1
|
De Lima EU, Dos Santos FF, Da Silva IC, De Lima CRA, Frutuoso VS, Caso GF, De Oliveira PR, Bezerra AK, Cerutti JM, Tamura RE, Ramos HE, de Rubio IGS. Reduced expression of FOXE1 in differentiated thyroid cancer, the contribution of CPG methylation, and their clinical relevance. Front Endocrinol (Lausanne) 2024; 15:1454349. [PMID: 39588344 PMCID: PMC11586194 DOI: 10.3389/fendo.2024.1454349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Introduction Forkhead box E1 (FOXE1) is a transcription factor with a crucial role in thyroid morphogenesis and differentiation. Promoter hypermethylation downregulates FOXE1 expression in different tumor types; nevertheless, its expression and relationship with methylation status in differentiated thyroid cancer (DTC) remain unclear. Methods A total of 33 pairs of matched samples of PTC tumors and non-tumors were included. Tumor cell cultures were treated with either 5-Aza-2'-deoxycytidine demethylating agent or dimethyl sulfoxide (DMSO). A real-time polymerase chain reaction (RT-PCR) and Western blotting were performed to assess FOXE1 expression. The methylation status was quantified using bisulfite sequencing. A luciferase gene assay was used to determine CpG-island functionality. Gene expression and promoter methylation of FOXE1 and FOXE1-regulated genes were also analyzed with data from The Cancer Genome Atlas (TCGA) thyroid samples. Results After demethylating treatment, increased FOXE1 mRNA was observed concomitantly with reduced promoter methylation of CpGisland2. A negative correlation between mRNA downregulation and an increased methylation level of CpGisland2 was observed in tumors. