151
|
Rendic SP, Peter Guengerich F. Human cytochrome P450 enzymes 5-51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition - toxic effects and benefits. Drug Metab Rev 2019; 50:256-342. [PMID: 30717606 DOI: 10.1080/03602532.2018.1483401] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 (P450, CYP) enzymes have long been of interest due to their roles in the metabolism of drugs, pesticides, pro-carcinogens, and other xenobiotic chemicals. They have also been of interest due to their very critical roles in the biosynthesis and metabolism of steroids, vitamins, and certain eicosanoids. This review covers the 22 (of the total of 57) human P450s in Families 5-51 and their substrate selectivity. Furthermore, included is information and references regarding inducibility, inhibition, and (in some cases) stimulation by chemicals. We update and discuss important aspects of each of these 22 P450s and questions that remain open.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- b Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
152
|
Baiocchi L, Zhou T, Liangpunsakul S, Lenci I, Santopaolo F, Meng F, Kennedy L, Glaser S, Francis H, Alpini G. Dual Role of Bile Acids on the Biliary Epithelium: Friend or Foe? Int J Mol Sci 2019; 20:1869. [PMID: 31014010 PMCID: PMC6514722 DOI: 10.3390/ijms20081869] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/11/2019] [Accepted: 04/13/2019] [Indexed: 12/12/2022] Open
Abstract
Bile acids are a family of amphipathic compounds predominantly known for their role in solubilizing and absorbing hydrophobic compounds (including liposoluble vitamins) in the intestine. Bile acids also are key signaling molecules and inflammatory agents that activate transcriptional factors and cell signaling pathways that regulate lipid, glucose, and energy metabolism in various human disorders, including chronic liver diseases. However, in the last decade increased awareness has been founded on the physiological and chemical heterogeneity of this category of compounds and their possible beneficial or injurious effects on the biliary tree. In this review, we provide an update on the current understanding of the molecular mechanism involving bile acid and biliary epithelium. The last achievements of the research in this field are summarized, focusing on the molecular aspects and the elements with relevance regarding human liver diseases.
Collapse
Affiliation(s)
- Leonardo Baiocchi
- Liver Unit, Department of Medicine, University of Rome "Tor Vergata", Viale Oxford 81, 00133 Rome, Italy.
| | - Tianhao Zhou
- Department of Medical Physiology, Texas A&M University, College of Medicine 702 SW HK Dodgen Loop, Temple, TX 76504, USA.
| | - Suthat Liangpunsakul
- Richard L. Roudebush VA Medical Center and Indiana University, Gastroenterology, Medicine 1481 W 10th street, Dedication Wing⁻Room C-7151, Indianapolis, IN 46202, USA.
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, 1481 W 10th street, Indianapolis, IN 46202, USA.
| | - Ilaria Lenci
- Liver Unit, Department of Medicine, University of Rome "Tor Vergata", Viale Oxford 81, 00133 Rome, Italy.
| | - Francesco Santopaolo
- Liver Unit, Department of Medicine, University of Rome "Tor Vergata", Viale Oxford 81, 00133 Rome, Italy.
| | - Fanyin Meng
- Richard L. Roudebush VA Medical Center and Indiana University, Gastroenterology, Medicine 1481 W 10th street, Dedication Wing⁻Room C-7151, Indianapolis, IN 46202, USA.
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, 1481 W 10th street, Indianapolis, IN 46202, USA.
| | - Lindsey Kennedy
- Richard L. Roudebush VA Medical Center and Indiana University, Gastroenterology, Medicine 1481 W 10th street, Dedication Wing⁻Room C-7151, Indianapolis, IN 46202, USA.
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University, College of Medicine 702 SW HK Dodgen Loop, Temple, TX 76504, USA.
| | - Heather Francis
- Richard L. Roudebush VA Medical Center and Indiana University, Gastroenterology, Medicine 1481 W 10th street, Dedication Wing⁻Room C-7151, Indianapolis, IN 46202, USA.
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, 1481 W 10th street, Indianapolis, IN 46202, USA.
| | - Gianfranco Alpini
- Richard L. Roudebush VA Medical Center and Indiana University, Gastroenterology, Medicine 1481 W 10th street, Dedication Wing⁻Room C-7151, Indianapolis, IN 46202, USA.
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, 1481 W 10th street, Indianapolis, IN 46202, USA.
| |
Collapse
|
153
|
Wilkens MR, Firmenich CS, Schnepel N, Muscher-Banse AS. A reduced protein diet modulates enzymes of vitamin D and cholesterol metabolism in young ruminants. J Steroid Biochem Mol Biol 2019; 186:196-202. [PMID: 30394334 DOI: 10.1016/j.jsbmb.2018.10.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/16/2018] [Accepted: 10/24/2018] [Indexed: 12/19/2022]
Abstract
Besides other adverse effects, a low protein diet has been shown to modulate cholesterol and vitamin D metabolism in monogastric species like rats and humans. As ruminants can increase the efficiency of the rumino-hepatic circulation of urea, it is assumed that goats should be able to compensate for a low dietary protein intake better. After a dietary protein restriction (9% vs. 20%) for six weeks, plasma concentrations of urea, albumin, 1,25-dihydroxyvitamin D3 and calcium were decreased, while plasma 25-hydroxyvitamin D3 (25-OHD3), and total cholesterol were significantly increased in young goats. Because this was not accompanied by any decrease in expression of CYP24A1 mRNA, we investigated mRNA expression of additional enzymes with known 24- and/or 25-hydroxylase activities (CYP2R1, CYP2J2, CYP3 A24, CYP27A1), receptors involved in their regulation (VDR, PXR, RXRα) and vitamin D binding protein (VDBP). CYP2R1expression was stimulated with the low dietary protein intake, negatively correlated with plasma urea and positively associated with serum 25-OHD3. The greater plasma concentrations of total cholesterol could be explained with the reduction of CYP2J2 and CYP27A1 expression. None of the receptors investigated were affected by the dietary protein restriction but mRNA expression of VDBP was slightly reduced. Taken together our results show that dietary protein restriction has an impact on vitamin D and cholesterol metabolism in ruminants, too. Therefore, further investigations are needed before dietary interventions aiming at diminishing nitrogen excretion can be implemented.
Collapse
Affiliation(s)
- Mirja R Wilkens
- Department of Physiology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173 Hannover, Germany
| | - Caroline S Firmenich
- Department of Physiology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173 Hannover, Germany
| | - Nadine Schnepel
- Department of Physiology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173 Hannover, Germany
| | - Alexandra S Muscher-Banse
- Department of Physiology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173 Hannover, Germany.
| |
Collapse
|
154
|
Ibrahim S, Dayoub R, Krautbauer S, Liebisch G, Wege AK, Melter M, Weiss TS. Bile acid-induced apoptosis and bile acid synthesis are reduced by over-expression of Augmenter of Liver Regeneration (ALR) in a STAT3-dependent mechanism. Exp Cell Res 2019; 374:189-197. [DOI: 10.1016/j.yexcr.2018.11.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/31/2018] [Accepted: 11/25/2018] [Indexed: 11/24/2022]
|
155
|
Parmentier C, Couttet P, Uteng M, Wolf A, Richert L. Transcriptomic Analysis of Cholestatic Compounds In Vitro. Methods Mol Biol 2019; 1981:175-186. [PMID: 31016655 DOI: 10.1007/978-1-4939-9420-5_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Drug-induced cholestasis is one of the most severe manifestations of drug-induced liver injury. Drug-induced cholestasis is characterized by an accumulation of endogenous metabolites normally excreted in the bile such as bile salts, cholesterol, bilirubin, or drug metabolites. The possibility to determine early in the drug development process whether a compound presents a risk of inducing drug-induced cholestasis is key information. Since preclinical repeated dose toxicity studies have limited predictive value, large efforts in identifying alternative in vitro models with improved prediction are being made. One of the best current models for in vitro human liver is primary human hepatocytes, and we recently reported that primary human hepatocytes can be kept as long-term cultures in 2D-sandwich configuration when regularly renewing the Matrigel overlay, thereby making the model useful for repeat exposure-related toxicities, as well as for the study of adaptive responses. This primary human hepatocyte culture system combined with transcriptomics carries the future promise to identify individual gene expression profiles predictive of increased drug-induced cholestasis risk.This chapter describes the various steps for culturing and exposing primary human hepatocytes to drugs during long-term 2D-sandwich culture, performing RNA extraction, gene chip assay and selecting hepatotoxic signature using the IPA software and highlighting genes involved in bile acid homeostasis.
Collapse
Affiliation(s)
| | | | - Marianne Uteng
- Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Armin Wolf
- The Janssen Pharmaceutical Companies of Johnson and Johnson, Beerse, Belgium
| | | |
Collapse
|
156
|
Gao Y, Li W, Chen J, Wang X, Lv Y, Huang Y, Zhang Z, Xu F. Pharmacometabolomic prediction of individual differences of gastrointestinal toxicity complicating myelosuppression in rats induced by irinotecan. Acta Pharm Sin B 2019; 9:157-166. [PMID: 30766787 PMCID: PMC6362258 DOI: 10.1016/j.apsb.2018.09.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/21/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022] Open
Abstract
Pharmacometabolomics has been already successfully used in toxicity prediction for one specific adverse effect. However in clinical practice, two or more different toxicities are always accompanied with each other, which puts forward new challenges for pharmacometabolomics. Gastrointestinal toxicity and myelosuppression are two major adverse effects induced by Irinotecan (CPT-11), and often show large individual differences. In the current study, a pharmacometabolomic study was performed to screen the exclusive biomarkers in predose serums which could predict late-onset diarrhea and myelosuppression of CPT-11 simultaneously. The severity and sensitivity differences in gastrointestinal toxicity and myelosuppression were judged by delayed-onset diarrhea symptoms, histopathology examination, relative cytokines and blood cell counts. Mass spectrometry-based non-targeted and targeted metabolomics were conducted in sequence to dissect metabolite signatures in predose serums. Eventually, two groups of metabolites were screened out as predictors for individual differences in late-onset diarrhea and myelosuppression using binary logistic regression, respectively. This result was compared with existing predictors and validated by another independent external validation set. Our study indicates the prediction of toxicity could be possible upon predose metabolic profile. Pharmacometabolomics can be a potentially useful tool for complicating toxicity prediction. Our findings also provide a new insight into CPT-11 precision medicine.
