1
|
Zhang Z, Zhang Q, Zhang Y, Lou Y, Ge L, Zhang W, Zhang W, Song F, Huang P. Role of sodium taurocholate cotransporting polypeptide (NTCP) in HBV-induced hepatitis: Opportunities for developing novel therapeutics. Biochem Pharmacol 2024; 219:115956. [PMID: 38049009 DOI: 10.1016/j.bcp.2023.115956] [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: 08/23/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
Hepatitis B is an infectious disease caused by the HBV virus. It presents a significant challenge for treatment due to its chronic nature and the potential for developing severe complications, including hepatocirrhosis and hepatocellular carcinoma. These complications not only cause physical and psychological distress to patients but also impose substantial economic and social burdens on both individuals and society as a whole. The internalization of HBV relies on endocytosis and necessitates the involvement of various proteins, including heparin sulfate proteoglycans, epidermal growth factor receptors, and NTCP. Among these proteins, NTCP is pivotal in HBV internalization and is primarily located in the liver's basement membrane. As a transporter of bile acids, NTCP also serves as a receptor facilitating HBV entry into cells. Numerous molecules have been identified to thwart HBV infection by stifling NTCP activity, although only a handful exhibit low IC50 values. In this systematic review, our primary focus dwells on the structure and regulation of NTCP, as well as the mechanism involved in HBV internalization. We underscore recent drug breakthroughs that specifically target NTCP to combat HBV infection. By shedding light on these advances, this review contributes novel insights into developing effective anti-HBV medications.
Collapse
Affiliation(s)
- Zhentao Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Qi Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Yutao Lou
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Luqi Ge
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wanli Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wen Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Feifeng Song
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
2
|
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: 3.5] [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
|
3
|
Cardinali CAEF, Martins YA, Prates RP, de Araújo EV, Costa Viana FJ, de Sousa ME, Bombardi EDC, Nunes MT. The bank robbery analogy as a first approach for understanding basic concepts of synthesis, transport, and mechanism of action of hormones. ADVANCES IN PHYSIOLOGY EDUCATION 2022; 46:724-727. [PMID: 36227110 DOI: 10.1152/advan.00158.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 06/16/2023]
Abstract
Learning endocrine physiology can be challenging. Some physiological concepts are abstract, making the process of learning more difficult for students. The comprehension of basic concepts, such as chemical hormone classification, is essential to understand the differences in synthesis, secretion, transport, and mechanism of action of hormones. To assist the students on this subject, we developed an analogy between the basic concepts of hormone synthesis, transport, and mechanism of action and a bank robbery as a first approach to engage and stimulate their learning process. In the analogy, the students are asked to help identify and characterize two bank robbery crews based on a set of evidence collected by the police. The goal is to identify the general profile of lipid- and water-soluble hormones synthesis, transport, and mechanism of action on target cells. When applying the activity, the students showed a great deal of interest in solving the crime and they seemed to understand the similarities between the analogy and the subject.NEW & NOTEWORTHY Two endocrine bank robbery crews are being searched by the police. As an endocrine system student, you have been summoned to help the police solve the robberies.
Collapse
Affiliation(s)
| | - Yandara A Martins
- Departamento de Fisiologia e Biofisica, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | | | | | | | - Eduardo da Cunha Bombardi
- Departamento de Ciencias Basicas, Faculdade de Odontologia de Araçatuba-FOA/UNESP, Araçatuba, Sao Paulo, Brazil
| | - Maria Tereza Nunes
- Departamento de Fisiologia e Biofisica, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
4
|
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
|
5
|
Vansell NR. Mechanisms by Which Inducers of Drug Metabolizing Enzymes Alter Thyroid Hormones in Rats. Drug Metab Dispos 2022; 50:508-517. [DOI: 10.1124/dmd.121.000498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/23/2021] [Indexed: 11/22/2022] Open
|
6
|
Abstract
Na+/taurocholate cotransporting polypeptide (NTCP) is important for the enterohepatic circulation of bile acids, which has been suggested to contribute to the long serum elimination half-lives of perfluoroalkyl substances in humans. We demonstrated that some perfluoroalkyl sulfonates are transported by NTCP; however, little was known about carboxylates. The purpose of this study was to determine if perfluoroalkyl carboxylates would interact with NTCP and potentially act as substrates. Sodium-dependent transport of [3H]-taurocholate was measured in human embryonic kidney cells (HEK293) stably expressing NTCP in the absence or presence of perfluoroalkyl carboxylates with varying chain lengths. PFCAs with 8 (PFOA), 9 (PFNA), and 10 (PFDA) carbons were the strongest inhibitors. Inhibition kinetics demonstrated competitive inhibition and indicated that PFNA was the strongest inhibitor followed by PFDA and PFOA. All three compounds are transported by NTCP, and kinetics experiments revealed that PFOA had the highest affinity for NTCP with a Km value of 1.8 ± 0.4 mM. The Km value PFNA was estimated to be 5.3 ± 3.5 mM and the value for PFDA could not be determined due to limited solubility. In conclusion, our results suggest that, in addition to sulfonates, perfluorinated carboxylates are substrates of NTCP and have the potential to interact with NTCP-mediated transport.
Collapse
|
7
|
Mancino G, Miro C, Di Cicco E, Dentice M. Thyroid hormone action in epidermal development and homeostasis and its implications in the pathophysiology of the skin. J Endocrinol Invest 2021; 44:1571-1579. [PMID: 33683663 PMCID: PMC8285348 DOI: 10.1007/s40618-020-01492-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
Thyroid hormones (THs) are key endocrine regulators of tissue development and homeostasis. They are constantly released into the bloodstream and help to regulate many cell functions. The principal products released by the follicular epithelial cells are T3 and T4. T4, which is the less active form of TH, is produced in greater amounts than T3, which is the most active form of TH. This mechanism highlights the importance of the peripheral regulation of TH levels that goes beyond the central axis. Skin, muscle, liver, bone and heart are finely regulated by TH. In particular, skin is among the target organs most influenced by TH, which is essential for skin homeostasis. Accordingly, skin diseases are associated with an altered thyroid status. Alopecia, dermatitis and vitiligo are associated with thyroiditis and alopecia and eczema are frequently correlated with the Graves' disease. However, only in recent decades have studies started to clarify the molecular mechanisms underlying the effects of TH in epidermal homeostasis. Herein, we summarize the most frequent clinical epidermal alterations linked to thyroid diseases and review the principal mechanisms involved in TH control of keratinocyte proliferation and functional differentiation. Our aim is to define the open questions in this field that are beginning to be elucidated thanks to the advent of mouse models of altered TH metabolism and to obtain novel insights into the physiopathological consequences of TH metabolism on the skin.
Collapse
Affiliation(s)
- G Mancino
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - C Miro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - E Di Cicco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - M Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy.
- CEINGE-Biotecnologie Avanzate Scarl, Naples, Italy.
| |
Collapse
|
8
|
Marty S, Beekhuijzen M, Charlton A, Hallmark N, Hannas BR, Jacobi S, Melching-Kollmuss S, Sauer UG, Sheets LP, Strauss V, Urbisch D, Botham PA, van Ravenzwaay B. Towards a science-based testing strategy to identify maternal thyroid hormone imbalance and neurodevelopmental effects in the progeny - part II: how can key events of relevant adverse outcome pathways be addressed in toxicological assessments? Crit Rev Toxicol 2021; 51:328-358. [PMID: 34074207 DOI: 10.1080/10408444.2021.1910625] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The current understanding of thyroid-related adverse outcome pathways (AOPs) with adverse neurodevelopmental outcomes in mammals has been reviewed. This served to establish if standard rodent toxicity test methods and in vitro assays allow identifying thyroid-related modes-of-action potentially leading to adverse neurodevelopmental outcomes, and the human relevance of effects - in line with the European Commission's Endocrine Disruptor Criteria. The underlying hypothesis is that an understanding of the key events of relevant AOPs provides insight into differences in incidence, magnitude, or species sensitivity of adverse outcomes. The rodent studies include measurements of serum thyroid hormones, thyroid gland pathology and neurodevelopmental assessments, but do not directly inform on specific modes-of-action. Opportunities to address additional non-routine parameters reflecting critical events of AOPs in toxicological assessments are presented. These parameters appear relevant to support the identification of specific thyroid-related modes-of-action, provided that prevailing technical limitations are overcome. Current understanding of quantitative key event relationships is often weak, but would be needed to determine if the triggering of a molecular initiating event will ultimately result in an adverse outcome. Also, significant species differences in all processes related to thyroid hormone signalling are evident, but the biological implications thereof (including human relevance) are often unknown. In conclusion, careful consideration of the measurement (e.g. timing, method) and interpretation of additional non-routine parameters is warranted. These findings will be used in a subsequent paper to propose a testing strategy to identify if a substance may elicit maternal thyroid hormone imbalance and potentially also neurodevelopmental effects in the progeny.
