1
|
Wang Y, Wang Z, Yu X, Wang X, Song J, Yu DJ, Ge F. MORE: a multi-omics data-driven hypergraph integration network for biomedical data classification and biomarker identification. Brief Bioinform 2024; 26:bbae658. [PMID: 39692449 DOI: 10.1093/bib/bbae658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
High-throughput sequencing methods have brought about a huge change in omics-based biomedical study. Integrating various omics data is possibly useful for identifying some correlations across data modalities, thus improving our understanding of the underlying biological mechanisms and complexity. Nevertheless, most existing graph-based feature extraction methods overlook the complementary information and correlations across modalities. Moreover, these methods tend to treat the features of each omics modality equally, which contradicts current biological principles. To solve these challenges, we introduce a novel approach for integrating multi-omics data termed Multi-Omics hypeRgraph integration nEtwork (MORE). MORE initially constructs a comprehensive hyperedge group by extensively investigating the informative correlations within and across modalities. Subsequently, the multi-omics hypergraph encoding module is employed to learn the enriched omics-specific information. Afterward, the multi-omics self-attention mechanism is then utilized to adaptatively aggregate valuable correlations across modalities for representation learning and making the final prediction. We assess MORE's performance on datasets characterized by message RNA (mRNA) expression, Deoxyribonucleic Acid (DNA) methylation, and microRNA (miRNA) expression for Alzheimer's disease, invasive breast carcinoma, and glioblastoma. The results from three classification tasks highlight the competitive advantage of MORE in contrast with current state-of-the-art (SOTA) methods. Moreover, the results also show that MORE has the capability to identify a greater variety of disease-related biomarkers compared to existing methods, highlighting its advantages in biomedical data mining and interpretation. Overall, MORE can be investigated as a valuable tool for facilitating multi-omics analysis and novel biomarker discovery. Our code and data can be publicly accessed at https://github.com/Wangyuhanxx/MORE.
Collapse
Affiliation(s)
- Yuhan Wang
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing 210094, China
| | - Zhikang Wang
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
| | - Xuan Yu
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong 999077, China
| | - Xiaoyu Wang
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing 210094, China
| | - Fang Ge
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan, Nanjing 210023, China
| |
Collapse
|
2
|
Ansari MM, Sahu SK, Singh TG, Singh SRJ, Kaur P. Evolving significance of kinase inhibitors in the management of Alzheimer's disease. Eur J Pharmacol 2024; 979:176816. [PMID: 39038637 DOI: 10.1016/j.ejphar.2024.176816] [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: 03/23/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's disease is a neurodegenerative problem with progressive loss of memory and other cognitive function disorders resulting in the imbalance of neurotransmitter activity and signaling progression, which poses the need of the potential therapeutic target to improve the intracellular signaling cascade brought by kinases. Protein kinase plays a significant and multifaceted role in the treatment of Alzheimer's disease, by targeting pathological mechanisms like tau hyperphosphorylation, neuroinflammation, amyloid-beta production and synaptic dysfunction. In this review, we thoroughly explore the essential protein kinases involved in Alzheimer's disease, detailing their physiological roles, regulatory impacts, and the newest inhibitors and compounds that are progressing into clinical trials. All the findings of studies exhibited the promising role of kinase inhibitors in the management of Alzheimer's disease. However, it still poses the need of addressing current challenges and opportunities involved with this disorder for the future perspective of kinase inhibitors in the management of Alzheimer's disease. Further study includes the development of biomarkers, combination therapy, and next-generation kinase inhibitors with increased potency and selectivity for its future prospects.
Collapse
Affiliation(s)
- Md Mustafiz Ansari
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | | | - Sovia R J Singh
- University Language Centre- Chitkara Business School, Chitkara University, Punjab, India
| | - Paranjeet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
3
|
Mechanisms of Sodium/Iodide Symporter-Mediated Mammary Gland Iodine Compensation during Lactation. Nutrients 2022; 14:nu14173592. [PMID: 36079849 PMCID: PMC9460413 DOI: 10.3390/nu14173592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
This research aimed to investigate the compensation mechanism of iodine deficiency and excess in the mammary gland during lactation. Female rats were divided into the low iodine group (LI), the normal iodine group (NI), the 10-fold high iodine group (10HI) and the 50-fold high iodine group (50HI). We measured the iodine levels in the urine, blood, milk, and mammary gland. The protein expression of sodium/iodide symporter (NIS), DPAGT1, and valosin-containing protein (VCP) in the mammary gland was also studied. The 24-hour urinary iodine concentration, serum total iodine concentration, serum non-protein-bound iodine concentration, breast milk iodine concentration, and mammary gland iodine content in the 50HI group were significantly higher than those in the NI group (p < 0.05). Compared with the NI group, NIS expression in the 50HI group significantly decreased (p < 0.05). DAPGT1 expression was significantly higher in the LI group than in the NI group (p < 0.05). The expression level of VCP was significantly increased in the 10HI and 50HI groups. In conclusion, milk iodine concentration is positively correlated with iodine intake, and the lactating mammary gland regulates the glycosylation and degradation of NIS by regulating DPAGT1 and VCP, thus regulating milk iodine level. However, the mammary gland has a limited role in compensating for iodine deficiency and excess.