Collapse
Key Words
- AUC-ROC, area under receiver operating characteristic
- BHB, β-hydroxybutyric acid
- Biomarkers
- C, control group
- CA, cholic acid
- CPT-11, irinotecan
- Complicating toxicity
- DBIL, direct bilirubin
- DCA, deoxycholic acid
- Diarrhea
- FDR, false discovery rate
- GCA, glycocholic acid
- Gastrointestinal toxicity
- IBIL, indirect bilirubin
- IT-TOF/MS, ion trap/time-offlight hybrid mass spectrometry
- Individual differences
- Irinotecan
- Lys, lysine
- MSTFA, N-methyl-N-trifluoroacetamide
- Metabolomics
- NS, non-sensitive group
- NSgt, non-sensitive for gastrointestinal toxicity
- NSmt, non-sensitive for myelosuppression toxicity
- OPLS-DA, orthogonal partial least-squares-discriminant analysis
- PCA, principal component analysis
- PLS-DA, partial least-squares-discriminant analysis
- Phe, phenylalanine
- Prediction
- QC, quality control
- RSD, relative standard deviation
- S, sensitive group
- Sgt, sensitive for gastrointestinal toxicity
- Smt, sensitive for myelosuppression toxicity
- T, CPT-11 treated group
- Trp, tryptophan
- UFLC, ultrafast liquid chromatography
- VIP, variable importance in the projection
- pFDR, false-discovery-rate-adjusted P value
Collapse
Affiliation(s)
- Yiqiao Gao
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Wei Li
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Jiaqing Chen
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Xu Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Yingtong Lv
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Yin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Fengguo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
157
|
Nguyen TT, Ung TT, Kim NH, Jung YD. Role of bile acids in colon carcinogenesis. World J Clin Cases 2018; 6:577-588. [PMID: 30430113 PMCID: PMC6232560 DOI: 10.12998/wjcc.v6.i13.577] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/15/2018] [Accepted: 10/11/2018] [Indexed: 02/05/2023] Open
Abstract
Bile acids (BAs) are cholesterol derivatives synthesized in the liver and then secreted into the intestine for lipid absorption. There are numerous scientific reports describing BAs, especially secondary BAs, as strong carcinogens or promoters of colon cancers. Firstly, BAs act as strong stimulators of colorectal cancer (CRC) initiation by damaging colonic epithelial cells, and inducing reactive oxygen species production, genomic destabilization, apoptosis resistance, and cancer stem cells-like formation. Consequently, BAs promote CRC progression via multiple mechanisms, including inhibiting apoptosis, enhancing cancer cell proliferation, invasion, and angiogenesis. There are diverse signals involved in the carcinogenesis mechanism of BAs, with a major role of epidermal growth factor receptor, and its down-stream signaling, involving mitogen-activated protein kinase, phosphoinositide 3-kinase/Akt, and nuclear factor kappa-light-chain-enhancer of activated B cells. BAs regulate numerous genes including the human leukocyte antigen class I gene, p53, matrix metalloprotease, urokinase plasminogen activator receptor, Cyclin D1, cyclooxygenase-2, interleukin-8, and miRNAs of CRC cells, leading to CRC promotion. These evidence suggests that targeting BAs is an efficacious strategies for CRC prevention and treatment.
Collapse
Affiliation(s)
- Thi Thinh Nguyen
- Department of Biochemistry, Chonnam National University Medical School, Jeonnam 58138, South Korea
| | - Trong Thuan Ung
- Department of Biochemistry, Chonnam National University Medical School, Jeonnam 58138, South Korea
| | - Nam Ho Kim
- Department of Nephrology, Chonnam National University Medical School, Gwangju 501-190, South Korea
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Jeonnam 58138, South Korea
| |
Collapse
|
158
|
Sun X, Jia Z. Microbiome modulates intestinal homeostasis against inflammatory diseases. Vet Immunol Immunopathol 2018; 205:97-105. [PMID: 30459007 DOI: 10.1016/j.vetimm.2018.10.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/21/2018] [Accepted: 10/27/2018] [Indexed: 02/07/2023]
Abstract
Eliminating prophylactic antibiotics in food animal production has exerted pressure on discovering antimicrobial alternatives (e.g. microbiome) to reduce elevated intestinal diseases. Intestinal tract is a complex ecosystem coupling host cells with microbiota. The microbiota and its metabolic activities and products are collectively called microbiome. Intestinal homeostasis is reached through dynamic and delicate crosstalk between host immunity and microbiome. However, this balance can be occasionally broken, which results in intestinal inflammatory diseases such as human Inflammatory Bowel Diseases, chicken necrotic enteritis, and swine postweaning diarrhea. In this review, we introduce the intestinal immune system, intestinal microbiome, and microbiome modulation of inflammation against intestinal diseases. The purpose of this review is to provide updated knowledge on host-microbe interaction and to promote using microbiome as new antimicrobial strategies to reduce intestinal diseases.
Collapse
Affiliation(s)
- Xiaolun Sun
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, 72701, United States.
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27402, United States
| |
Collapse
|
159
|
Kim T, Nason S, Holleman C, Pepin M, Wilson L, Berryhill TF, Wende AR, Steele C, Young ME, Barnes S, Drucker DJ, Finan B, DiMarchi R, Perez-Tilve D, Tschöp M, Habegger KM. Glucagon Receptor Signaling Regulates Energy Metabolism via Hepatic Farnesoid X Receptor and Fibroblast Growth Factor 21. Diabetes 2018; 67:1773-1782. [PMID: 29925501 PMCID: PMC6110317 DOI: 10.2337/db17-1502] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
Glucagon, an essential regulator of glucose and lipid metabolism, also promotes weight loss, in part through potentiation of fibroblast growth factor 21 (FGF21) secretion. However, FGF21 is only a partial mediator of metabolic actions ensuing from glucagon receptor (GCGR) activation, prompting us to search for additional pathways. Intriguingly, chronic GCGR agonism increases plasma bile acid levels. We hypothesized that GCGR agonism regulates energy metabolism, at least in part, through farnesoid X receptor (FXR). To test this hypothesis, we studied whole-body and liver-specific FXR-knockout (Fxr∆liver) mice. Chronic GCGR agonist (IUB288) administration in diet-induced obese (DIO) Gcgr, Fgf21, and Fxr whole-body or liver-specific knockout (∆liver) mice failed to reduce body weight when compared with wild-type (WT) mice. IUB288 increased energy expenditure and respiration in DIO WT mice, but not Fxr∆liver mice. GCGR agonism increased [14C]palmitate oxidation in hepatocytes isolated from WT mice in a dose-dependent manner, an effect blunted in hepatocytes from Fxr∆liver mice. Our data clearly demonstrate that control of whole-body energy expenditure by GCGR agonism requires intact FXR signaling in the liver. This heretofore-unappreciated aspect of glucagon biology has implications for the use of GCGR agonism in the therapy of metabolic disorders.
Collapse
MESH Headings
- Adiposity/drug effects
- Animals
- Anti-Obesity Agents/therapeutic use
- Calorimetry, Indirect
- Cells, Cultured
- Diet, High-Fat/adverse effects
- Energy Metabolism/drug effects
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Gene Expression Regulation/drug effects
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Liver/drug effects
- Mitochondria, Liver/enzymology
- Mitochondria, Liver/metabolism
- Obesity/drug therapy
- Obesity/etiology
- Obesity/metabolism
- Obesity/pathology
- Organ Specificity
- Oxidative Phosphorylation/drug effects
- Peptides/therapeutic use
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Glucagon/agonists
- Receptors, Glucagon/genetics
- Receptors, Glucagon/metabolism
- Signal Transduction/drug effects
- Weight Gain/drug effects
Collapse
Affiliation(s)
- Teayoun Kim
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Shelly Nason
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Cassie Holleman
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Mark Pepin
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, AL
| | - Landon Wilson
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL
| | - Taylor F Berryhill
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, AL
| | - Chad Steele
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Stephen Barnes
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN
| | - Richard DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN
- Department of Chemistry, Indiana University, Bloomington, IN
| | - Diego Perez-Tilve
- Division of Endocrinology, Diabetes and Metabolism, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
| | - Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - Kirk M Habegger
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
160
|
Navarro Suarez L, Brückner L, Rohn S. Electrochemical Oxidation of Primary Bile Acids: A Tool for Simulating Their Oxidative Metabolism? Int J Mol Sci 2018; 19:E2491. [PMID: 30142907 PMCID: PMC6165074 DOI: 10.3390/ijms19092491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/16/2018] [Accepted: 08/18/2018] [Indexed: 01/14/2023] Open
Abstract
Bile acids are a subgroup of sterols and important products of cholesterol catabolism in mammalian organisms. Modifications (e.g., oxidation and 7-dehydroxylation) are predominantly exerted by the intestinal microbiota. Bile acids can be found in almost all living organisms, and their concentration and metabolism can be used for the assessment of the pathological and nutritional status of an organism. Electrochemical oxidation is a rapid, relatively inexpensive approach to simulate natural metabolic redox processes in vitro. This technique further allows the identification of oxidative degradation pathways of individual substances, as well as the demonstration of binding studies of generated oxidation products with biologically relevant molecules. When coupling an electrochemical and a high-resolution mass spectrometric system, oxidation products can be generated and identified directly by non-targeted ESI-MS. Here, a method for the generation of oxidation products of the primary bile acids cholic acid and chenodeoxycholic acid was exemplarily developed. Most products and the highest intensities were observed at a pH value of 6. For cholic acid, a high potential of 3 V was necessary, while for chenodeoxycholic acid, a potential of 2.4 V led to a higher number of oxidation products. In a second approach, a binding study with glutathione was performed to simulate phase II metabolism. It was possible to detect signals of free glutathione, free bile acids, and adducts of both reactants. As the resulting mass spectra also showed some new signals of the oxidized bile acid, which could not be observed without glutathione, it can be assumed that glutathione is able to bind reactive oxidation species before reacting with other products.
Collapse
Affiliation(s)
- Laura Navarro Suarez
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany.
| | - Lea Brückner
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany.
| | - Sascha Rohn
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany.
| |
Collapse
|
161
|
Li CY, Dempsey JL, Wang D, Lee S, Weigel KM, Fei Q, Bhatt DK, Prasad B, Raftery D, Gu H, Cui JY. PBDEs Altered Gut Microbiome and Bile Acid Homeostasis in Male C57BL/6 Mice. Drug Metab Dispos 2018; 46:1226-1240. [PMID: 29769268 PMCID: PMC6053593 DOI: 10.1124/dmd.118.081547] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/11/2018] [Indexed: 12/14/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are persistent environmental contaminants with well characterized toxicities in host organs. Gut microbiome is increasingly recognized as an important regulator of xenobiotic biotransformation; however, little is known about its interactions with PBDEs. Primary bile acids (BAs) are metabolized by the gut microbiome into more lipophilic secondary BAs that may be absorbed and interact with certain host receptors. The goal of this study was to test our hypothesis that PBDEs cause dysbiosis and aberrant regulation of BA homeostasis. Nine-week-old male C57BL/6 conventional (CV) and germ-free (GF) mice were orally gavaged with corn oil (10 mg/kg), BDE-47 (100 μmol/kg), or BDE-99 (100 μmol/kg) once daily for 4 days (n = 3-5/group). Gut microbiome was characterized using 16S rRNA sequencing of the large intestinal content in CV mice. Both BDE-47 and BDE-99 profoundly decreased the alpha diversity of gut microbiome and differentially regulated 45 bacterial species. Both PBDE congeners increased Akkermansia muciniphila and Erysipelotrichaceae Allobaculum spp., which have been reported to have anti-inflammatory and antiobesity functions. Targeted metabolomics of 56 BAs was conducted in serum, liver, and small and large intestinal content of CV and GF mice. BDE-99 increased many unconjugated BAs in multiple biocompartments in a gut microbiota-dependent manner. This correlated with an increase in microbial 7α-dehydroxylation enzymes for secondary BA synthesis and increased expression of host intestinal transporters for BA absorption. Targeted proteomics showed that PBDEs downregulated host BA-synthesizing enzymes and transporters in livers of CV but not GF mice. In conclusion, there is a novel interaction between PBDEs and the endogenous BA-signaling through modification of the "gut-liver axis".
