocytes and human HepG2 cells. Comparative genomic analysis identified a conserved inverted repeat-1-like DNA sequence within a 300 base pair enhancer element of intron-1 in the human and mouse HEX/Hex gene. A combination of chromatin immunoprecipitation, electromobility shift assay, and transcriptional reporter assays demonstrated that FXR/Fxr binds to this element and mediates HEX/Hex transcriptional activation. CONCLUSION HEX/Hex is a novel bile acid-induced FXR/Fxr target gene during adaptation of hepatocytes to chronic bile acid exposure.
Collapse
Affiliation(s)
- Xiangbin Xing
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
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: 1160] [Impact Index Per Article: 77.3] [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
|
27
|
Horiuchi SI, Ishida S, Hongo T, Ishikawa Y, Miyajima A, Sawada JI, Ohno Y, Nakazawa K, Ozawa S. Global gene expression changes including drug metabolism and disposition induced by three-dimensional culture of HepG2 cells-Involvement of microtubules. Biochem Biophys Res Commun 2008; 378:558-62. [PMID: 19056338 DOI: 10.1016/j.bbrc.2008.11.088] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Accepted: 11/17/2008] [Indexed: 11/18/2022]
Abstract
Constitutive upregulation and a higher degree of induction of drug metabolism and disposition-related genes were found in a three-dimensional HepG2 culture. The upregulated genes are believed to be regulated by different regulatory factors. Global gene expression analysis using the Affymetrix GeneChip indicated that altered expression of microtubule-related genes may change the expressed levels of drug metabolizing and disposition genes. Stabilization of microtubule molecules with docetaxel, a tubulin-stabilizing agent, in the two-dimensional culture showed gene expression patterns similar to those found in the three-dimensional culture, indicating that the culture environment affects drug metabolism functions in HepG2 cells.
Collapse
Affiliation(s)
- Shin-Ichiro Horiuchi
- Division of Pharmacology, National Institute of Health Sciences, Kamiyoga 1-18-1, Setagaya-ku, Tokyo 158-8501, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Dial EJ, Darling RL, Lichtenberger LM. Importance of biliary excretion of indomethacin in gastrointestinal and hepatic injury. J Gastroenterol Hepatol 2008; 23:e384-9. [PMID: 18086111 DOI: 10.1111/j.1440-1746.2007.05266.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS A mechanism for protection of gastrointestinal (GI) and hepatic cells from damaging detergent actions of bile acids appears to involve the bile component, phosphatidylcholine (PC). Non-steroidal anti-inflammatory drugs (NSAIDs) induce intestinal injury in direct proportion to their ability to be excreted into bile, and are known to chemically associate with PC. We investigated the role of bile acids and PC in the mechanism of indomethacin-induced epithelial injury. METHODS Rats were injected orally or intravenously with radiolabeled indomethacin and their bile was collected over time for determination of NSAID secretion. Bile from rats treated with or without indomethacin was used in studies of red blood cell (RBC) hemolysis as a measure of membrane cytotoxicity. The bile salt, sodium deoxycholate (SDC), and indomethacin were tested alone and in combination with PC on RBC and on hepatic HepG2 cells. RESULTS Intravenously or orally given indomethacin was quantitatively excreted (approximately 50%) into bile over a 2-h study period. Bile from a rat treated with indomethacin or bile with exogenous indomethacin was cytotoxic to RBC, and the injury was prevented by the addition of PC. Hepatocytes exposed to SDC showed injury that could be dose-dependently prevented by PC, and reversed by indomethacin. CONCLUSIONS Biliary PC plays an important physiological role in protecting GI and hepatic epithelia from the cytotoxic actions of bile salts. The ability of NSAIDs excreted into the bile to associate with mixed bile salt micelles and reduce the protective action of the PC may be a critical component in the drugs' pathogenic mechanism.
