1
|
Li VW, Dong TS, Funes D, Hernandez L, Kushnir NR, Nair D, Jacobs JP, Reddy ST, Mayer EA, Chang L, Meriwether D. Mass spectrometric profiling of primary estrogens and estrogen metabolites in human stool and plasma partially elucidates the role of the gut microbiome in estrogen recycling. Mol Cell Endocrinol 2025; 603:112534. [PMID: 40180172 DOI: 10.1016/j.mce.2025.112534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Primary estrogens and estrogen metabolites are commonly measured in human plasma and serum, but there exist almost no recent reports for human stool. This knowledge gap limits our understanding of the relationships between systemic and gut estrogens. We developed a highly sensitive liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) method to determine, in human plasma and stool, the free and conjugated levels of estrone, estradiol, and estriol together with their additional hydroxyestrogen and methoxyestrogen metabolites. We investigated human stool and plasma estrogens in healthy control men; in follicular and luteal phase premenopausal women; and in postmenopausal women. Most estrogens were present in plasma and stool of all groups, while the plasma and stool levels of hydroxyestrogen and methoxyestrogen metabolites but not estrone were correlated. In stool, estrogens were higher in premenopausal women, with estrogens increasing across the menstrual cycle. We combined these LC-MS/MS measures with shotgun metagenomic sequencing of the stool microbiomes. Estrogen deconjugation enzyme gene copy numbers (β-glucuronidase and arylsulfatase) were higher in premenopausal women; while the gene copy number of β-glucuronidase + arylsulfatase, but not β-glucuronidase alone, correlated with deconjugated stool estrogens in all groups. Moreover, β-glucuronidase + arylsulfatase gene copy numbers correlated with combined plasma estrogens in men and with individual plasma estrogen metabolites in men and premenopausal women. These results support the hypothesis that gut microbial β-glucuronidase and arylsulfatase control the deconjugation of gut estrogens while modulating systemic levels through the uptake and recirculation of these deconjugated estrogens. The intestine may thus constitute an important additional compartment in estrogen physiology.
Collapse
Affiliation(s)
- Vince W Li
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-5347, United States
| | - Tien S Dong
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-5347, United States
| | - Diana Funes
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-5347, United States
| | - Laura Hernandez
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-5347, United States
| | - Nicole R Kushnir
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1752, United States
| | - Devika Nair
- College of Letters and Science, University of California Los Angeles, United States
| | - Jonathan P Jacobs
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-5347, United States
| | - Srinivasa T Reddy
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1679, United States
| | - Emeran A Mayer
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-5347, United States
| | - Lin Chang
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-5347, United States
| | - David Meriwether
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA, 90095-5347, United States.
| |
Collapse
|
2
|
Uoti A, Järvinen E, Sjöstedt N, Koenderink J, Finel M, Kidron H. Efflux and uptake of androgen sulfates using transporter-overexpressing HEK293 cells and membrane vesicles. J Pharm Sci 2025:103705. [PMID: 39993711 DOI: 10.1016/j.xphs.2025.103705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Hydrophilic steroid conjugates require active and facilitated transport mechanisms for their distribution into tissues and excretion from the body. The ATP-binding cassette (ABC) and solute carrier organic anion (SLCO) transporters involved in androgen sulfate (-S) disposition have been poorly characterized. In this study, we investigated the in vitro transport of testosterone-S, epitestosterone-S, dehydroepiandrosterone-S (DHEA-S), androsterone-S, and etiocholanolone-S by the multidrug resistance-associated proteins 2-4 (MRP2-4, ABCC2-4), breast cancer resistance protein (BCRP, ABCG2), and organic anion-transporting polypeptides (OATP) 1B1, 1B3, and 2B1 (SLCO1B1, SLCO1B3, and SLCO2B1) using human transporter-overexpressing HEK293 cells and membrane vesicles. We found testosterone-S, epitestosterone-S, and DHEA-S to be selectively transported by BCRP and/or MRP4, whereas all studied androgen sulfates were substrates of MRP3, OATP1B1, OATP1B3, and OATP2B1. MRP2 did not transport any of the studied compounds. Evaluation of transport kinetics revealed MRP4 to interact with its substrates at high to moderate affinity, whereas the observed affinities towards MRP3, BCRP, and OATPs were mostly moderate. These results help to build a better mechanistic understanding of the disposition of androgen sulfates in the human body. Additionally, this data may be used to assess the feasibility of androgen sulfates as additional biomarkers in doping detection.
Collapse
Affiliation(s)
- Arttu Uoti
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Erkka Järvinen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jan Koenderink
- Department of Pharmacy - Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Moshe Finel
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
3
|
Li S, Liu Y. Intestinal absorption mechanism and nutritional synergy promotion strategy of dietary flavonoids: transintestinal epithelial pathway mediated by intestinal transport proteins. Crit Rev Food Sci Nutr 2024:1-14. [PMID: 39086266 DOI: 10.1080/10408398.2024.2387320] [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: 08/02/2024]
Abstract
Dietary flavonoids exhibit a variety of physiological functions in regulating glucose and lipid metabolism, improving cardiovascular function, and enhancing stress resistance. However, poor intestinal absorption limits their health benefits. Previous studies on improving the absorption efficiency of flavonoids have focused on targeted release, enhanced gastrointestinal stability and prolonged retention time in digestive tract. But less attention has been paid to promoting the uptake and transport of flavonoids by intestinal epithelial cells through modulation of transporter protein-mediated pathways. Interestingly, some dietary nutrients have been found to modulate the expression or function of transporter proteins, thereby synergistically or antagonistically affecting flavonoid absorption. Therefore, this paper proposed an innovative regulatory strategy known as the "intestinal transport protein-mediated pathway" to promote intestinal absorption of dietary flavonoids. The flavonoid absorption mechanism in the intestinal epithelium, mediated by intestinal transport proteins, was summarized. The functional differences between the uptake transporter and efflux transporters during flavonoid trans-intestinal cellular transport were discussed. Finally, from the perspective of nutritional synergy promotion of absorption, the feasibility of promoting flavonoid intestinal absorption by regulating the expression/function of transport proteins through dietary nutrients was emphasized. This review provides a new perspective and developing precise dietary nutrient combinations for efficient dietary flavonoid absorption.