Collapse
|
4
|
Jhiang SM, Sipos JA. Na+/I- symporter expression, function, and regulation in non-thyroidal tissues and impact on thyroid cancer therapy. Endocr Relat Cancer 2021; 28:T167-T177. [PMID: 33974556 PMCID: PMC8419015 DOI: 10.1530/erc-21-0035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/20/2021] [Indexed: 11/08/2022]
Abstract
For the past 80 years, radioiodine (131I) has been used to ablate thyroid tissue not removed by surgery or to treat differentiated thyroid cancer that has metastasized to other parts of the body. However, the Na+/I- symporter (NIS), which mediates active iodide uptake into thyroid follicular cells, is also expressed in several non-thyroidal tissues. This NIS expression permits 131I accumulation and radiation damage in these non-target tissues, which accounts for the adverse effects of radioiodine therapy. We will review the data regarding the expression, function, and regulation of NIS in non-thyroidal tissues and explain the seemingly paradoxical adverse effects induced by 131I, the self-limited gastrointestinal adverse effects in contrast to the permanent salivary dysfunction that is seen after 131I therapy. We propose that prospective studies are needed to uncover the time-course of pathological processes underlying development and progression or ultimate resolution of 131I-induced salivary ductal obstruction and nasolacrimal duct obstruction. Finally, preventive measures and early therapeutic interventions that can be applied potentially to eliminate or alleviate long-term radioiodine adverse effects will be discussed.
Collapse
Affiliation(s)
- Sissy M Jhiang
- Department of Physiology and Cell Biology, The Ohio State University, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Jennifer A. Sipos
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| |
Collapse
|
5
|
Zhang L, Xu S, Cheng X, Wu J, Wang X, Wu L, Yu H, Bao J. Curcumin enhances the membrane trafficking of the sodium iodide symporter and augments radioiodine uptake in dedifferentiated thyroid cancer cells via suppression of the PI3K-AKT signaling pathway. Food Funct 2021; 12:8260-8273. [PMID: 34323243 DOI: 10.1039/d1fo01073e] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Radioactive iodine (RAI) is commonly used to treat differentiated thyroid cancer (DTC). A major challenge is the dedifferentiation of DTC with the loss of radioiodine uptake. Patients with distant metastases have persistent or recurrent disease and develop resistance to RAI therapy due to tumor dedifferentiation. Hence, tumor redifferentiation to restore sensitivity to RAI therapy is considered a promising strategy to overcome RAI resistance. In the present study, curcumin, a natural polyphenolic compound, was found to re-induce cell differentiation and increase the expression of thyroid-specific transcription factors, TTF-1, TTF-2 and transcriptional factor paired box 8 (PAX8), and iodide-metabolizing proteins, including thyroid stimulating hormone receptor (TSHR), thyroid peroxidase (TPO) and sodium iodide symporter (NIS) in dedifferentiated thyroid cancer cell lines, BCPAP and KTC-1. Importantly, curcumin enhanced NIS glycosylation and its membrane trafficking, resulting in a significant improvement of radioiodine uptake in vitro. Additionally, AKT knockdown phenocopied the restoration of thyroid-specific gene expression; however, ectopic expressed AKT inhibited curcumin-induced up-regulation of NIS protein, demonstrating that curcumin might improve radioiodine sensitivity via the inhibition of the PI3K-AKT-mTOR signaling pathway. Our study demonstrates that curcumin could represent a promising adjunctive therapy for restoring iodide avidity and improve radioiodine therapeutic efficacy in patients with RAI-refractory thyroid carcinoma.
