1
|
TGF-β, to target or not to target; to prevent thyroid cancer progression? Biochim Biophys Acta Rev Cancer 2022; 1877:188752. [PMID: 35728736 DOI: 10.1016/j.bbcan.2022.188752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 11/22/2022]
Abstract
Thyroid cancer (TC) is a common endocrine cancer with a rising incidence. Current treatment fails to eliminate aggressive thyroid tumours, prompting an investigation into the processes that cause disease progression. In this review, we provide insight into TGF-β driven epithelial to mesenchymal transition (EMT), summarizing the current literature surrounding thyroid carcinogenesis, and discuss the potential for therapeutic strategies targeting the TGF-β signalling pathway. Understanding the underlying mechanisms that regulate cancer stem cell (CSC) growth and TGF-β signalling may provide novel therapeutic approaches for highly resistant TCs.
Collapse
|
2
|
Qin J, Zhao N, Wang S, Liu S, Liu Y, Cui X, Wang S, Xiang Y, Fan C, Li Y, Shan Z, Teng W. Roles of Endogenous IL-10 and IL-10-Competent and CD5+ B Cells in Autoimmune Thyroiditis in NOD.H-2h4 Mice. Endocrinology 2020; 161:5802318. [PMID: 32152630 DOI: 10.1210/endocr/bqaa033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/05/2020] [Indexed: 12/14/2022]
Abstract
Interleukin (IL)-10 is a highly important anti-inflammatory cytokine in the immune system. CD1dhi and CD5+ B cells are both traditionally defined IL-10-secreting B cells. In recent years, a B cell group with combined markers of CD1dhi and CD5+ has been widely studied as it has been reported to suppress autoimmunity in mouse models of autoimmune diseases through IL-10 mechanisms. From the perspective of origination, CD1dhi and CD5+ B cells are developed from different B cell lineages. Whether the regulatory capacity of these 2 B cell groups is consistent with their ability to secrete IL-10 has not been determined. In this study, we generated IL-10 knockout NOD.H-2h4 mice to investigate the function of endogenous IL-10 in autoimmune thyroiditis and conducted adoptive transfer experiments to explore the respective roles of CD5+ and CD1dhi B cells. In our results, the IL-10-/- NOD.H-2h4 mice developed thyroiditis, similar to wild-type NOD.H-2h4 mice. The CD5+ B cells were more capable of secreting IL-10 than CD1dhi B cells in flow cytometric analysis, but the CD1dhi B cells showed more suppressive effects on thyroiditis development and autoantibody production, as well as Th17 cell response. In conclusion, endogenous IL-10 does not play an important role in autoimmune thyroiditis. CD1dhi B cells may play regulatory roles through mechanisms other than secreting IL-10.
Collapse
Affiliation(s)
- Jing Qin
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Zhao
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuo Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shanshan Liu
- Department of Emergency, The NO.202 Hospital of People's Liberation Army, Shenyang, Liaoning, China
| | - Yongping Liu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuejiao Cui
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shiwei Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Xiang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chenling Fan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yushu Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
3
|
Differences of the Structure of Immune Regulatory Cell Populations between Cellular Material from Sonographically Detected Focal Thyroid Lesions and Peripheral Blood in Humans. Int J Mol Sci 2019; 20:ijms20040918. [PMID: 30791564 PMCID: PMC6412456 DOI: 10.3390/ijms20040918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/11/2019] [Accepted: 02/16/2019] [Indexed: 01/30/2023] Open
Abstract
Focal thyroid lesions are common ultrasound findings with the estimated prevalence up to 67% of the population. They form characteristically enveloped regions with individual encapsulated microenvironment that may involve the specific distribution of immune system compounds—especially antigen presenting cells (APC). We analyzed and compared the most potent APC—plasmacytoid and conventional dendritic cells (DCs) subpopulations and three monocyte subpopulations as well as other immune cells—in peripheral blood and local blood of thyroid gland obtained parallelly in patients with focal thyroid lesions using flow cytometry. The analysis revealed significant differences in the distribution of main subsets of assessed cells between peripheral blood and biopsy material. The results support the existence of local, organ-specific immune reaction control networks within thyroid nodules.
Collapse
|
4
|
Ramírez-Moya J, Santisteban P. miRNA-Directed Regulation of the Main Signaling Pathways in Thyroid Cancer. Front Endocrinol (Lausanne) 2019; 10:430. [PMID: 31312183 PMCID: PMC6614345 DOI: 10.3389/fendo.2019.00430] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022] Open
Abstract
In the last two decades, great strides have been made in the study of microRNAs in development and in diseases such as cancer, as reflected in the exponential increase in the number of reviews on this topic including those on undifferentiated and well-differentiated thyroid cancer. Nevertheless, few reviews have focused on understanding the functional significance of the most up- or down-regulated miRNAs in thyroid cancer for the main signaling pathways hyperactivated in this tumor type. The aim of this review is to discuss the major miRNAs targeting proteins of the MAPK, PI3K, and TGFβ pathways, to define their mechanisms of action through the 3'UTR regions of their target genes, and to describe how they affect thyroid tumorigenesis through their actions on cell proliferation, migration, and invasion. Given the importance of miRNAs in cancer as diagnostic, prognostic and therapeutic candidates, a better understanding of this cross-talk might shed new light on the biomedical treatment of thyroid cancer.
Collapse
Affiliation(s)
- Julia Ramírez-Moya
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- *Correspondence: Pilar Santisteban
| |
Collapse
|
5
|
Oglio R, Thomasz L, Salvarredi L, Juvenal G, Pisarev M. Comparative effects of transforming growth factor beta isoforms on redox metabolism in thyroid cells. Mol Cell Endocrinol 2018; 470:168-178. [PMID: 29061379 DOI: 10.1016/j.mce.2017.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/19/2017] [Accepted: 10/19/2017] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Transforming growth factor beta (TGF-β) regulates thyroid function and growth. However, tumoral thyroid cells became resistant to this factor as they undifferentiated. Little is known about the effects of TGF-β isoforms. We compared the role of redox metabolism in the response to TGF-β isoforms between non tumoral and tumoral thyroid cells. METHODOLOGY AND RESULTS Differentiated rat thyroid cells (FRTL-5) and human thyroid follicular carcinoma cells (WRO) were treated with the three isoforms of TGF-β. TGF-β isoforms stopped cell cycle at different steps; G1 for FRTL-5 and G2/M for WRO. The three isoforms decreased cell viability and increased ROS accumulation in both cell lines. These effects were more pronounced in FRTL-5 than in WRO, and the isoform β1 was more potent in ROS production than the other two. TGF-β isoforms decreased total glutathione, catalase expression and it activity in both cell lines. Only in FRTL-5 the lipid peroxidation was demonstrated. Moreover, TGF-β1 decreased glutathione peroxidase and mitochondrial superoxide dismutase mRNA expression and increased mitochondrial ROS in FRTL-5, but no in WRO. Pretreatment with selenium increased glutathione peroxidase activity and decreased ROS production in WRO treated with TGF-β isoforms. Furthermore, selenium partially reversed the effect of TGF-β isoforms on cell viability only in WRO cells. The knockdown of endogenous NOX4 significantly reduced the TGF-β1 effect on cell viability in WRO but no in FRTL-5. CONCLUSION TGF-β disrupted the redox balance and increased ROS accumulation in both cell lines. FRTL-5 cells showed reduced antioxidant capacity and had a greater sensitivity to TGF-β isoforms, while WRO cells were more resistant. This observation provides new insights into the potential role of TGF-β in the redox regulation of thyroid cells.
