350 results on '"Sodium iodide symporter"'
Search Results
2. Advances in molecular mechanisms of iodine-131 therapy resistance in thyroid carcinoma
- Author
-
LIU Shiqi, WANG Hui, and FENG Fang
- Subjects
differentiated thyroid carcinoma ,dedifferentiation ,radioiodine ,sodium iodide symporter ,Medicine - Abstract
Thyroid cancer is the most common malignant tumor of the endocrine system, with differentiated thyroid carcinoma (DTC) accounting for over 90%. Most DTC patients have a good prognosis after systematic treatment, but a few develop dedifferentiation of primary tumor site or metastases, progressing to radioiodine-refractory DTC (RAIR-DTC), leading to significantly worse prognosis, which is a major cause of thyroid carcinoma-related mortality. Dysregulation of sodium iodide symporter (NIS) expression and function is the main reason for iodine-131 therapy resistance in thyroid carcinoma, influenced by genetic changes, epigenetic changes, tumor microenvironment, autophagy, and other factors. Genetic alterations such as the BRAFV600E mutation and RET/PTC chromosomal rearrangements activate oncogenic signaling pathways, directly or indirectly affecting NIS expression and its normal localization on the cell membrane. Epigenetic regulation modulates specific gene expression patterns, regulating NIS gene expression levels, thereby affecting the radioiodine uptake function of thyroid cells. Components in the tumor microenvironment, including immune cells, cytokines, and extracellular matrix, may also disrupt iodine uptake by reducing the expression levels of NIS and/or disrupting its normal function on the cell membrane. Additionally, autophagy, as an intracellular metabolic regulatory mechanism, can also modulate NIS expression and its intracellular distribution, thus impacting the radioiodine uptake and the sensitivity to iodine-131 therapy. Reviewing the roles of these factors in thyroid carcinoma dedifferentiation comprehensively can provide a more thorough understanding of the occurrence and progression of RAIR-DTC, aiding in the exploration of new therapeutic targets, improving prognosis, and providing more effective personalized treatment strategies for patients.
- Published
- 2024
- Full Text
- View/download PDF
3. Canine follicular cell and medullary thyroid carcinomas: Immunohistochemical characterization.
- Author
-
Jankovic, Jana, Tièche, Eve, Dettwiler, Martina, Hahn, Kerstin, Scheemaeker, Stephanie, Kessler, Martin, Daminet, Sylvie, Rottenberg, Sven, and Campos, Miguel
- Subjects
MEDULLARY thyroid carcinoma ,THYROTROPIN receptors ,IODIDE peroxidase ,SODIUM iodide ,THYROID gland tumors ,PROTEIN transport - Abstract
Research on modulation of iodine uptake by thyroid cells could help improve radioiodine treatment of dogs with thyroid tumors. The aim of this study was to characterize the immunohistochemical expression of thyroid transcription factor-1 (TTF-1), thyroglobulin, thyrotropin receptor (TSHR), sodium iodide symporter (NIS), pendrin, thyroid peroxidase (TPO), vimentin, and Ki-67 in follicular cell thyroid carcinomas (FTCs) and medullary thyroid carcinomas (MTCs), and to compare protein expression between FTC causing hyperthyroidism and FTC of euthyroid dogs. Immunohistochemistry was performed in 25 FTCs (9 follicular, 8 follicular-compact, and 8 compact) and 8 MTCs. FTCs and MTCs were positive for TTF-1, and expression was higher in FTCs of euthyroid dogs compared with FTCs of hyperthyroid dogs (P =.041). Immunolabeling for thyroglobulin was higher in follicular and follicular-compact FTCs compared with compact FTCs (P =.001), while vimentin expression was higher in follicular-compact FTCs compared with follicular FTCs (P =.011). The expression of TSHR, NIS, pendrin, and TPO was not significantly different among the different subtypes of FTCs or between FTCs causing hyperthyroidism and FTCs in euthyroid dogs. TSHR, NIS, pendrin, and TPO were also expressed in MTCs. Ki-67 labeling index was comparable between FTCs and MTCs, and between FTCs causing hyperthyroidism and FTCs in euthyroid dogs. Proteins of iodine transport were also expressed in canine MTCs, which could have implications for diagnosis and treatment. The different expression of thyroglobulin and vimentin between FTC histological subtypes could reflect variations in tumor differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Regulation of iodine-glucose flip-flop in SW1736 anaplastic thyroid cancer cell line
- Author
-
Heydarzadeh, S., Moshtaghie, A. A., Daneshpour, M., and Hedayati, M.
- Published
- 2024
- Full Text
- View/download PDF
5. Oncolytic Adenovirus for the Targeting of Paclitaxel-Resistant Breast Cancer Stem Cells.
- Author
-
Robert, Sacha, Roman Ortiz, Natasha Ivelisse, LaRocca, Christopher J., Ostrander, Julie Hanson, and Davydova, Julia
- Subjects
- *
CANCER stem cells , *BREAST cancer , *ADENOVIRUSES , *ESTROGEN receptors , *SODIUM iodide , *PACLITAXEL , *DISEASE risk factors , *BREAST - Abstract
Adjuvant systemic therapies effectively reduce the risk of breast cancer recurrence and metastasis, but therapy resistance can develop in some patients due to breast cancer stem cells (BCSCs). Oncolytic adenovirus (OAd) represents a promising therapeutic approach as it can specifically target cancer cells. However, its potential to target BCSCs remains unclear. Here, we evaluated a Cox-2 promoter-controlled, Ad5/3 fiber-modified OAd designed to encode the human sodium iodide symporter (hNIS) in breast cancer models. To confirm the potential of OAds to target BCSCs, we employed BCSC-enriched estrogen receptor-positive (ER+) paclitaxel-resistant (TaxR) cells and tumorsphere assays. OAd-hNIS demonstrated significantly enhanced binding and superior oncolysis in breast cancer cells, including ER+ cells, while exhibiting no activity in normal mammary epithelial cells. We observed improved NIS expression as the result of adenovirus death protein deletion. OAd-hNIS demonstrated efficacy in targeting TaxR BCSCs, exhibiting superior killing and hNIS expression compared to the parental cells. Our vector was capable of inhibiting tumorsphere formation upon early infection and reversing paclitaxel resistance in TaxR cells. Importantly, OAd-hNIS also destroyed already formed tumorspheres seven days after their initiation. Overall, our findings highlight the promise of OAd-hNIS as a potential tool for studying and targeting ER+ breast cancer recurrence and metastasis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. An Unlikely Source of Iodine Uptake: A Bronchogenic Cyst Masquerading as Metastatic Thyroid Cancer.
- Author
-
Dillon, Martha, Zielinski, Rachel, Worth, Jennifer, Sanders, Melinda, Ibrahim, Omar, and Vedere, Tarunya
- Subjects
- *
THYROID cancer , *METASTASIS , *CYSTS (Pathology) , *IODINE isotopes , *SODIUM iodide , *IODINE - Abstract
Radioactive iodine therapy and posttreatment scanning are essential components of differentiated thyroid carcinoma treatment and detection of metastatic disease. False-positive results can be seen on an I-131 scan and are important for clinicians to be aware of. Here, we present a case of a 33-year-old female with follicular thyroid carcinoma who was noted to have an area of moderate uptake in the chest on a whole-body scan following remnant ablation with 30 mCi of I-131 (1.11GBq) concerning for a metastatic hilar lymph node. This was determined to be a mediastinal bronchogenic cyst on surgical pathology. It has been previously proposed that the expression of sodium iodide symporters in some bronchogenic cysts could be the mechanism by which iodine uptake is seen within them. We were able to demonstrate positive immunohistochemical staining for both sodium iodide symporter and the associated paired box gene 8 transcription factor in the cyst sample, which supports the proposed theory. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Hubungan Ekspresi Sodium Iodide Symporter dengan Varian Histopatologi dan Stadium Papillary Thyroid Carcinoma
- Author
-
Rini Purnama Sari, yenita - yenita, Noza - Hilbertina, tofrizal - tofrizal, Daan - Khambri, and Husna - Yetti
- Subjects
sodium iodide symporter ,papillary thyroid carcinoma ,varian histopatologi ,stadium ,Medicine (General) ,R5-920 - Abstract
Abstrak Tujuan: Mengetahui hubungan ekspresi sodium iodide symporter dengan varian histopatologi dan stadium papillary thyroid carcinoma. Metode: Penelitian ini merupakan penelitian observasional dengan pendekatan cross sectional. Sampel penelitian ini adalah kasus papillary thyroid carcinoma dari laboratorium Patologi Anatomi RSUP Dr. M. Djamil Padang periode Januari 2022-Desember 2022 sebanyak 37 kasus. Varian histopatologi papillary thyroid carcinoma dinilai berdasarkan WHO 2017 dan stadium klinis dinilai berdasarkan TNM American Joint Committee on Cancer edisi ke VIII. Ekspresi NIS dinilai melalui pulasan imunohistokimia. Analisis bivariat dilakukan dengan uji Chi-square dengan hasil uji statistik dianggap bermakna jika p < 0,05. Hasil: Penelitian ini menunjukkan ekspresi NIS positif sebesar 13,5%. Analisis statistik menunjukkan adanya hubungan yang bermakna antara ekspresi NIS dengan varian histopatologi papillary thyroid carcinoma (p=0,03). Varian histopatologi tertentu menunjukkan tingkat ekspresi NIS yang lebih tinggi dibandingkan dengan varian lainnya. Namun, penelitian ini tidak menemukan hubungan antara ekspresi NIS dengan stadium papillary thyroid carcinoma (p =0,61). Kesimpulan: Terdapat hubungan antara ekspresi NIS dengan varian histopatologi. Ekspresi NIS tidak berhubungan dengan stadium papillary thyroid carcinoma. Kata kunci: sodium iodide symporter; papillary thyroid carcinoma; varian histopatologi; stadium
- Published
- 2024
- Full Text
- View/download PDF
8. Nuclear Imaging of the Thyroid Gland
- Author
-
O, Joo Hyun, Ha, Seunggyun, Kakudo, Kennichi, editor, Liu, Zhiyan, editor, Jung, Chan Kwon, editor, Hirokawa, Mitsuyoshi, editor, Bychkov, Andrey, editor, and Lai, Chiung-Ru, editor
- Published
- 2023
- Full Text
- View/download PDF
9. Evaluation of the diphenyl herbicide, oxyfluorfen, for effects on thyroid hormones in the juvenile rat
- Author
-
T.E. Stoker, G.D. DeVane, A.R. Buckalew, J.R. Bailey, J.L. Ford, and A.S. Murr
- Subjects
Thyroid hormones ,Oxyfluorfen ,Sodium iodide symporter ,Endocrine ,Toxicology. Poisons ,RA1190-1270 - Abstract
Recently, oxyfluorfen, a pre- and post-emergent diphenyl ether herbicide, was identified in our laboratory as an inhibitor of iodide uptake by the sodium iodide symporter (NIS), the first key step in the synthesis of thyroid hormones (THs). This inhibition was observed in vitro, using both a human NIS engineered cell line (hNIS-HEK293T-EPA) and a rat thyroid follicular cell line (FRTL-5). Oxyfluorfen was found to be a potent inhibitor of NIS activity with an EC50 of approximately 2 µM in both cell lines with no observed cytotoxicity at any concentration tested up to 100 μM. The current research tested the hypothesis that oxyfluorfen alters circulating concentrations of THs. This hypothesis was first tested in a pilot study with both juvenile male and female rats exposed to oxyfluorfen for 4 days at 0, 125, 250 and 500 mg/kg/day. Once we identified that this short-term 4-day oxyfluorfen exposure suppressed both total serum thyroxine (T4) and triiodothyronine (T3) at all doses, we tested seven lower concentrations of oxyfluorfen (0.8125 to 62.5 mg/kg day) in an 8-day exposure paradigm to more closely evaluate the dose–response. We found that oxyfluorfen suppressed serum T4 with a LOEL of 3.25 mg/kg/day and T3 with a LOEL 62.5 mg/kg/day. Analytical chemistry of the serum showed an accumulation over time following oral exposure to oxyfluorfen in both the 4- and 8-day groups. Analytical chemistry of the thyroid glands in the 8-day study revealed higher accumulation in the thyroid as compared to the serum (2 to 3- fold at 62.5 mg/kg). No changes in thyroid weight or serum TSH were observed following the 8-day exposure. This study is the first to demonstrate an effect of oxyfluorfen on serum thyroid hormones in the rat. Additional studies are needed to further evaluate the effects on thyroid homeostasis with extended exposures and the potential implications of the observed effects.