Collapse
Affiliation(s)
- Cindy Yanfei Li
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Joseph L Dempsey
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Dongfang Wang
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - SooWan Lee
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Kris M Weigel
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Qiang Fei
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Deepak Kumar Bhatt
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Bhagwat Prasad
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Daniel Raftery
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Haiwei Gu
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| | - Julia Yue Cui
- Departments of Environmental and Occupational Health Sciences (C.Y.F., J.L.D., S.L., K.M.W., J.Y.C.) and Pharmaceutics (D.K.B., B.P.) and Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine (D.W., Q.F., D.R.), University of Washington, Seattle, Washington; Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona (H.G.); Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China (D.W.); and Department of Chemistry, Jilin University, Changchun, Jilin Province, P. R. China (Q.F.)
| |
Collapse
|
162
|
Peroxisome Proliferator-Activated Receptor- γ Prevents Cholesterol Gallstone Formation in C57bl Mice by Regulating Bile Acid Synthesis and Enterohepatic Circulation. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7475626. [PMID: 30105244 PMCID: PMC6076980 DOI: 10.1155/2018/7475626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/13/2018] [Accepted: 06/27/2018] [Indexed: 12/28/2022]
Abstract
To investigate the role of the peroxisome proliferator-activated receptor-γ (PPARγ) in the progression of cholesterol gallstone disease (CGD), C57bl/6J mice were randomized to the following groups (n=7/group): L (lithogenic diet, LGD), LM (LGD+pioglitazone), CM (chow diet+pioglitazone), and NC (normal control, chow diet). Gallbladder stones were observed by microscopy. Histological gallbladder changes were assessed. Bile acids (BA) and cholesterol were measured in the serum, bile, and feces. Proteins and mRNA expression of genes involved in BA metabolism and enterohepatic circulation were assessed by western blotting and real-time RT-PCR. PPARγ activation was performed in LO2 cell by lentivirus transfection and in Caco2 cell by PPARγ agonist treatment. Downregulation of farnesoid X receptor (FXR) by small interference RNA (siRNA) was performed in L02 cells and Caco2 cells, respectively. Results showed that pharmacological activation of PPARγ by pioglitazone prevents cholesterol gallstone formation by increasing biliary BA synthesis and enterohepatic circulation. Activated PPARγ induced the expression of genes involved in enterohepatic circulation and bile acid synthesis (like PCG1α, BSEP, MRP2, MRP3, MRP4, NTCP, CYP7A1, CYP27A1, ASBT, OSTα, and OSTβ). Downregulation of FXR repressed expression of partial genes involved in BA enterohepatic circulation. These findings suggest a new function of PPARγ in preventing CGD by handling BA synthesis and transport through a FXR dependent or independent pathway.
Collapse
|
163
|
Enright EF, Griffin BT, Gahan CG, Joyce SA. Microbiome-mediated bile acid modification: Role in intestinal drug absorption and metabolism. Pharmacol Res 2018; 133:170-186. [DOI: 10.1016/j.phrs.2018.04.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/07/2018] [Accepted: 04/12/2018] [Indexed: 01/03/2023]
|
164
|
Abstract
Bile acids facilitate intestinal nutrient absorption and biliary cholesterol secretion to maintain bile acid homeostasis, which is essential for protecting liver and other tissues and cells from cholesterol and bile acid toxicity. Bile acid metabolism is tightly regulated by bile acid synthesis in the liver and bile acid biotransformation in the intestine. Bile acids are endogenous ligands that activate a complex network of nuclear receptor farnesoid X receptor and membrane G protein-coupled bile acid receptor-1 to regulate hepatic lipid and glucose metabolic homeostasis and energy metabolism. The gut-to-liver axis plays a critical role in the regulation of enterohepatic circulation of bile acids, bile acid pool size, and bile acid composition. Bile acids control gut bacteria overgrowth, and gut bacteria metabolize bile acids to regulate host metabolism. Alteration of bile acid metabolism by high-fat diets, sleep disruption, alcohol, and drugs reshapes gut microbiome and causes dysbiosis, obesity, and metabolic disorders. Gender differences in bile acid metabolism, FXR signaling, and gut microbiota have been linked to higher prevalence of fatty liver disease and hepatocellular carcinoma in males. Alteration of bile acid homeostasis contributes to cholestatic liver diseases, inflammatory diseases in the digestive system, obesity, and diabetes. Bile acid-activated receptors are potential therapeutic targets for developing drugs to treat metabolic disorders.
Collapse
Affiliation(s)
- John Y. L. Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Jessica M. Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
165
|
Chen MM, Hale C, Stanislaus S, Xu J, Véniant MM. FGF21 acts as a negative regulator of bile acid synthesis. J Endocrinol 2018; 237:139-152. [PMID: 29615519 DOI: 10.1530/joe-17-0727] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/26/2018] [Indexed: 12/17/2022]
Abstract
Fibroblast growth factor 21 (FGF21) is a potent regulator of glucose and lipid homeostasis in vivo; its most closely related subfamily member, FGF19, is known to be a critical negative regulator of bile acid synthesis. To delineate whether FGF21 also plays a functional role in bile acid metabolism, we evaluated the effects of short- and long-term exposure to native FGF21 and long-acting FGF21 analogs on hepatic signal transduction, gene expression and enterohepatic bile acid levels in primary hepatocytes and in rodent and monkey models. FGF21 acutely induced ERK phosphorylation and inhibited Cyp7A1 mRNA expression in primary hepatocytes and in different rodent models, although less potently than recombinant human FGF19. Long-term administration of FGF21 in mice fed a standard chow diet resulted in a 50-60% decrease in bile acid levels in the liver and small intestines and consequently a 60% reduction of bile acid pool size. In parallel, colonic and fecal bile acid was decreased, whereas fecal cholesterol and fatty acid excretions were elevated. The long-acting FGF21 analog showed superiority to recombinant human FGF21 and FGF19 in decreasing bile acid levels with long duration of effect action in mice. Long-term administration of the long-acting FGF21 analogs in obese cynomolgus monkeys suppressed plasma total bile acid and 7α-hydroxy-4-cholesten-3-one levels, a biomarker for bile acid synthesis. Collectively, these data reveal a previously unidentified role of FGF21 in bile acid metabolism as a negative regulator of bile acid synthesis.
Collapse
Affiliation(s)
| | | | | | | | - Murielle M Véniant
- Department of Cardiometabolic DisordersAmgen Inc., Thousand Oaks, California, USA
| |
Collapse
|
166
|
Quach HP, Dzekic T, Bukuroshi P, Pang KS. Potencies of vitamin D analogs, 1α-hydroxyvitamin D3
, 1α-hydroxyvitamin D2
and 25-hydroxyvitamin D3
, in lowering cholesterol in hypercholesterolemic mice in vivo. Biopharm Drug Dispos 2018; 39:196-204. [DOI: 10.1002/bdd.2126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/28/2018] [Accepted: 02/11/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Holly P. Quach
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy; University of Toronto; Toronto Ontario Canada
| | - Tamara Dzekic
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy; University of Toronto; Toronto Ontario Canada
| | - Paola Bukuroshi
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy; University of Toronto; Toronto Ontario Canada
| | - K. Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy; University of Toronto; Toronto Ontario Canada
| |
Collapse
|
167
|
Abstract
Steroids play vital roles in animal physiology across species, and the production of specific steroids is associated with particular internal biological functions. The internal functions of steroids are, in most cases, quite clear. However, an important feature of many steroids (their chemical stability) allows these molecules to play secondary, external roles as chemical messengers after their excretion via urine, feces, or other shed substances. The presence of steroids in animal excretions has long been appreciated, but their capacity to serve as chemosignals has not received as much attention. In theory, the blend of steroids excreted by an animal contains a readout of its own biological state. Initial mechanistic evidence for external steroid chemosensation arose from studies of many species of fish. In sea lampreys and ray-finned fishes, bile salts were identified as potent olfactory cues and later found to serve as pheromones. Recently, we and others have discovered that neurons in amphibian and mammalian olfactory systems are also highly sensitive to excreted glucocorticoids, sex steroids, and bile acids, and some of these molecules have been confirmed as mammalian pheromones. Steroid chemosensation in olfactory systems, unlike steroid detection in most tissues, is performed by plasma membrane receptors, but the details remain largely unclear. In this review, we present a broad view of steroid detection by vertebrate olfactory systems, focusing on recent research in fishes, amphibians, and mammals. We review confirmed and hypothesized mechanisms of steroid chemosensation in each group and discuss potential impacts on vertebrate social communication.
Collapse
|
168
|
Quach HP, Noh K, Hoi SY, Bruinsma A, Groothuis GMM, Li AP, Chow ECY, Pang KS. Alterations in gene expression in vitamin D-deficiency: Down-regulation of liver Cyp7a1 and renal Oat3 in mice. Biopharm Drug Dispos 2018; 39:99-115. [PMID: 29243851 DOI: 10.1002/bdd.2118] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/01/2017] [Accepted: 12/03/2017] [Indexed: 01/06/2023]
Abstract
The vitamin D-deficient model, established in the C57BL/6 mouse after 8 weeks of feeding vitamin D-deficient diets in the absence or presence of added calcium, was found associated with elevated levels of plasma parathyroid hormone (PTH) and plasma and liver cholesterol, and a reduction in cholesterol 7α-hydroxylase (Cyp7a1, rate-limiting enzyme for cholesterol metabolism) and renal Oat3 mRNA/protein expression levels. However, there was no change in plasma calcium and phosphate levels. Appraisal of the liver revealed an up-regulation of mRNA expressions of the small heterodimer partner (Shp) and attenuation of Cyp7a1, which contributed to hypercholesterolemia in vitamin D-deficiency. When vitamin D-sufficient or D-deficient mice were further rendered hypercholesterolemic with 3 weeks of feeding the respective, high fat/high cholesterol (HF/HC) diets, treatment with 1α,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ], active vitamin D receptor (VDR) ligand, or vitamin D (cholecalciferol) to HF/HC vitamin D-deficient mice lowered the cholesterol back to baseline levels. Cholecalciferol treatment partially restored renal Oat3 mRNA/protein expression back to that of vitamin D-sufficient mice. When the protein expression of protein kinase C (PKC), a known, negative regulator of Oat3, was examined in murine kidney, no difference in PKC expression was observed for any of the diets with/without 1,25(OH)2 D3 /cholecalciferol treatment, inferring that VDR regulation of renal Oat3 did not involve PKC in mice. As expected, plasma calcium levels were not elevated by cholecalciferol treatment of vitamin D-deficient mice, while 1,25(OH)2 D3 treatment led to hypercalcemia. In conclusion, vitamin D-deficiency resulted in down-regulation of liver Cyp7a1 and renal Oat3, conditions that are alleviated upon replenishment of cholecalciferol.
