1
|
Qin W, Zhang T, Ge M, Zhou H, Xu Y, Mu R, Huang C, Liu D, Huang B, Wang Q, Kong Q, Kong Q, Li F, Xiong W. Hepatic RACK1 deletion disturbs lipid and glucose homeostasis independently of insulin resistance. J Endocrinol 2022; 254:137-151. [PMID: 35608066 DOI: 10.1530/joe-22-0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 05/23/2022] [Indexed: 11/08/2022]
Abstract
Receptor for activated C kinase 1 (RACK1) is a versatile protein involved in multiple biological processes. In a previous study by Zhao et al., hepatic RACK1 deletion in mice led to an inhibition of autophagy, blocked autophagy-dependent lipolysis, and caused steatosis. Using the same mouse model (RACK1hep-/-), we revealed new roles of RACK1 in maintaining bile acid homeostasis and hepatic glucose uptake, which further affected circulatory lipid and glucose levels. To be specific, even under hepatic steatosis, the plasma lipids were generally reduced in RACK1hep-/- mouse, which was due to the suppression of intestinal lipid absorption. Accordingly, a decrease in total bile acid level was found in RACK1hep-/- livers, gallbladders, and small intestine tissues, and specific decrease of 12-hydroxylated bile acids was detected by liquid chromatography-mass spectrometry. Consistently, reduced expression of CYP8B1 was found. A decrease in hepatic glycogen storage was also observed, which might be due to the inhibited glucose uptake by GLUT2 insufficiency. Interestingly, RACK1-KO-inducing hepatic steatosis did not raise insulin resistance (IR) nor IR-inducing factors like endoplasmic reticulum stress and inflammation. In summary, this study uncovers that hepatic RACK1 might be required in maintaining bile acid homeostasis and glucose uptake in hepatocytes. This study also provides an additional case of hepatic steatosis disassociation with insulin resistance.
Collapse
Affiliation(s)
- Wanying Qin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
- University of the Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Ting Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
- University of the Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Mingxia Ge
- University of the Chinese Academy of Sciences, Beijing, People's Republic of China
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
| | - Huimin Zhou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
| | - Yuhui Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
| | - Rongfang Mu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
| | - Chaoguang Huang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, People's Republic of China
| | - Daowei Liu
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, People's Republic of China
| | - Bangrui Huang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
- University of the Chinese Academy of Sciences, Beijing, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, People's Republic of China
| | - Qian Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, People's Republic of China
| | - Qinghua Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
| | - Qingpeng Kong
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
| | - Fei Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
- Laboratory of Metabolomics and Drug-induced Liver Injury, Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Wenyong Xiong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research & Development of Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, People's Republic of China
| |
Collapse
|
2
|
Drzymała-Czyż S, Dziedzic K, Szwengiel A, Krzyżanowska-Jankowska P, Nowak JK, Nowicka A, Aringazina R, Drzymała S, Kashirskaya N, Walkowiak J. Serum bile acids in cystic fibrosis patients - glycodeoxycholic acid as a potential marker of liver disease. Dig Liver Dis 2022; 54:111-117. [PMID: 34305015 DOI: 10.1016/j.dld.2021.06.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) and CF-related liver disease can lead to disturbances in bile acid metabolism. AIM This study determined serum bile acid concentrations in CF to define their usefulness in liver disease assessment. METHODS Primary, secondary and conjugated bile acid levels were measured in three CF groups (25 patients each) exhibiting: liver cirrhosis, other liver disease, no liver disease, and in 25 healthy subjects (HS). RESULTS Bile acid levels were higher in CF patients than in HS, except for glycodeoxycholic acid (GDCA). However, bile acid concentrations did not differ between patients with cirrhosis and other liver involvement. GDCA and deoxycholic acid (DCA) differentiated CF patients with non-cirrhotic liver disease from those without liver disease (GDCA-AUC: 0.924, 95%CI 0.822-1.000, p<0.001; DCA-AUC: 0.867, 95%CI: 0.731-1.000, p<0.001). Principal component analysis revealed that in CF liver disease was related to GDCA, GGTP activity, severe genotype and pancreatic insufficiency. CONCLUSIONS A CF-specific bile acid profile was defined and shown to relate to liver disease. GDCA differentiates patients with non-cirrhotic liver involvement from those with no detectable liver disease. Hence, GDCA is a candidate for validation as a biomarker of non-cirrhotic progression of liver disease in CF.
Collapse
Affiliation(s)
- Sławomira Drzymała-Czyż
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznań, Poland; Department of Bromatology, Poznan University of Medical Sciences, Poznań, Poland
| | - Krzysztof Dziedzic
- Department of Food Science and Nutrition, Institute of Food Technology of Plant Origin, Poznań University of Life Sciences, Poznań, Poland
| | - Artur Szwengiel
- Department of Food Science and Nutrition, Institute of Food Technology of Plant Origin, Poznań University of Life Sciences, Poznań, Poland
| | | | - Jan K Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznań, Poland
| | - Agata Nowicka
- Department of Pulmonology, Allergology and Respiratory Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Raisa Aringazina
- Department of Internal Diseases No1, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Sylwia Drzymała
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznań, Poland
| | - Nataliya Kashirskaya
- Department of Genetic Epidemiology (Cystic Fibrosis Group), Research Centre for Medical Genetics, Moscow, Russia
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznań, Poland.
| |
Collapse
|
3
|
Matta K, Lefebvre T, Vigneau E, Cariou V, Marchand P, Guitton Y, Royer AL, Ploteau S, Le Bizec B, Antignac JP, Cano-Sancho G. Associations between persistent organic pollutants and endometriosis: A multiblock approach integrating metabolic and cytokine profiling. ENVIRONMENT INTERNATIONAL 2022; 158:106926. [PMID: 34649050 DOI: 10.1016/j.envint.2021.106926] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/09/2021] [Accepted: 10/06/2021] [Indexed: 05/09/2023]
Abstract
Humans are exposed daily to complex mixtures of chemical pollutants through their environment and diet, some of which have the potential to disrupt the bodies' natural endocrine functions and contribute to reproductive diseases like endometriosis. Increasing epidemiological and experimental evidence supports the association between endometriosis and certain persistent organic pollutants (POPs) like dioxins; however, little is known about the underlying linking mechanisms. The main objective of this study is to proof the methodological applicability and discovery potential of integrating ultra-trace mass spectrometry (MS) profiling of POP biomarkers and endogenous biomarker profiling (MS metabolomics and cytokines) in a case-control study for the etiological research of endometriosis. The approach is applied in a pilot clinical-based study conducted in France where women with and without surgically confirmed endometriosis were recruited. Serum samples were analysed with high-resolution MS for about 30 polychlorinated biphenyls (PCBs), organochlorinated pesticides and perfluoroalkyl substances (PFAS). About 600 serum metabolites and lipids were identified with targeted metabolomics using tandem MS with the Biocrates MxP® Quant 500 Kit. A panel of 4 pro-inflammatory cytokines were analysed using ELISA-based 4-PLEX analyser. Statistical analysis included a battery of variable selection approaches, multivariate logistic regression for single-chemical associations, Bayesian kernel machine regressions (BKMR) to identify mixture effects of POPs and a multiblock approach to identify shared biomarker signatures among high risk clusters. The results showed the positive associations between some POPs and endometriosis risk, including the pesticide trans-nonachlor Odds Ratio (95% Confidence Interval) 3.38 (2.06-5.98), p < 0.0001 and PCB 114 OR (95% CI) 1.83 (1.17-2.93), p = 0.009. The BKMR approach showed a tendency of a positive cumulative effect of the mixture, however trans-nonachlor exhibited significant associations within the mixture and interacted with other PCBs, strengthening the effects at highest concentrations. Finally, the multiblock analysis, relating the various blocks of data, revealed a latent cluster of women with higher risk of endometrioma presenting higher concentrations of trans-nonachlor, PCB 114 and dioxin-like toxic equivalents from PCBs, together with an increased inflammatory profile (i.e. elevated interleukin-8 and monocyte chemoattractant protein-1). It was also highlighted a specific metabolic pattern characterized by dysregulation of bile acid homeostasis and lipase activity. Further research will be required with larger sample size to confirm these findings and gain insight on the underlying mechanisms between POPs and endometriosis.
Collapse
Affiliation(s)
| | - Tiphaine Lefebvre
- LABERCA, Oniris, INRAE, 44307 Nantes, France; Faculty of Medicine, University of Nantes, Nantes, France; Department of Biology and Reproductive Medicine, University Hospital of Nantes, Nantes, France
| | | | | | | | | | | | - Stéphane Ploteau
- Faculty of Medicine, University of Nantes, Nantes, France; Service de Gynecologie-obstétrique, CIC FEA, Hôpital Mère Enfant, CHU Hôtel Dieu, Nantes, France
| | | | | | | |
Collapse
|
4
|
Li J, Zhu X, Zhang M, Zhang Y, Ye S, Leng Y, Yang T, Kong L, Zhang H. Limb expression 1-like (LIX1L) protein promotes cholestatic liver injury by regulating bile acid metabolism. J Hepatol 2021; 75:400-413. [PMID: 33746084 DOI: 10.1016/j.jhep.2021.02.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Cholestatic liver diseases comprise a variety of disorders of bile formation and/or flow which generally result in progressive hepatobiliary injury. Regulation of bile acid (BA) synthesis and homeostasis is a promising strategy for the treatment of cholestatic liver disease. Limb expression 1-like protein (LIX1L) plays an important role in post-transcriptional gene regulation, yet its role in cholestatic liver injury remains unclear. METHODS LIX1L expression was studied in patients with primary sclerosing cholangitis (PSC) or primary biliary cholangitis (PBC), and 3 murine models of cholestasis (bile duct ligation [BDL], Mdr2 knockout [Mdr2-/-], and cholic acid [CA] feeding). Lix1l knockout mice were employed to investigate the function of LIX1L in cholestatic liver diseases. Chromatin immunoprecipitation assays were performed to determine whether Egr-1 bound to the Lix1l promoter. MiRNA expression profiling was analyzed by microarray. An adeno-associated virus (AAV)-mediated hepatic delivery system was used to identify the function of miR-191-3p in vivo. RESULTS LIX1L expression was increased in the livers of patients with PSC and PBC, and in the 3 murine models, as well as in BA-stimulated primary mouse hepatocytes. BA-induced Lix1l upregulation was dependent on Egr-1, which served as a transcriptional activator. LIX1L deficiency attenuated cholestatic liver injury in BDL and Mdr2-/- mice. MiR-191-3p was the most reduced miRNA in livers of WT-BDL mice, while it was restored in Lix1l-/--BDL mice. MiR-191-3p targets and downregulates Lrh-1, thereby inhibiting Cyp7a1 and Cyp8b1 expression. AAV-mediated hepatic delivery of miR-191-3p significantly attenuated cholestatic liver injury in Mdr2-/- mice. CONCLUSIONS LIX1L deficiency alleviates cholestatic liver injury by inhibiting BA synthesis. LIX1L functions as a nexus linking BA/Egr-1 and miR-191-3p/LRH-1 signaling. LIX1L and miR-191-3p may be promising targets for the treatment of BA-associated hepatobiliary diseases. LAY SUMMARY Bile acid homeostasis can be impaired in cholestatic liver diseases. Our study identified a novel mechanism of positive feedback regulation in cholestasis. LIX1L and miR-191-3p represent potential therapeutic targets for cholestatic liver diseases.