Collapse
Affiliation(s)
- Sue Marty
- The Dow Chemical Company, Midland, MI, USA
| | | | | | | | | | | | | | - Ursula G Sauer
- Scientific Consultancy - Animal Welfare, Neubiberg, Germany
| | | | | | | | | | | |
Collapse
|
9
|
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
|
10
|
Zhang Q, He Z, Liu Z, Gong L. Integrated plasma and liver gas chromatography mass spectrometry and liquid chromatography mass spectrometry metabolomics to reveal physiological functions of sodium taurocholate cotransporting polypeptide (NTCP) with an Ntcp knockout mouse model. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1165:122531. [DOI: 10.1016/j.jchromb.2021.122531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 12/10/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022]
|
11
|
Venugopalan V, Al-Hashimi A, Rehders M, Golchert J, Reinecke V, Homuth G, Völker U, Manirajah M, Touzani A, Weber J, Bogyo MS, Verrey F, Wirth EK, Schweizer U, Heuer H, Kirstein J, Brix K. The Thyroid Hormone Transporter Mct8 Restricts Cathepsin-Mediated Thyroglobulin Processing in Male Mice through Thyroid Auto-Regulatory Mechanisms That Encompass Autophagy. Int J Mol Sci 2021; 22:ijms22010462. [PMID: 33466458 PMCID: PMC7796480 DOI: 10.3390/ijms22010462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
The thyroid gland is both a thyroid hormone (TH) generating as well as a TH responsive organ. It is hence crucial that cathepsin-mediated proteolytic cleavage of the precursor thyroglobulin is regulated and integrated with the subsequent export of TH into the blood circulation, which is enabled by TH transporters such as monocarboxylate transporters Mct8 and Mct10. Previously, we showed that cathepsin K-deficient mice exhibit the phenomenon of functional compensation through cathepsin L upregulation, which is independent of the canonical hypothalamus-pituitary-thyroid axis, thus, due to auto-regulation. Since these animals also feature enhanced Mct8 expression, we aimed to understand if TH transporters are part of the thyroid auto-regulatory mechanisms. Therefore, we analyzed phenotypic differences in thyroid function arising from combined cathepsin K and TH transporter deficiencies, i.e., in Ctsk-/-/Mct10-/-, Ctsk-/-/Mct8-/y, and Ctsk-/-/Mct8-/y/Mct10-/-. Despite the impaired TH export, thyroglobulin degradation was enhanced in the mice lacking Mct8, particularly in the triple-deficient genotype, due to increased cathepsin amounts and enhanced cysteine peptidase activities, leading to ongoing thyroglobulin proteolysis for TH liberation, eventually causing self-thyrotoxic thyroid states. The increased cathepsin amounts were a consequence of autophagy-mediated lysosomal biogenesis that is possibly triggered due to the stress accompanying intrathyroidal TH accumulation, in particular in the Ctsk-/-/Mct8-/y/Mct10-/- animals. Collectively, our data points to the notion that the absence of cathepsin K and Mct8 leads to excessive thyroglobulin degradation and TH liberation in a non-classical pathway of thyroid auto-regulation.
Collapse
Affiliation(s)
- Vaishnavi Venugopalan
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Alaa Al-Hashimi
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Maren Rehders
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Janine Golchert
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Vivien Reinecke
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Mythili Manirajah
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Adam Touzani
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Jonas Weber
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Matthew S. Bogyo
- Department of Pathology, School of Medicine, Stanford University, 300 Pasteur Dr., Stanford, CA 94305-5324, USA;
| | - Francois Verrey
- Physiologisches Institut, Universität Zürich, Winterthurerstr. 190, CH-8057 Zürich, Switzerland;
| | - Eva K. Wirth
- Berlin Institute of Health, Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Hessische Str. 3-4, Germany and DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, D-10115 Berlin, Germany;
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Medizinische Fakultät, Universität Bonn, Nußallee 11, D-53115 Bonn, Germany;
| | - Heike Heuer
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen (AöR), Universität Duisburg-Essen, Hufelandstr. 55, D-45147 Essen, Germany;
| | - Janine Kirstein
- Fachbereich 2 Biologie/Chemie, Faculty of Cell Biology, Universität Bremen, Leobener Straße 5, D-28359 Bremen, Germany;
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
- Correspondence: ; Tel.: +49-421-200-3246
| |
Collapse
|
12
|
Chung CW, Mo EY, Jung GS, Kim YH, Cho SW, Park DJ, Bae JM, Park YJ. Decreased Expression of Ileal Thyroid Hormone Transporters in a Hypothyroid Patient: A Case Report. Front Endocrinol (Lausanne) 2021; 12:664839. [PMID: 34122338 PMCID: PMC8187942 DOI: 10.3389/fendo.2021.664839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Malabsorption of levothyroxine (LT4) is a common problem faced in clinical practice. It is usually solved, if there are no complexities including gastrointestinal absorption disorder, by taking medicines on an empty stomach and avoiding foods interfering with LT4. Herein we present a rare case of a patient exhibiting malabsorption of LT4 with decreased membranous expression of ileal transporters. CASE The 22-Year-old female presented with sustained hypothyroid status despite medication of 7.8 μg/kg LT4. Medical history and LT4 absorption test (the absorption rate 8.4%) excluded pseudomalabsorption. No organic gastrointestinal disorder was found in the patient by blood chemistry, endoscopies, and abdominal computed tomography scan. The immunohistochemical analysis showed decreased membranous expression of LAT1 and LAT2 in distal ileum and ascending colon in the patient compared to 20 controls who have no thyroid disease. The expression of MCT8 in colon appeared at both nucleus and brush border in the patient, while it was limited to brush border in controls. The expression of other transporters was similar between the patient and controls. CONCLUSION The changes of the expression of LAT1 and LAT2 in this patient showing LT4 malabsorption might help to understand the role of intestinal transporters in the absorption of LT4 in humans. The functional relevance of the decrement of LAT1 and LAT2 in this patient remains to be elucidated.
