1
|
Hagenbuch B, Stieger B, Locher KP. Organic anion transporting polypeptides: Pharmacology, toxicology, structure, and transport mechanisms. Pharmacol Rev 2025; 77:100023. [PMID: 40148036 DOI: 10.1016/j.pharmr.2024.100023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/12/2024] [Indexed: 03/29/2025] Open
Abstract
Organic anion transporting polypeptides (OATPs) are membrane proteins that mediate the uptake of a wide range of substrates across the plasma membrane of various cells and tissues. They are classified into 6 subfamilies, OATP1 through OATP6. Humans contain 12 OATPs encoded by 11 solute carrier of organic anion transporting polypeptide (SLCO) genes: OATP1A2, OATP1B1, OATP1B3, the splice variant OATP1B3-1B7, OATP1C1, OATP2A1, OATP2B1, OATP3A1, OATP4A1, OATP4C1, OATP5A1, and OATP6A1. Most of these proteins are expressed in epithelial cells, where they mediate the uptake of structurally unrelated organic anions, cations, and even neutral compounds into the cytoplasm. The best-characterized members are OATP1B1 and OATP1B3, which have an important role in drug metabolism by mediating drug uptake into the liver and are involved in drug-drug interactions. In this review, we aimed to (1) provide a historical perspective on the identification of OATPs and their nomenclature and discuss their phylogenic relationships and molecular characteristics; (2) review the current knowledge of the broad substrate specificity and their role in drug disposition and drug-drug interactions, with a special emphasis on human hepatic OATPs; (3) summarize the different experimental systems that are used to study the function of OATPs and discuss their advantages and disadvantages; (4) review the available experimental 3-dimensional structures and examine how they can help elucidate the transport mechanisms of OATPs; and (5) finally, summarize the current knowledge of the regulation of OATP expression, discuss clinically important single-nucleotide polymorphisms, and outline challenges of physiologically based pharmacokinetic modeling and in vitro to in vivo extrapolation. SIGNIFICANCE STATEMENT: Organic anion transporting polypeptides (OATPs) are a family of 12 uptake transporters in the solute carrier superfamily. Several members, particularly the liver-expressed OATP1B1 and OATP1B3, are important drug transporters. They mediate the uptake of several endobiotics and xenobiotics, including statins and numerous other drugs, into hepatocytes, and their inhibition by other drugs or reduced expression due to single-nucleotide polymorphisms can lead to adverse drug effects. Their recently solved 3-dimensional structures should help to elucidate their transport mechanisms and broad substrate specificities.
Collapse
Affiliation(s)
- Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas.
| | - Bruno Stieger
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| | - Kaspar P Locher
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
2
|
Wanjari UR, Gopalakrishnan AV. Blood-testis barrier: a review on regulators in maintaining cell junction integrity between Sertoli cells. Cell Tissue Res 2024; 396:157-175. [PMID: 38564020 DOI: 10.1007/s00441-024-03894-7] [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] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
The blood-testis barrier (BTB) is formed adjacent to the seminiferous basement membrane. It is a distinct ultrastructure, partitioning testicular seminiferous epithelium into apical (adluminal) and basal compartments. It plays a vital role in developing and maturing spermatocytes into spermatozoa via reorganizing its structure. This enables the transportation of preleptotene spermatocytes across the BTB, from basal to adluminal compartments in the seminiferous tubules. Several bioactive peptides and biomolecules secreted by testicular cells regulate the BTB function and support spermatogenesis. These peptides activate various downstream signaling proteins and can also be the target themself, which could improve the diffusion of drugs across the BTB. The gap junction (GJ) and its coexisting junctions at the BTB maintain the immunological barrier integrity and can be the "gateway" during spermatocyte transition. These junctions are the possible route for toxicant entry, causing male reproductive dysfunction. Herein, we summarize the detailed mechanism of all the regulators playing an essential role in the maintenance of the BTB, which will help researchers to understand and find targets for drug delivery inside the testis.
Collapse
Affiliation(s)
- Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, PIN 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, PIN 632014, India.
| |
Collapse
|
3
|
Caron P, Declèves X. The Use of Levothyroxine Absorption Tests in Clinical Practice. J Clin Endocrinol Metab 2023; 108:1875-1888. [PMID: 36916146 DOI: 10.1210/clinem/dgad132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/10/2023] [Accepted: 03/06/2023] [Indexed: 03/15/2023]
Abstract
Although levothyroxine (LT4) is a widely prescribed drug, more than 30% of LT4-treated patients fail to achieve the recommended serum level of thyrotropin with a body weight-based dose of LT4. An LT4 absorption test (LT4AT) is part of the workup for confirming normal LT4 absorption or diagnosing malabsorption. We searched PubMed with the terms levothyrox*, L-T4, LT4, TT4, FT4, FT3, TT3, test, loading, uptake, absorp*, "absorb*, bioavailab*, bioequiv* malabsorb*, and pseudomalabsorb*. A total of 43 full-text publications were analyzed. The published procedures for LT4AT differ markedly in the test dose, formulation, test duration, frequency of blood collection, analyte (total thyroxine [TT4] or free thyroxine [FT4]), metric (absolute or relative peak or increment, or area under the curve) and the threshold for normal absorption. In a standardized LT4AT for routine use, the physician could advise the patient to not consume food, beverages, or medications the morning of the test; administer 1000 µg of LT4 in the patient's usual formulation as the test dose; ensure that the patient is supervised throughout the LT4AT; perform a 4-hour test, with hourly blood samples; assay FT4; and consider that normal LT4 absorption corresponds to an FT4 increment of more than 0.40 ng/dL (5.14 pmol/L) or a TT4 increment of more than 6 μg/dL (77.23 nmol/L) for a test dose of at least 300 µg, or a percentage TT4 absorption of more than 60%. If the test indicates abnormal LT4 absorption, the physician can increase the LT4 dose, change the formulation or administration route, and/or refer the patient to a gastroenterologist.
Collapse
Affiliation(s)
- Philippe Caron
- Service d'Endocrinologie, Maladies Métaboliques et Nutrition, Pôle Cardio-Vasculaire et Métabolique, Hôpital Larrey, CHU de Toulouse, Toulouse F-31059, France
| | - Xavier Declèves
- Service de Biologie du Médicament-Toxicologie, Hôpital Cochin, AP-HP, Paris F-75006, France
- INSERM UMR-S1144, Université Paris Cité, Paris F-75006, France
| |
Collapse
|
4
|
Mégier C, Dumery G, Luton D. Iodine and Thyroid Maternal and Fetal Metabolism during Pregnancy. Metabolites 2023; 13:metabo13050633. [PMID: 37233673 DOI: 10.3390/metabo13050633] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Thyroid hormones and iodine are required to increase basal metabolic rate and to regulate protein synthesis, long bone growth and neuronal maturation. They are also essential for protein, fat and carbohydrate metabolism regulation. Imbalances in thyroid and iodine metabolism can negatively affect these vital functions. Pregnant women are at risk of hypo or hyperthyroidism, in relation to or regardless of their medical history, with potential dramatic outcomes. Fetal development highly relies on thyroid and iodine metabolism and can be compromised if they malfunction. As the interface between the fetus and the mother, the placenta plays a crucial role in thyroid and iodine metabolism during pregnancy. This narrative review aims to provide an update on current knowledge of thyroid and iodine metabolism in normal and pathological pregnancies. After a brief description of general thyroid and iodine metabolism, their main modifications during normal pregnancies and the placental molecular actors are described. We then discuss the most frequent pathologies to illustrate the upmost importance of iodine and thyroid for both the mother and the fetus.
Collapse
Affiliation(s)
- Charles Mégier
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| | - Grégoire Dumery
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| | - Dominique Luton
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| |
Collapse
|
5
|
Uchida T, Shimamura M, Taka H, Kaga N, Miura Y, Nishida Y, Nagayama Y, Watada H. The Effect of Long-Term Inorganic Iodine on Intrathyroidal Iodothyronine Content and Gene Expression in Mice with Graves' Hyperthyroidism. Thyroid 2023; 33:330-337. [PMID: 36565031 PMCID: PMC10024588 DOI: 10.1089/thy.2022.0496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background: The main molecular mechanism underlying acute suppression of iodine organification in normal thyroids after an excessive iodine load, that is, the Wolff-Chaikoff effect, is assumed to be suppression of iodine oxidation and iodothyronine synthesis. However, the mechanism underlying chronic antithyroid action of inorganic iodine in Graves' disease is not fully understood. Using a mouse model of Graves' hyperthyroidism, we examined changes in iodothyronine content and gene expression profiles in the thyroid glands after inorganic iodine loading. Materials and Methods: Graves' hyperthyroidism was induced and maintained in BALB/c mice by repeated immunizations of recombinant adenovirus expressing the human thyrotropin (TSH) receptor A-subunit. Hyperthyroid mice were left untreated (GD-C; n = 8) or treated with inorganic iodine for 12 weeks (GD-NaI; n = 8). We used unimmunized BALB/c mice as a control group (n = 10). In each mouse, serum thyroxine (T4) levels were measured with enzyme-linked immunosorbent assay (ELISA) at 4-week intervals. The intrathyroidal iodothyronine content and gene expression levels were, respectively, evaluated by mass spectrometry and RNA sequencing (RNA-seq) at the end of the experimental period. Results: Serum T4 levels in the GD-C group remained higher than in the control group, whereas those in the GD-NaI group declined to normal levels during the experimental period. Intrathyroidal triiodothyronine (T3), reverse T3 (rT3), and T4 contents in the GD-C group were higher than the control group, and rT3 and T4 were further increased in the GD-NaI group. The observed alterations in iodothyronine levels in the thyroid and sera may be explained by altered expression levels of genes for iodothyronine biosynthetic molecules, their transporter, and deiodinases. Conclusion: In this mouse model of hyperthyroidism, higher intrathyroidal accumulation of T4 and reduced gene expression data of iodothyronine transporters in the GD-NaI group suggest that chronic antithyroid action of iodine in Graves' disease involves suppression of hormone secretion.
Collapse
Affiliation(s)
- Toyoyoshi Uchida
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Address correspondence to: Toyoyoshi Uchida, MD, PhD, Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Mika Shimamura
- Department of Molecular Medicine, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Hikari Taka
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Naoko Kaga
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshiki Miura
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuya Nishida
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuji Nagayama
- Department of Molecular Medicine, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Hirotaka Watada
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Thyroid Hormone Transporters in Pregnancy and Fetal Development. Int J Mol Sci 2022; 23:ijms232315113. [PMID: 36499435 PMCID: PMC9737226 DOI: 10.3390/ijms232315113] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Thyroid hormone is essential for fetal (brain) development. Plasma membrane transporters control the intracellular bioavailability of thyroid hormone. In the past few decades, 15 human thyroid hormone transporters have been identified, and among them, mutations in monocarboxylate transporter (MCT)8 and organic anion transporting peptide (OATP)1C1 are associated with clinical phenotypes. Different animal and human models have been employed to unravel the (patho)-physiological role of thyroid hormone transporters. However, most studies on thyroid hormone transporters focus on postnatal development. This review summarizes the research on the thyroid hormone transporters in pregnancy and fetal development, including their substrate preference, expression and tissue distribution, and physiological and pathophysiological role in thyroid homeostasis and clinical disorders. As the fetus depends on the maternal thyroid hormone supply, especially during the first half of pregnancy, the review also elaborates on thyroid hormone transport across the human placental barrier. Future studies may reveal how the different transporters contribute to thyroid hormone homeostasis in fetal tissues to properly facilitate development. Employing state-of-the-art human models will enable a better understanding of their roles in thyroid hormone homeostasis.