Collapse
Affiliation(s)
- Jie Li
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoyun Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Meihui Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yanqiu Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shengtao Ye
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yingrong Leng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ting Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
5
|
Hung C, Napoli E, Ross-Inta C, Graham J, Flores-Torres AL, Stanhope KL, Froment P, Havel PJ, Giulivi C. Ileal interposition surgery targets the hepatic TGF-β pathway, influencing gluconeogenesis and mitochondrial bioenergetics in the UCD-T2DM rat model of diabetes. FASEB J 2019; 33:11270-11283. [PMID: 31307210 DOI: 10.1096/fj.201802714r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Ileal interposition (IT) is a surgical procedure that increases the delivery of incompletely digested nutrients and biliary and pancreatic secretions to the distal intestinal mucosa. Here, we investigated the metabolic impact of this intervention in 2-mo-old prediabetic University of California, Davis type 2 diabetes mellitus rats by assessing liver gene expression at 1.5 mo post-IT surgery. Pathway analysis indicated decreased signaling via TGF-β/Smad (a family of proteins named mothers against decapentaplegic homologs), peroxisome proliferator-activated receptor (PPAR), and PI3K-Akt-AMPK-mechanistic target of rapamycin, likely targeting hepatic stellate cells because differentiation and activation of these cells is associated with decreased signaling via PPAR and TGF-β/Smad. IT surgery up-regulated the expression of genes involved in regulation of cholesterol and terpenoid syntheses and down-regulated those involved in glycerophospholipid metabolism [including cardiolipin (CL)], lipogenesis, and gluconeogenesis. Consistent with the down-regulation of the hepatic CL pathway, IT surgery produced a metabolic switch in liver, kidney cortex, and fat depots toward decreased mitochondrial fatty acid β-oxidation, the process required to fuel high energy-demanding pathways (e.g., gluconeogenesis and glyceroneogenesis), whereas opposite effects were observed in skeletal and cardiac muscles. This study demonstrates for the first time the presence of metabolic pathways that complement the effects of IT surgery to maximize its benefits and potentially identify similarly effective, durable, and less invasive therapeutic options for metabolic disease, including inhibitors of TGF-β signaling.-Hung, C., Napoli, E., Ross-Inta, C., Graham, J., Flores-Torres, A. L., Stanhope, K. L., Froment, P., Havel, P. J., Giulivi, C. Ileal interposition surgery targets the hepatic TGF-β pathway, influencing gluconeogenesis and mitochondrial bioenergetics in the UCD-T2DM rat model of diabetes.
Collapse
Affiliation(s)
- Connie Hung
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Catherine Ross-Inta
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - James Graham
- Department of Nutrition, University of California, Davis, Davis, California, USA
| | - Amanda L Flores-Torres
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA.,Department of Biochemistry, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Kimber L Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA.,Department of Nutrition, University of California, Davis, Davis, California, USA
| | - Pascal Froment
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Unité Mixte de Recherche (UMR) 85, Paris, France
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA.,Department of Nutrition, University of California, Davis, Davis, California, USA
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA.,Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis, Davis, California, USA
| |
Collapse
|
6
|
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
|
7
|
Valanejad L, Nadolny C, Shiffka S, Chen Y, You S, Deng R. Differential Feedback Regulation of Δ4-3-Oxosteroid 5β-Reductase Expression by Bile Acids. PLoS One 2017; 12:e0170960. [PMID: 28125709 PMCID: PMC5268776 DOI: 10.1371/journal.pone.0170960] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/14/2017] [Indexed: 02/08/2023] Open
Abstract
Δ4-3-oxosteroid 5β-reductase is member D1 of the aldo-keto reductase family 1 (AKR1D1), which catalyzes 5β-reduction of molecules with a 3-oxo-4-ene structure. Bile acid intermediates and most of the steroid hormones carry the 3-oxo-4-ene structure. Therefore, AKR1D1 plays critical roles in both bile acid synthesis and steroid hormone metabolism. Currently our understanding on transcriptional regulation of AKR1D1 under physiological and pathological conditions is very limited. In this study, we investigated the regulatory effects of primary bile acids, chenodeoxycholic acid (CDCA) and cholic acid (CA), on AKR1D1 expression. The expression levels of AKR1D1 mRNA and protein in vitro and in vivo following bile acid treatments were determined by real-time PCR and Western blotting. We found that CDCA markedly repressed AKR1D1 expression in vitro in human hepatoma HepG2 cells and in vivo in mice. On the contrary, CA significantly upregulated AKR1D1 expression in HepG2 cells and in mice. Further mechanistic investigations revealed that the farnesoid x receptor (FXR) signaling pathway was not involved in regulating AKR1D1 by bile acids. Instead, CDCA and CA regulated AKR1D1 through the mitogen-activated protein kinases/c-Jun N-terminal kinases (MAPK/JNK) signaling pathway. Inhibition of the MAPK/JNK pathway effectively abolished CDCA and CA-mediated regulation of AKR1D1. It was thus determined that AKR1D1 expression was regulated by CDCA and CA through modulating the MAPK/JNK signaling pathway. In conclusion, AKR1D1 expression was differentially regulated by primary bile acids through negative and positive feedback mechanisms. The findings indicated that both bile acid concentrations and compositions play important roles in regulating AKR1D1 expression, and consequently bile acid synthesis and steroid hormone metabolism.
Collapse
Affiliation(s)
- Leila Valanejad
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Christina Nadolny
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Stephanie Shiffka
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Yuan Chen
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Sangmin You
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Ruitang Deng
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, United States of America
| |
Collapse
|
8
|
|
9
|
Kakizuka S, Takeda T, Komiya Y, Koba A, Uchi H, Yamamoto M, Furue M, Ishii Y, Yamada H. Dioxin-Produced Alteration in the Profiles of Fecal and Urinary Metabolomes: A Change in Bile Acids and Its Relevance to Toxicity. Biol Pharm Bull 2016; 38:1484-95. [PMID: 26424014 DOI: 10.1248/bpb.b15-00235] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study investigated dioxin-induced changes in metabolomes in pubertal rat excrement. The administration of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or restricting dietary intake (pair-fed group) markedly altered the metabolomic profile including lipids, hormones, and vitamins in the urine and feces. TCDD caused an increase in the fecal chenodeoxycholic acid and taurocholic acid content and in urinary adrenaline and 17β-estradiol, while the urinary melatonin level was reduced by TCDD. These changes were not observed in the pair-fed group. In accordance with the elevated level of fecal bile acids, TCDD reduced the intestinal expression of the apical sodium-dependent bile salt transporter, which plays a role in resorbing bile acids from the bile duct. In addition, CYP7A1, a rate-limiting enzyme for bile acid biosynthesis, was attenuated by TCDD treatment, although TCDD induced hepatic CYP8B1, an enzyme essential for cholic acid synthesis. Supplying cholic acid or chenodeoxycholic acid to TCDD-exposed rats tended to restore the TCDD-produced reduction in serum triglycerides, whereas no similar trend was observed in wasting syndrome and lipid accumulation in the liver. These results suggest that: 1) TCDD alters the circulating levels of bile acids and hormones via a mechanism distinct from an attenuation in dietary intake, although the majority of TCDD-induced changes in nutrient contents in the excrement is due to a reduction in food intake; and 2) TCDD facilitates the excretion of bile acids and disrupts their biosynthesis, resulting in the disturbance of lipid homeostasis.
Collapse
Affiliation(s)
- Saki Kakizuka
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Heidker RM, Caiozzi GC, Ricketts ML. Grape Seed Procyanidins and Cholestyramine Differentially Alter Bile Acid and Cholesterol Homeostatic Gene Expression in Mouse Intestine and Liver. PLoS One 2016; 11:e0154305. [PMID: 27111442 PMCID: PMC4844140 DOI: 10.1371/journal.pone.0154305] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 04/12/2016] [Indexed: 01/05/2023] Open
Abstract
Bile acid (BA) sequestrants, lipid-lowering agents, may be prescribed as a monotherapy or combination therapy to reduce the risk of coronary artery disease. Over 33% of adults in the United States use complementary and alternative medicine strategies, and we recently reported that grape seed procyanidin extract (GSPE) reduces enterohepatic BA recirculation as a means to reduce serum triglyceride (TG) levels. The current study was therefore designed to assess the effects on BA, cholesterol and TG homeostatic gene expression following co-administration with GSPE and the BA sequestrant, cholestyramine (CHY). Eight-week old male C57BL/6 mice were treated for 4 weeks with either a control or 2% CHY-supplemented diet, after which, they were administered vehicle or GSPE for 14 hours. Liver and intestines were harvested and gene expression was analyzed. BA, cholesterol, non-esterified fatty acid and TG levels were also analyzed in serum and feces. Results reveal that GSPE treatment alone, and co-administration with CHY, regulates BA, cholesterol and TG metabolism differently than CHY administration alone. Notably, GSPE decreased intestinal apical sodium-dependent bile acid transporter (Asbt) gene expression, while CHY significantly induced expression. Administration with GSPE or CHY robustly induced hepatic BA biosynthetic gene expression, especially cholesterol 7α-hydroxylase (Cyp7a1), compared to control, while co-administration further enhanced expression. Treatment with CHY induced both intestinal and hepatic cholesterologenic gene expression, while co-administration with GSPE attenuated the CHY-induced increase in the liver but not intestine. CHY also induced hepatic lipogenic gene expression, which was attenuated by co-administration with GSPE. Consequently, a 25% decrease in serum TG levels was observed in the CHY+GSPE group, compared to the CHY group. Collectively, this study presents novel evidence demonstrating that GSPE provides additive and complementary efficacy as a lipid-lowering combination therapy in conjunction with CHY by attenuating hepatic cholesterol synthesis, enhancing BA biosynthesis and decreasing lipogenesis, which warrants further investigation.
Collapse
Affiliation(s)
- Rebecca M. Heidker
- Department of Agriculture, Nutrition and Veterinary Sciences, University of Nevada Reno, Reno, Nevada, United States of America
| | - Gianella C. Caiozzi
- Department of Agriculture, Nutrition and Veterinary Sciences, University of Nevada Reno, Reno, Nevada, United States of America
| | - Marie-Louise Ricketts
- Department of Agriculture, Nutrition and Veterinary Sciences, University of Nevada Reno, Reno, Nevada, United States of America
- * E-mail:
| |
Collapse
|
11
|
Alphonse PAS, Jones PJH. Revisiting Human Cholesterol Synthesis and Absorption: The Reciprocity Paradigm and its Key Regulators. Lipids 2015. [PMID: 26620375 DOI: 10.1007/s11745‐015‐4096‐7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hypercholesterolemia is a major risk factor for cardiovascular disease. Cholesterol homeostasis in the body is governed by the interplay between absorption, synthesis, and excretion or conversion of cholesterol into bile acids. A reciprocal relationship between cholesterol synthesis and absorption is known to regulate circulating cholesterol in response to dietary or therapeutic interventions. However, the degree to which these factors affect synthesis and absorption and the extent to which one vector shifts in response to the other are not thoroughly understood. Also, huge inter-individual variability exists in the manner in which the two systems act in response to any cholesterol-lowering treatment. Various factors are known to account for this variability and in light of recent experimental advances new players such as gene-gene interactions, gene-environmental effects, and gut microbiome hold immense potential in offering an explanation to the complex traits of inter-individual variability in human cholesterol metabolism. In this context, the objective of the present review is to provide an overview on cholesterol metabolism and discuss the role of potential factors such as genetics, epigenetics, epistasis, and gut microbiome, as well as other regulators in modulating cholesterol metabolism, especially emphasizing the reciprocal relationship between cholesterol synthesis and absorption. Furthermore, an evaluation of the implications of this push-pull mechanism on cholesterol-lowering strategies is presented.
Collapse
Affiliation(s)
- Peter A S Alphonse
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.
| | - Peter J H Jones
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada
- Food Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
12
|
Alphonse PAS, Jones PJH. Revisiting Human Cholesterol Synthesis and Absorption: The Reciprocity Paradigm and its Key Regulators. Lipids 2015; 51:519-36. [PMID: 26620375 DOI: 10.1007/s11745-015-4096-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/09/2015] [Indexed: 12/22/2022]
Abstract
Hypercholesterolemia is a major risk factor for cardiovascular disease. Cholesterol homeostasis in the body is governed by the interplay between absorption, synthesis, and excretion or conversion of cholesterol into bile acids. A reciprocal relationship between cholesterol synthesis and absorption is known to regulate circulating cholesterol in response to dietary or therapeutic interventions. However, the degree to which these factors affect synthesis and absorption and the extent to which one vector shifts in response to the other are not thoroughly understood. Also, huge inter-individual variability exists in the manner in which the two systems act in response to any cholesterol-lowering treatment. Various factors are known to account for this variability and in light of recent experimental advances new players such as gene-gene interactions, gene-environmental effects, and gut microbiome hold immense potential in offering an explanation to the complex traits of inter-individual variability in human cholesterol metabolism. In this context, the objective of the present review is to provide an overview on cholesterol metabolism and discuss the role of potential factors such as genetics, epigenetics, epistasis, and gut microbiome, as well as other regulators in modulating cholesterol metabolism, especially emphasizing the reciprocal relationship between cholesterol synthesis and absorption. Furthermore, an evaluation of the implications of this push-pull mechanism on cholesterol-lowering strategies is presented.
