1
|
Kumari P, Lungu-Mitea S, Novák J, Hilscherová K. Advancing in vitro assessment of iodide uptake inhibition: integrating a novel biotransformation pretreatment step. Arch Toxicol 2025:10.1007/s00204-025-04034-y. [PMID: 40355721 DOI: 10.1007/s00204-025-04034-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/19/2025] [Indexed: 05/14/2025]
Abstract
Thyroid hormones (TH) are essential for vertebrate development, growth, and metabolism. The increasing prevalence of anthropogenic chemicals with TH-disrupting potential highlights the urgent need for advanced methods to assess their impact on TH homeostasis. Inhibition of the sodium-iodide symporter (NIS) has been identified as a key molecular initiating event disrupting the TH system across species, with significant relevance for diagnostic and therapeutic applications in various carcinomas. This study presents in vitro bioassays for evaluating the effects of compounds on iodide uptake into cells, a critical step in TH production mediated by NIS. Two novel stably transfected human cell lines overexpressing human NIS were employed along with a rat thyroid cell model FRTL-5, using colorimetric Sandell-Kolthoff (SK) reaction for iodide detection. The results from 23 model compounds demonstrate comparability across various in vitro models and radioactivity-based assays. To enhance physiological relevance, an external biotransformation system (BTS) was integrated and optimized for live-cell compatibility without inducing cytotoxicity or interfering with the assay. Compounds identified as NIS inhibitors were evaluated using the BTS-augmented assay, which revealed that metabolic activity mitigated the inhibitory effects of some chemicals. The augmented assay exhibited strong concordance with in vivo and in silico biotransformation data. Protein sequence alignment confirmed high conservation of NIS functional domains across vertebrates, reinforcing the cross-species applicability of the findings. The SK-based NIS assay, with optional BTS integration, represents a sensitive, robust, and high-throughput amendable alternative to radioactivity-based methods, for characterizing the impacts of individual compounds and complex environmental mixtures on TH homeostasis.
Collapse
Affiliation(s)
- Puja Kumari
- Faculty of Science, RECETOX, Masaryk University, Kamenice 753/5, Pavilion A29, Kotlarska 2, 625 00, Brno, Czech Republic
| | - Sebastian Lungu-Mitea
- Faculty of Science, RECETOX, Masaryk University, Kamenice 753/5, Pavilion A29, Kotlarska 2, 625 00, Brno, Czech Republic
- Department of Biology and Environmental Science, Linnaeus University, SE-39182 Kalmar, Sweden
| | - Jiří Novák
- Faculty of Science, RECETOX, Masaryk University, Kamenice 753/5, Pavilion A29, Kotlarska 2, 625 00, Brno, Czech Republic
| | - Klára Hilscherová
- Faculty of Science, RECETOX, Masaryk University, Kamenice 753/5, Pavilion A29, Kotlarska 2, 625 00, Brno, Czech Republic.
| |
Collapse
|
2
|
Cook MA, Smailys JD, Ji K, Phelps SM, Tutol JN, Kim W, Ong WSY, Peng W, Maydew C, Zhang YJ, Dodani SC. NitrOFF: An engineered fluorescent biosensor to illuminate nitrate transport in living cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.22.644677. [PMID: 40166251 PMCID: PMC11957115 DOI: 10.1101/2025.03.22.644677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The duality of nitrate is nowhere best exemplified than in human physiology - a detrimental pollutant but also a protective nutrient and signaling ion - particularly as connected to reactive nitrogen oxides. Aside from limited insights into nitrate uptake and storage, foundational nitrate biology has lagged. Genetically encoded fluorescent biosensors can address this gap with real-time imaging. However, imaging technologies for mammalian cell applications remain rare. Here, we set out to design and engineer a two-domain chimera fusing the split green fluorescent protein EGFP and the nitrate recognition domain NreA from Staphylococcus carnosus. Over 7 rounds of directed evolution, 15 mutations were accumulated resulting in the functional biosensor NitrOFF. NitrOFF has a high degree of allosteric communication between the domains reflected in a turn-off intensiometric response (K d ≈ 9 μM). This was further reinforced by X-ray crystal structures of apo and nitrate bound NitrOFF, which revealed that the two domains undergo a large-scale conformational rearrangement that changes the relative positioning of the EGFP and NreA domains by 68.4°. Such a dramatic difference was triggered by the formation of a long helix at the engineered linker connecting the two domains, peeling the β7 strand off the EGFP and thus extinguishing the fluorescence upon nitrate binding. Finally, as a proof-of-concept, we highlighted the utility of this first-generation biosensor to monitor exogenous nitrate uptake and modulation in a human embryonic kidney (HEK) 293 cell line.