Collapse
Affiliation(s)
- Li Zhang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China. and Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China and School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Shichen Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Xian Cheng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Jing Wu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Xiaowen Wang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Liying Wu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Huixin Yu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Jiandong Bao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| |
Collapse
|
6
|
Oh JM, Ahn BC. Molecular mechanisms of radioactive iodine refractoriness in differentiated thyroid cancer: Impaired sodium iodide symporter (NIS) expression owing to altered signaling pathway activity and intracellular localization of NIS. Theranostics 2021; 11:6251-6277. [PMID: 33995657 PMCID: PMC8120202 DOI: 10.7150/thno.57689] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
The advanced, metastatic differentiated thyroid cancers (DTCs) have a poor prognosis mainly owing to radioactive iodine (RAI) refractoriness caused by decreased expression of sodium iodide symporter (NIS), diminished targeting of NIS to the cell membrane, or both, thereby decreasing the efficacy of RAI therapy. Genetic aberrations (such as BRAF, RAS, and RET/PTC rearrangements) have been reported to be prominently responsible for the onset, progression, and dedifferentiation of DTCs, mainly through the activation of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/AKT signaling pathways. Eventually, these alterations result in a lack of NIS and disabling of RAI uptake, leading to the development of resistance to RAI therapy. Over the past decade, promising approaches with various targets have been reported to restore NIS expression and RAI uptake in preclinical studies. In this review, we summarized comprehensive molecular mechanisms underlying the dedifferentiation in RAI-refractory DTCs and reviews strategies for restoring RAI avidity by tackling the mechanisms.
Collapse
|
7
|
Rathod M, Kelkar M, Valvi S, Salve G, De A. FOXA1 Regulation Turns Benzamide HDACi Treatment Effect-Specific in BC, Promoting NIS Gene-Mediated Targeted Radioiodine Therapy. MOLECULAR THERAPY-ONCOLYTICS 2020; 19:93-104. [PMID: 33102692 PMCID: PMC7554325 DOI: 10.1016/j.omto.2020.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/26/2020] [Indexed: 11/03/2022]
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.
Collapse
Affiliation(s)
- Maitreyi Rathod
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai 400094, India
| | - Madhura Kelkar
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai 400094, India
| | - Snehal Valvi
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India
| | - Girish Salve
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India
| | - Abhijit De
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai 400094, India
| |
Collapse
|
8
|
Wang H, Ma Z, Cheng X, Tuo B, Liu X, Li T. Physiological and Pathophysiological Roles of Ion Transporter-Mediated Metabolism in the Thyroid Gland and in Thyroid Cancer. Onco Targets Ther 2020; 13:12427-12441. [PMID: 33299328 PMCID: PMC7721308 DOI: 10.2147/ott.s280797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/17/2020] [Indexed: 12/21/2022] Open
Abstract
Thyroid cancer is the most common type of endocrine tumor and has shown an increasing annual incidence, especially among women. Patients with thyroid cancer have a good prognosis, with a high five-year survival rate; however, the recurrence rate and disease status of thyroid cancer remain a burden for patients, which compels us to further elucidate the pathogenesis of this disease. Recently, ion transporters have gradually become a hot topic in the field of thyroid gland biology and cancer research. Additionally, alterations in the metabolic state of tumor cells and protein molecules have gradually become the focus of scientific research. This review focuses on the progress in understanding the physiological and pathophysiological roles of ion transporter-mediated metabolism in both the thyroid gland and thyroid cancer. We also hope to shed light on new targets for the treatment and prognosis of thyroid cancer.
Collapse
Affiliation(s)
- Hu Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Zhiyuan Ma
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Xiaoming Cheng
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Digestive Disease Institute of Guizhou Province, Zunyi, People’s Republic of China
| | - Xuemei Liu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Digestive Disease Institute of Guizhou Province, Zunyi, People’s Republic of China
| | - Taolang Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| |
Collapse
|
9
|
Oh JM, Baek SH, Gangadaran P, Hong CM, Rajendran RL, Lee HW, Zhu L, Gopal A, Kalimuthu S, Jeong SY, Lee SW, Lee J, Ahn BC. A Novel Tyrosine Kinase Inhibitor Can Augment Radioactive Iodine Uptake Through Endogenous Sodium/Iodide Symporter Expression in Anaplastic Thyroid Cancer. Thyroid 2020; 30:501-518. [PMID: 31928162 DOI: 10.1089/thy.2018.0626] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
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.