- Published
- 2024
- Full Text
- View/download PDF
10. Dual EGFR- and TfR-targeted gene transfer for sodium iodide symporter gene therapy of glioblastoma
- Author
-
Rebekka Spellerberg, Teoman Benli-Hoppe, Carolin Kitzberger, Mara Hageneier, Nathalie Schwenk, Özgür Öztürk, Katja Steiger, Gabriele Multhoff, Matthias Eiber, Franz Schilling, Wolfgang A. Weber, Roland E. Kälin, Rainer Glass, Peter J. Nelson, Ernst Wagner, and Christine Spitzweg
- Subjects
sodium iodide symporter ,NIS ,radioiodine ,blood-brain barrier ,EGFR ,transferrin receptor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Sodium iodide symporter (NIS) gene transfer for active accumulation of iodide in tumor cells is a powerful theranostic strategy facilitating both diagnostic and therapeutic application of radioiodide. In glioblastoma (GBM), the blood-brain barrier (BBB) presents an additional delivery barrier for nucleic acid nanoparticles. In the present study, we designed dual-targeted NIS plasmid DNA complexes containing targeting ligands for the transferrin receptor (TfR) and the epidermal growth factor receptor (EGFR), thus providing the potential for active transport across the BBB followed by targeting of tumor cells. In vitro 125I transfection studies confirmed TfR- and EGFR-dependent transfection efficiency and NIS-specific iodide uptake of dual-targeted polyplexes. In vivo gene transfer in mice bearing orthotopic U87 GBM xenografts was assessed at 48 h after intravenous polyplex injection by positron emission tomography (PET) imaging using 18F-labeled tetrafluoroborate (TFB) as tracer. The tumoral 18F-TFB uptake of mice treated with dual-targeted polyplexes (0.56% ± 0.08% ID/mL) was significantly higher compared with mice treated with EGFR-mono-targeted (0.33% ± 0.03% ID/mL) or TfR-mono-targeted (0.27% ± 0.04% ID/mL) polyplexes. In therapy studies, application of 131I induced a superior therapeutic effect of the dual-targeted therapy, demonstrated by a significant delay in tumor growth and prolonged survival.
- Published
- 2022
- Full Text
- View/download PDF
11. High molar activity [18F]tetrafluoroborate synthesis for sodium iodide symporter imaging by PET
- Author
-
Dmitry Soloviev, Piotr Dzien, Agata Mackintosh, Gaurav Malviya, Gavin Brown, and David Lewis
- Subjects
NIS ,Reporter gene imaging ,Tetrafluoroborate ,Fluorine-18 ,Positron emission tomography ,Sodium iodide symporter ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract Background Sodium iodide symporter (NIS) imaging by positron emission tomography (PET) is gaining traction in nuclear medicine, with an increasing number of human studies being published using fluorine-18 radiolabelled tetrafluoroborate ([18F]TFB). Clinical success of any radiotracer relies heavily on its accessibility, which in turn depends on the availability of robust radiolabelling procedures providing a radiotracer in large quantities and of high radiopharmaceutical quality. Results Here we publish an improved radiolabelling method and quality control procedures for high molar activity [18F]TFB. The use of ammonium hydroxide for [18F]fluoride elution, commercially available boron trifluoride-methanol complex dissolved in acetonitrile as precursor and removal of unreacted [18F]fluoride on Florisil solid-phase extraction cartridges resulted in the reliable production of [18F]TFB on SYNTHRA and TRACERLAB FXFN automated synthesizers with radiochemical yields in excess of 30%, radiochemical purities in excess of 98% and molar activities in the range of 34–217 GBq/µmol at the end of synthesis. PET scanning of a mouse lung tumour model carrying a NIS reporter gene rendered images of high quality and improved sensitivity. Conclusions A novel automated radiosynthesis procedure for [18F]tetrafluoroborate has been developed that provides the radiotracer with high molar activity, suitable for preclinical imaging of NIS reporter gene.
- Published
- 2022
- Full Text
- View/download PDF
12. Evaluation of Single Dose and Fractionated Dose of I-131 Radiolabeled Nanoparticles for Triple-Negative Breast Cancer Treatment.
- Author
-
Marshall, Suphalak Khamruang, Kaewpradit, Nutnicha, Mudmarn, Tavadee, Buathong, Jirassaya, and Sriwirote, Palmuk
- Subjects
TRIPLE-negative breast cancer ,CYCLIN-dependent kinases ,CANCER treatment ,RADIOCHEMICAL purification ,SODIUM iodide ,ERYTHROCYTES - Abstract
Combination chemotherapy is still the standard clinical care for triple-negative breast cancer (TNBC). However, sodium iodide symporter (NIS) uptake by TNBC has opened the potential of NIS as a molecular target for radioiodine theranostic treatments. Radiolabeled poly(lactic-co-glycolic) acid nanocarrier (NINP) was developed for NIS targeted delivery of I-131 to MDA-MB-231 cells to overcome I-131 low uptake in cancer cells and rapid clearance. The NINP diameter of 237 nm has good particle size uniformity and excellent particle stability. Radiochemical purity, radioactive stability, and radiolabeling yield of NINPs over 72 h were >95%. Cytotoxicity confirmed fractionated NINPs over 72 h to be more effective in cell death than single-dose NINP and both single and fractionated Na
131 I. Cellular uptake in a three-dimensional spheroid confirmed that NINP fractionated-dose achieved ~4.8-fold-higher mean fluorescent intensity than Na131 I and ~2.7-fold greater reduction in cell viability compared to single-dose. The NINP fractionated-dose initiated greater cellular DNA damage to cells than single-dose NINP, resulting in inhibition of cell cycle progression, resulting in cell cycle progression being inhibited by cyclin-dependent kinases, which play a vital role in the control of MDA-MB-231 cell cycle. NINPs are biocompatible with blood, and were found to have no negative impact on red blood cells. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
13. Visualizing mast cell migration to tumor sites using sodium iodide symporter of nuclear medicine reporter gene
- Author
-
Seul-Gi Oh, Jun Young Choi, Jae-Eon Lee, SoYeon Jeon, Bo-Ra Lee, Kwang Hee Son, Sang Bong Lee, Beum-Soo An, Dae Youn Hwang, Seong-Jang Kim, Ki-Tae Ha, Jaetae Lee, and Yong Hyun Jeon
- Subjects
Mast cells ,Nuclear reporter gene imaging ,Sodium iodide symporter ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Purpose: Owing to the close relationship between mast cells and cancer progression, an imaging technique that can be applied in a clinical setting to explore the biological behavior of mast cells in the tumor microenvironment is needed. In this study, we visualized mast cell migration to lung tumor lesions in live mice using sodium iodide symporter (NIS) as a nuclear medicine reporter gene. Experimental design: The murine mast cell line MC-9 was infected with retrovirus including NIS, luciferase (as a surrogate marker for NIS), and Thy1.1 to generate MC-9/NFT cells. Radioiodine uptake was measured in MC-9/NFT cells, and an inhibition assay of radioiodine uptake using KCLO4 was also performed. Cell proliferation and FcεRI expression was examined in MC-9 and MC-9/NFT cells. The effect of mast cell-conditioned media (CM) on the proliferation of Lewis lung cancer (LLC) cells was examined. The migration level of MC-9/NFT cells was confirmed in the presence of serum-free media (SFM) and CM of cancer cells. After intravenous injection of MC-9/NFT cells into mice with an LLC tumor, I-124 PET/CT and biodistribution analysis was performed. Results: MC-9/NFT cells exhibited higher radioiodine avidity compared to parental MC-9 cells; this increased radioiodine avidity in MC-9/NFT cells was reduced to basal level by KCLO4. Levels of FcεRI expression and cell proliferation were not different in parental MC-9 cell and MC-9/ NFT cells. The CM of MC-9/NFT cells increased cancer cell proliferation relative to that of the SFM. The migration level of MC-9/NFT cells was higher in the CM than the SFM of LLC cells. PET/CT imaging with I-124 clearly showed infiltration of reporter mast cells in lung tumor at 24 h after transfer, which was consistent with the findings of the biodistribution examination. Conclusion: These findings suggest that the sodium iodide symporter can serve as a reliable nuclear medicine reporter gene for non-invasively imaging the biological activity of mast cells in mice with lung tumors. Visualizing mast cells in the tumor microenvironment via a nuclear medicine reporter gene would provide valuable insights into their biological functions.
