51
|
Zaborska KE, Lee SA, Garribay D, Cha E, Cummings BP. Deoxycholic acid supplementation impairs glucose homeostasis in mice. PLoS One 2018; 13:e0200908. [PMID: 30059528 PMCID: PMC6066200 DOI: 10.1371/journal.pone.0200908] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/22/2018] [Indexed: 12/14/2022] Open
Abstract
Bile acids are critical contributors to the regulation of whole body glucose homeostasis; however, the mechanisms remain incompletely defined. While the hydrophilic bile acid subtype, ursodeoxycholic acid, has been shown to attenuate hepatic endoplasmic reticulum (ER) stress and thereby improve glucose regulation in mice, the effect of hydrophobic bile acid subtypes on ER stress and glucose regulation in vivo is unknown. Therefore, we investigated the effect of the hydrophobic bile acid subtype, deoxycholic acid (DCA), on ER stress and glucose regulation. Eight week old C57BL/6J mice were fed a high fat diet supplemented with or without DCA. Glucose regulation was assessed by oral glucose tolerance and insulin tolerance testing. In addition, circulating bile acid profile and hepatic insulin and ER stress signaling were measured. DCA supplementation did not alter body weight or food intake, but did impair glucose regulation. Consistent with the impairment in glucose regulation, DCA increased the hydrophobicity of the circulating bile acid profile, decreased hepatic insulin signaling and increased hepatic ER stress signaling. Together, these data suggest that dietary supplementation of DCA impairs whole body glucose regulation by disrupting hepatic ER homeostasis in mice.
Collapse
Affiliation(s)
- Karolina E. Zaborska
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States of America
| | - Seon A. Lee
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States of America
| | - Darline Garribay
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States of America
| | - Eumee Cha
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States of America
| | - Bethany P. Cummings
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States of America
| |
Collapse
|
52
|
Zhang Y, LaCerte C, Kansra S, Jackson JP, Brouwer KR, Edwards JE. Comparative potency of obeticholic acid and natural bile acids on FXR in hepatic and intestinal in vitro cell models. Pharmacol Res Perspect 2018; 5. [PMID: 29226620 PMCID: PMC5723701 DOI: 10.1002/prp2.368] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/10/2017] [Indexed: 12/13/2022] Open
Abstract
Obeticholic acid (OCA) is a semisynthetic farnesoid X receptor (FXR) agonist, an analogue of chenodeoxycholic acid (CDCA) which is indicated for the treatment of primary biliary cholangitis (PBC) in combination with ursodeoxycholic acid (UDCA). OCA efficiently inhibits bile acid synthesis and promotes bile acid efflux via activating FXR-mediated mechanisms in a physiologically relevant in vitro cell system, Sandwich-cultured Transporter Certified ™ human primary hepatocytes (SCHH). The study herein evaluated the effects of UDCA alone or in combination with OCA in SCHH. UDCA (≤100 μmol/L) alone did not inhibit CYP7A1 mRNA, and thus, no reduction in the endogenous bile acid pool observed. UDCA ≤100 μmol/L concomitantly administered with 0.1 μmol/L OCA had no effect on bile acid synthesis beyond what was observed with OCA alone. Furthermore, this study evaluated human Caco-2 cells (clone C2BBe1) as in vitro intestinal models. Glycine conjugate of OCA increased mRNA levels of FXR target genes in Caco-2 cells, FGF-19, SHP, OSTα/β, and IBABP, but not ASBT, in a concentration-dependent manner, while glycine conjugate of UDCA had no effect on the expression of these genes. The results suggested that UDCA ≤100 μmol/L did not activate FXR in human primary hepatocytes or intestinal cell line Caco-2. Thus, co-administration of UDCA with OCA did not affect OCA-dependent pharmacological effects.
Collapse
Affiliation(s)
| | - Carl LaCerte
- Intercept Pharmaceuticals Inc., San Diego, CA, USA
| | | | | | | | | |
Collapse
|
53
|
Zhang Y, Lickteig AJ, Csanaky IL, Klaassen CD. Editor's Highlight: Clofibrate Decreases Bile Acids in Livers of Male Mice by Increasing Biliary Bile Acid Excretion in a PPARα-Dependent Manner. Toxicol Sci 2018; 160:351-360. [PMID: 28973556 DOI: 10.1093/toxsci/kfx191] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Fibrates and their receptor, namely peroxisome proliferator-activated receptor α (PPARα), have been reported to regulate bile acid (BA) synthesis and transport. However, the effect of fibrate treatment and PPARα activation on BA homeostasis remains controversial. In this study, both wild-type (WT) and PPARα-null male mice were treated with clofibrate (CLOF) for 4 days to evaluate the effects of short-term PPARα activation on BA homeostasis. Although a decrease in total BAs (ΣBAs) was observed in livers of CLOF-treated WT mice, it was not observed in PPARα-null mice. CLOF-mediated decrease in ΣBAs in the liver was not likely due to the reduction in BA synthesis or BA uptake, as evidenced by an increase in the BA synthetic enzyme (Cyp7a1) and 2 BA uptake transporters (Na (+)-taurocholate cotransporting polypeptide [Ntcp] and organic anion transporting polypeptide [Oatp]1b2). Instead, the decrease in liver BAs by CLOF is largely a result of increased biliary excretion of BAs, which was associated with a significant induction of the canalicular efflux transporter (bile salt export pump [Bsep]) in the liver. The PPARα-mediated increase in Cyp7a1 in CLOF-treated WT mice was not due to farnesoid X receptor (Fxr)-small heterodimer partner (Shp) signaling in the liver, but due to suppression of Fxr- fibroblast growth factor15 signaling in the ileum. Additionally, CLOF also suppressed intestinal BA transporters (apical sodium-dependent bile acid transporter and organic solute transporterβ) and cholesterol efflux transporters (Abcg5 and Abcg8) in a PPARα-dependent manner. In summary, this study provides the first comprehensive analysis on the effect of a short-term CLOF treatment on BA homeostasis, and revealed an essential role of PPARα in regulating BA synthesis, transport and signaling.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism
- Animals
- Bile Acids and Salts/metabolism
- Clofibrate/pharmacology
- Fibroblast Growth Factors/metabolism
- Gastrointestinal Agents/pharmacology
- Genotype
- Homeostasis
- Ileum/drug effects
- Ileum/metabolism
- Lipoproteins/metabolism
- Liver/drug effects
- Liver/metabolism
- Male
- Membrane Transport Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Organic Anion Transporters, Sodium-Dependent/metabolism
- PPAR alpha/agonists
- PPAR alpha/genetics
- PPAR alpha/metabolism
- Phenotype
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction/drug effects
- Steroid 17-alpha-Hydroxylase/metabolism
- Symporters/metabolism
- Time Factors
Collapse
Affiliation(s)
- Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Andrew J Lickteig
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Iván L Csanaky
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Division of Gastroenterology, Children's Mercy Hospital & Clinics, Kansas City, Missouri 64108
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160
| |
Collapse
|
54
|
Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, Agdashian D, Terabe M, Berzofsky JA, Fako V, Ritz T, Longerich T, Theriot CM, McCulloch JA, Roy S, Yuan W, Thovarai V, Sen SK, Ruchirawat M, Korangy F, Wang XW, Trinchieri G, Greten TF. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018; 360:360/6391/eaan5931. [PMID: 29798856 DOI: 10.1126/science.aan5931] [Citation(s) in RCA: 1003] [Impact Index Per Article: 143.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 02/27/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
Primary liver tumors and liver metastasis currently represent the leading cause of cancer-related death. Commensal bacteria are important regulators of antitumor immunity, and although the liver is exposed to gut bacteria, their role in antitumor surveillance of liver tumors is poorly understood. We found that altering commensal gut bacteria in mice induced a liver-selective antitumor effect, with an increase of hepatic CXCR6+ natural killer T (NKT) cells and heightened interferon-γ production upon antigen stimulation. In vivo functional studies showed that NKT cells mediated liver-selective tumor inhibition. NKT cell accumulation was regulated by CXCL16 expression of liver sinusoidal endothelial cells, which was controlled by gut microbiome-mediated primary-to-secondary bile acid conversion. Our study suggests a link between gut bacteria-controlled bile acid metabolism and liver antitumor immunosurveillance.
Collapse
Affiliation(s)
- Chi Ma
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Miaojun Han
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bernd Heinrich
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qiong Fu
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qianfei Zhang
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Milan Sandhu
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Agdashian
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Valerie Fako
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Ritz
- Institute of Pathology, University Hospital RWTH Aachen, Aachen 52074, Germany
| | - Thomas Longerich
- Institute of Pathology, University Hospital RWTH Aachen, Aachen 52074, Germany.,Institute of Pathology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Casey M Theriot
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - John A McCulloch
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Soumen Roy
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wuxing Yuan
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Leidos Biomedical Research, Inc, Microbiome Sequencing Core, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vishal Thovarai
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Leidos Biomedical Research, Inc, Microbiome Sequencing Core, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shurjo K Sen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Leidos Biomedical Research, Inc, Microbiome Sequencing Core, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Firouzeh Korangy
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,NCI CCR Liver Cancer Program, Bethesda, MD, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA. .,NCI CCR Liver Cancer Program, Bethesda, MD, USA
| |
Collapse
|
55
|
Gypenosides Altered Hepatic Bile Acids Homeostasis in Mice Treated with High Fat Diet. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:8098059. [PMID: 30105069 PMCID: PMC6076974 DOI: 10.1155/2018/8098059] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/18/2018] [Accepted: 03/29/2018] [Indexed: 02/08/2023]
Abstract
Gypenosides extracted from Gynostemma pentaphyllum (Thunb.) Makino have significant role in reducing serum lipid level and treating fatty liver diseases, however, without clear mechanism. As gypenosides share the similar core structures with bile acids (the endogenous ligands of nuclear receptor FXR), we hypothesize that gypenosides may improve hypercholesterolemia via FXR-mediated bile acids signaling. The present study was designed to validate the role of gypenosides in reducing levels of serum total cholesterol (TC) and low density lipoprotein cholesterol (LDL-C), as well as in regulating bile acids homeostasis and related gene expression levels. The C57BL/6 male mice were divided into four groups. Mice in groups ND and HFD were fed with normal diet and high fat diet for 38 weeks, respectively. In groups HFD+GP and HFD+ST, mice were fed with high fat diet for 38 weeks and treated with gypenosides and simvastatin (positive control) from weeks 16 to 38, respectively. Serum TC and LDL-C levels were assayed by commercially available kits. Expression levels of genes were tested by the quantitative real-time PCR. The LC-MS/MS was applied to quantify major bile acids in mice livers. Our results showed that gypenosides significantly decreased serum TC and LDL-C levels. The gene expression level of Shp was downregulated while the levels of Cyp7a1, Cyp8b1, Fxr, Lrh1, Jnk1/2, and Erk1/2 were upregulated by gypenosides. Indicated by LC-MS/MS technology, gypenosides increased the hepatic levels of several free bile acids and most taurine-conjugated bile acids while decreasing glycine-conjugated bile acids levels. In addition, gypenosides decreased the CA/CDCA ratio. Gypenosides may improve the abnormal lipid profile of HFD-fed mice via two pathways: (1) enhancing the bile acids biosynthesis from cholesterol; (2) decreasing the CA/CDCA ratio which is positively related to cholesterol absorption.