Collapse
Affiliation(s)
- Romina Oglio
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires 1429, Argentina.
| | - Lisa Thomasz
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires 1429, Argentina; CONICET, Argentina
| | - Leonardo Salvarredi
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires 1429, Argentina
| | - Guillermo Juvenal
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires 1429, Argentina; CONICET, Argentina
| | - Mario Pisarev
- CONICET, Argentina; Faculty of Medicine, University of Buenos Aires (UBA), Argentina.
| |
Collapse
|
6
|
Saenko VA, Rogounovitch TI. Genetic Polymorphism Predisposing to Differentiated Thyroid Cancer: A Review of Major Findings of the Genome-Wide Association Studies. Endocrinol Metab (Seoul) 2018; 33:164-174. [PMID: 29947173 PMCID: PMC6021315 DOI: 10.3803/enm.2018.33.2.164] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/04/2018] [Accepted: 05/10/2018] [Indexed: 11/30/2022] Open
Abstract
Thyroid cancer has one of the highest hereditary component among human malignancies as seen in medical epidemiology investigations, suggesting the potential meaningfulness of genetic studies. Here we review researches into genetic variations that influence the chance of developing non-familial differentiated thyroid cancer (DTC), focusing on the major findings of the genome-wide association studies (GWASs) of common single-nucleotide polymorphisms (SNPs). To date, eight GWAS have been performed, and the association of a number of SNPs have been reproduced in dozens of replication investigations across different ethnicities, including Korea and Japan. Despite the cumulative effect of the strongest SNPs demonstrates gradual increase in the risk for cancer and their association signals are statistically quite significant, the overall prediction ability for DTC appears to be very limited. Thus, genotyping of common SNPs only would be insufficient for evidence-based counseling in clinical setting at present. Further studies to include less significant and rare SNPs, non-SNP genetic information, gene-gene interactions, ethnicity, non-genetic and environmental factors, and development of more advanced computational algorithms are warranted to approach to personalized disease risk prediction and prognostication.
Collapse
Affiliation(s)
- Vladimir A Saenko
- Department of Radiation Molecular Epidemiology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan.
| | - Tatiana I Rogounovitch
- Department of Global Health, Medicine and Welfare, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
7
|
Ivanova K, Manolova I, Ignatova MM, Gulubova M. Immunohistochemical Expression of TGF-Β1, SMAD4, SMAD7, TGFβRII and CD68-Positive TAM Densities in Papillary Thyroid Cancer. Open Access Maced J Med Sci 2018; 6:435-441. [PMID: 29610597 PMCID: PMC5874362 DOI: 10.3889/oamjms.2018.105] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/21/2018] [Accepted: 01/22/2018] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND: Papillary thyroid carcinoma (PTC) accounts for 80% of the thyroid malignancies that are characterised by slow growth and an excellent prognosis. Over-expression of SMAD4 protein restores TGF-β signalling, determines a strong increase in anti-proliferative effect and reduces invasive potential of tumour cells expressing it. AIM: The study aimed to analyse the immunohistochemical expression of TGF-β1 and its downstream phosphorylated SMAD4, element and of the inhibitory SMAD7 PTC variants and their association with the localisation of TAMs within the tumour microenvironment. METHODS: For this retrospective study we investigated 69 patients immunohistochemistry with antibodies against TGF-β, TGF – β-RII, SMAD4, SMAD7, CD68+ macrophages. RESULTS: Patients with low infiltration with CD68+ cells in tumour stroma has significantly shorter survival (median of 129.267 months) compared to those with high CD68+ cells infiltration (p = 0.034). From the analysis of CD68+ cells in tumour border and tumour stroma correlated with expression of TGF-β1 / SMAD proteins, we observed that the positive expression of TGF-β1 in tumour cytoplasm, significantly correlated with increased number of CD68+ cells in tumour border (X2 = 5,945; p = 0.015). CONCLUSION: TGF-β enhances motility and stimulates recruitment of monocytes, macrophages and other immune cells while directly inhibiting their anti-tumour effector functions.
Collapse
Affiliation(s)
- Koni Ivanova
- Trakia University, Medical Faculty, Department of General and Clinical Pathology, Stara Zagora, Bulgaria
| | - Irena Manolova
- Trakia University, Medical Faculty, Department Molecular Biology, Immunology and Medical Genetics, Stara Zagora, Bulgaria
| | - Maria-Magdalena Ignatova
- Trakia University, Medical Faculty, Department of General and Clinical Pathology, Stara Zagora, Bulgaria
| | - Maya Gulubova
- Trakia University, Medical Faculty, Department of General and Clinical Pathology, Stara Zagora, Bulgaria
| |
Collapse
|
8
|
Braley-Mullen H, Yu S. NOD.H-2h4 mice: an important and underutilized animal model of autoimmune thyroiditis and Sjogren's syndrome. Adv Immunol 2015; 126:1-43. [PMID: 25727287 DOI: 10.1016/bs.ai.2014.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
NOD.H-2h4 mice express the K haplotype on the NOD genetic background. They spontaneously develop thyroiditis and Sjogren's syndrome, but they do not develop diabetes. Although autoimmune thyroid diseases and Sjogren's syndrome are highly prevalent autoimmune diseases in humans, there has been relatively little emphasis on the use of animal models of these diseases for understanding basic mechanisms involved in development and therapy of chronic organ-specific autoimmune diseases. The goal of this review is to highlight some of the advantages of NOD.H-2h4 mice for studying basic mechanisms involved in development of autoimmunity. NOD.H-2h4 mice are one of relatively few animal models that develop organ-specific autoimmune diseases spontaneously, i.e., without a requirement for immunization with antigen and adjuvant, and in both sexes in a relatively short period of time. Thyroiditis and Sjogren's syndrome in NOD.H-2h4 mice are chronic autoimmune diseases that develop relatively early in life and persist for the life of the animal. Because the animals do not become clinically ill, the NOD.H-2h4 mouse provides an excellent model to test therapeutic protocols over a long period of time. The availability of several mutant mice on this background provides a means to address the impact of particular cells and molecules on the autoimmune diseases. Moreover, to our knowledge, this is the only animal model in which the presence or absence of a single cytokine, IFN-γ, is sufficient to completely inhibit one autoimmune thyroid disease, with a completely distinct autoimmune thyroid disease developing when it is absent.
Collapse
Affiliation(s)
- Helen Braley-Mullen
- Departments of Medicine, Molecular Microbiology & Immunology, University of Missouri, Columbia, Missouri, USA.
| | - Shiguang Yu
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, Arkansas, USA
| |
Collapse
|
9
|
Doggui R, El Atia J. Iodine deficiency: Physiological, clinical and epidemiological features, and pre-analytical considerations. ANNALES D'ENDOCRINOLOGIE 2015; 76:59-66. [DOI: 10.1016/j.ando.2014.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/28/2014] [Accepted: 12/18/2014] [Indexed: 11/26/2022]
|
10
|
Jung JH, Jung J, Kim SK, Woo SH, Kang KM, Jeong BK, Jung MH, Kim JH, Hahm JR. Alpha lipoic acid attenuates radiation-induced thyroid injury in rats. PLoS One 2014; 9:e112253. [PMID: 25401725 PMCID: PMC4234464 DOI: 10.1371/journal.pone.0112253] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 10/08/2014] [Indexed: 12/31/2022] Open
Abstract
Exposure of the thyroid to radiation during radiotherapy of the head and neck is often unavoidable. The present study aimed to investigate the protective effect of α-lipoic acid (ALA) on radiation-induced thyroid injury in rats. Rats were randomly assigned to four groups: healthy controls (CTL), irradiated (RT), received ALA before irradiation (ALA + RT), and received ALA only (ALA, 100 mg/kg, i.p.). ALA was treated at 24 h and 30 minutes prior to irradiation. The neck area including the thyroid gland was evenly irradiated with 2 Gy per minute (total dose of 18 Gy) using a photon 6-MV linear accelerator. Greater numbers of abnormal and unusually small follicles in the irradiated thyroid tissues were observed compared to the controls and the ALA group on days 4 and 7 after irradiation. However, all pathologies were decreased by ALA pretreatment. The quantity of small follicles in the irradiated rats was greater on day 7 than day 4 after irradiation. However, in the ALA-treated irradiated rats, the numbers of small and medium follicles were significantly decreased to a similar degree as in the control and ALA-only groups. The PAS-positive density of the colloid in RT group was decreased significantly compared with all other groups and reversed by ALA pretreatment. The high activity index in the irradiated rats was lowered by ALA treatment. TGF-ß1 immunoreactivity was enhanced in irradiated rats and was more severe on the day 7 after radiation exposure than on day 4. Expression of TGF-ß1 was reduced in the thyroid that had undergone ALA pretreatment. Levels of serum pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) did not differ significantly between the all groups. This study provides that pretreatment with ALA decreased the severity of radiation-induced thyroid injury by reducing inflammation and fibrotic infiltration and lowering the activity index. Thus, ALA could be used to ameliorate radiation-induced thyroid injury.
