1
|
Xiao W, Li Z, Wang Y, Yongbo X, Li W, Li J, He M, Feng Y. Multiomics combined analysis reveals protective effect of 7-O-α-L-rhamnopyranosyl-kaempferol-3-O-β-D-glucopyranoside on autoimmune hepatitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156460. [PMID: 39923428 DOI: 10.1016/j.phymed.2025.156460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/03/2024] [Accepted: 02/01/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Autoimmune hepatitis (AIH) seriously endangers human health. Therefore, it is urgent to find new therapeutic drugs and targets for AIH. In this context, 7-O-α-L-rhamnopyranosyl-kaempferol-3-O-β-D-glucopyranoside (KGR), a flavonoid compound found in Embelia laeta (L.) Mez, has not been evaluated for its efficacy. OBJECTIVE This study aimed to investigate the therapeutic effect and mechanisms of KGR on AIH. RESEARCH DESIGN Concanavalin A (Con A) was used to establish a mouse AIH model. Molecular biology methods were used to evaluate the efficacy of KGR and transcriptomics, proteomics, and metabolomics were innovatively combined to revel the mechanism of action of KGR against AIH, which was verified by experiments. RESULTS Mouse liver sections demonstrated that KGR reduced the degree of degeneration and necrosis in liver cells in mice. Compared with the Con A group, KGR significantly reduced serum aminotransferase levels, inhibited the release of proinflammatory cytokines in the liver tissue, and inhibited oxidative stress (OS) by reducing malondialdehyde level and enhancing superoxide dismutase activity. Finally, multiomics revealed that primary bile acids synthesis and the FXR-TLR4/MYD88/JNK signaling pathway may be the regulatory targets of KGR. CONCLUSION The study results demonstrated that KGR inhibited OS and inflammatory responses by regulating primary bile acid synthesis and thereby inhibiting the FXR-TLR4/MYD88/JNK signaling pathway, and had a protective effect on Con A-induced AIH.
Collapse
Affiliation(s)
- Wei Xiao
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Zhiqiang Li
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Yilei Wang
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Xizi Yongbo
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Wanting Li
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Junmao Li
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, No. 56 Yangming Road, Nanchang 330006, Jiangxi, PR China
| | - Mingzhen He
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, No. 56 Yangming Road, Nanchang 330006, Jiangxi, PR China.
| | - Yulin Feng
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, No. 56 Yangming Road, Nanchang 330006, Jiangxi, PR China.
| |
Collapse
|
2
|
Fleishman JS, Kumar S. Bile acid metabolism and signaling in health and disease: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:97. [PMID: 38664391 PMCID: PMC11045871 DOI: 10.1038/s41392-024-01811-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
Bile acids, once considered mere dietary surfactants, now emerge as critical modulators of macronutrient (lipid, carbohydrate, protein) metabolism and the systemic pro-inflammatory/anti-inflammatory balance. Bile acid metabolism and signaling pathways play a crucial role in protecting against, or if aberrant, inducing cardiometabolic, inflammatory, and neoplastic conditions, strongly influencing health and disease. No curative treatment exists for any bile acid influenced disease, while the most promising and well-developed bile acid therapeutic was recently rejected by the FDA. Here, we provide a bottom-up approach on bile acids, mechanistically explaining their biochemistry, physiology, and pharmacology at canonical and non-canonical receptors. Using this mechanistic model of bile acids, we explain how abnormal bile acid physiology drives disease pathogenesis, emphasizing how ceramide synthesis may serve as a unifying pathogenic feature for cardiometabolic diseases. We provide an in-depth summary on pre-existing bile acid receptor modulators, explain their shortcomings, and propose solutions for how they may be remedied. Lastly, we rationalize novel targets for further translational drug discovery and provide future perspectives. Rather than dismissing bile acid therapeutics due to recent setbacks, we believe that there is immense clinical potential and a high likelihood for the future success of bile acid therapeutics.
Collapse
Affiliation(s)
- Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| |
Collapse
|
3
|
Sun Y, Zhang L, Jiang Z. The role of peroxisome proliferator-activated receptors in the regulation of bile acid metabolism. Basic Clin Pharmacol Toxicol 2024; 134:315-324. [PMID: 38048777 DOI: 10.1111/bcpt.13971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023]
Abstract
Bile acids are synthesized from cholesterol in the liver. Dysregulation of bile acid homeostasis, characterized by excessive accumulation in the liver, gallbladder and blood, can lead to hepatocellular damage and the development of cholestatic liver disease. Nuclear receptors play a crucial role in the control of bile acid metabolism by efficiently regulating bile acid synthesis and transport in the liver. Among these receptors, peroxisome proliferator-activated receptor (PPAR), a ligand-activated transcription factor belonging to the nuclear hormone receptor superfamily, controls the expression of genes involved in adipogenesis, lipid metabolism, inflammation and glucose homeostasis and has emerged as a potential therapeutic target for the treatment of the metabolic syndrome in the past two decades. Emerging evidence suggests that PPAR activation holds promise as a therapeutic target for cholestatic liver disease, as it affects both bile acid production and transport. This review provides a comprehensive overview of recent advances in elucidating the role of PPAR in the regulation of bile acid metabolism, highlighting the current position of PPAR agonists in the treatment of primary biliary cholangitis. By summarizing the specific regulatory effects of PPAR on bile acids, this review contributes to the exploration of novel therapeutic strategies for cholestatic liver diseases.