Collapse
|
56
|
Thakare R, Alamoudi JA, Gautam N, Rodrigues AD, Alnouti Y. Species differences in bile acids I. Plasma and urine bile acid composition. J Appl Toxicol 2018; 38:1323-1335. [DOI: 10.1002/jat.3644] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/11/2018] [Accepted: 04/11/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Rhishikesh Thakare
- Department of Pharmaceutical Sciences, College of Pharmacy; University of Nebraska Medical Center; Omaha NE 68198 USA
| | - Jawaher Abdullah Alamoudi
- Department of Pharmaceutical Sciences, College of Pharmacy; University of Nebraska Medical Center; Omaha NE 68198 USA
| | - Nagsen Gautam
- Department of Pharmaceutical Sciences, College of Pharmacy; University of Nebraska Medical Center; Omaha NE 68198 USA
| | - A. David Rodrigues
- Pharmacokinetics, Pharmacodynamics & Metabolism, Medicine Design, Pfizer Inc.; Groton CT 06340 USA
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, College of Pharmacy; University of Nebraska Medical Center; Omaha NE 68198 USA
| |
Collapse
|
57
|
Li J, Dawson PA. Animal models to study bile acid metabolism. Biochim Biophys Acta Mol Basis Dis 2018; 1865:895-911. [PMID: 29782919 DOI: 10.1016/j.bbadis.2018.05.011] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/19/2022]
Abstract
The use of animal models, particularly genetically modified mice, continues to play a critical role in studying the relationship between bile acid metabolism and human liver disease. Over the past 20 years, these studies have been instrumental in elucidating the major pathways responsible for bile acid biosynthesis and enterohepatic cycling, and the molecular mechanisms regulating those pathways. This work also revealed bile acid differences between species, particularly in the composition, physicochemical properties, and signaling potential of the bile acid pool. These species differences may limit the ability to translate findings regarding bile acid-related disease processes from mice to humans. In this review, we focus primarily on mouse models and also briefly discuss dietary or surgical models commonly used to study the basic mechanisms underlying bile acid metabolism. Important phenotypic species differences in bile acid metabolism between mice and humans are highlighted.
Collapse
Affiliation(s)
- Jianing Li
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322, United States
| | - Paul A Dawson
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322, United States.
| |
Collapse
|
58
|
Diabetic cognitive dysfunction is associated with increased bile acids in liver and activation of bile acid signaling in intestine. Toxicol Lett 2018; 287:10-22. [DOI: 10.1016/j.toxlet.2018.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/20/2017] [Accepted: 01/04/2018] [Indexed: 01/19/2023]
|
59
|
Csanaky IL, Lickteig AJ, Klaassen CD. Aryl hydrocarbon receptor (AhR) mediated short-term effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on bile acid homeostasis in mice. Toxicol Appl Pharmacol 2018. [PMID: 29452137 DOI: 10.1016/j.taap.2018.02.005.aryl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The effects of the most potent aryl hydrocarbon receptor (AhR) agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on bile acid (BA) homeostasis was examined in male and female wild-type and AhR-null mice shortly after 4-day exposure, rather than at a later time when secondary non-AhR dependent effects are more likely to occur. TCDD had similar effects on BA homeostasis in male and female mice. TCDD decreased the concentration of total-(Σ) BAs in liver by approximately 50% (all major BA categories except for the non-6,12-OH BAs), without decreasing the expression of the rate limiting BA synthetic enzyme (Cyp7a1) or altering the major BA regulatory pathways (FXR) in liver and intestine. Even though the Σ-BAs in liver were markedly decreased, the Σ-BAs excreted into bile were not altered. TCDD decreased the relative amount of 12-OH BAs (TCA, TDCA, CA, DCA) in bile and increased the biliary excretion of TCDCA and its metabolites (TαMCA, TUDCA); this was likely due to the decreased Cyp8b1 (12α-hydroxylase) in liver. The concentration of Σ-BAs in serum was not altered by TCDD, indicating that serum BAs do not reflect BA status in liver. However, proportions of individual BAs in serum reflected the decreased expression of Cyp8b1. All these TCDD-induced changes in BA homeostasis were absent in AhR-null mice. In summary, through the AhR, TCDD markedly decreases BA concentrations in liver and reduces the 12α-hydroxylation of BAs without altering Cyp7a1 and FXR signaling. The TCDD-induced decrease in Σ-BAs in liver did not result in a decrease in biliary excretion or serum concentrations of Σ-BAs.
Collapse
Affiliation(s)
- Iván L Csanaky
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Division of Gastroenterology, Children's Mercy Hospital, Kansas City, MO 64108; USA; Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Andrew J Lickteig
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
60
|
Csanaky IL, Lickteig AJ, Klaassen CD. Aryl hydrocarbon receptor (AhR) mediated short-term effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on bile acid homeostasis in mice. Toxicol Appl Pharmacol 2018; 343:48-61. [PMID: 29452137 DOI: 10.1016/j.taap.2018.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/25/2018] [Accepted: 02/12/2018] [Indexed: 01/05/2023]
Abstract
The effects of the most potent aryl hydrocarbon receptor (AhR) agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on bile acid (BA) homeostasis was examined in male and female wild-type and AhR-null mice shortly after 4-day exposure, rather than at a later time when secondary non-AhR dependent effects are more likely to occur. TCDD had similar effects on BA homeostasis in male and female mice. TCDD decreased the concentration of total-(Σ) BAs in liver by approximately 50% (all major BA categories except for the non-6,12-OH BAs), without decreasing the expression of the rate limiting BA synthetic enzyme (Cyp7a1) or altering the major BA regulatory pathways (FXR) in liver and intestine. Even though the Σ-BAs in liver were markedly decreased, the Σ-BAs excreted into bile were not altered. TCDD decreased the relative amount of 12-OH BAs (TCA, TDCA, CA, DCA) in bile and increased the biliary excretion of TCDCA and its metabolites (TαMCA, TUDCA); this was likely due to the decreased Cyp8b1 (12α-hydroxylase) in liver. The concentration of Σ-BAs in serum was not altered by TCDD, indicating that serum BAs do not reflect BA status in liver. However, proportions of individual BAs in serum reflected the decreased expression of Cyp8b1. All these TCDD-induced changes in BA homeostasis were absent in AhR-null mice. In summary, through the AhR, TCDD markedly decreases BA concentrations in liver and reduces the 12α-hydroxylation of BAs without altering Cyp7a1 and FXR signaling. The TCDD-induced decrease in Σ-BAs in liver did not result in a decrease in biliary excretion or serum concentrations of Σ-BAs.
Collapse
Affiliation(s)
- Iván L Csanaky
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Division of Gastroenterology, Children's Mercy Hospital, Kansas City, MO 64108; USA; Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Andrew J Lickteig
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
61
|
Zhu D, Sorg JA, Sun X. Clostridioides difficile Biology: Sporulation, Germination, and Corresponding Therapies for C. difficile Infection. Front Cell Infect Microbiol 2018; 8:29. [PMID: 29473021 PMCID: PMC5809512 DOI: 10.3389/fcimb.2018.00029] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/23/2018] [Indexed: 12/18/2022] Open
Abstract
Clostridioides difficile is a Gram-positive, spore-forming, toxin-producing anaerobe, and an important nosocomial pathogen. Due to the strictly anaerobic nature of the vegetative form, spores are the main morphotype of infection and transmission of the disease. Spore formation and their subsequent germination play critical roles in C. difficile infection (CDI) progress. Under suitable conditions, C. difficile spores will germinate and outgrow to produce the pathogenic vegetative form. During CDI, C. difficile produces toxins (TcdA and TcdB) that are required to initiate the disease. Meanwhile, it also produces spores that are responsible for the persistence and recurrence of C. difficile in patients. Recent studies have shed light on the regulatory mechanisms of C. difficile sporulation and germination. This review is to summarize recent advances on the regulation of sporulation/germination in C. difficile and the corresponding therapeutic strategies that are aimed at these important processes.
Collapse
Affiliation(s)
- Duolong Zhu
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Joseph A Sorg
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
62
|
von Hardenberg S, Gnewuch C, Schmitz G, Borlak J. ApoE is a major determinant of hepatic bile acid homeostasis in mice. J Nutr Biochem 2018; 52:82-91. [DOI: 10.1016/j.jnutbio.2017.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/13/2017] [Accepted: 09/09/2017] [Indexed: 12/27/2022]
|
63
|
Different effects of ursodeoxycholic acid on intrahepatic cholestasis in acute and recovery stages induced by alpha-naphthylisothiocyanate in mice. Toxicol Appl Pharmacol 2018; 342:69-78. [PMID: 29407775 DOI: 10.1016/j.taap.2018.01.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/18/2018] [Accepted: 01/27/2018] [Indexed: 01/03/2023]
Abstract
The aim of this study was to determine the effect of ursodeoxycholic acid (UDCA) on the alpha-naphthylisothiocyanate (ANIT)-induced acute and recovery stage of cholestasis model mice. In the acute stage of model mice, pretreatment with UDCA (25, 50, and 100 mg·kg-1, ig) for 12 days prior to ANIT administration (50 mg·kg-1, ig) resulted in the dramatic increase in serum biochemistry, with aggrevation of bile infarcts and hepatocyte necrosis. The elevation of beta-muricholic acid (β-MCA), cholic acid (CA), and taurocholic acid (TCA) in serum and liver, and reduction of these bile acids (BAs) in bile was observed. In contrast, in the recovery stage of model mice, treatment with UDCA (25, 50, and 100 mg·kg-1, ig) for 7 days after ANIT administration (50 mg·kg-1, ig) resulted in the significant decrease in levels of serum alanine aminotransferase (ALT) and total bile acid (TBA). Liver injury was attenuated, and the levels of TBA, CA, TCA, and β-MCA in the liver were significantly decreased. Additionally, UDCA can upregulate expression of BSEP, but it cannot upregulate expression of AE2. UDCA, which induced BSEP to increase bile acid-dependent bile flow, aggravated cholestasis and liver injury when the bile duct was obstructed in the acute stage of injury in model mice. In contrast, UDCA alleviated cholestasis and liver injury induced by ANIT when the obstruction was improved in the recovery stage.
Collapse
|
64
|
Impaired vagus function in rats suppresses bile acid synthesis in the liver by disrupting tight junctions and activating Fxr-Fgf15 signaling in the intestine. Biochem Biophys Res Commun 2018; 495:1490-1496. [DOI: 10.1016/j.bbrc.2017.11.201] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 11/29/2017] [Indexed: 12/12/2022]
|
65
|
Lee EA, Lee DI, Kim HY, Ahn SH, Seong HR, Jung WH, Kim KY, Kim S, Rhee SD. Cyp7a1 is continuously increased with disrupted Fxr-mediated feedback inhibition in hypercholesterolemic TALLYHO/Jng mice. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:20-25. [DOI: 10.1016/j.bbalip.2017.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 10/19/2022]
|
66
|
Zhang Y, Lickteig AJ, Csanaky IL, Klaassen CD. Activation of PPARα decreases bile acids in livers of female mice while maintaining bile flow and biliary bile acid excretion. Toxicol Appl Pharmacol 2017; 338:112-123. [PMID: 29175453 DOI: 10.1016/j.taap.2017.11.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/16/2017] [Accepted: 11/19/2017] [Indexed: 01/20/2023]
Abstract
Fibrates are hypolipidemic drugs that act as activators of peroxisome proliferator-activated receptor α (PPARα). In both humans and rodents, females were reported to be less responsive to fibrates than males. Previous studies on fibrates and PPARα usually involved male mice, but little has been done in females. The present study aimed to provide the first comprehensive analysis of the effects of clofibrate (CLOF) and PPARα on bile acid (BA) homeostasis in female mice. Study in WT male mice showed that a 4-day CLOF treatment increased liver weight, bile flow, and biliary BA excretion, but decreased total BAs in both serum and liver. In contrast, WT female mice were less susceptible to these CLOF-mediated responses observed in males. In WT female mice, CLOF decreased total BAs in the liver, but had little effect on the mRNAs of hepatic BA-related genes. Next, a comparative analysis between WT and PPARα-null female mice showed that lack of PPARα in female mice decreased total BAs in serum, but had little effect on total BAs in liver or bile. However, lack of PPARα in female mice increased mRNAs of BA synthetic enzymes (Cyp7a1, Cyp8b1, Cyp27a1, and Cyp7b1) and transporters (Ntcp, Oatp1a1, Oatp1b2, and Mrp3). Furthermore, the increase of Cyp7a1 in PPARα-null female mice was associated with an increase in liver Fxr-Shp-Lrh-1 signaling. In conclusion, female mice are resistant to CLOF-mediated effects on BA metabolism observed in males, which could be attributed to PPARα-mediated suppression in females on genes involved in BA synthesis and transport.
Collapse
Affiliation(s)
- Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China.
| | - Andrew J Lickteig
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160,USA
| | - Iván L Csanaky
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Division of Gastroenterology, Children's Mercy Hospital & Clinics, Kansas City, MO 64108; USA; Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160,USA.
| |
Collapse
|
67
|
Possible Role of Phosphatidylcholine and Sphingomyelin on Fumonisin B1-mediated Toxicity. Food Saf (Tokyo) 2017; 5:75-97. [PMID: 32231933 DOI: 10.14252/foodsafetyfscj.2017004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/29/2017] [Indexed: 12/21/2022] Open
Abstract
A major corn-related mycotoxin, fumonisin B1 (FB1), continues to attract attention of researchers as well as risk-assessors due to the diverse toxicological characteristics, including distinct target tissues in different animal species and opposite susceptibility in males and females in mice and rats. More than thirty years passed since the structure identification as a sphingoid-like chemical, but the causal mechanism of the toxicity remains obscure in spites of extensive studies. Considerable amounts of knowledge have been accumulated on the biochemical/toxicological actions of FB1, but the influence on lipid dynamics and mobilization in the body has not been focused well in relation to the FB1-mediated toxicity. Considerable influences of this toxin on mobilization of sphingolipids and phospholipids and also on adaptive changes in their compositions in tissues are implicated from recent studies on FB1-interacting ceramide synthases. Accumulated patho-physiological data also suggest a possible role of hepatic phospholipid on FB1-mediated toxicity. Thus, a mechanism of FB1-mediated toxicity is discussed in relation to the mobilization of phospholipids and sphingolipids in the body in this context.
Collapse
|
68
|
Thanissery R, Winston JA, Theriot CM. Inhibition of spore germination, growth, and toxin activity of clinically relevant C. difficile strains by gut microbiota derived secondary bile acids. Anaerobe 2017; 45:86-100. [PMID: 28279860 PMCID: PMC5466893 DOI: 10.1016/j.anaerobe.2017.03.004] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/02/2017] [Accepted: 03/04/2017] [Indexed: 12/18/2022]
Abstract
The changing epidemiology of Clostridium difficile infection over the past decades presents a significant challenge in the management of C. difficile associated diseases. The gastrointestinal tract microbiota provides colonization resistance against C. difficile, and growing evidence suggests that gut microbial derived secondary bile acids (SBAs) play a role. We hypothesized that the C. difficile life cycle; spore germination and outgrowth, growth, and toxin production, of strains that vary by age and ribotype will differ in their sensitivity to SBAs. C. difficile strains R20291 and CD196 (ribotype 027), M68 and CF5 (017), 630 (012), BI9 (001) and M120 (078) were used to define taurocholate (TCA) mediated spore germination and outgrowth, growth, and toxin activity in the absence and presence of gut microbial derived SBAs (deoxycholate, isodeoxycholate, lithocholate, isolithocholate, ursodeoxycholate, ω-muricholate, and hyodeoxycholate) found in the human and mouse large intestine. C. difficile strains varied in their rates of germination, growth kinetics, and toxin activity without the addition of SBAs. C. difficile M120, a highly divergent strain, had robust germination, growth, but significantly lower toxin activity compared to other strains. Many SBAs were able to inhibit TCA mediated spore germination and outgrowth, growth, and toxin activity in a dose dependent manner, but the level of inhibition and resistance varied across all strains and ribotypes. This study illustrates how clinically relevant C. difficile strains can have different responses when exposed to SBAs present in the gastrointestinal tract.
Collapse
Affiliation(s)
- Rajani Thanissery
- Department of Population Health and Pathobiology, College of Veterinary Medicine, Research Building 424, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607, United States.
| | - Jenessa A Winston
- Department of Population Health and Pathobiology, College of Veterinary Medicine, Research Building 424, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607, United States.
| | - Casey M Theriot
- Department of Population Health and Pathobiology, College of Veterinary Medicine, Research Building 424, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607, United States.
| |
Collapse
|
69
|
González-Peña D, Giménez L, de Ancos B, Sánchez-Moreno C. Role of dietary onion in modifying the faecal bile acid content in rats fed a high-cholesterol diet. Food Funct 2017; 8:2184-2192. [PMID: 28504277 DOI: 10.1039/c7fo00412e] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The determination of faecal bile patterns offers new opportunities in the search for non-invasive biomarkers of disease status. The objective of this study was to describe the shifts in faecal bile acid (BA) composition induced by feeding a high-cholesterol/cholic acid diet (HC) over 7 weeks of experimental feeding in Wistar rats, and to evaluate the effect of onion included as a functional ingredient (HCO). A HPLC-MS/MS method allowed the detection of 29 bile acids, 10 of which were tentatively identified and 12 confirmed and quantified by means of standards and calibration curves. The excretion of bile acids revealed a discriminating bile acid profile between the HC and HCO groups compared with the C group. HCO feeding indicated significant changes in specific primary and secondary BA in both the unconjugated and conjugated forms caused by the addition of the onion ingredient to the diet. The results suggest that the induction of microbiome modifications by the HC and HCO diets acts as a critical modifier of the faecal bile acid composition. These modifications might reflect and be linked to changes in the reabsorption of BA at an intestinal level and the process of BA deconjugation in the course of hypercholesterolemia.
Collapse
Affiliation(s)
- Diana González-Peña
- Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), ES-28040 Madrid, Spain.
| | - Lucía Giménez
- Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), ES-28040 Madrid, Spain.
| | - Begoña de Ancos
- Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), ES-28040 Madrid, Spain.
| | - Concepción Sánchez-Moreno
- Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), ES-28040 Madrid, Spain.
| |
Collapse
|
70
|
Feng L, Yuen YL, Xu J, Liu X, Chan MYC, Wang K, Fong WP, Cheung WT, Lee SST. Identification and characterization of a novel PPARα-regulated and 7α-hydroxyl bile acid-preferring cytosolic sulfotransferase mL-STL (Sult2a8). J Lipid Res 2017; 58:1114-1131. [PMID: 28442498 DOI: 10.1194/jlr.m074302] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 04/19/2017] [Indexed: 12/25/2022] Open
Abstract
PPARα has been known to play a pivotal role in orchestrating lipid, glucose, and amino acid metabolism via transcriptional regulation of its target gene expression during energy deprivation. Recent evidence has also suggested that PPARα is involved in bile acid metabolism, but how PPARα modulates the homeostasis of bile acids during fasting is still not clear. In a mechanistic study aiming to dissect the spectrum of PPARα target genes involved in metabolic response to fasting, we identified a novel mouse gene (herein named mL-STL for mouse liver-sulfotransferase-like) that shared extensive homology with the Sult2a subfamily of a superfamily of cytosolic sulfotransferases, implying its potential function in sulfonation. The mL-STL gene expressed predominantly in liver in fed state, but PPARα was required to sustain its expression during fasting, suggesting a critical role of PPARα in regulating the mL-STL-mediated sulfonation during fasting. Functional studies using recombinant His-tagged mL-STL protein revealed its narrow sulfonating activities toward 7α-hydroxyl primary bile acids, including cholic acid, chenodeoxycholic acid, and α-muricholic acid, and thus suggesting that mL-STL may be the major hepatic bile acid sulfonating enzyme in mice. Together, these studies identified a novel PPARα-dependent gene and uncovered a new role of PPARα as being an essential regulator in bile acid biotransformation via sulfonation during fasting.
Collapse
Affiliation(s)
- Lu Feng
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Yee-Lok Yuen
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Jian Xu
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Xing Liu
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Martin Yan-Chun Chan
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Kai Wang
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Wing-Ping Fong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Wing-Tai Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Susanna Sau-Tuen Lee
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| |
Collapse
|
71
|
Zeng H, Jiang Y, Chen P, Fan X, Li D, Liu A, Ma X, Xie W, Liu P, Gonzalez FJ, Huang M, Bi H. Schisandrol B protects against cholestatic liver injury through pregnane X receptors. Br J Pharmacol 2017; 174:672-688. [PMID: 28128437 DOI: 10.1111/bph.13729] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 01/22/2017] [Accepted: 01/23/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Currently, ursodeoxycholic acid and obeticholic acid are the only two FDA-approved drugs for cholestatic liver diseases. Thus, new therapeutic approaches need to be developed. Here we have evaluated the anti-cholestasis effects of Schisandrol B (SolB), a bioactive compound isolated from Schisandra sphenanthera. EXPERIMENTAL APPROACH Hepatoprotective effect of SolB against intrahepatic cholestasis, induced by lithocholic acid (LCA), was evaluated in mice. Metabolomic analysis and gene analysis were used to assess involvement of pregnane X receptor (PXR). Molecular docking, cell-based reporter gene analysis and knockout mice were used to demonstrate the critical role of the PXR pathway in the anti-cholestasis effects of SolB. KEY RESULTS SolB protected against LCA-induced intrahepatic cholestasis. Furthermore, therapeutic treatment with SolB decreased mortality in cholestatic mice. Metabolomics and gene analysis showed that SolB accelerated metabolism of bile acids, promoted bile acid efflux into the intestine, and induced hepatic expression of the PXR-target genes Cyp3a11, Ugt1a1, and Oatp2, which are involved in bile acid homeostasis. Mechanistic studies showed that SolB activated human PXR and up-regulated PXR target genes in human cell lines. Additionally, SolB did not protect Pxr-null mice from liver injury induced by intrahepatic cholestasis, thus providing genetic evidence that the effect of SolB was PXR-dependent. CONCLUSION AND IMPLICATIONS These findings provide direct evidence for the hepatoprotective effects of SolB against cholestasis by activating PXR. Therefore, SolB may provide a new and effective approach to the prevention and treatment of cholestatic liver diseases.