- Published
- 2023
- Full Text
- View/download PDF
14. The toxicological role of Myricetin in the progression of human anaplastic thyroid cancer SW1736 cell line.
- Author
-
Heydarzadeh, Shabnam, Moshtaghie, Ali Asghar, Daneshpour, Maryam, Pishdad, Reza, Farahani, Amin, and Hedayati, Mehdi
- Subjects
- *
ANAPLASTIC thyroid cancer , *IODINE isotopes , *TREATMENT effectiveness , *HYDROGEN bonding interactions , *SODIUM iodide - Abstract
Anaplastic thyroid cancer cells lack the capacity to effectively accumulate iodine and are therefore unresponsive to treatment with radioactive iodine. The main objective of this study was to examine the possible therapeutic effects of Myricetin on the SW1736 ATC cell line. In this study, we assessed the influence of Myricetin on iodide absorption, sodium iodide symporter gene expression, and apoptosis induction. The interaction between the 7UUY protein of NIS and Myricetin was investigated using AutoDock Vina. Assessment of cell viability was conducted with the MTT assay, whereas cell apoptosis was evaluated by flow cytometry using the Annexin V-FITC Apoptosis Detection kit. A spectrophotometric test based on the Sandell-Kolthoff reaction was conducted to assess the absorption of iodide by SW1736 cells. QRT-PCR analyses were used to assess the expression levels of NIS mRNA in SW1736 cells. The hydrogen bond interaction pattern created by PyMOL revealed the interactions between the target and ligand molecules. The results demonstrated that Myricetin-induced cell death is dependent on apoptosis in this type of thyroid cancer cell line. QRT-PCR analyses revealed significantly higher NIS mRNA (P < 0.001) levels in the Myricetin-treated group than in the non-treated group. Furthermore, Myricetin treatment significantly increased iodide uptake (P value = 0.0053) in the SW1736 thyroid cancer cell line compared to the control group. These findings suggest that Myricetin has potential as a therapeutic agent by promoting growth inhibition, enhancing NIS gene expression, and increasing iodide uptake in SW1736 cells. Additional research is necessary to clarify the fundamental mechanisms and to evaluate the efficacy of Myricetin in preclinical and clinical settings. • Anaplastic thyroid cancer cells are unresponsive to radioactive iodine treatment. • Targeting the iodide pathway sensitizes tumor cells to iodine therapy. • Myricetin enhances apoptosis, NIS gene expression, and iodide uptake in SW1736 cells. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
15. The sodium iodide symporter (NIS) as theranostic gene: its emerging role in new imaging modalities and non-viral gene therapy
- Author
-
Carolin Kitzberger, Rebekka Spellerberg, Volker Morath, Nathalie Schwenk, Kathrin A. Schmohl, Christina Schug, Sarah Urnauer, Mariella Tutter, Matthias Eiber, Franz Schilling, Wolfgang A. Weber, Sibylle Ziegler, Peter Bartenstein, Ernst Wagner, Peter J. Nelson, and Christine Spitzweg
- Subjects
Sodium iodide symporter ,[18F]tetrafluoroborate ,124I ,PET ,Glioblastoma ,Gene therapy ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Abstract Cloning of the sodium iodide symporter (NIS) in 1996 has provided an opportunity to use NIS as a powerful theranostic transgene. Novel gene therapy strategies rely on image-guided selective NIS gene transfer in non-thyroidal tumors followed by application of therapeutic radionuclides. This review highlights the remarkable progress during the last two decades in the development of the NIS gene therapy concept using selective non-viral gene delivery vehicles including synthetic polyplexes and genetically engineered mesenchymal stem cells. In addition, NIS is a sensitive reporter gene and can be monitored by high resolution PET imaging using the radiotracers sodium [124I]iodide ([124I]NaI) or [18F]tetrafluoroborate ([18F]TFB). We performed a small preclinical PET imaging study comparing sodium [124I]iodide and in-house synthesized [18F]TFB in an orthotopic NIS-expressing glioblastoma model. The results demonstrated an improved image quality using [18F]TFB. Building upon these results, we will be able to expand the NIS gene therapy approach using non-viral gene delivery vehicles to target orthotopic tumor models with low volume disease, such as glioblastoma. Trial registration not applicable.
- Published
- 2022
- Full Text
- View/download PDF
16. Propensity Score-Matched Analysis to Identify Pathways Associated with Loss of Sodium Iodide Symporter in Papillary Thyroid Cancer
- Author
-
Fang Lee, Chi-Yu Kuo, Chung-Hsin Tsai, and Shih-Ping Cheng
- Subjects
sodium iodide symporter ,thyroid cancer ,propensity score matching ,gene set enrichment analysis ,Biology (General) ,QH301-705.5 - Abstract
Sodium iodide symporter (NIS) expression in thyroid follicular cells plays an important role in normal physiology and radioactive iodine therapy for thyroid cancer. Loss of NIS expression is often seen in thyroid cancers and may lead to radioiodine refractoriness. To explore novel mechanisms of NIS repression beyond oncogenic drivers, clinical and RNA-seq data from the thyroid cancer dataset of The Cancer Genome Atlas were analyzed. Propensity score matching was used to control for various genetic background factors. We found that tumoral NIS expression was negatively correlated with tumor size. Additionally, low NIS expression was the only factor associated with recurrence-free survival in a Cox multivariate regression analysis. After matching for clinicopathologic profiles and driver mutations, the principal component analysis revealed distinct gene expressions between the high and low NIS groups. Gene set enrichment analysis suggested the downregulation of hedgehog signaling, immune networks, and cell adhesions. Positively enriched pathways included DNA replication, nucleotide excision repair, MYC, and Wnt/β-catenin pathways. In summary, we identified several potential targets which could be exploited to rescue the loss of NIS expression and develop redifferentiation strategies to facilitate radioactive iodine therapy for thyroid cancer.
- Published
- 2022
- Full Text
- View/download PDF
17. An Inverse Agonist of Estrogen-Related Receptor Gamma, GSK5182, Enhances Na + /I − Symporter Function in Radioiodine-Refractory Papillary Thyroid Cancer Cells.
- Author
-
Singh, Thoudam Debraj, Lee, Jae Eon, Son, Kwang Hee, Lee, Bo Ra, Kim, Sang Kyoon, Gulwani, Deepak, Sarangthem, Vijaya, and Jeon, Yong Hyun
- Subjects
- *
THROMBOPOIETIN receptors , *THYROID cancer , *ANAPLASTIC thyroid cancer , *CANCER cells , *MITOGEN-activated protein kinases , *IODINE isotopes - Abstract
Previously, we reported that an inverse agonist of estrogen-related receptor gamma (ERRγ), GSK5182, enhances sodium iodide (Na+/I−) symporter (NIS) function through mitogen-activated protein (MAP) kinase signaling in anaplastic thyroid cancer cells. This finding helped us to further investigate the effects of GSK5182 on NIS function in papillary thyroid cancer (PTC) refractory to radioactive iodine (RAI) therapy. Herein, we report the effects of ERRγ on the regulation of NIS function in RAI-resistant PTC cells using GSK5182. RAI-refractory BCPAP cells were treated with GK5182 for 24 h at various concentrations, and radioiodine avidity was determined with or without potassium perchlorate (KClO4) as an NIS inhibitor. We explored the effects of GSK5182 on ERRγ, the mitogen-activated protein (MAP) kinase pathway, and iodide metabolism-related genes. We examined whether the MAP pathway affected GSK5182-mediated NIS function using U0126, a selective MEK inhibitor. A clonogenic assay was performed to evaluate the cytotoxic effects of I-131. GSK5182 induced an increase in radioiodine avidity in a dose-dependent manner, and the enhanced uptake was completely inhibited by KClO4 in BCPAP cells. We found that ERRγ was downregulated and phosphorylated extracellular signal-regulated kinase (ERK)1/2 was upregulated in BCPAP cells, with an increase in total and membranous NIS and iodide metabolism-related genes. MEK inhibitors reversed the increase in radioiodine avidity induced by GSK5182. Clonogenic examination revealed the lowest survival in cells treated with a combination of GSK5182 and I-131 compared to those treated with either GSK518 or I-131 alone. We demonstrate that an inverse agonist of ERRγ, GSK5182, enhances the function of NIS protein via the modulation of ERRγ and MAP kinase signaling, thereby leading to increased responsiveness to radioiodine in RAI-refractory papillary thyroid cancer cells. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
18. Overexpression of Both Human Sodium Iodide Symporter (NIS) and BRG1-Bromodomain Synergistically Enhances Radioiodine Sensitivity by Stabilizing p53 through NPM1 Expression.