Collapse
Affiliation(s)
- Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Se Hwan Baek
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Liya Zhu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Arunnehru Gopal
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
10
|
Rathod M, Chatterjee S, Dutta S, Kalraiya R, Bhattacharyya D, De A. Mannose glycosylation is an integral step for NIS localization and function in human breast cancer cells. J Cell Sci 2019; 132:jcs.232058. [PMID: 31455607 DOI: 10.1242/jcs.232058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022] Open
Abstract
Chasing an intriguing biological question on the disparity of sodium iodide symporter (NIS, officially known as SLC5A5) expression and function in the clinical scenario of breast cancer, this study addresses key molecular defects involved. NIS in cancer patients has primarily been recorded to be a cytoplasmic protein, thus limiting the scope for targeted radio-iodine therapy. We developed NIS transgene-overexpressing MCF-7 breast cancer cells, and found a few clonal derivatives that show predominant expression of NIS in the plasma membrane. The majority of clones, however, showed cytosolic NIS expression over long passages. Cells expressing membranous NIS show unperturbed dynamic trafficking of NIS through secretory pathway organelles when compared to cells expressing cytoplasmic NIS or to parental cells. Further, treatment of cells expressing membranous NIS with specific glycosylation inhibitors highlighted the importance of inherent glycosylation processing and an 84 gene signature glycosylation RT-Profiler array revealed that clones expressing NIS in their membrane cluster separately compared to the other cells. We further confirm a role of three differentially expressed genes, i.e. MAN1B1, MAN1A1 and MAN2A1, in regulating NIS localization by RNA interference. Thus, this study shows the important role of mannosidase in N-glycosylation processing in order to correctly traffic NIS to the plasma membrane in breast cancer cells.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Maitreyi Rathod
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Sushmita Chatterjee
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India
| | - Shruti Dutta
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India
| | - Rajiv Kalraiya
- Glycobiology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Dibyendu Bhattacharyya
- Cell Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Abhijit De
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India .,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| |
Collapse
|
11
|
Ząbczyńska M, Kozłowska K, Pocheć E. Glycosylation in the Thyroid Gland: Vital Aspects of Glycoprotein Function in Thyrocyte Physiology and Thyroid Disorders. Int J Mol Sci 2018; 19:E2792. [PMID: 30227620 PMCID: PMC6163523 DOI: 10.3390/ijms19092792] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/07/2018] [Accepted: 09/14/2018] [Indexed: 02/08/2023] Open
Abstract
The key proteins responsible for hormone synthesis in the thyroid are glycosylated. Oligosaccharides strongly affect the function of glycosylated proteins. Both thyroid-stimulating hormone (TSH) secreted by the pituitary gland and TSH receptors on the surface of thyrocytes contain N-glycans, which are crucial to their proper activity. Thyroglobulin (Tg), the protein backbone for synthesis of thyroid hormones, is a heavily N-glycosylated protein, containing 20 putative N-glycosylated sites. N-oligosaccharides play a role in Tg transport into the follicular lumen, where thyroid hormones are produced, and into thyrocytes, where hyposialylated Tg is degraded. N-glycans of the cell membrane transporters sodium/iodide symporter and pendrin are necessary for iodide transport. Some changes in glycosylation result in abnormal activity of the thyroid and alteration of the metabolic clearance rate of hormones. Alteration of glycan structures is a pathological process related to the progression of chronic diseases such as thyroid cancers and autoimmunity. Thyroid carcinogenesis is accompanied by changes in sialylation and fucosylation, β1,6-branching of glycans, the content and structure of poly-LacNAc chains, as well as O-GlcNAcylation, while in thyroid autoimmunity the main processes affected are sialylation and fucosylation. The glycobiology of the thyroid gland is an intensively studied field of research, providing new data helpful in understanding the role of the sugar component in thyroid protein biology and disorders.
Collapse
Affiliation(s)
- Marta Ząbczyńska
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| | - Kamila Kozłowska
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| | - Ewa Pocheć
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| |
Collapse
|
12
|
Feng F, Yehia L, Ni Y, Chang YS, Jhiang SM, Eng C. A Nonpump Function of Sodium Iodide Symporter in Thyroid Cancer via Cross-talk with PTEN Signaling. Cancer Res 2018; 78:6121-6133. [PMID: 30217930 DOI: 10.1158/0008-5472.can-18-1954] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/15/2018] [Accepted: 09/11/2018] [Indexed: 01/21/2023]
Abstract
The sodium iodide symporter (NIS) is a classical iodide pump typically localized within the cell plasma membrane in thyroid cells, where NIS expression is believed to ensure success of mainstay radioiodide therapy in thyroid cancers. Although radioiodide uptake is generally reduced in thyroid cancer tissue, intracellular nonmembranous NIS has been reported to increase, suggesting that NIS serves a pump-independent function. Thyroid cancer is one of the major component cancers of Cowden syndrome, a subset of which is caused by germline mutations in PTEN In this study, we explored the noncanonical tumorigenic role of NIS in thyroid cancer cells in relation to PTEN signaling. PTEN knockdown in thyroid cancer cell lines stabilized intracellular NIS protein by promoting an interaction with NIS-LARG (leukemia-associated RhoA guanine exchange factor). Increased protein levels of cytoplasmic NIS enhanced RhoA activation and resulted in a promigration tumorigenic phenotype. Inhibition of NIS glycosylation through activation of the PI3K/AKT/mTOR signaling pathway contributed to mislocalization of NIS in the cytoplasm, facilitating its nonpump tumorigenic function through an interaction with LARG, which predominantly localized in the cytoplasm. Moreover, PTEN or PI3K/AKT/mTOR signaling could affect DPAGT1, a glycosylating enzyme involved in the initial step of N-linked glycosylation, to inhibit glycosylation of NIS. In summary, our results elucidate a pump-independent, protumorigenic role for NIS in thyroid cancer via its cross-talk with PTEN signaling.Significance: A novel pump-independent protumorigenic role of nonmembranous NIS challenges the presumption that radioiodine treatment of thyroid cancer is ineffective when transmembrane NIS is not expressed. Cancer Res; 78(21); 6121-33. ©2018 AACR.