Collapse
Affiliation(s)
- Mariah A. Cook
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080
| | | | - Ke Ji
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080
| | - Shelby M. Phelps
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080
| | - Jasmine N. Tutol
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080
| | - Wantae Kim
- McKetta Department of Chemical Engineering, University of Texas, Austin, TX 78712
| | - Whitney S. Y. Ong
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080
| | - Weicheng Peng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080
| | - Caden Maydew
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080
| | - Y. Jessie Zhang
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712
| | - Sheel C. Dodani
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080
| |
Collapse
|
3
|
Yu M, Deng Z, Wang K, Zhang X. Chloride channel-3 regulates sodium-iodide symporter expression and localization in the thyroids of mice on a high-iodide diet. Front Nutr 2025; 12:1537221. [PMID: 40191796 PMCID: PMC11968397 DOI: 10.3389/fnut.2025.1537221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Certain chloride channels and H+/Cl- antiporters, such as chloride channel 3 (ClC-3), are expressed at the apical pole of thyrocytes, facilitating iodide (I-) efflux. However, the relationship between ClC-3 and I- uptake remains unclear. Additionally, whether ClC-3 and reactive oxygen species (ROS) regulate sodium-iodide symporter (NIS) expression and localization under excessive I- conditions remain underexplored. Methods The expression and localization of ClC-3 in wild-type (WT), ClC-3 overexpression (OE) and ClC-3 knockout (KO) were detected by Western blotting (WB), immunohistochemistry and immunofluorescence, respectively. The 131I uptake of the thyroid was measured by thyroid function instrument. The expression and localization of NIS in normal and high iodide diet were detected, respectively. The role of ROS in the regulation of NIS by ClC-3 was observed. Results ClC-3 expressions in thyrocytes were primarily localized to the basolateral and lateral membranes, in both ClC-3 OE and WT mice groups under normal I- conditions. I- uptake was significantly higher in WT and ClC-3 OE mice than in the ClC-3 KO mice under normal I- conditions. The ClC-3 OE group exhibited a higher number of thyroid follicles with elevated NIS expression in the basolateral and lateral membranes than the WT and KO groups. In the ClC-3 KO group, the NIS was predominantly localized in the cytoplasm. In the WT group, NIS fluorescence intensity at the basolateral and lateral membranes increased after 48 h of excessive iodide exposure compared to 24 h. In ClC-3 OE mice, NIS, initially localized intracellularly after 24 h of excessive iodide exposure, was almost fully reintegrated into the basolateral and lateral membranes after 48 h. In contrast, in ClC-3 KO mice, NIS remained primarily cytoplasmic, with no significant change between 24 h and 48 h of I- excess. ROS fluorescence intensity was significantly higher in the ClC-3 OE group than those in the WT and KO groups after 24 h of I- excess. Pre-inhibition of ROS showed no significant differences in NIS localization or expression among the three groups after 24 h of I- excess. Discussion These findings suggest that ClC-3 may regulate NIS function via ROS signaling under excessive iodide conditions.
Collapse
Affiliation(s)
- Meisheng Yu
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiqin Deng
- Hand and Foot Surgery Department, The First Hospital Affiliated to Shenzhen University, Shenzhen, China
| | - Ke Wang
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Xi’an Medical University, Xi’an, China
| | - Xiangzhong Zhang
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Pesce E, Benitez-Gonzalez J, Tindall AJ, Lemkine GF, Robin-Duchesne B, Sachs LM, Pasquier EDD. Testing the sensitivity of the medaka Transgenic Eleuthero-embryonic THYroid-Specific assay (TETHYS) to different mechanisms of action. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 276:107081. [PMID: 39305711 DOI: 10.1016/j.aquatox.2024.107081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 11/12/2024]
Abstract
There are many concerns about the impacts of Endocrine-Disrupting Chemicals on both wildlife and human populations. A plethora of chemicals have been shown to interfere with the Hypothalamic-Pituitary-Thyroid (HPT) axis in vertebrates. Disruption of the HPT axis is one of main endocrine criteria considered for the regulation of chemicals, along with the estrogen axis, androgen axis and steroidogenesis (EATS). In response to these concerns, the Organization for Economic Cooperation and Development (OECD) initiated the validation of test guidelines (TGs) covering the EATS modalities. Regarding thyroid activity and/or disruption assessment, three OECD TGs are validated, all of them using amphibians. To date, no OECD TGs based on fish are available for the detection of Thyroid Active Chemicals (TACs). To fill this gap, we developed a new test for the detection of TACs, the TETHYS assay (Transgenic Eleuthero-embryonic THYroid-Specific assay). This assay uses a medaka (Oryzias latipes) transgenic line Tg(tg:eGFP) expressing Green Fluorescent Protein in the thyroid follicles, under the control of the thyroglobulin promoter. This assay is performed at eleuthero-embryonic life-stages with an exposure length of 72 h. In the present study, the following reference chemicals with known thyroid hormone system mechanism of action have been tested: methimazole, sodium perchlorate, sodium tetrafluoroborate, diclofenac, iopanoic acid, sobetirome, NH-3 and 1-850. Except for the thyroid receptor antagonists, all chemicals tested were identified as thyroid active, modifying the total fluorescence and the size of the thyroid follicles. To investigate the test specificity, we tested three chemicals presumed to be inert on the HPT axis: cefuroxime, abamectin and 17α-ethinylestradiol. All were found to be inactive in the TETHYS assay. This promising New Approach Methodology can serve as a foundation for the development of a new OECD TG in the frame of regulatory assessment of chemicals for thyroid activity.