Collapse
Affiliation(s)
- Peter A S Alphonse
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada. .,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.
| | - Peter J H Jones
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.,Food Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
13
|
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
|
14
|
Kim E, Kim S, Park Y. Sorghum extract exerts cholesterol-lowering effects through the regulation of hepatic cholesterol metabolism in hypercholesterolemic mice. Int J Food Sci Nutr 2015; 66:308-13. [PMID: 25582172 DOI: 10.3109/09637486.2014.1000839] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The purpose of this study is to investigate that sorghum extract (SE) exerts cholesterol-lowering effects through the regulation of hepatic cholesterol metabolism-related protein expression. C57BL/6 mice were fed a modified AIN-93G diet (NC) with saline, or a modified AIN-93G diet with 2% cholesterol and 0.25% cholic acid with either saline (HC) or 600 mg SE/kg body weight (HC-SE). Levels of total cholesterol and triglycerides in serum and liver were significantly lower in HC-SE than in HC. The expression of 3-hydroxy-3-methylglutaryl coenzyme A reductase, sterol regulatory elementary binding protein2 and fatty acid synthase were significantly lower, whereas phosphorylated AMP-activated protein kinase expression was significantly higher in HC-SE than in HC. Cholesterol 7-α hydroxylase expression was also significantly higher in mice given SE than in those given HC. These results suggest that the cholesterol-lowering effect of SE may be related to the regulation of hepatic cholesterol metabolism in this mouse model.
Collapse
Affiliation(s)
- Eunyoung Kim
- Department of Food and Nutrition, Hanyang University , Seoul , South Korea
| | | | | |
Collapse
|
15
|
Yang F, He Y, Liu HX, Tsuei J, Jiang X, Yang L, Wang ZT, Wan YJY. All-trans retinoic acid regulates hepatic bile acid homeostasis. Biochem Pharmacol 2014; 91:483-9. [PMID: 25175738 DOI: 10.1016/j.bcp.2014.08.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/18/2014] [Accepted: 08/19/2014] [Indexed: 12/11/2022]
Abstract
Retinoic acid (RA) and bile acids share common roles in regulating lipid homeostasis and insulin sensitivity. In addition, the receptor for RA (retinoid x receptor) is a permissive partner of the receptor for bile acids, farnesoid x receptor (FXR/NR1H4). Thus, RA can activate the FXR-mediated pathway as well. The current study was designed to understand the effect of all-trans RA on bile acid homeostasis. Mice were fed an all-trans RA-supplemented diet and the expression of 46 genes that participate in regulating bile acid homeostasis was studied. The data showed that all-trans RA has a profound effect in regulating genes involved in synthesis and transport of bile acids. All-trans RA treatment reduced the gene expression levels of Cyp7a1, Cyp8b1, and Akr1d1, which are involved in bile acid synthesis. All-trans RA also decreased the hepatic mRNA levels of Lrh-1 (Nr5a2) and Hnf4α (Nr2a1), which positively regulate the gene expression of Cyp7a1 and Cyp8b1. Moreover, all-trans RA induced the gene expression levels of negative regulators of bile acid synthesis including hepatic Fgfr4, Fxr, and Shp (Nr0b2) as well as ileal Fgf15. All-trans RA also decreased the expression of Abcb11 and Slc51b, which have a role in bile acid transport. Consistently, all-trans RA reduced hepatic bile acid levels and the ratio of CA/CDCA, as demonstrated by liquid chromatography-mass spectrometry. The data suggest that all-trans RA-induced SHP may contribute to the inhibition of CYP7A1 and CYP8B1, which in turn reduces bile acid synthesis and affects lipid absorption in the gastrointestinal tract.
Collapse
Affiliation(s)
- Fan Yang
- Institute of Chinese Material Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cai-Lun Road, Shanghai 201203, China; Department of Pathology and Laboratory Medicine, the University of California at Davis Medical Center, 4645, 2nd Avenue, Sacramento, CA 95817, USA.
| | - Yuqi He
- Department of Pathology and Laboratory Medicine, the University of California at Davis Medical Center, 4645, 2nd Avenue, Sacramento, CA 95817, USA.
| | - Hui-Xin Liu
- Department of Pathology and Laboratory Medicine, the University of California at Davis Medical Center, 4645, 2nd Avenue, Sacramento, CA 95817, USA.
| | - Jessica Tsuei
- Department of Pathology and Laboratory Medicine, the University of California at Davis Medical Center, 4645, 2nd Avenue, Sacramento, CA 95817, USA.
| | - Xiaoyue Jiang
- Thermo Fisher Scientific, 355 River Oaks Pkwy, San Jose, CA 95134, USA.
| | - Li Yang
- Institute of Chinese Material Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cai-Lun Road, Shanghai 201203, China.
| | - Zheng-Tao Wang
- Institute of Chinese Material Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cai-Lun Road, Shanghai 201203, China.
| | - Yu-Jui Yvonne Wan
- Institute of Chinese Material Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cai-Lun Road, Shanghai 201203, China; Department of Pathology and Laboratory Medicine, the University of California at Davis Medical Center, 4645, 2nd Avenue, Sacramento, CA 95817, USA.
| |
Collapse
|
16
|
Bonde Y, Plösch T, Kuipers F, Angelin B, Rudling M. Stimulation of murine biliary cholesterol secretion by thyroid hormone is dependent on a functional ABCG5/G8 complex. Hepatology 2012; 56:1828-37. [PMID: 22829162 PMCID: PMC3533177 DOI: 10.1002/hep.25861] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 05/15/2012] [Indexed: 12/07/2022]
Abstract
UNLABELLED Secretion of cholesterol into bile is important for the elimination of cholesterol from the body. Thyroid hormone (TH) increases biliary cholesterol secretion and hepatic gene expression of adenosine triphosphate (ATP)-binding cassette, subfamily G (WHITE), member 5 (ABCG5) and ATP-binding cassette, subfamily G (WHITE), member 8 (ABCG8), two half-transporters that act as a heterodimeric complex promoting sterol secretion. In addition, nuclear liver x receptor-alpha (LXRa), also regulated by TH, induces gene expression of ABCG5/G8. We here investigated if the TH-induced stimulation of biliary cholesterol secretion is mediated by the ABCG5/G8 complex in vivo, and if so, whether LXRa is involved. Mice homozygous for disruption of Abcg5 (Abcg5(-/-) ) or Lxra (Lxra(-/-) ) and their wild-type counterparts were treated with triiodothyronine (T3) for 14 days and compared to untreated mice of corresponding genetic backgrounds. Bile was collected by gallbladder cannulation, and liver samples were analyzed for gene expression levels. Basal biliary cholesterol secretion in Abcg5(-/-) mice was 72% lower than in Abcg5(+/+) mice. T3 treatment increased cholesterol secretion 3.1-fold in Abcg5(+/+) mice, whereas this response was severely blunted in Abcg5(-/-) mice. In contrast, biliary cholesterol secretion in T3-treated Lxra(+/+) and Lxra(-/-) mice was increased 3.5- and 2.6-fold, respectively, and did not differ significantly. CONCLUSIONS TH-induced secretion of cholesterol into bile is largely dependent on an intact ABCG5/G8 transporter complex, whereas LXRa is not critical for this effect.
Collapse
Affiliation(s)
- Ylva Bonde
- Metabolism Unit, Center for Endocrinology, Metabolism, and Diabetes, Department of Medicine, Karolinska University Hospital HuddingeStockholm, Sweden,Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Karolinska University Hospital HuddingeStockholm, Sweden
| | - Torsten Plösch
- Center for Liver, Digestive, and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, University of GroningenGroningen, The Netherlands
| | - Folkert Kuipers
- Center for Liver, Digestive, and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, University of GroningenGroningen, The Netherlands
| | - Bo Angelin
- Metabolism Unit, Center for Endocrinology, Metabolism, and Diabetes, Department of Medicine, Karolinska University Hospital HuddingeStockholm, Sweden,Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Karolinska University Hospital HuddingeStockholm, Sweden
| | - Mats Rudling
- Metabolism Unit, Center for Endocrinology, Metabolism, and Diabetes, Department of Medicine, Karolinska University Hospital HuddingeStockholm, Sweden,Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Karolinska University Hospital HuddingeStockholm, Sweden,Address reprint requests to: Mats Rudling, M.D., Professor, Metabolism Unit, Center for Endocrinology, Metabolism, and Diabetes, C2:94, Karolinska University Hospital Huddinge, Hälsovägen, Flemingsberg, S-141 86 Stockholm, Sweden E-mail: . fax: +4687110710
| |
Collapse
|
17
|
Kootte RS, Vrieze A, Holleman F, Dallinga-Thie GM, Zoetendal EG, de Vos WM, Groen AK, Hoekstra JBL, Stroes ES, Nieuwdorp M. The therapeutic potential of manipulating gut microbiota in obesity and type 2 diabetes mellitus. Diabetes Obes Metab 2012; 14:112-20. [PMID: 21812894 DOI: 10.1111/j.1463-1326.2011.01483.x] [Citation(s) in RCA: 244] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are attributed to a combination of genetic susceptibility and lifestyle factors. Their increasing prevalence necessitates further studies on modifiable causative factors and novel treatment options. The gut microbiota has emerged as an important contributor to the obesity--and T2DM--epidemic proposed to act by increasing energy harvest from the diet. Although obesity is associated with substantial changes in the composition and metabolic function of the gut microbiota, the pathophysiological processes remain only partly understood. In this review we will describe the development of the adult human microbiome and discuss how the composition of the gut microbiota changes in response to modulating factors. The influence of short-chain fatty acids, bile acids, prebiotics, probiotics, antibiotics and microbial transplantation is discussed from studies using animal and human models. Ultimately, we aim to translate these findings into therapeutic pathways for obesity and T2DM in humans.
Collapse
Affiliation(s)
- R S Kootte
- Department of Vascular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Miura T, Kimura N, Yamada T, Shimizu T, Nanashima N, Yamana D, Hakamada K, Tsuchida S. Sustained repression and translocation of Ntcp and expression of Mrp4 for cholestasis after rat 90% partial hepatectomy. J Hepatol 2011; 55:407-14. [PMID: 21167233 DOI: 10.1016/j.jhep.2010.11.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 11/05/2010] [Accepted: 11/12/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS To clarify the mechanism of persistent cholestasis after massive hepatectomy, the relationship between such cholestasis and the expression and localization of organic anion transporters for bile acids was examined in a rat model. METHODS Male Sprague-Dawley rats were subjected to 90% hepatectomy, and tissues were harvested at 0, 1, 3, and 7 days for microarray analysis, quantitative real-time polymerase chain reaction (RT-PCR), Western blotting, and immunohistochemistry to examine the expression of multidrug resistance protein 4 (Mrp4), bile salt export pump (Bsep), and sodium-dependent taurocholate cotransporting polypeptide (Ntcp). RESULTS Persistently elevated levels of serum bile acids were observed at days 3 and 7. RT-PCR and Western blotting indicated that the expression of Mrp4, a bile acid export pump located in the basolateral membrane, was increased at day 3. The expression of Ntcp, a transporter used to uptake bile acids from the sinusoids, was significantly decreased throughout the period. The levels of Bsep, an export pump localized to the canalicular membrane, were unchanged. Immunohistochemistry revealed the localization of Mrp4 and Bsep in the basolateral and canalicular membranes, respectively. On the other hand, at days 3 and 7, Ntcp was localized in the cytoplasm and was hardly detected in the basolateral membrane. CONCLUSIONS These results suggested that the sustained repression and translocation of Ntcp and the expression of Mrp4 at the basolateral membrane seem to be responsible for the high blood bile acids levels after massive hepatectomy.
