1
|
Guo J, Yang Y, Xu N, Li X, Yang Y, Feng W, Ye Y, Xu X, Cui J, Liu M, Huang Y. Pancreatic β-Cell TRAPδ Deficiency Reduces Insulin Production but Improves Insulin Sensitivity. Diabetes 2024; 73:1848-1861. [PMID: 39167635 DOI: 10.2337/db23-0984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 08/15/2024] [Indexed: 08/23/2024]
Abstract
The translocon-associated protein-δ (TRAPδ) plays a role in insulin biosynthesis within pancreatic β-cells. However, its pathophysiological significance in maintaining islet β-cell function and glucose homeostasis remains unclear. In this study, we generated a mouse model featuring pancreatic β-cell-specific deletion of TRAPδ (TRAPδ βKO). Our findings revealed that TRAPδ βKO resulted in decreased circulating insulin levels in mice fed either a normal chow diet or a high-fat diet. Multiple independent experiments established that although TRAPδ deletion reduced insulin content in the islets, it had no discernible effect on insulin gene expression, the insulin to proinsulin ratio, or the expression and glycosylation of the prohormone enzymes involved in proinsulin processing. These data suggest that TRAPδ does not play a pivotal role in the transcription of the insulin gene or proinsulin processing. However, untranslocated preproinsulin levels were significantly increased when islets were treated with a proteasomal inhibitor, suggesting that TRAPδ deficiency may hinder preproinsulin translocation, resulting in a rapid degradation of untranslocated preproinsulin that accounts for the decreased insulin production. Remarkably, despite the moderate decrease in circulating insulin levels in TRAPδ βKO mice, their glucose levels remained unaffected, indicating the presence of compensatory mechanisms that help maintain glucose homeostasis. Insulin tolerance tests further revealed improved insulin sensitivity, accompanied by upregulation of phosphorylated AKT in the peripheral tissues of TRAPδ βKO mice. Collectively, these data highlight the important role of TRAPδ in insulin biosynthesis and β-cell function. The moderate reduction in circulating insulin appears to promote insulin sensitivity in insulin target tissues. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Jiyun Guo
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yanshu Yang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ning Xu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ying Yang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wenli Feng
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yuanyuan Ye
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiaoxi Xu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jingqiu Cui
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yumeng Huang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
2
|
Suzuki Y, Sasamoto Y, Koyama T, Yoshijima C, Oda A, Nakatochi M, Kubo M, Momozawa Y, Uehara R, Ohno K. Relationship of hemoglobin level and plasma coproporphyrin-I concentrations as an endogenous probe for phenotyping OATP1B. Clin Transl Sci 2021; 14:1403-1411. [PMID: 33650309 PMCID: PMC8301560 DOI: 10.1111/cts.12996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/08/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Plasma coproporphyrin‐I (CP‐I) concentration is used as a sensitive and selective endogenous probe for phenotyping organic anion transporting polypeptides 1B (OATP1B) activity in many studies. CP‐I is produced in the process of heme synthesis, but the relationship between plasma CP‐I concentrations and heme synthesis activity is unknown. In this study, we evaluated the relationship between plasma CP‐I concentration and hemoglobin level as a biomarker of heme synthesis activity. The data of 391 subjects selected from the Japanese general population were analyzed. One hundred twenty‐six participants had OATP1B1*15 allele, 11 of whom were homozygous (OATP1B1*15/*15). Multiple regression analysis identified hemoglobin level as an independent variable associated with plasma CP‐I concentration (p < 0.0001). A significant positive correlation was observed between hemoglobin level and plasma CP‐I concentration in participants without OATP1B1*15 allele (n = 265; rs = 0.35, p < 0.0001) and with OATP1B1*15 allele (n = 126; rs =0.27, p = 0.0022). However, Kruskal–Wallis test showed no large difference in Kruskal–Wallis statistics between the distribution of plasma CP‐I concentrations and that of ratio of plasma CP‐I to hemoglobin among six OATP1B1 polymorphism groups. These findings suggest that the hemoglobin level seems to reflect biosynthesis of CP‐I. However, correction by hemoglobin level is not required when using basal plasma CP‐I concentration for phenotyping OATP1B activity.