Collapse
|
7
|
Chen Z, van der Sman AE, Groeneweg S, de Rooij LJ, Visser WE, Peeters RP, Meima ME. Thyroid Hormone Transporters in a Human Placental Cell Model. Thyroid 2022; 32:1129-1137. [PMID: 35699060 PMCID: PMC9526468 DOI: 10.1089/thy.2021.0503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Background: Fetal brain development in the first half of pregnancy is dependent on maternal thyroid hormone (TH), highlighting the importance of trans-placental TH transport. It is yet unclear which transporters are involved in this process. We aimed to identify the major TH transporters in a human placental cell model (BeWo cells). Methods: Messenger RNA expression of the known TH transporters (the monocarboxylate transporter [MCT]8, MCT10, the L-type amino acid transporter [LAT]1, LAT2, the organic anion transporting peptide [OATP]1A2 and OATP4A1) in BeWo cells and human placenta were determined by quantitative PCR. To determine the specificity and efficacy of transporter inhibitors, we first determined TH uptake at different inhibitor concentrations in African green monkey kidney fibroblast-like cells (COS1 cells) overexpressing TH transporters. We then tested TH uptake in BeWo cells in the presence or absence of the optimal inhibitor concentrations. Results: All tested TH transporters were expressed in human term placentas, whereas MCT8 was absent in BeWo cells. Both 2-amino-2-norbornanecarboxylic acid (BCH) and L-tryptophan at 1 mM inhibited LATs, whereas at the highest concentration (10 mM) L-tryptophan also inhibited MCT10. Verapamil inhibited OATP1A2 and less efficiently both MCTs, but not LATs. Both rifampicin and naringin reduced OATP1A2 activity. Finally, silychristin inhibited MCT8 at submicromolar concentrations and OATP1A2 partially only at the highest concentration tested (10 μM). In BeWo cells, verapamil reduced triiodothyronine (T3) uptake by 24%, BCH by 31%, and 1 mM L-tryptophan by 41%. The combination of BCH and verapamil additively decreased T3 uptake by 53% and the combination of BCH and 10 mM L-tryptophan by 60%, suggesting a major role for MCT10 and LATs in placental T3 uptake. Indeed, transfection of BeWo cells with MCT10-specific small interfering RNA significantly reduced T3 uptake. Only the combination of BCH and verapamil significantly reduced thyroxine (T4) uptake in BeWo cells, by 32%. Conclusions: Using pharmacological inhibitors, we show that MCT10 and LATs play a major role in T3 uptake in BeWo cells. T4 uptake appears independent of known TH transporters, suggesting the presence of, currently unknown, alternative transporter(s).
Collapse
Affiliation(s)
- Zhongli Chen
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A.S. Elise van der Sman
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Stefan Groeneweg
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Linda Johanna de Rooij
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - W. Edward Visser
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robin P. Peeters
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marcel E. Meima
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Address correspondence to: Marcel E. Meima, PhD, Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands
| |
Collapse
|
8
|
England-Mason G, Merrill SM, Gladish N, Moore SR, Giesbrecht GF, Letourneau N, MacIsaac JL, MacDonald AM, Kinniburgh DW, Ponsonby AL, Saffery R, Martin JW, Kobor MS, Dewey D. Prenatal exposure to phthalates and peripheral blood and buccal epithelial DNA methylation in infants: An epigenome-wide association study. ENVIRONMENT INTERNATIONAL 2022; 163:107183. [PMID: 35325772 DOI: 10.1016/j.envint.2022.107183] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/16/2022] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Prenatal exposure to phthalates has been associated with adverse health and neurodevelopmental outcomes. DNA methylation (DNAm) alterations may be a mechanism underlying these effects, but prior investigations of prenatal exposure to phthalates and neonatal DNAm profiles are limited to placental tissue and umbilical cord blood. OBJECTIVE Conduct an epigenome-wide association study (EWAS) of the associations between prenatal exposure to phthalates and DNAm in two accessible infant tissues, venous buffy coat blood and buccal epithelial cells (BECs). METHODS Participants included 152 maternal-infant pairs from the Alberta Pregnancy Outcomes and Nutrition (APrON) study. Maternal second trimester urine samples were analyzed for nine phthalate metabolites. Blood (n = 74) or BECs (n = 78) were collected from 3-month-old infants and profiled for DNAm using the Infinium HumanMethylation450 (450K) BeadChip. Robust linear regressions were used to investigate the associations between high (HMWPs) and low molecular weight phthalates (LMWPs) and change in methylation levels at variable Cytosine-phosphate-Guanine (CpG) sites in infant tissues, as well as the sensitivity of associations to potential confounders. RESULTS One candidate CpG in gene RNF39 reported by a previous study examining prenatal exposure to phthalates and cord blood DNAm was replicated. The EWAS identified 12 high-confidence CpGs in blood and another 12 in BECs associated with HMWPs and/or LMWPs. Prenatal exposure to bisphenol A (BPA) associated with two of the CpGs associated with HMWPs in BECs. DISCUSSION Prenatal exposure to phthalates was associated with DNAm variation at CpGs annotated to genes associated with endocrine hormone activity (i.e., SLCO4A1, TPO), immune pathways and DNA damage (i.e., RASGEF1B, KAZN, HLA-A, MYO18A, DIP2C, C1or109), and neurodevelopment (i.e., AMPH, NOTCH3, DNAJC5). Future studies that characterize the stability of these associations in larger samples, multiple cohorts, across tissues, and investigate the potential associations between these biomarkers and relevant health and neurodevelopmental outcomes are needed.
Collapse
Affiliation(s)
- Gillian England-Mason
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sarah M Merrill
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Nicole Gladish
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Sarah R Moore
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Gerald F Giesbrecht
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Psychology, Faculty of Arts, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicole Letourneau
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Faculty of Nursing, University of Calgary, Calgary, Alberta, Canada; Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Julia L MacIsaac
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Amy M MacDonald
- Alberta Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada
| | - David W Kinniburgh
- Alberta Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Anne-Louise Ponsonby
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Jonathan W Martin
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, Stockholm, Södermanland, Sweden
| | - Michael S Kobor
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada; Program in Child and Brain Development, CIFAR, Toronto, Ontario, Canada
| | - Deborah Dewey
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Owerko Centre, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada.
| |
Collapse
|
9
|
Wenzek C, Boelen A, Westendorf AM, Engel DR, Moeller LC, Führer D. The interplay of thyroid hormones and the immune system - where we stand and why we need to know about it. Eur J Endocrinol 2022; 186:R65-R77. [PMID: 35175936 PMCID: PMC9010816 DOI: 10.1530/eje-21-1171] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/17/2022] [Indexed: 11/08/2022]
Abstract
Over the past few years, growing evidence suggests direct crosstalk between thyroid hormones (THs) and the immune system. Components of the immune system were proposed to interfere with the central regulation of systemic TH levels. Conversely, THs regulate innate and adaptive immune responses as immune cells are direct target cells of THs. Accordingly, they express different components of local TH action, such as TH transporters or receptors, but our picture of the interplay between THs and the immune system is still incomplete. This review provides a critical overview of current knowledge regarding the interaction of THs and the immune system with the main focus on local TH action within major innate and adaptive immune cell subsets. Thereby, this review aims to highlight open issues which might help to infer the clinical relevance of THs in host defence in the context of different types of diseases such as infection, ischemic organ injury or cancer.
Collapse
Affiliation(s)
- Christina Wenzek
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Anita Boelen
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Astrid M Westendorf
- Institute for Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Daniel R Engel
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lars C Moeller
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Dagmar Führer
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Correspondence should be addressed to D Führer;
| |
Collapse
|
10
|
Schäfer AM, Meyer zu Schwabedissen HE, Grube M. Expression and Function of Organic Anion Transporting Polypeptides in the Human Brain: Physiological and Pharmacological Implications. Pharmaceutics 2021; 13:pharmaceutics13060834. [PMID: 34199715 PMCID: PMC8226904 DOI: 10.3390/pharmaceutics13060834] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022] Open
Abstract
The central nervous system (CNS) is an important pharmacological target, but it is very effectively protected by the blood–brain barrier (BBB), thereby impairing the efficacy of many potential active compounds as they are unable to cross this barrier. Among others, membranous efflux transporters like P-Glycoprotein are involved in the integrity of this barrier. In addition to these, however, uptake transporters have also been found to selectively uptake certain compounds into the CNS. These transporters are localized in the BBB as well as in neurons or in the choroid plexus. Among them, from a pharmacological point of view, representatives of the organic anion transporting polypeptides (OATPs) are of particular interest, as they mediate the cellular entry of a variety of different pharmaceutical compounds. Thus, OATPs in the BBB potentially offer the possibility of CNS targeting approaches. For these purposes, a profound understanding of the expression and localization of these transporters is crucial. This review therefore summarizes the current state of knowledge of the expression and localization of OATPs in the CNS, gives an overview of their possible physiological role, and outlines their possible pharmacological relevance using selected examples.
Collapse
Affiliation(s)
- Anima M. Schäfer
- Biopharmacy, Department Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland; (A.M.S.); (H.E.M.z.S.)
| | - Henriette E. Meyer zu Schwabedissen
- Biopharmacy, Department Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland; (A.M.S.); (H.E.M.z.S.)
| | - Markus Grube
- Center of Drug Absorption and Transport (C_DAT), Department of Pharmacology, University Medicine of Greifswald, 17489 Greifswald, Germany
- Correspondence: ; Tel./Fax: +49-3834-865636
| |
Collapse
|
11
|
Abstract
Thyroid hormone is essential for brain development and brain function in the adult. During development, thyroid hormone acts in a spatial and temporal-specific manner to regulate the expression of genes essential for normal neural cell differentiation, migration, and myelination. In the adult brain, thyroid hormone is important for maintaining normal brain function. Thyroid hormone excess, hyperthyroidism, and thyroid hormone deficiency, hypothyroidism, are associated with disordered brain function, including depression, memory loss, impaired cognitive function, irritability, and anxiety. Adequate thyroid hormone levels are required for normal brain function. Thyroid hormone acts through a cascade of signaling components: activation and inactivation by deiodinase enzymes, thyroid hormone membrane transporters, and nuclear thyroid hormone receptors. Additionally, the hypothalamic-pituitary-thyroid axis, with negative feedback of thyroid hormone on thyrotropin-releasing hormone (TRH) and thyroid-stimulating hormone (TSH) secretion, regulates serum thyroid hormone levels in a narrow range. Animal and human studies have shown both systemic and local reduction in thyroid hormone availability in neurologic disease and after brain trauma. Treatment with thyroid hormone and selective thyroid hormone analogs has resulted in a reduction in injury and improved recovery. This article will describe the thyroid hormone signal transduction pathway in the brain and the role of thyroid hormone in the aging brain, neurologic diseases, and the protective role when administered after traumatic brain injury. © 2021 American Physiological Society. Compr Physiol 11:1-21, 2021.