Collapse
Affiliation(s)
- Fang Feng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lamis Yehia
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Ying Ni
- Center for Clinical Genomics, Cleveland Clinic, Cleveland, Ohio
| | - Yi Seok Chang
- Department of Physiology and Cell Biology, and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Sissy Meihua Jhiang
- Department of Physiology and Cell Biology, and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio. .,Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Germline High Risk Cancer Focus Group, CASE Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
13
|
The rs2910164 Genetic Variant of miR-146a-3p Is Associated with Increased Overall Mortality in Patients with Follicular Variant Papillary Thyroid Carcinoma. Int J Mol Sci 2018; 19:ijms19030655. [PMID: 29495389 PMCID: PMC5877516 DOI: 10.3390/ijms19030655] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 02/18/2018] [Accepted: 02/20/2018] [Indexed: 11/17/2022] Open
Abstract
Aberrant expression of the sodium-iodide symporter (NIS) and the resistance to post-operative radioactive iodide treatment is a crucial cause of higher mortality of some thyroid cancer patients. In this study, we analyzed the impact of miR-146a on the expression and function of NIS and on the overall survival of thyroid cancer patients. The study included 2441 patients (2163 women; 278 men); including 359 cases with follicular variant of papillary thyroid carcinoma (fvPTC). miR:NIS interactions were analyzed in cell lines using in vivo binding and inhibition assays and radioactive iodine uptake assays. Tumor/blood DNA was used for rs2910164 genotyping. Overall survival was assessed retrospectively. In the results, we showed that miR-146a-3p directly binds to and inhibits NIS. Inhibition of miR-146a-3p restores the expression and function of NIS, increasing radioactive iodine uptake. Rs2910164 functional variant within miR-146a-3p is associated with increased overall mortality among fvPTC female patients. The deaths per 1000 person-years were 29.7 in CC carriers vs. 5.08 in GG/GC-carriers (HR = 6.21, p = 0.006). Higher mortality of CC vs. GG/GC carriers was also observed in patients with lower clinical stage (HR = 22.72, p < 0.001), smaller tumor size (pT1/pT2) (HR = 25.05, p < 0.001), lack of extrathyroidal invasion (HR = 9.03, p = 0.02), lack of nodular invasion (HR = 7.84, p = 0.002), lack of metastases (HR = 6.5, p = 0.005) and older (age at diagnosis >50 years) (HR = 7.8, p = 0.002). MiR-146a-3p underwent somatic mutations in 16.1% of analyzed specimens, mainly towards the deleterious C allele. In this report we propose a novel molecular marker of the clinical outcome of fvPTC patients. Rs2910164 increases the overall mortality with inhibition of NIS and disruption of radioiodine uptake as a possible mechanism.
Collapse
|
14
|
Lan L, Basourakos S, Cui D, Zuo X, Deng W, Huo L, Chen H, Zhang G, Deng L, Shi B, Luo Y. ATRA increases iodine uptake and inhibits the proliferation and invasiveness of human anaplastic thyroid carcinoma SW1736 cells: Involvement of β-catenin phosphorylation inhibition. Oncol Lett 2017; 14:7733-7738. [PMID: 29344218 PMCID: PMC5755144 DOI: 10.3892/ol.2017.7225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/10/2017] [Indexed: 12/31/2022] Open
Abstract
All-trans-retinoic acid (ATRA) can enhance iodine uptake capability of thyroid tumors, but the mechanisms remain poorly understood. The aim of the present study was to investigate the effects of ATRA on isotope susceptibility, proliferation and invasion of anaplastic thyroid carcinoma (ATC) and potential mechanisms. SW1736 cells were treated with 1 µmol/l ATRA or 1% ethanol for 5 days. A cell line stably expressing β-catenin-shRNA was established. An iodine uptake assay was performed using 125I. Proliferation and invasiveness were tested using MTT and Transwell assays, respectively. Western blotting was used to assess the expression of β-catenin, glycogen synthase kinase-3β (GSK-3β), sodium/iodine symporter (NIS) and proteins involved in epithelial-mesenchymal transition. Cells pretreated with ATRA were injected subcutaneously into SCID mice. Mice were intraperitoneally injected with 131I once on the first day of treatment, and tumor growth was then assessed. After 35 days of 131I treatment, ATRA-pretreated tumor volume and weight were decreased compared with the 131I alone group (163.32±19.57 vs. 332.06±21.37 mm3; 0.35±0.14 vs. 0.67±0.23 g, both P<0.05). Similar results were observed in the β-catenin shRNA-pretreated tumors. ATRA also increased the uptake of iodine by SW1736 cells (P<0.01), and similar results were observed in β-catenin shRNA cells. ATRA treatment decreased the cell proliferation and invasion compared with control cells (all P<0.05), similar to β-catenin shRNA. ATRA treatment decreased the expression of phosphorylated (p-)β-catenin, p-GSK-3β, vimentin, and fibronectin, and increased the expression of NIS and E-cadherin, compared with the control. ATRA increased the iodine uptake and inhibited the proliferation and invasion of SW1736 cells, involving β-catenin phosphorylation. In conclusion, ATRA could be used to improve the isotope sensitivity of ATC.