Collapse
Affiliation(s)
- Shuqiong Li
- College of Ocean Food and Biological Engineering, National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian, People's Republic of China
| | - Yixiang Liu
- College of Ocean Food and Biological Engineering, National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian, People's Republic of China
| |
Collapse
|
4
|
Courchesne M, Manrique G, Bernier L, Moussa L, Cresson J, Gutzeit A, Froehlich JM, Koh DM, Chartrand-Lefebvre C, Matoori S. Gender Differences in Pharmacokinetics: A Perspective on Contrast Agents. ACS Pharmacol Transl Sci 2024; 7:8-17. [PMID: 38230293 PMCID: PMC10789139 DOI: 10.1021/acsptsci.3c00116] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 01/18/2024]
Abstract
Gender is an important risk factor for adverse drug reactions. Women report significantly more adverse drug reactions than men. There is a growing consensus that gender differences in drug PK is a main contributor to higher drug toxicity in women. These differences stem from physiological differences (body composition, plasma protein concentrations, and liver and kidney function), drug interactions, and comorbidities. Contrast agents are widely used to enhance diagnostic performance in computed tomography and magnetic resonance imaging. Despite their broad use, these contrast agents can lead to important adverse reactions including hypersensitivity reactions, nephropathy, and hyperthyroidism. Importantly, female gender is one of the main risk factors for contrast agent toxicity. As these adverse reactions may be related to gender differences in PK, this perspective aims to describe distribution and elimination pathways of commonly used contrast agents and to critically discuss gender differences in these processes.
Collapse
Affiliation(s)
- Myriam Courchesne
- Faculté
de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montreal, Quebec H3T 1J4, Canada
| | - Gabriela Manrique
- Faculté
de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montreal, Quebec H3T 1J4, Canada
| | - Laurie Bernier
- Faculté
de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montreal, Quebec H3T 1J4, Canada
| | - Leen Moussa
- Faculté
de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montreal, Quebec H3T 1J4, Canada
| | - Jeanne Cresson
- Clinical
Research Group, Klus Apotheke Zurich, 8032 Zurich, Switzerland
| | - Andreas Gutzeit
- Department
of Health Sciences and Medicine, University
of Lucerne, Frohburgstaße 3, 6002 Luzern, Switzerland
- Institute
of Radiology and Nuclear Medicine and Breast Center St. Anna, Hirslanden Klinik St. Anna, 6006 Lucerne, Switzerland
- Department
of Radiology, Paracelsus Medical University, 5020 Salzburg, Austria
| | | | - Dow-Mu Koh
- Cancer Research
UK Clinical Magnetic Resonance Research Group, Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom
| | - Carl Chartrand-Lefebvre
- Radiology
Department, Centre Hospitalier de l’Université
de Montréal (CHUM), Montreal, Quebec H2X 3E4, Canada
- Centre
de Recherche du Centre Hospitalier de l’Université de
Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Simon Matoori
- Faculté
de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|
5
|
Gao Y, Deng H, Zhao Y, Li M, Wang L, Zhang Y. Gene Expression of Abcc2 and Its Regulation by Chicken Xenobiotic Receptor. TOXICS 2024; 12:55. [PMID: 38251011 PMCID: PMC10818656 DOI: 10.3390/toxics12010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024]
Abstract
Membrane transporter multidrug resistance-associated protein 2 (MRP2/Abcc2) exhibits high pharmaco-toxicological relevance because it exports multiple cytotoxic compounds from cells. However, no detailed information about the gene expression and regulation of MRP2 in chickens is yet available. Here, we sought to investigate the expression distribution of Abcc2 in different tissues of chicken and then determine whether Abcc2 expression is induced by chicken xenobiotic receptor (CXR). The bioinformatics analyses showed that MRP2 transporters have three transmembrane structural domains (MSDs) and two highly conserved nucleotide structural domains (NBDs), and a close evolutionary relationship with turkeys. Tissue distribution analysis indicated that Abcc2 was highly expressed in the liver, kidney, duodenum, and jejunum. When exposed to metyrapone (an agonist of CXR) and ketoconazole (an antagonist of CXR), Abcc2 expression was upregulated and downregulated correspondingly. We further confirmed that Abcc2 gene regulation is dependent on CXR, by overexpressing and interfering with CXR, respectively. We also demonstrated the induction of Abcc2 expression and the activity of ivermectin, with CXR being a likely mediator. Animal experiments demonstrated that metyrapone and ivermectin induced Abcc2 in the liver, kidney, and duodenum of chickens. Together, our study identified the gene expression of Abcc2 and its regulation by CXR in chickens, which may provide novel targets for the reasonable usage of veterinary drugs.