Collapse
Affiliation(s)
- Elise Pesce
- Laboratoire WatchFrog S.A., 1 Rue Pierre Fontaine, 91000 Évry, France; UMR 7221 Physiologie Moléculaire et Adaptation, CNRS, Muséum National d'Histoire Naturelle, CP32, 57 rue Cuvier, 75005 Paris, France
| | | | - Andrew J Tindall
- Laboratoire WatchFrog S.A., 1 Rue Pierre Fontaine, 91000 Évry, France
| | - Gregory F Lemkine
- Laboratoire WatchFrog S.A., 1 Rue Pierre Fontaine, 91000 Évry, France
| | | | - Laurent M Sachs
- UMR 7221 Physiologie Moléculaire et Adaptation, CNRS, Muséum National d'Histoire Naturelle, CP32, 57 rue Cuvier, 75005 Paris, France
| | | |
Collapse
|
5
|
Clavel Rolland N, Graslin F, Schorsch F, Pourcher T, Blanck O. Investigating the mechanisms of action of thyroid disruptors: A multimodal approach that integrates in vitro and metabolomic analysis. Toxicol In Vitro 2024; 100:105911. [PMID: 39069214 DOI: 10.1016/j.tiv.2024.105911] [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: 05/31/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
The thyroid gland, a vital component of the endocrine system, plays a pivotal role in regulating metabolic processes, growth, and development. To better characterize thyroid system disrupting chemicals (TSDC), we followed the next-generation risk assessment approach, which further considers the mechanistic profile of xenobiotics. We combined targeted in vitro testing with untargeted metabolomics. Four known TSDC, propyl-thiouracil (PTU), sodium perchlorate, triclosan, and 5-pregnen-3β-ol-20-one-16α‑carbonitrile (PCN) were investigated using rat in vitro models, including primary hepatocytes, PCCL3 cells, thyroid microsomes, and three-dimensional thyroid follicles. We confirmed each compound's mode of action, PTU inhibited thyroperoxidase activity and thyroid hormones secretion in thyroid cells model, sodium perchlorate induced a NIS-mediated iodide uptake decrease as triclosan to a lesser extent, and PCN activated expression and activity of hepatic enzymes (CYPs and UGTs) involved in thyroid hormones metabolism. In parallel, we characterized intracellular metabolites of interest. We identified disrupted basal metabolic pathways, but also metabolites directly linked to the compound's mode of action as tyrosine derivates for sodium perchlorate and triclosan, bile acids involved in beta-oxidation, and precursors of cytochrome P450 synthesis for PCN. This pilot study has provided metabolomic fingerprinting of dedicated TSDC exposures, which could be used to screen and differentiate specific modes of action.