Collapse
Affiliation(s)
- Yosuke Suzuki
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Japan
| | - Yuri Sasamoto
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chisato Yoshijima
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Japan
| | - Ayako Oda
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michiaki Kubo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Ritei Uehara
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keiko Ohno
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Japan
| |
Collapse
|
3
|
Lütjohann D, Stellaard F, Kerksiek A, Lötsch J, Oertel BG. Serum 4β-hydroxycholesterol increases during fluconazole treatment. Eur J Clin Pharmacol 2020; 77:659-669. [PMID: 33201347 PMCID: PMC8032583 DOI: 10.1007/s00228-020-03041-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/09/2020] [Indexed: 11/02/2022]
Abstract
PURPOSE The antifungal drugs ketoconazole and itraconazole reduce serum concentrations of 4β-hydroxycholesterol, which is a validated marker for hepatic cytochrome P450 (CYP) 3A4 activity. We tested the effect of another antifungal triazole agent, fluconazole, on serum concentrations of different sterols and oxysterols within the cholesterol metabolism to see if this inhibitory reaction is a general side effect of azole antifungal agents. METHODS In a prospective, double-blind, placebo-controlled, two-way crossover design, we studied 17 healthy subjects (nine men, eight women) who received 400 mg fluconazole or placebo daily for 8 days. On day 1 before treatment and on day 8 after the last dose, fasting blood samples were collected. Serum cholesterol precursors and oxysterols were measured by gas chromatography-mass spectrometry-selected ion monitoring and expressed as the ratio to cholesterol (R_sterol). RESULTS Under fluconazole treatment, serum R_lanosterol and R_24,25-dihydrolanosterol increased significantly without affecting serum cholesterol or metabolic downstream markers of hepatic cholesterol synthesis. Serum R_4β-, R_24S-, and R_27-hydroxycholesterol increased significantly. CONCLUSION Fluconazole inhibits the 14α-demethylation of lanosterol and 24,25-dihydrolanosterol, regulated by CYP51A1, without reduction of total cholesterol synthesis. The increased serum level of R_4β-hydroxycholesterol under fluconazole treatment is in contrast to the reductions observed under ketoconazole and itraconazole treatments. The question, whether this increase is caused by induction of CYP3A4 or by inhibition of the catabolism of 4β-hydroxycholesterol, must be answered by mechanistic in vitro and in vivo studies comparing effects of various azole antifungal agents on hepatic CYP3A4 activity.
Collapse
Affiliation(s)
- Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| | - Frans Stellaard
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor Stern Kai 7, 60590, Frankfurt, Germany
| | - Bruno G Oertel
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor Stern Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
4
|
Donkers JM, Roscam Abbing RLP, van Weeghel M, Levels JHM, Boelen A, Schinkel AH, Oude Elferink RPJ, van de Graaf SFJ. Inhibition of Hepatic Bile Acid Uptake by Myrcludex B Promotes Glucagon-Like Peptide-1 Release and Reduces Obesity. Cell Mol Gastroenterol Hepatol 2020; 10:451-466. [PMID: 32330730 PMCID: PMC7363705 DOI: 10.1016/j.jcmgh.2020.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Bile acids are important metabolic signaling molecules. Bile acid receptor activation promotes body weight loss and improves glycemic control. The incretin hormone GLP-1 and thyroid hormone activation of T4 to T3 have been suggested as important contributors. Here, we identify the hepatic bile acid uptake transporter Na+ taurocholate co-transporting polypeptide (NTCP) as target to prolong postprandial bile acid signaling. METHODS Organic anion transporting polypeptide (OATP)1a/1b KO mice with or without reconstitution with human OATP1B1 in the liver were treated with the NTCP inhibitor Myrcludex B for 3.5 weeks after the onset of obesity induced by high fat diet-feeding. Furthermore, radiolabeled T4 was injected to determine the role of NTCP and OATPs in thyroid hormone clearance from plasma. RESULTS Inhibition of NTCP by Myrcludex B in obese Oatp1a/1b KO mice inhibited hepatic clearance of bile acids from portal and systemic blood, stimulated GLP-1 secretion, reduced body weight, and decreased (hepatic) adiposity. NTCP inhibition did not affect hepatic T4 uptake nor lead to increased thyroid hormone activation. Myrcludex B treatment increased fecal energy output, explaining body weight reductions amongst unaltered food intake and energy expenditure. CONCLUSIONS Pharmacologically targeting hepatic bile acid uptake to increase bile acid signaling is a novel approach to treat obesity and induce GLP1- secretion.