Collapse
Affiliation(s)
- Yanhong Gao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Huacheng Deng
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Yuying Zhao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Mei Li
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Liping Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China;
| | - Yujuan Zhang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| |
Collapse
|
6
|
Deng F, Sjöstedt N, Santo M, Neuvonen M, Niemi M, Kidron H. Novel inhibitors of breast cancer resistance protein (BCRP, ABCG2) among marketed drugs. Eur J Pharm Sci 2023; 181:106362. [PMID: 36529162 DOI: 10.1016/j.ejps.2022.106362] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/11/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Drug-drug interactions (DDIs) are a major concern for the safe use of medications. Breast cancer resistance protein (BCRP) is a clinically relevant ATP-binding cassette (ABC) transporter for drug disposition. Inhibition of BCRP increases the plasma concentrations of BCRP substrate drugs, which potentially could lead to adverse drug reactions. The aim of the present study was to identify BCRP inhibitors amongst a library of 232 commonly used drugs and anticancer drugs approved by the United States Food and Drug Administration (FDA). BCRP inhibition studies were carried out using the vesicular transport assay. We found 75 drugs that reduced the relative transport activity of BCRP to less than 25% of the vehicle control and were categorized as strong inhibitors. The concentration required for 50% inhibition (IC50) was determined for 13 strong inhibitors that were previously poorly characterized for BCRP inhibition. The IC50 ranged from 1.1 to 11 µM, with vemurafenib, dabigatran etexilate and everolimus being the strongest inhibitors. According to the drug interaction guidance documents from the FDA and the European Medicines Agency (EMA), in vivo DDI studies are warranted if the theoretical intestinal luminal concentration of a drug exceeds its IC50 by tenfold. Here, the IC50 values for eight of the drugs were 100-fold lower than their theoretical intestinal luminal concentration. Moreover, a mechanistic static model suggested that vemurafenib, bexarotene, dabigatran etexilate, rifapentine, aprepitant, and ivacaftor could almost fully inhibit intestinal BCRP, increasing the exposure of concomitantly administered rosuvastatin over 90%. Therefore, clinical studies are warranted to investigate whether these drugs cause BCRP-mediated DDIs in humans.
Collapse
Affiliation(s)
- Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Tukholmankatu 8 C, P.O. Box 20, 00014, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Haartmaninkatu 8, P.O. Box 63, 00014, Finland
| | - Noora Sjöstedt
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland. Viikinkaari 5 E, P.O. Box 56, 00014, Finland
| | - Mariangela Santo
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland. Viikinkaari 5 E, P.O. Box 56, 00014, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Tukholmankatu 8 C, P.O. Box 20, 00014, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Haartmaninkatu 8, P.O. Box 63, 00014, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Tukholmankatu 8 C, P.O. Box 20, 00014, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Haartmaninkatu 8, P.O. Box 63, 00014, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Heidi Kidron
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland. Viikinkaari 5 E, P.O. Box 56, 00014, Finland.
| |
Collapse
|
7
|
Sex-specific effects of excipients on oral drug bioavailability. Int J Pharm 2022; 629:122365. [DOI: 10.1016/j.ijpharm.2022.122365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
|
8
|
Taskar KS, Yang X, Neuhoff S, Patel M, Yoshida K, Paine MF, Brouwer KL, Chu X, Sugiyama Y, Cook J, Polli JW, Hanna I, Lai Y, Zamek-Gliszczynski M. Clinical Relevance of Hepatic and Renal P-gp/BCRP Inhibition of Drugs: An International Transporter Consortium Perspective. Clin Pharmacol Ther 2022; 112:573-592. [PMID: 35612761 PMCID: PMC9436425 DOI: 10.1002/cpt.2670] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/16/2022] [Indexed: 12/11/2022]
Abstract
The role of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in drug-drug interactions (DDIs) and limiting drug absorption as well as restricting the brain penetration of drugs with certain physicochemical properties is well known. P-gp/BCRP inhibition by drugs in the gut has been reported to increase the systemic exposure to substrate drugs. A previous International Transporter Consortium (ITC) perspective discussed the feasibility of P-gp/BCRP inhibition at the blood-brain barrier and its implications. This ITC perspective elaborates and discusses specifically the hepatic and renal P-gp/BCRP (referred as systemic) inhibition of drugs and whether there is any consequence for substrate drug disposition. This perspective summarizes the clinical evidence-based recommendations regarding systemic P-gp and BCRP inhibition of drugs with a focus on biliary and active renal excretion pathways. Approaches to assess the clinical relevance of systemic P-gp and BCRP inhibition in the liver and kidneys included (i) curation of DDIs involving intravenously administered substrates or inhibitors; (ii) in vitro-to-in vivo extrapolation of P-gp-mediated DDIs at the systemic level; and (iii) curation of drugs with information available about the contribution of biliary excretion and related DDIs. Based on the totality of evidence reported to date, this perspective supports limited clinical DDI risk upon P-gp or BCRP inhibition in the liver or kidneys.