Collapse
Affiliation(s)
- Holly P Quach
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada, M5S 3M2
| | - Keumhan Noh
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada, M5S 3M2
| | - Stacie Y Hoi
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada, M5S 3M2
| | - Adrie Bruinsma
- Division of Pharmacokinetics, Toxicology and Targeting, Department of Pharmacy, University of Groningen, Groningen, The Netherlands, 9713, AV
| | - Geny M M Groothuis
- Division of Pharmacokinetics, Toxicology and Targeting, Department of Pharmacy, University of Groningen, Groningen, The Netherlands, 9713, AV
| | - Albert P Li
- In Vitro ADMET Laboratories, Columbia, Maryland, USA, 21045
| | - Edwin C Y Chow
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada, M5S 3M2
| | - K Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada, M5S 3M2
| |
Collapse
|
169
|
Petrescu AD, Kain J, Liere V, Heavener T, DeMorrow S. Hypothalamus-Pituitary-Adrenal Dysfunction in Cholestatic Liver Disease. Front Endocrinol (Lausanne) 2018; 9:660. [PMID: 30483216 PMCID: PMC6240761 DOI: 10.3389/fendo.2018.00660] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022] Open
Abstract
The Hypothalamic-Pituitary-Adrenal (HPA) axis has an important role in maintaining the physiological homeostasis in relation to external and internal stimuli. The HPA axis dysfunctions were extensively studied in neuroendocrine disorders such as depression and chronic fatigue syndrome but less so in hepatic cholestasis, cirrhosis or other liver diseases. The HPA axis controls many functions of the liver through neuroendocrine forward signaling pathways as well as negative feedback mechanisms, in health and disease. This review describes cell and molecular mechanisms of liver and HPA axis physiology and pathology. Evidence is presented from clinical and experimental model studies, demonstrating that dysfunctions of HPA axis are correlated with liver cholestatic disorders. The functional interactions of HPA axis with the liver and immune system in cases of bacterial and viral infections are also discussed. Proinflammatory cytokines stimulate glucocorticoid (GC) release by adrenals but they also inhibit bile acid (BA) efflux from liver. Chronic hepatic inflammation leads to cholestasis and impaired GC metabolism in the liver, so that HPA axis becomes depressed. Recently discovered interactions of GC with self-oscillating transcription factors that generate circadian rhythms of gene expression in brain and liver, in the context of GC replacement therapies, are also outlined.
Collapse
Affiliation(s)
- Anca D. Petrescu
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Temple, TX, United States
| | - Jessica Kain
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Temple, TX, United States
| | - Victoria Liere
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Temple, TX, United States
| | - Trace Heavener
- Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, TX, United States
| | - Sharon DeMorrow
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Temple, TX, United States
- Department of Internal Medicine, Texas A&M Health Science Center College of Medicine, Temple, TX, United States
- Department of Research Services, Central Texas Veterans Health Care System, Temple, TX, United States
- *Correspondence: Sharon DeMorrow
| |
Collapse
|
170
|
Affiliation(s)
- Mark M. Hughes
- School of Biochemistry and Immunology; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin Ireland
| | - Luke A.J. O'Neill
- School of Biochemistry and Immunology; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin Ireland
| |
Collapse
|
171
|
Di Ciaula A, Garruti G, Lunardi Baccetto R, Molina-Molina E, Bonfrate L, Wang DQH, Portincasa P. Bile Acid Physiology. Ann Hepatol 2017; 16:s4-s14. [PMID: 29080336 DOI: 10.5604/01.3001.0010.5493] [Citation(s) in RCA: 334] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/06/2017] [Indexed: 02/05/2023]
Abstract
The primary bile acids (BAs) are synthetized from colesterol in the liver, conjugated to glycine or taurine to increase their solubility, secreted into bile, concentrated in the gallbladder during fasting, and expelled in the intestine in response to dietary fat, as well as bio-transformed in the colon to the secondary BAs by the gut microbiota, reabsorbed in the ileum and colon back to the liver, and minimally lost in the feces. BAs in the intestine not only regulate the digestion and absorption of cholesterol, triglycerides, and fat-soluble vitamins, but also play a key role as signaling molecules in modulating epithelial cell proliferation, gene expression, and lipid and glucose metabolism by activating farnesoid X receptor (FXR) and G-protein-coupled bile acid receptor-1 (GPBAR-1, also known as TGR5) in the liver, intestine, muscle and brown adipose tissue. Recent studies have revealed the metabolic pathways of FXR and GPBAR-1 involved in the biosynthesis and enterohepatic circulation of BAs and their functions as signaling molecules on lipid and glucose metabolism.
Collapse
Affiliation(s)
| | - Gabriella Garruti
- Department of Emergency and Organ Transplants, Unit of Endocrinology, University of Bari Medical School, Bari, Italy
| | - Raquel Lunardi Baccetto
- Clinica Medica "A. Murri", Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, Bari, Italy
| | - Emilio Molina-Molina
- Clinica Medica "A. Murri", Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, Bari, Italy
| | - Leonilde Bonfrate
- Clinica Medica "A. Murri", Department of Biomedical Sciences & Human Oncology, University of Bari Aldo Moro Medical School, Bari, Italy
| | - David Q-H Wang
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Piero Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
172
|
Liu M, Liu C, Chen H, Huang X, Zeng X, Zhou J, Mi S. Prevention of cholesterol gallstone disease by schaftoside in lithogenic diet-induced C57BL/6 mouse model. Eur J Pharmacol 2017; 815:1-9. [PMID: 28993159 DOI: 10.1016/j.ejphar.2017.10.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/24/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022]
Abstract
Schaftoside (SS) is a bioactive compound present in the Herba Desmodii Styracifolii (DS), a herb that has been used to treat cholelithiasis and urolithiasis in Chinese medicine. Whether SS inhibits cholesterol (Ch) gallstone formation has not been investigated. This study examined the effects of oral intake of SS on Ch gallstone formation in C57BL/6 mice fed a lithogenic diet. The rate of gallstone formation was recorded. Levels of Ch, triglycerides (TG) and bile salts (BS) were measured in the bile and serum. Liver histopathology was examined microscopically, and mRNA expression levels of key genes involved in cholesterol and bile metabolism were determined by qPCR. Mice fed SS were protected against gallstone formation, had increased biliary levels of BS, and reduced biliary Ch levels, resulting in a lower Ch saturation index (CSI). In addition, mice fed SS had lower serum TG and Ch levels, increased mRNA expression of liver X receptor α, ATP binding cassette transporter 5/8 (ABCG5/8), and ileal bile acid binding protein (IBABP) in the ileum, and of farnesoid X receptor and bile salt export protein (BSEP) in the liver and ileum. SS also protected against histologically determined liver damage. Overall, these data indicate that SS protects against Ch gallstone formation in mice, and that the effect is mediated by activation of ileal liver X receptor α and hepatic farnesoid X receptor.
Collapse
Affiliation(s)
- Meijing Liu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, Guangdong Province, PR China
| | - Changhui Liu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, Guangdong Province, PR China
| | - Hao Chen
- College of food and drug, Anhui Science and Technology of University, Fengyang 233100, Anhui Province, PR China
| | - Xiaotao Huang
- Zhaoqing Medical College, Zhaoqing, Guangdong Province, PR China; Foshan University, Foshan, Guangdong Province, PR China.
| | - Xiaohui Zeng
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Province, PR China
| | - Juncheng Zhou
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Province, PR China
| | - Suiqing Mi
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, Guangdong Province, PR China
| |
Collapse
|
173
|
Rozenfeld JHK, Duarte EL, Oliveira TR, Lamy MT. Structural insights on biologically relevant cationic membranes by ESR spectroscopy. Biophys Rev 2017; 9:633-647. [PMID: 28836112 PMCID: PMC5662045 DOI: 10.1007/s12551-017-0304-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 07/28/2017] [Indexed: 12/26/2022] Open
Abstract
Cationic bilayers have been used as models to study membrane fusion, templates for polymerization and deposition of materials, carriers of nucleic acids and hydrophobic drugs, microbicidal agents and vaccine adjuvants. The versatility of these membranes depends on their structure. Electron spin resonance (ESR) spectroscopy is a powerful technique that employs hydrophobic spin labels to probe membrane structure and packing. The focus of this review is the extensive structural characterization of cationic membranes prepared with dioctadecyldimethylammonium bromide or diC14-amidine to illustrate how ESR spectroscopy can provide important structural information on bilayer thermotropic behavior, gel and fluid phases, phase coexistence, presence of bilayer interdigitation, membrane fusion and interactions with other biologically relevant molecules.
Collapse
Affiliation(s)
- Julio H K Rozenfeld
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, R. Botucatu 862, São Paulo, SP, 04023-062, Brazil
| | - Evandro L Duarte
- Instituto de Física, Universidade de São Paulo, R. do Matão 1371, São Paulo, SP, 05508-090, Brazil
| | - Tiago R Oliveira
- Centro de Engenharia, Modelagem e Ciências Sociais Aplicadas, Universidade Federal do ABC, R. Arcturus (Jd Antares), São Bernardo do Campo, SP, Brazil
| | - M Teresa Lamy
- Instituto de Física, Universidade de São Paulo, R. do Matão 1371, São Paulo, SP, 05508-090, Brazil.
| |
Collapse
|
174
|
Yan J, Xie G, Liang C, Hu Y, Zhao A, Huang F, Hu P, Liu P, Jia W, Wang X. Herbal medicine Yinchenhaotang protects against α-naphthylisothiocyanate-induced cholestasis in rats. Sci Rep 2017; 7:4211. [PMID: 28646179 PMCID: PMC5482856 DOI: 10.1038/s41598-017-04536-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/16/2017] [Indexed: 02/07/2023] Open
Abstract
Cholestasis is a clinical disorder defined as an impairment of bile flow, and that leads to toxic bile acid (BA) accumulation in hepatocytes. Here, we investigated the hepatoprotective effect of Yinchenhaotang (YCHT), a well-known formulae for the treatment of jaundice and liver disorders, against the cholestasis using the α-naphthylisothiocyanate (ANIT)-induced cholestasis in male Wistar rats. ANIT feeding induced significant cholestasis with substantially increased intrahepatic retention of hydrophobic BAs. The dynamic changes of serum and liver BAs indicated that YCHT was able to attenuate ANIT-induced BA perturbation, which is consistent with the histopathological findings that YCHT significantly decreased the liver damage. YCHT treatment substantially reduced serum alanine aminotransferase (ALT), alkaline phosphatase (AST), total bilirubin (TBIL) and direct bilirubin (DBIL) with minimal bile duct damage in the ANIT treated rats. Elevated mRNA expression of liver IL-6, IL-17A, IL-17F, TGF-β1, α-SMA, TGR5, NTCP, OATP1a1, and ileum ASBT and decreased liver IL-10, FXR, CAR, VDR, BSEP, MRP2, MRP3, MRP4 was also observed in ANIT-induced cholestasis but were attenuated or normalized by YCHT. Our results demonstrated that the BA profiles were significantly altered with ANIT intervention and YCHT possesses the hepatoprotective potential against cholestatic liver injury induced by hepatotoxin such as ANIT.
Collapse
Affiliation(s)
- Jingyu Yan
- E-institute of Shanghai Municipal Education Commission, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guoxiang Xie
- University of Hawaii Cancer Center, Honolulu, Hawaii, 96813, USA
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Chungeng Liang
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiyang Hu
- E-institute of Shanghai Municipal Education Commission, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Aihua Zhao
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Fengjie Huang
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ping Hu
- Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Ping Liu
- E-institute of Shanghai Municipal Education Commission, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Wei Jia
- University of Hawaii Cancer Center, Honolulu, Hawaii, 96813, USA.