Collapse
Affiliation(s)
- Naïs Clavel Rolland
- Université Côte d'Azur, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Transporter in Imaging and Radiotherapy in Oncology Laboratory (TIRO), School of Medicine, Nice, France; Bayer Crop Science, Sophia Antipolis, France
| | - Fanny Graslin
- Université Côte d'Azur, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Transporter in Imaging and Radiotherapy in Oncology Laboratory (TIRO), School of Medicine, Nice, France; Centre Antoine Lacassagne, Nice, France
| | | | - Thierry Pourcher
- Université Côte d'Azur, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Transporter in Imaging and Radiotherapy in Oncology Laboratory (TIRO), School of Medicine, Nice, France.
| | | |
Collapse
|
6
|
Haigis AC, Vergauwen L, LaLone CA, Villeneuve DL, O'Brien JM, Knapen D. Cross-species applicability of an adverse outcome pathway network for thyroid hormone system disruption. Toxicol Sci 2023; 195:1-27. [PMID: 37405877 DOI: 10.1093/toxsci/kfad063] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023] Open
Abstract
Thyroid hormone system disrupting compounds are considered potential threats for human and environmental health. Multiple adverse outcome pathways (AOPs) for thyroid hormone system disruption (THSD) are being developed in different taxa. Combining these AOPs results in a cross-species AOP network for THSD which may provide an evidence-based foundation for extrapolating THSD data across vertebrate species and bridging the gap between human and environmental health. This review aimed to advance the description of the taxonomic domain of applicability (tDOA) in the network to improve its utility for cross-species extrapolation. We focused on the molecular initiating events (MIEs) and adverse outcomes (AOs) and evaluated both their plausible domain of applicability (taxa they are likely applicable to) and empirical domain of applicability (where evidence for applicability to various taxa exists) in a THSD context. The evaluation showed that all MIEs in the AOP network are applicable to mammals. With some exceptions, there was evidence of structural conservation across vertebrate taxa and especially for fish and amphibians, and to a lesser extent for birds, empirical evidence was found. Current evidence supports the applicability of impaired neurodevelopment, neurosensory development (eg, vision) and reproduction across vertebrate taxa. The results of this tDOA evaluation are summarized in a conceptual AOP network that helps prioritize (parts of) AOPs for a more detailed evaluation. In conclusion, this review advances the tDOA description of an existing THSD AOP network and serves as a catalog summarizing plausible and empirical evidence on which future cross-species AOP development and tDOA assessment could build.
Collapse
Affiliation(s)
- Ann-Cathrin Haigis
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Lucia Vergauwen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Carlie A LaLone
- Great Lakes Toxicology and Ecology Division, United States Environmental Protection Agency, Duluth, Minnesota 55804, USA
| | - Daniel L Villeneuve
- Great Lakes Toxicology and Ecology Division, United States Environmental Protection Agency, Duluth, Minnesota 55804, USA
| | - Jason M O'Brien
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Dries Knapen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
7
|
Griebel-Thompson AK, Sands S, Chollet-Hinton L, Christifano D, Sullivan DK, Hull H, Carlson SE. A Scoping Review of Iodine and Fluoride in Pregnancy in Relation to Maternal Thyroid Function and Offspring Neurodevelopment. Adv Nutr 2023; 14:317-338. [PMID: 36796438 PMCID: PMC10229380 DOI: 10.1016/j.advnut.2023.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
Iodine (I), an essential nutrient, is important for thyroid function and therefore growth and development. Fluoride (F), also an essential nutrient, strengthens bones and teeth, and prevents childhood dental caries. Both severe and mild-to-moderate I deficiency and high F exposure during development are associated to decreased intelligence quotient with recent reports associating high levels of F exposure during pregnancy and infancy to low intelligence quotient. Both F and I are halogens, and it has been suggested that F may interfere with the role of I in thyroid function. We provide a scoping review of the literature on I and F exposure dur pregnancy and their individual effects on thyroid function and offspring neurodevelopment. We first discuss I intake and status in pregnancy and the relationship to thyroid function and offspring neurodevelopment. We follow with the F in pregnancy and offspring neurodevelopment. We then review the interaction between I and F on thyroid function. We searched for, and found only one study that assessed both I and F in pregnancy. We conclude more studies are needed.
Collapse
Affiliation(s)
- Adrianne K Griebel-Thompson
- Division of Health Services and Health Outcomes Research, Baby Health Behavior Lab, Children's Mercy Research Institute, Children's Mercy Hospital, Kansas City, MO, United States.