Collapse
Affiliation(s)
- Elizabeth J Dial
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Medical School, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
29
|
Deletion of the ileal basolateral bile acid transporter identifies the cellular sentinels that regulate the bile acid pool. Proc Natl Acad Sci U S A 2008; 105:4965-6. [PMID: 18362334 DOI: 10.1073/pnas.0801194105] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
30
|
Davis RA. Resolving the mechanism of bile acid negative-feedback regulation, a Journal of Lipid Research tradition. J Lipid Res 2008; 49:2-3. [PMID: 18159090 DOI: 10.1194/jlr.e700009-jlr200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
31
|
Carter BA, Taylor OA, Prendergast DR, Zimmerman TL, Von Furstenberg R, Moore DD, Karpen SJ. Stigmasterol, a soy lipid-derived phytosterol, is an antagonist of the bile acid nuclear receptor FXR. Pediatr Res 2007; 62:301-6. [PMID: 17622954 DOI: 10.1203/pdr.0b013e3181256492] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Phytosterols, components of soy-derived lipids, are among the proposed exacerbants of parenteral nutrition-associated cholestasis (PNAC). We investigated whether phytosterols contribute to bile acid (BA)-induced hepatocyte damage by antagonizing a nuclear receptor (NR) critically involved in hepatoprotection from cholestasis, FXR (farnesoid X receptor, NR1H4). In HepG2 cells, stigmasterol acetate (StigAc), a water-soluble Stig derivative, suppressed ligand-activated expression of FXR target genes involved in adaptation to cholestasis (i.e. BSEP, FGF-19, OSTalpha/beta). Furthermore, StigAc antagonized BA-activated, FXR target genes SHP and BSEP in FXR+/+, but not in FXR-/- mouse hepatocytes. Both Stig and StigAc inhibited BA-activated, FXR-dependent reporter gene expression in transfected HepG2 cells, whereas the most prevalent phytosterol in lipids, beta-sitosterol, had no inhibitory effect. Finally, among six ligand-activated NR-ligand binding domains (LBDs) tested, antagonism by StigAc was specific to only two (FXR and PXR, pregnane X receptor, NR1I2). We demonstrate that Stig, a phytosterol prevalent in soy-derived PN lipid solutions, is a potent in vitro antagonist of the NR for bile acids FXR.
Collapse
Affiliation(s)
- Beth A Carter
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Texas Children's Liver Center, Baylor College of Medicine, Houston 77030, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Kim I, Ahn SH, Inagaki T, Choi M, Ito S, Guo GL, Kliewer SA, Gonzalez FJ. Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine. J Lipid Res 2007; 48:2664-72. [PMID: 17720959 DOI: 10.1194/jlr.m700330-jlr200] [Citation(s) in RCA: 440] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Bile acid concentrations are controlled by a feedback regulatory pathway whereby activation of the farnesoid X receptor (FXR) represses transcription of both the CYP7A1 gene, encoding the rate-limiting enzyme in the classic bile acid synthesis pathway, and the CYP8B1 gene, required for synthesis of cholic acid. The tissue-specific roles of FXR were examined using liver- and intestine-specific FXR-null models. FXR deficiency in either liver (Fxr DeltaL) or intestine (Fxr DeltaIE) increased bile acid pool size. Treatment with the FXR-selective agonist GW4064 significantly repressed CYP7A1 in Fxr DeltaL mice but not Fxr DeltaIE mice, demonstrating that activation of FXR in intestine but not liver is required for short-term repression of CYP7A1 in liver. This intestinal-specific effect of FXR is likely mediated through induction of the hormone FGF15, which suppresses CYP7A1. In comparison to CYP7A1, FXR-mediated repression of CYP8B1 was more dependent on the presence of FXR in liver and less dependent on its presence in intestine. Consistent with these findings, recombinant FGF15 repressed CYP7A1 mRNA levels without affecting CYP8B1 expression. These data provide evidence that FXR-mediated repression of bile acid synthesis requires the complementary actions of FXR in both liver and intestine and reveal mechanistic differences in feedback repression of CYP7A1 and CYP8B1.
Collapse
Affiliation(s)
- Insook Kim
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Kuipers F, Stroeve JHM, Caron S, Staels B. Bile acids, farnesoid X receptor, atherosclerosis and metabolic control. Curr Opin Lipidol 2007; 18:289-97. [PMID: 17495603 DOI: 10.1097/mol.0b013e3281338d08] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Bile acids are amphiphilic molecules synthesized from cholesterol exclusively in the liver that are essential for effective absorption of dietary fat. In addition to this 'classical role', bile acids act as signalling molecules that control their own metabolism by activating the nuclear receptor, farnesoid X receptor. RECENT FINDINGS Recent work demonstrates that farnesoid X receptor exerts metabolic control beyond bile acid homeostasis, notably effects on HDL, triglyceride and glucose metabolism. Farnesoid X receptor influences insulin sensitivity of tissues that are not part of the enterohepatic circulation, for example, adipose tissue. Certain metabolic effects in the liver appear to be mediated via farnesoid X receptor-stimulated release of an intestinal growth factor. In addition, novel signalling pathways independent of farnesoid X receptor have been identified that may contribute to bile acid-mediated metabolic regulation. SUMMARY Farnesoid X receptor represents a potentially attractive target for treatment of various aspects of the metabolic syndrome and for prevention of atherosclerosis. Yet, in view of its pleiotropic effects and apparent species-specificity, it is evident that successful interference of the farnesoid X receptor signalling system will require the development of gene-specific and/or organ-specific farnesoid X receptor modulators and extensive testing in human models of disease.