Collapse
Affiliation(s)
- Jung Hwa Jung
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
| | - Jaehoon Jung
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
| | - Soo Kyoung Kim
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
| | - Seung Hoon Woo
- Department of Otolaryngology, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
| | - Ki Mun Kang
- Department of Radiation Oncology, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
| | - Bae-Kwon Jeong
- Department of Radiation Oncology, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
| | - Myeong Hee Jung
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Gyeongnam, Republic of Korea
| | - Jin Hyun Kim
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Gyeongnam, Republic of Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
- * E-mail: (JHK); (JRH)
| | - Jong Ryeal Hahm
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Gyeongnam, Republic of Korea
- * E-mail: (JHK); (JRH)
| |
Collapse
|
11
|
Brace MD, Wang J, Petten M, Bullock MJ, Makki F, Trites J, Taylor SM, Hart RD. Differential expression of transforming growth factor-beta in benign vs. papillary thyroid cancer nodules; a potential diagnostic tool? J Otolaryngol Head Neck Surg 2014; 43:22. [PMID: 25927212 PMCID: PMC4115165 DOI: 10.1186/s40463-014-0022-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 06/27/2014] [Indexed: 01/05/2023] Open
Abstract
Background Thyroid nodules are common, but only 5% of nodules are found to be malignant. In North America, the incidence of thyroid cancer is increasing. Fine needle aspirate (FNA) biopsy is the diagnostic test of choice. Unfortunately, up to 20% of FNAs are non-diagnostic. A specific molecular marker for thyroid cancer is desirable. Evidence suggests that cell signaling through transforming growth factor beta (TGF- β) is important in the development of thyroid cancer. We sought to compare the expression of TGF- β in malignant and benign thyroid nodules. Methods From 2008-present, thyroid nodule tissue from thyroidectomy specimens was prospectively collected and stored at −80°C. RNA extraction and reverse transcription was performed on 47 samples (24 papillary thyroid cancer and 23 benign nodules). Quantitative PCR using SYBR green was performed to detect TGF-β-1 and −2. Resulting CT values were normalized against β-actin. Gene expression was calculated using the 2-ΔCT method. Results A significantly greater expression of TGF- β1 (p < 0.0001) was detected in the group of malignant thyroid nodules compared to benign nodules. There was no difference in the expression of TGF- β2 (p = 0.4735) between the two groups. Conclusions In this study, we demonstrated that expression of TGF- β1 but not TGF- β2 is significantly increased in papillary thyroid cancer compared to benign thyroid nodules. This may serve as a potential diagnostic marker for papillary thyroid cancer.
Collapse
Affiliation(s)
- Matthew D Brace
- Department of Otolaryngology - Head and Neck Surgery; 3rd Floor Dickson Building, Victoria General Site, QEII Health Sciences Centre, 5820 University Ave., Halifax, Nova Scotia, B3H 2Y9, Canada.
| | - Jun Wang
- Department of Microbiology & Immunology, Department of Pathology, Department of Pediatrics, Canadian Center for Vaccinology, IWK Health Centre, Dr. Richard B. Goldbloom Research and Clinical Care Pavilion, 3rd Floor West, 5850/5980 University Avenue, PO Box 9700, Halifax, Nova Scotia, NS B3K 6R8, Canada.
| | - Mark Petten
- Department of Microbiology & Immunology, Department of Pathology, Department of Pediatrics, Canadian Center for Vaccinology, IWK Health Centre, Dr. Richard B. Goldbloom Research and Clinical Care Pavilion, 3rd Floor West, 5850/5980 University Avenue, PO Box 9700, Halifax, Nova Scotia, NS B3K 6R8, Canada.
| | - Martin J Bullock
- Department of Pathology; Dr. D. J. Mackenzie Building, 5788 University Avenue, Halifax, Nova Scotia, B3H 2Y9, Canada.
| | - Fawaz Makki
- Department of Otolaryngology - Head and Neck Surgery; 3rd Floor Dickson Building, Victoria General Site, QEII Health Sciences Centre, 5820 University Ave., Halifax, Nova Scotia, B3H 2Y9, Canada.
| | - Jonathan Trites
- Department of Otolaryngology - Head and Neck Surgery; 3rd Floor Dickson Building, Victoria General Site, QEII Health Sciences Centre, 5820 University Ave., Halifax, Nova Scotia, B3H 2Y9, Canada.
| | - S Mark Taylor
- Department of Otolaryngology - Head and Neck Surgery; 3rd Floor Dickson Building, Victoria General Site, QEII Health Sciences Centre, 5820 University Ave., Halifax, Nova Scotia, B3H 2Y9, Canada.
| | - Robert D Hart
- Department of Otolaryngology - Head and Neck Surgery; 3rd Floor Dickson Building, Victoria General Site, QEII Health Sciences Centre, 5820 University Ave., Halifax, Nova Scotia, B3H 2Y9, Canada.
| |
Collapse
|
12
|
Abstract
Immune responses against thyroid carcinomas have long been demonstrated and associations between inflammatory microenvironment and thyroid carcinomas repeatedly reported. This scenario has prompted scientists throughout the world to unveil how the inflammatory microenvironment is established in thyroid tumors and what is its influence on the outcome of patients with thyroid carcinoma. Many studies have reported the role of evasion from the immune system in tumor progression and reinforced the weakness of the innate immune response toward thyroid cancer spread in advanced stages. Translational studies have provided evidence that an increased density of tumor-associated macrophages in poorly differentiated thyroid carcinoma (DTC) is associated with an aggressive phenotype at diagnosis and decreased cancer-related survival, whereas well-DTC microenvironment enriched with macrophages is correlated with improved disease-free survival. It is possible that these different results are related to different microenvironments. Several studies have provided evidence that patients whose tumors are not infiltrated by lymphocytes present a high recurrence rate, suggesting that the presence of lymphocytes in the tumor microenvironment may favor the prognosis of patients with thyroid carcinoma. However, the effect of lymphocytes and other immune cells on patient outcome seems to result from complex interactions between the tumor and immune system, and the molecular pattern of cytokines and chemokines helps to explain the involvement of the immune system in thyroid tumor progression. The inflammatory microenvironment may help to characterize aggressive tumors and to identify patients who would benefit from a more invasive approach, probably sparing the vast majority of patients with an indolent disease from unnecessary procedures.
Collapse
Affiliation(s)
- Lucas Leite Cunha
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences, University of Campinas (FCM-Unicamp), Rua Tessália Vieira de Camargo 126, Barão Geraldo, Campinas, São Paulo, Brazil
| | | | | |
Collapse
|
13
|
Bianco AC, Anderson G, Forrest D, Galton VA, Gereben B, Kim BW, Kopp PA, Liao XH, Obregon MJ, Peeters RP, Refetoff S, Sharlin DS, Simonides WS, Weiss RE, Williams GR. American Thyroid Association Guide to investigating thyroid hormone economy and action in rodent and cell models. Thyroid 2014; 24:88-168. [PMID: 24001133 PMCID: PMC3887458 DOI: 10.1089/thy.2013.0109] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND An in-depth understanding of the fundamental principles that regulate thyroid hormone homeostasis is critical for the development of new diagnostic and treatment approaches for patients with thyroid disease. SUMMARY Important clinical practices in use today for the treatment of patients with hypothyroidism, hyperthyroidism, or thyroid cancer are the result of laboratory discoveries made by scientists investigating the most basic aspects of thyroid structure and molecular biology. In this document, a panel of experts commissioned by the American Thyroid Association makes a series of recommendations related to the study of thyroid hormone economy and action. These recommendations are intended to promote standardization of study design, which should in turn increase the comparability and reproducibility of experimental findings. CONCLUSIONS It is expected that adherence to these recommendations by investigators in the field will facilitate progress towards a better understanding of the thyroid gland and thyroid hormone dependent processes.