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Xiaoning Wang
- E-institute of Shanghai Municipal Education Commission, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
175
|
Zhu R, Hou Y, Sun Y, Li T, Fan J, Chen G, Wei J. Pectin Penta-Oligogalacturonide Suppresses Intestinal Bile Acids Absorption and Downregulates the FXR-FGF15 Axis in High-Cholesterol Fed Mice. Lipids 2017; 52:489-498. [PMID: 28474246 DOI: 10.1007/s11745-017-4258-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
Abstract
Haw pectin penta-oligogalacturonide (HPPS), purified from the hydrolysates of haw pectin, has important role in decreasing hepatic cholesterol accumulation and promoting bile acids (BA) excretion in the feces of mice fed a high-cholesterol diet (HCD). However, the mechanism is not clear. This study aims to investigate the effects of HPPS on BA reabsorption in ileum and biosynthesis in liver of mice. Results showed that HPPS increased fecal BA output by approximately 110%, but decreased ileal BA and the total BA pool size by approximately 47 and 36%, respectively, compared to HCD. Studies of molecular mechanism revealed that HPPS significantly decreased the mRNA and protein levels of farnesoid X receptor (FXR) in the small intestine of mice and inactivated the fibroblast growth factor 15 (FXR-FGF15) axis, which increased the mRNA and protein levels of CYP7A1 by approximately 204 and 104%, respectively, compared to HCD. Interestingly, the mRNA and protein levels of apical sodium-dependent bile acid transporter (ASBT) in the small intestine were approximately 128 and 73% higher in HPPS-fed mice than those in HCD-fed mice, respectively. However, no significant difference was detected for ASBT expression between HCD group and BA sequestrant cholestyramine group. These findings indicate that HPPS can suppress intestinal BA reabsorption and promoting hepatic BA biosynthesis. We speculated that HPPS could be ASBT competitive inhibitor rather than BA sequestrant in inhibiting BA reabsorption in ileum and improving cholesterol metabolism.
Collapse
Affiliation(s)
- Rugang Zhu
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang, 110036, China.
| | - Yuting Hou
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang, 110036, China
| | - Yandi Sun
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang, 110036, China
| | - Tuoping Li
- College of Food Science, Shenyang Agriculture University, Shenyang, 110032, China
| | - Jungang Fan
- Forestry Biotechnology and Analysis Test Center, Liaoning Academy of Forestry Sciences, Shenyang, 110032, China
| | - Gang Chen
- Forestry Biotechnology and Analysis Test Center, Liaoning Academy of Forestry Sciences, Shenyang, 110032, China
| | - Junxiu Wei
- Electrical Engineering and Automation, College of Light Industry, Liaoning University, Shenyang, 110032, China
| |
Collapse
|
176
|
Monte MJ, Alonso-Peña M, Briz O, Herraez E, Berasain C, Argemi J, Prieto J, Marin JJG. ACOX2 deficiency: An inborn error of bile acid synthesis identified in an adolescent with persistent hypertransaminasemia. J Hepatol 2017; 66:581-588. [PMID: 27884763 DOI: 10.1016/j.jhep.2016.11.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/27/2016] [Accepted: 11/02/2016] [Indexed: 02/01/2023]
Abstract
BACKGROUND & AIMS Acyl-CoA oxidase (ACOX2) is involved in the shortening of C27 cholesterol derivatives to generate C24 bile acids. Inborn errors affecting the rest of peroxisomal enzymes involved in bile acid biosynthesis have been described. Here we aimed at investigating the case of an adolescent boy with persistent hypertransaminasemia of unknown origin and suspected dysfunction in bile acid metabolism. METHODS Serum and urine samples were taken from the patient, his sister and parents and underwent HPLC-MS/MS and HPLC-TOF analyses. Coding exons in genes of interest were amplified by high-fidelity PCR and sequenced. Wild-type or mutated (mutACOX2) variants were overexpressed in human hepatoblastoma HepG2 cells to determine ACOX2 enzymatic activity, expression and subcellular location. RESULTS The patient's serum and urine showed negligible amounts of C24 bile acids, but augmented levels of C27 intermediates, mainly tauroconjugated trihydroxycholestanoic acid (THCA). Genetic analysis of enzymes potentially involved revealed a homozygous missense mutation (c.673C>T; R225W) in ACOX2. His only sister was also homozygous for this mutation and exhibited similar alterations in bile acid profiles. Both parents were heterozygous and presented normal C24 and C27 bile acid levels. Immunofluorescence studies showed similar protein size and peroxisomal localization for both normal and mutated variants. THCA biotransformation into cholic acid was enhanced in cells overexpressing ACOX2, but not in those overexpressing mutACOX2. Both cell types showed similar sensitivity to oxidative stress caused by C24 bile acids. In contrast, THCA-induced oxidative stress and cell death were reduced by overexpressing ACOX2, but not mutACOX2. CONCLUSION ACOX2 deficiency, a condition characterized by accumulation of toxic C27 bile acid intermediates, is a novel cause of isolated persistent hypertransaminasemia. LAY SUMMARY Elevation of serum transaminases is a biochemical sign of liver damage due to multiplicity of causes (viruses, toxins, autoimmunity, metabolic disorders). In rare cases the origin of this alteration remains unknown. We have identified by the first time in a young patient and his only sister a familiar genetic defect of an enzyme called ACOX2, which participates in the transformation of cholesterol into bile acids as a cause of increased serum transaminases in the absence of any other symptomatology. This treatable condition should be considered in the diagnosis of those patients where the cause of elevated transaminases remains obscure.
Collapse
Affiliation(s)
- Maria J Monte
- Experimental Hepatology and Drug Targeting (HEVEFARM), Institute for Biomedical Research (IBSAL), University of Salamanca, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Spain
| | - Marta Alonso-Peña
- Experimental Hepatology and Drug Targeting (HEVEFARM), Institute for Biomedical Research (IBSAL), University of Salamanca, Salamanca, Spain
| | - Oscar Briz
- Experimental Hepatology and Drug Targeting (HEVEFARM), Institute for Biomedical Research (IBSAL), University of Salamanca, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Spain
| | - Elisa Herraez
- Experimental Hepatology and Drug Targeting (HEVEFARM), Institute for Biomedical Research (IBSAL), University of Salamanca, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Spain
| | - Carmen Berasain
- Department of Medicine, Clinica Universidad de Navarra and Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Spain
| | - Josepmaria Argemi
- Department of Medicine, Clinica Universidad de Navarra and Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Jesus Prieto
- Department of Medicine, Clinica Universidad de Navarra and Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Spain.
| | - Jose J G Marin
- Experimental Hepatology and Drug Targeting (HEVEFARM), Institute for Biomedical Research (IBSAL), University of Salamanca, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Spain.
| |
Collapse
|
177
|
The transcriptional activity of hepatocyte nuclear factor 4 alpha is inhibited via phosphorylation by ERK1/2. PLoS One 2017; 12:e0172020. [PMID: 28196117 PMCID: PMC5308853 DOI: 10.1371/journal.pone.0172020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/30/2017] [Indexed: 12/13/2022] Open
Abstract
Hepatocyte nuclear factor 4 alpha (HNF4α) nuclear receptor is a master regulator of hepatocyte development, nutrient transport and metabolism. HNF4α is regulated both at the transcriptional and post-transcriptional levels by different mechanisms. Several kinases (PKA, PKC, AMPK) were shown to phosphorylate and decrease the activity of HNF4α. Activation of the ERK1/2 signalling pathway, inducing proliferation and survival, inhibits the expression of HNF4α. However, based on our previous results we hypothesized that HNF4α is also regulated at the post-transcriptional level by ERK1/2. Here we show that ERK1/2 is capable of directly phosphorylating HNF4α in vitro at several phosphorylation sites including residues previously shown to be targeted by other kinases, as well. Furthermore, we also demonstrate that phosphorylation of HNF4α leads to a reduced trans-activational capacity of the nuclear receptor in luciferase reporter gene assay. We confirm the functional relevance of these findings by demonstrating with ChIP-qPCR experiments that 30-minute activation of ERK1/2 leads to reduced chromatin binding of HNF4α. Accordingly, we have observed decreasing but not disappearing binding of HNF4α to the target genes. In addition, 24-hour activation of the pathway further decreased HNF4α chromatin binding to specific loci in ChIP-qPCR experiments, which confirms the previous reports on the decreased expression of the HNF4a gene due to ERK1/2 activation. Our data suggest that the ERK1/2 pathway plays an important role in the regulation of HNF4α-dependent hepatic gene expression.
Collapse
|
178
|
Evaluation of transcriptomic signature as a valuable tool to study drug-induced cholestasis in primary human hepatocytes. Arch Toxicol 2017; 91:2879-2893. [DOI: 10.1007/s00204-017-1930-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/11/2017] [Indexed: 12/22/2022]
|
179
|
Staley C, Weingarden AR, Khoruts A, Sadowsky MJ. Interaction of gut microbiota with bile acid metabolism and its influence on disease states. Appl Microbiol Biotechnol 2017; 101:47-64. [PMID: 27888332 PMCID: PMC5203956 DOI: 10.1007/s00253-016-8006-6] [Citation(s) in RCA: 390] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/09/2016] [Accepted: 11/11/2016] [Indexed: 01/18/2023]
Abstract
Primary bile acids serve important roles in cholesterol metabolism, lipid digestion, host-microbe interactions, and regulatory pathways in the human host. While most bile acids are reabsorbed and recycled via enterohepatic cycling, ∼5% serve as substrates for bacterial biotransformation in the colon. Enzymes involved in various transformations have been characterized from cultured gut bacteria and reveal taxa-specific distribution. More recently, bioinformatic approaches have revealed greater diversity in isoforms of these enzymes, and the microbial species in which they are found. Thus, the functional roles played by the bile acid-transforming gut microbiota and the distribution of resulting secondary bile acids, in the bile acid pool, may be profoundly affected by microbial community structure and function. Bile acids and the composition of the bile acid pool have historically been hypothesized to be associated with several disease states, including recurrent Clostridium difficile infection, inflammatory bowel diseases, metabolic syndrome, and several cancers. Recently, however, emphasis has been placed on how microbial communities in the dysbiotic gut may alter the bile acid pool to potentially cause or mitigate disease onset. This review highlights the current understanding of the interactions between the gut microbial community, bile acid biotransformation, and disease states, and addresses future directions to better understand these complex associations.
Collapse
Affiliation(s)
- Christopher Staley
- BioTechnology Institute, Center for Immunology University of Minnesota, Minneapolis, MN
| | - Alexa R Weingarden
- BioTechnology Institute, Center for Immunology University of Minnesota, Minneapolis, MN
| | - Alexander Khoruts
- BioTechnology Institute, Center for Immunology University of Minnesota, Minneapolis, MN
- Division of Gastroenterology, Department of Medicine, Center for Immunology University of Minnesota, Minneapolis, MN
| | - Michael J Sadowsky
- BioTechnology Institute, Center for Immunology University of Minnesota, Minneapolis, MN
- Department of Soil, Water and Climate, University of Minnesota, St. Paul, MN
| |
Collapse
|
180
|
JUŘICA J, DOVRTĚLOVÁ G, NOSKOVÁ K, ZENDULKA O. Bile Acids, Nuclear Receptors and Cytochrome P450. Physiol Res 2016; 65:S427-S440. [DOI: 10.33549/physiolres.933512] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This review summarizes the importance of bile acids (BA) as important regulators of various homeostatic mechanisms with detailed focus on cytochrome P450 (CYP) enzymes. In the first part, synthesis, metabolism and circulation of BA is summarized and BA are reviewed as physiological ligands of nuclear receptors which regulate transcription of genes involved in their metabolism, transport and excretion. Notably, PXR, FXR and VDR are the most important nuclear receptors through which BA regulate transcription of CYP genes involved in the metabolism of both BA and xenobiotics. Therapeutic use of BA and their derivatives is also briefly reviewed. The physiological role of BA interaction with nuclear receptors is basically to decrease production of toxic non-polar BA and increase their metabolic turnover towards polar BA and thus decrease their toxicity. By this, the activity of some drug-metabolizing CYPs is also influenced what could have clinically relevant consequences in cholestatic diseases or during the treatment with BA or their derivatives.