- Author
-
Na, Juri, Lee, Chul-Hee, Chung, June-Key, and Youn, Hyewon
- Subjects
- *
SODIUM iodide , *IODINE isotopes , *ARGININE deiminase , *GENETIC overexpression , *GENE therapy - Abstract
Improved therapeutic strategies are required to minimize side effects associated with radioiodine gene therapy to avoid unnecessary damage to normal cells and radiation-induced secondary malignancies. We previously reported that codon-optimized sodium iodide symporter (oNIS) enhances absorption of I-131 and that the brahma-associated gene 1 bromodomain (BRG1-BRD) causes inefficient DNA damage repair after high-energy X-ray therapy. To increase the therapeutic effect without applying excessive radiation, we considered the combination of oNIS and BRG1-BRD as gene therapy for the most effective radioiodine treatment. The antitumor effect of I-131 with oNIS or oNIS+BRD expression was examined by tumor xenograft models along with functional assays at the cellular level. The synergistic effect of both BRG1-BRD and oNIS gene overexpression resulted in more DNA double-strand breaks and led to reduced cell proliferation/survival rates after I-131 treatment, which was mediated by the p53/p21 pathway. We found increased p53, p21, and nucleophosmin 1 (NPM1) in oNIS- and BRD-expressing cells following I-131 treatment, even though the remaining levels of citrulline and protein arginine deiminase 4 (PAD4) were unchanged at the protein level. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
19. High molar activity [18F]tetrafluoroborate synthesis for sodium iodide symporter imaging by PET.
- Author
-
Soloviev, Dmitry, Dzien, Piotr, Mackintosh, Agata, Malviya, Gaurav, Brown, Gavin, and Lewis, David
- Subjects
SODIUM iodide ,TETRAFLUOROBORATES ,SOLID phase extraction ,POSITRON emission tomography ,REPORTER genes ,RADIOCHEMICAL purification ,RADIOACTIVE tracers - Abstract
Background: Sodium iodide symporter (NIS) imaging by positron emission tomography (PET) is gaining traction in nuclear medicine, with an increasing number of human studies being published using fluorine-18 radiolabelled tetrafluoroborate ([
18 F]TFB). Clinical success of any radiotracer relies heavily on its accessibility, which in turn depends on the availability of robust radiolabelling procedures providing a radiotracer in large quantities and of high radiopharmaceutical quality. Results: Here we publish an improved radiolabelling method and quality control procedures for high molar activity [18 F]TFB. The use of ammonium hydroxide for [18 F]fluoride elution, commercially available boron trifluoride-methanol complex dissolved in acetonitrile as precursor and removal of unreacted [18 F]fluoride on Florisil solid-phase extraction cartridges resulted in the reliable production of [18 F]TFB on SYNTHRA and TRACERLAB FXFN automated synthesizers with radiochemical yields in excess of 30%, radiochemical purities in excess of 98% and molar activities in the range of 34–217 GBq/µmol at the end of synthesis. PET scanning of a mouse lung tumour model carrying a NIS reporter gene rendered images of high quality and improved sensitivity. Conclusions: A novel automated radiosynthesis procedure for [18 F]tetrafluoroborate has been developed that provides the radiotracer with high molar activity, suitable for preclinical imaging of NIS reporter gene. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
20. Peripubertal exposure to oxyfluorfen, a diphenyl herbicide, delays pubertal development in the male rat by antagonizing androgen receptor activity.
- Author
-
Murr AS, Buckalew AR, Devane G, Bailey JR, Ford JL, Gray LE, and Stoker TE
- Subjects
- Animals, Male, Rats, Testosterone blood, Rats, Sprague-Dawley, Androgen Receptor Antagonists toxicity, Organ Size drug effects, Dose-Response Relationship, Drug, Prostate drug effects, Prostate metabolism, Thyrotropin blood, Seminal Vesicles drug effects, Seminal Vesicles metabolism, Thyroxine blood, Triiodothyronine blood, Endocrine Disruptors toxicity, Luteinizing Hormone blood, Herbicides toxicity, Receptors, Androgen metabolism, Receptors, Androgen drug effects, Sexual Maturation drug effects, Halogenated Diphenyl Ethers toxicity
- Abstract
We recently identified the herbicide oxyfluorfen as an inhibitor of iodide uptake by the sodium iodide symporter, a key step in thyroid hormone synthesis, using in vitro assays. We also observed a suppression of serum T4 and T3 in juvenile rats exposed orally to oxyfluorfen for 4 to 8-d. The purpose of the present study was to further evaluate the effects of an extended 31-d oral exposure using a male pubertal rat study (15 to 500 mg/kg). Oxyfluorfen delayed puberty at all doses (1.3 to 3.5-d) suppressing ventral prostate at 62.5 mg/kg and above and seminal vesicle weights at 31.25 mg/kg and above with no effect on testosterone or luteinizing hormone. Serum T4 and T3 were suppressed by all doses up to 80%, with a linear increase in serum TSH. Based on delayed puberty without changes in testosterone, we hypothesized that oxyfluorfen interferes with androgen receptor (AR) function. Results from our Hershberger study, with oxyfluorfen (62.5 and 125 mg/kg) co-treated with testosterone propionate (TP, 1 mg/kg) for 10-d showed 3 of 5 of the androgenic tissue weights were suppressed compared with TP alone indicating AR antagonism. We next confirmed this effect in an in vitro AR transcriptional activation reporter assay (0 to 20 μM) with 125 pM 5αDH-11-ketotestosterone and found concentration-dependent inhibition of AR luminescence activity (EC50 1.75 µM) without cytotoxicity. Thus, this study confirms the endocrine-disrupting mechanism of oxyfluorfen using in vitro and in vivo evaluations of the thyroid hormone and AR pathways. This abstract does not necessarily reflect U.S. EPA policy., (Published by Oxford University Press on behalf of the Society of Toxicology 2024.)
- Published
- 2025
- Full Text
- View/download PDF
21. Selective sodium iodide symporter (NIS) gene therapy of glioblastoma mediated by EGFR-targeted lipopolyplexes
- Author
-
Rebekka Spellerberg, Teoman Benli-Hoppe, Carolin Kitzberger, Simone Berger, Kathrin A. Schmohl, Nathalie Schwenk, Hsi-Yu Yen, Christian Zach, Franz Schilling, Wolfgang A. Weber, Roland E. Kälin, Rainer Glass, Peter J. Nelson, Ernst Wagner, and Christine Spitzweg
- Subjects
sodium iodide symporter ,NIS ,glioblastoma ,GBM ,gene therapy ,polyplexes ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Lipo-oligomers, post-functionalized with ligands to enhance targeting, represent promising new vehicles for the tumor-specific delivery of therapeutic genes such as the sodium iodide symporter (NIS). Due to its iodide trapping activity, NIS is a powerful theranostic tool for diagnostic imaging and the application of therapeutic radionuclides. 124I PET imaging allows non-invasive monitoring of the in vivo biodistribution of functional NIS expression, and application of 131I enables cytoreduction. In our experimental design, we used epidermal growth factor receptor (EGFR)-targeted polyplexes (GE11) initially characterized in vitro using 125I uptake assays. Mice bearing an orthotopic glioblastoma were treated subsequently with mono-dibenzocyclooctyne (DBCO)-PEG24-GE11/NIS or bisDBCO-PEG24-GE11/NIS, and 24–48 h later, 124I uptake was assessed by positron emission tomography (PET) imaging. The best-performing polyplex in the imaging studies was then selected for 131I therapy studies. The in vitro studies showed EGFR-dependent and NIS-specific transfection efficiency of the polyplexes. The injection of monoDBCO-PEG24-GE11/NIS polyplexes 48 h before 124I application was characterized to be the optimal regime in the imaging studies and was therefore used for an 131I therapy study, showing a significant decrease in tumor growth and a significant extension of survival in the therapy group. These studies demonstrate the potential of EGFR-targeted polyplex-mediated NIS gene therapy as a new strategy for the therapy of glioblastoma.
- Published
- 2021
- Full Text
- View/download PDF
22. Basolateral Sorting of the Sodium/Iodide Symporter Is Mediated by Adaptor Protein 1 Clathrin Adaptor Complexes.
- Author
-
Koumarianou, Petrina, Fernández-Méndez, Celia, Fajardo-Delgado, Dánae, Mielu, Lidia Mirella, Santisteban, Pilar, and De la Vieja, Antonio
- Subjects
- *
ADAPTOR proteins , *THYROID cancer , *CLATHRIN , *COATED vesicles , *TRANSFERRIN receptors , *MEMBRANE proteins - Abstract
Background: The sodium/iodide symporter (NIS) is a transmembrane protein located on the basolateral membrane of thyrocytes. Despite its physiological and clinical relevance, little is known about the mechanisms that mediate NIS subcellular sorting. In the present study, we examined NIS basolateral trafficking in vitro using non-thyroid and thyroid epithelial cells. Methods: Immunofluorescence and Western blotting were performed to analyze NIS subcellular location and function in cells grown in monolayers under unpolarized and/or polarized conditions. Strategic NIS residues were mutated, and binding of NIS to clathrin adaptor complexes was determined by immunoprecipitation. Results: We show that NIS reaches the plasma membrane (PM) through a thyrotropin-dependent mechanism 24 hours after treatment with the hormone. We demonstrate that NIS basolateral trafficking is a clathrin-mediated mechanism, in which the clathrin adaptor complexes AP-1 (A and B) sort NIS from the trans-Golgi network (TGN) and recycling endosomes (REs). Specifically, we show that the AP-1B μ1 subunit controls NIS basolateral sorting through common REs. In its absence, NIS is apically missorted but remains functional. Additionally, direct NIS basolateral transport from the TGN to the basolateral membrane is mediated by AP-1A through clathrin-coated vesicles that also carry the transferrin receptor. Loss of the μ1 subunit of AP-1A is functionally compensated by AP-1B. Furthermore, loss of both subunits diminishes NIS trafficking to the PM. Finally, we demonstrate that AP-1A binds to the L121 and LL562/563 residues on NIS, whereas AP-1B binds to L583. Conclusions: Our findings highlight the novel involvement of the clathrin-coated machinery in basolateral NIS trafficking. Given that AP-1A expression is reduced in tumors, and its expression correlates with that of NIS, these findings will help uncover new targets in thyroid cancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
23. Efficacy of Combination Therapy with Lenvatinib and Radioactive Iodine in Thyroid Cancer Preclinical Model.
- Author
-
Suzuki, Kensuke, Iwai, Hiroshi, Utsunomiya, Keita, Kono, Yumiko, Watabe, Tadashi, Kobayashi, Yoshiki, Bui, Dan Van, Sawada, Shunsuke, Yun, Yasutaka, Mitani, Akitoshi, Fukui, Kenta, Sakai, Haruka, Chu, Hanh Hong, Linh, Nguyen Manh, Tanigawa, Noboru, and Kanda, Akira
- Subjects
- *
THYROID cancer , *ANIMAL models in research , *SODIUM iodide , *IODINE isotopes , *CELL migration , *TUMOR growth - Abstract
Patients with differentiated thyroid cancer (DTC) usually have good prognosis, while those with advanced disease have poor clinical outcomes. This study aimed to investigate the antitumor effects of combination therapy with lenvatinib and 131I (CTLI) using three different types of DTC cell lines with different profiling of sodium iodide symporter (NIS) status. The radioiodine accumulation study revealed a significantly increased radioiodine uptake in K1-NIS cells after lenvatinib treatment, while there was almost no uptake in K1 and FTC-133 cells. However, lenvatinib administration before radioiodine treatment decreased radioiodine uptake of K1-NIS xenograft tumor in the in vivo imaging study. CTLI synergistically inhibited colony formation and DTC cell migration, especially in K1-NIS cells. Finally, 131I treatment followed by lenvatinib administration significantly inhibited tumor growth of the NIS-expressing thyroid cancer xenograft model. These results provide important clinical implications for the combined therapy that lenvatinib should be administered after 131I treatment to maximize the treatment efficacy. Our synergistic treatment effects by CTLI suggested its effectiveness for RAI-avid thyroid cancer, which retains NIS function. This potential combination therapy suggests a powerful and tolerable new therapeutic strategy for advanced thyroid cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
24. The sodium iodide symporter (NIS) as theranostic gene: its emerging role in new imaging modalities and non-viral gene therapy.