Collapse
Affiliation(s)
- Ling Lan
- Department of Endocrinology, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Spyros Basourakos
- Department of Genitourinary, Cancer Medicine, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Dai Cui
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xuemei Zuo
- Department of Genitourinary, Cancer Medicine, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Wei Deng
- Department of Endocrinology, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Lili Huo
- Department of Endocrinology, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Hailing Chen
- Department of Endocrinology, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Guoying Zhang
- Department of Endocrinology, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Lili Deng
- Department of Endocrinology, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Bingyin Shi
- Department of Endocrinology, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yong Luo
- Department of Genitourinary, Cancer Medicine, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA.,Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| |
Collapse
|
15
|
Ahn BC. Personalized Medicine Based on Theranostic Radioiodine Molecular Imaging for Differentiated Thyroid Cancer. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1680464. [PMID: 27239470 PMCID: PMC4864569 DOI: 10.1155/2016/1680464] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/13/2016] [Indexed: 12/31/2022]
Abstract
Molecular imaging based personalized therapy has been a fascinating concept for individualized therapeutic strategy, which is able to attain the highest efficacy and reduce adverse effects in certain patients. Theranostics, which integrates diagnostic testing to detect molecular targets for particular therapeutic modalities, is one of the key technologies that contribute to the success of personalized medicine. Although the term "theranostics" was used after the second millennium, its basic principle was applied more than 70 years ago in the field of thyroidology with radioiodine molecular imaging. Differentiated thyroid cancer, which arises from follicular cells in the thyroid, is the most common endocrine malignancy, and theranostic radioiodine has been successfully applied to diagnose and treat differentiated thyroid cancer, the applications of which were included in the guidelines published by various thyroid or nuclear medicine societies. Through better pathophysiologic understanding of thyroid cancer and advancements in nuclear technologies, theranostic radioiodine contributes more to modern tailored personalized management by providing high therapeutic effect and by avoiding significant adverse effects in differentiated thyroid cancer. This review details the inception of theranostic radioiodine and recent radioiodine applications for differentiated thyroid cancer management as a prototype of personalized medicine based on molecular imaging.
Collapse
Affiliation(s)
- Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50 Samduk-dong 2-ga, Jung-gu, Daegu 700-721, Republic of Korea
| |
Collapse
|
16
|
Chung T, Youn H, Yeom CJ, Kang KW, Chung JK. Glycosylation of Sodium/Iodide Symporter (NIS) Regulates Its Membrane Translocation and Radioiodine Uptake. PLoS One 2015; 10:e0142984. [PMID: 26599396 PMCID: PMC4658105 DOI: 10.1371/journal.pone.0142984] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/29/2015] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Human sodium/iodide symporter (hNIS) protein is a membrane glycoprotein that transports iodide ions into thyroid cells. The function of this membrane protein is closely regulated by post-translational glycosylation. In this study, we measured glycosylation-mediated changes in subcellular location of hNIS and its function of iodine uptake. METHODS HeLa cells were stably transfected with hNIS/tdTomato fusion gene in order to monitor the expression of hNIS. Cellular localization of hNIS was visualized by confocal microscopy of the red fluorescence of tdTomato. The expression of hNIS was evaluated by RT-PCR and immunoblot analysis. Functional activity of hNIS was estimated by radioiodine uptake. Cyclic AMP (cAMP) and tunicamycin were used to stimulate and inhibit glycosylation, respectively. In vivo images were obtained using a Maestro fluorescence imaging system. RESULTS cAMP-mediated Glycosylation of NIS resulted in increased expression of hNIS, stimulating membrane translocation, and enhanced radioiodine uptake. In contrast, inhibition of glycosylation by treatment with tunicamycin dramatically reduced membrane translocation of intracellular hNIS, resulting in reduced radioiodine uptake. In addition, our hNIS/tdTomato fusion reporter successfully visualized cAMP-induced hNIS expression in xenografted tumors from mouse model. CONCLUSIONS These findings clearly reveal that the membrane localization of hNIS and its function of iodine uptake are glycosylation-dependent, as our results highlight enhancement of NIS expression and glycosylation with subsequent membrane localization after cAMP treatment. Therefore, enhancing functional NIS by the increasing level of glycosylation may be suggested as a promising therapeutic strategy for cancer patients who show refractory response to conventional radioiodine treatment.