Collapse
Affiliation(s)
- Chae Won Chung
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Eun Young Mo
- Department of Internal Medicine, The Catholic University of Korea Incheon St. Mary’s Hospital, Incheon, South Korea
| | - Gyung Seo Jung
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Yoo Hyung Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sun Wook Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Do Joon Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
- *Correspondence: Young Joo Park, ; Jeong Mo Bae,
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
- *Correspondence: Young Joo Park, ; Jeong Mo Bae,
| |
Collapse
|
13
|
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: 102] [Impact Index Per Article: 25.5] [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
|
14
|
Weidner J, Jensen CH, Giske J, Eliassen S, Jørgensen C. Hormones as adaptive control systems in juvenile fish. Biol Open 2020; 9:bio046144. [PMID: 31996351 PMCID: PMC7044463 DOI: 10.1242/bio.046144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Growth is an important theme in biology. Physiologists often relate growth rates to hormonal control of essential processes. Ecologists often study growth as a function of gradients or combinations of environmental factors. Fewer studies have investigated the combined effects of environmental and hormonal control on growth. Here, we present an evolutionary optimization model of fish growth that combines internal regulation of growth by hormone levels with the external influence of food availability and predation risk. The model finds a dynamic hormone profile that optimizes fish growth and survival up to 30 cm, and we use the probability of reaching this milestone as a proxy for fitness. The complex web of interrelated hormones and other signalling molecules is simplified to three functions represented by growth hormone, thyroid hormone and orexin. By studying a range from poor to rich environments, we find that the level of food availability in the environment results in different evolutionarily optimal strategies of hormone levels. With more food available, higher levels of hormones are optimal, resulting in higher food intake, standard metabolism and growth. By using this fitness-based approach we also find a consequence of evolutionary optimization of survival on optimal hormone use. Where foraging is risky, the thyroid hormone can be used strategically to increase metabolic potential and the chance of escaping from predators. By comparing model results to empirical observations, many mechanisms can be recognized, for instance a change in pace-of-life due to resource availability, and reduced emphasis on reserves in more stable environments.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jacqueline Weidner
- University of Bergen, Department of Biological Sciences, Postboks 7803, N-5020 Bergen, Norway
| | | | - Jarl Giske
- University of Bergen, Department of Biological Sciences, Postboks 7803, N-5020 Bergen, Norway
| | - Sigrunn Eliassen
- University of Bergen, Department of Biological Sciences, Postboks 7803, N-5020 Bergen, Norway
| | - Christian Jørgensen
- University of Bergen, Department of Biological Sciences, Postboks 7803, N-5020 Bergen, Norway
| |
Collapse
|
15
|
Batistuzzo A, Ribeiro MO. Clinical and subclinical maternal hypothyroidism and their effects on neurodevelopment, behavior and cognition. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2020; 64:89-95. [PMID: 32187263 PMCID: PMC10522279 DOI: 10.20945/2359-3997000000201] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/29/2019] [Indexed: 11/23/2022]
Abstract
Clinical and subclinical hypothyroidism are the most common hormonal dysfunctions during pregnancy. Insufficient maternal thyroid hormones (THs) in the early stages of pregnancy can lead to severe impairments in the development of the central nervous system because THs are critical to central nervous system development. In the fetus and after birth, THs participate in neurogenic processes, cell differentiation, neuronal activation, axonal growth, dendritic arborization, synaptogenesis and myelination. Although treatment is simple and effective, approximately 30% of pregnant women in Brazil with access to prenatal care have their first consultation after the first trimester of pregnancy, and any delay in diagnosis and resulting treatment delay may lead to cognitive impairment in children. This review summarizes the effects of clinical and subclinical hypothyroidism on fetal neurodevelopment, behavior and cognition in humans and rodents. Arch Endocrinol Metab. 2020;64(1):89-95.
Collapse
Affiliation(s)
- Alice Batistuzzo
- Departamento de Pós-Graduação em Distúrbios do DesenvolvimentoCentro de Ciências Biológicas e da SaúdeUniversidade Presbiteriana MackenzieSão PauloSPBrasilDepartamento de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde (CCBS), Universidade Presbiteriana Mackenzie (UPM), São Paulo, SP, Brasil
| | - Miriam Oliveira Ribeiro
- Departamento de Pós-Graduação em Distúrbios do DesenvolvimentoCentro de Ciências Biológicas e da SaúdeUniversidade Presbiteriana MackenzieSão PauloSPBrasilDepartamento de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde (CCBS), Universidade Presbiteriana Mackenzie (UPM), São Paulo, SP, Brasil
| |
Collapse
|
16
|
Barreto-Chaves MLM, Senger N, Fevereiro MR, Parletta AC, Takano APC. Impact of hyperthyroidism on cardiac hypertrophy. Endocr Connect 2020; 9:EC-19-0543.R1. [PMID: 32101527 PMCID: PMC7159257 DOI: 10.1530/ec-19-0543] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/25/2020] [Indexed: 12/14/2022]
Abstract
The cardiac growth process (hypertrophy) is a crucial phenomenon conserved across a wide array of species and it is critically involved in maintenance of cardiac homeostasis. This process enables organism adaptation to changes of systemic demand and occurs due to a plethora of responses, depending on the type of signal or stimuli received. The growth of cardiac muscle cells in response to environmental conditions depends on the type, strength and duration of stimuli, and results in adaptive physiologic response or non-adaptive pathologic response. Thyroid hormones (TH) have a direct effect on the heart and induce a cardiac hypertrophy phenotype, which may evolve to heart failure. In this review, we summarize the literature on TH function in heart presenting results from experimental studies. We discuss the mechanistic aspects of TH associated with cardiac myocyte hypertrophy, increased cardiac myocyte contractility and electrical remodeling as well as the signaling pathways associated. In addition to classical crosstalk with the Sympathetic Nervous System (SNS), emerging work points to the new endocrine interaction between TH and Renin-Angiotensin System (RAS) is also explored. Given the inflammatory potential of the angiotensin II peptide, this new interaction may open the door for new therapeutic approaches that target key mechanisms responsible for TH-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- M L M Barreto-Chaves
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - N Senger
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - M R Fevereiro
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - A C Parletta
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - A P C Takano
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
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
|
18
|
Pan G. Roles of Hepatic Drug Transporters in Drug Disposition and Liver Toxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:293-340. [PMID: 31571168 DOI: 10.1007/978-981-13-7647-4_6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatic drug transporters are mainly distributed in parenchymal liver cells (hepatocytes), contributing to drug's liver disposition and elimination. According to their functions, hepatic transporters can be roughly divided into influx and efflux transporters, translocating specific molecules from blood into hepatic cytosol and mediating the excretion of drugs and metabolites from hepatic cytosol to blood or bile, respectively. The function of hepatic transport systems can be affected by interspecies differences and inter-individual variability (polymorphism). In addition, some drugs and disease can redistribute transporters from the cell surface to the intracellular compartments, leading to the changes in the expression and function of transporters. Hepatic drug transporters have been associated with the hepatic toxicity of drugs. Gene polymorphism of transporters and altered transporter expressions and functions due to diseases are found to be susceptible factors for drug-induced liver injury (DILI). In this chapter, the localization of hepatic drug transporters, their regulatory factors, physiological roles, and their roles in drug's liver disposition and DILI are reviewed.
Collapse
Affiliation(s)
- Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, Shanghai, China.
| |
Collapse
|
19
|
Little AG. Local Regulation of Thyroid Hormone Signaling. VITAMINS AND HORMONES 2018; 106:1-17. [DOI: 10.1016/bs.vh.2017.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
20
|
|
21
|
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: 1.4] [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
|
22
|
Hinz KM, Neef D, Rutz C, Furkert J, Köhrle J, Schülein R, Krause G. Molecular features of the L-type amino acid transporter 2 determine different import and export profiles for thyroid hormones and amino acids. Mol Cell Endocrinol 2017; 443:163-174. [PMID: 28108384 DOI: 10.1016/j.mce.2017.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 10/20/2022]
Abstract
The L-type amino acid transporter 2 (LAT2) imports amino acids (AA) and also certain thyroid hormones (TH), e.g. 3,3'-T2 and T3, but not rT3 and T4. We utilized LAT2 mutations (Y130A, N133S, F242W) that increase 3,3'-T2 import and focus here on import and export capacity for AA, T4, T3, BCH and derivatives thereof to delineate molecular features. Transport studies and analysis of competitive inhibition of import by radiolabelled TH and AA were performed in Xenopus laevis oocytes. Only Y130A, a pocket widening mutation, enabled import for T4 and increased it for T3. Mutant F242W showed increased 3,3'-T2 import but no import rates for other TH derivatives. No export was detected for any TH by LAT2-wild type (WT). Mutations Y130A and N133S enabled only the export of 3,3'-T2, while N133S also increased AA export. Thus, distinct molecular LAT2-features determine bidirectional AA transport but only an unidirectional 3,3'-T2 and T3 import.