Collapse
Affiliation(s)
- Yan-Yun Liu
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA.,Departments of Medicine and Physiology, Endocrinology, Diabetes and Metabolism Division, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Gregory A Brent
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA.,Departments of Medicine and Physiology, Endocrinology, Diabetes and Metabolism Division, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
12
|
Zafeiri A, Mitchell RT, Hay DC, Fowler PA. Over-the-counter analgesics during pregnancy: a comprehensive review of global prevalence and offspring safety. Hum Reprod Update 2020; 27:67-95. [PMID: 33118024 DOI: 10.1093/humupd/dmaa042] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/16/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Analgesia during pregnancy is often necessary. Due to their widespread availability, many mothers opt to use over-the-counter (OTC) analgesics. Those analgesic compounds and their metabolites can readily cross the placenta and reach the developing foetus. Evidence for safety or associations with adverse health outcomes is conflicting, limiting definitive decision-making for healthcare professionals. OBJECTIVE AND RATIONALE This review provides a detailed and objective overview of research in this field. We consider the global prevalence of OTC analgesia during pregnancy, explain the current mechanistic understanding of how analgesic compounds cross the placenta and reach the foetus, and review current research on exposure associations with offspring health outcomes. SEARCH METHODS A comprehensive English language literature search was conducted using PubMed and Scopus databases. Different combinations of key search terms were used including 'over-the-counter/non-prescription analgesics', 'pregnancy', 'self-medication', 'paracetamol', 'acetaminophen', 'diclofenac', 'aspirin', 'ibuprofen', 'in utero exposure', 'placenta drug transport', 'placental transporters', 'placenta drug metabolism' and 'offspring outcomes'. OUTCOMES This article examines the evidence of foetal exposure to OTC analgesia, starting from different routes of exposure to evidence, or the lack thereof, linking maternal consumption to offspring ill health. There is a very high prevalence of maternal consumption of OTC analgesics globally, which is increasing sharply. The choice of analgesia selected by pregnant women differs across populations. Location was also observed to have an effect on prevalence of use, with more developed countries reporting the highest consumption rates. Some of the literature focuses on the association of in utero exposure at different pregnancy trimesters and the development of neurodevelopmental, cardiovascular, respiratory and reproductive defects. This is in contrast to other studies which report no associations. WIDER IMPLICATIONS The high prevalence and the challenges of reporting exact consumption rates make OTC analgesia during pregnancy a pressing reproductive health issue globally. Even though some healthcare policy-making authorities have declared the consumption of some OTC analgesics for most stages of pregnancy to be safe, such decisions are often based on partial review of literature. Our comprehensive review of current evidence highlights that important knowledge gaps still exist. Those areas require further research in order to provide pregnant mothers with clear guidance with regard to OTC analgesic use during pregnancy.
Collapse
Affiliation(s)
- Aikaterini Zafeiri
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - David C Hay
- MRC Centre for Regenerative Medicine, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
13
|
Peng S, Li C, Xie X, Zhang X, Wang D, Lu X, Sun M, Meng T, Wang S, Jiang Y, Shan Z, Teng W. Divergence of Iodine and Thyroid Hormones in the Fetal and Maternal Parts of Human-Term Placenta. Biol Trace Elem Res 2020; 195:27-38. [PMID: 31502179 DOI: 10.1007/s12011-019-01834-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/11/2019] [Indexed: 01/29/2023]
Abstract
The human placenta is an important organ that forms a barrier where maternal and fetal exchange takes place. The placenta transport iodine to the fetal circulation by transfer of maternal iodine and deiodination of thyroid hormones (THs). The aim of the study was to examine the distribution of iodine and thyroid hormone transporters in the maternal and fetal sides of human-term placenta. A cross-sectional study was performed at the First Affiliated Hospital of China Medical University. Placental samples (maternal and fetal surfaces) were collected from 113 healthy-term pregnant women. The iodine content; the concentration of thyroxine (T4), triiodothyronine (T3), and reverse T3 (rT3); and the enzyme activity of placental type 2 iodothyronine deiodinase (D2) and D3 were examined. The mRNA and protein localization/expression of iodine and thyroid hormone transporters in the placenta were also studied. We also analyzed the association between expression level of Na+/I- symporter (NIS), thyroid hormone transporter protein, D3 activity in maternal and fetal surfaces of placenta with iodine content, and thyroid hormone levels. Iodine levels in placental samples from the maternal side were significantly higher than those in samples from the fetal side. T3 and T4 expression in fetal placenta was significantly lower than in maternal placenta. D3 activity in the fetal side of the placentas was significantly higher than that in the maternal side. The mRNA and protein expression of monocarboxylate transporters 8 (MCT8), L-amino acid transporters 1 (LAT1), organic anion transporting polypeptides 4A1 (OATP4A1), and TH binding protein transthyretin (TTR) were significantly increased in maternal side, while the NIS expression was higher in fetal side of human-term placenta. In conclusion, the enzymatic deiodination of thyroid hormones forms a barrier which reduces transplacental passage of the hormones and that the maternal part of the placenta is the primary factor in the mechanism regulating the hormonal transfer.
Collapse
Affiliation(s)
- Shiqiao Peng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Chenyan Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Xiaochen Xie
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Xiaomei Zhang
- Department of Endocrinology and Metabolism, Peking University International Hospital, Haidian, Beijing, 100000, People's Republic of China
| | - Danyang Wang
- Department of Endocrinology and Metabolism, The First Hospital of Dandong, Dandong, 118000, Liaoning, People's Republic of China
| | - Xixuan Lu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Manni Sun
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Tao Meng
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Shiwei Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Yaqiu Jiang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China.
| |
Collapse
|
14
|
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: 120] [Impact Index Per Article: 24.0] [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
|
15
|
Li TT, An JX, Xu JY, Tuo BG. Overview of organic anion transporters and organic anion transporter polypeptides and their roles in the liver. World J Clin Cases 2019; 7:3915-3933. [PMID: 31832394 PMCID: PMC6906560 DOI: 10.12998/wjcc.v7.i23.3915] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 02/05/2023] Open
Abstract
Organic anion transporters (OATs) and organic anion transporter polypeptides (OATPs) are classified within two SLC superfamilies, namely, the SLC22A superfamily and the SLCO superfamily (formerly the SLC21A family), respectively. They are expressed in many tissues, such as the liver and kidney, and mediate the absorption and excretion of many endogenous and exogenous substances, including various drugs. Most are composed of 12 transmembrane polypeptide chains with the C-terminus and the N-terminus located in the cell cytoplasm. OATs and OATPs are abundantly expressed in the liver, where they mainly promote the uptake of various endogenous substrates such as bile acids and various exogenous drugs such as antifibrotic and anticancer drugs. However, differences in the locations of glycosylation sites, phosphorylation sites, and amino acids in the OAT and OATP structures lead to different substrates being transported to the liver, which ultimately results in their different roles in the liver. To date, few articles have addressed these aspects of OAT and OATP structures, and we study further the similarities and differences in their structures, tissue distribution, substrates, and roles in liver diseases.
Collapse
Affiliation(s)
- Ting-Ting Li
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical University, Zunyi 563100, Guizhou Province, China
| | - Jia-Xing An
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical University, Zunyi 563100, Guizhou Province, China
| | - Jing-Yu Xu
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical University, Zunyi 563100, Guizhou Province, China
| | - Bi-Guang Tuo
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical University, Zunyi 563100, Guizhou Province, China
| |
Collapse
|
16
|
Cao L, Massey IY, Feng H, Yang F. A Review of Cardiovascular Toxicity of Microcystins. Toxins (Basel) 2019; 11:toxins11090507. [PMID: 31480273 PMCID: PMC6783932 DOI: 10.3390/toxins11090507] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 12/30/2022] Open
Abstract
The mortality rate of cardiovascular diseases (CVD) in China is on the rise. The increasing burden of CVD in China has become a major public health problem. Cyanobacterial blooms have been recently considered a global environmental concern. Microcystins (MCs) are the secondary products of cyanobacteria metabolism and the most harmful cyanotoxin found in water bodies. Recent studies provide strong evidence of positive associations between MC exposure and cardiotoxicity, representing a threat to human cardiovascular health. This review focuses on the effects of MCs on the cardiovascular system and provides some evidence that CVD could be induced by MCs. We summarized the current knowledge of the cardiovascular toxicity of MCs, with regard to direct cardiovascular toxicity and indirect cardiovascular toxicity. Toxicity of MCs is mainly governed by the increasing level of reactive oxygen species (ROS), oxidative stress in mitochondria and endoplasmic reticulum, the inhibition activities of serine/threonine protein phosphatase 1 (PP1) and 2A (PP2A) and the destruction of cytoskeletons, which finally induce the occurrence of CVD. To protect human health from the threat of MCs, this paper also puts forward some directions for further research.
Collapse
Affiliation(s)
- Linghui Cao
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Isaac Yaw Massey
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Hai Feng
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Fei Yang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China.
| |
Collapse
|
17
|
Hausner EA, Elmore SA, Yang X. Overview of the Components of Cardiac Metabolism. Drug Metab Dispos 2019; 47:673-688. [PMID: 30967471 PMCID: PMC7333657 DOI: 10.1124/dmd.119.086611] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Metabolism in organs other than the liver and kidneys may play a significant role in how a specific organ responds to chemicals. The heart has metabolic capability for energy production and homeostasis. This homeostatic machinery can also process xenobiotics. Cardiac metabolism includes the expression of numerous organic anion transporters, organic cation transporters, organic carnitine (zwitterion) transporters, and ATP-binding cassette transporters. Expression and distribution of the transporters within the heart may vary, depending on the patient's age, disease, endocrine status, and various other factors. Several cytochrome P450 (P450) enzyme classes have been identified within the heart. The P450 hydroxylases and epoxygenases within the heart produce hydroxyeicosatetraneoic acids and epoxyeicosatrienoic acids, metabolites of arachidonic acid, which are critical in regulating homeostatic processes of the heart. The susceptibility of the cardiac P450 system to induction and inhibition from exogenous materials is an area of expanding knowledge, as are the metabolic processes of glucuronidation and sulfation in the heart. The susceptibility of various transcription factors and signaling pathways of the heart to disruption by xenobiotics is not fully characterized but is an area with implications for disruption of normal postnatal development, as well as modulation of adult cardiac health. There are knowledge gaps in the timelines of physiologic maturation and deterioration of cardiac metabolism. Cross-species characterization of cardiac-specific metabolism is needed for nonclinical work of optimum translational value to predict possible adverse effects, identify sensitive developmental windows for the design and conduct of informative nonclinical and clinical studies, and explore the possibilities of organ-specific therapeutics.