Collapse
Affiliation(s)
- Taemoon Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
- Cancer Imaging Center, Seoul National University Hospital, Seoul, Korea
- * E-mail: (HY); (JKC)
| | - Chan Joo Yeom
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - June-Key Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
- * E-mail: (HY); (JKC)
| |
Collapse
|
17
|
Lakshmanan A, Wojcicka A, Kotlarek M, Zhang X, Jazdzewski K, Jhiang SM. microRNA-339-5p modulates Na+/I- symporter-mediated radioiodide uptake. Endocr Relat Cancer 2015; 22:11-21. [PMID: 25404690 PMCID: PMC4298451 DOI: 10.1530/erc-14-0439] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Na(+)/I(-) symporter (NIS)-mediated radioiodide uptake (RAIU) serves as the basis for targeted ablation of thyroid cancer remnants. However, many patients with thyroid cancer have reduced NIS expression/function and hence do not benefit from radioiodine therapy. microRNA (miR) has emerged as a promising therapeutic target in many diseases; yet, the role of miRs in NIS-mediated RAIU has not been investigated. In silico analysis was used to identify miRs that may bind to the 3'UTR of human NIS (hNIS). The top candidate miR-339-5p directly bound to the 3'UTR of hNIS. miR-339-5p overexpression decreased NIS-mediated RAIU in HEK293 cells expressing exogenous hNIS, decreased the levels of NIS mRNA, and RAIU in transretinoic acid/hydrocortisone (tRA/H)-treated MCF-7 human breast cancer cells as well as thyrotropin-stimulated PCCl3 rat thyroid cells. Nanostring nCounter rat miR expression assay was conducted to identify miRs deregulated by TGFβ, Akti-1/2, or 17-AAG known to modulate RAIU in PCCl3 cells. Among 38 miRs identified, 18 were conserved in humans. One of the 18 miRs, miR-195, was predicted to bind to the 3'UTR of hNIS and its overexpression decreased RAIU in tRA/H-treated MCF-7 cells. miR-339-5p was modestly increased in most papillary thyroid carcinomas (PTCs), yet miR-195 was significantly decreased in PTCs. Interestingly, the expression profiles of 18 miRs could be used to distinguish most PTCs from nonmalignant thyroid tissues. This is the first report, to our knowledge, demonstrating that hNIS-mediated RAIU can be modulated by miRs, and that the same miRs may also play roles in the development or maintenance of thyroid malignancy. Accordingly, miRs may serve as emerging targets to halt the progression of thyroid cancer and to enhance the efficacy of radioiodine therapy.
Collapse
Affiliation(s)
- Aparna Lakshmanan
- Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA
| | - Anna Wojcicka
- Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA
| | - Marta Kotlarek
- Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA
| | - Xiaoli Zhang
- Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA
| | - Krystian Jazdzewski
- Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA
| | - Sissy M Jhiang
- Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA Department of Physiology and Cell BiologyMolecularCellular and Developmental Biology Graduate Program, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, Ohio 43210, USAGenomic MedicineDepartment of General, Transplant, and Liver Surgery, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, PolLaboratory of Human Cancer GeneticsCentre of New Technologies, CENT, University of Warsaw, 02-089 Warsaw, PolCenter for BiostatisticsThe Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
18
|
Plantinga TS, Heinhuis B, Gerrits D, Netea MG, Joosten LAB, Hermus ARMM, Oyen WJG, Schweppe RE, Haugen BR, Boerman OC, Smit JWA, Netea-Maier RT. mTOR Inhibition promotes TTF1-dependent redifferentiation and restores iodine uptake in thyroid carcinoma cell lines. J Clin Endocrinol Metab 2014; 99:E1368-75. [PMID: 24712572 PMCID: PMC5393487 DOI: 10.1210/jc.2014-1171] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CONCEPT Redifferentiation of thyroid carcinoma cells has the potential to increase the efficacy of radioactive iodine therapy in treatment-refractory, nonmedullary thyroid carcinoma (TC), leading to an improved disease outcome. Mammalian target of rapamycin (mTOR) is a key regulator of cell fate affecting survival and differentiation, with autophagy and inflammation as prominent downstream pathways. METHODS The effects of mTOR inhibition were studied for its redifferentiation potential of the human TC cell lines BC-PAP, FTC133, and TPC1 by assessment of mRNA and protein expression of thyroid-specific genes and by performance of iodine uptake assays. RESULTS In thyroid transcription factor 1 (TTF1)-expressing cell lines, mTOR inhibition promoted redifferentiation of TC cells by the up-regulation of human sodium-iodine symporter mRNA and protein expression. Furthermore, these cells exhibited markedly elevated iodine uptake capacity. Surprisingly, this redifferentiation process was not mediated by autophagy induced during mTOR inhibition or by inflammatory mediators but through transcriptional effects at the level of TTF1 expression. Accordingly, small interfering RNA inhibition of TTF1 completely abrogated the induction of human sodium-iodine symporter by mTOR inhibition. CONCLUSION The present study has identified the TTF1-dependent molecular mechanisms through which the inhibition of mTOR leads to the redifferentiation of TC cells and subsequently to increased radioactive iodine uptake.