Collapse
Affiliation(s)
- Yuqing Sun
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Pharmaceutical Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Pharmaceutical Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Pharmaceutical Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
4
|
Yu X, Zhang Y, Cogliati B, Klaassen CD, Kumar S, Cheng X, Bu P. Distinct bile acid alterations in response to a single administration of PFOA and PFDA in mice. Toxicology 2024; 502:153719. [PMID: 38181850 PMCID: PMC10922993 DOI: 10.1016/j.tox.2023.153719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/22/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Per- and polyfluoroalkyl substances (PFASs), a group of synthetic chemicals that were once widely used for industrial purposes and in consumer products, are widely found in the environment and in human blood due to their extraordinary resistance to degradation. Once inside the body, PFASs can activate nuclear receptors such as PPARα and CAR. The present study aimed to investigate the impact of perfluorooctanoic acid (PFOA) and perfluorodecanoic acid (PFDA) on liver structure and functions, as well as bile acid homeostasis in mice. A single administration of 0.1 mmole/kg of PFDA, not PFOA, elevated serum ALT and bilirubin levels and caused cholestasis in WT mice. PFDA increased total and various bile acid species in serum but decreased them in the liver. Furthermore, in mouse livers, PFDA, not PFOA, down-regulated mRNA expression of uptake transporters (Ntcp, Oatp1a1, 1a4, 1b2, and 2b1) but induced efflux transporters (Bcrp, Mdr2, and Mrp2-4). In addition, PFDA, not PFOA, decreased Cyp7a1, 7b1, 8b1, and 27a1 mRNA expression in mouse livers with concomitant hepatic accumulation of cholesterol. In contrast, in PPARα-null mice, PFDA did not increase serum ALT, bilirubin, or total bile acids, but produced prominent hepatosteatosis; and the observed PFDA-induced expression changes of transporters and Cyps in WT mice were largely attenuated or abolished. In CAR-null mice, the observed PFDA-induced bile acid alterations in WT mice were mostly sustained. These results indicate that, at the dose employed, PFDA has more negative effects than PFOA on liver function. PPARα appears to play a major role in mediating most of PFDA-induced effects, which were absent or attenuated in PPARα-null mice. Lack of PPARα, however, exacerbated hepatic steatosis. Our findings indicate separated roles of PPARα in mediating the adaptive responses to PFDA: protective against hepatosteatosis but exacerbating cholestasis.
Collapse
Affiliation(s)
- Xiaoxiao Yu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Jamaica, NY 11439, United States
| | - Youcai Zhang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Bruno Cogliati
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029, United States; Department of Pathology, School of Veterinary Medicine and Animal Science, University of Sao Paulo, 05508-270, Sao Paulo, Brazil
| | - Curtis D Klaassen
- University of Kansas Medical Center, Kansas City, KS 66103, United States
| | - Sanaya Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Jamaica, NY 11439, United States
| | - Xingguo Cheng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Jamaica, NY 11439, United States
| | - Pengli Bu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Jamaica, NY 11439, United States.
| |
Collapse
|
5
|
Rausch M, Samodelov SL, Visentin M, Kullak-Ublick GA. The Farnesoid X Receptor as a Master Regulator of Hepatotoxicity. Int J Mol Sci 2022; 23:ijms232213967. [PMID: 36430444 PMCID: PMC9695947 DOI: 10.3390/ijms232213967] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The nuclear receptor farnesoid X receptor (FXR, NR1H4) is a bile acid (BA) sensor that links the enterohepatic circuit that regulates BA metabolism and elimination to systemic lipid homeostasis. Furthermore, FXR represents a real guardian of the hepatic function, preserving, in a multifactorial fashion, the integrity and function of hepatocytes from chronic and acute insults. This review summarizes how FXR modulates the expression of pathway-specific as well as polyspecific transporters and enzymes, thereby acting at the interface of BA, lipid and drug metabolism, and influencing the onset and progression of hepatotoxicity of varying etiopathogeneses. Furthermore, this review article provides an overview of the advances and the clinical development of FXR agonists in the treatment of liver diseases.
Collapse
|
6
|
Ma Y, Lu L, Tan K, Li Z, Guo T, Wu Y, Wu W, Zheng L, Fan F, Mo J, Gong Z. Reduced peroxisome proliferator-activated receptor-α and bile acid nuclear receptor NR1H4/FXR may affect the hepatic immune microenvironment of biliary atresia. Front Immunol 2022; 13:875593. [PMID: 36090996 PMCID: PMC9454303 DOI: 10.3389/fimmu.2022.875593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/03/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Biliary atresia (BA) is a childhood liver disease characterized by fibrous obstruction and obstruction of the extrahepatic biliary system and is one of the most common and serious biliary disorders in infants. Significant inflammation and fibrosis of the liver and biliary tract are the most prominent features, regardless of the initial damage to the BA. Abnormalities in innate or adaptive immunity have been found in human patients and mouse models of BA. We previously reported that children with BA had abnormal lipid metabolism, including free serum carnitine. OBJECTIVE To study gene and protein expression levels of the hepatic peroxisome proliferator-activated receptor-α (PPARα) signaling pathway and farnesoid X receptor (FXR) in BA and BA fibrosis, and assess their clinical values. METHODS Low expression of PPARα and NR1H4 (FXR) in BA were validated in the Gene Expression Omnibus database. Functional differences were determined by gene set enrichment analysis based on of PPARα and NR1H4 expression. BA patients from GSE46960 were divided into two clusters by using consensus clustering according to PPARα, NR1H4, and SMAD3 expression levels, and immunoinfiltration analysis was performed. Finally, 58 cases treated in our hospital were used for experimental verification. (IHC: 10 Biliary atresia, 10 choledochal cysts; PCR: 10 Biliary atresia, 14 choledochal cysts; WB: 10 Biliary atresia, 4 choledochal cysts). RESULTS Bioinformatics analysis showed that the expression of PPARα, CYP7A1 and NR1H4 (FXR) in the biliary atresia group was significantly lower than in the control group. More BA-specific pathways, including TGFβ signaling pathway, P53 signaling pathway, PI3K-AKT-mTOR signaling pathway, etc., are enriched in BA patients with low PPARα and NR1H4 expression. In addition, low NR1H4 expression is abundant in inflammatory responses, IL6/STAT3 signaling pathways, early estrogen responses, IL2 STAT5 signaling pathways, and TGFβ signaling pathways. The TGFβ signaling pathway was significant in both groups. According to the expression of PPARα, NR1H4 and SMAD3, a key node in TGFβ pathway, BA patients were divided into two clusters using consensus clustering. In cluster 2, SMAD3 expression was high, and PPARα and NR1H4 expression were low. In contrast to cluster 1, immune cell infiltration was higher in cluster 2, which was confirmed by immunohistochemistry. The mRNA and protein levels of PPARα and NR1H4 in BA patients were lower than in the control group by immunohistochemistry, Western blot analysis and real-time PCR. CONCLUSIONS The downregulation of PPARα and NR1H4 (FXR) signaling pathway may be closely related to biliary atresia.