Collapse
Affiliation(s)
- Elizabeth A Hausner
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| | - Susan A Elmore
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| | - Xi Yang
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| |
Collapse
|
18
|
Li AA, Makris SL, Marty MS, Strauss V, Gilbert ME, Blacker A, Zorrilla LM, Coder PS, Hannas B, Lordi S, Schneider S. Practical considerations for developmental thyroid toxicity assessments: What's working, what's not, and how can we do better? Regul Toxicol Pharmacol 2019; 106:111-136. [PMID: 31018155 DOI: 10.1016/j.yrtph.2019.04.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 12/26/2022]
Abstract
Thyroid hormones (THs; T3 and T4) play a role in development of cardiovascular, reproductive, immune and nervous systems. Thus, interpretation of TH changes from rodent studies (during pregnancy, in fetuses, neonates, and adults) is critical in hazard characterization and risk assessment. A roundtable session at the 2017 Society of Toxicology (SOT) meeting brought together academic, industry and government scientists to share knowledge and different perspectives on technical and data interpretation issues. Data from a limited group of laboratories were compiled for technical discussions on TH measurements, including good practices for reliable serum TH data. Inter-laboratory historical control data, derived from immunoassays or mass spectrometry methods, revealed: 1) assay sensitivities vary within and across methodologies; 2) TH variability is similar across animal ages; 3) laboratories generally achieve sufficiently sensitive TH quantitation levels, although issues remain for lower levels of serum TH and TSH in fetuses and postnatal day 4 pups; thus, assay sensitivity is critical at these life stages. Best practices require detailed validation of rat serum TH measurements across ages to establish assay sensitivity and precision, and identify potential matrix effects. Finally, issues related to data interpretation for biological understanding and risk assessment were discussed, but their resolution remains elusive.
Collapse
Affiliation(s)
- Abby A Li
- Exponent Inc., 1010 14th Street, San Francisco, CA, 94114, USA.
| | - Susan L Makris
- US Environmental Protection Agency Office of Research and Development, 1200 Pennsylvania Ave NW 8623R, Washington, DC, 20460, USA.
| | - M Sue Marty
- The Dow Chemical Company, Toxicology & Environmental Research and Consulting, 1803 Building, Midland, MI, 48674, USA.
| | - Volker Strauss
- BASF SE, Experimental Toxicology and Ecology, 67056, Ludwigshafen, Germany.
| | - Mary E Gilbert
- US Environmental Protection Agency, National Health Environmental Effects Research Lab, 109 T.W. Alexander Drive, MD B105 05, Research Triangle Park, NC, 27711, USA.
| | - Ann Blacker
- Bayer CropScience, P.O. Box 12014, RTP, NC, 27709, USA.
| | | | - Pragati S Coder
- Charles River Laboratories, Developmental and Reproductive Toxicology, 1407 George Road, Ashland, OH, 44805, USA.
| | - Bethany Hannas
- The Dow Chemical Company, Toxicology & Environmental Research and Consulting, 1803 Building, Midland, MI, 48674, USA.
| | - Sheri Lordi
- Charles River Laboratories International, 251 Ballardvale Street, Wilmington, MA, 01887, USA.
| | - Steffen Schneider
- BASF SE, Experimental Toxicology and Ecology, 67056, Ludwigshafen, Germany.
| |
Collapse
|
19
|
Lofthouse EM, Torrens C, Manousopoulou A, Nahar M, Cleal JK, O'Kelly IM, Sengers BG, Garbis SD, Lewis RM. Ursodeoxycholic acid inhibits uptake and vasoconstrictor effects of taurocholate in human placenta. FASEB J 2019; 33:8211-8220. [PMID: 30922127 PMCID: PMC6593889 DOI: 10.1096/fj.201900015rr] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intrahepatic cholestasis of pregnancy (ICP) causes increased transfer of maternal bile acids to the fetus and an increased incidence of sudden fetal death. Treatment includes ursodeoxycholic acid (UDCA), but it is not clear if UDCA protects the fetus. This study explores the placental transport of the bile acid taurocholate (TC) by the organic anion-transporting polypeptide, (OATP)4A1, its effects on the placental proteome and vascular function, and how these are modified by UDCA. Various methodological approaches including placental villous fragments and Xenopus laevis oocytes were used to investigate UDCA transport. Placental perfusions and myography investigated the effect of TC on vasculature. The effects of acute TC exposure on placental tissue were investigated using quantitative proteomics. UDCA inhibited OATP4A1 activity in placental villous fragments and oocytes. TC induced vasoconstriction in placental and rat vasculature, which was attenuated by UDCA. Quantitative proteomic analysis of villous fragments showed direct effects of TC on multiple placental pathways, including oxidative stress and autophagy. The effects of TC on the placental proteome and vasculature demonstrate how bile acids may cause fetal distress in ICP. UDCA inhibition of OATP4A1 suggests it will protect the mother and fetus against the vascular effects of TC by inhibiting its cellular uptake. UDCA may protect the fetus in ICP by inhibiting OATP4A1-mediated bile acid transfer and TC-induced placental vasoconstriction. Understanding the physiologic mechanisms of UDCA may allow better therapeutic interventions to be designed specifically for the fetus in the future.-Lofthouse, E. M., Torrens, C., Manousopoulou, A., Nahar, M., Cleal, J. K., O'Kelly, I. M., Sengers, B. G., Garbis, S. D., Lewis, R. M. Ursodeoxycholic acid inhibits uptake and vasoconstrictor effects of taurocholate in human placenta.
Collapse
Affiliation(s)
- Emma M Lofthouse
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Christopher Torrens
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | | | - Monica Nahar
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jane K Cleal
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Faculty of Engineering, University of Southampton, Southampton, United Kingdom; and
| | - Ita M O'Kelly
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Bram G Sengers
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom.,Faculty of Engineering, University of Southampton, Southampton, United Kingdom; and
| | - Spiros D Garbis
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom.,Division of Biology and Biological Engineering, Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, California, USA
| | - Rohan M Lewis
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
20
|
Genome-wide association meta-analysis for total thyroid hormone levels in Croatian population. J Hum Genet 2019; 64:473-480. [PMID: 30824882 DOI: 10.1038/s10038-019-0586-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/24/2019] [Accepted: 02/12/2019] [Indexed: 12/14/2022]
Abstract
Thyroid hormones (THs) are key regulators of cellular growth, development, and metabolism. The thyroid gland secretes two THs, thyroxine (T4) and triiodothyronine (T3), into the plasma where they are almost all bound reversibly to plasma proteins. Free forms of THs are metabolically active, however, they represent a very small fraction of total TH levels. No genome-wide studies have been performed to date on total TH levels, comprising of protein-bound and free forms of THs. To detect genetic variants associated with total TH levels, we carried out the first GWAS meta-analysis of total T4 levels in 1121 individuals from two Croatian cohorts (Split and Korcula). We also performed GWAS analyses of total T3 levels in 577 individuals and T3/T4 ratio in 571 individuals from the Split cohort. The top association in GWAS meta-analysis of total T4 was detected for an intronic variant within SLC22A9 gene (rs12282281, P = 4.00 × 10-7). Within the same region, a genome-wide significant variant (rs11822642, P = 2.50 × 10-8) for the T3/T4 ratio was identified. SLC22A9 encodes for an organic anion transporter protein expressed predominantly in the liver and belongs to the superfamily of solute carriers (SLC), a large group of transport membrane proteins. The transport of THs across the plasma membrane in peripheral tissues is facilitated by the membrane proteins, and all TH transport proteins known to date belong to the same SLC superfamily as SLC22A9. These results suggest a potential role for SLC22A9 as a novel transporter protein of THs.
Collapse
|
21
|
Virili C, Antonelli A, Santaguida MG, Benvenga S, Centanni M. Gastrointestinal Malabsorption of Thyroxine. Endocr Rev 2019; 40:118-136. [PMID: 30476027 DOI: 10.1210/er.2018-00168] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023]
Abstract
Levothyroxine, a largely prescribed drug with a narrow therapeutic index, is often a lifelong treatment. The therapeutic efficacy of T4 may be marred by behavioral, pharmacologic, and pathologic issues acting as interfering factors. Despite a continuous search for an optimal T4 treatment, a significant number of patients fail to show a complete chemical and/or clinical response to this reference dose of T4. Gastrointestinal malabsorption of oral T4 represents an emerging cause of refractory hypothyroidism and may be more frequent than previously reputed. In this review, we examine the pharmacologic features of T4 preparations and their linkage with the intestinal absorption of the hormone. We have stressed the major biochemical and pharmacologic characteristics of T4 and its interaction with the putative transporter at the intestinal level. We have examined the interfering role of nutrients, foods, and drugs on T4 absorption at the gastric and intestinal levels. The impact of gastrointestinal disorders on T4 treatment efficacy has been also analyzed, in keeping with the site of action and the interfering mechanisms. Based on the evidence obtained from the literature, we also propose a schematic diagnostic workup for the most frequent and often hidden gastrointestinal diseases impairing T4 absorption.
Collapse
Affiliation(s)
- Camilla Virili
- Endocrinology Unit, Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome, Latina, Italy
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Maria Giulia Santaguida
- Endocrinology Unit, Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome, Latina, Italy.,Endocrinology Unit, AUSL Latina, Latina, Italy
| | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine, University of Messina, Policlinico Universitario G. Martino, Messina, Italy.,Master Program on Childhood, Adolescent and Women's Endocrine Health, University of Messina, Policlinico Universitario G. Martino, Messina, Italy.,Interdepartmental Program of Molecular and Clinical Endocrinology, and Women's Endocrine Health, University Hospital, Policlinico Universitario G. Martino, Messina, Italy
| | - Marco Centanni
- Endocrinology Unit, Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome, Latina, Italy.,Endocrinology Unit, AUSL Latina, Latina, Italy
| |
Collapse
|
22
|
Stepien BK, Huttner WB. Transport, Metabolism, and Function of Thyroid Hormones in the Developing Mammalian Brain. Front Endocrinol (Lausanne) 2019; 10:209. [PMID: 31001205 PMCID: PMC6456649 DOI: 10.3389/fendo.2019.00209] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022] Open
Abstract
Ever since the discovery of thyroid hormone deficiency as the primary cause of cretinism in the second half of the 19th century, the crucial role of thyroid hormone (TH) signaling in embryonic brain development has been established. However, the biological understanding of TH function in brain formation is far from complete, despite advances in treating thyroid function deficiency disorders. The pleiotropic nature of TH action makes it difficult to identify and study discrete roles of TH in various aspect of embryogenesis, including neurogenesis and brain maturation. These challenges notwithstanding, enormous progress has been achieved in understanding TH production and its regulation, their conversions and routes of entry into the developing mammalian brain. The endocrine environment has to adjust when an embryo ceases to rely solely on maternal source of hormones as its own thyroid gland develops and starts to produce endogenous TH. A number of mechanisms are in place to secure the proper delivery and action of TH with placenta, blood-brain interface, and choroid plexus as barriers of entry that need to selectively transport and modify these hormones thus controlling their active levels. Additionally, target cells also possess mechanisms to import, modify and bind TH to further fine-tune their action. A complex picture of a tightly regulated network of transport proteins, modifying enzymes, and receptors has emerged from the past studies. TH have been implicated in multiple processes related to brain formation in mammals-neuronal progenitor proliferation, neuronal migration, functional maturation, and survival-with their exact roles changing over developmental time. Given the plethora of effects thyroid hormones exert on various cell types at different developmental periods, the precise spatiotemporal regulation of their action is of crucial importance. In this review we summarize the current knowledge about TH delivery, conversions, and function in the developing mammalian brain. We also discuss their potential role in vertebrate brain evolution and offer future directions for research aimed at elucidating TH signaling in nervous system development.
