1
|
Bintee B, Banerjee R, Hegde M, Vishwa R, Alqahtani MS, Abbas M, Alqahtani A, Rangan L, Sethi G, Kunnumakkara AB. Exploring bile acid transporters as key players in cancer development and treatment: Evidence from preclinical and clinical studies. Cancer Lett 2025; 609:217324. [PMID: 39571783 DOI: 10.1016/j.canlet.2024.217324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 12/01/2024]
Abstract
Bile acid transporters (BATs) are integral membrane proteins belonging to various families, such as solute carriers, organic anion transporters, and ATP-binding cassette families. These transporters play a crucial role in bile acid transportation within the portal and systemic circulations, with expression observed in tissues, including the liver, kidney, and small intestine. Bile acids serve as signaling molecules facilitating the absorption and reabsorption of fats and lipids. Dysregulation of bile acid concentration has been implicated in tumorigenesis, yet the role of BATs in this process remains underexplored. Emerging evidence suggests that BATs may modulate various stages of cancer progression, including initiation, development, proliferation, metastasis, and tumor microenvironment regulation. Targeting BATs using siRNAs, miRNAs, and small compound inhibitors in preclinical models and their polymorphisms are well-studied for transporters like BSEP, MDR1, MRP2, OATP1A2, etc., and have shed light on their involvement in tumorigenesis, particularly in cancers such as those affecting the liver and gastrointestinal tract. While BATs' role in diseases like Alagille syndrome, biliary atresia, and cirrhosis have been extensively studied, their implications in cancer warrant further investigation. This review highlights the expression and function of BATs in cancer development and emphasizes the potential of targeting these transporters as a novel therapeutic strategy for various malignancies.
Collapse
Affiliation(s)
- Bintee Bintee
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Ruchira Banerjee
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India; Applied Biodiversity Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Ravichandran Vishwa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Athba Alqahtani
- Research Centre, King Fahad Medical City, P.O. Box: 59046, Riyadh, 11525, Saudi Arabia
| | - Latha Rangan
- Applied Biodiversity Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
2
|
Hahn KR, Kwon HJ, Kim DW, Hwang IK, Yoon YS. Therapeutic Options of Crystallin Mu and Protein Disulfide Isomerase A3 for Cuprizone-Induced Demyelination in Mouse Hippocampus. Neurochem Res 2024; 49:3078-3093. [PMID: 39164609 PMCID: PMC11449959 DOI: 10.1007/s11064-024-04227-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024]
Abstract
This study investigates the changes in hippocampal proteomic profiles during demyelination and remyelination using the cuprizone model. Employing two-dimensional gel electrophoresis and liquid chromatography-tandem mass spectrometry for protein profiling, we observed significant alterations in the expression of ketimine reductase mu-crystallin (CRYM) and protein disulfide isomerase A3 precursor (PDIA3) following exposure to and subsequent withdrawal from cuprizone. Immunohistochemical staining validated these protein expression patterns in the hippocampus, revealing that both PDIA3 and CRYM were downregulated in the hippocampal CA1 region during demyelination and upregulated during remyelination. Additionally, we explored the potential protective effects of CRYM and PDIA3 against cuprizone-induced demyelination by synthesizing cell-permeable Tat peptide-fusion proteins (Tat-CRYM and Tat-PDIA3) to facilitate their crossing through the blood-brain barrier. Our results indicated that administering Tat-CRYM and Tat-PDIA3 mitigated the reduction in proliferating cell and differentiated neuroblast counts compared to the group receiving cuprizone alone. Notably, Tat-PDIA3 demonstrated significant effects in enhancing myelin basic protein expression alongside phosphorylation of CREB in the hippocampus, suggesting its potential therapeutic role in the prevention or treatment of demyelination, and by extension, in conditions such as multiple sclerosis.
Collapse
Affiliation(s)
- Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
- Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
3
|
Li S, Hao L, Deng J, Zhang J, Yu F, Ye F, Li N, Hu X. The Culprit Behind HBV-Infected Hepatocytes: NTCP. Drug Des Devel Ther 2024; 18:4839-4858. [PMID: 39494152 PMCID: PMC11529284 DOI: 10.2147/dddt.s480151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Hepatitis B virus (HBV) is a globally prevalent human DNA virus responsible for over 250 million cases of chronic liver infections, leading to conditions such as liver inflammation, cirrhosis and hepatocellular carcinoma (HCC). Sodium taurocholate co-transporting polypeptide (NTCP) is a transmembrane protein highly expressed in human hepatocytes and functions as a bile acid (BA) transporter. NTCP has been identified as the receptor that HBV and its satellite virus, hepatitis delta virus (HDV), use to enter hepatocytes. HBV entry into hepatocytes is tightly regulated by various signaling pathways, and NTCP plays an important role as the initial stage of HBV infection. NTCP acts as an initiation signal, causing metabolic changes in hepatocytes and facilitating the entry of HBV into hepatocytes. Thus, a comprehensive understanding of NTCP's role is crucial. In this review, we will examine the regulatory mechanisms governing HBV pre-S1 binding to liver membrane NTCP, the role of NTCP in HBV internalization, and the transcriptional and translational regulation of NTCP expression. Additionally, we will discuss clinical drugs targeting NTCP, including combination therapies involving NTCP inhibitors, and consider the safety of NTCP as a therapeutic target.
Collapse
Affiliation(s)
- Shenghao Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Liyuan Hao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Jiali Deng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Junli Zhang
- Department of Infectious Diseases, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu Province, People’s Republic of China
| | - Fei Yu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Fanghang Ye
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Na Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
4
|
Chen Z, Yildiz S, Markova B, de Rooij LJ, Leeuwenburgh S, Hamers T, Peeters RP, Heuer H, Meima ME, Visser WE. 3,3',5-Triiodothyroacetic Acid Transporters. Thyroid 2024; 34:1027-1037. [PMID: 38836423 DOI: 10.1089/thy.2023.0467] [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] [Indexed: 06/06/2024]
Abstract
Introduction: Thyroid hormone transporters are essential for thyroid hormones to enter target cells. Monocarboxylate transporter (MCT) 8 is a key transporter and is expressed at the blood-brain barrier (BBB), in neural cells and many other tissues. Patients with MCT8 deficiency have severe neurodevelopmental delays because of cerebral hypothyroidism and chronic sequelae of peripheral thyrotoxicosis. The T3 analog 3,3',5-triiodothyroacetic acid (TRIAC) rescued neurodevelopmental features in animal models mimicking MCT8 deficiency and improved key metabolic features in patients with MCT8 deficiency. However, the identity of the transporter(s) that facilitate TRIAC transport are unknown. Here, we screened candidate transporters that are expressed at the human BBB and/or brain-cerebrospinal fluid barrier and known thyroid hormone transporters for TRIAC transport. Materials and Methods: Plasma membrane expression was determined by cell surface biotinylation assays. Intracellular accumulation of 1 nM TRIAC was assessed in COS-1 cells expressing candidate transporters in Dulbecco's phosphate-buffered saline (DPBS)/0.1% glucose or Dulbecco's modified Eagle's medium (DMEM) with or without 0.1% bovine serum albumin (BSA). Expression of Slc22a8 was determined by fluorescent in situ hybridization in brain sections from wild-type and Mct8/Oatp1c1 knockout mice at postnatal days 12, 21, and 120. Results: In total, 59 plasma membrane transporters were selected for screening of TRIAC accumulation (n = 40 based on expression at the human BBB and/or brain-cerebrospinal fluid barrier and having small organic molecules as substrates; n = 19 known thyroid hormone transporters). Screening of the selected transporter panel showed that 18 transporters facilitated significant intracellular accumulation of TRIAC in DPBS/0.1% glucose or DMEM in the absence of BSA. In the presence of BSA, substantial transport was noted for SLCO1B1 and SLC22A8 (in DPBS/0.1% glucose and DMEM) and SLC10A1, SLC22A6, and SLC22A24 (in DMEM). The zebrafish and mouse orthologs of these transporters similarly facilitated intracellular accumulation of TRIAC. Highest Slc22a8 mRNA expression was detected in mouse brain capillary endothelial cells and choroid plexus epithelial cells at early postnatal time points, but was reduced at P120. Conclusions: Human SLC10A1, SLCO1B1, SLC22A6, SLC22A8, and SLC22A24 as well as their mouse and zebrafish orthologs are efficient TRIAC transporters. These findings contribute to the understanding of TRIAC treatment in patients with MCT8 deficiency and animal models thereof.
Collapse
Affiliation(s)
- Zhongli Chen
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Sena Yildiz
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Boyka Markova
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Linda J de Rooij
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Selmar Leeuwenburgh
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Timo Hamers
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, the Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Marcel E Meima
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus University Medical Center Rotterdam, the Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus University Medical Center Rotterdam, the Netherlands
| |
Collapse
|
5
|
Zakrzewicz D, Geyer J. Interactions of Na +/taurocholate cotransporting polypeptide with host cellular proteins upon hepatitis B and D virus infection: novel potential targets for antiviral therapy. Biol Chem 2023:hsz-2022-0345. [PMID: 37103224 DOI: 10.1515/hsz-2022-0345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/13/2023] [Indexed: 04/28/2023]
Abstract
Na+/taurocholate cotransporting polypeptide (NTCP) is a member of the solute carrier (SLC) family 10 transporters (gene symbol SLC10A1) and is responsible for the sodium-dependent uptake of bile salts across the basolateral membrane of hepatocytes. In addition to its primary transporter function, NTCP is the high-affinity hepatic receptor for hepatitis B (HBV) and hepatitis D (HDV) viruses and, therefore, is a prerequisite for HBV/HDV virus entry into hepatocytes. The inhibition of HBV/HDV binding to NTCP and internalization of the virus/NTCP receptor complex has become a major concept in the development of new antiviral drugs called HBV/HDV entry inhibitors. Hence, NTCP has emerged as a promising target for therapeutic interventions against HBV/HDV infections in the last decade. In this review, recent findings on protein-protein interactions (PPIs) between NTCP and cofactors relevant for entry of the virus/NTCP receptor complex are summarized. In addition, strategies aiming to block PPIs with NTCP to dampen virus tropism and HBV/HDV infection rates are discussed. Finally, this article suggests novel directions for future investigations evaluating the functional contribution of NTCP-mediated PPIs in the development and progression of HBV/HDV infection and subsequent chronic liver disorders.
Collapse
Affiliation(s)
- Dariusz Zakrzewicz
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| | - Joachim Geyer
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| |
Collapse
|
6
|
Lazcano I, Pech-Pool SM, Olvera A, García-Martínez I, Palacios-Pérez S, Orozco A. The importance of thyroid hormone signaling during early development: Lessons from the zebrafish model. Gen Comp Endocrinol 2023; 334:114225. [PMID: 36709002 DOI: 10.1016/j.ygcen.2023.114225] [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: 06/29/2022] [Revised: 11/16/2022] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
The zebrafish is an optimal experimental model to study thyroid hormone (TH) involvement in vertebrate development. The use of state-of-the-art zebrafish genetic tools available for the study of the effect of gene silencing, cell fate decisions and cell lineage differentiation have contributed to a more insightful comprehension of molecular, cellular, and tissue-specific TH actions. In contrast to intrauterine development, extrauterine embryogenesis observed in zebrafish has facilitated a more detailed study of the development of the hypothalamic-pituitary-thyroid axis. This model has also enabled a more insightful analysis of TH molecular actions upon the organization and function of the brain, the retina, the heart, and the immune system. Consequently, zebrafish has become a trendy model to address paradigms of TH-related functional and biomedical importance. We here compilate the available knowledge regarding zebrafish developmental events for which specific components of TH signaling are essential.
Collapse
Affiliation(s)
- I Lazcano
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - S M Pech-Pool
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - A Olvera
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - I García-Martínez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - S Palacios-Pérez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - A Orozco
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico; Escuela Nacional de Estudios Superiores, Unidad Juriquilla, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Querétaro 76230, Mexico.
| |
Collapse
|
7
|
Thyroid Hormone Transporters in Pregnancy and Fetal Development. Int J Mol Sci 2022; 23:ijms232315113. [PMID: 36499435 PMCID: PMC9737226 DOI: 10.3390/ijms232315113] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Thyroid hormone is essential for fetal (brain) development. Plasma membrane transporters control the intracellular bioavailability of thyroid hormone. In the past few decades, 15 human thyroid hormone transporters have been identified, and among them, mutations in monocarboxylate transporter (MCT)8 and organic anion transporting peptide (OATP)1C1 are associated with clinical phenotypes. Different animal and human models have been employed to unravel the (patho)-physiological role of thyroid hormone transporters. However, most studies on thyroid hormone transporters focus on postnatal development. This review summarizes the research on the thyroid hormone transporters in pregnancy and fetal development, including their substrate preference, expression and tissue distribution, and physiological and pathophysiological role in thyroid homeostasis and clinical disorders. As the fetus depends on the maternal thyroid hormone supply, especially during the first half of pregnancy, the review also elaborates on thyroid hormone transport across the human placental barrier. Future studies may reveal how the different transporters contribute to thyroid hormone homeostasis in fetal tissues to properly facilitate development. Employing state-of-the-art human models will enable a better understanding of their roles in thyroid hormone homeostasis.
Collapse
|
8
|
Aksoy O, Hantusch B, Kenner L. Emerging role of T3-binding protein μ-crystallin (CRYM) in health and disease. Trends Endocrinol Metab 2022; 33:804-816. [PMID: 36344381 DOI: 10.1016/j.tem.2022.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
Thyroid hormones are essential metabolic and developmental regulators that exert a huge variety of effects in different organs. Triiodothyronine (T3) and thyroxine (T4) are synthesized in the thyroid gland and constitute unique iodine-containing hormones that are constantly regulated by a homeostatic feedback mechanism. T3/T4 activity in cells is mainly determined by specific transporters, cytosolic binding proteins, deiodinases (DIOs), and nuclear receptors. Modulation of intracellular T3/T4 level contributes to the maintenance of this regulatory feedback. μ-Crystallin (CRYM) is an important intracellular high-affinity T3-binding protein that buffers the amount of T3 freely available in the cytosol, thereby controlling its action. In this review, we focus on the molecular and pathological properties of CRYM in thyroid hormone signaling, with emphasis on its critical role in malignancies.
Collapse
Affiliation(s)
- Osman Aksoy
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Brigitte Hantusch
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria; Unit for Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria; Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Kotsopoulou I, Vyas AK, Cory MJ, Chan CS, Jagarapu J, Gill S, Mudduluru M, Angelis D. Developmental changes of the fetal and neonatal thyroid gland and functional consequences on the cardiovascular system. J Perinatol 2022; 42:1576-1586. [PMID: 36376450 DOI: 10.1038/s41372-022-01559-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
Thyroid hormones play an important role in the development and function of the cardiac myocyte. Dysregulation of the thyroid hormone milieu affects the fetal cardiac cells via complex molecular mechanisms, either by altering gene expression or directly by affecting post-translational processes. This review offers a comprehensive summary of the effects of thyroid hormones on the developing cardiovascular system and its adaptation. Furthermore, we will highlight the gaps in knowledge and provide suggestions for future research.
Collapse
Affiliation(s)
- Ioanna Kotsopoulou
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arpita K Vyas
- Division of Pediatrics and Endocrinology, College of Medicine, California Northstate University, Elk Grove, CA, USA
| | - Melinda J Cory
- Division of Cardiology, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christina S Chan
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jawahar Jagarapu
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shamaila Gill
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Manjula Mudduluru
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dimitrios Angelis
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
10
|
Li Y, Zhou J, Li T. Regulation of the HBV Entry Receptor NTCP and its Potential in Hepatitis B Treatment. Front Mol Biosci 2022; 9:879817. [PMID: 35495620 PMCID: PMC9039015 DOI: 10.3389/fmolb.2022.879817] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Hepatitis B virus (HBV) is a globally prevalent human DNA virus responsible for more than 250 million cases of chronic liver infection, a condition that can lead to liver inflammation, cirrhosis, and hepatocellular carcinoma. Sodium taurocholate co-transporting polypeptide (NTCP), a transmembrane protein highly expressed in human hepatocytes and a mediator of bile acid transport, has been identified as the receptor responsible for the cellular entry of both HBV and its satellite, hepatitis delta virus (HDV). This has led to significant advances in our understanding of the HBV life cycle, especially the early steps of infection. HepG2-NTCP cells and human NTCP-expressing transgenic mice have been employed as the primary cell culture and animal models, respectively, for the study of HBV, and represent valuable approaches for investigating its basic biology and developing treatments for infection. However, the mechanisms involved in the regulation of NTCP transcription, translation, post-translational modification, and transport are still largely elusive. Improvements in our understanding of NTCP biology would likely facilitate the design of new therapeutic drugs for the prevention of the de novo infection of naïve hepatocytes. In this review, we provide critical findings regarding NTCP biology and discuss important questions that remain unanswered.
Collapse
Affiliation(s)
- Yan Li
- *Correspondence: Yan Li, ; Tianliang Li,
| | | | | |
Collapse
|
11
|
Multitasking Na+/Taurocholate Cotransporting Polypeptide (NTCP) as a Drug Target for HBV Infection: From Protein Engineering to Drug Discovery. Biomedicines 2022; 10:biomedicines10010196. [PMID: 35052874 PMCID: PMC8773476 DOI: 10.3390/biomedicines10010196] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 02/05/2023] Open
Abstract
Hepatitis B virus (HBV) infections are among the major public health concerns worldwide with more than 250 million of chronically ill individuals. Many of them are additionally infected with the Hepatitis D virus, a satellite virus to HBV. Chronic infection frequently leads to serious liver diseases including cirrhosis and hepatocellular carcinoma, the most common type of liver cancer. Although current antiviral therapies can control HBV replication and slow down disease progress, there is an unmet medical need to identify therapies to cure this chronic infectious disease. Lately, a noteworthy progress in fighting against HBV has been made by identification of the high-affinity hepatic host receptor for HBV and HDV, namely Na+/taurocholate cotransporting polypeptide (NTCP, gene symbol SLC10A1). Next to its primary function as hepatic uptake transporter for bile acids, NTCP is essential for the cellular entry of HBV and HDV into hepatocytes. Due to this high-ranking discovery, NTCP has become a valuable target for drug development strategies for HBV/HDV-infected patients. In this review, we will focus on a newly predicted three-dimensional NTCP model that was generated using computational approaches and discuss its value in understanding the NTCP’s membrane topology, substrate and virus binding taking place in plasma membranes. We will review existing data on structural, functional, and biological consequences of amino acid residue changes and mutations that lead to loss of NTCP’s transport and virus receptor functions. Finally, we will discuss new directions for future investigations aiming at development of new NTCP-based HBV entry blockers that inhibit HBV tropism in human hepatocytes.
Collapse
|
12
|
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
|
13
|
Oswald A, Chakraborty A, Ni Y, Wettengel JM, Urban S, Protzer U. Concentration of Na +-taurocholate-cotransporting polypeptide expressed after in vitro-transcribed mRNA transfection determines susceptibility of hepatoma cells for hepatitis B virus. Sci Rep 2021; 11:19799. [PMID: 34611272 PMCID: PMC8492621 DOI: 10.1038/s41598-021-99263-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/21/2021] [Indexed: 02/07/2023] Open
Abstract
Infection of hepatocytes by hepatitis B virus (HBV) depends on surface expression of its receptor Na+-taurocholate-cotransporting polypeptide (NTCP), but sufficient NTCP expression is lacking in most cell lines. NTCP can be introduced by plasmid transfection or transduction by viral vectors to render cells permissive for HBV. However, transient transfection of hepatocyte-derived cell lines is inefficient, resulting in inhomogeneous protein expression and does not allow to adapt the level of NTCP expression. We therefore utilized in vitro transcribed mRNA to introduce NTCP into cells. Optimization using alternative cap structures and nucleotide modifications rendered mRNA transfection into different non-hepatic and hepatic cell lines very efficient. After transfection of mRNA, surface expression and functionality of NTCP was demonstrated by staining with an N-terminal HBV-preS peptide and bile acid uptake. Introduction of NTCP by mRNA transfection increased susceptibility of hepatoma cells to HBV in a dose-dependent manner. Transfection of NTCP mRNA into non-liver cells, in contrast, supported bile acid uptake but did still not render the cells permissive for HBV, demonstrating the requirement for additional host factors. Introduction of candidate host factors by mRNA transfection will allow for fast and convenient analysis of the viral life cycle using a transient, but reliable expression system.
Collapse
Affiliation(s)
- Andreas Oswald
- Institute of Virology, School of Medicine, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
| | - Anindita Chakraborty
- Institute of Virology, School of Medicine, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
| | - Yi Ni
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Heidelberg, Heidelberg, Germany
| | - Jochen M Wettengel
- Institute of Virology, School of Medicine, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Heidelberg, Heidelberg, Germany
| | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany.
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany.
| |
Collapse
|
14
|
Floerl S, Kuehne A, Geyer J, Brockmoeller J, Tzvetkov MV, Hagos Y. Functional and Pharmacological Comparison of Human and Mouse Na +/Taurocholate Cotransporting Polypeptide (NTCP). SLAS DISCOVERY 2021; 26:1055-1064. [PMID: 34060352 DOI: 10.1177/24725552211017500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The Na+/taurocholate cotransporting polypeptide (NTCP) is located in the basolateral membrane of hepatocytes, where it transports bile acids from the portal blood back into hepatocytes. Furthermore, NTCP has a role for the hepatic transport of some drugs. Extrapolation of drug transport data from rodents to humans is not always possible, because species differences in the expression level, localization, affinity, and substrate selectivity of relevant transport proteins must be considered. In the present study, a functional comparison of human NTCP (hNTCP) and mouse Ntcp (mNtcp) showed similar Km values of 67 ± 10 µM and 104 ± 9 µM for the probe substrate estrone-3-sulfate as well as of 258 ± 42 µM and 199 ± 13 µM for the drug rosuvastatin, respectively. IC50 values for the probe inhibitor cyclosporine A were 3.1 ± 0.3 µM for hNTCP and 1.6 ± 0.4 µM for mNtcp. In a drug and pesticide inhibitory screening on both transporters, 4 of the 15 tested drugs (cyclosporine A, benzbromarone, MK571, and fluvastatin) showed high inhibitory potency, but only slight inhibition was observed for the 13 tested pesticides. Among these compounds, only four drugs and three pesticides showed significant differences in their inhibition pattern on hNTCP and mNtcp. Most pronounced was the difference for benzbromarone with a fivefold higher IC50 for mNtcp (27 ± 10 µM) than for hNTCP (5.5 ± 0.6 µM).In conclusion, we found a strong correlation between the transport kinetics and inhibition pattern among hNTCP and mNtcp. However, specific compounds, such as benzbromarone, showed clear species differences. Such species differences have to be considered when pharmacokinetic data are transferred from rodent to humans.
Collapse
Affiliation(s)
| | | | - Joachim Geyer
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Gießen, Germany
| | - Juergen Brockmoeller
- Institute of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | - Mladen V Tzvetkov
- Institute of Pharmacology Center of Drug Absorption and Transport (C_DAT), University Greifswald, Greifswald, Germany
| | | |
Collapse
|
15
|
Abstract
µ-Crystallin is a NADPH-regulated thyroid hormone binding protein encoded by the CRYM gene in humans. It is primarily expressed in the brain, muscle, prostate, and kidney, where it binds thyroid hormones, which regulate metabolism and thermogenesis. It also acts as a ketimine reductase in the lysine degradation pathway when it is not bound to thyroid hormone. Mutations in CRYM can result in non-syndromic deafness, while its aberrant expression, predominantly in the brain but also in other tissues, has been associated with psychiatric, neuromuscular, and inflammatory diseases. CRYM expression is highly variable in human skeletal muscle, with 15% of individuals expressing ≥13 fold more CRYM mRNA than the median level. Ablation of the Crym gene in murine models results in the hypertrophy of fast twitch muscle fibers and an increase in fat mass of mice fed a high fat diet. Overexpression of Crym in mice causes a shift in energy utilization away from glycolysis towards an increase in the catabolism of fat via β-oxidation, with commensurate changes of metabolically involved transcripts and proteins. The history, attributes, functions, and diseases associated with CRYM, an important modulator of metabolism, are reviewed.
Collapse
Affiliation(s)
- Christian J Kinney
- Department of Physiology School of Medicine, University of Maryland, Baltimore, Baltimore, MD 21201
| | - Robert J Bloch
- Department of Physiology School of Medicine, University of Maryland, Baltimore, Baltimore, MD 21201
| |
Collapse
|
16
|
Aksoy O, Pencik J, Hartenbach M, Moazzami AA, Schlederer M, Balber T, Varady A, Philippe C, Baltzer PA, Mazumder B, Whitchurch JB, Roberts CJ, Haitel A, Herac M, Susani M, Mitterhauser M, Marculescu R, Stangl‐Kremser J, Hassler MR, Kramer G, Shariat SF, Turner SD, Tichy B, Oppelt J, Pospisilova S, Hartenbach S, Tangermann S, Egger G, Neubauer HA, Moriggl R, Culig Z, Greiner G, Hoermann G, Hacker M, Heery DM, Merkel O, Kenner L. Thyroid and androgen receptor signaling are antagonized by μ-Crystallin in prostate cancer. Int J Cancer 2021; 148:731-747. [PMID: 33034050 PMCID: PMC7756625 DOI: 10.1002/ijc.33332] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/02/2020] [Indexed: 12/29/2022]
Abstract
Androgen deprivation therapy (ADT) remains a key approach in the treatment of prostate cancer (PCa). However, PCa inevitably relapses and becomes ADT resistant. Besides androgens, there is evidence that thyroid hormone thyroxine (T4) and its active form 3,5,3'-triiodo-L-thyronine (T3) are involved in the progression of PCa. Epidemiologic evidences show a higher incidence of PCa in men with elevated thyroid hormone levels. The thyroid hormone binding protein μ-Crystallin (CRYM) mediates intracellular thyroid hormone action by sequestering T3 and blocks its binding to cognate receptors (TRα/TRβ) in target tissues. We show in our study that low CRYM expression levels in PCa patients are associated with early biochemical recurrence and poor prognosis. Moreover, we found a disease stage-specific expression of CRYM in PCa. CRYM counteracted thyroid and androgen signaling and blocked intracellular choline uptake. CRYM inversely correlated with [18F]fluoromethylcholine (FMC) levels in positron emission tomography/magnetic resonance imaging of PCa patients. Our data suggest CRYM as a novel antagonist of T3- and androgen-mediated signaling in PCa. The role of CRYM could therefore be an essential control mechanism for the prevention of aggressive PCa growth.
Collapse
Affiliation(s)
- Osman Aksoy
- Department of PathologyMedical University ViennaViennaAustria
| | - Jan Pencik
- Department of PathologyMedical University ViennaViennaAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
- Present address: Jan Pencik, Molecular and Cell Biology LaboratoryThe Salk Institute for Biological StudiesLa JollaCaliforniaUSA
| | - Markus Hartenbach
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
| | - Ali A. Moazzami
- Department of Molecular Sciences, Uppsala BioCenterSwedish University of Agricultural SciencesUppsalaSweden
| | | | - Theresa Balber
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
- Ludwig Boltzmann Institute Applied DiagnosticsViennaAustria
- Department for Pharmaceutical Technology and BiopharmaceuticsUniversity of ViennaViennaAustria
| | - Adam Varady
- Department of PathologyMedical University ViennaViennaAustria
| | - Cecile Philippe
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
| | - Pascal A. Baltzer
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
| | | | | | | | - Andrea Haitel
- Department of PathologyMedical University ViennaViennaAustria
| | - Merima Herac
- Department of PathologyMedical University ViennaViennaAustria
| | - Martin Susani
- Department of PathologyMedical University ViennaViennaAustria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
- Ludwig Boltzmann Institute Applied DiagnosticsViennaAustria
| | - Rodrig Marculescu
- Department of Laboratory MedicineMedical University ViennaViennaAustria
| | | | | | - Gero Kramer
- Department of UrologyMedical University ViennaViennaAustria
| | - Shahrokh F. Shariat
- Department of UrologyMedical University ViennaViennaAustria
- Division of Urology, Department of Special SurgeryJordan University Hospital, The University of JordanAmmanJordan
- Institute for Urology and Reproductive HealthSechenov UniversityMoscowRussia
- Departments of UrologyWeill Cornell Medical CollegeNew YorkNew YorkUSA
- Department of UrologyUniversity of Texas SouthwesternDallasTexasUSA
- Department of Urology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Suzanne D. Turner
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of CambridgeCambridgeUK
- Center of Molecular Medicine, Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Boris Tichy
- Center of Molecular Medicine, Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Jan Oppelt
- Center of Molecular Medicine, Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Sarka Pospisilova
- Center of Molecular Medicine, Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Sabrina Hartenbach
- Histo Consulting Inc.UlmGermany
- Department of PathologyRudolfinerhaus Privatklinik GmbhViennaAustria
| | - Simone Tangermann
- Unit for Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
| | - Gerda Egger
- Department of PathologyMedical University ViennaViennaAustria
- Ludwig Boltzmann Institute Applied DiagnosticsViennaAustria
| | - Heidi A. Neubauer
- Institute of Animal Breeding and GeneticsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Richard Moriggl
- Institute of Animal Breeding and GeneticsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Zoran Culig
- Department of UrologyInnsbruck Medical UniversityInnsbruckAustria
| | - Georg Greiner
- Department of Laboratory MedicineMedical University ViennaViennaAustria
| | - Gregor Hoermann
- Department of Laboratory MedicineMedical University ViennaViennaAustria
- MLL Munich Leukemia LaboratoryMunichGermany
| | - Marcus Hacker
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
| | | | - Olaf Merkel
- Department of PathologyMedical University ViennaViennaAustria
| | - Lukas Kenner
- Department of PathologyMedical University ViennaViennaAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
- Unit for Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
- Christian Doppler Laboratory for Applied Metabolomics (CDL‐AM)Medical University of ViennaViennaAustria
| |
Collapse
|
17
|
Beaudoin JJ, Brouwer KLR, Malinen MM. Novel insights into the organic solute transporter alpha/beta, OSTα/β: From the bench to the bedside. Pharmacol Ther 2020; 211:107542. [PMID: 32247663 PMCID: PMC7480074 DOI: 10.1016/j.pharmthera.2020.107542] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022]
Abstract
Organic solute transporter alpha/beta (OSTα/β) is a heteromeric solute carrier protein that transports bile acids, steroid metabolites and drugs into and out of cells. OSTα/β protein is expressed in various tissues, but its expression is highest in the gastrointestinal tract where it facilitates the recirculation of bile acids from the gut to the liver. Previous studies established that OSTα/β is upregulated in liver tissue of patients with extrahepatic cholestasis, obstructive cholestasis, and primary biliary cholangitis (PBC), conditions that are characterized by elevated bile acid concentrations in the liver and/or systemic circulation. The discovery that OSTα/β is highly upregulated in the liver of patients with nonalcoholic steatohepatitis (NASH) further highlights the clinical relevance of this transporter because the incidence of NASH is increasing at an alarming rate with the obesity epidemic. Since OSTα/β is closely linked to the homeostasis of bile acids, and tightly regulated by the nuclear receptor farnesoid X receptor, OSTα/β is a potential drug target for treatment of cholestatic liver disease, and other bile acid-related metabolic disorders such as obesity and diabetes. Obeticholic acid, a semi-synthetic bile acid used to treat PBC, under review for the treatment of NASH, and in development for the treatment of other metabolic disorders, induces OSTα/β. Some drugs associated with hepatotoxicity inhibit OSTα/β, suggesting a possible role for OSTα/β in drug-induced liver injury (DILI). Furthermore, clinical cases of homozygous genetic defects in both OSTα/β subunits resulting in diarrhea and features of cholestasis have been reported. This review article has been compiled to comprehensively summarize the recent data emerging on OSTα/β, recapitulating the available literature on the structure-function and expression-function relationships of OSTα/β, the regulation of this important transporter, the interaction of drugs and other compounds with OSTα/β, and the comparison of OSTα/β with other solute carrier transporters as well as adenosine triphosphate-binding cassette transporters. Findings from basic to more clinically focused research efforts are described and discussed.
Collapse
Affiliation(s)
- James J Beaudoin
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - 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, USA.
| | - Melina M Malinen
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
18
|
Groeneweg S, van Geest FS, Peeters RP, Heuer H, Visser WE. Thyroid Hormone Transporters. Endocr Rev 2020; 41:5637505. [PMID: 31754699 DOI: 10.1210/endrev/bnz008] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ferdy S van Geest
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - W Edward Visser
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
19
|
Lu X, Liu L, Shan W, Kong L, Chen N, Lou Y, Zeng S. The Role of the Sodium-taurocholate Co-transporting Polypeptide (NTCP) and Bile Salt Export Pump (BSEP) in Related Liver Disease. Curr Drug Metab 2019; 20:377-389. [PMID: 31258056 DOI: 10.2174/1389200220666190426152830] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/10/2019] [Accepted: 03/26/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Sodium Taurocholate Co-transporting Polypeptide (NTCP) and Bile Salt Export Pump (BSEP) play significant roles as membrane transporters because of their presence in the enterohepatic circulation of bile salts. They have emerged as promising drug targets in related liver disease. METHODS We reviewed the literature published over the last 20 years with a focus on NTCP and BSEP. RESULTS This review summarizes the current perception about structure, function, genetic variation, and regulation of NTCP and BSEP, highlights the effects of their defects in some hepatic disorders, and discusses the application prospect of new transcriptional activators in liver diseases. CONCLUSION NTCP and BSEP are important proteins for transportation and homeostasis maintenance of bile acids. Further research is needed to develop new models for determining the structure-function relationship of bile acid transporters and screening for substrates and inhibitors, as well as to gain more information about the regulatory genetic mechanisms involved in the processes of liver injury.
Collapse
Affiliation(s)
- Xiaoyang Lu
- The First Affiliated Hospital, Zhejiang University, Zhejiang, China
| | - Lin Liu
- The First Affiliated Hospital, Zhejiang University, Zhejiang, China
| | - Wenya Shan
- The First Affiliated Hospital, Zhejiang University, Zhejiang, China
| | - Limin Kong
- The First Affiliated Hospital, Zhejiang University, Zhejiang, China
| | - Na Chen
- The First Affiliated Hospital, Zhejiang University, Zhejiang, China
| | - Yan Lou
- The First Affiliated Hospital, Zhejiang University, Zhejiang, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, China
| |
Collapse
|
20
|
Differential expression and immunoreactivity of thyroid hormone transporters MCT8 and OATP1C1 in rat ovary. Acta Histochem 2019; 121:151440. [PMID: 31561916 DOI: 10.1016/j.acthis.2019.151440] [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: 04/12/2019] [Revised: 08/02/2019] [Accepted: 09/03/2019] [Indexed: 11/20/2022]
Abstract
Thyroid hormones (THs) regulate several physiological processes in female mammals, many of which are related to reproduction such as steroidogenesis in the ovary, oocyte and granulosa cells maturation, follicular development and differentiation, and ovulation. THs actions require the presence of THs transporters to facilitate their cellular uptake and efflux. MCT8 and OATP1C1 are the principal THs transporters. The aim of the present study was to determine the gene expression and cellular localization of MCT8 and OATP1C1 in the rat ovary during the diestrus-II cycle phase. Ovaries of virgin adult rats were histologically processed. Reverse Transcription-PCR and immunohistochemistry analyses for MCT8 and OATP1C1 were done. MCT8 gene expression level was significantly higher (P ≤ 0.01) than that of OATP1C1 in the rat ovary. MCT8 and OATP1C1 were found in all types of ovarian cells but with different immunoreactivity. MCT8 showed stronger immunoreactivity in tertiary and Graafian follicles, corpus luteum and blood vessels, whereas OATP1C1's immunoreactivity was stronger in stroma cells, tunica albuginea, and blood vessels. Our results provide evidence that THs and their transporters are both necessary for ovarian function and that any alteration in these transporters could interfere with reproductive processes such as ovulation and steroidogenesis, compromising fertility.
Collapse
|
21
|
Zhang Y, Hagenbuch B. Protein-protein interactions of drug uptake transporters that are important for liver and kidney. Biochem Pharmacol 2019; 168:384-391. [PMID: 31381872 DOI: 10.1016/j.bcp.2019.07.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/31/2019] [Indexed: 12/28/2022]
Abstract
Drug uptake transporters are membrane proteins responsible for the trans-membrane transport of endo- and xenobiotics, including numerous drugs. They are important for the uptake of drugs into target tissues or into organs for metabolism and excretion. Many drug uptake transporters have a broad spectrum of structural-independent substrates, which make them vulnerable to drug-drug interactions. Recent studies have shown more and more complex pharmacokinetics involving transporters, and regulatory agencies now require studies to be performed to measure the involvement of transporters in drug development. A better understanding of the factors affecting the expression of transporters is needed. Despite many efforts devoted to the functional characterization of different drug uptake transporters, transporter in vitro to in vivo extrapolations are far from predicting the behavior under physiological conditions. There is an increasing number of uptake transporters demonstrated to form protein-protein interactions or to oligomerize. This raises the possibility that these interactions between or among transporters could help explaining the gap between in vitro and in vivo measurement of drug transporters. In this review, we summarized protein-protein interactions of drug uptake transporters that are important for pharmacokinetics, especially those in the liver and the kidneys.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States.
| |
Collapse
|
22
|
Rodríguez-Rodríguez A, Lazcano I, Sánchez-Jaramillo E, Uribe RM, Jaimes-Hoy L, Joseph-Bravo P, Charli JL. Tanycytes and the Control of Thyrotropin-Releasing Hormone Flux Into Portal Capillaries. Front Endocrinol (Lausanne) 2019; 10:401. [PMID: 31293518 PMCID: PMC6603095 DOI: 10.3389/fendo.2019.00401] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/06/2019] [Indexed: 12/17/2022] Open
Abstract
Central and peripheral mechanisms that modulate energy intake, partition and expenditure determine energy homeostasis. Thyroid hormones (TH) regulate energy expenditure through the control of basal metabolic rate and thermogenesis; they also modulate food intake. TH concentrations are regulated by the hypothalamus-pituitary-thyroid (HPT) axis, and by transport and metabolism in blood and target tissues. In mammals, hypophysiotropic thyrotropin-releasing hormone (TRH) neurons of the paraventricular nucleus of the hypothalamus integrate energy-related information. They project to the external zone of the median eminence (ME), a brain circumventricular organ rich in neuron terminal varicosities and buttons, tanycytes, other glial cells and capillaries. These capillary vessels form a portal system that links the base of the hypothalamus with the anterior pituitary. Tanycytes of the medio-basal hypothalamus express a repertoire of proteins involved in transport, sensing, and metabolism of TH; among them is type 2 deiodinase, a source of 3,3',5-triiodo-L-thyronine necessary for negative feedback on TRH neurons. Tanycytes subtypes are distinguished by position and phenotype. The end-feet of β2-tanycytes intermingle with TRH varicosities and terminals in the external layer of the ME and terminate close to the ME capillaries. Besides type 2 deiodinase, β2-tanycytes express the TRH-degrading ectoenzyme (TRH-DE); this enzyme likely controls the amount of TRH entering portal vessels. TRH-DE is rapidly upregulated by TH, contributing to TH negative feedback on HPT axis. Alterations in energy balance also regulate the expression and activity of TRH-DE in the ME, making β2-tanycytes a hub for energy-related regulation of HPT axis activity. β2-tanycytes also express TRH-R1, which mediates positive effects of TRH on TRH-DE activity and the size of β2-tanycyte end-feet contacts with the basal lamina adjacent to ME capillaries. These end-feet associations with ME capillaries, and TRH-DE activity, appear to coordinately control HPT axis activity. Thus, down-stream of neuronal control of TRH release by action potentials arrival in the external layer of the median eminence, imbricated intercellular processes may coordinate the flux of TRH into the portal capillaries. In conclusion, β2-tanycytes appear as a critical cellular element for the somatic and post-secretory control of TRH flux into portal vessels, and HPT axis regulation in mammals.
Collapse
Affiliation(s)
- Adair Rodríguez-Rodríguez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Iván Lazcano
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | - Edith Sánchez-Jaramillo
- Laboratorio de Neuroendocrinología Molecular, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Rosa María Uribe
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Lorraine Jaimes-Hoy
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Patricia Joseph-Bravo
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Jean-Louis Charli
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| |
Collapse
|
23
|
Kersseboom S, van Gucht ALM, van Mullem A, Brigante G, Farina S, Carlsson B, Donkers JM, van de Graaf SFJ, Peeters RP, Visser TJ. Role of the Bile Acid Transporter SLC10A1 in Liver Targeting of the Lipid-Lowering Thyroid Hormone Analog Eprotirome. Endocrinology 2017; 158:3307-3318. [PMID: 28938430 DOI: 10.1210/en.2017-00433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/14/2017] [Indexed: 12/20/2022]
Abstract
The thyroid hormone (TH) analog eprotirome (KB2115) was developed to lower cholesterol through selective activation of the TH receptor (TR) β1 in the liver. Interestingly, eprotirome shows low uptake in nonhepatic tissues, explaining its lipid-lowering action without adverse extrahepatic thyromimetic effects. Clinical trials have shown marked decreases in serum cholesterol levels. We explored the transport of eprotirome across the plasma membrane by members of three TH transporter families: monocarboxylate transporters MCT8 and MCT10; Na-independent organic anion transporters 1A2, 1B1, 1B3, 1C1, 2A1, and 2B1; and Na-dependent organic anion transporters SLC10A1 to SLC10A7. Cellular transport was studied in transfected COS1 cells using [14C]eprotirome and [125I]TH analogs. Of the 15 transporters tested initially, the liver-specific bile acid transporter SLC10A1 showed the highest eprotirome uptake (greater than a sevenfold induction after 60 minutes) as well as TRβ1-mediated transcriptional activity. Uptake of eprotirome by SLC10A1 was Na+ dependent and saturable with a Michaelis constant of 8 μM. Eprotirome transport was inhibited by known substrates for SLC10A1 (e.g., cholate and taurocholate), and by TH analogs such as triiodothyropropionic acid and triiodothyroacetic acid. However, no significant SLC10A1-mediated transport was observed of these [125I]TH analogs. We also studied the plasma disappearance and biliary excretion of [14C]eprotirome injected in control and Slc10a1 knockout mice. Although eprotirome is also transported by mouse Slc10a1, the pharmacokinetics of eprotirome were not affected by Slc10a1 deficiency. In conclusion, we have demonstrated that the liver-specific bile acid transporter SLC10A1 effectively transports eprotirome. However, Slc10a1 does not appear to be critical for the liver targeting of this TH analog in mice. Therefore, the importance of SLC10A1 for liver uptake of eprotirome in humans remains to be elucidated.
Collapse
Affiliation(s)
- Simone Kersseboom
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Anja L M van Gucht
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Alies van Mullem
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Giulia Brigante
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Stefania Farina
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Bo Carlsson
- Karo Bio AB, Novum Research Park, Huddinge S-141 57, Sweden
| | - Joanne M Donkers
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Amsterdam Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Amsterdam Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Robin P Peeters
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Theo J Visser
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| |
Collapse
|
24
|
王 鹤, 孙 鹏, 刘 克. 肝脏转运体表达和功能的变化对肝疾病的影响. Shijie Huaren Xiaohua Zazhi 2017; 25:1427-1437. [DOI: 10.11569/wcjd.v25.i16.1427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
转运体是药物吸收、分布、代谢和排泄的重要决定因素, 在肝脏表达尤为广泛. 肝脏转运体可以摄取大多数内源性物质、营养物质和外源性物质进入肝脏, 在肝脏内经过一系列的代谢转化, 最终将其外排入胆汁, 并由胆汁排到肝外. 越来越多的证据表明, 肝脏疾病状态下转运体的表达和功能会发生改变, 影响药物在体内的处置过程, 进而增加药物相互作用的可能性, 同时加大了疾病药物治疗的难度. 本文从肝脏摄取型和外排型转运体两方面出发, 针对肝脏转运体表达和功能的变化对肝疾病的影响作一综述.
Collapse
|
25
|
Pettersson H, Zarnegar B, Westin A, Persson V, Peuckert C, Jonsson J, Hallgren J, Kullander K. SLC10A4 regulates IgE-mediated mast cell degranulation in vitro and mast cell-mediated reactions in vivo. Sci Rep 2017; 7:1085. [PMID: 28439090 PMCID: PMC5430724 DOI: 10.1038/s41598-017-01121-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/22/2017] [Indexed: 11/24/2022] Open
Abstract
Mast cells act as sensors in innate immunity and as effector cells in adaptive immune reactions. Here we demonstrate that SLC10A4, also referred to as the vesicular aminergic-associated transporter, VAAT, modifies mast cell degranulation. Strikingly, Slc10a4 -/- bone marrow-derived mast cells (BMMCs) had a significant reduction in the release of granule-associated mediators in response to IgE/antigen-mediated activation, whereas the in vitro development of mast cells, the storage of the granule-associated enzyme mouse mast cell protease 6 (mMCP-6), and the release of prostaglandin D2 and IL-6 were normal. Slc10a4-deficient mice had a strongly reduced passive cutaneous anaphylaxis reaction and a less intense itching behaviour in response to the mast cell degranulator 48/80. Live imaging of the IgE/antigen-mediated activation showed decreased degranulation and that ATP was retained to a higher degree in mast cell granules lacking SLC10A4. Furthermore, ATP was reduced by two thirds in Slc10a4 -/- BMMCs supernatants in response to IgE/antigen. We speculate that SLC10A4 affects the amount of granule-associated ATP upon IgE/antigen-induced mast cell activation, which affect the release of granule-associated mast cell mediators. In summary, SLC10A4 acts as a regulator of degranulation in vitro and of mast cell-related reactions in vivo.
Collapse
Affiliation(s)
- Hanna Pettersson
- Department of Neuroscience, Uppsala University, Box 593, 751 24, Uppsala, Sweden
- Department of Organismal Biology, Uppsala University, Norbyv, 18A, 752 36, Uppsala, Sweden
| | - Behdad Zarnegar
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, 751 23, Uppsala, Sweden
| | - Annika Westin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, 751 23, Uppsala, Sweden
| | - Viktor Persson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, 751 23, Uppsala, Sweden
| | - Christiane Peuckert
- Department of Neuroscience, Uppsala University, Box 593, 751 24, Uppsala, Sweden
| | - Jörgen Jonsson
- Department of Neuroscience, Uppsala University, Box 593, 751 24, Uppsala, Sweden
| | - Jenny Hallgren
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, 751 23, Uppsala, Sweden.
| | - Klas Kullander
- Department of Neuroscience, Uppsala University, Box 593, 751 24, Uppsala, Sweden.
| |
Collapse
|
26
|
Abstract
Bile acids are potent signaling molecules that regulate glucose, lipid and energy homeostasis predominantly via the bile acid receptors farnesoid X receptor (FXR) and transmembrane G protein-coupled receptor 5 (TGR5). The sodium taurocholate cotransporting polypeptide (NTCP) and the apical sodium dependent bile acid transporter (ASBT) ensure an effective circulation of (conjugated) bile acids. The modulation of these transport proteins affects bile acid localization, dynamics and signaling. The NTCP-specific pharmacological inhibitor myrcludex B inhibits hepatic uptake of conjugated bile acids. Multiple ASBT-inhibitors are already in clinical trials to inhibit intestinal bile acid uptake. Here, we discuss current insights into the consequences of targeting bile acid uptake transporters on systemic and intestinal bile acid dynamics and discuss the possible therapeutic applications that evolve as a result.
Collapse
Affiliation(s)
- Davor Slijepcevic
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Stan F.J. van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands,Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands,*Stan F.J. van de Graaf, Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, NL-1105 BK Amsterdam (The Netherlands), E-Mail
| |
Collapse
|
27
|
The thyroid hormone nuclear receptors and the Wnt/β-catenin pathway: An intriguing liaison. Dev Biol 2017; 422:71-82. [DOI: 10.1016/j.ydbio.2017.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/26/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022]
|
28
|
Yu D, Zhang H, Lionarons DA, Boyer JL, Cai SY. Na +-taurocholate cotransporting polypeptide (NTCP/SLC10A1) ortholog in the marine skate Leucoraja erinacea is not a physiological bile salt transporter. Am J Physiol Regul Integr Comp Physiol 2017; 312:R477-R484. [PMID: 28077388 DOI: 10.1152/ajpregu.00302.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 01/05/2023]
Abstract
The Na+-dependent taurocholate cotransporting polypeptide (NTCP/SLC10A1) is a hepatocyte-specific solute carrier, which plays an important role in maintaining bile salt homeostasis in mammals. The absence of a hepatic Na+-dependent bile salt transport system in marine skate and rainbow trout raises a question regarding the function of the Slc10a1 gene in these species. Here, we have characterized the Slc10a1 gene in the marine skate, Leucoraja erinacea The transcript of skate Slc10a1 (skSlc10a1) encodes 319 amino acids and shares 46% identity to human NTCP (hNTCP) with similar topology to mammalian NTCP. SkSlc10a1 mRNA was mostly confined to the brain and testes with minimal expression in the liver. An FXR-bile salt reporter assay indicated that skSlc10a1 transported taurocholic acid (TCA) and scymnol sulfate, but not as effectively as hNTCP. An [3H]TCA uptake assay revealed that skSlc10a1 functioned as a Na+-dependent transporter, but with low affinity for TCA (Km = 92.4 µM) and scymnol sulfate (Ki = 31 µM), compared with hNTCP (TCA, Km = 5.4 µM; Scymnol sulfate, Ki = 3.5 µM). In contrast, the bile salt concentration in skate plasma was 2 µM, similar to levels seen in mammals. Interestingly, skSlc10a1 demonstrated transport activity for the neurosteroids dehydroepiandrosterone sulfate and estrone-3-sulfate at physiological concentration, similar to hNTCP. Together, our findings indicate that skSlc10a1 is not a physiological bile salt transporter, providing a molecular explanation for the absence of a hepatic Na+-dependent bile salt uptake system in skate. We speculate that Slc10a1 is a neurosteroid transporter in skate that gained its substrate specificity for bile salts later in vertebrate evolution.
Collapse
Affiliation(s)
- Dongke Yu
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Han Zhang
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Daniel A Lionarons
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - James L Boyer
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, New Haven, Connecticut; and.,Mount Desert Island Biological Laboratory, Salisbury Cove, Maine
| | - Shi-Ying Cai
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, New Haven, Connecticut; and
| |
Collapse
|
29
|
Chang SY, Weber EJ, Ness KV, Eaton DL, Kelly EJ. Liver and Kidney on Chips: Microphysiological Models to Understand Transporter Function. Clin Pharmacol Ther 2016; 100:464-478. [PMID: 27448090 DOI: 10.1002/cpt.436] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/13/2016] [Accepted: 07/15/2016] [Indexed: 12/19/2022]
Abstract
Because of complex cellular microenvironments of both the liver and kidneys, accurate modeling of transport function has remained a challenge, leaving a dire need for models that can faithfully recapitulate both the architecture and cell-cell interactions observed in vivo. The study of hepatic and renal transport function is a fundamental component of understanding the metabolic fate of drugs and xenobiotics; however, there are few in vitro systems conducive for these types of studies. For both the hepatic and renal systems, we provide an overview of the location and function of the most significant phase I/II/III (transporter) of enzymes, and then review current in vitro systems for the suitability of a transporter function study and provide details on microphysiological systems that lead the field in these investigations. Microphysiological modeling of the liver and kidneys using "organ-on-a-chip" technologies is rapidly advancing in transport function assessment and has emerged as a promising method to evaluate drug and xenobiotic metabolism. Future directions for the field are also discussed along with technical challenges encountered in complex multiple-organs-on-chips development.
Collapse
Affiliation(s)
- S Y Chang
- Department of Occupational and Environmental Health Sciences, University of Washington, Seattle, Washington, USA
| | - E J Weber
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Kp Van Ness
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - D L Eaton
- Department of Occupational and Environmental Health Sciences, University of Washington, Seattle, Washington, USA
| | - E J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
30
|
Abstract
The cellular influx and efflux of thyroid hormones are facilitated by transmembrane protein transporters. Of these transporters, monocarboxylate transporter 8 (MCT8) is the only one specific for the transport of thyroid hormones and some of their derivatives. Mutations in SLC16A2, the gene that encodes MCT8, lead to an X-linked syndrome with severe neurological impairment and altered concentrations of thyroid hormones. Histopathological analysis of brain tissue from patients who have impaired MCT8 function indicates that brain lesions start prenatally, and are most probably the result of cerebral hypothyroidism. A Slc16a2 knockout mouse model has revealed that Mct8 is an important mediator of thyroid hormone transport, especially T3, through the blood-brain barrier. However, unlike humans with an MCT8 deficiency, these mice do not have neurological impairment. One explanation for this discrepancy could be differences in expression of the T4 transporter OATP1C1 in the blood-brain barrier; OATP1C1 is more abundant in rodents than in primates and permits the passage of T4 in the absence of T3 transport, thus preventing full cerebral hypothyroidism. In this Review, we discuss the relevance of thyroid hormone transporters in health and disease, with a particular focus on the pathophysiology of MCT8 mutations.
Collapse
Affiliation(s)
- Juan Bernal
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Arturo Duperier 4, 28029 Madrid, Spain
| | - Ana Guadaño-Ferraz
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Arturo Duperier 4, 28029 Madrid, Spain
| | - Beatriz Morte
- Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Arturo Duperier 4, 28029 Madrid, Spain
| |
Collapse
|
31
|
Slijepcevic D, Kaufman C, Wichers CGK, Gilglioni EH, Lempp FA, Duijst S, de Waart DR, Oude Elferink RPJ, Mier W, Stieger B, Beuers U, Urban S, van de Graaf SFJ. Impaired uptake of conjugated bile acids and hepatitis b virus pres1-binding in na(+) -taurocholate cotransporting polypeptide knockout mice. Hepatology 2015; 62:207-19. [PMID: 25641256 PMCID: PMC4657468 DOI: 10.1002/hep.27694] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 01/07/2015] [Indexed: 12/14/2022]
Abstract
UNLABELLED The Na(+) -taurocholate cotransporting polypeptide (NTCP) mediates uptake of conjugated bile acids (BAs) and is localized at the basolateral membrane of hepatocytes. It has recently been recognized as the receptor mediating hepatocyte-specific entry of hepatitis B virus and hepatitis delta virus. Myrcludex B, a peptide inhibitor of hepatitis B virus entry, is assumed to specifically target NTCP. Here, we investigated BA transport and Myrcludex B binding in the first Slc10a1-knockout mouse model (Slc10a1 encodes NTCP). Primary Slc10a1(-/-) hepatocytes showed absence of sodium-dependent taurocholic acid uptake, whereas sodium-independent taurocholic acid uptake was unchanged. In vivo, this was manifested as a decreased serum BA clearance in all knockout mice. In a subset of mice, NTCP deficiency resulted in markedly elevated total serum BA concentrations, mainly composed of conjugated BAs. The hypercholanemic phenotype was rapidly triggered by a diet supplemented with ursodeoxycholic acid. Biliary BA output remained intact, while fecal BA excretion was reduced in hypercholanemic Slc10a1(-/-) mice, explained by increased Asbt and Ostα/β expression. These mice further showed reduced Asbt expression in the kidney and increased renal BA excretion. Hepatic uptake of conjugated BAs was potentially affected by down-regulation of OATP1A1 and up-regulation of OATP1A4. Furthermore, sodium-dependent taurocholic acid uptake was inhibited by Myrcludex B in wild-type hepatocytes, while Slc10a1(-/-) hepatocytes were insensitive to Myrcludex B. Finally, positron emission tomography showed a complete abrogation of hepatic binding of labeled Myrcludex B in Slc10a1(-/-) mice. CONCLUSION The Slc10a1-knockout mouse model supports the central role of NTCP in hepatic uptake of conjugated BAs and hepatitis B virus preS1/Myrcludex B binding in vivo; the NTCP-independent hepatic BA uptake machinery maintains a (slower) enterohepatic circulation of BAs, although it is occasionally insufficient to clear BAs from the circulation.
Collapse
Affiliation(s)
- Davor Slijepcevic
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, AMCAmsterdam, The Netherlands
| | - Christina Kaufman
- Department of Infectious Diseases and of Molecular Virology, University Hospital HeidelbergHeidelberg, Germany,Department of Nuclear Medicine, University Hospital HeidelbergHeidelberg, Germany
| | - Catharina GK Wichers
- Department of Molecular Cancer Research, Section of Metabolic Diseases, University Medical Center UtrechtUtrecht, The Netherlands
| | - Eduardo H Gilglioni
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, AMCAmsterdam, The Netherlands
| | - Florian A Lempp
- Department of Infectious Diseases and of Molecular Virology, University Hospital HeidelbergHeidelberg, Germany
| | - Suzanne Duijst
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, AMCAmsterdam, The Netherlands
| | - Dirk R de Waart
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, AMCAmsterdam, The Netherlands
| | - Ronald PJ Oude Elferink
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, AMCAmsterdam, The Netherlands
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital HeidelbergHeidelberg, Germany
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital ZurichZurich, Switzerland
| | - Ulrich Beuers
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, AMCAmsterdam, The Netherlands
| | - Stephan Urban
- Department of Infectious Diseases and of Molecular Virology, University Hospital HeidelbergHeidelberg, Germany,German Center for Infection Research, Heidelberg UniversityHeidelberg, Germany
| | - Stan FJ van de Graaf
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, AMCAmsterdam, The Netherlands
| |
Collapse
|
32
|
Schmidt S, Moncada M, Burger S, Geyer J. Expression, sorting and transport studies for the orphan carrier SLC10A4 in neuronal and non-neuronal cell lines and in Xenopus laevis oocytes. BMC Neurosci 2015; 16:35. [PMID: 26084360 PMCID: PMC4472396 DOI: 10.1186/s12868-015-0174-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 06/05/2015] [Indexed: 11/10/2022] Open
Abstract
Background SLC10A4 belongs to the solute carrier family SLC10 whose founding members are the Na+/taurocholate co-transporting polypeptide (NTCP, SLC10A1) and the apical sodium-dependent bile acid transporter (ASBT, SLC10A2). These carriers maintain the enterohepatic circulation of bile acids between the liver and the gut. SLC10A4 was identified as a novel member of the SLC10 carrier family with the highest phylogenetic relationship to NTCP. The SLC10A4 protein was detected in synaptic vesicles of cholinergic and monoaminergic neurons of the peripheral and central nervous system, suggesting a transport function for any kind of neurotransmitter. Therefore, in the present study, we performed systematic transport screenings for SLC10A4 and also aimed to identify the vesicular sorting domain of the SLC10A4 protein. Results We detected a vesicle-like expression pattern of the SLC10A4 protein in the neuronal cell lines SH-SY5Y and CAD. Differentiation of these cells to the neuronal phenotype altered neither SLC10A4 gene expression nor its vesicular expression pattern. Functional transport studies with different neurotransmitters, bile acids and steroid sulfates were performed in SLC10A4-transfected HEK293 cells, SLC10A4-transfected CAD cells and in Xenopus laevis oocytes. For these studies, transport by the dopamine transporter DAT, the serotonin transporter SERT, the choline transporter CHT1, the vesicular monoamine transporter VMAT2, the organic cation transporter Oct1, and NTCP were used as positive control. SLC10A4 failed to show transport activity for dopamine, serotonin, norepinephrine, histamine, acetylcholine, choline, acetate, aspartate, glutamate, gamma-aminobutyric acid, pregnenolone sulfate, dehydroepiandrosterone sulfate, estrone-3-sulfate, and adenosine triphosphate, at least in the transport assays used. When the C-terminus of SLC10A4 was replaced by the homologous sequence of NTCP, the SLC10A4-NTCP chimeric protein revealed clear plasma membrane expression in CAD and HEK293 cells. But this chimera also did not show any transport activity, even when the N-terminal domain of SLC10A4 was deleted by mutagenesis. Conclusions Although different kinds of assays were used to screen for transport function, SLC10A4 failed to show transport activity for a series of neurotransmitters and neuromodulators, indicating that SLC10A4 does not seem to represent a typical neurotransmitter transporter such as DAT, SERT, CHT1 or VMAT2.
Collapse
Affiliation(s)
- Stephanie Schmidt
- Institute of Pharmacology and Toxicology, Justus Liebig University of Giessen, Schubertstr. 81, 35392, Giessen, Germany.
| | - Marcela Moncada
- Institute of Pharmacology and Toxicology, Justus Liebig University of Giessen, Schubertstr. 81, 35392, Giessen, Germany.
| | - Simone Burger
- Institute of Pharmacology and Toxicology, Justus Liebig University of Giessen, Schubertstr. 81, 35392, Giessen, Germany.
| | - Joachim Geyer
- Institute of Pharmacology and Toxicology, Justus Liebig University of Giessen, Schubertstr. 81, 35392, Giessen, Germany.
| |
Collapse
|
33
|
Abstract
Thyroid hormones have an intimate relationship with cardiac function. Some of the most significant clinical signs and symptoms of thyroid disease are the cardiac manifestations. In both hypothyroidism and hyperthyroidism, the characteristic physiological effects of thyroid hormone can be understood from the actions at the molecular and cellular level. Here we explore topics from the metabolism and cellular effects of thyroid hormone to special considerations related to statin and amiodarone therapy for the alterations in thyroid hormone metabolism that accompany heart disease.
Collapse
|
34
|
Medici M, Visser WE, Visser TJ, Peeters RP. Genetic determination of the hypothalamic-pituitary-thyroid axis: where do we stand? Endocr Rev 2015; 36:214-44. [PMID: 25751422 DOI: 10.1210/er.2014-1081] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
For a long time it has been known that both hypo- and hyperthyroidism are associated with an increased risk of morbidity and mortality. In recent years, it has also become clear that minor variations in thyroid function, including subclinical dysfunction and variation in thyroid function within the reference range, can have important effects on clinical endpoints, such as bone mineral density, depression, metabolic syndrome, and cardiovascular mortality. Serum thyroid parameters show substantial interindividual variability, whereas the intraindividual variability lies within a narrow range. This suggests that every individual has a unique hypothalamus-pituitary-thyroid axis setpoint that is mainly determined by genetic factors, and this heritability has been estimated to be 40-60%. Various mutations in thyroid hormone pathway genes have been identified in persons with thyroid dysfunction or altered thyroid function tests. Because these causes are rare, many candidate gene and linkage studies have been performed over the years to identify more common variants (polymorphisms) associated with thyroid (dys)function, but only a limited number of consistent associations have been found. However, in the past 5 years, advances in genetic research have led to the identification of a large number of new candidate genes. In this review, we provide an overview of the current knowledge about the polygenic basis of thyroid (dys)function. This includes new candidate genes identified by genome-wide approaches, what insights these genes provide into the genetic basis of thyroid (dys)function, and which new techniques will help to further decipher the genetic basis of thyroid (dys)function in the near future.
Collapse
Affiliation(s)
- Marco Medici
- Rotterdam Thyroid Center, Department of Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
35
|
Marada VVVR, Flörl S, Kühne A, Müller J, Burckhardt G, Hagos Y. Interaction of human organic anion transporter 2 (OAT2) and sodium taurocholate cotransporting polypeptide (NTCP) with antineoplastic drugs. Pharmacol Res 2014; 91:78-87. [PMID: 25481222 DOI: 10.1016/j.phrs.2014.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/23/2014] [Accepted: 11/13/2014] [Indexed: 02/07/2023]
Abstract
The ability of an antineoplastic drug to exert its cytostatic effect depends largely on the balance between its uptake into and extrusion from the cancer cells. ATP driven efflux transporter proteins drive the export of antineoplastic drugs and play a pivotal role in the development of chemoresistance. As regards uptake transporters, comparably less is known on their impact in drug action. In the current study, we characterized the interactions of two uptake transporter proteins, expressed mainly in the liver; the organic anion transporter 2 (OAT2, encoded by the SLC22A7 gene) and the sodium taurocholate cotransporting polypeptide (NTCP, encoded by the SLC10A1 gene), stably transfected in human embryonic kidney cells, with some antineoplastic agents that are routinely being used in cancer chemotherapy. Whereas NTCP did not show any strong interactions with the cytostatics tested, we observed a very strong inhibition of OAT2 mediated [(3)H] cGMP uptake in the presence of bendamustine, irinotecan and paclitaxel. The Ki values of OAT2 for bendamustine, irinotecan and paclitaxel were determined to be 43.3±4.33μM, 26.4±2.34μM and 10.4±0.45μM, respectively. Incubation of bendamustine with OAT2 expressing cells increased the caspase-3 activity, and this increase was inhibited by simultaneous incubation with bendamustine and probenecid, a well-known inhibitor of OATs, suggesting that bendamustine is a substrate of OAT2. A higher accumulation of irinotecan was observed in OAT2 expressing cells compared to control pcDNA cells by HPLC analysis of cell lysates. The accumulation was diminished in the presence of cGMP, the substrate we used to functionally characterize OAT2, suggesting specificity of this uptake and the fact that OAT2 mediates uptake of irinotecan.