Collapse
Affiliation(s)
- Theo S Plantinga
- Departments of Internal Medicine (T.S.P., B.H., M.G.N., L.A.B.J., A.R.M.M.H., J.W.A.S., R.T.N.-M.) and Nuclear Medicine (D.G., W.J.G.O., O.C.B.) and Division of Endocrinology (T.S.P., A.R.M.M.H., J.W.A.S., R.T.N.-M.), Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; and Division of Endocrinology, Diabetes, and Metabolism (R.E.S., B.R.H.), University of Colorado Denver, Aurora, Colorado 80045
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Nicola JP, Reyna-Neyra A, Carrasco N, Masini-Repiso AM. Dietary iodide controls its own absorption through post-transcriptional regulation of the intestinal Na+/I- symporter. J Physiol 2012; 590:6013-26. [PMID: 23006481 DOI: 10.1113/jphysiol.2012.241307] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Dietary I(-) absorption in the gastrointestinal tract is the first step in I(-) metabolism. Given that I(-) is an essential constituent of the thyroid hormones, its concentrating mechanism is of significant physiological importance. We recently described the expression of the Na(+)/I(-) symporter (NIS) on the apical surface of the intestinal epithelium as a central component of the I(-) absorption system and reported reduced intestinal NIS expression in response to an I(-)-rich diet in vivo. Here, we evaluated the mechanism involved in the regulation of NIS expression by I(-) itself in enterocytes. Excess I(-) reduced NIS-mediated I(-) uptake in IEC-6 cells in a dose- and time-dependent fashion, which was correlated with a reduction of NIS expression at the plasma membrane. Perchlorate, a competitive inhibitor of NIS, prevented these effects, indicating that an increase in intracellular I(-) regulates NIS. Iodide induced rapid intracellular recruitment of plasma membrane NIS molecules and NIS protein degradation. Lower NIS mRNA levels were detected in response to I(-) treatment, although no transcriptional effect was observed. Interestingly, I(-) decreased NIS mRNA stability, affecting NIS translation. Heterologous green fluorescent protein-based reporter constructs revealed a significant repressive effect of the I(-)-targeting NIS mRNA 3 untranslated region. In conclusion, excess I(-) downregulates NIS expression in enterocytes by virtue of a complex mechanism. Our data suggest that I(-) regulates intestinal NIS mRNA expression at the post-transcriptional level as part of an autoregulatory effect of I(-) on its own metabolism.
Collapse
Affiliation(s)
- Juan Pablo Nicola
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, X5000HUA, Córdoba, Argentina
| | | | | | | |
Collapse
|
20
|
Stimulation of cultured h9 human embryonic stem cells with thyroid stimulating hormone does not lead to formation of thyroid-like cells. Stem Cells Int 2012; 2012:634914. [PMID: 22619683 PMCID: PMC3349263 DOI: 10.1155/2012/634914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 12/31/2011] [Indexed: 11/17/2022] Open
Abstract
The sodium-iodine symporter (NIS) is expressed on the cell membrane of many thyroid cancer cells, and is responsible for the radioactive iodine accumulation. However, treatment of anaplastic thyroid cancer is ineffective due to the low expression of NIS on cell membranes of these tumor cells. Human embryonic stem cells (ESCs) provide a potential vehicle to study the mechanisms of NIS expression regulation during differentiation. Human ESCs were maintained on feeder-independent culture conditions. RT-qPCR and immunocytochemistry were used to study differentiation marker expression, 125I uptake to study NIS function. We designed a two-step protocol for human ESC differentiation into thyroid-like cells, as was previously done for mouse embryonic stem cells. First, we obtained definitive endoderm from human ESCs. Second, we directed differentiation of definitive endoderm cells into thyroid-like cells using various factors, with thyroid stimulating hormone (TSH) as the main differentiating factor. Expression of pluripotency, endoderm and thyroid markers and 125I uptake were monitored throughout the differentiation steps. These approaches did not result in efficient induction of thyroid-like cells. We conclude that differentiation of human ESCs into thyroid cells cannot be induced by TSH media supplementation alone and most likely involves complicated developmental patterns that are yet to be understood.
Collapse
|