Collapse
Affiliation(s)
- Katrin M Hinz
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Dominik Neef
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Claudia Rutz
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Jens Furkert
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ralf Schülein
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Gerd Krause
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany.
| |
Collapse
|
23
|
Abstract
Transport of thyroid hormone (TH) across the plasma membrane is essential for intracellular TH metabolism and action, and this is mediated by specific transporter proteins. During the last two decades several transporters capable of transporting TH have been identified, including monocarboxylate transporter 8 (MCT8), MCT10 and organic anion transporting polypeptide 1C1 (OATP1C1). In particular MCT8 and OATP1C1 are important for the regulation of local TH activity in the brain and thus for brain development. MCT8 is a protein containing 12 transmembrane domains, and is encoded by the SLC16A2 gene located on the X chromosome. It facilitates both TH uptake and efflux across the cell membrane. Male subjects with hemizygous mutations in MCT8 are afflicted with severe intellectual and motor disability, also known as the Allan-Herndon-Dudley syndrome (AHDS), which goes together with low serum T4 and high T3 levels. This review concerns molecular and clinical aspects of MCT8 function.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - W Edward Visser
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Theo J Visser
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
24
|
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: 4.4] [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]
|
25
|
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
|
26
|
Visser TJ. Thyroid hormone transport across the placenta. ANNALES D'ENDOCRINOLOGIE 2016; 77:680-683. [PMID: 27659266 DOI: 10.1016/j.ando.2016.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 07/17/2016] [Indexed: 11/24/2022]
Affiliation(s)
- Theo J Visser
- Department of Internal Medicine, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.
| |
Collapse
|
27
|
Johannes J, Braun D, Kinne A, Rathmann D, Köhrle J, Schweizer U. Few Amino Acid Exchanges Expand the Substrate Spectrum of Monocarboxylate Transporter 10. Mol Endocrinol 2016; 30:796-808. [PMID: 27244477 PMCID: PMC5426580 DOI: 10.1210/me.2016-1037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/24/2016] [Indexed: 02/07/2023] Open
Abstract
Monocarboxylate transporters (MCTs) belong to the SLC16 family within the major facilitator superfamily of transmembrane transporters. MCT8 is a thyroid hormone transporter mutated in the Allan-Herndon-Dudley syndrome, a severe psychomotor retardation syndrome. MCT10 is closely related to MCT8 and is known as T-type amino acid transporter. Both transporters mediate T3 transport, but although MCT8 also transports rT3 and T4, these compounds are not efficiently transported by MCT10, which, in contrast, transports aromatic amino acids. Based on the 58% amino acid identity within the transmembrane regions among MCT8 and MCT10, we reasoned that substrate specificity may be primarily determined by a small number of amino acid differences between MCT8 and MCT10 along the substrate translocation channel. Inspecting the homology model of MCT8 and a structure-guided alignment between both proteins, we selected 8 amino acid positions and prepared chimeric MCT10 proteins with selected amino acids changed to the corresponding amino acids in MCT8. The MCT10 mutant harboring 8 amino acid substitutions was stably expressed in Madin-Darby canine kidney 1 cells and found to exhibit T4 transport activity. We then successively reduced the number of amino acid substitutions and eventually identified a minimal set of 2-3 amino acid exchanges which were sufficient to allow T4 transport. The resulting MCT10 chimeras exhibited KM values for T4 similar to MCT8 but transported T4 at a slower rate. The acquisition of T4 transport by MCT10 was associated with complete loss of the capacity to transport Phe, when Tyr184 was mutated to Phe.
Collapse
Affiliation(s)
- Jörg Johannes
- Institut für Biochemie und Molekularbiologie (J.J., D.B., U.S.), Rheinische Friedrich-Wilhelms-Universität, 53115 Bonn, Germany; and Institut für Experimentelle Endokrinologie (J.J., A.K., D.R., J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Doreen Braun
- Institut für Biochemie und Molekularbiologie (J.J., D.B., U.S.), Rheinische Friedrich-Wilhelms-Universität, 53115 Bonn, Germany; and Institut für Experimentelle Endokrinologie (J.J., A.K., D.R., J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Anita Kinne
- Institut für Biochemie und Molekularbiologie (J.J., D.B., U.S.), Rheinische Friedrich-Wilhelms-Universität, 53115 Bonn, Germany; and Institut für Experimentelle Endokrinologie (J.J., A.K., D.R., J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Daniel Rathmann
- Institut für Biochemie und Molekularbiologie (J.J., D.B., U.S.), Rheinische Friedrich-Wilhelms-Universität, 53115 Bonn, Germany; and Institut für Experimentelle Endokrinologie (J.J., A.K., D.R., J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Josef Köhrle
- Institut für Biochemie und Molekularbiologie (J.J., D.B., U.S.), Rheinische Friedrich-Wilhelms-Universität, 53115 Bonn, Germany; and Institut für Experimentelle Endokrinologie (J.J., A.K., D.R., J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie (J.J., D.B., U.S.), Rheinische Friedrich-Wilhelms-Universität, 53115 Bonn, Germany; and Institut für Experimentelle Endokrinologie (J.J., A.K., D.R., J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
28
|
Ortiga-Carvalho TM, Chiamolera MI, Pazos-Moura CC, Wondisford FE. Hypothalamus-Pituitary-Thyroid Axis. Compr Physiol 2016; 6:1387-428. [PMID: 27347897 DOI: 10.1002/cphy.c150027] [Citation(s) in RCA: 222] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The hypothalamus-pituitary-thyroid (HPT) axis determines the set point of thyroid hormone (TH) production. Hypothalamic thyrotropin-releasing hormone (TRH) stimulates the synthesis and secretion of pituitary thyrotropin (thyroid-stimulating hormone, TSH), which acts at the thyroid to stimulate all steps of TH biosynthesis and secretion. The THs thyroxine (T4) and triiodothyronine (T3) control the secretion of TRH and TSH by negative feedback to maintain physiological levels of the main hormones of the HPT axis. Reduction of circulating TH levels due to primary thyroid failure results in increased TRH and TSH production, whereas the opposite occurs when circulating THs are in excess. Other neural, humoral, and local factors modulate the HPT axis and, in specific situations, determine alterations in the physiological function of the axis. The roles of THs are vital to nervous system development, linear growth, energetic metabolism, and thermogenesis. THs also regulate the hepatic metabolism of nutrients, fluid balance and the cardiovascular system. In cells, TH actions are mediated mainly by nuclear TH receptors (210), which modify gene expression. T3 is the preferred ligand of THR, whereas T4, the serum concentration of which is 100-fold higher than that of T3, undergoes extra-thyroidal conversion to T3. This conversion is catalyzed by 5'-deiodinases (D1 and D2), which are TH-activating enzymes. T4 can also be inactivated by conversion to reverse T3, which has very low affinity for THR, by 5-deiodinase (D3). The regulation of deiodinases, particularly D2, and TH transporters at the cell membrane control T3 availability, which is fundamental for TH action. © 2016 American Physiological Society. Compr Physiol 6:1387-1428, 2016.
Collapse
Affiliation(s)
- Tania M Ortiga-Carvalho
- Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Maria I Chiamolera
- Department of Medicine, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Carmen C Pazos-Moura
- Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Fredic E Wondisford
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
29
|
|
30
|
Zibara K, El-Zein A, Joumaa W, El-Sayyad M, Mondello S, Kassem N. Thyroxine transfer from cerebrospinal fluid into choroid plexus and brain is affected by brefeldin A, low sodium, BCH, and phloretin, in ventriculo-cisternal perfused rabbits. Front Cell Dev Biol 2015; 3:60. [PMID: 26484343 PMCID: PMC4586509 DOI: 10.3389/fcell.2015.00060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/14/2015] [Indexed: 01/16/2023] Open
Abstract
Background: Thyroxine (T4) hormone is synthesized by the thyroid gland and then released into the systemic circulation where it binds to a number of proteins. Dysfunction in T4 transport mechanisms has been demonstrated in multiple central nervous system (CNS) diseases including Alzheimer's disease. In the presence of different compounds that inhibit potential T4 transport mechanisms, this study investigated the transfer of T4 from cerebrospinal fluid (CSF) into Choroid Plexus (CP) and other brain tissues. The compounds used were brefeldin A, low sodium artificial CSF (aCSF), BCH, phloretin, and taurocholate (TA). Methods: Radiolabeled T4 (125I-T4) was perfused continuously into the CSF and was assessed in several brain compartments with reference molecule 14C-mannitol and blue dextran, using the in vivo ventriculo-cisternal perfusion (V-C) technique in the rabbit. The aCSF containing the drug of interest was infused after 1 h of perfusion. Drugs were applied independently to the aCSF after 1 h of control perfusion. Results: Of interest, in presence of low sodium or BCH, the percentage recovery of 125I-T4, was increased compared to controls, with concomitant increase in T4 clearance. Conversely, brefeldin A, phloretin, and TA did not exert any significant effect on the recovery and clearance of 125I-T4 assessed in aCSF. On the other hand, the uptake of 125I-T4 into CP was raised by 18 fold compared to controls in the presence of brefeldin A. In addition, low sodium, BCH, or phloretin alone, enhanced the uptake of 125I-T4 by almost 3-fold, whereas TA did not show any significant effect. Finally, the uptake and distribution of 125I-T4 into other brain regions including ependymal region (ER) and caudate putamen (CAP) were significantly higher than in controls. Conclusion: Our study suggests the involvement of different mechanisms for the transfer of 125I-T4 from CSF into CP and other brain regions. This transfer may implicate sodium-dependent mechanisms, amino acid “L” system, or organic anion transporting polypeptide (OATP).