Collapse
Affiliation(s)
- Venkata V V R Marada
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany.
| | - Saskia Flörl
- PortaCellTec biosciences GmbH, Humboldtallee 23, 37073 Goettingen, Germany.
| | - Annett Kühne
- PortaCellTec biosciences GmbH, Humboldtallee 23, 37073 Goettingen, Germany.
| | - Judith Müller
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany.
| | - Gerhard Burckhardt
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany.
| | - Yohannes Hagos
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany; PortaCellTec biosciences GmbH, Humboldtallee 23, 37073 Goettingen, Germany.
| |
Collapse
|
36
|
Dietrich CG, Geier A. Effect of drug transporter pharmacogenetics on cholestasis. Expert Opin Drug Metab Toxicol 2014; 10:1533-51. [PMID: 25260651 DOI: 10.1517/17425255.2014.963553] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION The liver is the central place for the metabolism of drugs and other xenobiotics. In the liver cell, oxidation and conjugation of compounds take place, and at the same time, bile formation helps in extrusion of these compounds via the biliary route. A large number of transporters are responsible for drug uptake into the liver cell and excretion into bile or efflux to the sinusoidal blood. AREAS COVERED Genetic variants of these transporters and their transactivators contribute to changes in drug handling and are also responsible for cholestatic syndromes of different severity. This review summarizes the current knowledge regarding the influence of these genetic changes. The review covers progressive hereditary cholestatic syndromes as well as recurrent or transient cholestatic syndromes such as drug-induced liver injury, intrahepatic cholestasis of pregnancy, and benign recurrent intrahepatic cholestasis. EXPERT OPINION Polymorphisms in transporter genes are frequent. For clinically relevant cholestatic syndromes, it often requires a combination of genetic variants or acquired triggers such as pregnancy or drug treatment. In combination with other pathogenetic aspects, genetic variants in drug transporters may contribute to our understanding of not only cholestatic diseases such as primary sclerosing cholangitis or primary biliary cirrhosis, but also the natural course of chronic liver disease in general.
Collapse
|
37
|
Borel F, Hachi I, Palencia A, Gaillard MC, Ferrer JL. Crystal structure of mouse mu-crystallin complexed with NADPH and the T3 thyroid hormone. FEBS J 2014; 281:1598-612. [PMID: 24467707 DOI: 10.1111/febs.12726] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 01/17/2014] [Accepted: 01/20/2014] [Indexed: 01/17/2023]
Abstract
UNLABELLED Mu-crystallin (CRYM), first described as a structural component of the eye lens in marsupials, has been characterized as an NADPH-dependent cytosolic T3 thyroid hormone (triiodothyronine) binding protein. More recently, CRYM has also been associated with ketimine reductase activity. Here, we report three crystal structures: mouse CRYM (mCRYM) in its apo form, in a form complexed with NADPH, and in a form with both NADPH and triiodothyronine bound. Comparison of the apo and NADPH forms reveals a rearrangement of the protein upon NADPH binding that reduces the degrees of freedom of several residues and traps the conformation of the binding pocket in a more T3 competent state. These findings are in agreement with the cooperative mechanism identified using isothermal titration calorimetry. Our structure with T3 reveals for the first time the location of the hormone binding site and shows its detailed interactions. T3 binding involves mainly hydrophobic interactions. Only five residues, either directly or through bridging water molecules, are hydrogen bonded to the hormone. Using in silico docking analysis, a series of ring-containing hydrophobic molecules were identified as potential mCRYM ligands, suggesting that the specificity for the recognition of the hydrophobic part of the hormone might be low. This is in agreement with the ketimine reductase activity that has been identified for ovine CRYM, as it demonstrates how a protein known as a thyroid hormone transporter can accommodate the ringed molecules required for its ketimine reductase activity. In the light of our results, a putative role of CRYM in thyroid hormone metabolism is also discussed. STRUCTURED DIGITAL ABSTRACT CRYM and CRYM bind by x-ray crystallography (View interaction).
Collapse
Affiliation(s)
- Franck Borel
- Institut de Biologie Structurale, Université de Grenoble Alpes, CEA, CNRS, France
| | | | | | | | | |
Collapse
|
38
|
Bianco AC, Anderson G, Forrest D, Galton VA, Gereben B, Kim BW, Kopp PA, Liao XH, Obregon MJ, Peeters RP, Refetoff S, Sharlin DS, Simonides WS, Weiss RE, Williams GR. American Thyroid Association Guide to investigating thyroid hormone economy and action in rodent and cell models. Thyroid 2014; 24:88-168. [PMID: 24001133 PMCID: PMC3887458 DOI: 10.1089/thy.2013.0109] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND An in-depth understanding of the fundamental principles that regulate thyroid hormone homeostasis is critical for the development of new diagnostic and treatment approaches for patients with thyroid disease. SUMMARY Important clinical practices in use today for the treatment of patients with hypothyroidism, hyperthyroidism, or thyroid cancer are the result of laboratory discoveries made by scientists investigating the most basic aspects of thyroid structure and molecular biology. In this document, a panel of experts commissioned by the American Thyroid Association makes a series of recommendations related to the study of thyroid hormone economy and action. These recommendations are intended to promote standardization of study design, which should in turn increase the comparability and reproducibility of experimental findings. CONCLUSIONS It is expected that adherence to these recommendations by investigators in the field will facilitate progress towards a better understanding of the thyroid gland and thyroid hormone dependent processes.
Collapse
Affiliation(s)
- Antonio C. Bianco
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, Florida
| | - Grant Anderson
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota Duluth, Duluth, Minnesota
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Valerie Anne Galton
- Department of Physiology and Neurobiology, Dartmouth Medical School, Lebanon, New Hampshire
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Brian W. Kim
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, Florida
| | - Peter A. Kopp
- Division of Endocrinology, Metabolism, and Molecular Medicine, and Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiao Hui Liao
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - Maria Jesus Obregon
- Institute of Biomedical Investigation (IIB), Spanish National Research Council (CSIC) and Autonomous University of Madrid, Madrid, Spain
| | - Robin P. Peeters
- Division of Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Samuel Refetoff
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - David S. Sharlin
- Department of Biological Sciences, Minnesota State University, Mankato, Minnesota
| | - Warner S. Simonides
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Roy E. Weiss
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - Graham R. Williams
- Department of Medicine, Imperial College London, Hammersmith Campus, London, United Kingdom
| |
Collapse
|
39
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: transporters. Br J Pharmacol 2013; 170:1706-96. [PMID: 24528242 PMCID: PMC3892292 DOI: 10.1111/bph.12450] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Transporters are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
40
|
Bi YA, Qiu X, Rotter CJ, Kimoto E, Piotrowski M, Varma MV, EI-Kattan AF, Lai Y. Quantitative assessment of the contribution of sodium-dependent taurocholate co-transporting polypeptide (NTCP) to the hepatic uptake of rosuvastatin, pitavastatin and fluvastatin. Biopharm Drug Dispos 2013; 34:452-61. [DOI: 10.1002/bdd.1861] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 08/20/2013] [Accepted: 08/27/2013] [Indexed: 11/05/2022]
Affiliation(s)
- Yi-an Bi
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Global Research and Development; Groton; Connecticut; USA
| | - Xi Qiu
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Global Research and Development; Groton; Connecticut; USA
| | - Charles J. Rotter
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Global Research and Development; Groton; Connecticut; USA
| | - Emi Kimoto
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Global Research and Development; Groton; Connecticut; USA
| | - Mary Piotrowski
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Global Research and Development; Groton; Connecticut; USA
| | - Manthena V. Varma
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Global Research and Development; Groton; Connecticut; USA
| | - Ayman F. EI-Kattan
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Global Research and Development; Groton; Connecticut; USA
| | - Yurong Lai
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Global Research and Development; Groton; Connecticut; USA
| |
Collapse
|
41
|
The solute carrier family 10 (SLC10): beyond bile acid transport. Mol Aspects Med 2013; 34:252-69. [PMID: 23506869 DOI: 10.1016/j.mam.2012.07.004] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 06/13/2012] [Indexed: 12/22/2022]
Abstract
The solute carrier (SLC) family 10 (SLC10) comprises influx transporters of bile acids, steroidal hormones, various drugs, and several other substrates. Because the seminal transporters of this family, namely, sodium/taurocholate cotransporting polypeptide (NTCP; SLC10A1) and the apical sodium-dependent bile acid transporter (ASBT; SLC10A2), were primarily bile acid transporters, the term "sodium bile salt cotransporting family" was used for the SLC10 family. However, this notion became obsolete with the finding of other SLC10 members that do not transport bile acids. For example, the sodium-dependent organic anion transporter (SOAT; SLC10A6) transports primarily sulfated steroids. Moreover, NTCP was shown to also transport steroids and xenobiotics, including HMG-CoA inhibitors (statins). The SLC10 family contains four additional members, namely, P3 (SLC10A3; SLC10A3), P4 (SLC10A4; SLC10A4), P5 (SLC10A5; SLC10A5) and SLC10A7 (SLC10A7), several of which were unknown or considered hypothetical until approximately a decade ago. While their substrate specificity remains undetermined, great progress has been made towards their characterization in recent years. Explicitly, SLC10A4 may participate in vesicular storage or exocytosis of neurotransmitters or mastocyte mediators, whereas SLC10A5 and SLC10A7 may be involved in solute transport and SLC10A3 may have a role as a housekeeping protein. Finally, the newly found role of bile acids in glucose and energy homeostasis, via the TGR5 receptor, sheds new light on the clinical relevance of ASBT and NTCP. The present mini-review provides a brief summary of recent progress on members of the SLC10 family.
Collapse
|
42
|
Qiu X, Bi YA, Balogh LM, Lai Y. Absolute measurement of species differences in sodium taurocholate cotransporting polypeptide (NTCP/Ntcp) and its modulation in cultured hepatocytes. J Pharm Sci 2013; 102:3252-63. [PMID: 23657999 DOI: 10.1002/jps.23582] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/02/2013] [Accepted: 04/09/2013] [Indexed: 12/19/2022]
Abstract
Species differences among membrane transporters can be remarkable and difficult to properly assess by conventional methods. Herein, we employed the first use of stable isotope labeling in mammals or stable isotope-labeled peptides combined with mass spectrometry to identify species differences in sodium taurocholate cotransporting polypeptide (NTCP/Ntcp) protein expression in liver tissue and to characterize the modulation of protein expression in sandwich-cultured human (SCHH) and rat hepatocytes (SCRH). The lower limit of quantification was established to be 5 fmol on column with a standard curve that was linear up to 2000 fmol. The accuracy and precision were evaluated with three quality control samples and known amounts of synthetic proteotypic peptides that were spiked into the membrane protein extracts. The overall relative error and coefficient of variation were less than 10%. The expression of Ntcp in mouse and rat was significant higher than that in human (five-fold) and monkey (two-fold) and ranked as mouse > rat >> monkey > human. In the cultured hepatocytes, although significant downregulation of Ntcp expression in SCRH at day 5 after the culture was detected, NTCP expression in SCHH was comparable to the suspension hepatocytes. The results suggested that NTCP/Ntcp modulation in cultured hepatocytes is species specific.