Collapse
Affiliation(s)
- Yingxuan Ma
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Li Lu
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Kezhe Tan
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi Li
- Pathology Department, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Guo
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Yibo Wu
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Wu
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Lulu Zheng
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Feilong Fan
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayu Mo
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenhua Gong
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Zhou S, You H, Qiu S, Yu D, Bai Y, He J, Cao H, Che Q, Guo J, Su Z. A new perspective on NAFLD: Focusing on the crosstalk between peroxisome proliferator-activated receptor alpha (PPARα) and farnesoid X receptor (FXR). Biomed Pharmacother 2022; 154:113577. [PMID: 35988420 DOI: 10.1016/j.biopha.2022.113577] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 11/19/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is primarily caused by abnormal lipid metabolism and the accumulation of triglycerides in the liver. NAFLD is also associated with hepatic steatosis and nutritional and energy imbalances and is a chronic liver disease associated with a number of factors. Nuclear receptors play a key role in balancing energy and nutrient metabolism, and the peroxisome proliferator-activated receptor alpha (PPARα) and farnesoid X receptor (FXR) regulate lipid metabolism genes, controlling hepatocyte lipid utilization and regulating bile acid (BA) synthesis and transport. They play an important role in lipid metabolism and BA homeostasis. At present, PPARα and FXR are the most promising targets for the treatment of NAFLD among nuclear receptors. This review focuses on the crosstalk mechanisms and transcriptional regulation of PPARα and FXR in the pathogenesis of NAFLD and summarizes PPARα and FXR drugs in clinical trials, laying a theoretical foundation for the targeted treatment of NAFLD and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Shipeng Zhou
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huimin You
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shuting Qiu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dawei Yu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Jincan He
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd, Science City, Guangzhou 510663, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
8
|
Ye X, Zhang T, Han H. PPARα: A potential therapeutic target of cholestasis. Front Pharmacol 2022; 13:916866. [PMID: 35924060 PMCID: PMC9342652 DOI: 10.3389/fphar.2022.916866] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
The accumulation of bile acids in the liver leads to the development of cholestasis and hepatocyte injury. Nuclear receptors control the synthesis and transport of bile acids in the liver. Among them, the farnesoid X receptor (FXR) is the most common receptor studied in treating cholestasis. The activation of this receptor can reduce the amount of bile acid synthesis and decrease the bile acid content in the liver, alleviating cholestasis. Ursodeoxycholic acid (UDCA) and obeticholic acid (OCA) have a FXR excitatory effect, but the unresponsiveness of some patients and the side effect of pruritus seriously affect the results of UDCA or OCA treatment. The activator of peroxisome proliferator-activated receptor alpha (PPARα) has emerged as a new target for controlling the synthesis and transport of bile acids during cholestasis. Moreover, the anti-inflammatory effect of PPARα can effectively reduce cholestatic liver injury, thereby improving patients’ physiological status. Here, we will focus on the function of PPARα and its involvement in the regulation of bile acid transport and metabolism. In addition, the anti-inflammatory effects of PPARα will be discussed in some detail. Finally, we will discuss the application of PPARα agonists for cholestatic liver disorders.
Collapse
Affiliation(s)
- Xiaoyin Ye
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Zhang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Tong Zhang, ; Han Han,
| | - Han Han
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Tong Zhang, ; Han Han,
| |
Collapse
|
9
|
Tremmel R, Nies AT, van Eijck BAC, Handin N, Haag M, Winter S, Büttner FA, Kölz C, Klein F, Mazzola P, Hofmann U, Klein K, Hoffmann P, Nöthen MM, Gaugaz FZ, Artursson P, Schwab M, Schaeffeler E. Hepatic Expression of the Na+-Taurocholate Cotransporting Polypeptide Is Independent from Genetic Variation. Int J Mol Sci 2022; 23:ijms23137468. [PMID: 35806468 PMCID: PMC9267852 DOI: 10.3390/ijms23137468] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
The hepatic Na+-taurocholate cotransporting polypeptide NTCP/SLC10A1 is important for the uptake of bile salts and selected drugs. Its inhibition results in increased systemic bile salt concentrations. NTCP is also the entry receptor for the hepatitis B/D virus. We investigated interindividual hepatic SLC10A1/NTCP expression using various omics technologies. SLC10A1/NTCP mRNA expression/protein abundance was quantified in well-characterized 143 human livers by real-time PCR and LC-MS/MS-based targeted proteomics. Genome-wide SNP arrays and SLC10A1 next-generation sequencing were used for genomic analyses. SLC10A1 DNA methylation was assessed through MALDI-TOF MS. Transcriptomics and untargeted metabolomics (UHPLC-Q-TOF-MS) were correlated to identify NTCP-related metabolic pathways. SLC10A1 mRNA and NTCP protein levels varied 44-fold and 10.4-fold, respectively. Non-genetic factors (e.g., smoking, alcohol consumption) influenced significantly NTCP expression. Genetic variants in SLC10A1 or other genes do not explain expression variability which was validated in livers (n = 50) from The Cancer Genome Atlas. The identified two missense SLC10A1 variants did not impair transport function in transfectants. Specific CpG sites in SLC10A1 as well as single metabolic alterations and pathways (e.g., peroxisomal and bile acid synthesis) were significantly associated with expression. Inter-individual variability of NTCP expression is multifactorial with the contribution of clinical factors, DNA methylation, transcriptional regulation as well as hepatic metabolism, but not genetic variation.