| | - Scott Sands
- Department of Dietetics and Nutrition, Maternal and Infant Nutrition and Development Laboratory, University of Kansas Medical Center, Kansas City, United States
| | - Lynn Chollet-Hinton
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, United States
| | - Danielle Christifano
- Department of Dietetics and Nutrition, Maternal and Infant Nutrition and Development Laboratory, University of Kansas Medical Center, Kansas City, United States
| | - Debra K Sullivan
- Department of Dietetics and Nutrition, Maternal and Infant Nutrition and Development Laboratory, University of Kansas Medical Center, Kansas City, United States
| | - Holly Hull
- Department of Dietetics and Nutrition, Maternal and Infant Nutrition and Development Laboratory, University of Kansas Medical Center, Kansas City, United States
| | - Susan E Carlson
- Department of Dietetics and Nutrition, Maternal and Infant Nutrition and Development Laboratory, University of Kansas Medical Center, Kansas City, United States
| |
Collapse
|
8
|
Okamura T, Tsukamoto S, Okada M, Kikuchi T, Aizawa R, Wakizaka H, Nengaki N, Ogawa M, Ishii H, Zhang MR. 11C-Labeled Radiotracer for Noninvasive and Quantitative Assessment of the Thiocyanate Efflux System in the Brain. Bioconjug Chem 2022; 33:1654-1662. [PMID: 35951365 DOI: 10.1021/acs.bioconjchem.2c00277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Thiocyanate (SCN-) alters the potency of certain agonists for the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor, and dysfunctions in AMPA receptor signaling are considered to underlie a number of neurological diseases. While humans may be exposed to SCN- from the environment, including food sources, a carrier-mediated system transports SCN- from the brain into the blood and is an important regulator of SCN- distribution in the central nervous system. The assessment of this SCN- efflux system in the brain would thus be useful for understanding the mechanisms underlying the neurotoxicity of SCN- and for elucidating the relationship between the efflux system and brain diseases. However, the currently available technique for studying SCN- efflux is severely limited by its invasiveness. Here, we describe the development of a SCN- protracer, 9-pentyl-6-[11C]thiocyanatopurine ([11C]1), to overcome this limitation. [11C]1 was synthesized by the reaction of the iodo-precursor and [11C]SCN- or the reaction of the disulfide precursor with [11C]NH4CN. The protracer [11C]1 entered the brain after intravenous injection into mice and was rapidly metabolized to [11C]SCN-, which was then eliminated from the brain. The efflux of [11C]SCN- was dose-dependently inhibited by perchlorate, a monovalent anion, and the highest dose caused an 82% reduction in the efflux rate. Our findings demonstrate that [11C]1 can be used for the noninvasive and quantitative assessment of the SCN- efflux system in the brain.
Collapse
Affiliation(s)
- Toshimitsu Okamura
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Satoshi Tsukamoto
- Laboratory of Animal and Genome Sciences Section, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Maki Okada
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Tatsuya Kikuchi
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Ryutaro Aizawa
- Laboratory of Animal and Genome Sciences Section, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Hidekatsu Wakizaka
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Nobuki Nengaki
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan.,SHI Accelerator Service, Ltd., 1-17-6 Osaki, Shinagawa-ku, Tokyo 141-0032, Japan
| | - Masanao Ogawa
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan.,SHI Accelerator Service, Ltd., 1-17-6 Osaki, Shinagawa-ku, Tokyo 141-0032, Japan
| | - Hideki Ishii
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
9
|
Semenova AV, Sivolobova GF, Grazhdantseva AA, Agafonov AP, Kochneva GV. Reporter Transgenes for Monitoring the Antitumor Efficacy of Recombinant Oncolytic Viruses. Acta Naturae 2022; 14:46-56. [PMID: 36348722 PMCID: PMC9611865 DOI: 10.32607/actanaturae.11719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022] Open
Abstract
Accurate measurement of tumor size and margins is crucial for successful oncotherapy. In the last decade, non-invasive imaging modalities, including optical imaging using non-radioactive substrates, deep-tissue imaging with radioactive substrates, and magnetic resonance imaging have been developed. Reporter genes play the most important role among visualization tools; their expression in tumors and metastases makes it possible to track changes in the tumor growth and gauge therapy effectiveness. Oncolytic viruses are often chosen as a vector for delivering reporter genes into tumor cells, since oncolytic viruses are tumor-specific, meaning that they infect and lyse tumor cells without damaging normal cells. The choice of reporter transgenes for genetic modification of oncolytic viruses depends on the study objectives and imaging methods used. Optical imaging techniques are suitable for in vitro studies and small animal models, while deep-tissue imaging techniques are used to evaluate virotherapy in large animals and humans. For optical imaging, transgenes of fluorescent proteins, luciferases, and tyrosinases are used; for deep-tissue imaging, the most promising transgene is the sodium/iodide symporter (NIS), which ensures an accumulation of radioactive isotopes in virus-infected tumor cells. Currently, NIS is the only reporter transgene that has been shown to be effective in monitoring tumor virotherapy not only in preclinical but also in clinical studies.