Collapse
Affiliation(s)
- Joanne M Donkers
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Reinout L P Roscam Abbing
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Johannes H M Levels
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Anita Boelen
- Endocrinology Laboratory, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Alfred H Schinkel
- Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ronald P J Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
|
6
|
Xu Y, Zhong PP, Tao YY. Metabolic profiling of endogenous bile acids: a novel method to assess hepatoprotective effect of Tanreqing capsule on carbon-tetrachloride-induced liver injury in rats. Chin J Nat Med 2018; 16:271-283. [PMID: 29703327 DOI: 10.1016/s1875-5364(18)30057-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Indexed: 12/11/2022]
Abstract
Tanreqing (TRQ), a traditional Chinese medicine (TCM) formula, can alleviate liver injury and improve liver function. Its pharmacological mechanisms of actions are still unclear due to its complex components and multi-target natures. Metabolomic study is an effective approach to investigating drug pharmacological actions, new diagnostic markers, and potential mechanisms of actions. In the present study, a new strategy was used to evaluate the protective effect of TRQ capsule against carbon tetrachloride (CCl4)-induced hepatotoxicity in rats, by analyzing metabolic profiling of endogenous bile acids (BAs) along with biochemical and histological analyses. BAs concentrations were determined by ultra-performance liquid chromatography coupled with quadrupole mass spectrometry (UPLC-MS). Principal component analysis and partial least squares discriminant analysis were then employed to analyze the UPLC-MS results and compare the hepatoprotective effect of TRQ capsule in different groups at the doses of 0.36, 1.44, and 2.88 g·kg-1 body weight, respectively. Moreover, our results suggested that taurocholic acid (TCA) and taurohyodesoxycholic acid (THDCA) were the most important biochemical markers, which were indicative of CCl4-induced acute hepatic damage and hepatoprotective effect of TRQ capsule. Therefore, this new strategy would be an excellent alternative method for evaluating hepatoprotective effect and proposing potential mechanisms of action for other drugs as well.
Collapse
Affiliation(s)
- Ying Xu
- Shanghai Kaibao Pharmaceutical Co., Ltd., Shanghai 201401, China.
| | - Ping-Ping Zhong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yan-Yan Tao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Clinical Key Laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
7
|
Effect of maternal hypothyroidism during pregnancy on insulin resistance, lipid accumulation, and mitochondrial dysfunction in skeletal muscle of fetal rats. Biosci Rep 2018; 38:BSR20171731. [PMID: 29784871 PMCID: PMC6028760 DOI: 10.1042/bsr20171731] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 05/06/2018] [Accepted: 05/21/2018] [Indexed: 01/09/2023] Open
Abstract
The present study aimed to investigate the effect of maternal hypothyroidism during pregnancy on thyroid function of the fetal rat. Female Sprague–Dawley rats were randomized into two groups. Propylthiouracil (PTU) group received PTU in drinking water for 6 weeks (n=90), normal group received normal drinking water (n=50). The pregnant rats were obtained and had a cesarean-section to get at gestational ages of 8.5, 13, and 21 days, following blood samples and skeletal muscle were obtained from fetal rats. Levels of thyroid hormone, insulin, mitochondrial protein, and adipokines were detected using ELISA. Western blotting was performed to analyze mitochondria and insulin signal transduction-related protein in fetal rat skeletal muscle. Immunostaining of Periodic Acid-Schiff (PAS) and Oil Red O was used to observe the accumulation of muscle glycogen and lipid in the fetal rat. The results showed that the levels of thyroid hormone, insulin, insulin signal transduction-related protein, mitochondrial, and adipokines increased with the fetus developed, but had no statistical differences in the PTU group compared with the normal group. In conclusion, pregnant rats with hypothyroidism had no influence on insulin resistance (IR), lipid accumulation, and mitochondrial dysfunction in skeletal muscle of the fetal rats.