Collapse
|
23
|
Genome-wide analyses identify a role for SLC17A4 and AADAT in thyroid hormone regulation. Nat Commun 2018; 9:4455. [PMID: 30367059 PMCID: PMC6203810 DOI: 10.1038/s41467-018-06356-1] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/31/2018] [Indexed: 12/20/2022] Open
Abstract
Thyroid dysfunction is an important public health problem, which affects 10% of the general population and increases the risk of cardiovascular morbidity and mortality. Many aspects of thyroid hormone regulation have only partly been elucidated, including its transport, metabolism, and genetic determinants. Here we report a large meta-analysis of genome-wide association studies for thyroid function and dysfunction, testing 8 million genetic variants in up to 72,167 individuals. One-hundred-and-nine independent genetic variants are associated with these traits. A genetic risk score, calculated to assess their combined effects on clinical end points, shows significant associations with increased risk of both overt (Graves’ disease) and subclinical thyroid disease, as well as clinical complications. By functional follow-up on selected signals, we identify a novel thyroid hormone transporter (SLC17A4) and a metabolizing enzyme (AADAT). Together, these results provide new knowledge about thyroid hormone physiology and disease, opening new possibilities for therapeutic targets. Thyroid dysfunction is a common public health problem and associated with cardiovascular co-morbidities. Here, the authors carry out genome-wide meta-analysis for thyroid hormone (TH) levels, hyper- and hypothyroidism and identify SLC17A4 as a TH transporter and AADAT as a TH metabolizing enzyme.
Collapse
|
24
|
Nakamura Y, Nakanishi T, Tamai I. Membrane Transporters Contributing to PGE 2 Distribution in Central Nervous System. Biol Pharm Bull 2018; 41:1337-1347. [DOI: 10.1248/bpb.b18-00169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoshinobu Nakamura
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Takeo Nakanishi
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| |
Collapse
|
25
|
Meyer Zu Schwabedissen HE, Ferreira C, Schaefer AM, Oufir M, Seibert I, Hamburger M, Tirona RG. Thyroid Hormones Are Transport Substrates and Transcriptional Regulators of Organic Anion Transporting Polypeptide 2B1. Mol Pharmacol 2018; 94:700-712. [PMID: 29735582 DOI: 10.1124/mol.117.111161] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/02/2018] [Indexed: 01/06/2025] Open
Abstract
Levothyroxine replacement therapy forms the cornerstone of hypothyroidism management. Variability in levothyroxine oral absorption may contribute to the well-recognized large interpatient differences in required dose. Moreover, levothyroxine-drug pharmacokinetic interactions are thought to be caused by altered oral bioavailability. Interestingly, little is known regarding the mechanisms contributing to levothyroxine absorption in the gastrointestinal tract. Here, we aimed to determine whether the intestinal drug uptake transporter organic anion transporting polypeptide 2B1 (OATP2B1) may be involved in facilitating intestinal absorption of thyroid hormones. We also explored whether thyroid hormones regulate OATP2B1 gene expression. In cultured Madin-Darby Canine Kidney II/OATP2B1 cells and in OATP2B1-transfected Caco-2 cells, thyroid hormones were found to inhibit OATP2B1-mediated uptake of estrone-3-sulfate. Competitive counter-flow experiments evaluating the influence on the cellular accumulation of estrone-3-sulfate in the steady state indicated that thyroid hormones were substrates of OATP2B1. Additional evidence that thyroid hormones were OATP2B1 substrates was provided by OATP2B1-dependent stimulation of thyroid hormone receptor activation in cell-based reporter assays. Bidirectional transport studies in intestinal Caco-2 cells showed net absorptive flux of thyroid hormones, which was attenuated by the presence of the OATP2B1 inhibitor, atorvastatin. In intestinal Caco-2 and LS180 cells, but not in liver Huh-7 or HepG2 cells, OATP2B1 expression was induced by treatment with thyroid hormones. Reporter gene assays revealed thyroid hormone receptor α-mediated transactivation of the SLCO2B1 1b and the SLCO2B1 1e promoters. We conclude that thyroid hormones are substrates and transcriptional regulators of OATP2B1. These insights provide a potential mechanistic basis for oral levothyroxine dose variability and drug interactions.
Collapse
Affiliation(s)
- Henriette E Meyer Zu Schwabedissen
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Celio Ferreira
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Anima M Schaefer
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Mouhssin Oufir
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Isabell Seibert
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Matthias Hamburger
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Rommel G Tirona
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| |
Collapse
|
26
|
Ferreira C, Prestin K, Hussner J, Zimmermann U, Meyer Zu Schwabedissen HE. PDZ domain containing protein 1 (PDZK1), a modulator of membrane proteins, is regulated by the nuclear receptor THRβ. Mol Cell Endocrinol 2018; 461:215-225. [PMID: 28928085 DOI: 10.1016/j.mce.2017.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/25/2017] [Accepted: 09/13/2017] [Indexed: 01/17/2023]
Abstract
Genome wide association studies revealed single nucleotide polymorphisms (SNP) located within the promoter of PDZ domain containing protein 1 (PDZK1) to be associated with serum uric acid levels. Since modulation of transporters and particularly of membrane proteins involved in uric acid handling by PDZK1 has previously been reported, the aim of this study was to analyze the impact of the polymorphisms rs1967017, rs1471633, and rs12129861 on promoter activity and thereby transcription of PDZK1. Cell-based reporter gene assays showed transactivation of the PDZK1-promoter by triiodothyronine mediated by thyroid hormone receptors (THR) α and β. In silico analysis verified localization of the polymorphism rs1967017 within the most likely THR binding site whose deletion reduced THR-mediated transactivation. Furthermore, our study shows regulation of PDZK1 by thyroid hormones, thereby providing a mechanistic basis for the previously reported associations between thyroid hormone status and uric acid homeostasis.
Collapse
Affiliation(s)
- Celio Ferreira
- Department of Pharmaceutical Sciences, Biopharmacy, University of Basel, 4056 Basel, Switzerland
| | - Katharina Prestin
- Department of Pharmaceutical Sciences, Biopharmacy, University of Basel, 4056 Basel, Switzerland
| | - Janine Hussner
- Department of Pharmaceutical Sciences, Biopharmacy, University of Basel, 4056 Basel, Switzerland
| | - Uwe Zimmermann
- Clinic for Urology, University Medicine Greifswald, Greifswald, Germany
| | | |
Collapse
|
27
|
Sasaki K, Tachikawa M, Uchida Y, Hirano S, Kadowaki F, Watanabe M, Ohtsuki S, Terasaki T. ATP-Binding Cassette Transporter A Subfamily 8 Is a Sinusoidal Efflux Transporter for Cholesterol and Taurocholate in Mouse and Human Liver. Mol Pharm 2018; 15:343-355. [DOI: 10.1021/acs.molpharmaceut.7b00679] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Kazunari Sasaki
- Membrane Transport
and Drug Targeting Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masanori Tachikawa
- Membrane Transport
and Drug Targeting Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Yasuo Uchida
- Membrane Transport
and Drug Targeting Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Satoshi Hirano
- Membrane Transport
and Drug Targeting Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Fumito Kadowaki
- Membrane Transport
and Drug Targeting Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Michitoshi Watanabe
- Membrane Transport
and Drug Targeting Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8555, Japan
| | - Tetsuya Terasaki
- Membrane Transport
and Drug Targeting Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| |
Collapse
|
28
|
Younis IR, Ahmed MA, Burman KD, Soldin OP, Jonklaas J. Stable Isotope Pharmacokinetic Studies Provide Insight into Effects of Age, Sex, and Weight on Levothyroxine Metabolism. Thyroid 2018. [DOI: 10.1089/thy.2017.0380 pmid: 29212434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Islam R. Younis
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Mariam A. Ahmed
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Kenneth D. Burman
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC
| | - Offie P. Soldin
- Departments of Medicine, Oncology, Physiology, and Biophysics, Georgetown University Medical Center, Washington, DC
| | - Jacqueline Jonklaas
- Division of Endocrinology, Georgetown University Medical Center, Washington, DC
| |
Collapse
|
29
|
Younis IR, Ahmed MA, Burman KD, Soldin OP, Jonklaas J. Stable Isotope Pharmacokinetic Studies Provide Insight into Effects of Age, Sex, and Weight on Levothyroxine Metabolism. Thyroid 2018; 28:41-49. [PMID: 29212434 PMCID: PMC5770123 DOI: 10.1089/thy.2017.0380] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND This study sought to determine whether levothyroxine pharmacokinetics (PKs) are affected by age, weight, and sex. METHODS A PK study was performed after administration of a tracer dose of carbon-13-labeled LT4 (13C-LT4). The study was conducted at an academic medical center. Adults of any age being treated with levothyroxine for hypothyroidism were enrolled in the study. A single dose of 13C-LT4 was administered. Eighteen serial plasma samples were collected. One sample was obtained before the 13C-LT4 dose, and the majority of the remaining samples were collected over the 120-hour period post dosing. 13C-LT4 concentration was quantified using liquid chromatography tandem mass spectrometry. PK analysis was conducted using a linear log trapezoidal non-compartmental analysis using Phoenix 6.4. RESULTS Eight males and 33 females with a median age of 50 years (range 22-78 years) and median weight of 65.9 kg (range 50-150 kg) were enrolled in the study. The median 13C-LT4 dose administered was 100 μg (range 70-300 μg). The median oral clearance rate (CL/F), apparent volume of distribution (V/F), time to peak concentration (Tmax), and dose-normalized peak concentration (Cmax) of 13C-LT4 were estimated to be 0.712 L/h, 164.9 L, 4 h, and 7.5 ng/L/μg, respectively. The dose-normalized area under the concentration-time curve from time 0 to 120 hours and half-life of the terminal distribution phase were 0.931 ng.h/mL/μg and 172.2 h, respectively. There was no significant difference in any 13C-LT4 PK parameter between patients aged >60 years (n = 10) and patients aged ≤60 years (n = 31), nor was there a relationship between age as a continuous variable and 13C-LT4 PK parameters. Sex only affected CL/F, V/F, and dose-normalized Cmax in univariate analyses. However, after adjusting for weight, sex was no longer a significant covariate. Weight was a significant predictor for CL/F, V/F and dose-normalized Cmax of 13C-LT4 in multivariate analyses. CONCLUSION Prior studies suggest that patient age affects levothyroxine dose requirement. This study did not identify an effect of age and suggests that age-related changes in levothyroxine pharmacokinetics may be mediated by age-related weight differences. Physicians should consider a patient's weight, rather than age, for estimating levothyroxine dosage requirement.