Collapse
Affiliation(s)
- Kunal S. Taskar
- Drug Metabolism and Pharmacokinetics, IVIVT, GlaxoSmithKline, Stevenage, UK
| | - Xinning Yang
- Office of Clinical Pharmacology, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD
| | - Sibylle Neuhoff
- Certara UK Ltd, Simcyp Division, 1 Concourse Way, Level 2-Acero, Sheffield, S1 2BJ, UK
| | - Mitesh Patel
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Kenta Yoshida
- Clinical Pharmacology, Genentech Early Research and Development, South San Francisco, CA 94080, USA
| | - Mary F. Paine
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA
| | - Kim L.R. Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Xiaoyan Chu
- Department of ADME and Discovery Toxicology, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033 USA
| | - Yuichi Sugiyama
- Laboratory of Quantitative System PK/Pharmacodynamics, School of Pharmacy, Kioicho campus, Josai International University, Tokyo 102-0093, Japan
| | - Jack Cook
- Clinical Pharmacology, Global Product Development, Pfizer Inc., Groton, Connecticut, USA
| | - Joseph W. Polli
- Global Medical Sciences, ViiV Healthcare, Research Triangle Park NC USA
| | - Imad Hanna
- Pharmacokinetic Sciences-Oncology, Novartis Institute for Biomedical Research, East Hanover, NJ
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc. Foster City, CA USA
| | | |
Collapse
|
9
|
He L, Xu C, Wang Z, Duan S, Xu J, Li C, Yao X, Gonzalez FJ, Qin Z, Yao Z. Identification of naturally occurring inhibitors in Xian-Ling-Gu-Bao capsule against the glucuronidation of estrogens. Front Pharmacol 2022; 13:935685. [PMID: 35991901 PMCID: PMC9386001 DOI: 10.3389/fphar.2022.935685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Xian-Ling-Gu-Bao (XLGB) capsule, a well-known traditional Chinese medicine prescription, is widely used for the treatment of osteoporosis. It could significantly increase the levels of estrogen in ovariectomized rats and mice. However, this working mechanism has not been well elucidated. Considering that UDP-glucuronosyltransferase (UGT) enzymes are the important enzymes that inactivate and regulate estrogen activity in vivo, this study aimed to identify the bioactive compounds from XLGB against the glucuronidation of estrogens. First, thirty compounds were considered as candidate bioactive compounds based on our previous studies including pharmacological evaluation, chemical profiles, and metabolic profiles. Second, the characteristics of estrogen glucuronidation by uridine diphosphate glucuronic acid (UDPGA)-supplemented human liver microsomes (HLM), human intestine microsomes (HIM), and expressed UGT enzymes were determined, and the incubation systems of their key UGT enzymes were optimized. Then, inhibitory effects and mechanisms of XLGB and its main compounds toward the key UGT isozymes were further investigated. As a result, estrogen underwent efficient glucuronidation by HLM and HIM. UGT1A10, 1A1, and 2B7 were mainly responsible for the glucuronidation of estrone, β-estradiol, and estriol, respectively. For E1 and E2, UGT1A10 and 1A1 tended to mediate estrogen-3-O-glucuronidation, while UGT2B7 preferred catalyzing estrogen-16-O-glucuronidation. Furthermore, the incubation system for active UGT isoforms was optimized including Tris-HCl buffer, detergents, MgCl2 concentration, β-glucuronidase inhibitors, UDPGA concentration, protein concentration, and incubation time. Based on optimal incubation conditions, eleven, nine, and nine compounds were identified as the potent inhibitors for UGT1A10, 1A1, and 2B7, respectively (IC50 < 4.97 μM and Ki < 3.35 μM). Among them, six compounds (bavachin, isobavachin, isobavachalcone, neobavaisoflavone, corylifol A, and icariside II) simultaneously demonstrated potent inhibitory effects against these three active enzymes. Prenylated flavanols from Epimedium brevicornu Maxim., prenylated flavonoids from Psoralea corylifolia L., and salvianolic acids from Salvia miltiorrhiza Bge. were characterized as the most important and effective compounds. The identification of potent natural inhibitors of XLGB against the glucuronidation of estrogen laid an important foundation for the pharmacodynamic material basis.
Collapse
Affiliation(s)
- Liangliang He
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Chunxia Xu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Ziying Wang
- School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Shuyi Duan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinjin Xu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Chuan Li
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinsheng Yao
- College of Pharmacy, Jinan University, Guangzhou, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development Ministry of P.R. China, Jinan University, Guangzhou, China
| | | | - Zifei Qin
- College of Pharmacy, Jinan University, Guangzhou, China
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhihong Yao, ; Zifei Qin,
| | - Zhihong Yao
- College of Pharmacy, Jinan University, Guangzhou, China
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development Ministry of P.R. China, Jinan University, Guangzhou, China
- *Correspondence: Zhihong Yao, ; Zifei Qin,
| |
Collapse
|
10
|
Järvinen E, Deng F, Kiander W, Sinokki A, Kidron H, Sjöstedt N. The Role of Uptake and Efflux Transporters in the Disposition of Glucuronide and Sulfate Conjugates. Front Pharmacol 2022; 12:802539. [PMID: 35095509 PMCID: PMC8793843 DOI: 10.3389/fphar.2021.802539] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Glucuronidation and sulfation are the most typical phase II metabolic reactions of drugs. The resulting glucuronide and sulfate conjugates are generally considered inactive and safe. They may, however, be the most prominent drug-related material in the circulation and excreta of humans. The glucuronide and sulfate metabolites of drugs typically have limited cell membrane permeability and subsequently, their distribution and excretion from the human body requires transport proteins. Uptake transporters, such as organic anion transporters (OATs and OATPs), mediate the uptake of conjugates into the liver and kidney, while efflux transporters, such as multidrug resistance proteins (MRPs) and breast cancer resistance protein (BCRP), mediate expulsion of conjugates into bile, urine and the intestinal lumen. Understanding the active transport of conjugated drug metabolites is important for predicting the fate of a drug in the body and its safety and efficacy. The aim of this review is to compile the understanding of transporter-mediated disposition of phase II conjugates. We review the literature on hepatic, intestinal and renal uptake transporters participating in the transport of glucuronide and sulfate metabolites of drugs, other xenobiotics and endobiotics. In addition, we provide an update on the involvement of efflux transporters in the disposition of glucuronide and sulfate metabolites. Finally, we discuss the interplay between uptake and efflux transport in the intestine, liver and kidneys as well as the role of transporters in glucuronide and sulfate conjugate toxicity, drug interactions, pharmacogenetics and species differences.