Collapse
Affiliation(s)
- Roman Tremmel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Anne T. Nies
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
- iFIT Cluster of Excellence (EXC2180) “Image Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany
| | - Barbara A. C. van Eijck
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Niklas Handin
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (N.H.); (F.Z.G.); (P.A.)
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Florian A. Büttner
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Charlotte Kölz
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Franziska Klein
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Pascale Mazzola
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Kathrin Klein
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany; (P.H.); (M.M.N.)
- Division of Medical Genetics, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany; (P.H.); (M.M.N.)
- Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany
| | - Fabienne Z. Gaugaz
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (N.H.); (F.Z.G.); (P.A.)
| | - Per Artursson
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (N.H.); (F.Z.G.); (P.A.)
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
- iFIT Cluster of Excellence (EXC2180) “Image Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany
- Departments of Clinical Pharmacology, and of Pharmacy and Biochemistry, University of Tuebingen, 72076 Tuebingen, Germany
- Correspondence: ; Tel.: +49-711-8101-3700
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
- iFIT Cluster of Excellence (EXC2180) “Image Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
10
|
Jiao TY, Ma YD, Guo XZ, Ye YF, Xie C. Bile acid and receptors: biology and drug discovery for nonalcoholic fatty liver disease. Acta Pharmacol Sin 2022; 43:1103-1119. [PMID: 35217817 PMCID: PMC9061718 DOI: 10.1038/s41401-022-00880-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/25/2022] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), a series of liver metabolic disorders manifested by lipid accumulation within hepatocytes, has become the primary cause of chronic liver diseases worldwide. About 20%-30% of NAFLD patients advance to nonalcoholic steatohepatitis (NASH), along with cell death, inflammation response and fibrogenesis. The pathogenesis of NASH is complex and its development is strongly related to multiple metabolic disorders (e.g. obesity, type 2 diabetes and cardiovascular diseases). The clinical outcomes include liver failure and hepatocellular cancer. There is no FDA-approved NASH drug so far, and thus effective therapeutics are urgently needed. Bile acids are synthesized in hepatocytes, transported into the intestine, metabolized by gut bacteria and recirculated back to the liver by the enterohepatic system. They exert pleiotropic roles in the absorption of fats and regulation of metabolism. Studies on the relevance of bile acid disturbance with NASH render it as an etiological factor in NASH pathogenesis. Recent findings on the functional identification of bile acid receptors have led to a further understanding of the pathophysiology of NASH such as metabolic dysregulation and inflammation, and bile acid receptors are recognized as attractive targets for NASH treatment. In this review, we summarize the current knowledge on the role of bile acids and the receptors in the development of NAFLD and NASH, especially the functions of farnesoid X receptor (FXR) in different tissues including liver and intestine. The progress in the development of bile acid and its receptors-based drugs for the treatment of NASH including bile acid analogs and non-bile acid modulators on bile acid metabolism is also discussed.
Collapse
Affiliation(s)
- Ting-Ying Jiao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yuan-di Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Zhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yun-Fei Ye
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
11
|
Reversal of NAFLD After VSG Is Independent of Weight-Loss but RYGB Offers More Efficacy When Maintained on a High-Fat Diet. Obes Surg 2022; 32:2010-2022. [PMID: 35419698 DOI: 10.1007/s11695-022-06053-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/28/2022] [Accepted: 04/07/2022] [Indexed: 12/11/2022]
Abstract
PURPOSE Bariatric surgery is emerging as an effective treatment for obesity and the metabolic syndrome. Recently, we demonstrated that Roux-en-Y gastric bypass (RYGB), but not vertical sleeve gastrectomy (VSG), resulted in improvements to white adipose physiology and enhanced brown adipose functioning. Since beneficial alterations to liver health are also expected after bariatric surgery, comparing the post-operative effects of RYGB and VSG on liver physiology is essential to their application in the treatment of non-alcoholic fatty liver disease (NAFLD). MATERIALS AND METHODS The effects of RYGB and VSG on liver physiology were compared using diet induced mouse model of obesity. High-fat diet (HFD) was administered for 12 weeks after surgery and alterations to liver physiology were assessed. RESULTS Both RYGB and VSG showed decreased liver weight as well as reductions to hepatic cholesterol and triglyceride levels. There were demonstrable improvements to NAFLD activity score (NAS) and fibrosis stage scoring after both surgeries. In RYGB, these beneficial changes to liver function resulted from the downregulation of pro-fibrotic and upregulation anti-fibrotic genes, as well as increased fatty acid oxidation and bile acid flux. For VSG, though similar alterations were observed, they were less potent. However, VSG did significantly downregulate pro-fibrotic genes and showed increased glycogen content paralleled by decreased glycogenolysis which may have contributed to the resolution of NAFLD. CONCLUSION RYGB and VSG improve liver physiology and function, but RYGB is more efficacious. Resolutions of NAFLD in RYGB and VSG are achieved through different processes, independent of weight loss.