Collapse
Affiliation(s)
- Hang Zeng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yiming Jiang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Pan Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaomei Fan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Dongshun Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Aiming Liu
- Medical School of Ningbo University, Ningbo, China
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peiqing Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Min Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Huichang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
72
|
Bu P, Le Y, Zhang Y, Zhang Y, Cheng X. Berberine-induced Inactivation of Signal Transducer and Activator of Transcription 5 Signaling Promotes Male-specific Expression of a Bile Acid Uptake Transporter. J Biol Chem 2017; 292:4602-4613. [PMID: 28154180 DOI: 10.1074/jbc.m116.757567] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/29/2017] [Indexed: 01/05/2023] Open
Abstract
Sodium-taurocholate co-transporting polypeptide (Ntcp/NTCP) is the major uptake transporter of bile salts in mouse and human livers. In certain diseases, including endotoxemia, cholestasis, diabetes, and hepatocarcinoma, Ntcp/NTCP expression is markedly reduced, which interferes with enterohepatic circulation of bile salts, impairing the absorption of lipophilic compounds. Therefore, normal Ntcp/NTCP expression in the liver is physiologically important. Berberine is an herbal medicine used historically to improve liver function and has recently been shown to repress STAT signaling. However, berberine effects on Ntcp/NTCP expression are unknown, prompting use to investigate this possible connection. Our results showed that berberine dose-dependently increased Ntcp expression in male mouse liver and decreased taurocholic acid levels in serum but increased them in the liver. In mouse and human hepatoma cells, berberine induced Ntcp/NTCP mRNA and protein expression and increased cellular uptake of [3H] taurocholate. Mechanistically, berberine decreased nuclear protein levels of phospho-JAK2 and phospho-STAT5, thus disrupting the JAK2-STAT5 signaling. Moreover, berberine stimulated luciferase reporter expression from the mouse Ntcp promoter when one putative STAT5 response element (RE) (-1137 bp) was deleted and from the human NTCP promoter when three putative STAT5REs (-2898, -2164, and -691 bp) were deleted. Chromatin immunoprecipitation demonstrated that berberine decreased binding of phospho-STAT5 protein to the-2164 and -691 bp STAT5REs in the human NTCP promoter. In summary, berberine-disrupted STAT5 signaling promoted mouse and human Ntcp/NTCP expression, resulting in enhanced bile acid uptake. Therefore, berberine may be a therapeutic candidate compound for maintaining bile acid homeostasis.
Collapse
Affiliation(s)
- Pengli Bu
- From the Departments of Pharmaceutical Sciences and.,Biological Sciences, St. John's University, Queens, New York 11439 and
| | - Yuan Le
- From the Departments of Pharmaceutical Sciences and
| | - Yue Zhang
- From the Departments of Pharmaceutical Sciences and
| | - Youcai Zhang
- the School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | | |
Collapse
|
73
|
Zhang F, Qin H, Zhao Y, Wei Y, Xi L, Rao Z, Zhang J, Ma Y, Duan Y, Wu X. Effect of cholecystectomy on bile acids as well as relevant enzymes and transporters in mice: Implication for pharmacokinetic changes of rifampicin. Eur J Pharm Sci 2017; 96:141-153. [DOI: 10.1016/j.ejps.2016.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 12/19/2022]
|
74
|
Wahlström A, Kovatcheva-Datchary P, Ståhlman M, Khan MT, Bäckhed F, Marschall HU. Induction of farnesoid X receptor signaling in germ-free mice colonized with a human microbiota. J Lipid Res 2016; 58:412-419. [PMID: 27956475 DOI: 10.1194/jlr.m072819] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/06/2016] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota influences the development and progression of metabolic diseases partly by metabolism of bile acids (BAs) and modified signaling through the farnesoid X receptor (FXR). In this study, we aimed to determine how the human gut microbiota metabolizes murine BAs and affects FXR signaling in colonized mice. We colonized germ-free mice with cecal content from a mouse donor or feces from a human donor and euthanized the mice after short-term (2 weeks) or long-term (15 weeks) colonization. We analyzed the gut microbiota and BA composition and expression of FXR target genes in ileum and liver. We found that cecal microbiota composition differed between mice colonized with mouse and human microbiota and was stable over time. Human and mouse microbiota reduced total BA levels similarly, but the humanized mice produced less secondary BAs. The human microbiota was able to reduce the levels of tauro-β-muricholic acid and induce expression of FXR target genes Fgf15 and Shp in ileum after long-term colonization. We show that a human microbiota can change BA composition and induce FXR signaling in colonized mice, but the levels of secondary BAs produced are lower than in mice colonized with a mouse microbiota.
Collapse
Affiliation(s)
- Annika Wahlström
- Department of Molecular and Clinical Medicine and Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Petia Kovatcheva-Datchary
- Department of Molecular and Clinical Medicine and Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine and Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Muhammad-Tanweer Khan
- Department of Molecular and Clinical Medicine and Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- Department of Molecular and Clinical Medicine and Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden .,Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine and Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
75
|
Rudling M. Understanding mouse bile acid formation: Is it time to unwind why mice and rats make unique bile acids? J Lipid Res 2016; 57:2097-2098. [PMID: 27777318 DOI: 10.1194/jlr.c072876] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Mats Rudling
- Metabolism Unit and KI/AZ Integrated Cardio Metabolic Center, Department of Medicine, Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute at Karolinska University Hospital Huddinge, S-141 86 Stockholm, Sweden
| |
Collapse
|
76
|
Guo Y, Zhang Y, Huang W, Selwyn FP, Klaassen CD. Dose-response effect of berberine on bile acid profile and gut microbiota in mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:394. [PMID: 27756364 PMCID: PMC5070223 DOI: 10.1186/s12906-016-1367-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/29/2016] [Indexed: 02/04/2023]
Abstract
Background Berberine (BBR) is a traditional antimicrobial herbal medicine. Recently, BBR has gained popularity as a supplement to lower blood lipids, cholesterol and glucose. Bile acids (BAs) are known to regulate blood levels of triglycerides, cholesterol, glucose and energy homeostasis, and gut flora play an important role in BA metabolism. However, whether BBR alters BAs metabolism or dose-response effect of BBR on gut flora is unknown. Methods In this study, the effects of various doses of BBR on the concentrations of BAs in liver and serum of male C57BL/6 mice were determined by UPLC-MS/MS, and the expression of BA-related genes, as well as the amount of 32 of the most abundant gut bacterial species in the terminal ileum and large intestine of male C57BL/6 mice were quantified by RT-PCR and Quantigene 2.0 Reagent System, respectively. Results Unconjugated BAs and total BAs were significantly altered by BBR in serum but not in liver. Increased primary BAs (βMCA, TβMCA and TUDCA) and decreased secondary BAs (DCA, LCA and the T-conjugates) were observed in livers and serum of mice fed BBR. The expression of BA-synthetic enzymes (Cyp7a1 and 8b1) and uptake transporter (Ntcp) increased 39-400 % in liver of mice fed the higher doses of BBR, whereas nuclear receptors and efflux transporters were not markedly altered. In addition, Bacteroides were enriched in the terminal ileum and large bowel of mice treated with BBR. Conclusion The present study indicated that various doses of BBR have effects on BA metabolism and related genes as well as intestinal flora, which provides insight into many pathways of BBR effects. Electronic supplementary material The online version of this article (doi:10.1186/s12906-016-1367-7) contains supplementary material, which is available to authorized users.
Collapse
|
77
|
Sun L, Beggs K, Borude P, Edwards G, Bhushan B, Walesky C, Roy N, Manley MW, Gunewardena S, O'Neil M, Li H, Apte U. Bile acids promote diethylnitrosamine-induced hepatocellular carcinoma via increased inflammatory signaling. Am J Physiol Gastrointest Liver Physiol 2016; 311:G91-G104. [PMID: 27151938 PMCID: PMC4967172 DOI: 10.1152/ajpgi.00027.2015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/09/2016] [Indexed: 01/31/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common hepatic malignancy and the third leading cause of cancer related deaths. Previous studies have implicated bile acids in pathogenesis of HCC, but the mechanisms are not known. We investigated the mechanisms of HCC tumor promotion by bile acids the diethylnitrosamine (DEN)-initiation-cholic acid (CA)-induced tumor promotion protocol in mice. The data show that 0.2% CA treatment resulted in threefold increase in number and size of DEN-induced liver tumors. All tumors observed in DEN-treated mice were well-differentiated HCCs. The HCCs observed in DEN-treated CA-fed mice exhibited extensive CD3-, CD20-, and CD45-positive inflammatory cell aggregates. Microarray-based global gene expression studies combined with Ingenuity Pathway Analysis revealed significant activation of NF-κB and Nanog in the DEN-treated 0.2% CA-fed livers. Further studies showed significantly higher TNF-α and IL-1β mRNA, a marked increase in total and phosphorylated-p65 and phosphorylated IκBα (degradation form) in livers of DEN-treated 0.2% CA-fed mice. Treatment of primary mouse hepatocytes with various bile acids showed significant induction of stemness genes including Nanog, KLF4, Sox2, and Oct4. Quantification of total and 20 specific bile acids in liver, and serum revealed a tumor-associated bile acid signature. Finally, quantification of total serum bile acids in normal, cirrhotic, and HCC human samples revealed increased bile acids in serum of cirrhotic and HCC patients. Taken together, these data indicate that bile acids are mechanistically involved pathogenesis of HCC and may promote HCC formation via activation of inflammatory signaling.
Collapse
Affiliation(s)
- Lina Sun
- 1Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas; ,5Department of Hepatic Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Kevin Beggs
- 1Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas;
| | - Prachi Borude
- 1Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas;
| | - Genea Edwards
- 1Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas;
| | - Bharat Bhushan
- 1Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas;
| | - Chad Walesky
- 1Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas;
| | - Nairita Roy
- 1Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas;
| | - Michael W. Manley
- 1Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas;
| | - Sumedha Gunewardena
- 2Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas; ,3Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas;
| | - Maura O'Neil
- 4Department of Pathology, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Hua Li
- 5Department of Hepatic Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Udayan Apte
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas;
| |
Collapse
|
78
|
Bile acids in drug induced liver injury: Key players and surrogate markers. Clin Res Hepatol Gastroenterol 2016; 40:257-266. [PMID: 26874804 DOI: 10.1016/j.clinre.2015.12.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 12/21/2015] [Accepted: 12/27/2015] [Indexed: 02/04/2023]
Abstract
Bile acid research has gained great momentum since the role of bile acids as key signaling molecules in the enterohepatic circulation was discovered. Their physiological function in regulating their own homeostasis, as well as energy and lipid metabolism make them interesting targets for the pharmaceutical industry in the context of diseases such as bile acid induced diarrhea, bile acid induced cholestasis or nonalcoholic steatohepatitis. Changes in bile acid homeostasis are also linked to various types of drug-induced liver injury (DILI). However, the key question whether bile acids are surrogate markers for monitoring DILI or key pathogenic players in the onset and progression of DILI is under intense investigation. The purpose of this review is to summarize the different facets of bile acids in the context of normal physiology, hereditary defects of bile acid transport and DILI.
Collapse
|
79
|
Lickteig AJ, Csanaky IL, Pratt-Hyatt M, Klaassen CD. Activation of Constitutive Androstane Receptor (CAR) in Mice Results in Maintained Biliary Excretion of Bile Acids Despite a Marked Decrease of Bile Acids in Liver. Toxicol Sci 2016; 151:403-18. [PMID: 26984780 DOI: 10.1093/toxsci/kfw054] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Activation of Constitutive Androstane Receptor (CAR) protects against bile acid (BA)-induced liver injury. This study was performed to determine the effect of CAR activation on bile flow, BA profile, as well as expression of BA synthesis and transport genes. Synthetic CAR ligand 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) was administered to mice for 4 days. BAs were quantified by UPLC-MS/MS (ultraperformance liquid chromatography-tandem mass spectrometry). CAR activation decreases total BAs in livers of male (49%) and female mice (26%), largely attributable to decreases of the 12α-hydroxylated BA taurocholic acid (T-CA) (males (M) 65%, females (F) 45%). Bile flow in both sexes was increased by CAR activation, and the increases were BA-independent. CAR activation did not alter biliary excretion of total BAs, but overall BA composition changed. Excretion of muricholic (6-hydroxylated) BAs was increased in males (101%), and the 12α-OH proportion of biliary BAs was decreased in both males (37%) and females (28%). The decrease of T-CA in livers of males and females correlates with the decreased mRNA of the sterol 12α-hydroxylase Cyp8b1 in males (71%) and females (54%). As a response to restore BAs to physiologic concentrations in liver, mRNA of Cyp7a1 is upregulated following TCPOBOP (males 185%, females 132%). In ilea, mRNA of the negative feedback regulator Fgf15 was unaltered by CAR activation, indicating biliary BA excretion was sufficient to maintain concentrations of total BAs in the small intestine. In summary, the effects of CAR activation on BAs in male and female mice are quite similar, with a marked decrease in the major BA T-CA in the liver.