Collapse
Affiliation(s)
- Erkka Järvinen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alli Sinokki
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
11
|
Gwinn JK, Uhlig S, Ivanova L, Fæste CK, Kryuchkov F, Robertson A. In Vitro Glucuronidation of Caribbean Ciguatoxins in Fish: First Report of Conjugative Ciguatoxin Metabolites. Chem Res Toxicol 2021; 34:1910-1925. [PMID: 34319092 PMCID: PMC9215509 DOI: 10.1021/acs.chemrestox.1c00181] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ciguatoxins (CTX) are potent marine neurotoxins, which can bioaccumulate in seafood, causing a severe and prevalent human illness known as ciguatera poisoning (CP). Despite the worldwide impact of ciguatera, effective disease management is hindered by a lack of knowledge regarding the movement and biotransformation of CTX congeners in marine food webs, particularly in the Caribbean and Western Atlantic. In this study we investigated the hepatic biotransformation of C-CTX across several fish and mammalian species through a series of in vitro metabolism assays focused on phase I (CYP P450; functionalization) and phase II (UGT; conjugation) reactions. Using liquid chromatography high-resolution mass spectrometry to explore potential C-CTX metabolites, we observed two glucuronide products of C-CTX-1/-2 and provided additional evidence from high-resolution tandem mass spectrometry to support their identification. Chemical reduction experiments confirmed that the metabolites were comprised of four distinct glucuronide products with the sugar attached at two separate sites on C-CTX-1/-2 and excluded the C-56 hydroxyl group as the conjugation site. Glucuronidation is a novel biotransformation pathway not yet reported for CTX or other related polyether phycotoxins, yet its occurrence across all fish species tested suggests that it could be a prevalent and important detoxification mechanism in marine organisms. The absence of glucuronidation observed in this study for both rat and human microsomes suggests that alternate biotransformation pathways may be dominant in higher vertebrates.
Collapse
Affiliation(s)
- Jessica Kay Gwinn
- School of Marine and Environmental Sciences, University of South Alabama, Mobile, Alabama 36688, United States
- Dauphin Island Sea Lab, Dauphin Island, Alabama 36528, United States
| | - Silvio Uhlig
- Toxinology Research Group, Norwegian Veterinary Institute, Ås NO-1431, Norway
| | - Lada Ivanova
- Toxinology Research Group, Norwegian Veterinary Institute, Ås NO-1431, Norway
| | | | - Fedor Kryuchkov
- Toxinology Research Group, Norwegian Veterinary Institute, Ås NO-1431, Norway
| | - Alison Robertson
- School of Marine and Environmental Sciences, University of South Alabama, Mobile, Alabama 36688, United States
- Dauphin Island Sea Lab, Dauphin Island, Alabama 36528, United States
| |
Collapse
|
12
|
Fashe M, Yi M, Sueyoshi T, Negishi M. Sex-specific expression mechanism of hepatic estrogen inactivating enzyme and transporters in diabetic women. Biochem Pharmacol 2021; 190:114662. [PMID: 34157297 DOI: 10.1016/j.bcp.2021.114662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 12/21/2022]
Abstract
Circulating estrogens levels significantly decrease in menopause and levels off in postmenopausal women. Accordingly, the liver represses levels of enzymes and membrane transporters, thereby decreasing capability of inactivating and excreting estrogens. Women increasingly develop type 2 diabetes during or after menopause. Estrogens are known to promote liver diseases in these women. Here, we have found that the estrogen inactivating sulfotransferase (SULT1E1) and an ATP-binding cassette subfamily G member 2 (ABCG2), a gene encoding breast cancer resistance protein that exports sulfated estrogens, increased their expression levels in diabetic women but not men. For the sulfotransferase gene, phosphorylated nuclear receptors ERα and RORα, at Ser212 and Ser100, respectively, bind their response elements to activate the SULT1E1 promoter in women. This coordinated increase in estrogen inactivation and excretion, and the phosphorylated nuclear receptor-mediated gene activation could be a defense mechanism against toxicities of estrogens through inactivation and excretion in the livers of women.
Collapse
Affiliation(s)
- Muluneh Fashe
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| | - MyeongJin Yi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Tatsuya Sueyoshi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Masahiko Negishi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
13
|
Chapa R, Li CY, Basit A, Thakur A, Ladumor MK, Sharma S, Singh S, Selen A, Prasad B. Contribution of Uptake and Efflux Transporters to Oral Pharmacokinetics of Furosemide. ACS OMEGA 2020; 5:32939-32950. [PMID: 33403255 PMCID: PMC7774078 DOI: 10.1021/acsomega.0c03930] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/03/2020] [Indexed: 05/17/2023]
Abstract
Furosemide is a widely used diuretic for treating excessive fluid accumulation caused by disease conditions like heart failure and liver cirrhosis. Furosemide tablet formulation exhibits variable pharmacokinetics (PK) with bioavailability ranging from 10 to almost 100%. To explain the variable absorption, we integrated the physicochemical, in vitro dissolution, permeability, distribution, and the elimination parameters of furosemide in a physiologically-based pharmacokinetic (PBPK) model. Although the intravenous PBPK model reasonably described the observed in vivo PK data, the reported low passive permeability failed to capture the observed data after oral administration. To mechanistically justify this discrepancy, we hypothesized that transporter-mediated uptake contributes to the oral absorption of furosemide in conjunction with passive permeability. Our in vitro results confirmed that furosemide is a substrate of intestinal breast cancer resistance protein (BCRP), multidrug resistance-associated protein 4 (MRP4), and organic anion transporting polypeptide 2B1 (OATP2B1), but it is not a substrate of P-glycoprotein (P-gp) and MRP2. We then estimated the net transporter-mediated intestinal uptake and integrated it into the PBPK model under both fasting and fed conditions. Our in vitro data and PBPK model suggest that the absorption of furosemide is permeability-limited, and OATP2B1 and MRP4 are important for its permeability across intestinal membrane. Further, as furosemide has been proposed as a probe substrate of renal organic anion transporters (OATs) for assessing clinical drug-drug interactions (DDIs) during drug development, the confounding effects of intestinal transporters identified in this study on furosemide PK should be considered in the clinical transporter DDI studies.