Collapse
Affiliation(s)
- Antonio C. Bianco
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, Florida
| | - Grant Anderson
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota Duluth, Duluth, Minnesota
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Valerie Anne Galton
- Department of Physiology and Neurobiology, Dartmouth Medical School, Lebanon, New Hampshire
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Brian W. Kim
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, Florida
| | - Peter A. Kopp
- Division of Endocrinology, Metabolism, and Molecular Medicine, and Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiao Hui Liao
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - Maria Jesus Obregon
- Institute of Biomedical Investigation (IIB), Spanish National Research Council (CSIC) and Autonomous University of Madrid, Madrid, Spain
| | - Robin P. Peeters
- Division of Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Samuel Refetoff
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - David S. Sharlin
- Department of Biological Sciences, Minnesota State University, Mankato, Minnesota
| | - Warner S. Simonides
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Roy E. Weiss
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois
| | - Graham R. Williams
- Department of Medicine, Imperial College London, Hammersmith Campus, London, United Kingdom
| |
Collapse
|
14
|
Liu Z, Yang Y, Zhang Y, Ye X, Wang L, Xu G. Stomatin-like protein 2 is associated with the clinicopathological features of human papillary thyroid cancer and is regulated by TGF-β in thyroid cancer cells. Oncol Rep 2013; 31:153-60. [PMID: 24190591 DOI: 10.3892/or.2013.2833] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 10/28/2013] [Indexed: 11/06/2022] Open
Abstract
Papillary thyroid cancer (PTC) accounts for 80-90% of all cases of thyroid malignancies. Stomatin-like protein 2 (SLP-2) is a novel member of the stomatin superfamily and is found in several types of human tumors. However, whether it is expressed in human PTC is unknown. In the present study, we aimed to explore the diagnostic value of SLP-2 in patients with PTC and to investigate whether SLP-2 expression is regulated by transforming growth factor-β (TGF-β), a cytokine which plays an important role in PTC tumorigenesis. A total of 107 patients consisting of 99 cases of classical and 8 cases of follicular variant PTC was examined. The expression of SLP-2 mRNA and protein was examined by immunohistochemistry (IHC) and qPCR, respectively. We found that SLP-2 was overexpressed in human PTC. The expression of SLP-2 was significantly associated with clinicopathological features of the PTC cases. Particularly, increased SLP-2 expression was mainly correlated with primary tumors >1 cm in size, with late stage tumors and with metastatic lymph nodes. The expression of SLP-2 was correlated with the expression of Ki-67, a cell proliferation marker, in PTC tissues as detected by IHC. SLP-2 was upregulated by TGF-β1 in PTC cells as evaluated by western blotting. The present data revealed for the first time that patients with PTC exhibited SLP-2 overexpression that was associated with clinicopathological features. The correlation between SLP-2 expression and proliferation marker Ki-67 may be characteristic of PTC and may reflect PTC progression. SLP-2 was upregulated by TGF-β1, indicating a possible role of SLP-2 in PTC tumorigenesis. Our data suggest that SLP-2 may be considered as a useful diagnostic marker and therapeutic target for PTC.
Collapse
Affiliation(s)
- Zebing Liu
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | | | | | | | | | | |
Collapse
|
15
|
Colin IM, Denef JF, Lengelé B, Many MC, Gérard AC. Recent insights into the cell biology of thyroid angiofollicular units. Endocr Rev 2013; 34:209-38. [PMID: 23349248 PMCID: PMC3610675 DOI: 10.1210/er.2012-1015] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 11/07/2012] [Indexed: 01/06/2023]
Abstract
In thyrocytes, cell polarity is of crucial importance for proper thyroid function. Many intrinsic mechanisms of self-regulation control how the key players involved in thyroid hormone (TH) biosynthesis interact in apical microvilli, so that hazardous biochemical processes may occur without detriment to the cell. In some pathological conditions, this enzymatic complex is disrupted, with some components abnormally activated into the cytoplasm, which can lead to further morphological and functional breakdown. When iodine intake is altered, autoregulatory mechanisms outside the thyrocytes are activated. They involve adjacent capillaries that, together with thyrocytes, form the angiofollicular units (AFUs) that can be considered as the functional and morphological units of the thyroid. In response to iodine shortage, a rapid expansion of the microvasculature occurs, which, in addition to nutrients and oxygen, optimizes iodide supply. These changes are triggered by angiogenic signals released from thyrocytes via a reactive oxygen species/hypoxia-inducible factor/vascular endothelial growth factor pathway. When intra- and extrathyrocyte autoregulation fails, other forms of adaptation arise, such as euthyroid goiters. From onset, goiters are morphologically and functionally heterogeneous due to the polyclonal nature of the cells, with nodules distributed around areas of quiescent AFUs containing globules of compact thyroglobulin (Tg) and surrounded by a hypotrophic microvasculature. Upon TSH stimulation, quiescent AFUs are activated with Tg globules undergoing fragmentation into soluble Tg, proteins involved in TH biosynthesis being expressed and the local microvascular network extending. Over time and depending on physiological needs, AFUs may undergo repetitive phases of high, moderate, or low cell and tissue activity, which may ultimately culminate in multinodular goiters.
Collapse
Affiliation(s)
- Ides M Colin
- Pôle de Morphologie, Institut de Recherche Expérimentale et Clinique, Secteur des Sciences de la Santé, Université Catholique de Louvain (UCL), UCL-5251, 52 Avenue E. Mounier, B-1200, Bruxelles, Belgium.
| | | | | | | | | |
Collapse
|
16
|
TGF-beta/Smad pathway and BRAF mutation play different roles in circumscribed and infiltrative papillary thyroid carcinoma. Virchows Arch 2012; 460:587-600. [DOI: 10.1007/s00428-012-1234-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 03/13/2012] [Accepted: 03/26/2012] [Indexed: 10/28/2022]
|
17
|
MicroRNA miR-146b-5p regulates signal transduction of TGF-β by repressing SMAD4 in thyroid cancer. Oncogene 2011; 31:1910-22. [PMID: 21874046 DOI: 10.1038/onc.2011.381] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
MicroRNAs (miRNA) are small non-coding RNAs involved in post-transcriptional gene regulation that have crucial roles in several types of tumors, including papillary thyroid carcinoma (PTC). miR-146b-5p is overexpressed in PTCs and is regarded as a relevant diagnostic marker for this type of cancer. A computational search revealed that miR-146b-5p putatively binds to the 3' untranslated region (UTR) of SMAD4, an important member of the transforming growth factor β (TGF-β) signaling pathway. The TGF-β pathway is a negative regulator of thyroid follicular cell growth, and the mechanism by which thyroid cancer cells evade its inhibitory signal remains unclear. We questioned whether the modulation of the TGF-β pathway by miR-146b-5p can contribute to thyroid tumorigenesis. Luciferase reporter assay confirmed the direct binding of miR-146b-5p on the SMAD4 3'UTR. Specific inhibition of miR-146b-5p with a locked nucleic acid-modified anti-miR-146b oligonucleotide significantly increased SMAD4 levels in the human papillary carcinoma cell lines, TPC-1 and BCPAP. Moreover, suppression of miR-146b-5p increased the cellular response to the TGF-β anti-proliferative signal, significantly decreasing the proliferation rate. The overexpression of miR-146b-5p in normal rat follicular PCCL3 cells decreased SMAD4 levels and disrupted TGF-β signal transduction. MiR-146b-5p overexpression in PCCL3 cells also significantly increased cell proliferation in the absence of thyroid-stimulating hormone and conferred resistance to TGF-β-mediated cell-cycle arrest. Additionally, the activation of thyroid most common oncogenes RET/PTC3 and BRAF in PCCL3 cells upregulated miR-146b-5p expression. Our results confirm the oncogenic role of miR-146b-5p in thyroid follicular cells and contribute to knowledge regarding the modulation of TGF-β signal transduction by miRNAs in PTCs.