Collapse
|
8
|
Abstract
It has been known for a long time that thyroid hormones have prominent effects on hepatic fatty acid and cholesterol synthesis and metabolism. Indeed, hypothyroidism has been associated with increased serum levels of triglycerides and cholesterol as well as non-alcoholic fatty liver disease (NAFLD). Advances in areas such as cell imaging, autophagy and metabolomics have generated a more detailed and comprehensive picture of thyroid-hormone-mediated regulation of hepatic lipid metabolism at the molecular level. In this Review, we describe and summarize the key features of direct thyroid hormone regulation of lipogenesis, fatty acid β-oxidation, cholesterol synthesis and the reverse cholesterol transport pathway in normal and altered thyroid hormone states. Thyroid hormone mediates these effects at the transcriptional and post-translational levels and via autophagy. Given these potentially beneficial effects on lipid metabolism, it is possible that thyroid hormone analogues and/or mimetics might be useful for the treatment of metabolic diseases involving the liver, such as hypercholesterolaemia and NAFLD.
Collapse
Affiliation(s)
- Rohit A. Sinha
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Brijesh K. Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Paul M. Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
9
|
Templeman NM, Flibotte S, Chik JHL, Sinha S, Lim GE, Foster LJ, Nislow C, Johnson JD. Reduced Circulating Insulin Enhances Insulin Sensitivity in Old Mice and Extends Lifespan. Cell Rep 2018; 20:451-463. [PMID: 28700945 DOI: 10.1016/j.celrep.2017.06.048] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/01/2017] [Accepted: 06/19/2017] [Indexed: 12/20/2022] Open
Abstract
The causal relationships between insulin levels, insulin resistance, and longevity are not fully elucidated. Genetic downregulation of insulin/insulin-like growth factor 1 (Igf1) signaling components can extend invertebrate and mammalian lifespan, but insulin resistance, a natural form of decreased insulin signaling, is associated with greater risk of age-related disease in mammals. We compared Ins2+/- mice to Ins2+/+ littermate controls, on a genetically stable Ins1 null background. Proteomic and transcriptomic analyses of livers from 25-week-old mice suggested potential for healthier aging and altered insulin sensitivity in Ins2+/- mice. Halving Ins2 lowered circulating insulin by 25%-34% in aged female mice, without altering Igf1 or circulating Igf1. Remarkably, decreased insulin led to lower fasting glucose and improved insulin sensitivity in aged mice. Moreover, lowered insulin caused significant lifespan extension, observed across two diverse diets. Our study indicates that elevated insulin contributes to age-dependent insulin resistance and that limiting basal insulin levels can extend lifespan.
Collapse
Affiliation(s)
- Nicole M Templeman
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Stephane Flibotte
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jenny H L Chik
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Sunita Sinha
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Gareth E Lim
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Corey Nislow
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
10
|
Kang L, Si L, Rao J, Li D, Wu Y, Wu S, Wu M, He S, Zhu W, Wu Y, Xu J, Li G, Huang J. Polygoni Multiflori Radix derived anthraquinones alter bile acid disposition in sandwich-cultured rat hepatocytes. Toxicol In Vitro 2017; 40:313-323. [PMID: 28161596 DOI: 10.1016/j.tiv.2017.01.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 01/30/2023]
Abstract
Hepatic adverse reaction associated with Polygoni Multiflori Radix (PMR) has been frequently reported in recent years. Highly-enriched anthraquinones (AQs) in PMR, such as emodin, chrysophanol and physcion, have been found to be hepatotoxic. In the present study, sandwich-cultured rat hepatocytes (SCRHs) were employed to investigate the effect of individual and combined AQs on the disposition of endogenous bile acids (BAs) and exogenous probe substrates including deuterium-labeled taurocholate (d5-TCA), glycochenodeoxycholic acid (d4-GCDCA) and 5 (and 6)-carboxy-2',7'-dichlorofluorescein (CDF). Emodin and chrysophanol significantly inhibited bile salt export pump and multidrug resistance-associated protein 2 (Mrp2), respectively, as evidenced by decreased biliary excretion index (BEI) of d5-TCA and CDF. Moreover, basolateral efflux transporters were inhibited by all individual and combined AQs. As a result, cellular accumulation of total and specific endogenous BAs were significantly elevated by individual AQs, alone or combined. In addition, down-regulation of Mrps in both gene and protein levels by AQs served as another critical contributing factor for BA accumulation in SCRHs. To be noted, subsequent adaptive gene regulation, including reduced Ntcp expression, upregulated Bsep levels, and downregulated Cyp8b1, alleviated, to a certain extent, but not prevented from toxic BA accumulation. In summary, all three AQs of interest are likely to alter BA disposition through direct inhibition of BA transporters as well as regulated expression of BA transporters and enzymes.