- Author
-
Kitzberger, Carolin, Spellerberg, Rebekka, Morath, Volker, Schwenk, Nathalie, Schmohl, Kathrin A., Schug, Christina, Urnauer, Sarah, Tutter, Mariella, Eiber, Matthias, Schilling, Franz, Weber, Wolfgang A., Ziegler, Sibylle, Bartenstein, Peter, Wagner, Ernst, Nelson, Peter J., and Spitzweg, Christine
- Subjects
GENE therapy ,SODIUM iodide ,HIGH resolution imaging ,REPORTER genes ,MESENCHYMAL stem cells ,POSITRON emission tomography ,RADIOACTIVE tracers - Abstract
Cloning of the sodium iodide symporter (NIS) in 1996 has provided an opportunity to use NIS as a powerful theranostic transgene. Novel gene therapy strategies rely on image-guided selective NIS gene transfer in non-thyroidal tumors followed by application of therapeutic radionuclides. This review highlights the remarkable progress during the last two decades in the development of the NIS gene therapy concept using selective non-viral gene delivery vehicles including synthetic polyplexes and genetically engineered mesenchymal stem cells. In addition, NIS is a sensitive reporter gene and can be monitored by high resolution PET imaging using the radiotracers sodium [
124 I]iodide ([124 I]NaI) or [18 F]tetrafluoroborate ([18 F]TFB). We performed a small preclinical PET imaging study comparing sodium [124 I]iodide and in-house synthesized [18 F]TFB in an orthotopic NIS-expressing glioblastoma model. The results demonstrated an improved image quality using [18 F]TFB. Building upon these results, we will be able to expand the NIS gene therapy approach using non-viral gene delivery vehicles to target orthotopic tumor models with low volume disease, such as glioblastoma. Trial registration not applicable. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
25. Targeted Next-Generation Sequencing of Congenital Hypothyroidism-Causative Genes Reveals Unexpected Thyroglobulin Gene Variants in Patients with Iodide Transport Defect.
- Author
-
Bernal Barquero, Carlos Eduardo, Geysels, Romina Celeste, Jacques, Virginie, Carro, Gerardo Hernán, Martín, Mariano, Peyret, Victoria, Abregú, María Celeste, Papendieck, Patricia, Masini-Repiso, Ana María, Savagner, Frédérique, Chiesa, Ana Elena, Citterio, Cintia E., and Nicola, Juan Pablo
- Subjects
- *
GENETIC variation , *NUCLEOTIDE sequencing , *RECESSIVE genes , *TRANSPORTATION of patients , *CONGENITAL hypothyroidism , *THYROGLOBULIN , *THYROID gland - Abstract
Congenital iodide transport defect is an uncommon autosomal recessive disorder caused by loss-of-function variants in the sodium iodide symporter (NIS)-coding SLC5A5 gene and leading to dyshormonogenic congenital hypothyroidism. Here, we conducted a targeted next-generation sequencing assessment of congenital hypothyroidism-causative genes in a cohort of nine unrelated pediatric patients suspected of having a congenital iodide transport defect based on the absence of 99mTc-pertechnetate accumulation in a eutopic thyroid gland. Although, unexpectedly, we could not detect pathogenic SLC5A5 gene variants, we identified two novel compound heterozygous TG gene variants (p.Q29* and c.177-2A>C), three novel heterozygous TG gene variants (p.F1542Vfs*20, p.Y2563C, and p.S523P), and a novel heterozygous DUOX2 gene variant (p.E1496Dfs*51). Splicing minigene reporter-based in vitro assays revealed that the variant c.177-2A>C affected normal TG pre-mRNA splicing, leading to the frameshift variant p.T59Sfs*17. The frameshift TG variants p.T59Sfs*17 and p.F1542Vfs*20, but not the DUOX2 variant p.E1496Dfs*51, were predicted to undergo nonsense-mediated decay. Moreover, functional in vitro expression assays revealed that the variant p.Y2563C reduced the secretion of the TG protein. Our investigation revealed unexpected findings regarding the genetics of congenital iodide transport defects, supporting the existence of yet to be discovered mechanisms involved in thyroid hormonogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
26. Bone Marrow Mesenchymal Stem Cell-Mediated Radiosensitive Promoter-Combined Sodium Iodide Symporter for the Treatment of Breast Cancer.
- Author
-
Zhang, Lu, Xi, Yue, Guo, Rui, Miao, Ying, Chen, Hong, Zhang, Min, and Li, Biao
- Subjects
- *
SODIUM iodide , *ADIPOGENESIS , *BONE marrow , *REVERSE transcriptase polymerase chain reaction , *BREAST cancer , *BONE growth , *TRIPLE-negative breast cancer - Abstract
To develop genetically engineered bone marrow mesenchymal stem cells (BMSCs) that carries a radiotherapy gene to target triple-negative breast cancer (TNBC) and to evaluate the efficacy of radiation damage within the tumor microenvironment. The early growth response protein1 (Egr1)-human sodium iodide symporter (hNIS) gene was transfected into BMSCs by lentiviral transfection and the expression levels were evaluated by quantitative reverse transcription polymerase chain reaction. Transwell and adipogenesis and osteogenesis assays were performed to determine the targeting properties and adipogenic and osteogenic characteristics of the transgenic stem cells. The uptake of radioiodine and the efflux characteristics of the transgenic stem cells were determined by iodine uptake experiments. 131I-SPECT imaging was used to determine the characteristics of targeting to TNBC and to quantify the iodine uptake of transgenic stem cells in vivo. The effects of 131I treatment on BMSCs were characterized using tumor growth, immune cell infiltration, and tumor invasion endpoints based on immunohistochemistry and flow cytometry analysis of tumor samples. BMSCs-Egr1-hNIS cells abundantly express hNIS after radiation induction and are chemotactically attracted to TNBC tumors. Iodine uptake of BMSCs-Egr1-hNIS gradually increased with increasing induction concentrations and times. When the inductive concentration of 131I was >100 μCi/mL and lasted for 36 h, the rate of iodine uptake in cells increased. In vitro, the radioiodine quickly flowed out from cells within 20 min but in vivo, the rate of radioiodine loss was significantly slower and occurred over 24 h. After 131I therapy, tumor growth was inhibited, white blood cells infiltrated into tumor site and the levels of invasion-related cytokines significantly decreased. BMSCs-Egr1-hNIS-mediated 131I therapy can achieve precisely targeted radiotherapy to inhibit tumor growth, promote immune cell infiltration to the tumor sites, and reduce the invasiveness and metastasis characteristics of tumor cells. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
27. Targeting GLI1 Transcription Factor for Restoring Iodine Avidity with Redifferentiation in Radioactive-Iodine Refractory Thyroid Cancers.
- Author
-
Oh, Ji Min, Rajendran, Ramya Lakshmi, Gangadaran, Prakash, Hong, Chae Moon, Jeong, Ju Hye, Lee, Jaetae, and Ahn, Byeong-Cheol
- Subjects
- *
THERAPEUTIC use of iodine , *DRUG efficacy , *BIOCHEMISTRY , *THYROID gland tumors , *GENE expression , *PHENOMENOLOGY , *RADIOACTIVE elements , *DESCRIPTIVE statistics , *TRANSCRIPTION factors , *IODINE - Abstract
Simple Summary: Thyroid cancers have an excellent prognosis by standard therapy of surgery followed by radioactive-iodine therapy. However, metastatic thyroid cancers do not response to radioactive-iodine therapy by losing iodine avidity. Therefore, reversing iodine avidity to metastatic thyroid cancers gives a new chance of applying radioactive-iodine therapy to the cancers. In the current study, we found that GLI1 knockdown can revert iodine non-avid thyroid cancers to iodine avid cancers by increasing expression of thyroid-specific proteins. Restoration of iodine avidity in thyroid cancers makes the cancers sensitive to radioactive-iodine therapy again. Therefore, the GLI1 can be a potential therapeutic target of radioactive-iodine resistant thyroid cancers. Radioactive-iodine (RAI) therapy is the mainstay for patients with recurrent and metastatic thyroid cancer. However, many patients exhibit dedifferentiation characteristics along with lack of sodium iodide symporter (NIS) functionality, low expression of thyroid-specific proteins, and poor RAI uptake, leading to poor prognosis. Previous studies have demonstrated the effect of GLI family zinc finger 1 (GLI1) inhibition on tumor growth and apoptosis. In this study, we investigated the role of GLI1 in the context of redifferentiation and improvement in the efficacy of RAI therapy for thyroid cancer. We evaluated GLI1 expression in several thyroid cancer cell lines and selected TPC-1 and SW1736 cell lines showing the high expression of GLI. We performed GLI1 knockdown and evaluated the changes of thyroid-specific proteins expression, RAI uptake and I-131-mediated cytotoxicity. The effect of GANT61 (GLI1 inhibitor) on endogenous NIS expression was also assessed. Endogenous NIS expression upregulated by inhibiting GLI1, in addition, increased expression level in plasma membrane. Also, GLI1 knockdown increased expression of thyroid-specific proteins. Restoration of thyroid-specific proteins increased RAI uptake and I-131-mediated cytotoxic effect. Treatment with GANT61 also increased expression of endogenous NIS. Targeting GLI1 can be a potential strategy with redifferentiation for restoring RAI avidity in dedifferentiated thyroid cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
28. FOXA1 Regulation Turns Benzamide HDACi Treatment Effect-Specific in BC, Promoting NIS Gene-Mediated Targeted Radioiodine Therapy
- Author
-
Maitreyi Rathod, Madhura Kelkar, Snehal Valvi, Girish Salve, and Abhijit De
- Subjects
sodium iodide symporter ,breast cancer ,benzamide ,HDAC inhibitor ,transcriptional factor ,radio-iodine therapy, FOXA1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Human sodium iodide symporter (NIS) gene mediated radio-ablation is a successful procedure in thyroid cancer clinics. In recent years, natural expression of NIS is reported in breast cancer (BC) cases but is yet to make its mark as a therapeutic procedure in BC clinics. A pre-exposure to histone deacetylase (HDAC) inhibitors to amplify endogenous NIS expression was attempted, but achieving cancer tissue-specific enhancement of NIS in patients is an important challenge to win. Here, for the first time, we show that a benzamide class of HDACi (bHDACi) can significantly induce NIS gene expression and function (p < 0.05) in BC cells with minimal off-target effects. Transcription factor (TF) profiler and promoter binding array reveals 22 TFs differentially activated by CI-994, of which FOXA1 is identified as a unique and positive regulator of NIS. Clonogenic assay shows reduced survival with bHDACi + 131I combination treatment. Further, AR-42 and MS-275 treatment shows enhanced NIS expression in an orthotopic breast tumor model. Combining bHDACi with 1 mCi 131I shows 40% drop in signal (p < 0.05), indicating enhanced radio-ablation effect. Cerenkov imaging revealed higher accumulation of 131I in MS-275-treated tumors. Thus, bHDACi-mediated selective enhancement ensuring minimal off-target effect is a step further toward using NIS as a therapeutic target for BC.
- Published
- 2020
- Full Text
- View/download PDF
29. In Vivo Myoblasts Tracking Using the Sodium Iodide Symporter Gene Expression in Dogs
- Author
-
Isabel Punzón, David Mauduit, Bryan Holvoet, Jean-Laurent Thibaud, Pauline de Fornel, Christophe M. Deroose, Nicolas Blanchard-Gutton, Jean-Thomas Vilquin, Maurilio Sampaolesi, Inès Barthélémy, and Stéphane Blot
- Subjects
myoblast transplantation ,sodium iodide symporter ,SPECT/CT ,in vivo imaging ,GRMD ,cell therapy ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Stem cell-based therapies are a promising approach for the treatment of degenerative muscular diseases; however, clinical trials have shown inconclusive and even disappointing results so far. Noninvasive cell monitoring by medicine imaging could improve the understanding of the survival and biodistribution of cells following injection. In this study, we assessed the canine sodium iodide symporter (cNIS) reporter gene as an imaging tool to track by single-photon emission computed tomography (SPECT/CT) transduced canine myoblasts after intramuscular (IM) administrations in dogs. cNIS-expressing cells kept their myogenic capacities and showed strong 99 mTc-pertechnetate (99 mTcO4−) uptake efficiency both in vitro and in vivo. cNIS expression allowed visualization of cells by SPECT/CT along time: 4 h, 48 h, 7 days, and 30 days after IM injection; biopsies collected 30 days post administration showed myofiber’s membranes expressing cNIS. This study demonstrates that NIS can be used as a reporter to track cells in vivo in the skeletal muscle of large animals. Our results set a proof of concept of the benefits NIS-tracking tool may bring to the already challenging cell-based therapies arena in myopathies and pave the way to a more efficient translation to the clinical setting from more accurate pre-clinical results.
- Published
- 2020
- Full Text
- View/download PDF
30. Radioiodine Therapy During Breastfeeding
- Author
-
Demir, Hakan, Özülker, Tamer, editor, Adaş, Mine, editor, and Günay, Semra, editor
- Published
- 2019
- Full Text
- View/download PDF
31. The sodium iodide symporter (NIS): novel applications for radionuclide imaging and treatment.
- Author
-
Spitzweg, C., Nelson, P. J., Wagner, E., Bartenstein, P., Weber, W. A., Schwaiger, M., and Morris, J. C.
- Subjects
- *
RADIONUCLIDE imaging , *SODIUM iodide , *THYROID cancer , *REPORTER genes , *GENE therapy , *GENETIC transformation - Abstract
Cloning of the sodium iodide symporter (NIS) 25 years ago has o pened an exciting chapter in molecular thyroidology with the characterization of NIS as one of the most powerful theranostic genes and the development of a promising gene therapy strategy based on image-guided selective NIS gene transfer in non-thyroidal tumors followed by application of 131I or alternative radionuclides, such as 188Re and 211At. Over the past two decades, significant progress has been made in the development of the NIS gene therapy concept, from local NIS gene delivery towards promising new applications in disseminated disease, in particular through the use of oncolytic viruses, non-viral polyplexes, and genetically engineered MSCs as highly effective, highly selective and flexible gene delivery vehicles. In addition to allowing the robust therapeutic application of radioiodine in non-thyroid cancer settings, these studies have also been able to take advantage of NIS as a sensitive reporter gene that allows tempo ral and spatial monitoring of vector biodistribution, replication, and elimination - critically important issues for preclinical development and clinical translation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
32. Characterization of Genetic Variants in the SLC5A5 Gene and Associations With Breast Milk Iodine Concentration in Lactating Women of African Descent: The NUPED Study
- Author
-
Sicelosethu S. Siro, Jeannine Baumgartner, Maryke Schoonen, Jennifer Ngounda, Linda Malan, Elizabeth A. Symington, Cornelius M. Smuts, and Lizelle Zandberg
- Subjects
breast milk ,iodine concentration ,lactating women ,SLC5A5 gene ,sodium iodide symporter ,urinary iodine concentration ,Nutrition. Foods and food supply ,TX341-641 - Abstract
Background: The sodium iodide symporter is responsible for the transfer of iodine into breast milk and is encoded for by the SLC5A5 gene. The role of genetic variants in the SLC5A5 gene locus in relation to the transfer of iodine from plasma into breast milk in healthy lactating individuals has, to our knowledge, not been explored.Objective: To identify and characterize possible genetic variants of the SLC5A5 gene in women of African descent living in urban South Africa, and to study associations with breast milk iodine concentrations (BMIC) in lactating women.Methods: This study is affiliated to the Nutrition during Pregnancy and Early Development (NuPED) cohort study (n = 250 enrolled pregnant women). In a randomly selected sub-sample of 32 women, the SLC5A5 gene was sequenced to identify known and novel variants. Of the identified variants, genotyping of selected variants was performed in all pregnant women who gave consent for genetic analyses (n = 246), to determine the frequency of the variants in the study sample. Urinary iodine concentration (UIC) in spot urine samples and BMIC were measured to determine iodine status. Associations of SLC5A5 genetic variants with BMIC were studied in lactating women (n = 55).Results: We identified 27 variants from sequencing of gene exomes and 10 variants were selected for further study. There was a significant difference in BMIC between the genotypes of the rs775249401 variant (P = 0.042), with the homozygous GG group having lower BMIC [86.8 (54.9–167.9) μg/L] compared to the (A) allele carriers rs775249401(AG+AA) [143.9 (122.4–169.3) μg/L] (P = 0.042). Of the rs775249401(GG), 49% had UIC
- Published
- 2021
- Full Text
- View/download PDF
33. Defending Effects of Iodide Transfer in Placental Barrier Against Maternal Iodine Deficiency.
- Author
-
Sun, Yina, Han, Yuanyuan, Qian, Ming, Li, Yongmei, Ye, Yan, Lin, Laixiang, and Liu, Yuanjun
- Subjects
- *
IODINE , *IODINE deficiency , *PREGNANCY proteins , *AMNIOTIC liquid , *SODIUM iodide , *MESSENGER RNA - Abstract
Objective: Placental iodide transport is necessary for maintaining an adequate iodide supply to the developing fetus. We hypothesized that compounds from the placental barrier can compensate for decreases in maternal iodine intake and normalize fetal iodine levels. Methods: Pregnant rats administered different amounts of iodine (1.24, 2.5, 5, or 10 μg/day) were evaluated on gestational day (gd) 16 and 20. The iodine levels in maternal blood, amniotic fluid (AF), and placental tissue were estimated using As-Ce catalytic spectrophotometry. The protein and/or messenger RNA (mRNA) levels of sodium iodide symporter (NIS), pendrin, alpha-smooth muscle actin (α-SMA), and CD31 in the placental labyrinth, trophoblast cells isolated using laser capture microdissection (LCM), and/or fetomaternal thyroid were detected using immunoblotting, real-time polymerase chain reaction, and/or immunohistochemistry. Results: When iodine intake was reduced, iodine levels in maternal blood gradually decreased; however, placental iodine levels were not significantly different between groups on gd16 and gd20. Minimal changes were observed in AF iodine levels on gd16, and a mild decreasing trend was observed (iodine dose, 10 to 1.24 μg/day) on gd20. NIS protein, which was linearly distributed along the basolateral membrane of maternal–fetal thyroid follicles, gradually increased with decreasing iodine levels. Regarding iodine deficiency in the placental labyrinth on gd16 and gd20, pendrin and glycosylated NIS proteins were significantly upregulated in a dose-dependent manner. However, the mRNA levels were unchanged. Furthermore, the conversion of NIS protein from the nonglycosylated to the glycosylated form increased. In trophoblast cells isolated using LCM, PDS mRNA levels increased in the 1.24-μg/day group on gd16 but not NIS mRNA levels. There was a smaller α-SMA+ area in the labyrinth zone on gd16 and gd20; however, the proportional CD31+ area increased on gd16 and reduced on gd20 with decreased iodine levels. Conclusions: All mechanisms upregulating the expression of iodine transporters and changes in villous stroma and microvessel area in the placental labyrinth can promote iodide transfer from mother to fetus in iodine deficiency, especially before the onset of fetal thyroid function. Compensatory NIS protein regulation in the placenta against decreased iodine intake mainly occurs during translation and glycosylation modification after translation. Pendrin may be more important than NIS in the mediation of placental iodide transport. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
34. A review of species differences in the control of, and response to, chemical-induced thyroid hormone perturbations leading to thyroid cancer.