Collapse
|
18
|
Thomasz L, Oglio R, Randi AS, Fernandez M, Dagrosa MA, Cabrini RL, Juvenal GJ, Pisarev MA. Biochemical changes during goiter induction by methylmercaptoimidazol and inhibition by delta-iodolactone in rat. Thyroid 2010; 20:1003-13. [PMID: 20825298 DOI: 10.1089/thy.2009.0257] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND We have demonstrated that the administration of delta-iodolactone (i.e., 5-iodo-delta lactone) of arachidonic acid (IL-delta), a mediator in thyroid autoregulation, prevents goiter induction by methylmercaptoimidazol (MMI) in rats. Other studies have shown that transforming growth factor beta-1 (TGF-beta1) mimics some of the actions of excess iodide, but its participation in autoregulation is disputed. The present studies were performed to test the hypotheses that IL-delta decreases thyroid growth by inhibition of cell proliferation and/or by stimulation of apoptosis due to oxidative stress, that TGF-beta is stimulated by an excess of iodide and by IL-delta, and that c-Myc and c-Fos expression are upregulated during goiter induction and downregulated during goiter inhibition. METHODS Rats were treated with MMI alone or together with iodide or IL-delta. Thyroid weight, cell number, cell proliferation, apoptosis, and oxidative stress were determined. Proliferating cell nuclear antigen (PCNA), TGF-beta1, TGF-beta3, c-Myc, and c-Fos were measured by Western blot. RESULTS MMI caused a progressive increase in thyroid weight accompanied by an increase in cell number, asymmetry of the ploidy histograms, and PCNA, c-Fos, and c-Myc expression. In addition, an early increase of apoptosis was observed. Peroxides as well as glutathione peroxidase and catalase activities were also increased in goitrous animals. The inhibitory action of IL-delta on goiter formation was accompanied by the inhibition of cell proliferation evidenced by a significant decrease in cell number, PCNA expression, and asymmetry of the ploidy histograms. A transient stimulation of apoptosis after 7 days of treatment was also observed. MMI administration stimulated TGF-beta1 but not TGF-beta3 synthesis. IL-delta alone caused a slight increase of TGF-beta3 but not TGF-beta1, whereas potassium iodide (KI) stimulated both isoforms and MMI reversed KI effect on TGF-beta1 expression but not on TGF-beta3. CONCLUSIONS The goiter inhibitory action of IL-delta is due to the inhibition of cell proliferation and the transient stimulation of apoptosis. This latter action does not involve oxidative stress. TGF-beta1 does not play a role in the autoregulatory pathway mediated by IL-delta. Iodide stimulates TGF-beta3 without the need of being organified. These results suggest that there may be more than one pathway involved in the autoregulatory mechanism.
Collapse
Affiliation(s)
- Lisa Thomasz
- Nuclear Biochemistry Division, Department of Radiobiology, National Atomic Energy Commission, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Miranda ERD, De Marco L, Soares MMS. Splicing variants impact in thyroid normal physiology and pathological conditions. ACTA ACUST UNITED AC 2010; 53:709-15. [PMID: 19893912 DOI: 10.1590/s0004-27302009000600003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2009] [Accepted: 07/20/2009] [Indexed: 11/22/2022]
Abstract
RNA splicing is an essential, precisely regulated process that occurs after gene transcription and before mRNA translation, in which introns may be removed and exons, retained. Variability in splicing patterns is a major source of protein diversity from the genome and function to generate a tremendously diverse proteome from a relatively small number of genes. Changes in splice site choice can determine different effects on the encoded protein. Small changes in peptide sequence can alter ligand binding, enzymatic activity, allosteric regulation, or protein localization. Errors in splicing regulation have been implicated in a number of different disease states. This study reviewed the mechanisms of splicing and their repercussion in endocrinology, emphasizing its importance in some thyroid physiological and pathological conditions.
Collapse
|
20
|
Matsuo SE, Fiore APZP, Siguematu SM, Ebina KN, Friguglietti CUM, Ferro MC, Kulcsar MAV, Kimura ET. Expression of SMAD proteins, TGF-beta/activin signaling mediators, in human thyroid tissues. ACTA ACUST UNITED AC 2010; 54:406-12. [DOI: 10.1590/s0004-27302010000400010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2009] [Accepted: 02/24/2010] [Indexed: 12/31/2022]
Abstract
OBJECTIVE: To investigate the expression of SMAD proteins in human thyroid tissues since the inactivation of TGF-β/activin signaling components is reported in several types of cancer. Phosphorylated SMAD 2 and SMAD3 (pSMAD2/3) associated with the SMAD4 induce the signal transduction generated by TGF-β and activin, while SMAD7 inhibits this intracellular signaling. Although TGF-β and activin exert antiproliferative roles in thyroid follicular cells, thyroid tumors express high levels of these proteins. MATERIALS AND METHODS: The protein expression of SMADs was evaluated in multinodular goiter, follicular adenoma, papillary and follicular carcinomas by immunohistochemistry. RESULTS: The expression of pSMAD2/3, SMAD4 and SMAD7 was observed in both benign and malignant thyroid tumors. Although pSMAD2/3, SMAD4 and SMAD7 exhibited high cytoplasmic staining in carcinomas, the nuclear staining of pSMAD2/3 was not different between benign and malignant lesions. CONCLUSIONS: The finding of SMADs expression in thyroid cells and the presence of pSMAD2/3 and SMAD4 proteins in the nucleus of tumor cells indicates propagation of TGF-β/activin signaling. However, the high expression of the inhibitory SMAD7, mostly in malignant tumors, could contribute to the attenuation of the SMADs antiproliferative signaling in thyroid carcinomas.
Collapse
|
21
|
Riesco-Eizaguirre G, Rodríguez I, De la Vieja A, Costamagna E, Carrasco N, Nistal M, Santisteban P. The BRAFV600E oncogene induces transforming growth factor beta secretion leading to sodium iodide symporter repression and increased malignancy in thyroid cancer. Cancer Res 2009; 69:8317-25. [PMID: 19861538 DOI: 10.1158/0008-5472.can-09-1248] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The activating mutation BRAF(V600E) is a frequent genetic event in papillary thyroid carcinomas (PTC) that predicts a poor prognosis, leading to loss of sodium/iodide symporter (NIS) expression and subsequent radioiodide-refractory metastatic disease. The molecular basis of such an aggressive behavior induced by BRAF remains unclear. Here, we show a mechanism through which BRAF induces NIS repression and promotes epithelial to mesenchimal transition and invasion based on the operation of an autocrine transforming growth factor (TGF)beta loop. BRAF induces secretion of functional TGFbeta and blocking TGFbeta/Smad signaling at multiple levels rescues BRAF-induced NIS repression. Although this mechanism is MAP/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK independent, secreted TGFbeta cooperates with MEK-ERK signaling in BRAF-induced cell migration, Matrigel invasion, and EMT. Consistent with this process, TGFbeta and other key components of TGFbeta signaling, such as TbetaRII and pSmad2, are overexpressed in human PTC, suggesting a widespread activation of this pathway by locally released TGFbeta. Moreover, this high TGFbeta/Smad activity is associated with PTC invasion, nodal metastasis, and BRAF status. Interestingly, TGFbeta is overexpressed in the invasive front, whereas NIS is preferentially expressed in the central regions of the tumors, suggesting that this negative correlation between TGFbeta and NIS occurs locally inside the tumor. Our study describes a novel mechanism of NIS repression in thyroid cancer and provides evidence that TGFbeta may play a key role in promoting radioiodide resistance and tumor invasion during PTC progression.