Collapse
Affiliation(s)
- Li Kang
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Luqin Si
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Hubei Engineering Research Center for Novel Drug Delivery Systems, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jing Rao
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Dan Li
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ya Wu
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Sanlan Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Minghui Wu
- Malcom Randall VA Medical Center, Gainesville, FL 32608, USA
| | - Sijie He
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wenwen Zhu
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yang Wu
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jiaqiang Xu
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Gao Li
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Hubei Engineering Research Center for Novel Drug Delivery Systems, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jiangeng Huang
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Hubei Engineering Research Center for Novel Drug Delivery Systems, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
11
|
Abstract
Thyroid hormones (TH) are endocrine messengers essential for normal development and function of virtually every vertebrate. The hypothalamic-pituitary-thyroid axis is exquisitely modulated to maintain nearly constant TH (T4 and T3) levels in circulation. However peripheral tissues and the CNS control the intracellular availability of TH, suggesting that circulating concentrations of TH are not fully representative of what each cell type sees. Indeed, recent work in the field has identified that TH transporters, deiodinases and thyroid hormone receptor coregulators can strongly control tissue-specific sensitivity to a set amount of TH. Furthermore, the mechanism by which the thyroid hormone receptors regulate target gene expression can vary by gene, tissue and cellular context. This review will highlight novel insights into the machinery that controls the cellular response to TH, which include unique signaling cascades. These findings shed new light into the pathophysiology of human diseases caused by abnormal TH signaling.
Collapse
Affiliation(s)
- Arturo Mendoza
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Xiang T, Yang Z, Sun B, Luo H, Zhang S, Ren B, Chen X, Zhou X, Chen Z. Traditional Chinese medicine: Pivotal role of the spleen in the metabolism of aristolochic acid I in rats is dependent on oatp2a1. Mol Med Rep 2016; 14:3243-50. [DOI: 10.3892/mmr.2016.5612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 06/27/2016] [Indexed: 11/06/2022] Open
|
13
|
Lin JZ, Martagón AJ, Cimini SL, Gonzalez DD, Tinkey DW, Biter A, Baxter JD, Webb P, Gustafsson JÅ, Hartig SM, Phillips KJ. Pharmacological Activation of Thyroid Hormone Receptors Elicits a Functional Conversion of White to Brown Fat. Cell Rep 2015; 13:1528-37. [PMID: 26586443 PMCID: PMC4662916 DOI: 10.1016/j.celrep.2015.10.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/31/2015] [Accepted: 10/07/2015] [Indexed: 01/06/2023] Open
Abstract
The functional conversion of white adipose tissue (WAT) into a tissue with brown adipose tissue (BAT)-like activity, often referred to as "browning," represents an intriguing strategy for combating obesity and metabolic disease. We demonstrate that thyroid hormone receptor (TR) activation by a synthetic agonist markedly induces a program of adaptive thermogenesis in subcutaneous WAT that coincides with a restoration of cold tolerance to cold-intolerant mice. Distinct from most other browning agents, pharmacological TR activation dissociates the browning of WAT from activation of classical BAT. TR agonism also induces the browning of white adipocytes in vitro, indicating that TR-mediated browning is cell autonomous. These data establish TR agonists as a class of browning agents, implicate the TRs in the browning of WAT, and suggest a profound pharmacological potential of this action.