Collapse
|
12
|
Choudhuri S, Klaassen CD. Molecular Regulation of Bile Acid Homeostasis. Drug Metab Dispos 2022; 50:425-455. [PMID: 34686523 DOI: 10.1124/dmd.121.000643] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022] Open
Abstract
Bile acids have been known for decades to aid in the digestion and absorption of dietary fats and fat-soluble vitamins in the intestine. The development of gene knockout mice models and transgenic humanized mouse models have helped us understand other functions of bile acids, such as their role in modulating fat, glucose, and energy metabolism, and in the molecular regulation of the synthesis, transport, and homeostasis of bile acids. The G-protein coupled receptor TGR5 regulates the bile acid induced alterations of intermediary metabolism, whereas the nuclear receptor FXR regulates bile acid synthesis and homeostasis. However, this review indicates that unidentified factors in addition to FXR must exist to aid in the regulation of bile acid synthesis and homeostasis. SIGNIFICANCE STATEMENT: This review captures the present understanding of bile acid synthesis, the role of bile acid transporters in the enterohepatic circulation of bile acids, the role of the nuclear receptor FXR on the regulation of bile acid synthesis and bile acid transporters, and the importance of bile acids in activating GPCR signaling via TGR5 to modify intermediary metabolism. This information is useful for developing drugs for the treatment of various hepatic and intestinal diseases, as well as the metabolic syndrome.
Collapse
Affiliation(s)
- Supratim Choudhuri
- Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland (S.C.) and Department of Pharmacology, Toxicology, and Therapeutics, School of Medicine, University of Kansas, Kansas City, Kansas (C.D.K.)
| | - Curtis D Klaassen
- Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland (S.C.) and Department of Pharmacology, Toxicology, and Therapeutics, School of Medicine, University of Kansas, Kansas City, Kansas (C.D.K.)
| |
Collapse
|
13
|
Mice with a deficiency in Peroxisomal Membrane Protein 4 (PXMP4) display mild changes in hepatic lipid metabolism. Sci Rep 2022; 12:2512. [PMID: 35169201 PMCID: PMC8847483 DOI: 10.1038/s41598-022-06479-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/31/2022] [Indexed: 11/08/2022] Open
Abstract
Peroxisomes play an important role in the metabolism of a variety of biomolecules, including lipids and bile acids. Peroxisomal Membrane Protein 4 (PXMP4) is a ubiquitously expressed peroxisomal membrane protein that is transcriptionally regulated by peroxisome proliferator-activated receptor α (PPARα), but its function is still unknown. To investigate the physiological function of PXMP4, we generated a Pxmp4 knockout (Pxmp4-/-) mouse model using CRISPR/Cas9-mediated gene editing. Peroxisome function was studied under standard chow-fed conditions and after stimulation of peroxisomal activity using the PPARα ligand fenofibrate or by using phytol, a metabolite of chlorophyll that undergoes peroxisomal oxidation. Pxmp4-/- mice were viable, fertile, and displayed no changes in peroxisome numbers or morphology under standard conditions. Also, no differences were observed in the plasma levels of products from major peroxisomal pathways, including very long-chain fatty acids (VLCFAs), bile acids (BAs), and BA intermediates di- and trihydroxycholestanoic acid. Although elevated levels of the phytol metabolites phytanic and pristanic acid in Pxmp4-/- mice pointed towards an impairment in peroxisomal α-oxidation capacity, treatment of Pxmp4-/- mice with a phytol-enriched diet did not further increase phytanic/pristanic acid levels. Finally, lipidomic analysis revealed that loss of Pxmp4 decreased hepatic levels of the alkyldiacylglycerol class of neutral ether lipids, particularly those containing polyunsaturated fatty acids. Together, our data show that while PXMP4 is not critical for overall peroxisome function under the conditions tested, it may have a role in the metabolism of (ether)lipids.
Collapse
|
14
|
Liu J, Fan Y, Yu H, Xu T, Zhang C, Zhou L, Li G, Zhang Y. Allopurinol Protects Against Cholestatic Liver Injury in Mice Not Through Depletion of Uric Acid. Toxicol Sci 2021; 181:295-305. [PMID: 33749747 DOI: 10.1093/toxsci/kfab034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cholestasis is one of the most severe manifestations of liver injury and has limited therapeutic options. Allopurinol (AP), an inhibitor of uric acid (UA) synthesis, was reported to prevent liver damage in several liver diseases. However, whether AP protects against intrahepatic cholestatic liver injury and what is the role of UA in the pathogenesis of cholestasis remain unknown. In this study, we reported that AP attenuated liver injury in a mouse model of intrahepatic cholestasis induced by alpha-naphthylisothiocyanate (ANIT). AP showed no significant effect on glutathione depletion, inflammation, or bile acid metabolism in livers of ANIT-treated mice. Instead, AP significantly improved fatty acid β-oxidation in livers of ANIT-treated mice, which was associated with activation of PPARα. The protective effect of AP on cholestatic liver injury was not attributable to the depletion of UA, because both exogenous and endogenous UA prevented liver injury in ANIT-treated mice via inhibition of NF-kB-mediated inflammation. In conclusion, the present study provides a new perspective for the therapeutic use of AP and the role of UA in cholestatic liver injury.