Collapse
Affiliation(s)
- Garcilaso Riesco-Eizaguirre
- Instituto de Investigaciones Biomédicas Alberto Sols Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
22
|
Pisarev MA, Thomasz L, Juvenal GJ. Role of transforming growth factor beta in the regulation of thyroid function and growth. Thyroid 2009; 19:881-92. [PMID: 19645615 DOI: 10.1089/thy.2007.0303] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Transforming growth factor beta (TGF-beta) exists in nature as three isoforms. They exert their effects by binding to a type II receptor located at the cell membrane. The TGF-beta-type II receptor complex then recruits type I receptor, and this new complex stimulates the phosphorylation of Smads 2 and 3, which are subsequently transferred to the nucleus, where they regulate gene transcription. The thyroid gland expresses the TGF-beta1 gene mRNA and synthesizes the protein, which under physiologic conditions regulates thyroid growth and function. Different studies have demonstrated that TGF-beta1 inhibits cell proliferation and a number of functional parameters. These include cyclic adenosine monophosphate (AMP) formation, iodine uptake and organification, hormone secretion, and the expression of thyroglobulin, thyroid peroxidase, and Na(+)/I(-) symporter. The expression of the TGF-beta1 gene and protein may be stimulated by iodine under normal conditions. Since TGF-beta1 mimics some of the inhibitory actions of iodine, its participation in thyroid autoregulation has been proposed; however, this concept is still debated. In thyroid tumors, the inhibitory action of TGF-beta1 on cell proliferation is progressively lost as the tumor becomes more undifferentiated. The alterations in the signaling pathway of TGF-beta1 are not the same in tumors from different species. Even within the same species, such as the pig thyroid, the results may be different depending on whether monolayers or follicular suspensions are employed. The data suggest that it is not entirely possible to apply the results obtained in animal studies to normal or pathological human thyroid tissue. More studies are required to provide the information needed to develop treatments, based on targeting the signaling pathway of TGF-beta1, for undifferentiated thyroid cancer and other thyroid diseases.
Collapse
Affiliation(s)
- Mario A Pisarev
- Department of Radiobiology, University of Buenos Aires School of Medicine, Argentina.
| | | | | |
Collapse
|
23
|
Ruggeri RM, Sciacchitano S, Vitale A, Cardelli P, Galletti M, Vitarelli E, Barresi G, Benvenga S, Trimarchi F, Trovato M. Serum hepatocyte growth factor is increased in Hashimoto's thyroiditis whether or not it is associated with nodular goiter as compared with healthy non-goitrous individuals. J Endocrinol Invest 2009; 32:465-9. [PMID: 19468262 DOI: 10.1007/bf03346487] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Some growth factors and cytokines are known to cooperate with TSH in thyroid nodular growth, but few data are available on their circulating levels in Hashimoto's thyroiditis (HT). AIM To evaluate in HT patients whether thyroid nodules are associated with variations in serum levels of hepatocyte growth factor (HGF) and interleukin-6 (IL-6). SUBJECTS AND METHODS Serum levels of HGF and IL-6 were measured by enzyme-linked immunosorbent assay in 176 euthyroid subjects, subdivided into 4 groups: A) HT patients with nodular goiter (no.=42); B) non-goitrous HT patients (no.=36); C) non-HT patients with nodular goiter (no.=48), and D) healthy subjects without thyroid disease (no.=50). RESULTS The highest concentrations of serumHGF were found in patients with nodular goiter, irrespective of the presence of associated HT (groups A and C). Nevertheless, in group A serum HGF levels were significantly higher than in group C (860.8+/-333.6 pg/ml vs 691.5+/-156 pg/ml, p<0.01). Moreover, though serum HGF levels in group B (578.3+/-217 pg/ml) were lower than in group A, they were significantly higher than in healthy controls (group D, 512.7+/-170.4 pg/ml, p<0.001). Serum IL-6 levels were similar in the two HT groups (A and B), and increased with respect to groups C and D. CONCLUSIONS Serum HGF is increased in HT, especially associated to thyroid nodules, as compared with healthy non-goitrous individuals.
Collapse
Affiliation(s)
- R M Ruggeri
- Unit of Endocrinology, Clinical-Experimental Department of Medicine and Pharmacology, University of Messina, Messina, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Yu S, Sharp GC, Braley-Mullen H. TGF-beta promotes thyroid epithelial cell hyperplasia and fibrosis in IFN-gamma-deficient NOD.H-2h4 mice. THE JOURNAL OF IMMUNOLOGY 2008; 181:2238-45. [PMID: 18641364 DOI: 10.4049/jimmunol.181.3.2238] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
IFN-gamma(-/-)NOD.H-2h4 mice given 0.05% NaI in their water develop severe thyroid epithelial cell (thyrocyte) hyperplasia and proliferation (TEC H/P) and fibrosis. Proliferating thyrocytes of IFN-gamma(-/-) mice with TEC H/P produce TGF-beta as demonstrated by immunohistochemical staining and in situ hybridization. Strong expression of activating phosphorylated Smad-2/3 and weak expression of inhibitory Smad-7 by proliferating thyrocytes correlate with the severity of TEC H/P. Splenocytes from IFN-gamma(-/-) mice with severe TEC H/P transfer severe TEC H/P to IFN-gamma(-/-)NOD.H-2h4.SCID mice. Mice given anti-TGF-beta had markedly reduced thyrocyte proliferation and decreased fibrosis compared with mouse Ig-treated controls, suggesting that TGF-beta plays an important role in development of TEC H/P induced by activated splenocytes. Moreover, transgenic IFN-gamma(-/-)NOD.H-2h4 mice expressing TGF-beta on thyrocytes all develop fibrosis and moderate to severe TEC H/P with accelerated kinetics, directly demonstrating a role for TGF-beta in severe TEC H/P and fibrosis.
Collapse
Affiliation(s)
- Shiguang Yu
- Department of Veterans Affairs Research Service, Columbia, MO 65212, USA
| | | | | |
Collapse
|
25
|
Kimura ET, Matsuo SE, Ricarte-Filho JC. TGFbeta, activina e sinalização SMAD em câncer de tiróide. ACTA ACUST UNITED AC 2007; 51:683-9. [PMID: 17891231 DOI: 10.1590/s0004-27302007000500005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Accepted: 05/29/2007] [Indexed: 11/22/2022]
Abstract
TGFbeta e activina são membros da superfamília TGFbeta e desempenham um amplo papel no desenvolvimento, proliferação e apoptose. Estes fatores de crescimento exercem seus efeitos biológicos ligando-se a receptores de membrana do tipo I e do tipo II que transduzem a sinalização até o núcleo através da fosforilação das proteínas R-SMADs (SMAD 2/3) e co-SMADs (SMAD4). O controle apropriado da via de TGFbeta/activina ainda depende da regulação negativa exercida pelo SMAD inibitório (SMAD7) e pelas enzimas E3 de ubiquitinação (Smurfs). Fisiologicamente, TGFbeta e activina atuam como potentes inibidores da proliferação na célula folicular tiroidiana. Desta forma, alterações de receptores e componentes da via de sinalização SMAD estão associadas a diferentes tipos de tumores. Desde que TGFbeta e activina geram sua sinalização intracelular utilizando os mesmos componentes da via SMAD, o desequilíbrio desta via prejudica dois processos anti-mitogênicos da célula. Nesta revisão, enfocamos aspectos que indicam o mecanismo de resistência ao efeito inibitório de TGFbeta e activina ocasionado pelo desequilíbrio da via de sinalização SMAD nas neoplasias da tiróide.
Collapse
Affiliation(s)
- Edna T Kimura
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, SP, Brazil.
| | | | | |
Collapse
|
26
|
Matsuo SE, Martins L, Leoni SG, Hajjar D, Ricarte-Filho JCM, Ebina KN, Kimura ET. Marcadores biológicos de tumores tiroidianos. ACTA ACUST UNITED AC 2004; 48:114-25. [PMID: 15611824 DOI: 10.1590/s0004-27302004000100013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Um marcador biológico ideal deve ser específico e sensível para identificar o tipo tumoral e caracterizar o estágio da progressão neoplásica. Os tumores de tiróide originam-se de dois tipos celulares: 1) carcinoma medular originário de células parafoliculares; e 2) as neoplasias de células epiteliais foliculares, que incluem bócio, adenomas, carcinomas diferenciados (carcinoma papilífero e carcinoma folicular) e carcinoma indiferenciado (carcinoma anaplásico). O comportamento biológico distinto faz com que cada tipo tumoral necessite de uma conduta terapêutica específica. O conhecimento acumulado ao longo destes anos, utilizando métodos de biologia molecular e, mais recentemente, a genômica, identificou mutações específicas de câncer de tiróide e, atualmente, entendemos muito das alterações que ocorrem na expressão de fatores de crescimento, seus receptores e proteínas sinalizadoras intracelular nas neoplasias tiroidianas. Contudo, apesar desses, até o momento não dispomos de um marcador eficiente que auxilie no diagnóstico e prognóstico e, conseqüentemente, para indicação de uma terapêutica mais adequada. Nesta revisão, discutiremos os principais aspectos relacionados à tumorigênese tiroidiana, avaliando o potencial destes fatores como marcador em neoplasia folicular de tiróide.