Collapse
Affiliation(s)
- Jean Z Lin
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, TX 77030, USA; Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77004, USA
| | - Alexandro J Martagón
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, TX 77030, USA; Escuela de Biotecnología y Alimentos, Instituto Tecnológico y de Estudios Superiores de Monterrey, 64849 Monterrey, NL, Mexico
| | - Stephanie L Cimini
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Daniel D Gonzalez
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, TX 77030, USA; Escuela de Biotecnología y Alimentos, Instituto Tecnológico y de Estudios Superiores de Monterrey, 64849 Monterrey, NL, Mexico
| | - David W Tinkey
- Comparative Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Amadeo Biter
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - John D Baxter
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Paul Webb
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jan-Åke Gustafsson
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, TX 77030, USA; Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77004, USA
| | - Sean M Hartig
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin J Phillips
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
14
|
Chu X, Shih SJ, Shaw R, Hentze H, Chan GH, Owens K, Wang S, Cai X, Newton D, Castro-Perez J, Salituro G, Palamanda J, Fernandis A, Ng CK, Liaw A, Savage MJ, Evers R. Evaluation of cynomolgus monkeys for the identification of endogenous biomarkers for hepatic transporter inhibition and as a translatable model to predict pharmacokinetic interactions with statins in humans. Drug Metab Dispos 2015; 43:851-63. [PMID: 25813937 DOI: 10.1124/dmd.115.063347] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/26/2015] [Indexed: 12/31/2022] Open
Abstract
Inhibition of hepatic transporters such as organic anion transporting polypeptides (OATPs) 1B can cause drug-drug interactions (DDIs). Determining the impact of perpetrator drugs on the plasma exposure of endogenous substrates for OATP1B could be valuable to assess the risk for DDIs early in drug development. As OATP1B orthologs are well conserved between human and monkey, we assessed in cynomolgus monkeys the endogenous OATP1B substrates that are potentially suitable to assess DDI risk in humans. The effect of rifampin (RIF), a potent inhibitor for OATP1B, on plasma exposure of endogenous substrates of hepatic transporters was measured. From the 18 biomarkers tested, RIF (18 mg/kg, oral) caused significant elevation of plasma unconjugated and conjugated bilirubin, which may be attributed to inhibition of cOATP1B1 and cOATP1B3 based on in vitro to in vivo extrapolation analysis. To further evaluate whether cynomolgus monkeys are a suitable translational model to study OATP1B-mediated DDIs, we determined the inhibitory effect of RIF on in vitro transport and pharmacokinetics of rosuvastatin (RSV) and atorvastatin (ATV). RIF strongly inhibited the uptake of RSV and ATV by cOATP1B1 and cOATP1B3 in vitro. In agreement with clinical observations, RIF (18 mg/kg, oral) significantly decreased plasma clearance and increased the area under the plasma concentration curve (AUC) of intravenously administered RSV by 2.8- and 2.7-fold, and increased the AUC and maximum plasma concentration of orally administered RSV by 6- and 10.3-fold, respectively. In contrast to clinical findings, RIF did not significantly increase plasma exposure of either intravenous or orally administered ATV, indicating species differences in the rate-limiting elimination pathways.
Collapse
Affiliation(s)
- Xiaoyan Chu
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Shian-Jiun Shih
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Rachel Shaw
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Hannes Hentze
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Grace H Chan
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Karen Owens
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Shubing Wang
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Xiaoxin Cai
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Deborah Newton
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Jose Castro-Perez
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Gino Salituro
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Jairam Palamanda
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Aaron Fernandis
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Choon Keow Ng
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Andy Liaw
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Mary J Savage
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Raymond Evers
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| |
Collapse
|
15
|
Martagón AJ, Lin JZ, Cimini SL, Webb P, Phillips KJ. The amelioration of hepatic steatosis by thyroid hormone receptor agonists is insufficient to restore insulin sensitivity in ob/ob mice. PLoS One 2015; 10:e0122987. [PMID: 25849936 PMCID: PMC4388544 DOI: 10.1371/journal.pone.0122987] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/26/2015] [Indexed: 12/13/2022] Open
Abstract
Thyroid hormone receptor (TR) agonists have been proposed as therapeutic agents to treat non-alcoholic fatty liver disease (NAFLD) and insulin resistance. We investigated the ability of the TR agonists GC-1 and KB2115 to reduce hepatic steatosis in ob/ob mice. Both compounds markedly reduced hepatic triglyceride levels and ameliorated hepatic steatosis. However, the amelioration of fatty liver was not sufficient to improve insulin sensitivity in these mice and reductions in hepatic triglycerides did not correlate with improvements in insulin sensitivity or glycemic control. Instead, the effects of TR activation on glycemia varied widely and were found to depend upon the time of treatment as well as the compound and dosage used. Lower doses of GC-1 were found to further impair glycemic control, while a higher dose of the same compound resulted in substantially improved glucose tolerance and insulin sensitivity, despite all doses being equally effective at reducing hepatic triglyceride levels. Improvements in glycemic control and insulin sensitivity were observed only in treatments that also increased body temperature, suggesting that the induction of thermogenesis may play a role in mediating these beneficial effects. These data illustrate that the relationship between TR activation and insulin sensitivity is complex and suggests that although TR agonists may have value in treating NAFLD, their effect on insulin sensitivity must also be considered.