Collapse
Affiliation(s)
- Jing Liu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yang Fan
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Hang Yu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Tong Xu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Chunze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, China
| | - Lijun Zhou
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Gentao Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China.,School of Pharmacy, Weifang Medical University, Shandong 261053, China
| | - Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
15
|
Ning Y, Xu F, Xin R, Yao F. Palmatine regulates bile acid cycle metabolism and maintains intestinal flora balance to maintain stable intestinal barrier. Life Sci 2020; 262:118405. [PMID: 32926925 DOI: 10.1016/j.lfs.2020.118405] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Palmatine (PAL) is a natural isoquinoline alkaloid that has been widely used in the pharmaceutical field. The current study aimed to investigate the function of PAL in improving hyperlipidemia induced by high-fat diet (HFD) in rats. METHODS Biochemical analysis of triglyceride (TG), total cholesterol (TC), low-density lipoprotein-cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDLC) was performed on rats. Total bile acid (TBA) and stool TC and TBA were also measured to assess the changes in total bile acid excretion. RT-qPCR was employed to detect the expression of genes related to bile acid metabolism, and the Western blot assay was used to detect the levels of CYP7A1, ZO-1, ZO-2, and Claudin-1. The siRNA experiment was employed to further investigate whether PAL regulated CYP7A1 through PPARα. Lipopolysaccharide (LPS) and FITC-dextran (FD-4) were also tested to assess the intestinal permeability. RESULTS AL-treated rats had lower TC, TG, LDL-C levels, lower serum TBA levels, and increased fecal TBA and TC levels. Furthermore, CYP7A1 protein expression was up-regulated in PAL-treated rats. Additionally, PAL regulated bile acid metabolism by up-regulating the expression of CYP7A1 and PPARα and down-regulating the expression of FXR. Besides, the area of plasma FD-4 and LPS content in the PAL group were reduced, and the expression of proteins ZO-1, ZO-2 and Claudin-1 related to intestinal permeability was increased. CONCLUSION All in all, PAL could mediate the PPARα-CYP7A1 pathway to maintain the balance of intestinal flora, regulate the bile acid metabolism, and reduce the blood lipids of rats, thereby protecting against hyperlipidemia.
Collapse
Affiliation(s)
- Yayuan Ning
- Department of Cardiology, the Second Hospital of Jilin University, Changchun 130041, PR China
| | - Fei Xu
- Department of Acupuncture, the Second Hospital of Jilin University, Changchun 130041, PR China
| | - Rui Xin
- Department of Radiology, the Second Hospital of Jilin University, Changchun 130041, PR China
| | - Fang Yao
- Department of Cardiology, the Second Hospital of Jilin University, Changchun 130041, PR China.
| |
Collapse
|
16
|
Zhang Y, Lickteig AJ, Liu J, Csanaky IL, Klaassen CD. Effects of ablation and activation of Nrf2 on bile acid homeostasis in male mice. Toxicol Appl Pharmacol 2020; 403:115170. [PMID: 32738332 DOI: 10.1016/j.taap.2020.115170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 01/16/2023]
Abstract
The role of nuclear factor erythroid 2-related factor 2 (Nrf2) in bile acid (BA) homeostasis remains controversial. In this study, activation of Nrf2 was achieved either pharmacologically by CDDO-imidazolide (CDDO-Im) or genetically through a "gene dose-response" model consisting of Nrf2-null, wild-type (WT), Keap1-knockdown (Keap1-KD), and Keap1-hepatocyte knockout (Keap1-HKO) mice. In WT mice, CDDO-Im increased bile flow and decreased hepatic BAs, which was associated with a down-regulation of the canalicular BA efflux transporter Bsep and an increase in biliary BA excretion. In contrast, hepatic Bsep and biliary BA excretion were not altered in Keap1-KD or Keap1-HKO mice, suggesting that Nrf2 is not important for regulating Bsep or BA-dependent bile flow. In contrast, hepatic Mrp2 and Mrp3 were up-regulated by both pharmacological and genetic activations of Nrf2. Furthermore, ileal BA transporters (Asbt and Ostβ) and cholesterol transporters (Abcg5 and Abcg8) were down-regulated by both pharmacological and genetic activations of Nrf2, suggesting a role of Nrf2 in intestinal absorption of BAs and cholesterol. In Nrf2-null mice, CDDO-Im down-regulated hepatic BA uptake transporters (Ntcp, Oatp1a1, and Oatp1b2), leading to a 39-fold increase of serum BAs. To conclude, the present study demonstrates that activation of Nrf2 in mice up-regulates Mrp2 and Mrp3 in the liver and down-regulates BA and cholesterol transporters in the intestine.
Collapse
Affiliation(s)
- Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China.
| | - Andrew J Lickteig
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jing Liu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Iván L Csanaky
- Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Division of Gastroenterology, Children's Mercy Hospital & Clinics, Kansas City, MO 64108, USA; Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
17
|
Yang Y, Zhao Y, Li W, Wu Y, Wang X, Wang Y, Liu T, Ye T, Xie Y, Cheng Z, He J, Bai P, Zhang Y, Ouyang L. Emerging targets and potential therapeutic agents in non-alcoholic fatty liver disease treatment. Eur J Med Chem 2020; 197:112311. [PMID: 32339855 DOI: 10.1016/j.ejmech.2020.112311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 03/29/2020] [Accepted: 04/04/2020] [Indexed: 02/08/2023]
Abstract
Nonalcoholic Fatty Liver Disease (NAFLD) is the most common chronic liver disease in the world, which is characterized by liver fat accumulation unrelated to excessive drinking. Indeed, it attracts growing attention and becomes a global health problem. Due to the complexity of the NAFLD pathogenic mechanism, no related drugs were approved by Food and Drug Administration (FDA) till now. However, it is encouraging that a series of candidate drugs have entered the clinical trial stage with expectation to treat NAFLD. In this review, we summarized the main pathways and pathogenic mechanisms of NAFLD, as well as introduced the main potential therapeutic targets and the corresponding compounds involved in metabolism, inflammation and fibrosis. Furthermore, we also discuss the progress of these compounds, such as drug design and optimization, the choice of pharmacological properties and druglikeness, and the analysis of structure-activity relationship. This review offers a medium on future drug design and development, to be beneficial to relevant studies.