Collapse
Affiliation(s)
- Takuya Miura
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
The gut microbiota enhances the host’s metabolic capacity for processing nutrients and drugs and modulate the activities of multiple pathways in a variety of organ systems. We have probed the systemic metabolic adaptation to gut colonization for 20 days following exposure of axenic mice (n = 35) to a typical environmental microbial background using high-resolution 1H nuclear magnetic resonance (NMR) spectroscopy to analyze urine, plasma, liver, kidney, and colon (5 time points) metabolic profiles. Acquisition of the gut microbiota was associated with rapid increase in body weight (4%) over the first 5 days of colonization with parallel changes in multiple pathways in all compartments analyzed. The colonization process stimulated glycogenesis in the liver prior to triggering increases in hepatic triglyceride synthesis. These changes were associated with modifications of hepatic Cyp8b1 expression and the subsequent alteration of bile acid metabolites, including taurocholate and tauromuricholate, which are essential regulators of lipid absorption. Expression and activity of major drug-metabolizing enzymes (Cyp3a11 and Cyp2c29) were also significantly stimulated. Remarkably, statistical modeling of the interactions between hepatic metabolic profiles and microbial composition analyzed by 16S rRNA gene pyrosequencing revealed strong associations of the Coriobacteriaceae family with both the hepatic triglyceride, glucose, and glycogen levels and the metabolism of xenobiotics. These data demonstrate the importance of microbial activity in metabolic phenotype development, indicating that microbiota manipulation is a useful tool for beneficially modulating xenobiotic metabolism and pharmacokinetics in personalized health care. Gut bacteria have been associated with various essential biological functions in humans such as energy harvest and regulation of blood pressure. Furthermore, gut microbial colonization occurs after birth in parallel with other critical processes such as immune and cognitive development. Thus, it is essential to understand the bidirectional interaction between the host metabolism and its symbionts. Here, we describe the first evidence of an in vivo association between a family of bacteria and hepatic lipid metabolism. These results provide new insights into the fundamental mechanisms that regulate host-gut microbiota interactions and are thus of wide interest to microbiological, nutrition, metabolic, systems biology, and pharmaceutical research communities. This work will also contribute to developing novel strategies in the alteration of host-gut microbiota relationships which can in turn beneficially modulate the host metabolism.
Collapse
|
20
|
Daher R, Yazbeck T, Jaoude JB, Abboud B. Consequences of dysthyroidism on the digestive tract and viscera. World J Gastroenterol 2009; 15:2834-8. [PMID: 19533804 PMCID: PMC2699000 DOI: 10.3748/wjg.15.2834] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Thyroid hormones define basal metabolism throughout the body, particularly in the intestine and viscera. Gastrointestinal manifestations of dysthyroidism are numerous and involve all portions of the tract. Thyroid hormone action on motility has been widely studied, but more complex pathophysiologic mechanisms have been indicated by some studies although these are not fully understood. Both thyroid hormone excess and deficiency can have similar digestive manifestations, such as diarrhea, although the mechanism is different in each situation. The liver is the most affected organ in both hypo- and hyperthyroidism. Specific digestive diseases may be associated with autoimmune thyroid processes, such as Hashimoto’s thyroiditis and Grave’s disease. Among them, celiac sprue and primary biliary cirrhosis are the most frequent although a clear common mechanism has never been proven. Overall, thyroid-related digestive manifestations were described decades ago but studies are still needed in order to confirm old concepts or elucidate undiscovered mechanisms. All practitioners must be aware of digestive symptoms due to dysthyroidism in order to avoid misdiagnosis of rare but potentially lethal situations.
Collapse
|
21
|
Yu BZ, Kaimal R, Bai S, El Sayed KA, Tatulian SA, Apitz RJ, Jain MK, Deng R, Berg OG. Effect of guggulsterone and cembranoids of Commiphora mukul on pancreatic phospholipase A(2): role in hypocholesterolemia. JOURNAL OF NATURAL PRODUCTS 2009; 72:24-28. [PMID: 19102680 DOI: 10.1021/np8004453] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Guggulsterone (7) and cembranoids (8-12) from Commiphora mukul stem bark resin guggul were shown to be specific modulators of two independent sites that are also modulated by bile salts (1-6) to control cholesterol absorption and catabolism. Guggulsterone (7) antagonized the chenodeoxycholic acid (3)-activated nuclear farnesoid X receptor (FXR), which regulates cholesterol metabolism in the liver. The cembranoids did not show a noticeable effect on FXR, but lowered the cholate (1)-activated rate of human pancreatic IB phospholipase A2 (hPLA2), which controls gastrointestinal absorption of fat and cholesterol. Analysis of the data using a kinetic model has suggested an allosteric mechanism for the rate increase of hPLA2 by cholate and also for the rate-lowering effect by certain bile salts or cembranoids on the cholate-activated hPLA2 hydrolysis of phosphatidylcholine vesicles. The allosteric inhibition of PLA2 by certain bile salts and cembranoids showed some structural specificity. Biophysical studies also showed specific interaction of the bile salts with the interface-bound cholate-activated PLA2. Since cholesterol homeostasis in mammals is regulated by FXR in the liver for metabolism and by PLA2 in the intestine for absorption, modulation of PLA2 and FXR by bile acids and selected guggul components suggests novel possibilities for hypolipidemic and hypocholesterolemic therapies.
Collapse
Affiliation(s)
- Bao-Zhu Yu
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19713, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kőhalmy K, Rozman D, Pascussi JM, Sárváry E, Monostory K. Crosstalk between cholesterol homeostasis and drug metabolism. Orv Hetil 2008; 149:1283-9. [DOI: 10.1556/oh.2008.28329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Napjainkban a cardiovascularis megbetegedések vezető halálozási oknak számítanak világszerte. A szív- és érrendszeri megbetegedések kialakulásában jelentős szerepet játszik a magas szérumkoleszterin-szint, illetve az atherosclerosis. A vér koleszterinszintjének csökkentésével kedvezően befolyásolható a káros folyamatok kialakulása, és a már kialakult betegségekben is javulás érhető el. Az általánosan alkalmazott sztatinalapú gyógyszeres terápia ade novokoleszterin-bioszintézist gátolja a májban. Más hatóanyagok (például ezetimib) a koleszterin táplálékból történő felszívódását gátolják. A leghatékonyabb megoldást ezek kombinált alkalmazása jelentheti. A gyógyszeres terápia során azonban figyelembe kell venni, hogy számos vegyület (gyógyszer) képes specifikusan megváltoztatni – a koleszterinhomeosztázis fenntartásában szerepet játszó enzimek mellett – a gyógyszer-metabolizáló enzimek indukciójával a citokróm P450 enzimek mennyiségét is (például sztatinok), ami a terápia módosítását teszi szükségessé. A koleszterin-anyagcsere és a gyógyszer-metabolizmus szabályozásában ugyanis több kapcsolódási pont is található. A kapcsolat az úgynevezett nukleáris receptorokon keresztül valósul meg, ezért a koleszterinhomeosztázis és a gyógyszer-metabolizmus közti összefüggés megértése és ismereteink bővítése elengedhetetlen egy megfelelő terápiás stratégia kidolgozásához, illetve új gyógyszerek fejlesztéséhez.
Collapse
Affiliation(s)
- Krisztina Kőhalmy
- 1 Magyar Tudományos Akadémia, Kémiai Kutatóközpont Farmakobiokémiai Osztály Budapest Pusztaszeri út 59–67. 1025
| | | | - Jean-Marc Pascussi
- 3 Université Montpellier Institut National de la Santé et de la Recherche Médicale Montpellier Franciaország
| | - Enikő Sárváry
- 4 Semmelweis Egyetem, Általános Orvostudományi Kar Transzplantációs és Sebészeti Klinika Budapest
| | - Katalin Monostory
- 1 Magyar Tudományos Akadémia, Kémiai Kutatóközpont Farmakobiokémiai Osztály Budapest Pusztaszeri út 59–67. 1025
| |
Collapse
|
23
|
Li Y, Mezei O, Shay NF. Human and murine hepatic sterol-12-alpha-hydroxylase and other xenobiotic metabolism mRNA are upregulated by soy isoflavones. J Nutr 2007; 137:1705-12. [PMID: 17585019 DOI: 10.1093/jn/137.7.1705] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The transport and metabolism of xenobiotics is controlled by the drug transporters and drug-metabolizing enzymes in the liver and small intestine. Expression of these genes is 1 factor affecting the half-life of drugs and xenobiotics. Isoflavone-containing soyfood products and supplements are promoted to treat several different health conditions, including improvement of blood lipid profiles. Because relatively high isoflavone intake may be possible via use of supplements, we tested the hypothesis that isoflavones regulate the expression of genes critical to drug transport and metabolism. Using a gene array screening method, 2 drug transporters, Multidrug restistant-1 and Multidrug-related protein-2; 3 phase I enzymes, cytochrome 1A1, 3A4, and 8B1; and 2 phase II enzymes, carbohydrate sulfotransferase-5 and glutathione-sulfotransferase-2, were upregulated 3-fold or more of the initial expression levels in primary human hepatocytes exposed to soy isoflavones for 48 h. Isoflavone-related induction of 12-alpha-hydroxylase (CYP8B1) was further studied in other in vitro and murine in vivo models. Transfection studies suggest that isoflavones may act as a weak activating ligand for hepatocyte nuclear factor 4alpha, which in turn may activate the transcription of CYP8B1. The action of soy isoflavones on CYP8B1 may increase the conversion of cholesterol into bile acids and enhance synthesis of cholic acid. These isoflavone-induced changes in gene expression may help explain how isoflavones modulate cholesterol metabolism.
Collapse
Affiliation(s)
- Yilan Li
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.
| | | | | |
Collapse
|
24
|
Souidi M, Scanff P, Grison S, Gourmelon P, Aigueperse J. Effects of ionizing radiation on the activity of the major hepatic enzymes implicated in bile acid biosynthesis in the rat. C R Biol 2007; 330:861-70. [PMID: 18068644 DOI: 10.1016/j.crvi.2007.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Revised: 03/22/2007] [Accepted: 03/22/2007] [Indexed: 10/23/2022]
Abstract
In the days following high-dose radiation exposure, damage to small intestinal mucosa is aggravated by changes in the bile acid pool reaching the gut. Intestinal bile acid malabsorption, as described classically, may be associated with altered hepatic bile acid biosynthesis, which was the objective of this work. The activity of the main rate-limiting enzymes implicated in the bile acid biosynthesis were evaluated in the days following an 8-Gy gamma(60)Co total body irradiation of rats, with concomitant determination of biliary bile acid profiles and intestinal bile acid content. Modifications of biliary bile acid profiles, observed as early as the first post-irradiation day, were most marked at the third and fourth day, and resulted in an increased hydrophobicity index. In parallel, the intestinal bile acids' content was enhanced and hepatic enzymatic activities leading to bile acids were changed. A marked increase of sterol 12 alpha-hydroxylase and decrease of oxysterol 7 alpha-hydroxylase activity was observed at day 3, whereas both cholesterol 7 alpha-hydroxylase and oxysterol 7 alpha-hydroxylase activities were decreased at day 4 after irradiation. These results show, for the first time, radiation-induced modifications of hepatic enzymatic activities implicated in bile acid biosynthesis and suggest that they are mainly a consequence of radiation-altered intestinal absorption, which induces a physiological response of the enterohepatic bile acid recirculation.