Collapse
Affiliation(s)
| | | | | | - O. ZENDULKA
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno. Czech Republic
| |
Collapse
|
181
|
|
182
|
Henkel AS, LeCuyer B, Olivares S, Green RM. Endoplasmic Reticulum Stress Regulates Hepatic Bile Acid Metabolism in Mice. Cell Mol Gastroenterol Hepatol 2016; 3:261-271. [PMID: 28275692 PMCID: PMC5331781 DOI: 10.1016/j.jcmgh.2016.11.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/01/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS Cholestasis promotes endoplasmic reticulum (ER) stress in the liver, however, the effect of ER stress on hepatic bile acid metabolism is unknown. We aim to determine the effect of ER stress on hepatic bile acid synthesis and transport in mice. METHODS ER stress was induced pharmacologically in C57BL/6J mice and human hepatoma (HepG2) cells. The hepatic expression of genes controlling bile acid synthesis and transport was determined. To measure the activity of the primary bile acid synthetic pathway, the concentration of 7α-hydroxy-4-cholesten-3-1 was measured in plasma. RESULTS Induction of ER stress in mice and HepG2 cells rapidly suppressed the hepatic expression of the primary bile acid synthetic enzyme, cholesterol 7α-hydroxylase. Plasma levels of 7α-hydroxy-4-cholesten-3-1 were reduced in mice subjected to ER stress, indicating impaired bile acid synthesis. Induction of ER stress in mice and HepG2 cells increased expression of the bile salt export pump (adenosine triphosphate binding cassette [Abc]b11) and a bile salt efflux pump (Abcc3). The observed regulation of Cyp7a1, Abcb11, and Abcc3 occurred in the absence of hepatic inflammatory cytokine activation and was not dependent on activation of hepatic small heterodimer partner or intestinal fibroblast growth factor 15. Consistent with suppressed bile acid synthesis and enhanced bile acid export from hepatocytes, prolonged ER stress decreased the hepatic bile acid content in mice. CONCLUSIONS Induction of ER stress in mice suppresses bile acid synthesis and enhances bile acid removal from hepatocytes independently of established bile acid regulatory pathways. These data show a novel function of the ER stress response in regulating bile acid metabolism.
Collapse
Key Words
- 7α-Hydroxy-4-Cholesten-3-1
- ABC, adenosine triphosphate binding cassette
- Bile Acid Synthesis
- C4, 7α-hydroxy-4-cholesten-3-1
- CYP7A1, cholesterol 7α-hydroxylase
- Cyp7a1
- DMEM, Dulbecco's modified Eagle medium
- DMSO, dimethyl sulfoxide
- ER, endoplasmic reticulum
- ERK, extracellular signaling-regulated kinase
- FGF, fibroblast growth factor
- FXR, farnesoid X receptor
- IL, interleukin
- IRE1α, inositol requiring enzyme 1α
- JNK, c-Jun-N-terminal kinase
- NTCP, sodium/taurocholate cotransporter
- RIDD, regulated inositol requiring enzyme 1α–dependent messenger RNA decay
- SHP, small heterodimer partner
- UPR, unfolded protein response
- Unfolded Protein Response
- mRNA, messenger RNA
Collapse
Affiliation(s)
- Anne S. Henkel
- Correspondence Address correspondence to: Anne S. Henkel, MD, 320 East Superior Street, Tarry 15-705, Chicago, Illinois 60611. fax: (312) 908-9032.320 East Superior StreetTarry 15-705ChicagoIllinois 60611
| | | | | | | |
Collapse
|
183
|
Wang Y, Han Y, Chai F, Xiang H, Huang T, Kou S, Han B, Gong X, Ye X. The antihypercholesterolemic effect of columbamine from Rhizoma Coptidis in HFHC-diet induced hamsters through HNF-4α/FTF-mediated CYP7A1 activation. Fitoterapia 2016; 115:111-121. [DOI: 10.1016/j.fitote.2016.09.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/25/2016] [Accepted: 09/30/2016] [Indexed: 01/04/2023]
|
184
|
Chen Q, Yang X, Zhang H, Kong X, Yao L, Cui X, Zou Y, Fang F, Yang J, Chang Y. Metformin impairs systemic bile acid homeostasis through regulating SIRT1 protein levels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:101-112. [PMID: 27816442 DOI: 10.1016/j.bbamcr.2016.10.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 10/24/2016] [Accepted: 10/30/2016] [Indexed: 01/04/2023]
Abstract
Metformin is widely used to treat hyperglycemia. However, metformin treatment may induce intrahepatic cholestasis and liver injury in a few patients with type II diabetes through an unknown mechanism. Here we show that metformin decreases SIRT1 protein levels in primary hepatocytes and liver. Both metformin-treated wild-type C57 mice and hepatic SIRT1-mutant mice had increased hepatic and serum bile acid levels. However, metformin failed to change systemic bile acid levels in hepatic SIRT1-mutant mice. Molecular mechanism study indicates that SIRT1 directly interacts with and deacetylates Foxa2 to inhibit its transcriptional activity on expression of genes involved in bile acids synthesis and transport. Hepatic SIRT1 mutation elevates Foxa2 acetylation levels, which promotes Foxa2 binding to and activating genes involved in bile acids metabolism, impairing hepatic and systemic bile acid homeostasis. Our data clearly suggest that hepatic SIRT1 mediates metformin effects on systemic bile acid metabolism and modulation of SIRT1 activity in liver may be an attractive approach for treatment of bile acid-related diseases such as cholestasis.
Collapse
Affiliation(s)
- Qi Chen
- 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; Hangzhou Center for Disease Control and Prevention, Zhejiang, People's Republic of China
| | - Xiaoying Yang
- 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
| | - Huabing Zhang
- 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
| | - Xingxing Kong
- 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
| | - Lu Yao
- 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
| | - Xiaona Cui
- 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
| | - Yongkang Zou
- 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
| | - Fude Fang
- 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
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Yongsheng Chang
- 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
|
185
|
Kulig W, Cwiklik L, Jurkiewicz P, Rog T, Vattulainen I. Cholesterol oxidation products and their biological importance. Chem Phys Lipids 2016; 199:144-160. [DOI: 10.1016/j.chemphyslip.2016.03.001] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 12/14/2022]
|
186
|
Do HJ, Lee YS, Ha MJ, Cho Y, Yi H, Hwang YJ, Hwang GS, Shin MJ. Beneficial effects of voglibose administration on body weight and lipid metabolism via gastrointestinal bile acid modification. Endocr J 2016; 63:691-702. [PMID: 27349182 DOI: 10.1507/endocrj.ej15-0747] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This study was designed with the goal of examining the effects of voglibose administration on body weight and lipid metabolism and underlying mechanism high fat diet-induced obese mice. Male C57BL/6 mice were randomly assigned to one of four groups: a control diet (CTL), high-fat diet (HF), high-fat diet supplemented with voglibose (VO), and high fat diet pair-fed group (PF). After 12 weeks, the following characteristics were investigated: serum lipid and glucose levels, serum polar metabolite profiles, and expression levels of genes involved in lipid and bile acid metabolism. In addition, pyrosequencing was used to analyze the composition of gut microbiota found in feces. Total body weight gain was significantly lower in the VO group than in the CTL, HF, and PF groups. The VO group exhibited improved metabolic profiles including those of blood glucose, triglyceride, and total cholesterol levels. The 12-week voglibose administration decreased the ratio of Firmicutes to Bacteroidetes found in feces. Circulating levels of taurocholic and cholic acid were significantly higher in the VO group than in the HF and CTL groups. Deoxycholic acid levels tended to be higher in the VO group than in the HF group. Voglibose administration downregulated expression levels of CYP8B1 and HNF4α genes and upregulated those of PGC1α, whereas FXRα was not affected. Voglibose administration elicits changes in the composition of the intestinal microbiota and circulating metabolites, which ultimately has systemic effects on body weight and lipid metabolism in mice.
Collapse
Affiliation(s)
- Hyun Ju Do
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 136-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Yun C, Yin T, Shatzer K, Burrin DG, Cui L, Tu Y, Hu M. Determination of 7α-OH cholesterol by LC-MS/MS: Application in assessing the activity of CYP7A1 in cholestatic minipigs. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1025:76-82. [PMID: 27218859 PMCID: PMC5358015 DOI: 10.1016/j.jchromb.2016.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 05/01/2016] [Accepted: 05/03/2016] [Indexed: 01/29/2023]
Abstract
An LC-MS/MS method was developed and validated to determine 7α-OH cholesterol in liver microsome. This method was convenient and fast with high specificity and sensitivity. Briefly, a gradient elution was performed on a Synergi polar-C18 column (50×4.6mm i.d., 3μm). The mobile phase (consisting of 0.1% HCOOH solution and acetonitrile) eluted in gradient at a flow rate of 1ml/min. MS detection was operated on APCI (+) mode; the MRM transitions for 7α-OH cholesterol and D7-cholesterol (I.S.) were 385.1≥159.1 and 376.4≥266.3, respectively. The linear response range of 7α-OH cholesterol was covered from 1.563 to 100.0ng/ml. All of the validation items meet the requirement of FDA guidance for bioanalytical method validation. This method was applied to enzymatic studies for determination of cholesterol 7alpha-hydroxylation activity catalyzed by CYP7A1 in the cholestatic minipigs liver microsomes.
Collapse
Affiliation(s)
- Changhong Yun
- Department of Pharmaceutics, University of Houston, 1441 Moursund St. Houston, TX 77030, United States
| | - Taijun Yin
- Department of Pharmaceutics, University of Houston, 1441 Moursund St. Houston, TX 77030, United States
| | - Katherine Shatzer
- Department of Pharmaceutics, University of Houston, 1441 Moursund St. Houston, TX 77030, United States
| | - Douglas G Burrin
- USDA Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates St. Houston, TX 77030, United States
| | - Liwei Cui
- USDA Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates St. Houston, TX 77030, United States
| | - Yifan Tu
- Department of Pharmaceutics, University of Houston, 1441 Moursund St. Houston, TX 77030, United States
| | - Ming Hu
- Department of Pharmaceutics, University of Houston, 1441 Moursund St. Houston, TX 77030, United States.
| |
Collapse
|
188
|
Mörk LM, Strom SC, Mode A, Ellis EC. Addition of Dexamethasone Alters the Bile Acid Composition by Inducing CYP8B1 in Primary Cultures of Human Hepatocytes. J Clin Exp Hepatol 2016; 6:87-93. [PMID: 27493455 PMCID: PMC4963319 DOI: 10.1016/j.jceh.2016.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/22/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Primary human hepatocytes offer the best human in vitro model for studies on human liver cell metabolism. Investigators use a variety of different media supplements and matrix biocoatings and the type of culture system used may influence the outcome. OBJECTIVES To optimize in vitro conditions for primary human hepatocytes with regard to bile acid synthesis. METHODS Human hepatocytes were isolated and cultured on collagen type I or EHS matrigel in cell media with or without dexamethasone. The glucocorticoid receptor (GR) antagonist RU486 was used to elucidate the involvement of GR. RESULTS Hepatocytes cultured on EHS matrigel produced more bile acids and expressed higher levels of cholesterol 7α-hydroxylase (CYP7A1) than cells cultured on rat tail collagen. Supplementation with dexamethasone increased the formation of cholic acid (CA) and decreased chenodeoxycholic acid formation. In line with these results, the mRNA expression of sterol 12α-hydroxylase (CYP8B1) increased following dexamethasone treatment. Surprisingly, the mRNA expression of CYP7A1 and CYP27A1 was not increased to the same extent. By using the GR antagonist RU486, we concluded that CYP8B1 induction is mediated via a GR-independent pathway. An altered expression of retinoid-related orphan receptor (ROR) α and ROR α target gene Glucose-6-phosphatase (G6Pase) suggests that ROR α signaling may regulate CYP8B1 expression. CONCLUSION Primary human hepatocytes have an increased bile acid synthesis rate when cultured on matrigel as compared to collagen. Exposure to glucocorticoid hormones stimulates the expression of CYP8B1, leading to an increased formation of CA and alteration of the bile acid composition. The effect is most likely mediated through a GR-independent pathway, possibly through ROR α.