Collapse
Affiliation(s)
- Revathi Chapa
- Department
of Pharmaceutics, University of Washington, Seattle, Washington 98195-0005, United States
| | - Cindy Yanfei Li
- Department
of Pharmaceutics, University of Washington, Seattle, Washington 98195-0005, United States
| | - Abdul Basit
- College
of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Aarzoo Thakur
- National
Institute of Pharmaceutical
Education and Research (NIPER), SAS Nagar, Punjab 160062, India
| | - Mayur K Ladumor
- Department
of Pharmaceutics, University of Washington, Seattle, Washington 98195-0005, United States
- National
Institute of Pharmaceutical
Education and Research (NIPER), SAS Nagar, Punjab 160062, India
| | - Sheena Sharma
- College
of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
- National
Institute of Pharmaceutical
Education and Research (NIPER), SAS Nagar, Punjab 160062, India
| | - Saranjit Singh
- National
Institute of Pharmaceutical
Education and Research (NIPER), SAS Nagar, Punjab 160062, India
| | - Arzu Selen
- Office
of Testing and Research, Office of Pharmaceutical Quality, CDER/ FDA, Silver
Spring, Maryland 20903-1058, United States
| | - Bhagwat Prasad
- College
of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| |
Collapse
|
14
|
Bangma J, Szilagyi J, Blake BE, Plazas C, Kepper S, Fenton SE, Fry RC. An assessment of serum-dependent impacts on intracellular accumulation and genomic response of per- and polyfluoroalkyl substances in a placental trophoblast model. ENVIRONMENTAL TOXICOLOGY 2020; 35:1395-1405. [PMID: 32790152 PMCID: PMC7738272 DOI: 10.1002/tox.23004] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/07/2020] [Accepted: 07/11/2020] [Indexed: 05/18/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS), a class of environmental contaminants, have been detected in human placenta and cord blood. The mechanisms driving PFAS-induced effects on the placenta and adverse pregnancy outcomes are not well understood. This study investigated the impact of perfluorooctanesulfonic acid (PFOS), perfluorooctanoic acid (PFOA), and a replacement PFAS known as hexafluoropropylene oxide dimer acid (HFPO-DA, tradename GenX) on placental trophoblasts in vitro. Several key factors were addressed. First, PFAS levels in cell culture reagents at baseline were quantified. Second, the role of supplemental media serum in intracellular accumulation of PFAS in a human trophoblast (JEG3) cell line was established. Finally, the impact of PFAS on the expression of 96 genes involved in proper placental function in JEG3 cells was evaluated. The results revealed that serum-free media (SFM) contained no detectable PFAS. In contrast, fetal bovine serum-supplemented media (SSM) contained PFNA, PFUdA, PFTrDA, and 6:2 FTS, but these PFAS were not detected internally in cells. Intracellular accumulation following 24 hr treatments was significantly higher when cultured in SFM compared to SSM for PFOS and PFOA, but not HFPO-DA. Treatment with PFAS was associated with gene expression changes (n = 32) in pathways vital to placental function, including viability, syncytialization, inflammation, transport, and invasion/mesenchymal transition. Among the most robust PFAS-associated changes were those observed in the known apoptosis-related genes, BAD and BAX. These results suggest a complex relationship between PFAS, in vitro culture conditions, and altered expression of key genes necessary for proper placentation.
Collapse
Affiliation(s)
- Jacqueline Bangma
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John Szilagyi
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Bevin E. Blake
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of the National Toxicology Program (DNTP), NTP Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, North Carolina, USA
| | - Cinthya Plazas
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stewart Kepper
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Suzanne E. Fenton
- Division of the National Toxicology Program (DNTP), NTP Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, North Carolina, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Corresponding author: Rebecca Fry, PhD, , Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
15
|
The Relative Importance of the Small Intestine and the Liver in Phase II Metabolic Transformations and Elimination of p-Nitrophenol Administered in Different Doses in the Rat. Sci Pharm 2020. [DOI: 10.3390/scipharm88040051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Intestinal and hepatic function have been investigated in phase II metabolic reactions and elimination of p-nitrophenol (PNP) in the rat. A jejunal loop was cannulated and recirculated with isotonic solutions containing PNP in different concentrations (0, 20, 100, 500, 1000 µM). Samples were obtained from the perfusate at given intervals. To investigate the metabolic and excretory functions of the liver, the bile duct was cannulated, and the bile was collected. Metabolites of PNP were determined by validated HPLC (high pressure liquid chromatography) methods. The results demonstrated the relative importance of the small intestine and the liver in phase II metabolic transformations and elimination of PNP. There were significant differences between the luminal and biliary appearances of p-nitrophenol-glucuronide (PNP-G) and p-nitrophenol–sulfate (PNP-S). The PNP-G appeared in the intestinal lumen at the lower PNP concentrations (20 µM and 100 µM) at higher rate than in the bile. No significant difference was found between the intestinal and the biliary excretion of PNP-G when PNP was administered at a concentration of 500 µM. However, a reverse ratio of these parameters was observed at the administration of 1000 µM PNP. The results indicated that both the small intestine and the liver might play an important role in phase II metabolic reactions and elimination of PNP. However, the relative importance of the small intestine and the liver can be dependent on the dose of drugs.
Collapse
|
16
|
Deng F, Ghemtio L, Grazhdankin E, Wipf P, Xhaard H, Kidron H. Binding Site Interactions of Modulators of Breast Cancer Resistance Protein, Multidrug Resistance-Associated Protein 2, and P-Glycoprotein Activity. Mol Pharm 2020; 17:2398-2410. [PMID: 32496785 PMCID: PMC7497665 DOI: 10.1021/acs.molpharmaceut.0c00155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
ATP-binding cassette (ABC)-transporters
protect tissues by pumping
their substrates out of the cells in many physiological barriers,
such as the blood–brain barrier, intestine, liver, and kidney.