Collapse
Affiliation(s)
- Maâmar Souidi
- Institut de radioprotection et de sûreté nucléaire, direction de la radioprotection de l'homme, service de radiobiologie et d'épidémiologie, IRSN, 92262 Fontenay-aux-Roses cedex, France
| | | | | | | | | |
Collapse
|
25
|
Henríquez-Hernández LA, Flores-Morales A, Santana-Farré R, Axelson M, Nilsson P, Norstedt G, Fernández-Pérez L. Role of pituitary hormones on 17alpha-ethinylestradiol-induced cholestasis in rat. J Pharmacol Exp Ther 2007; 320:695-705. [PMID: 17108234 DOI: 10.1124/jpet.106.113209] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Estrogens cause intrahepatic cholestasis in susceptible women during pregnancy, after administration of oral contraceptives, or during postmenopausal hormone replacement therapy. 17alpha-Ethinylestradiol (EE) is a synthetic estrogen widely used to cause experimental cholestasis in rodents with the aim of examining molecular mechanisms involved in this disease. EE actions on the liver are thought to be mediated by estrogen receptor alpha (ERalpha) and pituitary hormones. We tested this hypothesis by analyzing metabolic changes induced by EE in livers from hypophysectomized (HYPOX) and hypothyroid rats. Microarray studies revealed that the number of genes regulated by EE was increased almost 4-fold in HYPOX rat livers compared with intact males. Little overlap was apparent between the effects of EE in intact and HYPOX rats, demonstrating that pituitary hormones play a critical role in the hepatic effects of EE. Consistently, hypophysectomy protects the liver against induction by EE of serum bilirubin and alkaline phosphatase, two markers of cholestasis and hepatotoxicity and modulates the effects of EE on several genes involved in bile acid homeostasis (e.g., FXR, SHP, BSEP, and Cyp8b1). Finally, we demonstrate a novel mechanism of action of EE through binding and negative regulation of glucocorticoid receptor-mediated transcription. In summary, pituitary- and ERalpha-independent mechanisms contribute to development of EE-induced changes in liver transcriptome. Such mechanisms may be relevant when this model of EE-induced cholestasis is evaluated. The observation that the pharmacological effects of estrogen in liver differ in the absence or presence of the pituitary could be clinically relevant, because different drugs that block actions of pituitary hormones are now available.
Collapse
Affiliation(s)
- L A Henríquez-Hernández
- Molecular Endocrinology Group, Department of Clinical Science, University of Las Palmas of Gran Canaria, Spain
| | | | | | | | | | | | | |
Collapse
|
26
|
Wang J, Olin M, Rozell B, Björkhem I, Einarsson C, Eggertsen G, Gåfvels M. Differential hepatocellular zonation pattern of cholesterol 7α-hydroxylase (Cyp7a1) and sterol 12α-hydroxylase (Cyp8b1) in the mouse. Histochem Cell Biol 2007; 127:253-61. [PMID: 17237956 DOI: 10.1007/s00418-006-0239-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2006] [Indexed: 12/13/2022]
Abstract
The synthesis of primary bile acids is confined to the hepatocytes. This study aimed to evaluate the expression pattern within the liver architecture of the rate-limiting enzyme of the neutral pathway, cholesterol 7alpha-hydroxylase (Cyp7a1), and sterol 12alpha-hydroxylase (Cyp8b1), the enzyme necessary for the synthesis of cholic acid. Specific Cyp8b1 and Cyp7a1 peptide antiserums were used for immunohistochemical staining of livers from wild type and Cyp8b1 null mice, the latter instead expressing beta-galactosidase (beta-Gal) as a replacement reporter gene. Cyp8b1 was mainly expressed in the hepatocytes in a zonal pattern surrounding the central vein while the areas surrounding the portal zones showed much lower levels. The zonation was maintained in cholic acid-depleted mice using beta-Gal as a reporter protein. Cyp7a1 expression in wild type mice also showed a zonal distribution pattern, although less distinct, with a maximal expression within a 1-2 cell thick layer of hepatocytes surrounding the central vein. In Cyp8b1 null mice, a more intense staining was obtained, in accordance with the higher expression level of Cyp7a1, although the overall expression pattern was maintained. Our results in mice indicate possible differences in the regulation of the cellular zonation of Cyp7a1 and Cyp8b1. Also, cholic acid affects the set-point of Cyp7a1 expression but not its zonal distribution.
Collapse
Affiliation(s)
- Jin Wang
- Department of Laboratory Medicine (H3), Division of Clinical Chemistry, C1-74, Karolinska Institutet and Karolinska University Hospital, 141 86 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The nuclear receptors CAR and PXR were first characterized as xenosensing transcription factors regulating the induction of phase I and II xenobiotic-metabolizing enzymes as well as transporters in response to exogenous stimuli. It has now become clear, however, that these receptors cross-talk with endogenous stimuli as well, which extends their regulation to various physiological processes such as energy metabolism and cell growth. As recognition of the function of these receptors has widened, the molecular mechanism of their regulation has evolved from simple protein-DNA binding to regulation by complex protein-protein interactions. Novel mechanisms as to how xenobiotic exposure alters hepatic metabolic pathways such as gluconeogenesis and beta-oxidation have emerged. At the same time, the molecular mechanism of how endogenous stimuli, such as insulin, regulate xenobiotc metabolism via CAR and PXR have also become evident.
Collapse
Affiliation(s)
| | - MASAHIKO NEGISHI
- Corresponding author: Masahiko Negishi, Ph.D., Tel: 919-541-2404, Fax: 919-541-0696, E-mail:
| |
Collapse
|
28
|
Abstract
By participating in pathways of cholesterol biosynthesis and elimination, different cytochrome P450 (P450 or CYP) enzymes play an important role in maintenance of cholesterol homeostasis. CYP51 is involved in cholesterol biosynthesis, whereas CYP 7A1, 27A1, 46A1, 7B1, 39A1, and 8B1 are the key enzymes in cholesterol catabolism to bile acids, the major route of cholesterol elimination in mammals. Cholesterol transformations to steroid hormones are also initiated by the P450 enzyme CYP11A1. Finally, one of the major drug-metabolizing P450s CYP3A4 seems to contribute to bile acid biosynthesis as well. The 9 P450s will be the focus of this review and assessed as drug targets for cholesterol lowering.
Collapse
Affiliation(s)
- Irina A Pikuleva
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1031, USA.
| |
Collapse
|
29
|
Santosa S, Varady KA, AbuMweis S, Jones PJH. Physiological and therapeutic factors affecting cholesterol metabolism: does a reciprocal relationship between cholesterol absorption and synthesis really exist? Life Sci 2006; 80:505-14. [PMID: 17113605 DOI: 10.1016/j.lfs.2006.10.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 09/18/2006] [Accepted: 10/10/2006] [Indexed: 01/06/2023]
Abstract
Cholesterol absorption and synthesis contribute to maintaining cholesterol homeostasis. Several physiological and therapeutic factors affect cholesterol homeostasis, including: genetics, circadian rhythm, body weight, plant sterols, ezetimibe, and statin therapy. The present objective is to determine the main vector, i.e. cholesterol absorption or synthesis, affected by each of these factors, and to examine whether an alteration in one vector is linked to a reciprocal change in the other. Current techniques used to assess cholesterol absorption and synthesis are also reviewed. Review of physiological factors affecting cholesterol metabolism suggest a reciprocal relationship between these two vectors. Carriers of the E2 isoform of apolipoprotein E and ATP binding cassette (ABC) G8 19H (exon 1 mutation) show a decrease in cholesterol absorption accompanied by a corresponding increase in synthesis. Circadian rhythm affects cholesterol synthesis, however, its effect on absorption has yet to be established. Obese subjects show an increase in cholesterol synthesis with a subsequent decrease in cholesterol absorption. Weight loss down regulates cholesterol synthesis, but has little or no effect on absorption. In the case of therapeutic factors, plant sterols and stanols inhibit cholesterol absorption, which results in a compensatory increase in synthesis. Ezetimibe also decreases intestinal absorption, while reciprocally increasing synthesis. Statin therapy down regulates synthesis, which is accompanied by a rise in absorption. These findings suggest that a change in one vector, fairly consistently, results in a compensatory and opposing change in the other. An understanding of this reciprocal relationship between cholesterol absorption and synthesis may allow for the development of more effective interventions for dyslipidemic disorders.
Collapse
Affiliation(s)
- Sylvia Santosa
- School of Dietetics and Human Nutrition, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
30
|
Inoue Y, Yu AM, Yim SH, Ma X, Krausz KW, Inoue J, Xiang CC, Brownstein MJ, Eggertsen G, Björkhem I, Gonzalez FJ. Regulation of bile acid biosynthesis by hepatocyte nuclear factor 4alpha. J Lipid Res 2005; 47:215-27. [PMID: 16264197 PMCID: PMC1413576 DOI: 10.1194/jlr.m500430-jlr200] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hepatocyte nuclear factor 4alpha (HNF4alpha) regulates many genes that are preferentially expressed in liver. Mice lacking hepatic expression of HNF4alpha (HNF4alphaDeltaL) exhibited markedly increased levels of serum bile acids (BAs) compared with HNF4alpha-floxed (HNF4alphaF/F) mice. The expression of genes involved in the hydroxylation and side chain beta-oxidation of cholesterol, including oxysterol 7alpha-hydroxylase, sterol 12alpha-hydroxylase (CYP8B1), and sterol carrier protein x, was markedly decreased in HNF4alphaDeltaL mice. Cholesterol 7alpha-hydroxylase mRNA and protein were diminished only during the dark cycle in HNF4alphaDeltaL mice, whereas expression in the light cycle was not different between HNF4alphaDeltaL and HNF4alphaF/F mice. Because CYP8B1 expression was reduced in HNF4alphaDeltaL mice, it was studied in more detail. In agreement with the mRNA levels, CYP8B1 enzyme activity was absent in HNF4alphaDeltaL mice. An HNF4alpha binding site was found in the mouse Cyp8b1 promoter that was able to direct HNF4alpha-dependent transcription. Surprisingly, cholic acid-derived BAs, produced as a result of CYP8B1 activity, were still observed in the serum and gallbladder of these mice. These studies reveal that HNF4alpha plays a central role in BA homeostasis by regulation of genes involved in BA biosynthesis, including hydroxylation and side chain beta-oxidation of cholesterol in vivo.
Collapse
Affiliation(s)
- Yusuke Inoue
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute
| | - Ai-Ming Yu
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute
| | - Sun Hee Yim
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute
| | - Xiaochao Ma
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute
| | | | - Junko Inoue
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute
| | - Charlie C. Xiang
- Laboratory of Genetics, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Michael J. Brownstein
- Laboratory of Genetics, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Gösta Eggertsen
- Department of Medical Laboratory Sciences and Technology, Huddinge University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Ingemar Björkhem
- Department of Medical Laboratory Sciences and Technology, Huddinge University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute
| |
Collapse
|
31
|
Murphy C, Parini P, Wang J, Björkhem I, Eggertsen G, Gåfvels M. Cholic acid as key regulator of cholesterol synthesis, intestinal absorption and hepatic storage in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2005; 1735:167-75. [PMID: 15994119 DOI: 10.1016/j.bbalip.2005.06.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2004] [Revised: 05/18/2005] [Accepted: 06/06/2005] [Indexed: 10/25/2022]
Abstract
To study the effects of cholic acid (CA) feeding on hepatic cholesterol metabolism, male sterol 12alpha-hydroxylase (CYP8B1) knockout (-/-) mice and wildtype controls (+/+) were fed either a control diet or the same diet supplemented with CA (0.1% or 0.5% w/w) or cholesterol (1% w/w). During feeding of the control diet, cholesterol synthesis was increased in CYP8B1-/- compared to +/+ mice. Both cholesterol and CA feeding down regulated mRNA expression of cholesterogenic genes and hepatic de novo cholesterol synthesis as also reflected by a concomitant decrease in the nuclear factor SREBP-2 precursor protein and increased hepatic free cholesterol levels. Mice with an intact CYP8B1 gene (CYP8B1+/+ and C57Bl/6 mice) accumulated higher concentrations of cholesteryl esters (24- and 25-fold, respectively) in their livers compared to CYP8B1-/- mice (8-fold). Feeding of CA increased intestinal cholesterol absorption in CYP8B1+/+ mice by 23% and in CYP8B1-/- mice by 50%. While plasma cholesterol did not differ between CYP8B1+/+ and -/- mice under control conditions and cholesterol feeding a decrease was seen in CYP8B1-/- but not CYP8B1+/+ mice fed CA. This study indicates that CA is an important determinant for intestinal cholesterol absorption and that the levels of the transcription factor SREBP-2 in the liver are dependent upon the combined effect of CA on intestinal cholesterol absorption and CYP7A1. The possibility is discussed that inhibition of CYP8B1 and thus CA synthesis may be beneficial for the treatment of hyperlipidemic disorders.