Collapse
Affiliation(s)
- Kazem Zibara
- ER045, PRASE, Faculty of Sciences-I, Lebanese University Beirut, Lebanon ; Department of Biology, Faculty of Sciences-I, Lebanese University Beirut, Lebanon
| | - Ali El-Zein
- ER045, PRASE, Faculty of Sciences-I, Lebanese University Beirut, Lebanon ; Physics Department, Faculty of Sciences, Lebanese University Beirut, Lebanon
| | - Wissam Joumaa
- Department of Biology, Faculty of Sciences-I, Lebanese University Beirut, Lebanon
| | | | | | - Nouhad Kassem
- ER045, PRASE, Faculty of Sciences-I, Lebanese University Beirut, Lebanon ; Department of Pharmacology and Therapeutics, Institute of Pharmaceutical Science, King's College London London, UK
| |
Collapse
|
31
|
Engels K, Rakov H, Zwanziger D, Moeller LC, Homuth G, Köhrle J, Brix K, Führer D. Differences in Mouse Hepatic Thyroid Hormone Transporter Expression with Age and Hyperthyroidism. Eur Thyroid J 2015; 4:81-6. [PMID: 26601077 PMCID: PMC4640301 DOI: 10.1159/000381020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 02/16/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Clinical features of thyroid dysfunction vary with age, and an oligosymptomatic presentation of hyperthyroidism is frequently observed in the elderly. This suggests age modulation of thyroid hormone (TH) action, which may occur, for example, by alterations in TH production, metabolism and/or TH action in target organs. OBJECTIVES In this paper, we address possible changes in TH transporter expression in liver tissues as a mechanism of age-dependent variation in TH action. METHODS Chronic hyperthyroidism was induced in 4- and 20-month-old C57BL6/NTac male mice (n = 8-10) by intraperitoneal injections of 1 µg/g body weight L-thyroxine (T4) every 48 h over 7 weeks. Control animals were injected with PBS. Total RNA was isolated from liver samples for analysis of the TH transporter and TH-responsive gene expression. TH concentrations were determined in mice sera. RESULTS Baseline serum free T4 (fT4) concentrations were significantly higher in euthyroid young compared to old mice. T4 treatment increased total T4, fT4 and free triiodothyronine to comparable concentrations in young and old mice. In the euthyroid state, TH transporter expression was significantly higher in old than in young mice, except for Mct8 and Oatp1a1 expression levels. Hyperthyroidism resulted in upregulation of Mct10, Lat1 and Lat2 in liver tissue, while Oatp1a1, Oatp1b2 and Oatp1a4 expression was downregulated. This effect was preserved in old animals. CONCLUSION Here, we show age-dependent differences in TH transporter mRNA expression in the euthyroid and hyperthyroid state of mice focusing on the liver as a classical TH target organ.
Collapse
Affiliation(s)
- Kathrin Engels
- Department of Endocrinology and Metabolism, University Hospital Essen, Essen, Germany
| | - Helena Rakov
- Department of Endocrinology and Metabolism, University Hospital Essen, Essen, Germany
| | - Denise Zwanziger
- Department of Endocrinology and Metabolism, University Hospital Essen, Essen, Germany
| | - Lars C. Moeller
- Department of Endocrinology and Metabolism, University Hospital Essen, Essen, Germany
| | - Georg Homuth
- Department of Functional Genomics, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Josef Köhrle
- Institute of Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Dagmar Führer
- Department of Endocrinology and Metabolism, University Hospital Essen, Essen, Germany
- *Dagmar Führer, MD, PhD, Department of Endocrinology and Metabolism and Division of Laboratory Research, University Hospital Essen, Hufelandstrasse 55, DE-45147 Essen (Germany), E-Mail
| |
Collapse
|
32
|
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: 6.7] [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
|
33
|
Alkemade A. Thyroid hormone and the developing hypothalamus. Front Neuroanat 2015; 9:15. [PMID: 25750617 PMCID: PMC4335174 DOI: 10.3389/fnana.2015.00015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/02/2015] [Indexed: 01/12/2023] Open
Abstract
Thyroid hormone (TH) plays an essential role in normal brain development and function. Both TH excess and insufficiency during development lead to structural brain abnormalities. Proper TH signaling is dependent on active transport of the prohormone thyroxine (T4) across the blood-brain-barrier and into brain cells. In the brain T4 undergoes local deiodination into the more active 3,3′,5-triiodothyronine (T3), which binds to nuclear TH receptors (TRs). TRs are already expressed during the first trimester of pregnancy, even before the fetal thyroid becomes functional. Throughout pregnancy, the fetus is largely dependent on the maternal TH supply. Recent studies in mice have shown that normal hypothalamic development requires intact TH signaling. In addition, the development of the human lateral hypothalamic zone coincides with a strong increase in T3 and TR mRNA concentrations in the brain. During this time the fetal hypothalamus already shows evidence for TH signaling. Expression of components crucial for central TH signaling show a specific developmental timing in the human hypothalamus. A coordinated expression of deiodinases in combination with TH transporters suggests that TH concentrations are regulated to prevent untimely maturation of brain cells. Even though the fetus depends on the maternal TH supply, there is evidence suggesting a role for the fetal hypothalamus in the regulation of TH serum concentrations. A decrease in expression of proteins involved in TH signaling towards the end of pregnancy may indicate a lower fetal TH demand. This may be relevant for the thyrotropin (TSH) surge that is usually observed after birth, and supports a role for the hypothalamus in the regulation of TH concentrations during the fetal period anticipating birth.
Collapse
Affiliation(s)
- Anneke Alkemade
- Amsterdam Brain and Cognition Center, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
34
|
Schweizer U, Johannes J, Bayer D, Braun D. Structure and function of thyroid hormone plasma membrane transporters. Eur Thyroid J 2014; 3:143-53. [PMID: 25538896 PMCID: PMC4224232 DOI: 10.1159/000367858] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/26/2014] [Indexed: 01/25/2023] Open
Abstract
Thyroid hormones (TH) cross the plasma membrane with the help of transporter proteins. As charged amino acid derivatives, TH cannot simply diffuse across a lipid bilayer membrane, despite their notorious hydrophobicity. The identification of monocarboxylate transporter 8 (MCT8, SLC16A2) as a specific and very active TH transporter paved the way to the finding that mutations in the MCT8 gene cause a syndrome of psychomotor retardation in humans. The purpose of this review is to introduce the current model of transmembrane transport and highlight the diversity of TH transmembrane transporters. The interactions of TH with plasma transfer proteins, T3 receptors, and deiodinase are summarized. It is shown that proteins may bind TH owing to their hydrophobic character in hydrophobic cavities and/or by specific polar interaction with the phenolic hydroxyl, the aminopropionic acid moiety, and by weak polar interactions with the iodine atoms. These findings are compared with our understanding of how TH transporters interact with substrate. The presumed effects of mutations in MCT8 on protein folding and transport function are explained in light of the available homology model.