Collapse
Affiliation(s)
- Andrew J Lickteig
- *Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Iván L Csanaky
- *Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160; Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Hospital & Clinics, Kansas City, Missouri 64108; Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Matthew Pratt-Hyatt
- *Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Curtis D Klaassen
- *Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160; *Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160;
| |
Collapse
|
80
|
Kong B, Zhu Y, Li G, Williams JA, Buckley K, Tawfik O, Luyendyk JP, Guo GL. Mice with hepatocyte-specific FXR deficiency are resistant to spontaneous but susceptible to cholic acid-induced hepatocarcinogenesis. Am J Physiol Gastrointest Liver Physiol 2016; 310:G295-302. [PMID: 26744468 PMCID: PMC4773826 DOI: 10.1152/ajpgi.00134.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 12/12/2015] [Indexed: 01/31/2023]
Abstract
Farnesoid X receptor (FXR) belongs to the nuclear receptor superfamily with its endogenous ligands bile acids. Mice with whole body FXR deficiency develop liver tumors spontaneously, but the underlying mechanism is unclear. Moreover, it is unknown whether FXR deficiency in liver alone serves as a tumor initiator or promoter during liver carcinogenesis. This study aims to evaluate the effects of hepatocyte-specific FXR deficiency (FXR(hep-/-)) in liver tumor formation. The results showed that FXR(hep-/-) mice did not show spontaneous liver tumorigenesis with aging (up to 24 mo of age). Therefore FXR(hep-/-) mice were fed a bile acid (cholic acid)-containing diet alone or along with a liver tumor initiator, diethylnitrosamine (DEN). Thirty weeks later, no tumors were found in wild-type or FXR(hep-/-) mice without any treatment or with DEN only. However, with cholic acid, while only some wild-type mice developed tumors, all FXR(hep-/-) mice presented with severe liver injury and tumors. Interestingly, FXR(hep-/-) mouse livers increased basal expression of tumor suppressor p53 protein, apoptosis, and decreased basal cyclin D1 expression, which may prevent tumor development in FXR(hep-/-) mice. However, cholic acid feeding reversed these effects in FXR(hep-/-) mice, which is associated with an increased cyclin D1 and decreased cell cycle inhibitors. More in-depth analysis indicates that the increased in cell growth might result from disturbance of the MAPK and JAK/Stat3 signaling pathways. In conclusion, this study shows that hepatic FXR deficiency may only serve as a tumor initiator, and increased bile acids is required for tumor formation likely by promoting cell proliferation.
Collapse
Affiliation(s)
- Bo Kong
- 1Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey;
| | - Yan Zhu
- 2Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China;
| | - Guodong Li
- 3Department of General Surgery, the Fourth Hospital of Harbin Medical University, Harbin; ,4Division of Biobank Research, Department of General Surgery, the Fourth Hospital of Harbin Medical University, Harbin;
| | - Jessica A. Williams
- 5Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas;
| | - Kyle Buckley
- 1Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey;
| | - Ossama Tawfik
- 6Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas; and
| | - James P. Luyendyk
- 7Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Grace L. Guo
- 1Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey;
| |
Collapse
|
81
|
Mice Abundant in Muricholic Bile Acids Show Resistance to Dietary Induced Steatosis, Weight Gain, and to Impaired Glucose Metabolism. PLoS One 2016; 11:e0147772. [PMID: 26824238 PMCID: PMC4732983 DOI: 10.1371/journal.pone.0147772] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/07/2016] [Indexed: 11/19/2022] Open
Abstract
High endogenous production of, or treatment with muricholic bile acids, strongly reduces the absorption of cholesterol. Mice abundant in muricholic bile acids may therefore display an increased resistance against dietary induced weight gain, steatosis, and glucose intolerance due to an anticipated general reduction in lipid absorption. To test this hypothesis, mice deficient in steroid 12-alpha hydroxylase (Cyp8b1-/-) and therefore abundant in muricholic acids were monitored for 11 weeks while fed a high fat diet. Food intake and body and liver weights were determined, and lipids in liver, serum and feces were measured. Further, responses during oral glucose and intraperitoneal insulin tolerance tests were evaluated. On the high fat diet, Cyp8b1-/- mice displayed less weight gain compared to wildtype littermates (Cyp8b1+/+). In addition, liver enlargement with steatosis and increases in serum LDL-cholesterol were strongly attenuated in Cyp8b1-/- mice on high fat diet. Fecal excretion of cholesterol was increased and there was a strong trend for doubled fecal excretion of free fatty acids, while excretion of triglycerides was unaltered, indicating dampened lipid absorption. On high fat diet, Cyp8b1-/- mice also presented lower serum glucose levels in response to oral glucose gavage or to intraperitoneal insulin injection compared to Cyp8b1+/+. In conclusion, following exposure to a high fat diet, Cyp8b1-/- mice are more resistant against weight gain, steatosis, and to glucose intolerance than Cyp8b1+/+ mice. Reduced lipid absorption may in part explain these findings. Overall, the results suggest that muricholic bile acids may be beneficial against the metabolic syndrome.
Collapse
|
82
|
Zhan L, Yang I, Kong B, Shen J, Gorczyca L, Memon N, Buckley BT, Guo GL. Dysregulation of bile acid homeostasis in parenteral nutrition mouse model. Am J Physiol Gastrointest Liver Physiol 2016; 310:G93-G102. [PMID: 26564717 PMCID: PMC4719060 DOI: 10.1152/ajpgi.00252.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/07/2015] [Indexed: 01/31/2023]
Abstract
Long-term parenteral nutrition (PN) administration can lead to PN-associated liver diseases (PNALD). Although multiple risk factors have been identified for PNALD, to date, the roles of bile acids (BAs) and the pathways involved in BA homeostasis in the development and progression of PNALD are still unclear. We have established a mouse PN model with IV infusion of PN solution containing soybean oil-based lipid emulsion (SOLE). Our results showed that PN altered the expression of genes involved in a variety of liver functions at the mRNA levels. PN increased liver gene expression of Cyp7a1 and markedly decreased that of Cyp8b1, Cyp7b1, Bsep, and Shp. CYP7A1 and CYP8B1 are important for synthesizing the total amount of BAs and regulating the hydrophobicity of BAs, respectively. Consistently, both the levels and the percentages of primary BAs as well as total non-12α-OH BAs increased significantly in the serum of PN mice compared with saline controls, whereas liver BA profiles were largely similar. The expression of several key liver-X receptor-α (LXRα) target genes involved in lipid synthesis was also increased in PN mouse livers. Retinoid acid-related orphan receptor-α (RORα) has been shown to induce the expression of Cyp8b1 and Cyp7b1, as well as to suppress LXRα function. Western blot showed significantly reduced nuclear migration of RORα protein in PN mouse livers. This study shows that continuous PN infusion with SOLE in mice leads to dysregulation of BA homeostasis. Alterations of liver RORα signaling in PN mice may be one of the mechanisms implicated in the pathogenesis of PNALD.
Collapse
Affiliation(s)
- Le Zhan
- 1Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey;
| | - Ill Yang
- 2EOHSI/Chemical Analytical Core Laboratory, Rutgers University, Piscataway, New Jersey;
| | - Bo Kong
- 1Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey;
| | - Jianliang Shen
- 1Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey;
| | - Ludwik Gorczyca
- 1Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey;
| | - Naureen Memon
- 3Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Brian T. Buckley
- 2EOHSI/Chemical Analytical Core Laboratory, Rutgers University, Piscataway, New Jersey;
| | - Grace L. Guo
- 1Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey;
| |
Collapse
|
83
|
Bile Acid Signaling Is Involved in the Neurological Decline in a Murine Model of Acute Liver Failure. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:312-23. [PMID: 26683664 DOI: 10.1016/j.ajpath.2015.10.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 10/06/2015] [Accepted: 10/09/2015] [Indexed: 12/13/2022]
Abstract
Hepatic encephalopathy is a serious neurological complication of liver failure. Serum bile acids are elevated after liver damage and may disrupt the blood-brain barrier and enter the brain. Our aim was to assess the role of serum bile acids in the neurological complications after acute liver failure. C57Bl/6 or cytochrome p450 7A1 knockout (Cyp7A1(-/-)) mice were fed a control, cholestyramine-containing, or bile acid-containing diet before azoxymethane (AOM)-induced acute liver failure. In parallel, mice were given an intracerebroventricular infusion of farnesoid X receptor (FXR) Vivo-morpholino before AOM injection. Liver damage, neurological decline, and molecular analyses of bile acid signaling were performed. Total bile acid levels were increased in the cortex of AOM-treated mice. Reducing serum bile acids via cholestyramine feeding or using Cyp7A1(-/-) mice reduced bile acid levels and delayed AOM-induced neurological decline, whereas cholic acid or deoxycholic acid feeding worsened AOM-induced neurological decline. The expression of bile acid signaling machinery apical sodium-dependent bile acid transporter, FXR, and small heterodimer partner increased in the frontal cortex, and blocking FXR signaling delayed AOM-induced neurological decline. In conclusion, circulating bile acids may play a pathological role during hepatic encephalopathy, although precisely how they dysregulate normal brain function is unknown. Strategies to minimize serum bile acid concentrations may reduce the severity of neurological complications associated with liver failure.
Collapse
|
84
|
Selwyn FP, Csanaky IL, Zhang Y, Klaassen CD. Importance of Large Intestine in Regulating Bile Acids and Glucagon-Like Peptide-1 in Germ-Free Mice. Drug Metab Dispos 2015; 43:1544-56. [PMID: 26199423 PMCID: PMC4576674 DOI: 10.1124/dmd.115.065276] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/20/2015] [Indexed: 12/25/2022] Open
Abstract
It is known that 1) elevated serum bile acids (BAs) are associated with decreased body weight, 2) elevated glucagon-like peptide-1 (GLP-1) levels can decrease body weight, and 3) germ-free (GF) mice are resistant to diet-induced obesity. The purpose of this study was to test the hypothesis that a lack of intestinal microbiota results in more BAs in the body, resulting in increased BA-mediated transmembrane G protein-coupled receptor 5 (TGR5) signaling and increased serum GLP-1 as a mechanism of resistance of GF mice to diet-induced obesity. GF mice had 2- to 4-fold increased total BAs in the serum, liver, bile, and ileum. Fecal excretion of BAs was 63% less in GF mice. GF mice had decreased secondary BAs and increased taurine-conjugated BAs, as anticipated. Surprisingly, there was an increase in non-12α-OH BAs, namely, β-muricholic acid, ursodeoxycholic acid (UDCA), and their taurine conjugates, in GF mice. Further, in vitro experiments confirmed that UDCA is a primary BA in mice. There were minimal changes in the mRNA of farnesoid X receptor target genes in the ileum (Fibroblast growth factor 15, small heterodimer protein, and ileal bile acid-binding protein), in the liver (small heterodimer protein, liver receptor homolog-1, and cytochrome P450 7a1), and BA transporters (apical sodium dependent bile acid transporter, organic solute transporter α, and organic solute transporter β) in the ileum of GF mice. Surprisingly, there were marked increases in BA transporters in the large intestine. Increased GLP-1 levels and gallbladder size were observed in GF mice, suggesting activation of TGR5 signaling. In summary, the GF condition results in increased expression of BA transporters in the colon, resulting in 1) an increase in total BA concentrations in tissues, 2) a change in BA composition to favor an increase in non-12α-OH BAs, and 3) activation of TGR5 signaling with increased gallbladder size and GLP-1.