Collapse
Affiliation(s)
- Islam R. Younis
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Mariam A. Ahmed
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Kenneth D. Burman
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC
| | - Offie P. Soldin
- Departments of Medicine, Oncology, Physiology, and Biophysics, Georgetown University Medical Center, Washington, DC
| | - Jacqueline Jonklaas
- Division of Endocrinology, Georgetown University Medical Center, Washington, DC
| |
Collapse
|
30
|
Geyer J, Bakhaus K, Bernhardt R, Blaschka C, Dezhkam Y, Fietz D, Grosser G, Hartmann K, Hartmann MF, Neunzig J, Papadopoulos D, Sánchez-Guijo A, Scheiner-Bobis G, Schuler G, Shihan M, Wrenzycki C, Wudy SA, Bergmann M. The role of sulfated steroid hormones in reproductive processes. J Steroid Biochem Mol Biol 2017; 172:207-221. [PMID: 27392637 DOI: 10.1016/j.jsbmb.2016.07.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
Abstract
Sulfated steroid hormones, such as dehydroepiandrosterone sulfate or estrone-3-sulfate, have long been regarded as inactive metabolites as they cannot activate classical steroid receptors. Some of them are present in the blood circulation at quite high concentrations, but generally sulfated steroids exhibit low membrane permeation due to their hydrophilic properties. However, sulfated steroid hormones can actively be imported into specific target cells via uptake carriers, such as the sodium-dependent organic anion transporter SOAT, and, after hydrolysis by the steroid sulfatase (so-called sulfatase pathway), contribute to the overall regulation of steroid responsive organs. To investigate the biological significance of sulfated steroid hormones for reproductive processes in humans and animals, the research group "Sulfated Steroids in Reproduction" was established by the German Research Foundation DFG (FOR1369). Projects of this group deal with transport of sulfated steroids, sulfation of free steroids, desulfation by the steroid sulfatase, effects of sulfated steroids on steroid biosynthesis and membrane receptors as well as MS-based profiling of sulfated steroids in biological samples. This review and concept paper presents key findings from all these projects and provides a broad overview over the current research on sulfated steroid hormones in the field of reproduction.
Collapse
Affiliation(s)
- Joachim Geyer
- Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany.
| | - Katharina Bakhaus
- Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Rita Bernhardt
- Institute of Biochemistry, Saarland University, Saarbrücken, Germany
| | - Carina Blaschka
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Yaser Dezhkam
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Daniela Fietz
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Gary Grosser
- Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Katja Hartmann
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Michaela F Hartmann
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Jens Neunzig
- Institute of Biochemistry, Saarland University, Saarbrücken, Germany
| | - Dimitrios Papadopoulos
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - Alberto Sánchez-Guijo
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - Gerhard Schuler
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Mazen Shihan
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - Christine Wrenzycki
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Stefan A Wudy
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Martin Bergmann
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| |
Collapse
|
31
|
Ban MJ, Ji SH, Lee CK, Bae SB, Kim HJ, Ahn TS, Lee MS, Baek MJ, Jeong D. Solute carrier organic anion transporter family member 4A1 (SLCO4A1) as a prognosis marker of colorectal cancer. J Cancer Res Clin Oncol 2017; 143:1437-1447. [PMID: 28378090 DOI: 10.1007/s00432-017-2393-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/12/2017] [Indexed: 12/29/2022]
Abstract
PURPOSE Solute carrier organic anion transporter family member 4A1 (SLCO4A1) is involved in the transport of various compounds, including sugars, bile salts, organic acids, metal ions, amine compounds, and estrogen. SLCO4A1 is highly expressed in several cancers and a gender bias has been observed in colorectal cancer (CRC). We investigated SLCO4A1 expression, its prognostic value in patients with CRC, and its role in CRC cell proliferation and metastasis. METHODS SLCO4A1 expression was assessed by immunohistochemistry (IHC) on specimens from 84 patients with CRC. The association of SLCO4A1 expression with clinicopathological features was examined. To confirm the biological role of SLCO4A1 in CRC, four CRC cell lines expressing SLCO4A1 were used and SLCO4A1 expression was knocked down by siRNA. Cell proliferation, MTT, migration, invasion, and semisolid agar colony formation assays were performed. RESULTS SLCO4A1 was overexpressed in 32% of the CRC samples. SLCO4A1 overexpression and pathologic T stage were independent prognostic factors of decreased survival (P = 0.021). Kaplan-Meier analysis indicated a decreased cumulative survival for patients highly expressing SLCO4A1 compared to patients showing low SLCO4A1 expression (Log-rank test, P = 0.025). In cell lines, SLCO4A1 knockdown resulted in a significant decrease of viability, invasion, and migration when compared to control cells. Semisolid colony formation assay indicated that SLCO4A1-knocked down cells presented poor carcinogenic abilities compared to control cells. CONCLUSIONS SLCO4A1 may be a valuable marker of poor prognostic for CRC. Furthermore, SLCO4A1 plays an important role in CRC cell proliferation, migration, invasion, and carcinogenesis.
Collapse
Affiliation(s)
- Myung Jin Ban
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea
- Department of Medicine, The Graduate School of Yonsei University, Seoul, Republic of Korea
| | - Sang Hee Ji
- Soonchunhyang Medical Science Research Institute, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea
| | - Chi-Kyu Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea
| | - Sang Byung Bae
- Department of Oncology, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea
| | - Han Jo Kim
- Department of Oncology, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea
| | - Tae Sung Ahn
- Department of Surgery, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea
| | - Moon Soo Lee
- Department of Surgery, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea
| | - Moo-Jun Baek
- Department of Surgery, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea.
| | - Dongjun Jeong
- Soonchunhyang Medical Science Research Institute, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea.
- Department of Pathology, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam-do, Republic of Korea.
| |
Collapse
|
32
|
Abstract
Transporters in proximal renal tubules contribute to the disposition of numerous drugs. Furthermore, the molecular mechanisms of tubular secretion have been progressively elucidated during the past decades. Organic anions tend to be secreted by the transport proteins OAT1, OAT3 and OATP4C1 on the basolateral side of tubular cells, and multidrug resistance protein (MRP) 2, MRP4, OATP1A2 and breast cancer resistance protein (BCRP) on the apical side. Organic cations are secreted by organic cation transporter (OCT) 2 on the basolateral side, and multidrug and toxic compound extrusion (MATE) proteins MATE1, MATE2/2-K, P-glycoprotein, organic cation and carnitine transporter (OCTN) 1 and OCTN2 on the apical side. Significant drug-drug interactions (DDIs) may affect any of these transporters, altering the clearance and, consequently, the efficacy and/or toxicity of substrate drugs. Interactions at the level of basolateral transporters typically decrease the clearance of the victim drug, causing higher systemic exposure. Interactions at the apical level can also lower drug clearance, but may be associated with higher renal toxicity, due to intracellular accumulation. Whereas the importance of glomerular filtration in drug disposition is largely appreciated among clinicians, DDIs involving renal transporters are less well recognized. This review summarizes current knowledge on the roles, quantitative importance and clinical relevance of these transporters in drug therapy. It proposes an approach based on substrate-inhibitor associations for predicting potential tubular-based DDIs and preventing their adverse consequences. We provide a comprehensive list of known drug interactions with renally-expressed transporters. While many of these interactions have limited clinical consequences, some involving high-risk drugs (e.g. methotrexate) definitely deserve the attention of prescribers.
Collapse
Affiliation(s)
- Anton Ivanyuk
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland.
| | - Françoise Livio
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland
| | - Jérôme Biollaz
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland
| | - Thierry Buclin
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland
| |
Collapse
|
33
|
Zhou F, Zhu L, Wang K, Murray M. Recent advance in the pharmacogenomics of human Solute Carrier Transporters (SLCs) in drug disposition. Adv Drug Deliv Rev 2017; 116:21-36. [PMID: 27320645 DOI: 10.1016/j.addr.2016.06.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/01/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
Drug pharmacokinetics is influenced by the function of metabolising enzymes and influx/efflux transporters. Genetic variability of these genes is known to impact on clinical therapies. Solute Carrier Transporters (SLCs) are the primary influx transporters responsible for the cellular uptake of drug molecules, which consequently, impact on drug efficacy and toxicity. The Organic Anion Transporting Polypeptides (OATPs), Organic Anion Transporters (OATs) and Organic Cation Transporters (OCTs/OCTNs) are the most important SLCs involved in drug disposition. The information regarding the influence of SLC polymorphisms on drug pharmacokinetics is limited and remains a hot topic of pharmaceutical research. This review summarises the recent advance in the pharmacogenomics of SLCs with an emphasis on human OATPs, OATs and OCTs/OCTNs. Our current appreciation of the degree of variability in these transporters may contribute to better understanding the inter-patient variation of therapies and thus, guide the optimisation of clinical treatments.
Collapse
|
34
|
Ghoneim RH, Kojovic D, Piquette-Miller M. Impact of endotoxin on the expression of drug transporters in the placenta of HIV-1 transgenic (HIV-Tg) rats. Eur J Pharm Sci 2017; 102:94-102. [PMID: 28274777 DOI: 10.1016/j.ejps.2017.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/06/2017] [Accepted: 03/03/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Inflammatory responses in HIV (+) patients may be exacerbated due to reports of subclinical endotoxemia and existing immune dysregulation. As inflammation has been reported to mediate changes in the expression of transporters, this could be potentiated in pregnant HIV (+) women. Similar to humans, the HIV-Tg rat model develops immune dysregulation and chronic AIDS-like conditions. Our objective was to examine the expression of placental drug transporters in HIV-Tg rats in response to low-dose endotoxin. METHODS Pregnant HIV-Tg rats or wild-type littermates (WT) were treated with low dose bacterial endotoxin 0.1mg/kg (n=8) or 0.25mg/kg (n=4-6) on GD18 and placentas were harvested 18h later. Placental and hepatic expression of transporters and cytokines were examined using qRT-PCR and Western blotting. RESULTS As compared to WT, endotoxin administration increased the hepatic and placental expression of IL-6 and TNF-α to a greater extent in HIV-Tg rats (p<0.05). The placental mRNA and protein expression of Abcb1a and Slco2b1 was significantly decreased in endotoxin-treated HIV-Tg but not WT rats and downregulation of Slco4a1 mRNA was more pronounced in the HIV-Tg group (p<0.05). These changes significantly correlated with the placental expression of pro-inflammatory cytokines. Abcc3 mRNA expression was increased in the placenta of endotoxin treated WT rats only, while placental expression of Abcc1, Abcc2 and Abcc4 was not significantly affected in both WT and HIV rats. Endotoxin imposed a pronounced downregulation in the hepatic expression of Abcb1a, Abcc2, Abcc4, Abcg2, Slco1a1, Slco1b2 and Slc29a1 in both HIV-Tg and WT rats; however, Abcb1b expression was increased in HIV but not WT rats. CONCLUSION Our results indicate that low-dose endotoxin resulted in an augmented inflammatory response in HIV-Tg rats accompanied with significant changes in the placental expression of several drug transporters. Our data suggests that subclinical endotoxemia and other co-existing infections may alter the placental transfer of drugs in the HIV population.