Collapse
Affiliation(s)
- Yu Yang
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yu Zhao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenzhen Li
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yuyao Wu
- West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Wang
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yijie Wang
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Tingmei Liu
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Tinghong Ye
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Zhiqiang Cheng
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jun He
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| | - Peng Bai
- Department of Forensic Genetics, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
| | - Yiwen Zhang
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| | - Liang Ouyang
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| |
Collapse
|
18
|
Cӑtoi AF, Pârvu AE, Mironiuc A, Silaghi H, Pop ID, Andreicuț AD. Ultra-Early and Early Changes in Bile Acids and Insulin After Sleeve Gastrectomy Among Obese Patients. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E757. [PMID: 31766784 PMCID: PMC6955910 DOI: 10.3390/medicina55120757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE In obese patients, sleeve gastrectomy (SG) has shown mixed results on bile acid (BA) values. The aim of our study was to examine the potential ultra-early and early changes of the circulating total BA in relation with the changes of insulin resistance (IR) in obese patients submitted to laparoscopic SG. Materials and Methods: Twenty-four obese subjects were investigated for body mass index (BMI), total fasting BA, insulin, homeostasis model assessment of insulin resistance (HOMA-IR), and leptin before and at 7 and 30 d after SG. Results: After surgery, mean BMI decreased at the first (p < 0.001) and at the second time point (p < 0.001) relative to baseline. Total fasting BA values did not change significantly at 7 d (p = 0.938) and at 30 d (p = 0.289) after SG. No significant changes were found at 7 d (p = 0.194, p = 0.34) and 30 d (p = 0.329, p = 0.151) after surgery regarding fasting insulin and HOMA-IR, respectively. However, a trend of increased total fasting BA and decreased fasting insulin and HOMA- after laparoscopic SG has been found. Negative correlations between total fasting BA and insulin (r = -0.807, p = 0.009), HOMA-IR (r = -0.855, p = 0.014), and blood glucose (r = -0.761, p = 0.047), respectively, were observed at one month after SG. Conclusion: In conclusion, here, we found a lack of significant changes in total fasting BA, insulin, and HOMA-IR ultra-early and early after SG, which precluded us to consider a possible relation between the variations of BA and IR. However, the presence of the tendency for total fasting BA to increase and for insulin and HOMA-IR to decrease, as well as of the negative correlations one month after laparoscopic SG, suggest that this surgery brings about some changes that point towards the existence, and possibly towards the restoration, at least to some extent, of the link between BA and glucose metabolism.
Collapse
Affiliation(s)
- Adriana Florinela Cӑtoi
- Department of Pathophysiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.E.P.); (A.D.A.)
| | - Alina Elena Pârvu
- Department of Pathophysiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.E.P.); (A.D.A.)
| | - Aurel Mironiuc
- 2nd Surgical Clinic, Department of Surgery, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Horațiu Silaghi
- 5th Surgical Clinic, Department of Surgery, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Ioana Delia Pop
- Department of Exact Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania;
| | - Andra Diana Andreicuț
- Department of Pathophysiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.E.P.); (A.D.A.)
| |
Collapse
|
19
|
Lickteig AJ, Zhang Y, Klaassen CD, Csanaky IL. Effects of Absence of Constitutive Androstane Receptor (CAR) on Bile Acid Homeostasis in Male and Female Mice. Toxicol Sci 2019; 171:132-145. [PMID: 31225615 PMCID: PMC6735724 DOI: 10.1093/toxsci/kfz143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 01/05/2023] Open
Abstract
Accumulation of BAs in hepatocytes has a role in liver disease and also in drug-induced liver injury. The Constitutive Androstane Receptor (CAR) has been shown to protect against BA-induced liver injury. The polymorphism of CAR has recently been shown to modify the pharmacokinetics and pharmacodynamics of various drugs. Thus it was hypothesized that polymorphism of CAR may also influence BA homeostasis. Using CAR-null and WT mice, this study modeled the potential consequences of CAR polymorphism on BA homeostasis. Our previous study showed that chemical activation of CAR decreases the total BA concentrations in livers of mice. Surprisingly the absence of CAR also decreased the BA concentrations in livers of mice, but to a lesser extent than in CAR-activated mice. Neither CAR activation nor elimination of CAR altered the biliary excretion of total BAs, but CAR activation increased the proportion of 6-OH BAs (TMCA), whereas the lack of CAR increased the excretion of TCA, TCDCA and TDCA. Serum BA concentrations did not parallel the decrease in BA concentrations in the liver in either the mice after CAR activation or mice lacking CAR. Gene expression of BA synthesis, transporter and regulator genes were mainly similar in livers of CAR-null and WT mice. In summary, CAR activation decreases primarily the 12-OH BA concentrations in liver, whereas lack of CAR decreases the concentrations of 6-OH BAs in liver. In bile, CAR activation increases the biliary excretion of 6-OH BAs, whereas absence of CAR increases the biliary excretion of 12-OH BAs and TCDCA.
Collapse
Affiliation(s)
- Andrew J Lickteig
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P. R. China
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Iván L Csanaky
- Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Division of Gastroenterology, Children's Mercy Hospital, Kansas City, Missouri, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
20
|
Xie C, Takahashi S, Brocker CN, He S, Chen L, Xie G, Jang K, Gao X, Krausz KW, Qu A, Levi M, Gonzalez FJ. Hepatocyte peroxisome proliferator-activated receptor α regulates bile acid synthesis and transport. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1396-1411. [PMID: 31195146 DOI: 10.1016/j.bbalip.2019.05.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/05/2019] [Accepted: 05/28/2019] [Indexed: 12/12/2022]
Abstract
Peroxisome proliferator-activated receptor alpha (PPARα) controls lipid homeostasis through regulation of lipid transport and catabolism. PPARα activators are clinically used for hyperlipidemia treatment. The role of PPARα in bile acid (BA) homeostasis is beginning to emerge. Herein, Ppara-null and hepatocyte-specific Ppara-null (Ppara∆Hep) as well as the respective wild-type mice were treated with the potent PPARα agonist Wy-14,643 (Wy) and global metabolomics performed to clarify the role of hepatocyte PPARα in the regulation of BA homeostasis. Levels of all serum BAs were markedly elevated in Wy-treated wild-type mice but not in Ppara-null and Ppara∆Hep mice. Gene expression analysis showed that PPARα activation (1) down-regulated the expression of sodium-taurocholate acid transporting polypeptide and organic ion transporting polypeptide 1 and 4, responsible for the uptake of BAs into the liver; (2) decreased the expression of bile salt export pump transporting BA from hepatocytes into the bile canaliculus; (3) upregulated the expression of multidrug resistance-associated protein 3 and 4 transporting BA from hepatocytes into the portal vein. Moreover, there was a notable increase in the compositions of serum, hepatic and biliary cholic acid and taurocholic acid following Wy treatment, which correlated with the upregulated expression of the Cyp8b1 gene encoding sterol 12α-hydroxylase. The effects of Wy were identical between the Ppara∆Hep and Ppara-null mice. Hepatocyte PPARα controlled BA synthesis and transport not only via direct transcriptional regulation but also via crosstalk with hepatic farnesoid X receptor signaling. These findings underscore a key role for hepatocyte PPARα in the control of BA homeostasis.