Collapse
Affiliation(s)
- Felcy Pavithra Selwyn
- Departments of Pharmacology, Toxicology, and Therapeutics (F.P.S., Y.Z.) and Internal Medicine (I.L.C., C.D.K.), University of Kansas Medical Center, Kansas City, Kansas
| | - Iván L Csanaky
- Departments of Pharmacology, Toxicology, and Therapeutics (F.P.S., Y.Z.) and Internal Medicine (I.L.C., C.D.K.), University of Kansas Medical Center, Kansas City, Kansas
| | - Youcai Zhang
- Departments of Pharmacology, Toxicology, and Therapeutics (F.P.S., Y.Z.) and Internal Medicine (I.L.C., C.D.K.), University of Kansas Medical Center, Kansas City, Kansas
| | - Curtis D Klaassen
- Departments of Pharmacology, Toxicology, and Therapeutics (F.P.S., Y.Z.) and Internal Medicine (I.L.C., C.D.K.), University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
85
|
Fu ZD, Cui JY, Klaassen CD. The Role of Sirt1 in Bile Acid Regulation during Calorie Restriction in Mice. PLoS One 2015; 10:e0138307. [PMID: 26372644 PMCID: PMC4570809 DOI: 10.1371/journal.pone.0138307] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 08/28/2015] [Indexed: 12/11/2022] Open
Abstract
Sirtuin 1 (Sirt1) is an NAD+-dependent protein deacetylase that is proposed to mediate many health-promoting effects of calorie restriction (CR). We recently reported that short-term CR increased the bile acid (BA) pool size in mice, likely due to increased BA synthesis in liver. Given the important role of Sirt1 in the regulation of glucose, lipid, as well as BA metabolism, we hypothesized that the CR-induced increase in BAs is Sirt1-dependent. To address this, the present study utilized genetically-modified mice that were Sirt1 loss of function (liver knockout, LKO) or Sirt1 gain of function (whole body-transgenic, TG). Three genotypes of mice (Sirt1-LKO, wild-type, and Sirt1-TG) were each randomly divided into ad libitum or 40% CR feeding for one month. BAs were extracted from various compartments of the enterohepatic circulation, followed by BA profiling by UPLC-MS/MS. CR increased the BA pool size and total BAs in serum, gallbladder, and small intestine. The CR-induced increase in BA pool size correlated with the tendency of increase in the expression of the rate-limiting BA-synthetic enzyme Cyp7a1. However, in contrast to the hypothesis, the CR-induced increase in BA pool size and Cyp7a1 expression was still observed with ablated expression of Sirt1 in liver, and completely suppressed with whole-body overexpression of Sirt1. Furthermore, in terms of BA composition, CR increased the ratio of 12α-hydroxylated BAs regardless of Sirt1 genotypes. In conclusion, the CR-induced alterations in BA pool size, BA profiles, and expression of BA-related genes do not appear to be dependent on Sirt1.
Collapse
Affiliation(s)
- Zidong Donna Fu
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States of America
- Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, Heilongjiang Province, People's Republic of China, 150081
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States of America
| | - Curtis D. Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| |
Collapse
|
86
|
Saracut C, Molnar C, Russu C, Todoran N, Vlase L, Turdean S, Voidazan S, Copotoiu C. Secondary bile acids effects in colon pathology. Experimental mice study. Acta Cir Bras 2015; 30:624-31. [DOI: 10.1590/s0102-865020150090000007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/25/2015] [Indexed: 12/12/2022] Open
Affiliation(s)
| | - Calin Molnar
- University of Medicine and Pharmacy Tg-Mures, Romania
| | | | | | - Laurian Vlase
- University of Medicine and Pharmacy Cluj Napoca, Romania
| | - Sabin Turdean
- University of Medicine and Pharmacy Cluj Napoca, Romania
| | | | | |
Collapse
|
87
|
Rudling M, Bonde Y. Stimulation of apical sodium-dependent bile acid transporter expands the bile acid pool and generates bile acids with positive feedback properties. Dig Dis 2015; 33:376-81. [PMID: 26045272 DOI: 10.1159/000371690] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Bile acid synthesis has been considered a prototype for how a physiological process is controlled by end product feedback inhibition. By this feedback inhibition, bile acid concentrations are kept within safe ranges. However, careful examination of published rodent data strongly suggests that bile acid synthesis is also under potent positive feedback control by hydrophilic bile acids. KEY MESSAGES Current concepts on the regulation of bile acid synthesis are derived from mouse models. Recent data have shown that mice have farnesoid X receptor (FXR) antagonistic bile acids capable of quenching responses elicited by FXR agonistic bile acids. This is important to recognize to understand the regulation of bile acid synthesis in the mouse, and in particular to clarify if mouse model findings are valid also in the human situation. CONCLUSIONS In addition to classic end product feedback inhibition, regulation of bile acid synthesis in the mouse largely appears also to be driven by changes in hepatic levels of murine bile acids such as α- and β-muricholic acids. This has not been previously recognized. Stimulated bile acid synthesis or induction of the apical sodium-dependent bile acid transporter in the intestine, increase the availability of chenodeoxycholic acid in the liver, thereby promoting hepatic conversion of this bile acid into muricholic acids. Recognition of these mechanisms is essential for understanding the regulation of bile acid synthesis in the mouse, and for our awareness of important species differences in the regulation of bile acid synthesis in mice and humans.
Collapse
Affiliation(s)
- Mats Rudling
- Metabolism Unit, Department of Endocrinology, Metabolism and Diabetes, KI/AZ Integrated CardioMetabolic Center, Department of Medicine, and Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
88
|
Huang F, Wang T, Lan Y, Yang L, Pan W, Zhu Y, Lv B, Wei Y, Shi H, Wu H, Zhang B, Wang J, Duan X, Hu Z, Wu X. Deletion of mouse FXR gene disturbs multiple neurotransmitter systems and alters neurobehavior. Front Behav Neurosci 2015; 9:70. [PMID: 25870546 PMCID: PMC4378301 DOI: 10.3389/fnbeh.2015.00070] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 03/03/2015] [Indexed: 12/14/2022] Open
Abstract
Farnesoid X receptor (FXR) is a nuclear hormone receptor involved in bile acid synthesis and homeostasis. Dysfunction of FXR is involved in cholestasis and atherosclerosis. FXR is prevalent in liver, gallbladder, and intestine, but it is not yet clear whether it modulates neurobehavior. In the current study, we tested the hypothesis that mouse FXR deficiency affects a specific subset of neurotransmitters and results in an unique behavioral phenotype. The FXR knockout mice showed less depressive-like and anxiety-related behavior, but increased motor activity. They had impaired memory and reduced motor coordination. There were changes of glutamatergic, GABAergic, serotoninergic, and norepinephrinergic neurotransmission in either hippocampus or cerebellum. FXR deletion decreased the amount of the GABA synthesis enzyme GAD65 in hippocampus but increased GABA transporter GAT1 in cerebral cortex. FXR deletion increased serum concentrations of many bile acids, including taurodehydrocholic acid, taurocholic acid, deoxycholic acid (DCA), glycocholic acid (GCA), tauro-α-muricholic acid, tauro-ω-muricholic acid, and hyodeoxycholic acid (HDCA). There were also changes in brain concentrations of taurocholic acid, taurodehydrocholic acid, tauro-ω-muricholic acid, tauro-β-muricholic acid, deoxycholic acid, and lithocholic acid (LCA). Taken together, the results from studies with FXR knockout mice suggest that FXR contributes to the homeostasis of multiple neurotransmitter systems in different brain regions and modulates neurobehavior. The effect appears to be at least partially mediated by bile acids that are known to cross the blood-brain barrier (BBB) inducing potential neurotoxicity.
Collapse
Affiliation(s)
- Fei Huang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Tingting Wang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Yunyi Lan
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Li Yang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Weihong Pan
- Blood-Brain Barrier Group, Pennington Biomedical Research Center Baton Rouge, LA, USA
| | - Yonghui Zhu
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Boyang Lv
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Yuting Wei
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Hailian Shi
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Hui Wu
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Beibei Zhang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Jie Wang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Xiaofeng Duan
- Pharmacy Department, Shanghai East Hospital Shanghai, China
| | - Zhibi Hu
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Xiaojun Wu
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, the State Administration of TCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine Shanghai, China
| |
Collapse
|
89
|
Song P, Rockwell CE, Cui JY, Klaassen CD. Individual bile acids have differential effects on bile acid signaling in mice. Toxicol Appl Pharmacol 2015; 283:57-64. [PMID: 25582706 DOI: 10.1016/j.taap.2014.12.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 12/10/2014] [Accepted: 12/15/2014] [Indexed: 01/22/2023]
Abstract
Bile acids (BAs) are known to regulate BA synthesis and transport by the farnesoid X receptor in the liver (FXR-SHP) and intestine (FXR-Fgf15). However, the relative importance of individual BAs in regulating these processes is not known. Therefore, mice were fed various doses of five individual BAs, including cholic acid (CA), chenodeoxycholic acid (CDCA), deoxoycholic acid (DCA), lithocholic acid (LCA), and ursodeoxycholic acid (UDCA) in their diets at various concentrations for one week to increase the concentration of one BA in the enterohepatic circulation. The mRNA of BA synthesis and transporting genes in liver and ileum were quantified. In the liver, the mRNA of SHP, which is the prototypical target gene of FXR, increased in mice fed all concentrations of BAs. In the ileum, the mRNA of the intestinal FXR target gene Fgf15 was increased at lower doses and to a higher extent by CA and DCA than by CDCA and LCA. Cyp7a1, the rate-limiting enzyme in BA synthesis, was decreased more by CA and DCA than CDCA and LCA. Cyp8b1, the enzyme that 12-hydroxylates BAs and is thus responsible for the synthesis of CA, was decreased much more by CA and DCA than CDCA and LCA. Surprisingly, neither a decrease in the conjugated BA uptake transporter (Ntcp) nor increase in BA efflux transporter (Bsep) was observed by FXR activation, but an increase in the cholesterol efflux transporter (Abcg5/Abcg8) was observed with FXR activation. Thus in conclusion, CA and DCA are more potent FXR activators than CDCA and LCA when fed to mice, and thus they are more effective in decreasing the expression of the rate limiting gene in BA synthesis Cyp7a1 and the 12-hydroxylation of BAs Cyp8b1, and are also more effective in increasing the expression of Abcg5/Abcg8, which is responsible for biliary cholesterol excretion. However, feeding BAs do not alter the mRNA or protein levels of Ntcp or Bsep, suggesting that the uptake or efflux of BAs is not regulated by FXR at physiological and pharmacological concentrations of BAs.
Collapse
Affiliation(s)
- Peizhen Song
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA.
| | - Cheryl E Rockwell
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA.
| | - Julia Yue Cui
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA.