Collapse
Affiliation(s)
- Ragia H Ghoneim
- Leslie Dan Faculty of Pharmacy, University of Toronto, ON, Canada
| | - Dea Kojovic
- Leslie Dan Faculty of Pharmacy, University of Toronto, ON, Canada
| | | |
Collapse
|
35
|
Affiliation(s)
- Eleni Kotsampasakou
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14 1090 Vienna Austria
| | - Gerhard F. Ecker
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14 1090 Vienna Austria
| |
Collapse
|
36
|
Kovacsics D, Patik I, Özvegy-Laczka C. The role of organic anion transporting polypeptides in drug absorption, distribution, excretion and drug-drug interactions. Expert Opin Drug Metab Toxicol 2016; 13:409-424. [PMID: 27783531 DOI: 10.1080/17425255.2017.1253679] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION The in vivo fate and effectiveness of a drug depends highly on its absorption, distribution, metabolism, excretion and toxicity (ADME-Tox). Organic anion transporting polypeptides (OATPs) are membrane proteins involved in the cellular uptake of various organic compounds, including clinically used drugs. Since OATPs are significant players in drug absorption and distribution, modulation of OATP function via pharmacotherapy with OATP substrates/inhibitors, or modulation of their expression, affects drug pharmacokinetics. Given their cancer-specific expression, OATPs may also be considered anticancer drug targets. Areas covered: We describe the human OATP family, discussing clinically relevant consequences of altered OATP function. We offer a critical analysis of published data on the role of OATPs in ADME and in drug-drug interactions, especially focusing on OATP1A2, 1B1, 1B3 and 2B1. Expert opinion: Four members of the OATP family, 1A2, 1B1, 1B3 and 2B1, have been characterized in detail. As biochemical and pharmacological knowledge on the other OATPs is lacking, it seems timely to direct research efforts towards developing the experimental framework needed to investigate the transport mechanism and substrate specificity of the poorly described OATPs. In addition, elucidating the role of OATPs in tumor development and therapy response are critical avenues for further research.
Collapse
Affiliation(s)
- Daniella Kovacsics
- a Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - Izabel Patik
- a Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - Csilla Özvegy-Laczka
- a Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| |
Collapse
|
37
|
Gnocchi D, Steffensen KR, Bruscalupi G, Parini P. Emerging role of thyroid hormone metabolites. Acta Physiol (Oxf) 2016; 217:184-216. [PMID: 26748938 DOI: 10.1111/apha.12648] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 07/28/2015] [Accepted: 01/03/2016] [Indexed: 12/15/2022]
Abstract
Thyroid hormones (THs) are essential for the regulation of development and metabolism in key organs. THs produce biological effects both by directly affecting gene expression through the interaction with nuclear receptors (genomic effects) and by activating protein kinases and/or ion channels (short-term effects). Such activations can be either direct, in the case of ion channels, or mediated by membrane or cytoplasmic receptors. Short-term-activated signalling pathways often play a role in the regulation of genomic effects. Several TH intermediate metabolites, which were previously considered without biological activity, have now been associated with a broad range of actions, mostly attributable to short-term effects. Here, we give an overview of the physiological roles and mechanisms of action of THs, focusing on the emerging position that TH metabolites are acquiring as important regulators of physiology and metabolism.
Collapse
Affiliation(s)
- D. Gnocchi
- Division of Clinical Chemistry; Department of Laboratory Medicine; Karolinska Institutet at Karolinska University Hospital Huddinge; Stockholm Sweden
| | - K. R. Steffensen
- Division of Clinical Chemistry; Department of Laboratory Medicine; Karolinska Institutet at Karolinska University Hospital Huddinge; Stockholm Sweden
| | - G. Bruscalupi
- Department of Biology and Biotechnology ‘Charles Darwin’; Sapienza University of Rome; Rome Italy
| | - P. Parini
- Division of Clinical Chemistry; Department of Laboratory Medicine; Karolinska Institutet at Karolinska University Hospital Huddinge; Stockholm Sweden
- Metabolism Unit; Department of Medicine; Karolinska Institutet at Karolinska University Hospital Huddinge; Stockholm Sweden
| |
Collapse
|
38
|
|
39
|
Abstract
The cellular influx and efflux of thyroid hormones are facilitated by transmembrane protein transporters. Of these transporters, monocarboxylate transporter 8 (MCT8) is the only one specific for the transport of thyroid hormones and some of their derivatives. Mutations in SLC16A2, the gene that encodes MCT8, lead to an X-linked syndrome with severe neurological impairment and altered concentrations of thyroid hormones. Histopathological analysis of brain tissue from patients who have impaired MCT8 function indicates that brain lesions start prenatally, and are most probably the result of cerebral hypothyroidism. A Slc16a2 knockout mouse model has revealed that Mct8 is an important mediator of thyroid hormone transport, especially T3, through the blood-brain barrier. However, unlike humans with an MCT8 deficiency, these mice do not have neurological impairment. One explanation for this discrepancy could be differences in expression of the T4 transporter OATP1C1 in the blood-brain barrier; OATP1C1 is more abundant in rodents than in primates and permits the passage of T4 in the absence of T3 transport, thus preventing full cerebral hypothyroidism. In this Review, we discuss the relevance of thyroid hormone transporters in health and disease, with a particular focus on the pathophysiology of MCT8 mutations.
Collapse
Affiliation(s)
- Juan Bernal
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Arturo Duperier 4, 28029 Madrid, Spain
| | - Ana Guadaño-Ferraz
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Arturo Duperier 4, 28029 Madrid, Spain
| | - Beatriz Morte
- Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Arturo Duperier 4, 28029 Madrid, Spain
| |
Collapse
|
40
|
Medici M, Visser WE, Visser TJ, Peeters RP. Genetic determination of the hypothalamic-pituitary-thyroid axis: where do we stand? Endocr Rev 2015; 36:214-44. [PMID: 25751422 DOI: 10.1210/er.2014-1081] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
For a long time it has been known that both hypo- and hyperthyroidism are associated with an increased risk of morbidity and mortality. In recent years, it has also become clear that minor variations in thyroid function, including subclinical dysfunction and variation in thyroid function within the reference range, can have important effects on clinical endpoints, such as bone mineral density, depression, metabolic syndrome, and cardiovascular mortality. Serum thyroid parameters show substantial interindividual variability, whereas the intraindividual variability lies within a narrow range. This suggests that every individual has a unique hypothalamus-pituitary-thyroid axis setpoint that is mainly determined by genetic factors, and this heritability has been estimated to be 40-60%. Various mutations in thyroid hormone pathway genes have been identified in persons with thyroid dysfunction or altered thyroid function tests. Because these causes are rare, many candidate gene and linkage studies have been performed over the years to identify more common variants (polymorphisms) associated with thyroid (dys)function, but only a limited number of consistent associations have been found. However, in the past 5 years, advances in genetic research have led to the identification of a large number of new candidate genes. In this review, we provide an overview of the current knowledge about the polygenic basis of thyroid (dys)function. This includes new candidate genes identified by genome-wide approaches, what insights these genes provide into the genetic basis of thyroid (dys)function, and which new techniques will help to further decipher the genetic basis of thyroid (dys)function in the near future.
Collapse
Affiliation(s)
- Marco Medici
- Rotterdam Thyroid Center, Department of Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
41
|
Lu J, Michaud V, Moya LGG, Gaudette F, Leung YH, Turgeon J. Effects of β-blockers and tricyclic antidepressants on the activity of human organic anion transporting polypeptide 1A2 (OATP1A2). J Pharmacol Exp Ther 2015; 352:552-8. [PMID: 25563901 DOI: 10.1124/jpet.114.219287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
The organic anion transporting polypeptide 1A2 (OATP1A2), a membrane drug transporter expressed on important organs (such as the brain, kidney, and intestine) may be a key element in the disposition of drugs. Previous studies demonstrated that it could transport a broad spectrum of substrates, including endogenous molecules and clinically relevant drugs, such as several β-blockers and 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors. The primary objective of this study was to investigate OATP1A2 transport activity using rosuvastatin as a probe substrate and evaluate competitive inhibition of its transport by β-blockers. Rosuvastatin transport was saturable, with a Km of 60.2 µM. With the exception of carvedilol (IC50 of 3.2 µM), all of the other β-blockers that were evaluated had a small or insignificant effect on OATP1A2-mediated uptake of rosuvastatin. Carvedilol differs from the other β-blockers by the tricyclic moiety in its chemical structure. As a secondary objective, the transport of a series of tricyclic compounds by OATP1A2 and their potential for rosuvastatin transport inhibition were evaluated. Tricyclic compounds were not OATP1A2 substrates. On the other hand, tricyclic compounds with a short aliphatic amine chain inhibited OATP1A2-mediated rosuvastatin transport. Our data suggest that these drugs may modulate the transport of OATP1A2 substrates and may affect drug actions.
Collapse
Affiliation(s)
- Jennifer Lu
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., F.G., Y.H.L., J.T.); and Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., Y.H.L., J.T.)
| | - Veronique Michaud
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., F.G., Y.H.L., J.T.); and Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., Y.H.L., J.T.)
| | - Liliam Gabriela Guilarte Moya
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., F.G., Y.H.L., J.T.); and Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., Y.H.L., J.T.)
| | - Fleur Gaudette
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., F.G., Y.H.L., J.T.); and Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., Y.H.L., J.T.)
| | - Yat Hei Leung
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., F.G., Y.H.L., J.T.); and Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., Y.H.L., J.T.)
| | - Jacques Turgeon
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., F.G., Y.H.L., J.T.); and Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada (J.L., V.M., L.G.G.M., Y.H.L., J.T.)
| |
Collapse
|
42
|
Nakanishi T, Tamai I. Putative roles of organic anion transporting polypeptides (OATPs) in cell survival and progression of human cancers. Biopharm Drug Dispos 2014; 35:463-84. [DOI: 10.1002/bdd.1915] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 08/01/2014] [Accepted: 08/12/2014] [Indexed: 01/19/2023]
Affiliation(s)
- Takeo Nakanishi
- Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences; Kanazawa University; Kakuma-machi Kanazawa 920-1192 Japan
| | - Ikumi Tamai
- Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences; Kanazawa University; Kakuma-machi Kanazawa 920-1192 Japan
| |
Collapse
|
43
|
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: 3.7] [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
|
44
|
Radulović Z, Porter LM, Kim TK, Mulenga A. Comparative bioinformatics, temporal and spatial expression analyses of Ixodes scapularis organic anion transporting polypeptides. Ticks Tick Borne Dis 2014; 5:287-98. [PMID: 24582512 DOI: 10.1016/j.ttbdis.2013.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 12/01/2013] [Accepted: 12/01/2013] [Indexed: 01/06/2023]
Abstract
Organic anion-transporting polypeptides (Oatps) are an integral part of the detoxification mechanism in vertebrates and invertebrates. These cell surface proteins are involved in mediating the sodium-independent uptake and/or distribution of a broad array of organic amphipathic compounds and xenobiotic drugs. This study describes bioinformatics and biological characterization of 9 Oatp sequences in the Ixodes scapularis genome. These sequences have been annotated on the basis of 12 transmembrane domains, consensus motif D-X-RW-(I,V)-GAWW-X-G-(F,L)-L, and 11 conserved cysteine amino acid residues in the large extracellular loop 5 that characterize the Oatp superfamily. Ixodes scapularis Oatps may regulate non-redundant cross-tick species conserved functions in that they did not cluster as a monolithic group on the phylogeny tree and that they have orthologs in other ticks. Phylogeny clustering patterns also suggest that some tick Oatp sequences transport substrates that are similar to those of body louse, mosquito, eye worm, and filarial worm Oatps. Semi-quantitative RT-PCR analysis demonstrated that all 9 I. scapularis Oatp sequences were expressed during tick feeding. Ixodes scapularis Oatp genes potentially regulate functions during early and/or late-stage tick feeding as revealed by normalized mRNA profiles. Normalized transcript abundance indicates that I. scapularis Oatp genes are strongly expressed in unfed ticks during the first 24h of feeding and/or at the end of the tick feeding process. Except for 2 I. scapularis Oatps, which were expressed in the salivary glands and ovaries, all other genes were expressed in all tested organs, suggesting the significance of I. scapularis Oatps in maintaining tick homeostasis. Different I. scapularis Oatp mRNA expression patterns were detected and discussed with reference to different physiological states of unfed and feeding ticks.