Collapse
Affiliation(s)
- Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 2089, United States of America; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China.
| | - Shogo Takahashi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 2089, United States of America; Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC, United States of America.
| | - Chad N Brocker
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 2089, United States of America.
| | - Shijun He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China.
| | - Li Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Guomin Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China.
| | - Katrina Jang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 2089, United States of America.
| | - Xiaoxia Gao
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 2089, United States of America.
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 2089, United States of America.
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China.
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC, United States of America.
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 2089, United States of America.
| |
Collapse
|
21
|
Dempsey JL, Wang D, Siginir G, Fei Q, Raftery D, Gu H, Yue Cui J. Pharmacological Activation of PXR and CAR Downregulates Distinct Bile Acid-Metabolizing Intestinal Bacteria and Alters Bile Acid Homeostasis. Toxicol Sci 2019; 168:40-60. [PMID: 30407581 PMCID: PMC6821357 DOI: 10.1093/toxsci/kfy271] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome regulates important host metabolic pathways including xenobiotic metabolism and intermediary metabolism, such as the conversion of primary bile acids (BAs) into secondary BAs. The nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are well-known regulators for xenobiotic biotransformation in liver. However, little is known regarding the potential effects of PXR and CAR on the composition and function of the gut microbiome. To test our hypothesis that activation of PXR and CAR regulates gut microbiota and secondary BA synthesis, 9-week-old male conventional and germ-free mice were orally gavaged with corn oil, PXR agonist PCN (75 mg/kg), or CAR agonist TCPOBOP (3 mg/kg) once daily for 4 days. PCN and TCPOBOP decreased two taxa in the Bifidobacterium genus, which corresponded with decreased gene abundance of the BA-deconjugating enzyme bile salt hydrolase. In liver and small intestinal content of germ-free mice, there was a TCPOBOP-mediated increase in total, primary, and conjugated BAs corresponding with increased Cyp7a1 mRNA. Bifidobacterium, Dorea, Peptociccaceae, Anaeroplasma, and Ruminococcus positively correlated with T-UDCA in LIC, but negatively correlated with T-CDCA in serum. In conclusion, PXR and CAR activation downregulates BA-metabolizing bacteria in the intestine and modulates BA homeostasis in a gut microbiota-dependent manner.
Collapse
Affiliation(s)
- Joseph L Dempsey
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105
| | - Dongfang Wang
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
- Chongqing Blood Center, Chongqing 400015, P.R. China
| | - Gunseli Siginir
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105
| | - Qiang Fei
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
- Department of Chemistry, Jilin University, Changchun, Jilin Province 130061, P.R. China
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona 85004
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105
| |
Collapse
|
22
|
Zhang Y, Lickteig AJ, Csanaky IL, Klaassen CD. Activation of PPARα decreases bile acids in livers of female mice while maintaining bile flow and biliary bile acid excretion. Toxicol Appl Pharmacol 2017; 338:112-123. [PMID: 29175453 DOI: 10.1016/j.taap.2017.11.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/16/2017] [Accepted: 11/19/2017] [Indexed: 01/20/2023]
Abstract
Fibrates are hypolipidemic drugs that act as activators of peroxisome proliferator-activated receptor α (PPARα). In both humans and rodents, females were reported to be less responsive to fibrates than males. Previous studies on fibrates and PPARα usually involved male mice, but little has been done in females. The present study aimed to provide the first comprehensive analysis of the effects of clofibrate (CLOF) and PPARα on bile acid (BA) homeostasis in female mice. Study in WT male mice showed that a 4-day CLOF treatment increased liver weight, bile flow, and biliary BA excretion, but decreased total BAs in both serum and liver. In contrast, WT female mice were less susceptible to these CLOF-mediated responses observed in males. In WT female mice, CLOF decreased total BAs in the liver, but had little effect on the mRNAs of hepatic BA-related genes. Next, a comparative analysis between WT and PPARα-null female mice showed that lack of PPARα in female mice decreased total BAs in serum, but had little effect on total BAs in liver or bile. However, lack of PPARα in female mice increased mRNAs of BA synthetic enzymes (Cyp7a1, Cyp8b1, Cyp27a1, and Cyp7b1) and transporters (Ntcp, Oatp1a1, Oatp1b2, and Mrp3). Furthermore, the increase of Cyp7a1 in PPARα-null female mice was associated with an increase in liver Fxr-Shp-Lrh-1 signaling. In conclusion, female mice are resistant to CLOF-mediated effects on BA metabolism observed in males, which could be attributed to PPARα-mediated suppression in females on genes involved in BA synthesis and transport.
Collapse
Affiliation(s)
- Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China.
| | - Andrew J Lickteig
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160,USA
| | - Iván L Csanaky
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Division of Gastroenterology, Children's Mercy Hospital & Clinics, Kansas City, MO 64108; USA; Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160,USA.
| |
Collapse
|