Collapse
Affiliation(s)
- Alexandro J. Martagón
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, Texas, United States of America
- Escuela de Biotecnología y Alimentos, Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, NL, Mexico
| | - Jean Z. Lin
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, Texas, United States of America
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, United States of America
| | - Stephanie L. Cimini
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Paul Webb
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Kevin J. Phillips
- Diabetes and Metabolic Disease Program, Houston Methodist Research Institute, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
16
|
Meyer Zu Schwabedissen HE, Boettcher K, Steiner T, Schwarz UI, Keiser M, Kroemer HK, Siegmund W. OATP1B3 is expressed in pancreatic β-islet cells and enhances the insulinotropic effect of the sulfonylurea derivative glibenclamide. Diabetes 2014; 63:775-84. [PMID: 24150606 DOI: 10.2337/db13-1005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Organic anion transporting polypeptide OATP1B3 is a membrane-bound drug transporter that facilitates cellular entry of a variety of substrates. Most of the previous studies focused on its hepatic expression and function in hepatic drug elimination. In this study, we report expression of OATP1B3 in human pancreatic tissue, with the abundance of the transporter localized in the islets of Langerhans. Transport studies using OATP1B3-overexpressing MDCKII cells revealed significant inhibition of the cellular uptake of the known substrate cholecystokinin-8 in the presence of the insulinotropic antidiabetes compounds tolbutamide, glibenclamide, glimepiride, and nateglinide and identified glibenclamide as a novel substrate of OATP1B3. Sulfonylurea derivatives exert their insulinotropic effect by binding to the SUR1 subunit of the KATP channels inducing insulin secretion in β-cells. Here, we show that transient overexpression of human OATP1B3 in a murine β-cell line (MIN6)-which exhibits glucose and glibenclamide-sensitive insulin secretion-significantly enhances the insulinotropic effect of glibenclamide without affecting glucose-stimulated insulin secretion. Taken together, our data provide evidence that the drug transporter OATP1B3 functions as a determinant of the insulinotropic effect of glibenclamide on the tissue level. Changes in transport activity based on drug-drug interactions or genetic variability may therefore influence glibenclamide efficacy.
Collapse
|
17
|
Goldberg AS, Tirona RG, Asher LJ, Kim RB, Van Uum SHM. Ciprofloxacin and rifampin have opposite effects on levothyroxine absorption. Thyroid 2013; 23:1374-8. [PMID: 23647409 DOI: 10.1089/thy.2013.0014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Levothyroxine (L-T4) absorption varies between individuals, and can be affected by various concomitantly administered drugs. Case reports have indicated an association between cotreatment with ciprofloxacin or rifampin and hypothyroidism in patients on a stable L-T4 dose. METHODS The effects of two antibiotics on T4 absorption were prospectively assessed in a double-blind, randomized, crossover fashion. Eight healthy volunteers received 1000 μg L-T4 combined with placebo, ciprofloxacin 750 mg, or rifampin 600 mg as single doses. We measured total plasma thyroxine (T4) concentrations over a 6-hour period after dosing using liquid chromatography-tandem mass spectrometry. For each study arm, areas under the T4 plasma concentration-time curve (T4 AUCs) were compared. RESULTS Coadministration of ciprofloxacin significantly decreased the T4 AUC by 39% (p = 0.035), while, surprisingly, rifampin significantly increased T4 AUC by 25% (p = 0.003). CONCLUSION Intestinal absorption of L-T4 is differentially affected by acute coadministration of ciprofloxacin or rifampin. Mechanistic studies focused on intestinal and possibly hepatic thyroid hormone transporters are required to explain the observed drug interactions with L-T4.