Collapse
Affiliation(s)
- A. V. Semenova
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| | - G. F. Sivolobova
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| | - A. A. Grazhdantseva
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| | - A. P. Agafonov
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| | - G. V. Kochneva
- Federal Budgetary Research Institution «State Research Center of Virology and Biotechnology «Vector», Koltsovo, Novosibirsk region, 630559, Russia
| |
Collapse
|
10
|
Concilio SC, Russell SJ, Peng KW. A brief review of reporter gene imaging in oncolytic virotherapy and gene therapy. Mol Ther Oncolytics 2021; 21:98-109. [PMID: 33981826 PMCID: PMC8065251 DOI: 10.1016/j.omto.2021.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reporter gene imaging (RGI) can accelerate development timelines for gene and viral therapies by facilitating rapid and noninvasive in vivo studies to determine the biodistribution, magnitude, and durability of viral gene expression and/or virus infection. Functional molecular imaging systems used for this purpose can be divided broadly into deep-tissue and optical modalities. Deep-tissue modalities, which can be used in animals of any size as well as in human subjects, encompass single photon emission computed tomography (SPECT), positron emission tomography (PET), and functional/molecular magnetic resonance imaging (f/mMRI). Optical modalities encompass fluorescence, bioluminescence, Cerenkov luminescence, and photoacoustic imaging and are suitable only for small animal imaging. Here we discuss the mechanisms of action and relative merits of currently available reporter gene systems, highlighting the strengths and weaknesses of deep tissue versus optical imaging systems and the hardware/reagents that are used for data capture and processing. In light of recent technological advances, falling costs of imaging instruments, better availability of novel radioactive and optical tracers, and a growing realization that RGI can give invaluable insights across the entire in vivo translational spectrum, the approach is becoming increasingly essential to facilitate the competitive development of new virus- and gene-based drugs.
Collapse
Affiliation(s)
| | | | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
11
|
Jin Y, Liu B, Younis MH, Huang G, Liu J, Cai W, Wei W. Next-Generation Molecular Imaging of Thyroid Cancer. Cancers (Basel) 2021; 13:3188. [PMID: 34202358 PMCID: PMC8268517 DOI: 10.3390/cancers13133188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
An essential aspect of thyroid cancer (TC) management is personalized and precision medicine. Functional imaging of TC with radioiodine and [18F]FDG has been frequently used in disease evaluation for several decades now. Recently, advances in molecular imaging have led to the development of novel tracers based on aptamer, peptide, antibody, nanobody, antibody fragment, and nanoparticle platforms. The emerging targets-including HER2, CD54, SHP2, CD33, and more-are promising targets for clinical translation soon. The significance of these tracers may be realized by outlining the way they support the management of TC. The provided examples focus on where preclinical investigations can be translated. Furthermore, advances in the molecular imaging of TC may inspire the development of novel therapeutic or theranostic tracers. In this review, we summarize TC-targeting probes which include transporter-based and immuno-based imaging moieties. We summarize the most recent evidence in this field and outline how these emerging strategies may potentially optimize clinical practice.
Collapse
Affiliation(s)
- Yuchen Jin
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, China
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Beibei Liu
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People’s Hospital Affiliatede to Shanghai Jiao Tong University, Shanghai 200233, China;
| | - Muhsin H. Younis
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| | - Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| |
Collapse
|
12
|
Di Dalmazi G, Giuliani C. Plant constituents and thyroid: A revision of the main phytochemicals that interfere with thyroid function. Food Chem Toxicol 2021; 152:112158. [PMID: 33789121 DOI: 10.1016/j.fct.2021.112158] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 01/06/2023]
Abstract
In the past few decades, there has been a lot of interest in plant constituents for their antioxidant, anti-inflammatory, anti-microbial and anti-proliferative properties. However, concerns have been raised on their potential toxic effects particularly when consumed at high dose. The anti-thyroid effects of some plant constituents have been known for some time. Indeed, epidemiological observations have shown the causal association between staple food based on brassicaceae or soybeans and the development of goiter and/or hypothyroidism. Herein, we review the main plant constituents that interfere with normal thyroid function such as cyanogenic glucosides, polyphenols, phenolic acids, and alkaloids. In detail, we summarize the in vitro and in vivo studies present in the literature, focusing on the compounds that are more abundant in foods or that are available as dietary supplements. We highlight the mechanism of action of these compounds on thyroid cells by giving a particular emphasis to the experimental studies that can be significant for human health. Furthermore, we reveal that the anti-thyroid effects of these plant constituents are clinically evident only when they are consumed in very large amounts or when their ingestion is associated with other conditions that impair thyroid function.