Collapse
Key Words
- BSEP, bile salt export pump
- CA, cholic acid
- CDCA, chenodeoxycholic acid
- CYP27A1, sterol 27α-hydroxylase
- CYP7A1, cholesterol 7α-hydroxylase
- CYP8B1, sterol 12α-hydroxylase
- FXR, farnesoid X receptor
- G6Pase, glucose-6-phosphatase
- GR, glucocorticoid receptor
- NTCP, Na+-taurocholate cotransporting polypeptide
- PXR, pregnane X receptor
- ROR, retinoid-related orphan receptor
- chenodeoxycholic acid
- cholic acid
- dexamethasone
- matrigel
- primary hepatocytes
Collapse
Affiliation(s)
- Lisa-Mari Mörk
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden,Address for correspondence: Lisa-Mari Mörk, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, F82, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden. Tel.: +46 8 585 83062; fax: +46 8 585 82912.
| | - Stephen C. Strom
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, Sweden
| | - Agneta Mode
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Ewa C.S. Ellis
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
189
|
Sorg UR, Behnke K, Degrandi D, Reich M, Keitel V, Herebian D, Deenen R, Beyer M, Schultze JL, Köhrer K, Gabbert HE, Mayatepek E, Häussinger D, Pfeffer K. Cooperative role of lymphotoxin β receptor and tumor necrosis factor receptor p55 in murine liver regeneration. J Hepatol 2016; 64:1108-1117. [PMID: 26708145 DOI: 10.1016/j.jhep.2015.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 11/24/2015] [Accepted: 12/08/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS The liver exhibits a unique capacity for regeneration in response to injury. Lymphotoxin-β receptor (LTβR), a core member of the tumor necrosis factor (TNF)/tumor necrosis factor receptor (TNFR) superfamily is known to play an important role in this process. However, the function of LTβR during pathophysiological alterations and its molecular mechanisms during liver regeneration are so far ill-characterized. METHODS LTβR(-/-) mice were subjected to 70% hepatectomy and liver regeneration capacity, bile acid profiles, and transcriptome analysis were performed. RESULTS LTβR(-/-) deficient mice suffered from increased and prolonged liver tissue damage after 70% hepatectomy, accompanied by deregulated bile acid homeostasis. Pronounced differences in the expression patterns of genes relevant for bile acid synthesis and recirculation were observed. LTβR and TNFRp55 share downstream signalling elements. Therefore, LTβR(-/-) mice were treated with etanercept to create mice functionally deficient in both signalling pathways. Strikingly, the combined blockade of TNFRp55 and LTβR signalling leads to complete failure of liver regeneration resulting in death within 24 to 48h after PHx. Transcriptome analysis revealed a marked disparity in gene expression programs in livers of LTβR(-/-) and etanercept-treated LTβR(-/-) vs. wild-type animals after PHx. Murinoglobulin 2 was identified as a significantly differentially regulated gene. CONCLUSIONS LTβR is essential for efficient liver regeneration and cooperates with TNFRp55 in this process. Differences in survival kinetics strongly suggest distinct functions for these two cytokine receptors in liver regeneration. Failure of TNFR and LTβR signalling renders liver regeneration impossible.
Collapse
Affiliation(s)
- Ursula R Sorg
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Kristina Behnke
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany; Molecular Medicine II, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Daniel Degrandi
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Maria Reich
- Department of Internal Medicine, Gastroenterology, Hepatology and Infectious Diseases, University Children's Hospital, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Verena Keitel
- Department of Internal Medicine, Gastroenterology, Hepatology and Infectious Diseases, University Children's Hospital, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - René Deenen
- Center for Biological and Medical Research, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Marc Beyer
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Karl Köhrer
- Center for Biological and Medical Research, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Helmut E Gabbert
- Institute of Pathology, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Dieter Häussinger
- Department of Internal Medicine, Gastroenterology, Hepatology and Infectious Diseases, University Children's Hospital, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany.
| |
Collapse
|
190
|
Cheng Y, El-Kattan A, Zhang Y, Ray AS, Lai Y. Involvement of Drug Transporters in Organ Toxicity: The Fundamental Basis of Drug Discovery and Development. Chem Res Toxicol 2016; 29:545-63. [DOI: 10.1021/acs.chemrestox.5b00511] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yaofeng Cheng
- Pharmaceutical
Candidate Optimization, Bristol-Myers Squibb Company, 3551 Lawrenceville
Road, Princeton, New Jersey 08540, United States
| | - Ayman El-Kattan
- Department
of Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., 610 Main
Street, Cambridge, Massachusetts 02139, United States
| | - Yan Zhang
- Drug
Metabolism and Biopharmaceutics, Incyte Corporation, 1801 Augustine
Cutoff, Wilmington, Delaware 19803, United States
| | - Adrian S. Ray
- Department
of Drug Metabolism, Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Yurong Lai
- Pharmaceutical
Candidate Optimization, Bristol-Myers Squibb Company, 3551 Lawrenceville
Road, Princeton, New Jersey 08540, United States
| |
Collapse
|
191
|
Nie YF, Hu J, Yan XH. Cross-talk between bile acids and intestinal microbiota in host metabolism and health. J Zhejiang Univ Sci B 2016; 16:436-46. [PMID: 26055905 DOI: 10.1631/jzus.b1400327] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bile acid (BA) is de novo synthesized exclusively in the liver and has direct or indirect antimicrobial effects. On the other hand, the composition and size of the BA pool can be altered by intestinal microbiota via the biotransformation of primary BAs to secondary BAs, and subsequently regulate the nuclear farnesoid X receptor (FXR; NR1H4). The BA-activated FXR plays important roles in BA synthesis and metabolism, glucose and lipid metabolism, and even hepatic autophagy. BAs can also play a role in the interplays among intestinal microbes. In this review, we mainly discuss the interactions between BAs and intestinal microbiota and their roles in regulating host metabolism, and probably the autophagic signaling pathway.
Collapse
Affiliation(s)
- Yang-fan Nie
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | | | | |
Collapse
|
192
|
Cheese intake lowers plasma cholesterol concentrations without increasing bile acid excretion. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2016. [DOI: 10.1016/j.jnim.2015.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
193
|
Synergetic cholesterol-lowering effects of main alkaloids from Rhizoma Coptidis in HepG2 cells and hypercholesterolemia hamsters. Life Sci 2016; 151:50-60. [PMID: 26876917 DOI: 10.1016/j.lfs.2016.02.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 02/02/2016] [Accepted: 02/10/2016] [Indexed: 01/23/2023]
Abstract
AIMS Hyperlipidemia contributes to the progression of cardiovascular diseases. Main alkaloids from Rhizoma Coptidis including berberine (BBR), coptisine (COP), palmatine (PAL), epiberberine (EPI) and jatrorrhizine (JAT), improved dyslipidemia in hypercholesterolemic hamsters to a different degree. In this study, HepG2 cells and hypercholesterolemic hamsters were used to investigate the synergetic cholesterol-lowering efficacy of these five main alkaloids. MAIN METHODS The cellular lipid and cholesterol accumulation and in HepG2 cells were evaluated by Oil Red O staining and HPLC analysis. LDL receptor, 3-Hydroxy-3-methylglutaryl CoA reductase (HMGCR) and cholesterol 7-alpha-hydroxylase (CYP7A1) that involving cholesterol metabolism in HepG2 cells were measured by qRT-PCR, western blot and immunofluorescence analysis. The serum profiles including total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-c) and high-density lipoprotein cholesterol (HDL-c), as well as TC and total bile acids (TBA) of feces in hypercholesterolemic hamsters were also measured. KEY FINDING As compared to single alkaloids, the combination of five main alkaloids (COM) reduced the lipid and cholesterol accumulation in HepG2 cells more effectively and performed an advantageous effect on controlling TC, TG, LDL-c and HDL-c in hypercholesterolemic hamsters. More effective reduction of TBA and TC levels in feces of hamsters were achieved after the administration of COM. These effects were derived from the up-regulation of LDL receptor and CYP7A1, as well as HMGCR downregulation. SIGNIFICANCE Our results demonstrated that COM showed a synergetic cholesterol-lowering efficacy, which was better than single alkaloids and it might be considered as a potential therapy for hypercholesterolemia.
Collapse
|
194
|
Interdependence of nutrient metabolism and the circadian clock system: Importance for metabolic health. Mol Metab 2016; 5:133-152. [PMID: 26977390 PMCID: PMC4770266 DOI: 10.1016/j.molmet.2015.12.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 12/15/2015] [Accepted: 12/29/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND While additional research is needed, a number of large epidemiological studies show an association between circadian disruption and metabolic disorders. Specifically, obesity, insulin resistance, cardiovascular disease, and other signs of metabolic syndrome all have been linked to circadian disruption in humans. Studies in other species support this association and generally reveal that feeding that is not in phase with the external light/dark cycle, as often occurs with night or rotating shift workers, is disadvantageous in terms of energy balance. As food is a strong driver of circadian rhythms in the periphery, understanding how nutrient metabolism drives clocks across the body is important for dissecting out why circadian misalignment may produce such metabolic effects. A number of circadian clock proteins as well as their accessory proteins (such as nuclear receptors) are highly sensitive to nutrient metabolism. Macronutrients and micronutrients can function as zeitgebers for the clock in a tissue-specific way and can thus impair synchrony between clocks across the body, or potentially restore synchrony in the case of circadian misalignment. Circadian nuclear receptors are particularly sensitive to nutrient metabolism and can alter tissue-specific rhythms in response to changes in the diet. Finally, SNPs in human clock genes appear to be correlated with diet-specific responses and along with chronotype eventually may provide valuable information from a clinical perspective on how to use diet and nutrition to treat metabolic disorders. SCOPE OF REVIEW This article presents a background of the circadian clock components and their interrelated metabolic and transcriptional feedback loops, followed by a review of some recent studies in humans and rodents that address the effects of nutrient metabolism on the circadian clock and vice versa. We focus on studies in which results suggest that nutrients provide an opportunity to restore or, alternatively, can destroy synchrony between peripheral clocks and the central pacemaker in the brain as well as between peripheral clocks themselves. In addition, we review several studies looking at clock gene SNPs in humans and the metabolic phenotypes or tendencies associated with particular clock gene mutations. MAJOR CONCLUSIONS Targeted use of specific nutrients based on chronotype has the potential for immense clinical utility in the future. Macronutrients and micronutrients have the ability to function as zeitgebers for the clock by activating or modulating specific clock proteins or accessory proteins (such as nuclear receptors). Circadian clock control by nutrients can be tissue-specific. With a better understanding of the mechanisms that support nutrient-induced circadian control in specific tissues, human chronotype and SNP information might eventually be used to tailor nutritional regimens for metabolic disease treatment and thus be an important part of personalized medicine's future.