Collapse
Affiliation(s)
- Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- *Prof. Dr. Ulrich Schweizer, Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Nussallee 11, DE-53115 Bonn (Germany), E-Mail
| | - Jörg Johannes
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dorothea Bayer
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Doreen Braun
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
35
|
Abstract
The description of two novel human defects in the last ten years has uncovered new aspects of thyroid hormone physiology with regard to cell-membrane transport and intracellular metabolism. Mutations in the X-linked monocarboxylate transporter 8 (MCT8) gene result in an invalidating neurodevelopmental phenotype in males and pathognomonic thyroid functions tests with high T3, low rT3, low or low normal T4, and normal or slightly high TSH. Recessive mutations in the selenocysteine insertion sequence binding protein 2 (SBP2) gene present a variable clinical phenotype depending on the severity of the defect and its consequences on the selenoprotein hierarchy. Most characteristic is the thyroid phenotype of low serum T3, high T4, high rT3, and slightly elevated TSH levels. Herein we review all known cases of MCT8 and SBP2 deficiency and describe each disease in terms of the clinical, biochemical, genetic, and therapeutic aspects.
Collapse
Affiliation(s)
- Jiao Fu
- Department of Medicine, University of Chicago Medical Center, 5841 S. Maryland Avenue MC3090, Room M369, Chicago, IL 60637, USA; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an 710061, People's Republic of China.
| | - Alexandra M Dumitrescu
- Department of Medicine, University of Chicago Medical Center, 5841 S. Maryland Avenue MC3090, Room M369, Chicago, IL 60637, USA.
| |
Collapse
|
36
|
Richardson VM, Ferguson SS, Sey YM, DeVito MJ. In vitrometabolism of thyroxine by rat and human hepatocytes. Xenobiotica 2013; 44:391-403. [DOI: 10.3109/00498254.2013.847990] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
37
|
Hagenbuch B, Stieger B. The SLCO (former SLC21) superfamily of transporters. Mol Aspects Med 2013; 34:396-412. [PMID: 23506880 DOI: 10.1016/j.mam.2012.10.009] [Citation(s) in RCA: 260] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 09/19/2012] [Indexed: 01/04/2023]
Abstract
The members of the organic anion transporting polypeptide superfamily (OATPs) are classified within the SLCO solute carrier family. All functionally well characterized members are predicted to have 12 transmembrane domains and are sodium-independent transport systems that mediate the transport of a broad range of endo- as well as xenobiotics. Substrates are mainly amphipathic organic anions with a molecular weight of more than 300Da, but some of the known transported substrates are also neutral or even positively charged. Among the well characterized substrates are numerous drugs including statins, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, antibiotics, antihistaminics, antihypertensives and anticancer drugs. Based on their amino acid sequence identities, the different OATPs cluster into families (in general with more than 40% amino acid sequence identity) and subfamilies (more than 60% amino acid identity). With the sequencing of genomes from different species and the computerized prediction of encoded proteins more than 300 OATPs can be found in the databases, however only a fraction of them have been identified in humans, rodents, and some additional species important for pharmaceutical research like the rhesus monkey (Macaca mulatta), the dog (Canis lupus familiaris) and the pig (Sus scrofa). These OATPs form 6 families (OATP1-OATP6) and 13 subfamilies. In this review we try to summarize what is currently known about OATPs with respect to endogenous substrates, tissue distribution, transport mechanisms, regulation of expression, structure-function relationship and mutations and polymorphisms.
Collapse
Affiliation(s)
- Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | |
Collapse
|
38
|
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: 95] [Impact Index Per Article: 8.6] [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
|
39
|
Colucci P, Yue CS, Ducharme M, Benvenga S. A Review of the Pharmacokinetics of Levothyroxine for the Treatment of Hypothyroidism. EUROPEAN ENDOCRINOLOGY 2013; 9:40-47. [PMID: 30349610 DOI: 10.17925/ee.2013.09.01.40] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 02/17/2013] [Indexed: 12/24/2022]
Abstract
Thyroxine hormone has been recognised since the early part of the nineteenth century and levothyroxine has been available since the mid-nineteenth century as a replacement for deficient thyroid hormones. While levothyroxine remains the staple treatment for hypothyroidism even to this day, its optimal use can be challenging. As is often the case with older drugs, the pharmacokinetics of levothyroxine is often under-appreciated or misunderstood and many factors influence the optimal dosing of levothyroxine. This article will review the pharmacokinetics of levothyroxine in the treatment of hypothyroidism and highlight major concepts that should aid both clinicians and researchers.
Collapse
Affiliation(s)
| | - Corinne Seng Yue
- Principal Scientist, Learn and Confirm Inc. and PhD Candidate, Faculty of Pharmacy, University of Montreal
| | - Murray Ducharme
- President and CEO, Learn and Confirm Inc., St Laurent, Canada and Associate Professor, Faculty of Pharmacy, University of Montreal, Montreal, Canada
| | - Salvatore Benvenga
- Professor of Medicine, Director, Master Program on Childhood, Adolescent and Women's Endocrine Health, and Chief, Interdepartmental Program of Molecular & Clinical Endocrinology and Women's Endocrine Healt, University of Messina, Messina, Italy
| |
Collapse
|
40
|
Antonini D, Sibilio A, Dentice M, Missero C. An Intimate Relationship between Thyroid Hormone and Skin: Regulation of Gene Expression. Front Endocrinol (Lausanne) 2013; 4:104. [PMID: 23986743 PMCID: PMC3749490 DOI: 10.3389/fendo.2013.00104] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/05/2013] [Indexed: 12/23/2022] Open
Abstract
Skin is the largest organ of the human body and plays a key role in protecting the individual from external insults. The barrier function of the skin is performed primarily by the epidermis, a self-renewing stratified squamous epithelium composed of cells that undergo a well-characterized and finely tuned process of terminal differentiation. By binding to their receptors thyroid hormones (TH) regulate epidermal cell proliferation, differentiation, and homeostasis. Thyroid dysfunction has multiple classical manifestations at skin level. Several TH-responsive genes, as well as genes critical for TH metabolism and action, are expressed at epidermal level. The role of TH in skin is still controversial, although it is generally recognized that TH signaling is central for skin physiology and homeostasis. Here we review the data on the epidermis and its function in relation to TH metabolism and regulation of gene expression. An understanding of the cellular and molecular basis of TH action in epidermal cells may lead to the identification of putative therapeutical targets for treatment of skin disorders.
Collapse
Affiliation(s)
| | - Annarita Sibilio
- Department of Clinical Medicine Surgery, University of Naples Federico II, Napoli, Italy
| | - Monica Dentice
- Department of Clinical Medicine Surgery, University of Naples Federico II, Napoli, Italy
| | - Caterina Missero
- CEINGE Biotecnologie Avanzate, Napoli, Italy
- Fondazione IRCCS SDN, Napoli, Italy
- *Correspondence: Caterina Missero, CEINGE Biotecnologie Avanzate, via Gaetano Salvatore 486, Napoli 80145, Italy e-mail:
| |
Collapse
|
41
|
Schweizer U, Köhrle J. Function of thyroid hormone transporters in the central nervous system. Biochim Biophys Acta Gen Subj 2012; 1830:3965-73. [PMID: 22890106 DOI: 10.1016/j.bbagen.2012.07.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 07/12/2012] [Accepted: 07/30/2012] [Indexed: 01/24/2023]
Abstract
BACKGROUND Iodothyronines are charged amino acid derivatives that cannot passively cross a phospholipid bilayer. Transport of thyroid hormones across plasma membranes is mediated by integral membrane proteins belonging to several gene families. These transporters therefore allow or limit access of thyroid hormones into brain. Since thyroid hormones are essential for brain development and cell differentiation, it is expected that genetic deficiency of such transporters would result in neurodevelopmental derangements. SCOPE OF REVIEW We introduce concepts of thyroid hormone transport into the brain and into brain cells. Important thyroid hormone transmembrane transporters are presented along with their expression patterns in different brain cell types. A focus is placed on monocarboxylate transporter 8 (MCT8) which has been identified as an essential thyroid hormone transporter in humans. Mutations in MCT8 underlie one of the first described X-linked mental retardation syndromes, the Allan-Herndon-Dudley syndrome. MAJOR CONCLUSIONS Thyroid hormone transporter molecules are expressed in a developmental and cell type-specific pattern. Any thyroid hormone molecule has to cross consecutively the luminal and abluminal membranes of the capillary endothelium, enter astrocytic foot processes, and leave the astrocyte through the plasma membrane to finally cross another plasma membrane on its way towards its target nucleus. GENERAL SIGNIFICANCE We can expect more transporters being involved in or contributing to in neurodevelopmental or neuropsychiatric disease. Due to their expression in cellular components regulating the hypothalamus-pituitary-thyroid axis, mutations and polymorphisms are expected to impact on negative feedback regulation and hormonal setpoints. This article is part of a Special Issue entitled Thyroid hormone signalling.