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA.
| |
Collapse
|
90
|
Jones RD, Lopez AM, Tong EY, Posey KS, Chuang JC, Repa JJ, Turley SD. Impact of physiological levels of chenodeoxycholic acid supplementation on intestinal and hepatic bile acid and cholesterol metabolism in Cyp7a1-deficient mice. Steroids 2015; 93:87-95. [PMID: 25447797 PMCID: PMC4297738 DOI: 10.1016/j.steroids.2014.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/07/2014] [Indexed: 01/07/2023]
Abstract
Mice deficient in cholesterol 7α-hydroxylase (Cyp7a1) have a diminished bile acid pool (BAP) and therefore represent a useful model for investigating the metabolic effects of restoring the pool with a specific BA. Previously we carried out such studies in Cyp7a1(-/-) mice fed physiological levels of cholic acid (CA) and achieved BAP restoration, along with an increased CA enrichment, at a dietary level of just 0.03% (w/w). Here we demonstrate that in Cyp7a1(-/-) mice fed chenodeoxycholic acid (CDCA) at a level of 0.06% (w/w), the BAP was restored to normal size and became substantially enriched with muricholic acid (MCA) (>70%), leaving the combined contribution of CA and CDCA to be <15%. This resulted in a partial to complete reversal of the main changes in cholesterol and BA metabolism associated with Cyp7a1 deficiency such as an elevated rate of intestinal sterol synthesis, an enhanced level of mRNA for Cyp8b1 in the liver, and depressed mRNA levels for Ibabp, Shp and Fgf15 in the distal small intestine. When Cyp7a1(-/-) and matching Cyp7a1(+/+) mice were fed a diet with added cholesterol (0.2%) (w/w), either alone, or also containing CDCA (0.06%) (w/w) or CA (0.03%) (w/w) for 18days, the hepatic total cholesterol concentrations (mg/g) in the Cyp7a1(-/-) mice were 26.9±3.7, 16.4±0.9 and 47.6±1.9, respectively, vs. 4.9±0.4, 5.0±0.7 and 6.4±1.9, respectively in the corresponding Cyp7a1(+/+) controls. These data affirm the importance of using moderate levels of dietary BA supplementation to elicit changes in hepatic cholesterol metabolism through shifts in BAP size and composition.
Collapse
Affiliation(s)
- Ryan D Jones
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| | - Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| | - Ernest Y Tong
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| | - Kenneth S Posey
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| | - Jen-Chieh Chuang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| | - Joyce J Repa
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States; Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| |
Collapse
|
91
|
Fu ZD, Cui JY, Klaassen CD. Atorvastatin induces bile acid-synthetic enzyme Cyp7a1 by suppressing FXR signaling in both liver and intestine in mice. J Lipid Res 2014; 55:2576-86. [PMID: 25278499 DOI: 10.1194/jlr.m053124] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Statins are effective cholesterol-lowering drugs to treat CVDs. Bile acids (BAs), the end products of cholesterol metabolism in the liver, are important nutrient and energy regulators. The present study aims to investigate how statins affect BA homeostasis in the enterohepatic circulation. Male C57BL/6 mice were treated with atorvastatin (100 mg/kg/day po) for 1 week, followed by BA profiling by ultra-performance LC-MS/MS. Atorvastatin decreased BA pool size, mainly due to less BA in the intestine. Surprisingly, atorvastatin did not alter total BAs in the serum or liver. Atorvastatin increased the ratio of 12α-OH/non12α-OH BAs. Atorvastatin increased the mRNAs of the BA-synthetic enzymes cholesterol 7α-hydroxylase (Cyp7a1) (over 10-fold) and cytochrome P450 27a1, the BA uptake transporters Na⁺/taurocholate cotransporting polypeptide and organic anion transporting polypeptide 1b2, and the efflux transporter multidrug resistance-associated protein 2 in the liver. Noticeably, atorvastatin suppressed the expression of BA nuclear receptor farnesoid X receptor (FXR) target genes, namely small heterodimer partner (liver) and fibroblast growth factor 15 (ileum). Furthermore, atorvastatin increased the mRNAs of the organic cation uptake transporter 1 and cholesterol efflux transporters Abcg5 and Abcg8 in the liver. The increased expression of BA-synthetic enzymes and BA transporters appear to be a compensatory response to maintain BA homeostasis after atorvastatin treatment. The Cyp7a1 induction by atorvastatin appears to be due to suppressed FXR signaling in both the liver and intestine.
Collapse
Affiliation(s)
- Zidong Donna Fu
- Departments of Pharmacology, Toxicology, and Therapeutics University of Kansas Medical Center, Kansas City, KS 66160 Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, Heilongjiang Province, People's Republic of China 150081
| | - Julia Yue Cui
- Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Curtis D Klaassen
- Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| |
Collapse
|
92
|
Dawson PA, Karpen SJ. Intestinal transport and metabolism of bile acids. J Lipid Res 2014; 56:1085-99. [PMID: 25210150 DOI: 10.1194/jlr.r054114] [Citation(s) in RCA: 379] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Indexed: 12/17/2022] Open
Abstract
In addition to their classical roles as detergents to aid in the process of digestion, bile acids have been identified as important signaling molecules that function through various nuclear and G protein-coupled receptors to regulate a myriad of cellular and molecular functions across both metabolic and nonmetabolic pathways. Signaling via these pathways will vary depending on the tissue and the concentration and chemical structure of the bile acid species. Important determinants of the size and composition of the bile acid pool are their efficient enterohepatic recirculation, their host and microbial metabolism, and the homeostatic feedback mechanisms connecting hepatocytes, enterocytes, and the luminal microbiota. This review focuses on the mammalian intestine, discussing the physiology of bile acid transport, the metabolism of bile acids in the gut, and new developments in our understanding of how intestinal metabolism, particularly by the gut microbiota, affects bile acid signaling.
Collapse
Affiliation(s)
- Paul A Dawson
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322
| | - Saul J Karpen
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322
| |
Collapse
|
93
|
Liu J, Lu H, Lu YF, Lei X, Cui JY, Ellis E, Strom SC, Klaassen CD. Potency of individual bile acids to regulate bile acid synthesis and transport genes in primary human hepatocyte cultures. Toxicol Sci 2014; 141:538-46. [PMID: 25055961 DOI: 10.1093/toxsci/kfu151] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Bile acids (BAs) are known to regulate their own homeostasis, but the potency of individual bile acids is not known. This study examined the effects of cholic acid (CA), chenodeoxycholic acid (CDCA), deoxycholic acid (DCA), lithocholic acid (LCA) and ursodeoxycholic acid (UDCA) on expression of BA synthesis and transport genes in human primary hepatocyte cultures. Hepatocytes were treated with the individual BAs at 10, 30, and 100μM for 48 h, and RNA was extracted for real-time PCR analysis. For the classic pathway of BA synthesis, BAs except for UDCA markedly suppressed CYP7A1 (70-95%), the rate-limiting enzyme of bile acid synthesis, but only moderately (35%) down-regulated CYP8B1 at a high concentration of 100μM. BAs had minimal effects on mRNA of two enzymes of the alternative pathway of BA synthesis, namely CYP27A1 and CYP7B1. BAs increased the two major target genes of the farnesoid X receptor (FXR), namely the small heterodimer partner (SHP) by fourfold, and markedly induced fibroblast growth factor 19 (FGF19) over 100-fold. The BA uptake transporter Na(+)-taurocholate co-transporting polypeptide was unaffected, whereas the efflux transporter bile salt export pump was increased 15-fold and OSTα/β were increased 10-100-fold by BAs. The expression of the organic anion transporting polypeptide 1B3 (OATP1B3; sixfold), ATP-binding cassette (ABC) transporter G5 (ABCG5; sixfold), multidrug associated protein-2 (MRP2; twofold), and MRP3 (threefold) were also increased, albeit to lesser degrees. In general, CDCA was the most potent and effective BA in regulating these genes important for BA homeostasis, whereas DCA and CA were intermediate, LCA the least, and UDCA ineffective.
Collapse
Affiliation(s)
- Jie Liu
- University of Kansas Medical Center, Kansas City, Kansas 66160 Zunyi Medical College, Zunyi, China
| | - Hong Lu
- University of Kansas Medical Center, Kansas City, Kansas 66160 Upstate Medical University, Syracuse, New York 13210
| | - Yuan-Fu Lu
- University of Kansas Medical Center, Kansas City, Kansas 66160 Zunyi Medical College, Zunyi, China
| | - Xiaohong Lei
- University of Kansas Medical Center, Kansas City, Kansas 66160 Upstate Medical University, Syracuse, New York 13210
| | - Julia Yue Cui
- University of Kansas Medical Center, Kansas City, Kansas 66160
| | | | - Stephen C Strom
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden University of Pittsburgh Medical Center, Pittsburgh, PA USA
| | | |
Collapse
|
94
|
Raveendran VV, Kassel KM, Smith DD, Luyendyk JP, Williams KJ, Cherian R, Reed GA, Flynn CA, Csanaky IL, Lickteig AL, Pratt-Hyatt MJ, Klaassen CD, Dileepan KN. H1-antihistamines exacerbate high-fat diet-induced hepatic steatosis in wild-type but not in apolipoprotein E knockout mice. Am J Physiol Gastrointest Liver Physiol 2014; 307:G219-28. [PMID: 24852568 PMCID: PMC4101675 DOI: 10.1152/ajpgi.00027.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We examined the effects of two over-the-counter H1-antihistamines on the progression of fatty liver disease in male C57Bl/6 wild-type and apolipoprotein E (ApoE)-/- mice. Mice were fed a high-fat diet (HFD) for 3 mo, together with administration of either cetirizine (4 mg/kg body wt) or fexofenadine (40 mg/kg body wt) in drinking water. Antihistamine treatments increased body weight gain, gonadal fat deposition, liver weight, and hepatic steatosis in wild-type mice but not in ApoE-/- mice. Lobular inflammation, acute inflammation, and necrosis were not affected by H1-antihistamines in either genotype. Serum biomarkers of liver injury tended to increase in antihistamine-treated wild-type mice. Serum level of glucose was increased by fexofenadine, whereas lipase was increased by cetirizine. H1-antihistamines reduced the mRNA expression of ApoE and carbohydrate response element-binding protein in wild-type mice, without altering the mRNA expression of sterol regulatory element-binding protein 1c, fatty acid synthase, or ApoB100, in either genotype. Fexofenadine increased both triglycerides and cholesterol ester, whereas cetirizine increased only cholesterol ester in liver, with a concomitant decrease in serum triglycerides by both antihistamines in wild-type mice. Antihistamines increased hepatic levels of conjugated bile acids in wild-type mice, with the effect being significant in fexofenadine-treated animals. The increase was associated with changes in the expression of organic anion transport polypeptide 1b2 and bile salt export pump. These results suggest that H1-antihistamines increase the progression of fatty liver disease in wild-type mice, and there seems to be an association between the severity of disease, presence of ApoE, and increase in hepatic bile acid levels.