Collapse
Affiliation(s)
- Charlotte Murphy
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska University Hospital, 14186 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
32
|
Lu Y, Heydel JM, Li X, Bratton S, Lindblom T, Radominska-Pandya A. Lithocholic acid decreases expression of UGT2B7 in Caco-2 cells: a potential role for a negative farnesoid X receptor response element. Drug Metab Dispos 2005; 33:937-46. [PMID: 15821044 PMCID: PMC2652669 DOI: 10.1124/dmd.104.003061] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human UDP-glucuronosyltransferase (UGT) 2B7 is the major isoform catalyzing the glucuronidation of a variety of endogenous compounds including bile acids. To determine the role of bile acids in the regulation of UGT2B7 expression, Caco-2 cells were incubated with the natural human farnesoid X receptor (hFXR) ligand, chenodeoxycholic acid, as well as the secondary bile acid, lithocholic acid, derived from chenodeoxycholic acid. Incubation of Caco-2 cells with lithocholic acid in the absence of exogenous hFXR resulted in a dose-dependent down-regulation of UGT2B7 mRNA levels, with an IC(50) of 13 microM. Similar down-regulation was also observed with chenodeoxycholic acid; however, much higher concentrations were required. Transient transfection of Caco-2 cells with hFXR suppressed UGT2B7 mRNA expression both in the absence and presence of ligand. UGT2B7 promoter transfection experiments and deletion/mutation analysis showed that lithocholic acid-activated hFXR decreased UGT2B7 promoter activity via a negative hFXR response element (NFRE) located between nucleotides -148 and -134. Cotransfection with hFXR and/or human retinoid X receptor further enhanced the repression. Electrophoretic mobility shift assays additionally confirmed the role of NFRE in UGT2B7 down-regulation by lithocholic acid. These findings suggest that lithocholic acid, an activator of nuclear hFXR, acts as a negative regulator of UGT2B7 expression, indicating that hFXR may play an essential role in lithocholic acid homeostasis through negative regulation of this UGT that is involved in lithocholic acid biotransformation. Therefore, it is postulated that lithocholic acid toxicity may be due to down-regulation of genes involved in its detoxification, including UGT2B7, leading to limited excretion of lithocholic acid from the body.
Collapse
Affiliation(s)
- Yuan Lu
- Department of Biochemistry, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | | | | | | | | |
Collapse
|
33
|
Wang J, Greene S, Eriksson LC, Rozell B, Reihnér E, Einarsson C, Eggertsen G, Gåfvels M. Human sterol 12α-hydroxylase (CYP8B1) is mainly expressed in hepatocytes in a homogenous pattern. Histochem Cell Biol 2005; 123:441-6. [PMID: 15891895 DOI: 10.1007/s00418-005-0779-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2005] [Indexed: 11/26/2022]
Abstract
The liver is the only organ where the complete synthesis of bile acids takes place. The present study was undertaken to investigate whether regional differences exist within the individual human hepatic lobuli regarding the pattern of expression of sterol 12alpha-hydroxylase (CYP8B1), a key enzyme in bile acid synthesis. A specific anti-human CYP8B1 peptide antiserum was developed and used for Western blotting and hepatic immunostaining of livers from various patients. CYP8B1 in human liver was expressed in the cytoplasm of hepatocytes with an even nonzonal distribution within the liver lobulus. Pericentral expression was confirmed for CYP2E1. A weak staining was noted in cholangiocytes and Kupffer cells. Previous studies on hepatic CYP27A1 and CYP7A1 in rats have shown a zonal expression, primarily in the pericentral region. Our studies indicate a different pattern for CYP8B1 expression in human liver, which was even rather than zonal.
Collapse
Affiliation(s)
- Jin Wang
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska University Hospital Huddinge, C1:74, 141 86 Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Jahan A, Chiang JYL. Cytokine regulation of human sterol 12alpha-hydroxylase (CYP8B1) gene. Am J Physiol Gastrointest Liver Physiol 2005; 288:G685-95. [PMID: 15550563 DOI: 10.1152/ajpgi.00207.2004] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sterol 12alpha-hydroxylase (CYP8B1) catalyzes cholic acid synthesis in the liver and is feedback inhibited by bile acids. In addition to activating farnesoid X receptor (nuclear receptor subfamily 1H4), bile acids also induce inflammatory cytokines in hepatocytes. The objective of this study was to investigate the mechanism by which inflammatory cytokines inhibit human CYP8B1 gene transcription. Real-time PCR assays revealed that both chenodeoxycholic acid (CDCA) and interleukin-1beta (IL-1beta) markedly reduced CYP8B1, cholesterol 7alpha-hydroxylase CYP7A1 and hepatic nuclear factor 4alpha (HNF4alpha) mRNA expression levels in human primary hepatocytes. However, CDCA induced, but IL-1beta reduced, small heterodimer partner (SHP) mRNA expression. IL-1beta inhibited human CYP8B1 reporter activity only in liver cells, and a c-Jun NH(2)-terminal kinase (JNK)-specific inhibitor-blocked IL-1beta inhibition. Activated JNK1 or c-Jun inhibited, whereas their dominant negative forms blocked, IL-1beta inhibition of CYP8B1 transcription. Mutagenesis analyses mapped an IL-1beta response element to a previously identified bile acid response element, which contains an HNF4alpha binding site. A dominant negative HNF4alpha inhibited CYP8B1 gene transcription and ectopically expressed HNF4alpha blocked IL-1beta inhibition. Furthermore, IL-1beta inhibited HNF4alpha gene transcription, protein expression, and binding to the CYP8B1 gene. JNK1 phosphorylated HNF4alpha and a JNK-specific inhibitor blocked the IL-1beta inhibition of HNF4alpha expression. These results suggest that IL-1beta inhibits CYP8B1 gene transcription via a mitogen-activated protein kinase/JNK pathway that inhibits HNF4alpha gene expression and its DNA-binding ability. This mechanism may play an important role in the adaptive response to inflammatory cytokines and in the protection of the liver during cholestasis.
Collapse
Affiliation(s)
- Asmeen Jahan
- Dept. of Biochemistry and Molecular Pathology, Northeastern Ohio Universities College of Medicine, 4209 State Route 44, P. O Box 95, Rootstown, OH 44272, USA
| | | |
Collapse
|
35
|
Chen W, Suruga K, Nishimura N, Gouda T, Lam VN, Yokogoshi H. Comparative regulation of major enzymes in the bile acid biosynthesis pathway by cholesterol, cholate and taurine in mice and rats. Life Sci 2005; 77:746-57. [PMID: 15936349 DOI: 10.1016/j.lfs.2004.11.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2004] [Accepted: 11/23/2004] [Indexed: 10/25/2022]
Abstract
These enzymes play important roles in the biosynthesis of bile acids. They are cholesterol 7alpha-hydroxylase (CYP7A1), the rate limiting enzyme in the classic pathway, sterol 12alpha-hydroxylase (CYP8B1), the key enzyme for synthesis of cholic acid (CA), and sterol 27-hydroxylase (CYP27), the initial enzyme in the alternative pathway. In the present study, the susceptibility of these three enzymes to dietary cholesterol and cholate, and the cholesterol lowering effect of taurine were determined in male C57BL/6 mice and Wistar rats. Both mice and rats were divided into 6 groups: control group (N), high cholesterol diet group (C), high cholesterol and cholate diet group (CB), and their 1% taurine-supplemented groups (NT, CT, CBT, respectively). After animals were fed with the respective diets for one week, the mRNA levels of CYP7A1 increased in the C-group compared with those of the N-group, and decreased in the CB-group compared with those of the C-group in both mice and rats. But the extent of decrease is different between the two species. CYP8B1 was also markedly repressed by cholate in mice, but not in rats. These results are consistent with the changes in serum and liver cholesterol concentrations. Taurine significantly increased CYP7A1 mRNA levels in the CBT-group compared with the CB-group in both animal models, with a subsequent decrease in serum and liver cholesterol levels and increase in fecal bile acid excretion. Up-regulated CYP8B1 was also observed after taurine supplementation in the CBT-group in mice. No increase in CYP7A1 was produced by taurine in the CT-group compared with that of the C-group in mice, although the changes of serum and liver cholesterol and fecal bile acids indicated taurine showed an efficient cholesterol lowering effect. In addition, CYP27 was induced in both C- and CB-groups of rats but not of mice, and no changes were produced by taurine. The overall results suggest that there are differences between mice and rats in susceptibility of the three enzymes to dietary cholesterol and cholate, and taurine induced CYP7A1 to produce its cholesterol-lowering effect only in the presence of cholate in the cholesterol diet.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Bile Acids and Salts/biosynthesis
- Blotting, Northern
- Cholates/pharmacology
- Cholestanetriol 26-Monooxygenase
- Cholesterol 7-alpha-Hydroxylase/genetics
- Cholesterol 7-alpha-Hydroxylase/metabolism
- Cholesterol, Dietary/blood
- Cholesterol, Dietary/pharmacology
- Cloning, Molecular
- Cytochrome P-450 Enzyme System/metabolism
- DNA Primers
- DNA, Complementary/genetics
- Diet
- Electrophoresis, Agar Gel
- Feces/chemistry
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/genetics
- Liver/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Rats
- Rats, Wistar
- Species Specificity
- Steroid 12-alpha-Hydroxylase/genetics
- Steroid 12-alpha-Hydroxylase/metabolism
- Steroid Hydroxylases/genetics
- Steroid Hydroxylases/metabolism
- Taurine/pharmacology
Collapse
Affiliation(s)
- Wen Chen
- Laboratory of Nutritional Biochemistry, School of Food and Nutritional Sciences, and COE Program in the 21st Century, The University of Shizuoka, Shizuoka 422-8526, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Dieter MZ, Maher JM, Cheng X, Klaassen CD. Expression and regulation of the sterol half-transporter genes ABCG5 and ABCG8 in rats. Comp Biochem Physiol C Toxicol Pharmacol 2004; 139:209-18. [PMID: 15683829 DOI: 10.1016/j.cca.2004.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Revised: 11/03/2004] [Accepted: 11/03/2004] [Indexed: 11/22/2022]
Abstract
The ABCG5 and ABCG8 genes encode half-transporter proteins that heterodimerize to form a transporter of plant sterols and cholesterol. The purpose of this study was to examine the expression and regulation of ABCG5 and ABCG8 at the mRNA level in Sprague-Dawley rats. Both ABCG5 and ABCG8 mRNA were expressed primarily in rat small intestine and liver, and gender-specific differences in expression were observed. The effects of treatment with a battery of microsomal enzyme inducers on ABCG5 and ABCG8 mRNA were examined; most treatments had no effect, but of three PXR ligands, PCN was an effective inducer, spironolactone was repressive, and dexamethasone was ineffective. The effects of a 1% cholesterol diet on the regulation of rat ABCG5 and ABCG8 were also examined, and compared with those in C57BL/6 mice. Cholesterol caused a suppression of ABCG5 and ABCG8 mRNA in rat liver, but the same treatment increased the expression of these genes in mouse liver. ABCG5 and ABCG8 mRNA was also induced by cholesterol in rat ileum, but not mouse ileum. These results suggest variation between rats and mice in regulatory mechanisms controlling ABCG5 and ABCG8 expression, and may explain some differences in lipid metabolism observed between these two species.