Collapse
|
195
|
Liu G, Wu X, Jia G, Zhao H, Chen X, Wu C, Wang J. Effects of glutamine against oxidative stress in the metabolome of rats—new insight. RSC Adv 2016. [DOI: 10.1039/c6ra14469a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glutamine exerts potential functions against the harmful effects of oxidative stress on animals.
Collapse
Affiliation(s)
- Guangmang Liu
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - XianJian Wu
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Gang Jia
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Hua Zhao
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Xiaoling Chen
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Caimei Wu
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- China
- Key Laboratory for Animal Disease-Resistance Nutrition of China
| | - Jing Wang
- Maize Research Institute
- Sichuan Agricultural University
- Chengdu 611130
- China
| |
Collapse
|
196
|
Transcriptional profiling suggests that Nevirapine and Ritonavir cause drug induced liver injury through distinct mechanisms in primary human hepatocytes. Chem Biol Interact 2015; 255:31-44. [PMID: 26626330 DOI: 10.1016/j.cbi.2015.11.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/28/2015] [Accepted: 11/20/2015] [Indexed: 12/25/2022]
Abstract
Drug induced liver injury (DILI), a major cause of pre- and post-approval failure, is challenging to predict pre-clinically due to varied underlying direct and indirect mechanisms. Nevirapine, a non-nucleoside reverse transcriptase inhibitor (NNRTI) and Ritonavir, a protease inhibitor, are antiviral drugs that cause clinical DILI with different phenotypes via different mechanisms. Assessing DILI in vitro in hepatocyte cultures typically requires drug exposures significantly higher than clinical plasma Cmax concentrations, making clinical interpretations of mechanistic pathway changes challenging. We previously described a system that uses liver-derived hemodynamic blood flow and transport parameters to restore primary human hepatocyte biology, and drug responses at concentrations relevant to in vivo or clinical exposure levels. Using this system, primary hepatocytes from 5 human donors were exposed to concentrations approximating clinical therapeutic and supra-therapeutic levels of Nevirapine (11.3 and 175.0 μM) and Ritonavir (3.5 and 62.4 μM) for 48 h. Whole genome transcriptomics was performed by RNAseq along with functional assays for metabolic activity and function. We observed effects at both doses, but a greater number of genes were differentially expressed with higher probability at the toxic concentrations. At the toxic doses, both drugs showed direct cholestatic potential with Nevirapine increasing bile synthesis and Ritonavir inhibiting bile acid transport. Clear differences in antigen presentation were noted, with marked activation of MHC Class I by Nevirapine and suppression by Ritonavir. This suggests CD8+ T cell involvement for Nevirapine and possibly NK Killer cells for Ritonavir. Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Unlike Ritonavir, Nevirapine did not increase fatty acid synthesis or activate the respiratory electron chain with simultaneous mitochondrial uncoupling supporting clinical reports of a lower propensity for steatosis. This in vitro study offers insights into the disparate direct and immune-mediated toxicity mechanisms underlying Nevirapine and Ritonavir toxicity in the clinic.
Collapse
|
197
|
Alphonse PAS, Jones PJH. Revisiting Human Cholesterol Synthesis and Absorption: The Reciprocity Paradigm and its Key Regulators. Lipids 2015. [PMID: 26620375 DOI: 10.1007/s11745‐015‐4096‐7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hypercholesterolemia is a major risk factor for cardiovascular disease. Cholesterol homeostasis in the body is governed by the interplay between absorption, synthesis, and excretion or conversion of cholesterol into bile acids. A reciprocal relationship between cholesterol synthesis and absorption is known to regulate circulating cholesterol in response to dietary or therapeutic interventions. However, the degree to which these factors affect synthesis and absorption and the extent to which one vector shifts in response to the other are not thoroughly understood. Also, huge inter-individual variability exists in the manner in which the two systems act in response to any cholesterol-lowering treatment. Various factors are known to account for this variability and in light of recent experimental advances new players such as gene-gene interactions, gene-environmental effects, and gut microbiome hold immense potential in offering an explanation to the complex traits of inter-individual variability in human cholesterol metabolism. In this context, the objective of the present review is to provide an overview on cholesterol metabolism and discuss the role of potential factors such as genetics, epigenetics, epistasis, and gut microbiome, as well as other regulators in modulating cholesterol metabolism, especially emphasizing the reciprocal relationship between cholesterol synthesis and absorption. Furthermore, an evaluation of the implications of this push-pull mechanism on cholesterol-lowering strategies is presented.
Collapse
Affiliation(s)
- Peter A S Alphonse
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.
| | - Peter J H Jones
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada
- Food Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
198
|
Escalona A, Muñoz R, Irribarra V, Solari S, Allende F, Francisco Miquel J. Bile acids synthesis decreases after laparoscopic sleeve gastrectomy. Surg Obes Relat Dis 2015; 12:763-769. [PMID: 26948941 DOI: 10.1016/j.soard.2015.11.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Bariatric surgery is the most effective treatment alternative in morbid obesity. The mechanisms contributing to these benefits remain poorly understood. Bile acids (BAs) are mediators of different regulatory functions in glucose and cholesterol homeostasis and energy expenditure. Recent evidence suggests that BAs are critically important for the beneficial effects of sleeve gastrectomy (SG). OBJECTIVES The aim of this study was to evaluate the effect of SG on BA synthesis. SETTING University Hospital. Santiago, Chile. METHODS Obese patients were evaluated before and after SG (1, 3, 6, and 12 months). BA synthesis was evaluated through the serum marker, 7 α-hydroxy-4-cholesten-3-one (C4). Primary and secondary BA and C4 were determined by high performance liquid chromatography coupled with tandem mass spectrometry detection (HPLC-MS/MS). RESULTS From June 2013 to January 2014, 19 patients (age 37.6±7.8 years; BMI 35.8±3.5 kg/m(2); 79% female) were included in this study. Mean weight loss at 1, 3, 6, and 12 months was 11.3, 17.5, 23.6, and 25.4 kg, respectively, equivalent to 11.8, 18.6, 24.8, and 26.9 of total body water percentage (%TBW) (P<.0001), respectively and 43.2, 68.2, 91, and 98.8 of percentage of excess weight loss (%EWL), respectively (P<.001). Serum C4 levels at baseline, 1, 3, 6, and 12 months were 23.4±21.1, 4.9±8.2, 8.7±12.1, 13.8±12.9, and 18.8±16.8 ng/mL (P<.0001), respectively. Fibroblast growth factor 19 (FGF19) levels at baseline, 1, 3, 6, and 12 months were 71±33.3, 130.5±66.2, 117.8±57.2, 134.6±91.7, and 124.3±85.9 pg/mL (P = .019), respectively. CONCLUSION Serum levels of C4 decrease after SG, indicating a reduction in the synthesis of BA. FGF19 may play a role in decreasing BA synthesis. Further studies are necessary to characterize the effect of bariatric surgery on BA homeostasis.
Collapse
Affiliation(s)
- Alex Escalona
- Department of Surgery. Faculty of Medicine, Universidad de los Andes, Santiago, Chile.
| | - Rodrigo Muñoz
- Department of Digestive Surgery, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Veronica Irribarra
- Department of Nutrition, Diabetes and Metabolism, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sandra Solari
- Department of Clinical Laboratories, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fidel Allende
- Department of Clinical Laboratories, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Francisco Miquel
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
199
|
Cole AJ, Teigen LM, Jahansouz C, Earthman CP, Sibley SD. The Influence of Bariatric Surgery on Serum Bile Acids in Humans and Potential Metabolic and Hormonal Implications: a Systematic Review. Curr Obes Rep 2015; 4:441-50. [PMID: 26335653 DOI: 10.1007/s13679-015-0171-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent research suggests a mechanistic role for bile acids (BA) in the metabolic improvement following bariatric surgery. It is believed that the hormonal and metabolic effects associated with changes in systemic BAs may be related to the farnesoid X receptor (FXR) and a G-protein coupled receptor (TGR5). This systematic review examines changes in systemic BAs following bariatric procedures. Studies were included if they reported the measurement of systemic BAs in humans at at least one time point after bariatric surgery. Eleven papers were identified that met the inclusion criteria. Seven studies reported the effect of Roux-en-Y gastric bypass (RYGB) on fasting BAs. The majority (6/7) reported that fasting BAs increased after RYGB. Data regarding fasting BAs after vertical sleeve gastrectomy (VSG) and laparoscopic gastric banding (LAGB) are inconsistent. Data regarding post-prandial BA changes after RYGB, VSG, and LAGB are also inconsistent. More research is needed to investigate the connection between BAs and the metabolic improvement seen after bariatric surgery.
Collapse
Affiliation(s)
- Abigail J Cole
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, 1334 Eckles Avenue, Saint Paul, MN, 55108, USA.
| | - Levi M Teigen
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, 1334 Eckles Avenue, Saint Paul, MN, 55108, USA.
| | - Cyrus Jahansouz
- Medical School, Department of Surgery, University of Minnesota-Twin Cities, 420 Delaware Street SE, Minneapolis, MN, 55404, USA.
| | - Carrie P Earthman
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, 1334 Eckles Avenue, Saint Paul, MN, 55108, USA.
| | - Shalamar D Sibley
- Department of Medicine, Division of Endocrinology, University of Minnesota-Twin Cities, 516 Delaware St. SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
200
|
Alphonse PAS, Jones PJH. Revisiting Human Cholesterol Synthesis and Absorption: The Reciprocity Paradigm and its Key Regulators. Lipids 2015; 51:519-36. [PMID: 26620375 DOI: 10.1007/s11745-015-4096-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/09/2015] [Indexed: 12/22/2022]
Abstract
Hypercholesterolemia is a major risk factor for cardiovascular disease. Cholesterol homeostasis in the body is governed by the interplay between absorption, synthesis, and excretion or conversion of cholesterol into bile acids. A reciprocal relationship between cholesterol synthesis and absorption is known to regulate circulating cholesterol in response to dietary or therapeutic interventions. However, the degree to which these factors affect synthesis and absorption and the extent to which one vector shifts in response to the other are not thoroughly understood. Also, huge inter-individual variability exists in the manner in which the two systems act in response to any cholesterol-lowering treatment. Various factors are known to account for this variability and in light of recent experimental advances new players such as gene-gene interactions, gene-environmental effects, and gut microbiome hold immense potential in offering an explanation to the complex traits of inter-individual variability in human cholesterol metabolism. In this context, the objective of the present review is to provide an overview on cholesterol metabolism and discuss the role of potential factors such as genetics, epigenetics, epistasis, and gut microbiome, as well as other regulators in modulating cholesterol metabolism, especially emphasizing the reciprocal relationship between cholesterol synthesis and absorption. Furthermore, an evaluation of the implications of this push-pull mechanism on cholesterol-lowering strategies is presented.
Collapse
Affiliation(s)
- Peter A S Alphonse
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada. .,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.
| | - Peter J H Jones
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.,Food Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|