These substrates include various endogenous metabolites, but, in addition,
ABC transporters recognize a wide range of compounds, therefore affecting
the disposition and elimination of clinically used drugs and their
metabolites. Although numerous ABC-transporter inhibitors are known,
the underlying mechanism of inhibition is not well characterized.
The aim of this study is to deepen our understanding of transporter
inhibition by studying the molecular basis of ligand recognition.
In the current work, we compared the effect of 44 compounds on the
active transport mediated by three ABC transporters: breast cancer
resistance protein (BCRP and ABCG2), multidrug-resistance associated
protein (MRP2 and ABCC2), and P-glycoprotein (P-gp and ABCB1). Eight
compounds were strong inhibitors of all three transporters, while
the activity of 36 compounds was transporter-specific. Of the tested
compounds, 39, 25, and 11 were considered as strong inhibitors, while
1, 4, and 11 compounds were inactive against BCRP, MRP2, and P-gp,
respectively. In addition, six transport-enhancing stimulators were
observed for P-gp. In order to understand the observed selectivity,
we compared the surface properties of binding cavities in the transporters
and performed structure–activity analysis and computational
docking of the compounds to known binding sites in the transmembrane
domains and nucleotide-binding domains. Based on the results, the
studied compounds are more likely to interact with the transmembrane
domain than the nucleotide-binding domain. Additionally, the surface
properties of the substrate binding site in the transmembrane domains
of the three transporters were in line with the observed selectivity.
Because of the high activity toward BCRP, we lacked the dynamic range
needed to draw conclusions on favorable interactions; however, we
identified amino acids in both P-gp and MRP2 that appear to be important
for ligand recognition.
Collapse
Affiliation(s)
- Feng Deng
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| | - Leo Ghemtio
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| | - Evgeni Grazhdankin
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| | - Peter Wipf
- Department of Chemistry, The Center for Chemical Methodologies and Library Development, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Henri Xhaard
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| |
Collapse
|
17
|
Järvinen E, Kidron H, Finel M. Human efflux transport of testosterone, epitestosterone and other androgen glucuronides. J Steroid Biochem Mol Biol 2020; 197:105518. [PMID: 31704245 DOI: 10.1016/j.jsbmb.2019.105518] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/25/2019] [Accepted: 10/26/2019] [Indexed: 01/01/2023]
Abstract
Several drug-metabolizing enzymes are known to control androgen homeostasis in humans. UDP-glucuronosyltransferases convert androgens to glucuronide conjugates in the liver and intestine, which enables subsequent elimination of these conjugated androgens via urine. The most important androgen is testosterone, while others are the testosterone metabolites androsterone and etiocholanolone, and the testosterone precursor dehydroepiandrosterone. Epitestosterone is another endogenous androgen, which is included as a crucial marker in urine doping tests. Since glucuronide conjugates are hydrophilic, efflux transporters mediate their excretion from tissues. In this study, we employed the membrane vesicle assay to identify the efflux transporters for glucuronides of androsterone, dehydroepiandrosterone, epitestosterone, etiocholanolone and testosterone. The human hepatic and intestinal transporters MRP2 (ABCC2), MRP3 (ABCC3), MRP4 (ABCC4), BCRP (ABCG2) and MDR1 (ABCB1) were studied in vitro. Of these transporters, only MRP2 and MRP3 transported the androgen glucuronides investigated. In kinetic analyses, MRP3 transported glucuronides of androsterone, epitestosterone and etiocholanolone at low Km values, between 0.4 and 4 μM, while the Km values for glucuronides of testosterone and dehydroepiandrosterone were 14 and 51 μM, respectively. MRP2 transported the glucuronides at lower affinity, as indicated by Km values over 100 μM. Interestingly, the MRP2-mediated transport of androsterone and epitestosterone glucuronides was best described by sigmoidal kinetics. The inability of BCRP to transport any of the androgen glucuronides investigated is drastically different from its highly active transport of several estrogen conjugates. Our results explain the transporter-mediated disposition of androgen glucuronides in humans, and shed light on differences between the human efflux transporters MRP2, MRP3, MRP4, BCRP and MDR1.
Collapse
Affiliation(s)
- Erkka Järvinen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland.
| | - Heidi Kidron
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Finland
| | - Moshe Finel
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland
| |
Collapse
|
18
|
Transporters in the Mammary Gland-Contribution to Presence of Nutrients and Drugs into Milk. Nutrients 2019; 11:nu11102372. [PMID: 31590349 PMCID: PMC6836069 DOI: 10.3390/nu11102372] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/19/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
A large number of nutrients and bioactive ingredients found in milk play an important role in the nourishment of breast-fed infants and dairy consumers. Some of these ingredients include physiologically relevant compounds such as vitamins, peptides, neuroactive compounds and hormones. Conversely, milk may contain substances-drugs, pesticides, carcinogens, environmental pollutants-which have undesirable effects on health. The transfer of these compounds into milk is unavoidably linked to the function of transport proteins. Expression of transporters belonging to the ATP-binding cassette (ABC-) and Solute Carrier (SLC-) superfamilies varies with the lactation stages of the mammary gland. In particular, Organic Anion Transporting Polypeptides 1A2 (OATP1A2) and 2B1 (OATP2B1), Organic Cation Transporter 1 (OCT1), Novel Organic Cation Transporter 1 (OCTN1), Concentrative Nucleoside Transporters 1, 2 and 3 (CNT1, CNT2 and CNT3), Peptide Transporter 2 (PEPT2), Sodium-dependent Vitamin C Transporter 2 (SVCT2), Multidrug Resistance-associated Protein 5 (ABCC5) and Breast Cancer Resistance Protein (ABCG2) are highly induced during lactation. This review will focus on these transporters overexpressed during lactation and their role in the transfer of products into the milk, including both beneficial and harmful compounds. Furthermore, additional factors, such as regulation, polymorphisms or drug-drug interactions will be described.