Collapse
Affiliation(s)
- Ulrich Schweizer
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | | |
Collapse
|
42
|
Abe S, Namba N, Abe M, Fujiwara M, Aikawa T, Kogo M, Ozono K. Monocarboxylate transporter 10 functions as a thyroid hormone transporter in chondrocytes. Endocrinology 2012; 153:4049-58. [PMID: 22719050 DOI: 10.1210/en.2011-1713] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thyroid hormone is essential for normal proliferation and differentiation of chondrocytes. Thus, untreated congenital hypothyroidism is marked by severe short stature. The monocarboxylate transporter 8 (MCT8) is a highly specific transporter for thyroid hormone. The hallmarks of Allan-Herndon-Dudley syndrome, caused by MCT8 mutations, are severe psychomotor retardation and elevated T(3) levels. However, growth is mostly normal. We therefore hypothesized that growth plate chondrocytes use transporters other than MCT8 for thyroid hormone uptake. Extensive analysis of thyroid hormone transporter mRNA expression in mouse chondrogenic ATDC5 cells revealed that monocarboxylate transporter 10 (Mct10) was most abundantly expressed among the transporters known to be highly specific for thyroid hormone, namely Mct8, Mct10, and organic anion transporter 1c1. Expression levels of Mct10 mRNA diminished with chondrocyte differentiation in these cells. Accordingly, Mct10 mRNA was expressed most abundantly in the growth plate resting zone chondrocytes in vivo. Small interfering RNA-mediated knockdown of Mct10 mRNA in ATDC5 cells decreased [(125)I]T(3) uptake up to 44% compared with negative control (P < 0.05). Moreover, silencing Mct10 mRNA expression abolished the known effects of T(3), i.e. suppression of proliferation and enhancement of differentiation, in ATDC5 cells. These results suggest that Mct10 functions as a thyroid hormone transporter in chondrocytes and can explain at least in part why Allan-Herndon-Dudley syndrome patients do not exhibit significant growth impairment.
Collapse
Affiliation(s)
- Sanae Abe
- Department of Pediatrics, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
43
|
Martin LA, Wilson DT, Reuhl KR, Gallo MA, Klaassen CD. Polychlorinated biphenyl congeners that increase the glucuronidation and biliary excretion of thyroxine are distinct from the congeners that enhance the serum disappearance of thyroxine. Drug Metab Dispos 2011; 40:588-95. [PMID: 22187485 DOI: 10.1124/dmd.111.042796] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Polychlorinated biphenyl (PCB) congeners differentially reduce serum thyroxine (T(4)) in rats, but little is known about their ability to affect biliary excretion of T(4). Thus, male Sprague-Dawley rats were orally administered Aroclor-1254, Aroclor-1242 (32 mg/kg per day), PCB-95, PCB-99, PCB-118 (16 mg/kg per day), PCB-126 (40 μg/kg per day), 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (3.9 μg/kg per day), or corn oil for 7 days. Twenty-four hours after the last dose, [(125)I]T(4) was administered intravenously, and blood, bile, and urine samples were collected for quantifying [(125)I]T(4) and in bile [(125)I]T(4) metabolites. Serum T(4) concentrations were reduced by all treatments, but dramatic reductions occurred in response to Aroclor-1254, PCB-99 [phenobarbital (PB)-type congener], and PCB-118 (mixed-type congener). None of the treatments increased urinary excretion of [(125)I]T(4). Aroclor-1254, PCB-118, TCDD, and PCB-126 (TCDD-type congener) increased biliary excretion of T(4)-glucuronide by 850, 756, 710, and 573%, respectively, corresponding to marked induction of hepatic UDP-glucuronosyltransferase (UGT) activity toward T(4). PCB-95 and PCB-99 did not induce UGT activity; therefore, the increased biliary excretion of T(4)-glucuronide was related to the affinity of congeners for the aryl hydrocarbon receptor. The disappearance of [(125)I]T(4) from serum was rapid (within 15-min) and was increased by Aroclor-1254, PCB-99 and PCB-118. Thus, reductions in serum T(4) in response to PCBs did not always correspond with UGT activity toward T(4) or with increased biliary excretion of T(4)-glucuronide. The rapid disappearance of [(125)I]T(4) from the serum of rats treated with PB-like PCBs suggests that increased tissue uptake of T(4) is an additional mechanism by which PCBs may reduce serum T(4).
Collapse
Affiliation(s)
- L A Martin
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | |
Collapse
|
44
|
Abstract
Thyroid hormones (TH) are essential for the development of the human brain, growth and cellular metabolism. Investigation of TH transporters became one of the emerging fields in thyroid research after the discovery of inactivating mutations in the Monocarboxylate transporter 8 (MCT8), which was found to be highly specific for TH transport. However, additional transmembrane transporters are also very important for TH uptake and efflux in different cell types. They transport TH as secondary substrates and include the aromatic amino acid transporting MCT10, the organic anion transporting polypeptides (e.g. OATP1C1, OATP1A2, OPTP1A4) and the large neutral amino acid transporters (LAT1 and LAT2). These TH transporters characteristically possess 12 transmembrane spanners but due to the strong differing sequences between the three transporter families we assume an identical conformation is not very likely. In contrast to the others, the LAT family members form a heterodimer with the escort protein 4F2hc/CD98. A comparison of sequence proportions, locations and types of functional sensitive features for TH transport discovered by mutations, revealed that transport sensitive charged residues occur as conserved amino acids only within each family of the transporter types but not in all putative TH transporters. Based on the lack of highly conserved sensitive charged residues throughout the three transporter families as a common counterpart for the amino acid moiety of the substrates, we conclude that the molecular transport mechanism is likely organized either a) by different molecular determinants in the divergent transporter types or b) the counterparts for the substrates` amino acid moiety at the transporter are not any charged side chains but other proton acceptors or donators. However, positions of transport sensitive residues coincide at transmembrane helix 8 in the TH transporter MCT8, OATP1C1 and another amino acid transporter, the L-cystine and L-glutamate exchanger xCT, which is highly homologous to LAT1 and LAT2. Here we review the data available and compare similarities and differences between these primary and secondary TH transporters regarding sequences, topology, potential structures, trafficking to the plasma membrane, molecular features and locations of transport sensitive functionalities. Thereby, we focus on TH transporters occurring in the blood-brain barrier.
Collapse
Affiliation(s)
- Anita Kinne
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Roessle-Str, 10, 13125 Berlin, Germany.
| | | | | |
Collapse
|
45
|
A possible mechanism for 2,2',4,4',5,5'-hexachlorobiphenyl-mediated decrease in serum thyroxine level in mice. Toxicol Appl Pharmacol 2011; 254:48-55. [PMID: 21569787 DOI: 10.1016/j.taap.2011.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 04/23/2011] [Accepted: 04/25/2011] [Indexed: 11/22/2022]
Abstract
Serum total thyroxine (T₄) level was markedly decreased, without significant increases in the levels of hepatic T₄-UDP-glucuronosyltransferase (T₄-UGT) and serum thyroid-stimulating hormone, 3 days after treatment with 2,2',4,4',5,5'-hexachlorobiphenyl (CB153) (100mg/kg, ip) in both 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-sensitive C57BL/6 and TCDD-resistant DBA/2 mice. Likewise, in either strain of mice, no CB153-mediated changes in the binding levels of [(125)I]T₄ to serum proteins, such as transthyretin, albumin, and thyroxine binding globulin, were observed, while in CB153-pretreated C57BL/6 mice, but not in CB153-pretreated DBA/2 mice, the levels of biliary [(125)I]₄T and [(125)I]T₄-glucuronide at 90-120 min after injection of [(125)I]T₄ slightly increased, as compared with those in the corresponding control mice. Concerning tissue distribution of [(125)I]T₄, liver-selective increases in the [(125)I]T₄ accumulation by CB153-pretreatment were observed in both C57BL/6 and DBA/2 mice, and the hepatic levels of [(125)I]T₄ in the C57BL/6 and DBA/2 mice became more than 44% and 34% of the [(125)I]T₄ dosed, respectively. The present findings indicated that the CB153-mediated decreases in the level of serum total T₄in C57BL/6 and DBA/2 mice occur mainly through an increase in the accumulation of T₄ in the liver.