Collapse
Affiliation(s)
- Alyse S Goldberg
- 1 Division of Endocrinology and Metabolism, Department of Medicine, Schulich School of Medicine and Dentistry, Western University , London, Canada
| | | | | | | | | |
Collapse
|
18
|
König J, Müller F, Fromm MF. Transporters and drug-drug interactions: important determinants of drug disposition and effects. Pharmacol Rev 2013; 65:944-66. [PMID: 23686349 DOI: 10.1124/pr.113.007518] [Citation(s) in RCA: 403] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Uptake and efflux transporters determine plasma and tissue concentrations of a broad variety of drugs. They are localized in organs such as small intestine, liver, and kidney, which are critical for drug absorption and elimination. Moreover, they can be found in important blood-tissue barriers such as the blood-brain barrier. Inhibition or induction of drug transporters by coadministered drugs can alter pharmacokinetics and pharmacodynamics of the victim drugs. This review will summarize in particular clinically observed drug-drug interactions attributable to inhibition or induction of intestinal export transporters [P-glycoprotein (P-gp), breast cancer resistance protein (BCRP)], to inhibition of hepatic uptake transporters [organic anion transporting polypeptides (OATPs)], or to inhibition of transporter-mediated [organic anion transporters (OATs), organic cation transporter 2 (OCT2), multidrug and toxin extrusion proteins (MATEs), P-gp] renal secretion of xenobiotics. Available data on the impact of nutrition on transport processes as well as genotype-dependent, transporter-mediated drug-drug interactions will be discussed. We will also present and discuss data on the variable extent to which information on the impact of transporters on drug disposition is included in summaries of product characteristics of selected countries (SPCs). Further work is required regarding a better understanding of the role of the drug metabolism-drug transport interplay for drug-drug interactions and on the extrapolation of in vitro findings to the in vivo (human) situation.
Collapse
Affiliation(s)
- Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Clinical Pharmacology and Clinical Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | | | | |
Collapse
|
19
|
Ramachandran A, Betts G, Bhana S, Helme G, Blick C, Moller-Levet C, Saunders E, Valentine H, Pepper S, Miller CJ, Buffa F, Harris AL, West CML. An in vivo hypoxia metagene identifies the novel hypoxia inducible factor target gene SLCO1B3. Eur J Cancer 2013; 49:1741-51. [PMID: 23352438 DOI: 10.1016/j.ejca.2012.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/23/2012] [Accepted: 12/04/2012] [Indexed: 01/24/2023]
Abstract
A hypoxia-associated gene signature (metagene) was previously derived via in vivo data-mining. In this study, we aimed to investigate whether this approach could identify novel hypoxia regulated genes. From an initial list of nine genes, three were selected for further study (BCAR1, IGF2BP2 and SLCO1B3). Ten cell lines were exposed to hypoxia and interrogated for the expression of the three genes. All three genes were hypoxia inducible in at least one of the 10 cell lines with SLCO1B3 induced in seven. SLCO1B3 was studied further using chromatin immunoprecipitation and luciferase assays to investigate hypoxia inducible factor (HIF) dependent transcription. Two functional HIF response elements were identified within intron 1 of the gene. The functional importance of SLCO1B3 was studied by gene knockdown experiments followed by cell growth assays, flow cytometry and Western blotting. SLCO1B3 knockdown reduced cell size and 3-dimensional spheroid volume, which was associated with decreased activation of the mammalian target of rapamycin (mTOR) pathway. Finally, Oncomine analysis revealed that head and neck and colorectal tumours had higher levels of SLCO1B3 compared to normal tissue. Thus, the knowledge based approach for deriving gene signatures can identify novel biologically relevant genes.
Collapse
Affiliation(s)
- Anassuya Ramachandran
- Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|