Collapse
|
19
|
Li CY, Basit A, Gupta A, Gáborik Z, Kis E, Prasad B. Major glucuronide metabolites of testosterone are primarily transported by MRP2 and MRP3 in human liver, intestine and kidney. J Steroid Biochem Mol Biol 2019; 191:105350. [PMID: 30959153 PMCID: PMC7075494 DOI: 10.1016/j.jsbmb.2019.03.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/25/2019] [Accepted: 03/30/2019] [Indexed: 01/29/2023]
Abstract
Testosterone glucuronide (TG), androsterone glucuronide (AG), etiocholanolone glucuronide (EtioG) and dihydrotestosterone glucuronide (DHTG) are the major metabolites of testosterone (T), which are excreted in urine and bile. Glucuronides can be deconjugated to active androgen in gut lumen after biliary excretion, which in turn can affect physiological levels of androgens. The goal of this study was to quantitatively characterize the mechanisms by which TG, AG, EtioG and DHTG are eliminated from liver, intestine, and kidney utilizing relative expression factor (REF) approach. Using vesicular transport assay with recombinant human MRP2, MRP3, MRP4, MDR1 and BCRP, we first identified that TG, AG, EtioG, and DHTG were primarily substrates of MRP2 and MRP3, although lower levels of transport were also observed with MDR1 and BCRP vesicles. The transport kinetic analyses revealed higher intrinsic clearances of TG by MRP2 and MRP3 as compared to that of DHTG, AG, and EtioG. MRP3 exhibited higher affinity for the transport of the studied glucuronides than MRP2. We next quantified the protein abundances of these efflux transporters in vesicles and compared the same with pooled total membrane fractions isolated from human tissues by quantitative LC-MS/MS proteomics. The fractional contribution of individual transporters (ft) was estimated by proteomics-based physiological scaling factors, i.e., transporter abundance in whole tissue versus vesicles, and corrected for inside-out vesicles (determined by 5'-nucleotidase assay). The glucuronides of inactive androgens, AG and EtioG were preferentially transported by MRP3, whereas the glucuronides of active androgens, TG and DHTG were mainly transported by MRP2 in liver. Efflux by bile canalicular transport may indicate the potential role of enterohepatic recirculation in regulating the circulating active androgens after deconjugation in the gut. In intestine, MRP3 possibly contributes most to the efflux of these glucuronides. In kidney, all studied glucuronides seemed to be preferentially effluxed by MRP2 and MDR1 (for EtioG). These REF based analysis need to be confirmed with in vivo findings. Overall, characterization of the efflux mechanisms of T glucuronide metabolites is important for predicting the androgen disposition and interindividual variability, including drug-androgen interaction in humans. The mechanistic data can be extrapolated to other androgen relevant organs (e.g. prostate, testis and placenta) by integrating these data with quantitative tissue proteomics data.
Collapse
Affiliation(s)
- Cindy Yanfei Li
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Abdul Basit
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Anshul Gupta
- Amgen Research, Department of Pharmacokinetics and Drug Metabolism, Cambridge, MA, USA
| | | | - Emese Kis
- SOLVO Biotechnology, Budapest, Hungary
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
20
|
Sjöstedt N, Salminen TA, Kidron H. Endogenous, cholesterol-activated ATP-dependent transport in membrane vesicles from Spodoptera frugiperda cells. Eur J Pharm Sci 2019; 137:104963. [PMID: 31226387 DOI: 10.1016/j.ejps.2019.104963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/04/2019] [Accepted: 06/17/2019] [Indexed: 12/19/2022]
Abstract
Transport proteins of the ATP-binding cassette (ABC) family are found in all kingdoms of life. In humans, several ABC efflux transporters play a role in drug disposition and excretion. Therefore, in vitro methods have been developed to characterize the substrate and inhibitor properties of drugs with respect to these transporters. In the vesicular transport assay, transport is studied using inverted membrane vesicles produced from transporter overexpressing cell lines of both mammalian and insect origin. Insect cell expression systems benefit from a higher expression compared to background, but are not as well characterized as their mammalian counterparts regarding endogenous transport. Therefore, the contribution of this transport in the assay might be underappreciated. In this study, endogenous transport in membrane vesicles from Spodoptera frugiperda -derived Sf9 cells was characterized using four typical substrates of human ABC transporters: 5(6)-carboxy-2,'7'-dichlorofluorescein (CDCF), estradiol-17β-glucuronide, estrone sulfate and N-methyl-quinidine. Significant ATP-dependent transport was observed for three of the substrates with cholesterol-loading of the vesicles, which is sometimes used to improve the activity of human transporters expressed in Sf9 cells. The highest effect of cholesterol was on CDCF transport, and this transport in the cholesterol-loaded Sf9 vesicles was time and concentration dependent with a Km of 8.06 ± 1.11 μM. The observed CDCF transport was inhibited by known inhibitors of human ABCC transporters, but not by ABCB1 and ABCG2 inhibitors verapamil and Ko143, respectively. Two candidate genes for ABCC-type transporters in the S. frugiperda genome (SfABCC2 and SfABCC3) were identified based on sequence analysis as a hypothesis to explain the observed endogenous ABCC-type transport in Sf9 vesicles. Although further studies are needed to verify the role of SfABCC2 and SfABCC3 in Sf9 vesicles, the findings of this study highlight the need to carefully characterize background transport in Sf9 derived membrane vesicles to avoid false positive substrate findings for human ABC transporters studied with this overexpression system.
Collapse
Affiliation(s)
- Noora Sjöstedt
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| | - Tiina A Salminen
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Heidi Kidron
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|