- Author
-
Foster, John R., Tinwell, Helen, and Melching-Kollmuss, Stephanie
- Abstract
This review summarises the current state of knowledge regarding the physiology and control of production of thyroid hormones, the effects of chemicals in perturbing their synthesis and release that result in thyroid cancer. It does not consider the potential neurodevelopmental consequences of low thyroid hormones. There are a number of known molecular initiating events (MIEs) that affect thyroid hormone synthesis in mammals and many chemicals are able to activate multiple MIEs simultaneously. AOP analysis of chemical-induced thyroid cancer in rodents has defined the key events that predispose to the development of rodent cancer and many of these will operate in humans under appropriate conditions, if they were exposed to high enough concentrations of the affecting chemicals. There are conditions however that, at the very least, would indicate significant quantitative differences in the sensitivity of humans to these effects, with rodents being considerably more sensitive to thyroid effects by virtue of differences in the biology, transport and control of thyroid hormones in these species as opposed to humans where turnover is appreciably lower and where serum transport of T4/T3 is different to that operating in rodents. There is heated debate around claimed qualitative differences between the rodent and human thyroid physiology, and significant reservations, both scientific and regulatory, still exist in terms of the potential neurodevelopmental consequences of low thyroid hormone levels at critical windows of time. In contrast, the situation for the chemical induction of thyroid cancer, through effects on thyroid hormone production and release, is less ambiguous with both theoretical, and actual data, showing clear dose-related thresholds for the key events predisposing to chemically induced thyroid cancer in rodents. In addition, qualitative differences in transport, and quantitative differences in half life, catabolism and turnover of thyroid hormones, exist that would not operate under normal situations in humans. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
35. Review of the possible association between thyroid and breast carcinoma
- Author
-
Liangbo Dong, Jun Lu, Bangbo Zhao, Weibin Wang, and Yupei Zhao
- Subjects
Thyroid cancer ,Breast cancer ,Iodine ,Sodium iodide symporter ,Thyroid hormone ,Thyroid hormone receptor ,Surgery ,RD1-811 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Thyroid and breast cancer are two of the malignant diseases with highest incidence in females. Based on clinical experience, breast and thyroid cancer often occur metachronously or synchronously. Therefore, thyroid and breast cancer might share some common etiological factors. The relationship between these diseases has attracted substantial attention, and because these two glands are both regulated by the hypothalamic-pituitary axis, such a relationship is not surprising. A study of this relationship will be useful for obtaining a better understanding of the mechanism by which these two malignancies co-occur. Main body This study reviewed the progress in research on the roles of iodine intake, folate metabolism, obesity, gonadal hormones, and thyroid hormone in thyroid and breast cancer. These studies evaluating the etiological roles of these factors in linking breast and thyroid cancer might also improve our understanding and identify new therapeutic approaches, such as sodium/iodide symporter-mediated radioiodine therapy and thyroid-stimulating hormone receptor antagonists, for breast cancer. In addition, some specific treatments for each cancer, such as radiotherapy for breast cancer or radioactive iodine therapy for thyroid cancer, might be risk factors for secondary malignances, including breast and thyroid cancer. Conclusions Studies of the precise relationship between the co-occurrence of breast and thyroid cancer will certainly improve our understanding of the biological behaviors of these two malignancies and direct evidence-based clinical practice.
- Published
- 2018
- Full Text
- View/download PDF
36. Iodide Transporters in the Endometrium: A Potential Diagnostic Marker for Women with Recurrent Pregnancy Failures.
- Author
-
Bilal, Mahmood Y., Dambaeva, Svetlana, Brownstein, David, Kwak-Kim, Joanne, Gilman-Sachs, Alice, Beaman, Kenneth D., Bilal, Mahmood Yousif, Beaman, Kenneth D, and Bilal, Mahmood Y
- Subjects
- *
ENDOMETRIUM , *IODIDES , *IODINE deficiency , *GERM cells , *PREGNANCY - Abstract
Objective: The element iodine is an essential nutrient utilized by the thyroid glands, and deficiency of this element has been linked to reproductive failures. Iodide transporters are also present in reproductive tissues and cells of embryonic origin such as the endometrium and trophoblasts, respectively. The aim of this study is to understand if levels of iodide transporters are linked to pregnancy outcomes.Subjects and Methods: RNA derived from endometrial biopsies from controls or women with recurrent reproductive failures was analyzed utilizing RT-PCR and targeted RNASeq.Results: When compared to controls, women with 2 or more reproductive failures had a significant increase (>5 fold) in mRNA levels of the iodine transporters NIS and PENDRIN, but not thyroglobulin when probed vis RT-PCR. Targeted RNASeq analysis confirmed these findings when another group of patients were analyzed.Conclusion: These findings suggest possible abnormal iodine metabolism and a deficiency of iodine in endometrial tissues from some of the women with reproductive failures. We hypothesize from these findings that inorganic iodide and/or iodine is required for optimal cellular function in reproductive tissues, and that iodide transporters may potentially be used as a marker for infertility or for probing potential localized iodine deficiency that may not present in a typical thyroid panel analysis. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
37. A Novel Tyrosine Kinase Inhibitor Can Augment Radioactive Iodine Uptake Through Endogenous Sodium/Iodide Symporter Expression in Anaplastic Thyroid Cancer.
- Author
-
Oh, Ji Min, Baek, Se Hwan, Gangadaran, Prakash, Hong, Chae Moon, Rajendran, Ramya Lakshmi, Lee, Ho Won, Zhu, Liya, Gopal, Arunnehru, Kalimuthu, Senthilkumar, Jeong, Shin Young, Lee, Sang-Woo, Lee, Jaetae, and Ahn, Byeong-Cheol
- Subjects
- *
IODINE isotopes , *ANAPLASTIC thyroid cancer , *PROTEIN-tyrosine kinases , *KINASE inhibitors , *TREATMENT effectiveness , *SODIUM iodide , *THYROID cancer - Abstract
Background: Radioactive iodine (RAI) therapy is an important strategy in the treatment of thyroid cancer. However, anaplastic thyroid cancer (ATC), a rare malignancy, exhibits severe dedifferentiation characteristics along with a lack of sodium iodide symporter (NIS) expression and function. Therefore, RAI therapy is ineffective and contributes toward poor prognosis of these patients. Recently, small-molecule tyrosine kinase inhibitors (TKIs) have been used to treat thyroid cancer patients for restoring NIS expression and function and RAI uptake capacity. However, most results reported thus far are associated with differentiated thyroid cancer. In this study, we identified a new TKI and investigated its effects on cell redifferentiation, NIS function, and RAI therapy in ATC. Methods: We identified a new TKI, "5-(5-{4H, 5H,6H-cyclopenta[b]thiophen-2-yl}-1,3,4-oxadiazol-2-yl)-1-methyl-1,2-dihydropyridin-2-one" (CTOM-DHP), using a high-throughput screening system. CTOM-DHP was exposed to 8505C ATC cells at different concentrations and time points. Concentrations of 12.5 and 25 μM and an incubation time of 72 hours were chosen as the conditions for subsequent NIS promoter assays and NIS mRNA and protein expression experiments. In addition, we examined factors related to iodide metabolism after CTOM-DHP treatment as well as the signaling pathways mediating the effects of CTOM-DHP on endogenous NIS expression. RAI uptake and 131I cytotoxicity effects caused by CTOM-DHP pretreatment were also evaluated in vitro and in vivo. Results: Promoter assays as well as mRNA and protein expression analyses confirmed that NIS expression was augmented by treatment of 8505C ATC cells with CTOM-DHP. Moreover, CTOM-DHP treatment robustly increased the expression of other thyroid-specific proteins and thyroid transcription factors related to iodide metabolism. Enhancement of NIS function was demonstrated by an increase in 125I uptake and 131I cytotoxicity. Increased endogenous NIS expression was associated with the inhibition of PI3K/Akt and MAPK signaling pathways. In vivo results also demonstrated an increase in NIS promoter activity and RAI avidity in response to CTOM-DHP treatment. Furthermore, 131I-mediated therapeutic effects preferentially improved in a tumor xenograft mice model. Conclusions: CTOM-DHP, a new TKI identified in this study, enhances endogenous NIS expression and thereby is a promising compound for restoring RAI avidity in ATC. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
38. A Historical Perspective on the Use of Radionuclides for Therapy
- Author
-
Beylergil, Volkan, Carrasquillo, Jorge A., Weber, Wolfgang A., Larson, Steven M., Brady, Luther W., Series editor, Lu, Jiade J., Series editor, Nieder, Carsten, Series editor, and Baum, Richard P., editor
- Published
- 2014
- Full Text
- View/download PDF
39. Porous Graphene Oxide Decorated Ion Selective Electrode for Observing Across-Cytomembrane Ion Transport
- Author
-
Shihui Hu, Rong Zhang, and Yunfang Jia
- Subjects
porous graphene oxide ,interfacial micro-environment ,ion selective electrode ,ion transport ,sodium iodide symporter ,Chemical technology ,TP1-1185 - Abstract
The technology for measuring cytomembrane ion transport is one of the necessities in modern biomedical research due to its significance in the cellular physiology, the requirements for the non-invasive and easy-to-operate devices have driven lots of efforts to explore the potential electrochemical sensors. Herein, we would like to evidence the exploitation of the porous graphene oxide (PGO) decorated ion selective electrode (ISE) as a detector to capture the signal of cytomembrane ion transport. The tumor cells (MDAMB231, A549 and HeLa) treated by iodide uptake operation, with and without the sodium-iodide-symporter (NIS) expression, are used as proofs of concept. It is found that under the same optimized experimental conditions, the changed output voltages of ISEs before and after the cells’ immobilization are in close relation with the NIS related ion’s across-membrane transportation, including I−, Na+ and Cl−. The explanation for the measured results is proposed by clarifying the function of the PGO scaffold interfacial micro-environment (IME), that is, in this spongy-like micro-space, the NIS related minor ionic fluctuations can be accumulated and amplified for ISE to probe. In conclusion, we believe the integration of the microporous graphene derivatives-based IME and ISE may pave a new way for observing the cytomembrane ionic activities.