Collapse
Affiliation(s)
- Matthew Z Dieter
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | |
Collapse
|
37
|
Yang Y, Eggertsen G, Gåfvels M, Andersson U, Einarsson C, Björkhem I, Chiang JYL. Mechanisms of cholesterol and sterol regulatory element binding protein regulation of the sterol 12α-hydroxylase gene (CYP8B1). Biochem Biophys Res Commun 2004; 320:1204-10. [PMID: 15249218 DOI: 10.1016/j.bbrc.2004.06.069] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Indexed: 10/26/2022]
Abstract
Sterol 12alpha-hydroxylase (CYP8B1) is an obligatory enzyme for the synthesis of cholic acid and regulation of liver bile acid synthesis and intestine cholesterol absorption. The present study evaluates the roles for sterol regulatory element binding proteins (SREBPs) in the regulation of the CYP8B1 gene. Cholesterol feeding of mice and rats decreased the activity of CYP8B1, contrary to the up-regulation of cholesterol 7alpha-hydroxylase (CYP7A1). Cholesterol feeding also reduced mRNA levels for SREBP-1 but not for SREBP-2 in rat livers. Cholesterol and 25-hydroxycholesterol decreased the CYP8B1/luciferase reporter activity. Co-transfection of SREBP-1a and -1c stimulated CYP8B1 promoter activity, while SREBP-2 did not have any effects. Electrophoretic mobility shift assay and mutagenesis analyses identified several functional sterol regulatory elements (SRE) and E-box motifs in the rat CYP8B1 promoter. Our results indicate that SREBP-1a and -1c enhance transcription of the CYP8B1 gene through binding to SRE. Cholesterol loading reduces SREBP-1 mRNA expression in addition to reducing functional SREBP-1 protein, and results in decreasing CYP8B1 gene transcription.
Collapse
Affiliation(s)
- Yizeng Yang
- Division of Clinical Chemistry, Karolinska Institute at Huddinge University Hospital, Huddinge S-141 86, Sweden
| | | | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- John Y L Chiang
- Department of Biochemistry and Molecular Pathology, Northeastern Ohio Universities College of Medicine, 4209 State Route 44, P. O. Box 95, Rootstown, OH 44272, USA.
| |
Collapse
|
39
|
del Castillo-Olivares A, Campos JA, Pandak WM, Gil G. The role of alpha1-fetoprotein transcription factor/LRH-1 in bile acid biosynthesis: a known nuclear receptor activator that can act as a suppressor of bile acid biosynthesis. J Biol Chem 2004; 279:16813-21. [PMID: 14766742 DOI: 10.1074/jbc.m400646200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Two key regulatory enzymes in the bile acid biosynthesis pathway are cholesterol 7alpha-hydroxylase/CYP7A1 (7alpha-hydroxylase) and sterol 12alpha-hydroxylase/CYP8B1 (12alpha-hydroxylase). It has been shown previously that hepatocyte nuclear factor-4alpha (HNF-4) and the alpha(1)-fetoprotein transcription factor (FTF) are activators of 7alpha-and 12alpha-hydroxylase transcription and that the small heterodimer partner (SHP) suppresses bile acid biosynthesis by heterodimerizing with FTF. However, the role of FTF in bile acid biosynthesis has been studied only in tissue culture systems. In heterozygous FTF knockout mice, 7alpha- and 12alpha-hydroxylase genes were expressed at 5-7-fold higher levels than in wild-type mice, an apparent direct contradiction to previous in vitro observations. This higher expression of the 7alpha- and 12alpha-hydroxylase genes resulted in a 33% higher bile acid pool in their gallbladders, bile more enriched in cholic acid, and a 13% decrease in plasma cholesterol levels. Adenovirus-mediated FTF overexpression in wild-type mice resulted in 10-fold lower expression of the 7alpha- and 12alpha-hydroxylase genes and up to 8-fold higher SHP expression, highlighting the dual role that FTF plays in different promoters. Shorter overexpression times still resulted in lower 7alpha- and 12alpha-hydroxylase expression, but unchanged SHP expression, suggesting that two different mechanisms are involved in the FTF-mediated suppression of 7alpha- and 12alpha-hydroxylase expression. This FTF-mediated suppression of the expression of two bile acid biosynthesis genes resulted in a 3-fold lower rate of bile acid synthesis in a rat bile fistula animal model. Based on these observations and on protein binding studies performed in vitro and by chromatin immunoprecipitation, we hypothesize that FTF has two synergetic effects that contribute to its role in bile acid biosynthesis: 1) it has the ability to activate the expression of SHP, which in turn heterodimerizes and suppresses FTF transactivation activity; and 2) it occupies the FTF/HNF-4 recognition site within the 7alpha- and 12alpha-hydroxylase promoters, which can otherwise be occupied by a factor (HNF-4) that cannot be suppressed by SHP.
Collapse
Affiliation(s)
- Antonio del Castillo-Olivares
- Department of Biochemistry, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia 23298-0614, USA
| | | | | | | |
Collapse
|
40
|
Scanff P, Souidi M, Grison S, Griffiths NM, Gourmelon P. Alteration of the enterohepatic recirculation of bile acids in rats after exposure to ionizing radiation. Can J Physiol Pharmacol 2004; 82:114-24. [PMID: 15052292 DOI: 10.1139/y03-131] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this work was to study acute alterations of the enterohepatic recirculation (EHR) of bile acids 3 days after an 8-Gy radiation exposure in vivo in the rat by a washout technique. Using this technique in association with HPLC analysis, the EHR of the major individual bile acids was determined in control and irradiated animals. Ex vivo ileal taurocholate absorption was also studied in Ussing chambers. Major hepatic enzyme activities involved in bile acid synthesis were also measured. Measurements of bile acid intestinal content and intestinal absorption efficiency calculation from washout showed reduced intestinal absorption with significant differences from one bile acid to another: absorption of taurocholate and tauromuricholate was decreased, whereas absorption of the more hydrophobic taurochenodeoxycholate was increased, suggesting that intestinal passive diffusion was enhanced, whereas ileal active transport might be reduced. Basal hepatic secretion was increased only for taurocholate, in accordance with the marked increase of CYP8B1 activity in the liver. The results are clearly demonstrate that concomitantly with radiation-induced intestinal bile acid malabsorption, hepatic bile acid synthesis and secretion are also changed. A current working model for pathophysiological changes in enterohepatic recycling after irradiation is thus proposed.Key words: irradiation, bile acids, intestine, liver, enterohepatic recycling.
Collapse
Affiliation(s)
- P Scanff
- Institut de Radioprotection et de Sûreté Nucléaire, Direction de la RadioProtection de l'Homme, Service de Radiobiologie et d'Epidemiologie, IRSN, B.P. n 17, F-92262 Fontenay-aux-Roses CEDEX, France.
| | | | | | | | | |
Collapse
|
41
|
Drover VAB, Agellon LB. Regulation of the human cholesterol 7alpha-hydroxylase gene (CYP7A1) by thyroid hormone in transgenic mice. Endocrinology 2004; 145:574-81. [PMID: 14592954 DOI: 10.1210/en.2003-0993] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thyroid hormones exert significant changes in the metabolism of bile acids. However, in humans, the effect of thyroid hormone on cholesterol 7alpha-hydroxylase (cyp7a), the rate- controlling enzyme in the classical bile acid biosynthetic pathway, remains poorly understood and has been difficult to study directly in vivo. Previous studies from our laboratory have shown that the activity of the human cholesterol 7alpha-hydroxylase gene promoter is repressed by T(3) in cultured cells. Accordingly, we hypothesized that T(3) would negatively regulate human CYP7A1 gene expression in vivo. We tested this hypothesis by inducing hypo- and hyperthyroidism in transgenic mice expressing the human CYP7A1 gene. Hypothyroidism did not affect the abundance of human cyp7a mRNA in transgenic mice. In hyperthyroid male mice, human cyp7a mRNA abundance was decreased. No significant change in cyp7a mRNA abundance was observed in hyperthyroid female mice. Gender differences in the amount of cholesterol and bile acids in gallbladder bile were also observed. The data indicate that thyroid hormone can repress the human CYP7A1 gene in transgenic mice, but this effect is dependent on gender in this in vivo model.
Collapse
Affiliation(s)
- Victor A B Drover
- Canadian Institutes of Health Research Group in Molecular and Cell Biology of Lipids, Heritage Medical Research Centre, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
| | | |
Collapse
|
42
|
Lundell K, Wikvall K. Gene structure of pig sterol 12alpha-hydroxylase (CYP8B1) and expression in fetal liver: comparison with expression of taurochenodeoxycholic acid 6alpha-hydroxylase (CYP4A21). Biochim Biophys Acta Mol Cell Biol Lipids 2004; 1634:86-96. [PMID: 14643796 DOI: 10.1016/j.bbalip.2003.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cholic acid is the major trihydroxy bile acid formed in most mammals. The domestic pig (Sus scrofa) is an exception. The bile of adult pig is devoid of cholic acid whereas hyocholic acid is found in amounts equal to that of cholic acid in humans. The pathway leading to formation of hyocholic acid is believed to be species-specific and to have evolved in the pig to compensate for a nonexistent or deficient cholic acid biosynthesis. However, a high level of cholic acid has recently been found in the bile of fetal pig. Here we describe that a gene encoding the key enzyme in cholic acid biosynthesis, the sterol 12alpha-hydroxylase (CYP8B1), is in fact present in the pig genome. The deduced amino acid sequence shows 81% identity to the human and rabbit orthologues. CYP8B1 mRNA is expressed at significant levels in fetal pig liver. Both CYP8B1 and the key enzyme in hyocholic acid formation, taurochenodeoxycholic acid 6alpha-hydroxylase (CYP4A21), were found to be expressed in pig liver in a developmental-dependent but opposite fashion.
Collapse
Affiliation(s)
- Kerstin Lundell
- Division of Biochemistry, Department of Pharmaceutical Biosciences, University of Uppsala, Box 578, S-751 23 Uppsala, Sweden.
| | | |
Collapse
|
43
|
Abstract
The synthesis and excretion of bile acids comprise the major pathway of cholesterol catabolism in mammals. Synthesis provides a direct means of converting cholesterol, which is both hydrophobic and insoluble, into a water-soluble and readily excreted molecule, the bile acid. The biosynthetic steps that accomplish this transformation also confer detergent properties to the bile acid, which are exploited by the body to facilitate the secretion of cholesterol from the liver. This role in the elimination of cholesterol is counterbalanced by the ability of bile acids to solubilize dietary cholesterol and essential nutrients and to promote their delivery to the liver. The synthesis of a full complement of bile acids requires 17 enzymes. The expression of selected enzymes in the pathway is tightly regulated by nuclear hormone receptors and other transcription factors, which ensure a constant supply of bile acids in an ever changing metabolic environment. Inherited mutations that impair bile acid synthesis cause a spectrum of human disease; this ranges from liver failure in early childhood to progressive neuropathy in adults.