Collapse
Affiliation(s)
- Sílvia E Matsuo
- Departamento de Histologia & Embriologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP
| | | | | | | | | | | | | |
Collapse
|
27
|
Affiliation(s)
- Nicholas J Sarlis
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas--M. D. Anderson Cancer Center, Houston, Texas 77082, USA
| | | |
Collapse
|
28
|
Bravo SB, Pampín S, Cameselle-Teijeiro J, Carneiro C, Domínguez F, Barreiro F, Alvarez CV. TGF-β-induced apoptosis in human thyrocytes is mediated by p27kip1 reduction and is overridden in neoplastic thyrocytes by NF-κB activation. Oncogene 2003; 22:7819-30. [PMID: 14586408 DOI: 10.1038/sj.onc.1207029] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Millions of people worldwide suffer goiter, a proliferative disease of the follicular cells of the thyroid that may become neoplastic. Thyroid neoplasms have low proliferative index, low apoptotic index and a high incidence of metastasis. TGF-beta is overexpressed in thyroid follicular tumor cells. To investigate the role of TGF-beta in thyroid tumor progression, we established cultures of human thyrocytes from different proliferative pathologies (Grave's disease, multinodular goiter, follicular adenoma, papillary carcinoma), lymph node metastasis, and a normal thyroid sample. All cultures maintained the thyrocyte phenotype. TGF-beta induced cell-cycle arrest in all cultures, in contrast with results reported for other epithelial tumors. In deprived medium, TGF-beta induced apoptosis in normal thyrocyte cultures and all neoplastic cultures except the metastatic cultures. This apoptosis was mediated by a reduction in p27kip1 levels, inducing cell-cycle initiation. Antisense p27 expression induced apoptosis in the absence of TGF-beta. By contrast, in cells in which p27 was overexpressed, TGF-beta had a survival effect. In growth medium, a net survival effect occurs in neoplastic thyrocytes only, not normal thyrocytes, due to activation of the NF-kappaB survival program. Together, these findings suggest that (a) thyroid neoplasms are due to reduced apoptosis, not increased division, in line with the low proliferative index of these pathologies, and (b) TGF-beta induces apoptosis in normal thyrocytes via p27 reduction, but that in neoplastic thyrocytes this effect is overridden by activation of the NF-kappaB program.
Collapse
Affiliation(s)
- Susana B Bravo
- Department of Physiology, School of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | | | | | | | | | | | | |
Collapse
|
29
|
Cerutti JM, Ebina KN, Matsuo SE, Martins L, Maciel RMB, Kimura ET. Expression of Smad4 and Smad7 in human thyroid follicular carcinoma cell lines. J Endocrinol Invest 2003; 26:516-21. [PMID: 12952364 DOI: 10.1007/bf03345213] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Smad proteins have been shown tomediate the signal transduction pathway downstream of the transforming growth factor beta (TGFbeta). TGFbeta induces the phosphorylation of Smad2 and Smad3 which associate with Smad4 and translocate to the nucleus where they regulate gene transcription; besides these stimulatory Smads, the inhibitory Smads, Smad6 and Smad7, oppose signaling by blocking receptors and interrupting the phosphorylation of Smads2/3. The loss of TGFbeta-sensitivity, caused by inactivation of components of TGFbeta signaling, as Smad4, underlies a wide variety of human disorders, including cancer. In addition, the overexpression of the inhibitory Smad7, which prevents the phosphorylation of Smad2/3 and consequently inhibits TGFbeta signaling pathways, was observed in some diseases. In the present study we investigated the expression of Smad4 and Smad7 in thyroid cell lines (NPA papillary carcinoma, WRO follicular carcinoma and ARO anaplastic carcinoma) by RT-PCR and immunocytochemistry. Our results show that Smad4 was expressed in all thyroid cell lines and controls analyzed, differently from other classes of tumors where Smad4 expression was deleted. On the other hand, Smad7 was overexpressed in ARO anaplastic cell line, the most malignant follicular thyroid carcinoma. Our data suggest that the abrogation of the TGFbeta response by Smad7 overexpression may be a mechanism for the tumor aggressiveness observed in undifferentiated thyroid tumors.
Collapse
Affiliation(s)
- J M Cerutti
- Laboratory of Molecular Endocrinology, Division of Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | |
Collapse
|
30
|
Matsuo SE, Ebina KN, Kulcsar MAV, Friguglietti CUM, Kimura ET. Activin betaB expression in rat experimental goiter and human thyroid tumors. Thyroid 2003; 13:239-47. [PMID: 12729472 DOI: 10.1089/105072503321582033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Activins are dimeric proteins of the transforming growth factor beta superfamily, which exhibit multiple functions in gonadal and extragonadal tissues. Expression of activin A, composed of two betaA subunits, has been shown in the thyroid, whereas there has been no study regarding activin B (betaBbetaB) in this gland. In other tissues, such as the gonads, pancreas, and adrenal cortex, expression of both activin betaA and activin betaB has been described. In this study, we detected activin betaB mRNA and protein expression using reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry in rat experimental goiter and in human thyroid, including multinodular goiter, follicular adenoma, papillary carcinoma, and follicular carcinoma. Activin betaA mRNA and protein expression was also investigated in rat and human thyroid tissue. The expression of both activin betaB and activin betaA was highest in rat methimazole-induced goiter and in human follicular adenoma, and papillary and follicular carcinomas when compared with multinodular goiter and normal thyroid tissue. The increased expression of activin betaB as well as activin betaA, observed in this study, suggests that activin B and activin A may be involved in the proliferative and neoplastic processes of the thyroid.
Collapse
Affiliation(s)
- Sílvia E Matsuo
- Department of Histology and Embryology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
31
|
Abstract
The traditional view holds that hyperplasia of endocrine glands is secondary to oversecretion of a trophic hormone. However, in most cases, the mechanism underlying this growth is the spontaneous proliferation of benign neoplasias. Pathologists still depend on subtle morphological criteria to delineate and further classify these tumours. Owing to their variable architecture, a bewildering nomenclature has emerged for these tumours, exemplified by the many names applied to the goitrous thyroid gland: hyperplasia, adenomatous goitre, adenomatoid nodules, benign nodular thyroid disease, adenoma, etc. This article reviews the evidence suggesting that: (1) the varied types of benign neogeneration of endocrine tissue, the spectrum of which ranges from 'simple hyperplasia' to 'true adenoma', involve the same process; (2) even clonality of a growing lesion cannot distinguish hyperplasia from neoplasia; and (3) the basic processes in both cases are not different from those that cause benign tumours in other organs.
Collapse
Affiliation(s)
- Michael Derwahl
- Dept Medicine, St Hedwig Kliniken, and Humboldt University Berlin, Grosse Hamburger Str. 5-10, D-10115, Berlin, Germany.