Collapse
|
46
|
Stieger B. The role of the sodium-taurocholate cotransporting polypeptide (NTCP) and of the bile salt export pump (BSEP) in physiology and pathophysiology of bile formation. Handb Exp Pharmacol 2011:205-59. [PMID: 21103971 DOI: 10.1007/978-3-642-14541-4_5] [Citation(s) in RCA: 200] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bile formation is an important function of the liver. Bile salts are a major constituent of bile and are secreted by hepatocytes into bile and delivered into the small intestine, where they assist in fat digestion. In the small intestine, bile salts are almost quantitatively reclaimed and transported back via the portal circulation to the liver. In the liver, hepatocytes take up bile salts and secrete them again into bile for ongoing enterohepatic circulation. Uptake of bile salts into hepatocytes occurs largely in a sodium-dependent manner by the sodium taurocholate cotransporting polypeptide NTCP. The transport properties of NTCP have been extensively characterized. It is an electrogenic member of the solute carrier family of transporters (SLC10A1) and transports predominantly bile salts and sulfated compounds, but is also able to mediate transport of additional substrates, such as thyroid hormones, drugs and toxins. It is highly regulated under physiologic and pathophysiologic conditions. Regulation of NTCP copes with changes of bile salt load to hepatocytes and prevents entry of cytotoxic bile salts during liver disease. Canalicular export of bile salts is mediated by the ATP-binding cassette transporter bile salt export pump BSEP (ABCB11). BSEP constitutes the rate limiting step of hepatocellular bile salt transport and drives enterohepatic circulation of bile salts. It is extensively regulated to keep intracellular bile salt levels low under normal and pathophysiologic situations. Mutations in the BSEP gene lead to severe progressive familial intrahepatic cholestasis. The substrates of BSEP are practically restricted to bile salts and their metabolites. It is, however, subject to inhibition by endogenous metabolites or by drugs. A sustained inhibition will lead to acquired cholestasis, which can end in liver injury.
Collapse
Affiliation(s)
- Bruno Stieger
- Division of Clinical Pharmacology and Toxicology, University Hospital, 8091, Zurich, Switzerland.
| |
Collapse
|
47
|
König J. Uptake transporters of the human OATP family: molecular characteristics, substrates, their role in drug-drug interactions, and functional consequences of polymorphisms. Handb Exp Pharmacol 2011:1-28. [PMID: 21103967 DOI: 10.1007/978-3-642-14541-4_1] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Organic anion transporting polypeptides (OATPs, gene family: SLC21/SLCO) mediate the uptake of a broad range of substrates including several widely prescribed drugs into cells. Drug substrates for members of the human OATP family include HMG-CoA-reductase inhibitors (statins), antibiotics, anticancer agents, and cardiac glycosides. OATPs are expressed in a variety of different tissues including brain, intestine, liver, and kidney, suggesting that these uptake transporters are important for drug absorption, distribution, and excretion. Because of their wide tissue distribution and broad substrate spectrum, altered transport kinetics, for example, due to drug-drug interactions or due to the functional consequences of genetic variations (polymorphisms), can contribute to the interindividual variability of drug effects. Therefore, the molecular characteristics of human OATP family members, the role of human OATPs in drug-drug interactions, and the in vitro analysis of the functional consequences of genetic variations in SLCO genes encoding OATP proteins are the focus of this chapter.
Collapse
Affiliation(s)
- Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
48
|
Richardson TA, Klaassen CD. Disruption of thyroid hormone homeostasis in Ugt1a-deficient Gunn rats by microsomal enzyme inducers is not due to enhanced thyroxine glucuronidation. Toxicol Appl Pharmacol 2010; 248:38-44. [PMID: 20655938 DOI: 10.1016/j.taap.2010.07.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 07/10/2010] [Accepted: 07/14/2010] [Indexed: 11/25/2022]
Abstract
Microsomal enzyme inducers (MEI) that increase UDP-glucuronosyltransferases (UGTs) are thought to increase glucuronidation of thyroxine (T(4)), thus reducing serum T(4), and subsequently increasing thyroid stimulating hormone (TSH). Ugt1a1 and Ugt1a6 mediate T(4) glucuronidation. Therefore, this experiment determined the involvement of Ugt1a enzymes in increased T(4) glucuronidation, decreased serum T(4), and increased TSH after MEI treatment. Male Wistar and Ugt1a-deficient Wistar (Gunn) rats were fed a control diet or diet containing pregnenolone-16α-carbonitrile (PCN; 800 ppm), 3-methylcholanthrene (3-MC; 200 ppm), or Aroclor 1254 (PCB; 100 ppm) for 7 days. Serum T(4), triiodothyronine (T(3)), and TSH concentrations, hepatic T(4)/T(3) glucuronidation, and thyroid histology and follicular cell proliferation were investigated. PCN, 3-MC, and PCB treatments decreased serum T(4), whereas serum T(3) was maintained in both Gunn and Wistar rats (except for PCB treatment). TSH was increased in Wistar and Gunn rats after PCN (130 and 277%) or PCB treatment (72 and 60%). T(4) glucuronidation in Wistar rats was increased after PCN (298%), 3-MC (85%), and PCB (450%), but was extremely low in Gunn rats, and unchanged after MEI. T(3) glucuronidation was increased after PCN (121%) or PCB (58%) in Wistar rats, but only PCN increased T(3) glucuronidation in Gunn rats (43%). PCN treatment induced thyroid morphological changes and increased follicular cell proliferation in both strains. These data demonstrate that T(4) glucuronidation cannot be increased in Ugt1a-deficient Gunn rats. Thus, the decrease in serum T(4), increase in TSH, and increase in thyroid cell proliferation after MEI are not dependent on increased T(4) glucuronidation, and cannot be attributed to Ugt1a enzymes.
Collapse
Affiliation(s)
- Terrilyn A Richardson
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | |
Collapse
|
49
|
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: 270] [Impact Index Per Article: 19.3] [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
|
50
|
Abstract
Adequate levels of iodine, a trace element variably distributed on the earth, are required for the synthesis of the thyroid hormones thyroxine (T4) and triiodothyronine (T3). The iodide cycle consists of a series of transport, oxidation and coupling steps in thyroid follicular cells to produce thyroid hormone. The sodium/iodide symporter (NIS) transports iodide into the thyrocyte. Competitive inhibitors of NIS, such as perchlorate and thiocyanate, can decrease the entrance of iodide into the follicular cell. Pendrin is the primary protein that is responsible for iodide efflux out of the thyrocyte and into the follicular lumen. T4 is deiodinated in target tissues to produce the active form of thyroid hormone, T3, and other metabolites. Exposure to excessive iodine or chronic iodine deficiency may result in various clinical disorders. The Wolff-Chaikoff effect and Jöd-Basedow phenomenon describe mechanisms of thyroid autoregulation and dysregulation, respectively, during iodine excess. Population studies have determined that iodine deficiency exists in approximately 38% of the world's population, is the leading cause of preventable mental retardation, and is of particular concern to women and their infants. Finally, the unique role of iodine utilization in thyroid physiology has applications in many important clinical areas.
Collapse
Affiliation(s)
- Angela Leung
- a Boston University Medical Center, Section of Endocrinology, Diabetes, and Nutrition, 88 East Newton Street, Evans 201, Boston, MA 02118, USA
| | - Elizabeth N Pearce
- a Boston University Medical Center, Section of Endocrinology, Diabetes, and Nutrition, 88 East Newton Street, Evans 201, Boston, MA 02118, USA
| | - Lewis E Braverman
- a Boston University Medical Center, Section of Endocrinology, Diabetes, and Nutrition, 88 East Newton Street, Evans 201, Boston, MA 02118, USA
- b
| |
Collapse
|