Collapse
Affiliation(s)
- David W Russell
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390-9046, USA.
| |
Collapse
|
44
|
Souidi M, Dubrac S, Parquet M, Milliat F, Férézou J, Sérougne C, Loison C, Riottot M, Boudem N, Bécue T, Lutton C. Effects of dietary 27-hydroxycholesterol on cholesterol metabolism and bile acid biosynthesis in the hamster. Can J Physiol Pharmacol 2003; 81:854-63. [PMID: 14614521 DOI: 10.1139/y03-079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
27-hydroxycholesterol (27OH-Chol) is an important endogenous oxysterol resulting from the action of sterol 27-hydroxylase (CYP27A1) on cholesterol in the liver and numerous extrahepatic tissues. It may act as a modulator of cholesterol and bile acid metabolism. The effects of 27OH-Chol on the main enzymes and receptors of cholesterol metabolism were investigated by feeding male hamsters a diet supplemented with 27OH-Chol (0.1% w/w) for 1 week. Intestinal scavenger class B, type I (SR-BI) protein level was decreased (65%), but hepatic expression was increased (+34%). Liver 3β-hydroxy-3β-methyl glutaryl coenzyme A reductase (58%), cholesterol 7α-hydroxylase (54%), oxysterol 7α-hydroxylase (44%), and sterol 12α-hydroxylase (70%) activities were all decreased. Bile acid composition was changed (fourfold increase in the chenodeoxycholic/cholic acid ratio). This study demonstrates that dietary 27OH-Chol modulates major enzymes of cholesterol metabolism and alters the biliary bile acid profile, making it more hydrophobic, at least at this level of intake. Its effects on SR-BI protein levels are organ dependent. The properties of 27OH-Chol or its metabolites on cholesterol metabolism probably result from the activation of specific transcription factors. Key words: cholesterol 7α-hydroxylase (CYP7A1), sterol 12α-hydroxylase (CYP8B1), sterol 27-hydroxylase (CYP27A1), 3β-hydroxy-3β-methyl glutaryl coenzyme A reductase (HMGCoAR), scavenger receptor class B type I (SR-BI).
Collapse
Affiliation(s)
- Maâmar Souidi
- Laboratory of Physiology and Nutrition, Bâtiment 447, Université Paris-Sud, 91405 Orsay CEDEX, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Yao L, Dawson PA, Woollett LA. Increases in biliary cholesterol-to-bile acid ratio in pregnant hamsters fed low and high levels of cholesterol. Am J Physiol Gastrointest Liver Physiol 2003; 284:G263-8. [PMID: 12529268 DOI: 10.1152/ajpgi.00332.2002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gallstones develop when the secretion of cholesterol is elevated compared with the secretion of bile acids into bile. One of the risk factors for the formation of gallstones is pregnancy. Because the pregnancy-induced increase in hepatic cholesterol synthesis rates could play a critical role in the development of cholesterol stones, the aim of the present study was to determine whether stone formation, as assessed by the ratio of cholesterol to bile acids in bile, could be ablated by blocking the pregnancy-induced increase in hepatic sterol synthesis rates. Golden Syrian hamsters were fed either ground chow or chow supplemented with 0.5% cholesterol for 3 wk and studied in the nonpregnant state or in late gestation. In chow-fed animals, a 1.6-fold increase in the ratio of cholesterol to bile acids occurred simultaneously with a sevenfold increase in hepatic sterol synthesis rate and a ninefold increase in the amount of newly synthesized cholesterol secreted into the bile in late gestation. In the cholesterol-fed dams, an increase in the ratio of cholesterol to bile acids occurred even with the lack of induction of hepatic sterol synthesis rates during pregnancy. Thus it appears that the marked induction of hepatic sterol synthesis rates during gestation is not essential for the pregnancy-induced cholesterol saturation of bile when cholesterol is fed to animals.
Collapse
Affiliation(s)
- Lihang Yao
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio 45267-0529, USA
| | | | | |
Collapse
|
46
|
Kosters A, Jirsa M, Groen AK. Genetic background of cholesterol gallstone disease. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1637:1-19. [PMID: 12527402 DOI: 10.1016/s0925-4439(02)00173-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cholesterol gallstone formation is a multifactorial process involving a multitude of metabolic pathways. The primary pathogenic factor is hypersecretion of free cholesterol into bile. For people living in the Western Hemisphere, this is almost a normal condition, certainly in the elderly, which explains the very high incidence of gallstone disease. It is probably because the multifactorial background genes responsible for the high incidence have not yet been identified, despite the fact that genetic factors clearly play a role. Analysis of the many pathways involved in biliary cholesterol secretion reveals many potential candidates and considering the progress in unraveling the regulatory mechanisms of the responsible genes, identification of the primary gallstone genes will be successful in the near future.
Collapse
Affiliation(s)
- Astrid Kosters
- Department of Experimental Hepatology, AMC Liver Center S1-172, Academic Medical Center, Meibergdreef 69-71, 1105 BK, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
47
|
Matsuzaki Y, Bouscarel B, Ikegami T, Honda A, Doy M, Ceryak S, Fukushima S, Yoshida S, Shoda J, Tanaka N. Selective inhibition of CYP27A1 and of chenodeoxycholic acid synthesis in cholestatic hamster liver. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1588:139-48. [PMID: 12385778 DOI: 10.1016/s0925-4439(02)00157-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The aim of this study was to explore the regulation of serum cholic acid (CA)/chenodeoxycholic acid (CDCA) ratio in cholestatic hamster induced by ligation of the common bile duct for 48 h. The serum concentration of total bile acids and CA/CDCA ratio were significantly elevated, and the serum proportion of unconjugated bile acids to total bile acids was reduced in the cholestatic hamster similar to that in patients with obstructive jaundice. The hepatic CA/CDCA ratio increased from 3.6 to 11.0 (P<0.05) along with a 2.9-fold elevation in CA concentration (P<0.05) while the CDCA level remained unchanged. The hepatic mRNA and protein level as well as microsomal activity of the cholesterol 7alpha-hydroxylase, 7alpha-hydroxy-4-cholesten-3-one 12alpha-hydroxylase and 5beta-cholestane-3alpha,7alpha,12alpha-triol 25-hydroxylase were not significantly affected in cholestatic hamsters. In contrast, the mitochondrial activity and enzyme mass of the sterol 27-hydroxylase were significantly reduced, while its mRNA levels remained normal in bile duct-ligated hamster. In conclusion, bile acid biosynthetic pathway via mitochondrial sterol 27-hydroxylase was preferentially inhibited in bile duct-ligated hamsters. The suppression of CYP27A1 is, at least in part, responsible for the relative decreased production of CDCA and increased CA/CDCA ratio in the liver, bile and serum of cholestatic hamsters.
Collapse
Affiliation(s)
- Yasushi Matsuzaki
- Department of Gastroenterology and Hepatology, Institute of Clinical Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba City, 305-8575 Ibaraki, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Gerbod-Giannone MC, Del Castillo-Olivares A, Janciauskiene S, Gil G, Hylemon PB. Suppression of cholesterol 7alpha-hydroxylase transcription and bile acid synthesis by an alpha1-antitrypsin peptide via interaction with alpha1-fetoprotein transcription factor. J Biol Chem 2002; 277:42973-80. [PMID: 12223476 DOI: 10.1074/jbc.m205089200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
alpha1-Antitrypsin (alpha1-AT) is a serum protease inhibitor that is synthesized mainly in the liver, and its rate of synthesis markedly increases in response to inflammation. This increase in alpha1-AT synthesis results in an increase in peptides, like its carboxyl-terminal C-36 peptide (C-36), resulting from alpha1-AT cleavage by proteases. Atherosclerosis is a form of chronic inflammation, and one of the risk factors is elevated plasma cholesterol levels. Because of the correlation between atherosclerosis, plasma cholesterol content, inflammation, and alpha1-AT rate of synthesis, we investigated the effect of the C-36 serpin peptide on hepatic bile acid biosynthesis. We discovered that C-36 is a powerful and specific transcriptional down-regulator of bile acid synthesis in primary rat hepatocytes, through inhibition of the cholesterol 7alpha-hydroxylase/CYP7A1 (7alpha-hydroxylase) promoter. Mice injected with the C-36 peptide also showed a decrease in 7alpha-hydroxylase mRNA. A mutated but very similar peptide did not have any effect on 7alpha-hydroxylase mRNA or its promoter. The sterol 12alpha-hydroxylase/CYP8B1 (12alpha-hydroxylase) promoter is also down-regulated by the C-36 peptide in HepG2 cells but not by the mutated peptide. The DNA element involved in the C-36-mediated regulation of 7alpha- and 12alpha-hydroxylase promoters mapped to the alpha1-fetoprotein transcription factor (FTF) site in both promoters. The C-36 peptide prevented binding of FTF to its target DNA recognition site by direct interaction with FTF. We hypothesize that the C-36 peptide specifically interacts with FTF and induces a conformational change that results in loss of its DNA binding ability, which results in suppression of 7alpha- and 12alpha-hydroxylase transcription. These results suggest that peptides derived from specific serum proteins may alter hepatic gene expression in a highly specific manner.
Collapse
Affiliation(s)
- Marie-Christine Gerbod-Giannone
- Department of Microbiology and Immunology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
Bile acids derived from cholesterol and oxysterols derived from cholesterol and bile acid synthesis pathways are signaling molecules that regulate cholesterol homeostasis in mammals. Many nuclear receptors play pivotal roles in the regulation of bile acid and cholesterol metabolism. Bile acids activate the farnesoid X receptor (FXR) to inhibit transcription of the gene for cholesterol 7alpha-hydroxylase, and stimulate excretion and transport of bile acids. Therefore, FXR is a bile acid sensor that protects liver from accumulation of toxic bile acids and xenobiotics. Oxysterols activate the liver orphan receptors (LXR) to induce cholesterol 7alpha-hydroxylase and ATP-binding cassette family of transporters and thus promote reverse cholesterol transport from the peripheral tissues to the liver for degradation to bile acids. LXR also induces the sterol response element binding protein-1c that regulates lipogenesis. Therefore, FXR and LXR play critical roles in coordinate control of bile acid, cholesterol, and triglyceride metabolism to maintain lipid homeostasis. Nuclear receptors and bile acid/oxysterol-regulated genes are potential targets for developing drug therapies for lowering serum cholesterol and triglycerides and treating cardiovascular and liver diseases.
Collapse
Affiliation(s)
- John Y L Chiang
- Department of Biochemistry and Molecular Pathology, Northeastern Ohio Universities College of Medicine, Rootstown, Ohio 44272, USA.
| |
Collapse
|
50
|
Yang Y, Zhang M, Eggertsen G, Chiang JYL. On the mechanism of bile acid inhibition of rat sterol 12alpha-hydroxylase gene (CYP8B1) transcription: roles of alpha-fetoprotein transcription factor and hepatocyte nuclear factor 4alpha. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1583:63-73. [PMID: 12069850 DOI: 10.1016/s1388-1981(02)00186-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The sterol 12alpha-hydroxylase (CYP8B1) is a key enzyme of the bile acid biosynthetic pathway. It regulates the composition of bile acids in bile, i.e. ratio between cholic acid (CA) and chenodeoxycholic acid (CDCA). In similarity with cholesterol 7alpha-hydroxylase (CYP7A1), this enzyme is subjected to a negative feedback regulation by bile acids. It has been recently reported that bile acid-activated farnesoid X receptor (FXR) induces the small heterodimer partner (SHP) that interacts with alpha-fetoprotein transcription factor (FTF) and down-regulates CYP7A1 transcription. We studied whether the same mechanism also regulated rat CYP8B1 gene transcription. Feeding rats with CDCA caused a 40-50% decrease of CYP8B1 and hepatocyte nuclear factor 4alpha (HNF4alpha) mRNA expression levels. This was associated with an increase in FTF mRNA expression, but SHP mRNA expression was not altered. Electrophoretic mobility shift assay (EMSA) and transient transfection assay of promoter/reporter genes coupled to mutagenesis analysis identified a putative bile acid response element (BARE) that has an HNF4alpha binding site embedded in two overlapping FTF binding sites. Mutation of the HNF4alpha binding site markedly reduced basal promoter activity and its repression by bile acids. Cotransfection with FTF strongly repressed CYP8B1 transcription. Interestingly, HNF4alpha could overcome the inhibitory effects of FTF and bile acids. We conclude that FTF and HNF4alpha not only play critical roles on CYP8B1 gene transcription, but also mediate bile acid feedback inhibition. This study reveals a novel mechanism by which bile acids inhibit rat CYP8B1 gene transcription by inducing FTF and inhibiting HNF4alpha expression.
Collapse
Affiliation(s)
- Yizeng Yang
- Department of Biochemistry and Molecular Pathology, Northeastern Ohio Universities College of Medicine, Rootstown 44272, USA
| | | | | | | |
Collapse
|