Collapse
Affiliation(s)
- Zeljko Radulović
- Texas A & M University AgriLife Research, Department of Entomology, 2475 TAMU, College Station, TX 77843, USA
| | - Lindsay M Porter
- Texas A & M University AgriLife Research, Department of Entomology, 2475 TAMU, College Station, TX 77843, USA
| | - Tae K Kim
- Texas A & M University AgriLife Research, Department of Entomology, 2475 TAMU, College Station, TX 77843, USA
| | - Albert Mulenga
- Texas A & M University AgriLife Research, Department of Entomology, 2475 TAMU, College Station, TX 77843, USA.
| |
Collapse
|
45
|
Functional Expression of Drug Transporters in Glial Cells. PHARMACOLOGY OF THE BLOOD BRAIN BARRIER: TARGETING CNS DISORDERS 2014; 71:45-111. [DOI: 10.1016/bs.apha.2014.06.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
46
|
Popovic M, Zaja R, Fent K, Smital T. Molecular characterization of zebrafish Oatp1d1 (Slco1d1), a novel organic anion-transporting polypeptide. J Biol Chem 2013; 288:33894-33911. [PMID: 24126916 DOI: 10.1074/jbc.m113.518506] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The organic anion-transporting polypeptide (OATP/Oatp) superfamily includes a group of polyspecific transporters that mediate transport of large amphipathic, mostly anionic molecules across cell membranes of eukaryotes. OATPs/Oatps are involved in the disposition and elimination of numerous physiological and foreign compounds. However, in non-mammalian species, the functional properties of Oatps remain unknown. We aimed to elucidate the role of Oatp1d1 in zebrafish to gain insights into the functional and structural evolution of the OATP1/Oatp1 superfamily. We show that diversification of the OATP1/Oatp1 family occurs after the emergence of jawed fish and that the OATP1A/Oatp1a and OATP1B/Oatp1b subfamilies appeared at the root of tetrapods. The Oatp1d subfamily emerged in teleosts and is absent in tetrapods. The zebrafish Oatp1d1 is similar to mammalian OATP1A/Oatp1a and OATP1B/Oatp1b members, with the main physiological role in transport and balance of steroid hormones. Oatp1d1 activity is dependent upon pH gradient, which could indicate bicarbonate exchange as a mode of transport. Our analysis of evolutionary conservation and structural properties revealed that (i) His-79 in intracellular loop 3 is conserved within OATP1/Oatp1 family and is crucial for the transport activity; (ii) N-glycosylation impacts membrane targeting and is conserved within the OATP1/Oatp1 family with Asn-122, Asn-133, Asn-499, and Asn-512 residues involved; (iii) the evolutionarily conserved cholesterol recognition interaction amino acid consensus motif is important for membrane localization; and (iv) Oatp1d1 is present in dimeric and possibly oligomeric form in the cell membrane. In conclusion, we describe the first detailed characterization of a new Oatp transporter in zebrafish, offering important insights into the functional evolution of the OATP1/Oatp1 family and the physiological role of Oatp1d1.
Collapse
Affiliation(s)
- Marta Popovic
- Laboratory for Molecular Ecotoxicology, Division for Marine and Environmental Research, Rudjer Boskovic Institute, Bijenicka 54, 10 000 Zagreb, Croatia
| | - Roko Zaja
- Laboratory for Molecular Ecotoxicology, Division for Marine and Environmental Research, Rudjer Boskovic Institute, Bijenicka 54, 10 000 Zagreb, Croatia
| | - Karl Fent
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Gründenstrasse 40, CH-4132 Muttenz, Switzerland; Department of Environmental System Sciences, Swiss Federal Institute of Technology (ETHZ), CH-8092 Zürich, Switzerland
| | - Tvrtko Smital
- Laboratory for Molecular Ecotoxicology, Division for Marine and Environmental Research, Rudjer Boskovic Institute, Bijenicka 54, 10 000 Zagreb, Croatia.
| |
Collapse
|
47
|
Muzzio AM, Noyes PD, Stapleton HM, Lema SC. Tissue distribution and thyroid hormone effects on mRNA abundance for membrane transporters Mct8, Mct10, and organic anion-transporting polypeptides (Oatps) in a teleost fish. Comp Biochem Physiol A Mol Integr Physiol 2013; 167:77-89. [PMID: 24113777 DOI: 10.1016/j.cbpa.2013.09.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 11/18/2022]
Abstract
Many of the actions of thyroid hormones (THs) occur via TH binding to intracellular receptors. Although it was long thought that THs diffused passively across plasma membranes, it is now recognized that cellular entry is mediated by a variety of membrane transporter proteins. In this study, we identified cDNAs encoding the TH transporters monocarboxylate transferases 8 (mct8) and 10 (mct10) as well as eight distinct organic anion-transporting polypeptide (oatp) proteins from fathead minnow (Pimephales promelas). Analysis of the tissue distribution of transporter mRNAs revealed that mct8 and mct10 transcripts were both abundant in liver, but also present at lower levels in brain, gonad and other tissues. Transcripts encoding oatp1c1 were highly abundant in brain, liver and gonad, and exhibited significant sex differences in the liver and gonad. Treatment of adult male minnows with 3,5,3'-triiodothyronine (T3) or the goitrogen methimazole altered gene transcript abundance for several transporters. Fish given exogenous T3 had reduced mct8 and oapt1c1 mRNA levels in the liver compared to methimazole-treated fish. In the brain, transcripts for mct8, mct10, oatp2b1, and oatp3a1 were each reduced in abundance in fish with elevated T3. As a whole, these results provide evidence that TH status influences the transcriptional dynamics of mct8, mct10 and several Oatp genes including oatp1c1 in teleost fish.
Collapse
Affiliation(s)
- Amanda M Muzzio
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, USA
| | | | | | | |
Collapse
|
48
|
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: 282] [Impact Index Per Article: 23.5] [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
|
49
|
Vasilopoulou E, Loubière LS, Heuer H, Trajkovic-Arsic M, Darras VM, Visser TJ, Lash GE, Whitley GS, McCabe CJ, Franklyn JA, Kilby MD, Chan SY. Monocarboxylate transporter 8 modulates the viability and invasive capacity of human placental cells and fetoplacental growth in mice. PLoS One 2013; 8:e65402. [PMID: 23776477 PMCID: PMC3680392 DOI: 10.1371/journal.pone.0065402] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 04/25/2013] [Indexed: 11/22/2022] Open
Abstract
Monocarboxylate transporter 8 (MCT8) is a well-established thyroid hormone (TH) transporter. In humans, MCT8 mutations result in changes in circulating TH concentrations and X-linked severe global neurodevelopmental delay. MCT8 is expressed in the human placenta throughout gestation, with increased expression in trophoblast cells from growth-restricted pregnancies. We postulate that MCT8 plays an important role in placental development and transplacental TH transport. We investigated the effect of altering MCT8 expression in human trophoblast in vitro and in a Mct8 knockout mouse model. Silencing of endogenous MCT8 reduced T3 uptake into human extravillous trophoblast-like cells (SGHPL-4; 40%, P<0.05) and primary cytotrophoblast (15%, P<0.05). MCT8 over-expression transiently increased T3 uptake (SGHPL-4∶30%, P<0.05; cytotrophoblast: 15%, P<0.05). Silencing MCT8 did not significantly affect SGHPL-4 invasion, but with MCT8 over-expression T3 treatment promoted invasion compared with no T3 (3.3-fold; P<0.05). Furthermore, MCT8 silencing increased cytotrophoblast viability (∼20%, P<0.05) and MCT8 over-expression reduced cytotrophoblast viability independently of T3 (∼20%, P<0.05). In vivo, Mct8 knockout reduced fetal:placental weight ratios compared with wild-type controls at gestational day 18 (25%, P<0.05) but absolute fetal and placental weights were not significantly different. The volume fraction of the labyrinthine zone of the placenta, which facilitates maternal-fetal exchange, was reduced in Mct8 knockout placentae (10%, P<0.05). However, there was no effect on mouse placental cell proliferation in vivo. We conclude that MCT8 makes a significant contribution to T3 uptake into human trophoblast cells and has a role in modulating human trophoblast cell invasion and viability. In mice, Mct8 knockout has subtle effects upon fetoplacental growth and does not significantly affect placental cell viability probably due to compensatory mechanisms in vivo.
Collapse
Affiliation(s)
- Elisavet Vasilopoulou
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Laurence S. Loubière
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Heike Heuer
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany
| | | | - Veerle M. Darras
- Laboratory of Comparative Endocrinology, Katholieke Universiteit, Leuven, Belgium
| | | | - Gendie E. Lash
- Reproductive and Vascular Biology Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Guy S. Whitley
- Division of Biomedical Sciences, St George’s University of London, London, United Kingdom
| | - Christopher J. McCabe
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jayne A. Franklyn
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Mark D. Kilby
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shiao Y. Chan
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Yang W, Zhang Y, Fu F, Li R. High-resolution array-comparative genomic hybridization profiling reveals 20q13.33 alterations associated with ovarian endometriosis. Gynecol Endocrinol 2013; 29:603-7. [PMID: 23656391 DOI: 10.3109/09513590.2013.788632] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE The purpose of this study is to investigate the potential genetic alterations at DNA level in patients with ovarian endometriosis by high-resolution array-based comparative genomic hybridization (array-CGH) analysis. METHODS Following the laparoscopic surgical and the post-operative pathological examination, genomic DNA was extracted from endometriomas of 11 women with endometriosis and endometrial tissue of the controls and analyzed by array-CGH. Real-time PCR was used for confirmation the result of array-CGH analysis and detected the DNA copy number variations of the eutopic endometrium from the five patients with the duplication in 20q13.33 region. RESULTS All 11 patients with ovarian endometriosis were diagnosed through the laparoscopic surgical and the post-operative pathological examination. We found occurrence of genomic duplication at 20q13.33 chromosomal region with gain of GATA5 and SLCO4A1 genes in 5 of 11 endometriomas from patients. CONCLUSION The results of the present study suggest that there was 20q13.33 duplication in women with ovarian endometriosis. This effect might be due to the alterations of GATA5 and SLCO4A1 genes in the gain region, through involving the metabolism of the steroid hormone.
Collapse
Affiliation(s)
- Wenqing Yang
- Department of Obstetrics and Gynecology, Xiangya Hospital of Central South University, Changsha, China
| | | | | | | |
Collapse
|