Collapse
Affiliation(s)
- Folkert Kuipers
- Center for Liver, Digestive and Metabolic Diseases, University Medical Center Groningen, The Netherlands.
| | | | | | | |
Collapse
|
34
|
Cariou B, Staels B. FXR: a promising target for the metabolic syndrome? Trends Pharmacol Sci 2007; 28:236-43. [PMID: 17412431 DOI: 10.1016/j.tips.2007.03.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2006] [Revised: 02/12/2007] [Accepted: 03/21/2007] [Indexed: 12/23/2022]
Abstract
The metabolic syndrome is an insulin-resistant state that is characterized by a cluster of cardiovascular risk factors, including abdominal obesity, hyperglycemia, elevated blood pressure and combined dyslipidemia. In this review, we discuss the role of the bile-acid-activated farnesoid X receptor (FXR) in the modulation of the metabolic syndrome. Owing to its regulatory actions in lipid and glucose homeostasis, FXR is a potential pharmacological target. Moreover, the observation that FXR also influences endothelial function and atherosclerosis indicates a regulatory role in the cardiovascular complications that are associated with the metabolic syndrome. The pharmacological activation of FXR leads to a complex response that integrates beneficial actions and potentially undesirable side-effects. Thus, the identification of selective FXR modulators (selective bile acid receptor modulators) is required for the development of compounds that can be used to treat the metabolic syndrome.
Collapse
Affiliation(s)
- Bertrand Cariou
- Centre Hospitalier Universitaire Hôtel-Dieu, Nantes 44093, France.
| | | |
Collapse
|
35
|
Fickert P, Moustafa T, Trauner M. Primary sclerosing cholangitis--the arteriosclerosis of the bile duct? Lipids Health Dis 2007; 6:3. [PMID: 17254334 PMCID: PMC1796546 DOI: 10.1186/1476-511x-6-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 01/25/2007] [Indexed: 12/11/2022] Open
Abstract
Primary sclerosing cholangitis (PSC) is a chronic inflammatory disease of unknown aetiology affecting the large bile ducts and characterized by periductal fibrosis and stricture formation, which ultimately result in biliary cirrhosis and liver failure. Arteriosclerosis involves the accumulation of altered lipids and lipoproteins in large arteries; this drives inflammation and fibrosis and ultimately leads to narrowing of the arteries and hypoperfusion of dependent organs and tissues. Knowledge of the causative factors is crucial to the understanding of disease mechanisms and the development of specific treatment. Based on pathogenetic similarities between PSC and arteriosclerosis, we hypothesize that PSC represents "arteriosclerosis of the bile duct" initiated by toxic biliary lipids. This hypothesis is based on common molecular, cellular, and morphological features providing the conceptual framework for a deeper understanding of their pathogenesis. This hypothesis should stimulate translational research to facilitate the search for novel treatment strategies for both diseases.
Collapse
Affiliation(s)
- Peter Fickert
- Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Tarek Moustafa
- Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Michael Trauner
- Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| |
Collapse
|
36
|
Lee FY, Lee H, Hubbert ML, Edwards PA, Zhang Y. FXR, a multipurpose nuclear receptor. Trends Biochem Sci 2006; 31:572-80. [PMID: 16908160 DOI: 10.1016/j.tibs.2006.08.002] [Citation(s) in RCA: 259] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Revised: 07/14/2006] [Accepted: 08/03/2006] [Indexed: 12/20/2022]
Abstract
The farnesoid X receptor (FXR) is a ligand-activated transcription factor and a member of the nuclear receptor superfamily. In the past six years, remarkable inroads have been made into determining the functional importance of FXR. This receptor has been shown to have crucial roles in controlling bile acid homeostasis, lipoprotein and glucose metabolism, hepatic regeneration, intestinal bacterial growth and the response to hepatotoxins. Thus, the development of FXR agonists might prove useful for the treatment of diabetes, cholesterol gallstones, and hepatic and intestinal toxicity.
Collapse
Affiliation(s)
- Florence Y Lee
- Department of Biological Chemistry, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|