| | | |
Collapse
|
32
|
Nilsson E, Doraiswamy V, Parrott JA, Skinner MK. Expression and action of transforming growth factor beta (TGFbeta1, TGFbeta2, TGFbeta3) in normal bovine ovarian surface epithelium and implications for human ovarian cancer. Mol Cell Endocrinol 2001; 182:145-55. [PMID: 11514049 DOI: 10.1016/s0303-7207(01)00584-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The majority of ovarian tumors are derived from the single layer of epithelial cells on the surface of the ovary termed the ovarian surface epithelium (OSE). Stromal cell-OSE interactions are postulated to be an important aspect of normal OSE biology and the biology of ovarian cancer. Transforming growth factor beta (TGFbeta) has been shown to often be a mesenchymal cell-derived growth factor that mediates stromal cell-epithelial cell interactions in a variety of different tissues. The current study investigates the expression and action of TGFbeta isoforms (TGFbeta1, TGFbeta2, and TGFbeta3) in OSE and the underlying stroma in both normal bovine and human tumor tissues. Normal bovine ovaries are similar to human ovaries and are used as a model system to investigate normal OSE and stromal cell functions. All three TGFbeta isoforms and their receptor, transforming growth factor beta receptor type II (TGFbetaRII), proteins were found to be detected in the OSE from normal bovine ovaries using immunohistochemistry. Ovarian stromal tissue also contained positive immunostaining for TGFbeta isoforms and TGFbetaRII. RNA was collected from normal bovine OSE and ovarian stromal cells to examine TGFbeta gene expression. TGFbeta1, TGFbeta2, and TGFbeta3 transcripts were detected in both freshly isolated and cultured bovine OSE and stromal cells by a sensitive quantitative polymerase chain reaction assay. TGFbeta1 and TGFbeta2 mRNA levels were found to be present at similar levels in freshly isolated OSE and stroma. Interestingly, TGFbeta3 mRNA levels were significantly higher in freshly isolated OSE than stromal cells. All but TGFbeta3 mRNA in OSE increased when the cells were cultured. Observations indicate that normal bovine OSE and stroma cells express the three TGFbeta isoforms in vivo and in vitro. Human ovarian tumors from stage II, stage III and stage IV cases were found to express TGFbeta1, TGFbeta2, TGFbeta3 and TGFbetaRII protein primarily in the epithelial cell component by immunohistochemistry analysis. The stromal cell component of the human ovarian tumors contained little or no TGFbeta or TGFbetaRII immunostaining. TGFbeta actions on bovine OSE and stromal cells were also investigated. TGFbeta was found to inhibit the growth of OSE, but not stromal cells. To further examine the actions of TGFbeta on OSE, the expression of two growth factors previously shown to be expressed by OSE were analyzed. TGFbeta1 was found to stimulate the expression of both keratinocyte growth factor (KGF) and kit ligand/stem cell factor (KL) by bovine OSE. Therefore, TGFbeta actions on OSE will likely promote a cascade of cell-cell interactions and cellular responses involving multiple growth factors. The effects of regulatory agents on TGFbeta expression by the bovine OSE were examined. Transforming growth factor alpha (TGFalpha) stimulated TGFbeta1 expression, TGFbeta1 stimulated TGFbeta2 expression, and follicle stimulating hormone (FSH) stimulated TGFbeta3 expression. These results demonstrate that TGFbeta isoforms are regulated differently by the regulatory agents tested. In summary, all the TGFbeta isoforms are differentially expressed by the OSE and TGFbeta appears to have an important role in regulating OSE and possibly stromal-OSE interactions. A complex network of endocrine and paracrine interactions appears to influence the expression and actions of TGFbeta on OSE. Abnormal expression and/or action of TGFbeta is postulated to in part be involved in the onset and progression of ovarian cancer.
Collapse
Affiliation(s)
- E Nilsson
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, WA 99163-4231, USA
| | | | | | | |
Collapse
|
33
|
Furlanetto TW, Nunes RB, Sopelsa AM, Maciel RM. Estradiol decreases iodide uptake by rat thyroid follicular FRTL-5 cells. Braz J Med Biol Res 2001; 34:259-63. [PMID: 11175503 DOI: 10.1590/s0100-879x2001000200015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Estradiol has well-known indirect effects on the thyroid. A direct effect of estradiol on thyroid follicular cells, increasing cell growth and reducing the expression of the sodium-iodide symporter gene, has been recently reported. The aim of the present investigation was to study the effect of estradiol on iodide uptake by thyroid follicular cells, using FRTL-5 cells as a model. Estradiol decreased basal iodide uptake by FRTL-5 cells from control levels of 2.490 +/- 0.370 to 2.085 +/- 0.364 pmol I-/microg DNA at 1 ng/ml (P<0.02), to 1.970 +/- 0.302 pmol I-/microg DNA at 10 ng/ml (P<0.003), and to 2.038 +/- 0.389 pmol I-/microg DNA at 100 ng/ml (P<0.02). In addition, 4 ng/ml estradiol decreased iodide uptake induced by 0.02 mIU/ml thyrotropin from 8.678 +/- 0.408 to 7.312 +/- 0.506 pmol I-/microg DNA (P<0.02). A decrease in iodide uptake by thyroid cells caused by estradiol has not been described previously and may have a role in goiter pathogenesis.
Collapse
Affiliation(s)
- T W Furlanetto
- Divisão de Medicina Interna, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil.
| | | | | | | |
Collapse
|
34
|
Moretti F, Nanni S, Pontecorvi A. Molecular pathogenesis of thyroid nodules and cancer. BAILLIERE'S BEST PRACTICE & RESEARCH. CLINICAL ENDOCRINOLOGY & METABOLISM 2000; 14:517-39. [PMID: 11289733 DOI: 10.1053/beem.2000.0101] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumours derived from the thyroid follicular epithelium represent an informative model for understanding the molecular pathogenesis of multistage tumourigenesis, which is the prevailing theory on cancer development and progression nowadays. The early stages of thyroid tumour development appear to be the consequence of the activation or 'de novo' expression of several proto-oncogenes or growth factor receptors, such as ras, ret, NTRK, met, gsp and the thyrotropin (TSH) receptor. Alterations in the expression pattern of these genes are associated with the development of differentiated neoplasms, ranging from benign toxic adenomas (gsp and TSH receptor), to follicular (ras) and papillary (ret/PTC, NTRK, met) carcinomas. They may all be considered to be early events of thyroid cell transformation and, for some, experimental evidence derived from gene transfer studies supports this hypothesis. Alterations in tumour suppressor genes (p53, Rb) are associated instead with the most aggressive and poorly differentiated forms of thyroid cancer, indicating that, in the thyroid tumourigenic process, they represent late genetic events. Specific environmental factors (iodine deficiency, ionizing radiations) have been shown to play a crucial role in promoting the development of thyroid cancer, influencing both its genotypic and phenotypic features. Interestingly, a high percentage of genetic lesions causing thyroid cancer originate from gene rearrangements and chromosomal translocations (ret/PTC, NTRK, Pax-8/PPARgamma) a finding which, being a rare event in most epithelial tumours, makes the molecular pathogenesis of thyroid cancer unique. The uninterrupted flow of information on the molecular genetics of thyroid nodules and cancer will broaden the correlation between genotype and phenotype and will also provide important information for the development of more accurate preoperative diagnostic tools and more efficient treatment choices for the different forms of thyroid cancer.
Collapse
Affiliation(s)
- F Moretti
- Institute of Experimental Medicine, National Research Council
| | | | | |
Collapse
|
35
|
Singh B, Lim D, Cigudosa JC, Ghossein R, Shaha AR, Poluri A, Wreesmann VB, Tuttle M, Shah JP, Rao PH. Screening for genetic aberrations in papillary thyroid cancer by using comparative genomic hybridization. Surgery 2000; 128:888-93;discussion 893-4. [PMID: 11114620 DOI: 10.1067/msy.2000.110847] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Determination of the genetic composition of papillary thyroid cancers may help explain differences in observed clinical behavior. Comparative genomic hybridization (CGH) is a novel molecular cytogenetic assay that allows simultaneous detection of gains, losses, and amplification of genetic information, making it an ideal screening tool. The aim of this study was to identify genetic aberrations occurring in papillary thyroid cancers by using CGH analysis. METHODS CGH analysis was performed on 21 individual cases of papillary thyroid cancers. Nonparametric statistical comparisons were performed with the Fisher exact test. RESULTS Genetic abnormalities were identified by CGH in 10 of 21 cases (48%). A recurrent pattern of aberrations was seen in cases where genetic changes were detected, involving losses at chromosome arms 1p and 9q and chromosomes 17, 19, and 22, and gains at chromosome 4 and chromosome arms 5q, 6q, 9q, and 13q. The loss of chromosome 22 was unique to younger patients (P =.05) and was associated with a higher rate of regional lymphatic metastasis (19% vs 80%, P =.02). CONCLUSIONS Two genetically unique groups of patients were identified by using CGH analysis. One group had no detectable aberrations; the other had a recurrent pattern of aberrations, localizing to the identical chromosomal loci. This pattern of aberrations suggests that the involved loci may contain genes important in thyroid carcinogenesis. The clinical significance of the presence of copy number changes detected by CGH needs to be determined. In addition, molecular cloning of involved genes in each of the aberrations is warranted.
Collapse
Affiliation(s)
- B Singh
- Head and Neck Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|