Collapse
Affiliation(s)
- Giulia Di Dalmazi
- Center for Advanced Studies and Technology (CAST) and Department of Medicine and Aging Science, University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy; Department of Medicine and Aging Science, Translational Medicine PhD Program, University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy.
| | - Cesidio Giuliani
- Center for Advanced Studies and Technology (CAST) and Department of Medicine and Aging Science, University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|
13
|
Holloway N, Riley B, MacKenzie DS. Expression of the sodium iodide symporter (NIS) in reproductive and neural tissues of teleost fish. Gen Comp Endocrinol 2021; 300:113632. [PMID: 33002449 DOI: 10.1016/j.ygcen.2020.113632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/30/2020] [Accepted: 09/20/2020] [Indexed: 11/26/2022]
Abstract
Iodine, an essential component of thyroid hormones, can only be obtained through the diet. The sodium iodide symporter (NIS) transports iodide across mammalian intestinal and thyroid epithelia to deliver iodide for thyroid hormone production. Using reverse transcription-polymerase chain reaction (RT-PCR) we confirmed that mRNA for a homolog of mammalian NIS is expressed in comparable locations, both sub-pharyngeal thyroid tissue and intestine, in multiple teleost fish species, supporting a conserved mechanism for intestinal-thyroid iodine transport across vertebrates. To determine when in embryogenesis NIS expression is initiated we utilized in situ hybridization (ISH) during development of zebrafish (Danio rerio) embryos. This revealed expression of nis as early as 2 days post fertilization (dpf) along the dorsal surface of the yolk sac, suggesting a function to import iodine from yolk. To evaluate the potential for maternal deposition of iodine in yolk, RT-PCR and further in situ staining of ovarian tissue in gravid female zebrafish confirmed NIS mRNA presence in the ooplasm and granulosa layer of early stage follicles. This further suggests that maternally-deposited NIS mRNA may be available for early embryogenesis. Unexpectedly, ISH in embryos revealed robust nis expression in the central nervous system throughout days 2-5 days post fertilization, with adult whole brain ISH localizing expression in the hypothalamus, cerebellum, and optic tectum. RT-PCR on whole brain tissue from five species of adult fish representing three taxonomic orders likewise revealed robust CNS expression. These unexpected locations of nis expression suggest novel, as yet undescribed reproductive and neural functions of NIS in teleost species.
Collapse
Affiliation(s)
- Nicholas Holloway
- Department of Biology, 3258 TAMU, Texas A&M University, College Station, TX 77843, USA.
| | - Bruce Riley
- Department of Biology, 3258 TAMU, Texas A&M University, College Station, TX 77843, USA
| | - Duncan S MacKenzie
- Department of Biology, 3258 TAMU, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
14
|
Concilio SC, Suksanpaisan L, Pham L, Peng KW, Russell SJ. Improved Noninvasive In Vivo Tracking of AAV-9 Gene Therapy Using the Perchlorate-Resistant Sodium Iodide Symporter from Minke Whale. Mol Ther 2020; 29:236-243. [PMID: 33038323 PMCID: PMC7791078 DOI: 10.1016/j.ymthe.2020.09.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 01/12/2023] Open
Abstract
The sodium iodide symporter (NIS) is widely used as a reporter gene to noninvasively monitor the biodistribution and durability of vector-mediated gene expression via gamma scintigraphy, single-photon emission computed tomography (SPECT), and positron-emission tomography (PET). However, the approach is limited by background signal due to radiotracer uptake by endogenous NIS-expressing tissues. In this study, using the SPECT tracer pertechnetate (99mTcO4) and the PET tracer tetrafluoroborate (B18F4), in combination with the NIS inhibitor perchlorate, we compared the transport properties of human NIS and minke whale (Balaenoptera acutorostrata scammoni) NIS in vitro and in vivo. Based on its relative resistance to perchlorate, the NIS protein from minke whale appeared to be the superior candidate reporter gene. SPECT and PET imaging studies in nude mice challenged with NIS-encoding adeno-associated virus (AAV)-9 vectors confirmed that minke whale NIS, in contrast to human and endogenous mouse NIS, continues to function as a reliable reporter even when background radiotracer uptake by endogenous NIS is blocked by perchlorate.