- Published
- 2020
- Full Text
- View/download PDF
40. In Vivo Imaging of MSCs
- Author
-
Bulte, Jeff W. M., Hematti, Peiman, editor, and Keating, Armand, editor
- Published
- 2013
- Full Text
- View/download PDF
41. Leveraging molecular targeted drugs and immune checkpoint inhibitors treat advanced thyroid carcinoma to achieve thyroid carcinoma redifferentiation.
- Author
-
Su JY, Huang T, Zhang JL, Lu JH, Wang ML, Yan J, Lin RB, Lin SY, and Wang J
- Abstract
Thyroid cancer can be classified into three different types based on the degree of differentiation: well-differentiated, poorly differentiated, and anaplastic thyroid carcinoma. Well-differentiated thyroid cancer refers to cancer cells that closely resemble normal thyroid cells, while poorly differentiated and anaplastic thyroid carcinoma are characterized by cells that have lost their resemblance to normal thyroid cells. Advanced thyroid carcinoma, regardless of its degree of differentiation, is known to have a higher likelihood of disease progression and is generally associated with a poor prognosis. However, the process through which well-differentiated thyroid carcinoma transforms into anaplastic thyroid carcinoma, also known as "dedifferentiation", has been a subject of intensive research. In recent years, there have been significant breakthroughs in the treatment of refractory advanced thyroid cancer. Clinical studies have been conducted to evaluate the efficacy and safety of molecular targeted drugs and immune checkpoint inhibitors in the treatment of dedifferentiated thyroid cancer. These drugs work by targeting specific molecules or proteins in cancer cells to inhibit their growth or by enhancing the body's immune response against the cancer cells. This article aims to explore some of the possible mechanisms behind the dedifferentiation process in well-differentiated thyroid carcinoma. It also discusses the clinical effects of molecular targeted drugs and immune checkpoint inhibitors in thyroid cancer patients with different degrees of differentiation. Furthermore, it offers insights into the future trends in the treatment of advanced thyroid cancer, highlighting the potential for improved outcomes and better patient care., Competing Interests: None., (AJCR Copyright © 2024.)
- Published
- 2024
42. Genetic Engineering of Therapeutic Cells with the Sodium Iodide Symporter (NIS) to Enable Noninvasive In Vivo Therapy Tracking.
- Author
-
Grimsdell B, Saleem A, Volpe A, and Fruhwirth GO
- Subjects
- Humans, Tomography, Emission-Computed, Single-Photon, Genetic Engineering, Positron-Emission Tomography methods, Symporters genetics, Symporters metabolism
- Abstract
Noninvasive long-term imaging of therapeutic cells in preclinical models can be achieved through introducing a reporter gene into the cells of interest. Despite important recent developments such as gene editing, cell engineering based on lentiviruses remains a mainstream tool for gene transfer applicable to a variety of different cell types.In this chapter, we describe how to use lentivirus-based genetic engineering to render different candidate cell therapies in vivo traceable by radionuclide imaging. We illustrate this reporter gene technology using the sodium iodide symporter (NIS), which is compatible with both positron emission tomography (PET) and single-photon emission computed tomography (SPECT). For preclinical experimentation, we fused NIS with a suitable fluorescent protein such as monomeric GFP or RFP to streamline cell line generation and downstream analyses of ex vivo tissue samples. We present protocols for reporter gene engineering of human cardiac progenitor cells, regulatory T cells, and effector T cells as well as for the characterization experiments required to validate NIS-fluorescent protein reporter function in these candidate therapeutic cells., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
43. Nasolacrimal Duct Obstruction and Lacrimal Surgery in Cancer Patients
- Author
-
Savar, Aaron, Esmaeli, Bita, and Esmaeli, Bita, editor
- Published
- 2011
- Full Text
- View/download PDF
44. Iodine and brain metabolism
- Author
-
Verheesen, R. H., Schweitzer, C. M., Preedy, Victor R., editor, Watson, Ronald Ross, editor, and Martin, Colin R., editor
- Published
- 2011
- Full Text
- View/download PDF
45. TGFB1-driven mesenchymal stem cell-mediated NIS gene transfer.
- Author
-
Schug, Christina, Urnauer, Sarah, Jaeckel, Carsten, Schmohl, Kathrin A., Tutter, Mariella, Steiger, Katja, Schwenk, Nathalie, Schwaiger, Markus, Wagner, Ernst, Nelson, Peter J., and Spitzweg, Christine
- Subjects
- *
GENETIC transformation , *MESENCHYMAL stem cells , *CANCER treatment , *PERCHLORATES , *TUMOR growth - Abstract
Based on their excellent tumor-homing capacity, genetically engineered mesenchymal stem cells (MSCs) are under investigation as tumor-selective gene delivery vehicles. Transgenic expression of the sodium iodide symporter (NIS) in genetically engineered MSCs allows noninvasive tracking of MSC homing by imaging of functional NIS expression as well as therapeutic application of 131I. The use of tumor stroma-activated promoters can improve tumor-specific MSC-mediated transgene delivery. The essential role of transforming growth factor B1 (TGFB1) and the SMAD downstream target in the signaling between tumor and the surrounding stroma makes the biology of this pathway a potential option to better control NIS expression within the tumor milieu. Bone marrow-derived MSCs were stably transfected with a NIS-expressing plasmid driven by a synthetic SMAD-responsive promoter (SMAD-NIS-MSCs). Radioiodide uptake assays revealed a 4.9-fold increase in NIS-mediated perchlorate-sensitive iodide uptake in SMAD-NIS-MSCs after TGFB1 stimulation compared to unstimulated cells demonstrating the successful establishment of MSCs, which induce NIS expression in response to activation of TGFB1 signaling using a SMAD-responsive promoter. 123I-scintigraphy revealed significant tumor-specific radioiodide accumulation and thus NIS expression after systemic application of SMAD-NIS-MSCs into mice harboring subcutaneous tumors derived from the human hepatocellular carcinoma (HCC) cell line HuH7, which express TGFB1. 131I therapy in SMAD-NIS-MSCs-treated mice demonstrated a significant delay in tumor growth and prolonged survival. Making use of the tumoral TGFB1 signaling network in the context of MSC-mediated NIS gene delivery is a promising approach to foster tumor stroma-selectivity of NIS transgene expression and tailor NIS-based gene therapy to TGFB1-rich tumor environments. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
46. Human sodium iodide transporter gene‑mediated imaging and therapy of mouse glioma, comparison between 188 Re and 131 I.
- Author
-
Guo, Rui, Xi, Yun, Zhang, Min, Miao, Ying, Zhang, Miao, and Li, Biao
- Subjects
- *
GLIOMA treatment , *CANCER radiotherapy , *SODIUM cotransport systems , *SODIUM iodide , *CANCER cells , *TREATMENT effectiveness - Abstract
Novel treatment options are urgently required for patients with glioma who are not effectively treated through standard therapy. Human sodium iodide symporter (hNIS) is a molecular target of certain tumors types. Compared with 131I, 188Re possesses a higher energy and shorter half‑life; therefore, the effects of 188Re and 131I were compared in hNIS‑mediated gene imaging and therapy in the present study. Recombinant human brain glioma cell line U87 was transfected with a recombinant lentiviral vector containing hNIS (U87‑hNIS). U87‑0 cell line transfected with blank lentivirus was prepared as a control. In vitro, the 188Re and 131I uptake of U87‑hNIS cells were 21.3‑times and 25.9‑times that of the control groups, however the excretion rate of the two nuclides was very rapid, and the half‑life was only ~4 min. Sodium perchlorate inhibited hNIS‑mediated 188Re and 131I uptake to levels observed in the control groups. 188Re and 131I were able to kill U87‑hNIS cells selectively, with a survival of only 21.6 and 36.2%, respectively. U87‑hNIS nude mice appeared to accumulate 188Re, with a ratio of radioactivity counts between tumor and non‑tumor sites of ~13.5 compared with 10.3 of 131I 1 h after radionuclide injection. In contrast with in vitro studies, U87‑hNIS cells demonstrated a notable increase in 188Re retention in vivo, even 24 h after 188Re injection. U87‑hNIS cells also exhibited increased 131I retention in vivo; however, as the time increased, 131I was rapidly released with the tumor no longer able to be imaged 24 h after 131I injection. Following treatment, U87‑hNIS tumors experienced a volume reduction of 24.1%, whereas U87‑0 cells demonstrated an increase of 28.8%. 188Re and 131I were revealed to be effective at decreasing tumor volume compared with the control. However, 188Re was significantly more potent compared with 131I (P<0.01). The present study indicated that the U87‑hNIS cell line is sufficient to induce specific 188Re and 131I uptake, which may kill cells in vitro and in vivo. 188Re exhibited an increased retention time in vivo compared with 131I, which facilitates the imaging and therapy of U87‑hNIS tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
47. DNA Microarray Analysis of Effects of TSH, Iodide, Cytokines, and Therapeutic Agents on Gene Expression in Cultured Human Thyroid Follicles
- Author
-
Sato, Kanji, Yamazaki, Kazuko, Yamada, Emiko, Conn, P. Michael, editor, Handwerger, Stuart, editor, and Aronow, Bruce, editor
- Published
- 2008
- Full Text
- View/download PDF
48. Enzymes/Transporters
- Author
-
Boy, Regine Garcia, Knapp, Eva-Maria, Eisenhut, Michael, Haberkorn, Uwe, Mier, Walter, Hofmann, F., editor, Semmler, Wolfhard, editor, and Schwaiger, Markus, editor
- Published
- 2008
- Full Text
- View/download PDF
49. PET and SPECT
- Author
-
Haberkorn, Uwe, Hofmann, F., editor, Semmler, Wolfhard, editor, and Schwaiger, Markus, editor
- Published
- 2008
- Full Text
- View/download PDF
50. Future Trends: Molecular PET
- Author
-
Mohnike, Wolfgang, editor, Hör, Gustav, editor, and Schelbert, Heinrich R., editor
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.