Collapse
Affiliation(s)
- Xi Qiu
- Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Global Research & Development, Groton Laboratories, Pfizer Inc., Groton, Connecticut 06340, USA
| | | | | | | |
Collapse
|
43
|
Döring B, Lütteke T, Geyer J, Petzinger E. The SLC10 carrier family: transport functions and molecular structure. CURRENT TOPICS IN MEMBRANES 2013. [PMID: 23177985 DOI: 10.1016/b978-0-12-394316-3.00004-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The SLC10 family represents seven genes containing 1-12 exons that encode proteins in humans with sequence lengths of 348-477 amino acids. Although termed solute carriers (SLCs), only three out of seven (i.e. SLC10A1, SLC10A2, and SLC10A6) show sodium-dependent uptake of organic substrates across the cell membrane. These include the uptake of bile salts, sulfated steroids, sulfated thyroidal hormones, and certain statin drugs by SLC10A1 (Na(+)-taurocholate cotransporting polypeptide (NTCP)), the uptake of bile salts by SLC10A2 (apical sodium-dependent bile acid transporter (ASBT)), and uptake of sulfated steroids and sulfated taurolithocholate by SLC10A6 (sodium-dependent organic anion transporter (SOAT)). The other members of the family are orphan carriers not all localized in the cell membrane. The name "bile acid transporter family" arose because the first two SLC10 members (NTCP and ASBT) are carriers for bile salts that establish their enterohepatic circulation. In recent years, information has been obtained on their 2D and 3D membrane topology, structure-transport relationships, and on the ligand and sodium-binding sites. For SLC10A2, the putative 3D morphology was deduced from the crystal structure of a bacterial SLC10A2 analog, ASBT(NM). This information was used in this chapter to calculate the putative 3D structure of NTCP. This review provides first an introduction to recent knowledge about bile acid synthesis and newly found bile acid hormonal functions, and then describes step-by-step each individual member of the family in terms of expression, localization, substrate pattern, as well as protein topology with emphasis on the three functional SLC10 carrier members.
Collapse
Affiliation(s)
- Barbara Döring
- SLC10 family research group, Institute of Pharmacology and Toxicology, Justus Liebig University Giessen, Biomedical Research Center (BFS), Giessen, Germany
| | | | | | | |
Collapse
|
44
|
Murk AJ, Rijntjes E, Blaauboer BJ, Clewell R, Crofton KM, Dingemans MML, Furlow JD, Kavlock R, Köhrle J, Opitz R, Traas T, Visser TJ, Xia M, Gutleb AC. Mechanism-based testing strategy using in vitro approaches for identification of thyroid hormone disrupting chemicals. Toxicol In Vitro 2013; 27:1320-46. [PMID: 23453986 DOI: 10.1016/j.tiv.2013.02.012] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 02/07/2013] [Accepted: 02/18/2013] [Indexed: 11/16/2022]
Abstract
The thyroid hormone (TH) system is involved in several important physiological processes, including regulation of energy metabolism, growth and differentiation, development and maintenance of brain function, thermo-regulation, osmo-regulation, and axis of regulation of other endocrine systems, sexual behaviour and fertility and cardiovascular function. Therefore, concern about TH disruption (THD) has resulted in strategies being developed to identify THD chemicals (THDCs). Information on potential of chemicals causing THD is typically derived from animal studies. For the majority of chemicals, however, this information is either limited or unavailable. It is also unlikely that animal experiments will be performed for all THD relevant chemicals in the near future for ethical, financial and practical reasons. In addition, typical animal experiments often do not provide information on the mechanism of action of THDC, making it harder to extrapolate results across species. Relevant effects may not be identified in animal studies when the effects are delayed, life stage specific, not assessed by the experimental paradigm (e.g., behaviour) or only occur when an organism has to adapt to environmental factors by modulating TH levels. Therefore, in vitro and in silico alternatives to identify THDC and quantify their potency are needed. THDC have many potential mechanisms of action, including altered hormone production, transport, metabolism, receptor activation and disruption of several feed-back mechanisms. In vitro assays are available for many of these endpoints, and the application of modern '-omics' technologies, applicable for in vivo studies can help to reveal relevant and possibly new endpoints for inclusion in a targeted THDC in vitro test battery. Within the framework of the ASAT initiative (Assuring Safety without Animal Testing), an international group consisting of experts in the areas of thyroid endocrinology, toxicology of endocrine disruption, neurotoxicology, high-throughput screening, computational biology, and regulatory affairs has reviewed the state of science for (1) known mechanisms for THD plus examples of THDC; (2) in vitro THD tests currently available or under development related to these mechanisms; and (3) in silico methods for estimating the blood levels of THDC. Based on this scientific review, the panel has recommended a battery of test methods to be able to classify chemicals as of less or high concern for further hazard and risk assessment for THD. In addition, research gaps and needs are identified to be able to optimize and validate the targeted THD in vitro test battery for a mechanism-based strategy for a decision to opt out or to proceed with further testing for THD.
Collapse
Affiliation(s)
- AlberTinka J Murk
- Wageningen University, Sub-department of Toxicology, Tuinlaan 5, 6703 HE Wageningen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Thyroid hormone has profound effects on the heart and cardiovascular system. This article describes the cellular mechanisms by which thyroid hormone acts at the level of the cardiac myocyte and the vascular smooth muscle cell to alter phenotype and physiology. Because it is well established that thyroid hormone, specifically T(3), acts on almost every cell and organ in the body, studies on the regulation of thyroid hormone transport into cardiac and vascular tissue have added clinical significance. The characteristic changes in cardiovascular hemodynamics and metabolism that accompany thyroid disease states can then be best understood at the cellular level.
Collapse
Affiliation(s)
- Sara Danzi
- Department of Biological Sciences and Geology, Queensborough Community College, Bayside, NY 11364, USA
| | | |
Collapse
|
46
|
Greupink R, Dillen L, Monshouwer M, Huisman MT, Russel FGM. Interaction of fluvastatin with the liver-specific Na+ -dependent taurocholate cotransporting polypeptide (NTCP). Eur J Pharm Sci 2011; 44:487-96. [PMID: 21945488 DOI: 10.1016/j.ejps.2011.09.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 08/15/2011] [Accepted: 09/09/2011] [Indexed: 11/16/2022]
Abstract
It has been reported that polymorphisms in the organic anion transporting polypeptide 1B1 (OATP1B1, SLCO1B1) result in decreased hepatic uptake of simvastatin carboxy acid, the active metabolite of simvastatin. This is not the case for fluvastatin and it has been hypothesized that for this drug other hepatic uptake pathways exist. Here, we studied whether Na(+)-dependent taurocholate co-transporting polypeptide (NTCP, SLC10A1) can be an alternative hepatic uptake route for fluvastatin. Chinese Hamster Ovary cells transfected with human NTCP (CHO-NTCP) were used to investigate the inhibitory effect of fluvastatin and other statins on [(3)H]-taurocholic acid uptake ([(3)H]-TCA). Statin uptake by CHO-NTCP and cryopreserved human hepatocytes was assessed via LC-MS/MS. Fluvastatin appeared to be a potent and competitive inhibitor of [(3)H]-TCA uptake (IC(50) of 40μM), pointing to an interaction at the level of the bile acid binding pocket of NTCP. The inhibitory action of other statins was also studied, which revealed that statin inhibitory potency increased with molecular descriptors of lipophilicity: calculated logP (r(2)=0.82, p=0.034), logD(7.4) (r(2)=0.77, p=0.0001). Studies in CHO-NTCP cells showed that fluvastatin was indeed an NTCP substrate (K(m) 250±30μM, V(max) 1340±50ng/mg total cell protein/min). However, subsequent studies revealed that at clinically relevant plasma concentrations, NTCP contributed minimally to overall accumulation in human hepatocytes. In conclusion, fluvastatin interacts with NTCP at the level of the bile acid binding pocket and is an NTCP substrate. However, under normal conditions, NTCP-mediated uptake of this drug seems not to be a significant hepatocellular uptake pathway.
Collapse
Affiliation(s)
- Rick Greupink
- Department of Pharmacology and Toxicology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, The Netherlands
| | | | | | | | | |
Collapse
|
47
|
Stieger B, Geier A. Genetic variations of bile salt transporters as predisposing factors for drug-induced cholestasis, intrahepatic cholestasis of pregnancy and therapeutic response of viral hepatitis. Expert Opin Drug Metab Toxicol 2011; 7:411-25. [PMID: 21320040 DOI: 10.1517/17425255.2011.557067] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Drug-induced cholestasis, intrahepatic cholestasis of pregnancy and viral hepatitis are acquired forms of liver disease. Cholestasis is a pathophysiologic state with impaired bile formation and subsequent accumulation of bile salts in hepatocytes. The bile salt export pump (BSEP) (ABCB11) is the key export system for bile salts from hepatocytes. AREAS COVERED This article provides an introduction into the physiology of bile formation followed by a summary of the current knowledge on the key bile salt transporters, namely, the sodium-taurocholate co-transporting polypeptide NTCP, the organic anion transporting polypeptides (OATPs), BSEP and the multi-drug resistance protein 3. The pathophysiologic consequences of altered functions of these transporters, with an emphasis on molecular and genetic aspects, are then discussed. EXPERT OPINION Knowledge of the role of hepatocellullar transporters, especially BSEP, in acquired cholestasis is continuously increasing. A common variant of BSEP (p.V444A) is now a well-established susceptibility factor for acquired cholestasis and recent evidence suggests that the same variant also influences the therapeutic response and disease progression of viral hepatitis C. Studies in large independent cohorts are now needed to confirm the relevance of p.V444A. Genome-wide association studies should lead to the identification of additional genetic factors underlying cholestatic liver disease.
Collapse
Affiliation(s)
- Bruno Stieger
- University Hospital Zurich, Division of Clinical Pharmacology and Toxicology, 8091 Zurich, Switzerland.
| | | |
Collapse
|
48
|
Visser WE, Friesema ECH, Visser TJ. Minireview: thyroid hormone transporters: the knowns and the unknowns. Mol Endocrinol 2010; 25:1-14. [PMID: 20660303 DOI: 10.1210/me.2010-0095] [Citation(s) in RCA: 275] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The effects of thyroid hormone (TH) on development and metabolism are exerted at the cellular level. Metabolism and action of TH take place intracellularly, which require transport of the hormone across the plasma membrane. This process is mediated by TH transporter proteins. Many TH transporters have been identified at the molecular level, although a few are classified as specific TH transporters, including monocarboxylate transporter (MCT)8, MCT10, and organic anion-transporting polypeptide 1C1. The importance of TH transporters for physiology has been illustrated dramatically by the causative role of MCT8 mutations in males with psychomotor retardation and abnormal serum TH concentrations. Although Mct8 knockout animals have provided insight in the mechanisms underlying parts of the endocrine phenotype, they lack obvious neurological abnormalities. Thus, the pathogenesis of the neurological abnormalities in males with MCT8 mutations is not fully understood. The prospects of identifying other transporters and transporter-based syndromes promise an exciting future in the TH transporter field.
Collapse
Affiliation(s)
- W Edward Visser
- Erasmus University Medical Center, Molewaterplein 50, Rotterdam, The Netherlands
| | | | | |
Collapse
|