Collapse
Affiliation(s)
- Susanna C Concilio
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Linh Pham
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Imanis Life Sciences, LLC, Rochester, MN 55901, USA
| | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Imanis Life Sciences, LLC, Rochester, MN 55901, USA.
| |
Collapse
|
15
|
Lisco G, De Tullio A, Giagulli VA, De Pergola G, Triggiani V. Interference on Iodine Uptake and Human Thyroid Function by Perchlorate-Contaminated Water and Food. Nutrients 2020; 12:E1669. [PMID: 32512711 PMCID: PMC7352877 DOI: 10.3390/nu12061669] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Perchlorate-induced natrium-iodide symporter (NIS) interference is a well-recognized thyroid disrupting mechanism. It is unclear, however, whether a chronic low-dose exposure to perchlorate delivered by food and drinks may cause thyroid dysfunction in the long term. Thus, the aim of this review was to overview and summarize literature results in order to clarify this issue. METHODS Authors searched PubMed/MEDLINE, Scopus, Web of Science, institutional websites and Google until April 2020 for relevant information about the fundamental mechanism of the thyroid NIS interference induced by orally consumed perchlorate compounds and its clinical consequences. RESULTS Food and drinking water should be considered relevant sources of perchlorate. Despite some controversies, cross-sectional studies demonstrated that perchlorate exposure affects thyroid hormone synthesis in infants, adolescents and adults, particularly in the case of underlying thyroid diseases and iodine insufficiency. An exaggerated exposure to perchlorate during pregnancy leads to a worse neurocognitive and behavioral development outcome in infants, regardless of maternal thyroid hormone levels. DISCUSSION AND CONCLUSION The effects of a chronic low-dose perchlorate exposure on thyroid homeostasis remain still unclear, leading to concerns especially for highly sensitive patients. Specific studies are needed to clarify this issue, aiming to better define strategies of detection and prevention.
Collapse
Affiliation(s)
- Giuseppe Lisco
- ASL Brindisi, Unit of Endocrinology, Metabolism & Clinical Nutrition, Hospital “A. Perrino”, Strada per Mesagne 7, 72100 Brindisi, Puglia, Italy;
| | - Anna De Tullio
- Interdisciplinary Department of Medicine—Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari “Aldo Moro”, School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Puglia, Italy; (A.D.T.); (V.A.G.)
| | - Vito Angelo Giagulli
- Interdisciplinary Department of Medicine—Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari “Aldo Moro”, School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Puglia, Italy; (A.D.T.); (V.A.G.)
- Clinic of Endocrinology and Metabolic Disease, Conversano Hospital, Via Edmondo de Amicis 36, 70014 Conversano, Bari, Puglia, Italy
| | - Giovanni De Pergola
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Puglia, Italy;
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine—Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari “Aldo Moro”, School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Puglia, Italy; (A.D.T.); (V.A.G.)
| |
Collapse
|
16
|
Oncolytic measles virus therapy enhances tumor antigen-specific T-cell responses in patients with multiple myeloma. Leukemia 2020; 34:3310-3322. [PMID: 32327728 PMCID: PMC7581629 DOI: 10.1038/s41375-020-0828-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/13/2020] [Accepted: 03/30/2020] [Indexed: 12/24/2022]
Abstract
Oncolytic virus therapy leads to immunogenic death of virus-infected tumor cells and this has been shown in preclinical models to enhance the cytotoxic T-lymphocyte response against tumor-associated antigens (TAAs), leading to killing of uninfected tumor cells. To investigate whether oncolytic virotherapy can increase immune responses to tumor antigens in human subjects, we studied T-cell responses against a panel of known myeloma TAAs using PBMC samples obtained from ten myeloma patients before and after systemic administration of an oncolytic measles virus encoding sodium iodide symporter (MV-NIS). Despite their prior exposures to multiple immunosuppressive antimyeloma treatment regimens, T-cell responses to some of the TAAs were detectable even before measles virotherapy. Measurable baseline T-cell responses against MAGE-C1 and hTERT were present. Furthermore, MV-NIS treatment significantly (P < 0.05) increased T-cell responses against MAGE-C1 and MAGE-A3. Interestingly, one patient who achieved complete remission after MV-NIS therapy had strong baseline T-cell responses both to measles virus proteins and to eight of the ten tested TAAs. Our data demonstrate that oncolytic virotherapy can function as an antigen agnostic vaccine, increasing cytotoxic T-lymphocyte responses against TAAs in patients with multiple myeloma, providing a basis for continued exploration of this modality in combination with immune checkpoint blockade.
Collapse
|