Collapse
Affiliation(s)
| | | | - Donald D. Smith
- 1Division of Allergy, Clinical Immunology, and Rheumatology,
| | - James P. Luyendyk
- 2Department of Pharmacology, Toxicology, and Therapeutics, ,5Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Kurt J. Williams
- 5Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Rachel Cherian
- 4Veterans Affairs Medical Center, Kansas City, Missouri;
| | | | | | - Iván L. Csanaky
- 3Division of Gastroenterology, Hepatology, and Motility, Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas;
| | - Andrew L. Lickteig
- 3Division of Gastroenterology, Hepatology, and Motility, Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas;
| | - Matthew J. Pratt-Hyatt
- 3Division of Gastroenterology, Hepatology, and Motility, Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas;
| | - Curtis D. Klaassen
- 3Division of Gastroenterology, Hepatology, and Motility, Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas;
| | | |
Collapse
|
95
|
Fickert P, Pollheimer MJ, Beuers U, Lackner C, Hirschfield G, Housset C, Keitel V, Schramm C, Marschall HU, Karlsen TH, Melum E, Kaser A, Eksteen B, Strazzabosco M, Manns M, Trauner M, for the International PSC Study Group (IPSCSG). Characterization of animal models for primary sclerosing cholangitis (PSC). J Hepatol 2014; 60:1290-303. [PMID: 24560657 PMCID: PMC4517670 DOI: 10.1016/j.jhep.2014.02.006] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/01/2014] [Accepted: 02/08/2014] [Indexed: 01/17/2023]
Abstract
Primary sclerosing cholangitis (PSC) is a chronic cholangiopathy characterized by biliary fibrosis, development of cholestasis and end stage liver disease, high risk of malignancy, and frequent need for liver transplantation. The poor understanding of its pathogenesis is also reflected in the lack of effective medical treatment. Well-characterized animal models are utterly needed to develop novel pathogenetic concepts and study new treatment strategies. Currently there is no consensus on how to evaluate and characterize potential PSC models, which makes direct comparison of experimental results and effective exchange of study material between research groups difficult. The International Primary Sclerosing Cholangitis Study Group (IPSCSG) has therefore summarized these key issues in a position paper proposing standard requirements for the study of animal models of PSC.
Collapse
Affiliation(s)
- Peter Fickert
- Research Unit for Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria; Institute of Pathology, Medical University of Graz, Austria.
| | - Marion J. Pollheimer
- Research Unit for Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria,Institute of Pathology, Medical University of Graz, Austria
| | - Ulrich Beuers
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Centre, University of Amsterdam, The Netherlands
| | | | - Gideon Hirschfield
- Centre for Liver Research, Institute of Biomedical Research, School of Immunity and Infection, University of Birmingham, UK
| | - Chantal Housset
- UPMC Univ Paris 06 & INSERM, UMR-S 938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| | - Verena Keitel
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University Düsseldorf Germany
| | | | - Hanns-Ulrich Marschall
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, The Sahlgrenska Academy, Sweden
| | - Tom H. Karlsen
- Division of Gastroenterology and Hepatology, Department of Medicine, Rikshospitalet, Oslo, Norway,Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway,Research Institute of Internal Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway,Division of Gastroenterology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Espen Melum
- Division of Gastroenterology and Hepatology, Department of Medicine, Rikshospitalet, Oslo, Norway,Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway,Research Institute of Internal Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Addenbrooek's Hospital, UK
| | - Bertus Eksteen
- Centre for Liver Research, MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, and The Queen Elizabeth Hospital, University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - Mario Strazzabosco
- Section of Gastroenterology, University of Milan-Bicocca, Milan, Italy,Liver Center, Yale University School of Medicine, United States
| | - Michael Manns
- Division of Gastroenterology, Hepatology and Endocrinology, Medical University Hannover, Germany
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria.
| | | |
Collapse
|
96
|
Bile acid supplementation improves established liver steatosis in obese mice independently of glucagon-like peptide-1 secretion. J Physiol Biochem 2014; 70:667-74. [DOI: 10.1007/s13105-014-0336-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 04/23/2014] [Indexed: 12/17/2022]
|
97
|
Woolbright BL, Li F, Xie Y, Farhood A, Fickert P, Trauner M, Jaeschke H. Lithocholic acid feeding results in direct hepato-toxicity independent of neutrophil function in mice. Toxicol Lett 2014; 228:56-66. [PMID: 24742700 DOI: 10.1016/j.toxlet.2014.04.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/01/2014] [Accepted: 04/02/2014] [Indexed: 02/07/2023]
Abstract
Lithocholic acid (LCA) supplementation in the diet results in intrahepatic cholestasis and bile infarcts. Previously we showed that an innate immune response is critical for cholestatic liver injury in the bile duct ligated mice. Thus, the purpose of this study was to investigate the role of neutrophils in the mechanism of liver injury caused by feeding mice a diet containing LCA. C57BL/6 mice were given control or 1% LCA containing diet for 24-96 h and then examined for parameters of hepatotoxicity. Plasma ALT levels were significantly increased by 48 h after LCA feeding, which correlated with both neutrophil recruitment to the liver and upregulation of numerous pro-inflammatory genes. The injury was confirmed by histology. Deficiency in intercellular adhesion molecule-1 (ICAM-1) expression or inhibition of neutrophil function failed to protect against the injury. Bile acid levels were quantified in plasma and bile of LCA-fed mice after 48 and 96 h. Only the observed biliary levels of taurochenodeoxycholic acid and potentially tauro-LCA caused direct cytotoxicity in mouse hepatocytes. These data support the conclusion that neutrophil recruitment occurs after the onset of bile acid-induced necrosis in LCA-fed animals, and is not a primary mechanism of cell death when cholestasis occurs through accumulation of hydrophobic bile acids.
Collapse
Affiliation(s)
- Benjamin L Woolbright
- Department of Pharmacology, Toxicology & Therapeutics, Kansas University Medical Center, USA
| | - Feng Li
- Department of Pharmacology, Toxicology & Therapeutics, Kansas University Medical Center, USA
| | - Yuchao Xie
- Department of Pharmacology, Toxicology & Therapeutics, Kansas University Medical Center, USA
| | - Anwar Farhood
- Department of Pathology, St. David's North Austin Medical Center, Austin, TX 78756, USA
| | - Peter Fickert
- Research Unit for Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine, Graz, Austria; Department of Pathology, Medical University of Graz, Graz, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, Kansas University Medical Center, USA.
| |
Collapse
|
98
|
Zhang Y, Limaye PB, Renaud HJ, Klaassen CD. Effect of various antibiotics on modulation of intestinal microbiota and bile acid profile in mice. Toxicol Appl Pharmacol 2014; 277:138-45. [PMID: 24657338 DOI: 10.1016/j.taap.2014.03.009] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/10/2014] [Accepted: 03/12/2014] [Indexed: 12/24/2022]
Abstract
Antibiotic treatments have been used to modulate intestinal bacteria and investigate the role of intestinal bacteria on bile acid (BA) homeostasis. However, knowledge on which intestinal bacteria and bile acids are modified by antibiotics is limited. In the present study, mice were administered various antibiotics, 47 of the most abundant bacterial species in intestine, as well as individual BAs in plasma, liver, and intestine were quantified. Compared to the two antibiotic combinations (vancomycin+imipenem and cephalothin+neomycin), the three single antibiotics (metronidazole, ciprofloxacin and aztreonam) have less effect on intestinal bacterial profiles, and thus on host BA profiles and mRNA expression of genes that are important for BA homeostasis. The two antibiotic combinations decreased the ratio of Firmicutes to Bacteroidetes in intestine, as well as most secondary BAs in serum, liver and intestine. Additionally, the two antibiotic combinations significantly increased mRNA of the hepatic BA uptake transporters (Ntcp and Oatp1b2) and canalicular BA efflux transporters (Bsep and Mrp2), but decreased mRNA of the hepatic BA synthetic enzyme Cyp8b1, suggesting an elevated enterohepatic circulation of BAs. Interestingly, the two antibiotic combinations tended to have opposite effect on the mRNAs of most intestinal genes, which tended to be inhibited by vancomycin+imipenem but stimulated by cephalothin+neomycin. To conclude, the present study clearly shows that various antibiotics have distinct effects on modulating intestinal bacteria and host BA metabolism.
Collapse
Affiliation(s)
- Youcai Zhang
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Pallavi B Limaye
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Helen J Renaud
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| |
Collapse
|
99
|
Hu X, Bonde Y, Eggertsen G, Rudling M. Muricholic bile acids are potent regulators of bile acid synthesis via a positive feedback mechanism. J Intern Med 2014; 275:27-38. [PMID: 24118394 DOI: 10.1111/joim.12140] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Bile acid (BA) synthesis is regulated by negative feedback end-product inhibition, initiated by farnesoid X receptors (FXRs) in liver and gut. Studies on cholic acid (CA)-free Cyp8b1(-/-) mice have concluded that CA is a potent suppressor of BA synthesis. Cyp8b1(-/-) mice have increased BA synthesis and an enlarged BA pool, a phenotype shared with bile-duct-ligated, antibiotics-administered and with germ-free mice. Studies on such mice have concluded BA synthesis is induced due to reduced hormonal signalling by fibroblast growth factor (FGF)15 from intestine to liver. A mutual finding in these models is that potent FXR-agonistic BAs are reduced. We hypothesized that the absence of the potent FXR agonist deoxycholic acid (DCA) may be important for the induction of BA synthesis in these situations. DESIGN Two of these models were investigated, antibiotic treatment and Cyp8b1(-/-) mice and their combination. Secondary BA formation was inhibited by ampicillin (AMP) given to wild-type and Cyp8b1(-/-) mice. We then administered CA, chenodeoxycholic acid (CDCA) or DCA to AMP-treated Cyp8b1(-/-) mice. RESULTS Our data show that the phenotype of AMP-treated wild-type mice resembles that of Cyp8b1(-/-) mice with fourfold induced Cyp7a1 expression, increased intestinal apical sodium-dependent BA transporter expression and increased hepatic BA levels. We also show that reductions in the FXR-agonistic BAs CDCA, CA, DCA or lithocholic acid cannot explain this phenotype; instead, it is likely due to increases in levels of α- and β-muricholic BAs and ursodeoxycholic acid, three FXR-antagonistic BAs. CONCLUSIONS Our findings reveal a potent positive feedback mechanism for regulation of BA synthesis in mice that appears to be sufficient without endocrine effects of FGF15 on Cyp7a1. This mechanism will be fundamental in understanding BA metabolism in both mice and humans.
Collapse
Affiliation(s)
- X Hu
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | | |
Collapse
|
100
|
Pathak P, Li T, Chiang JYL. Retinoic acid-related orphan receptor α regulates diurnal rhythm and fasting induction of sterol 12α-hydroxylase in bile acid synthesis. J Biol Chem 2013; 288:37154-65. [PMID: 24226095 DOI: 10.1074/jbc.m113.485987] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sterol 12α-hydroxylase (CYP8B1) is required for cholic acid synthesis and plays a critical role in intestinal cholesterol absorption and pathogenesis of cholesterol gallstone, dyslipidemia, and diabetes. In this study we investigated the underlying mechanism of fasting induction and circadian rhythm of CYP8B1 by a cholesterol-activated nuclear receptor and core clock gene retinoic acid-related orphan receptor α (RORα). Fasting stimulated, whereas restricted-feeding reduced expression of CYP8B1 mRNA and protein. However, fasting and feeding had little effect on the diurnal rhythm of RORα mRNA expression, but fasting increased RORα protein levels by cAMP-activated protein kinase A-mediated phosphorylation and stabilization of the protein. Adenovirus-mediated gene transduction of RORα to mice strongly induced CYP8B1 expression, and increased liver cholesterol and 12α-hydroxylated bile acids in the bile acid pool and serum. A reporter assay identified a functional RORα response element in the CYP8B1 promoter. RORα recruited cAMP response element-binding protein-binding protein (CBP) to stimulate histone acetylation on the CYP8B1 gene promoter. In conclusion, RORα is a key regulator of diurnal rhythm and fasting induction of CYP8B1, which regulates bile acid composition and serum and liver cholesterol levels. Antagonizing RORα activity may be a therapeutic strategy for treating inflammatory diseases such as non-alcoholic fatty liver disease and type 2 diabetes.
Collapse
Affiliation(s)
- Preeti Pathak
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio 44272 and
| | | | | |
Collapse
|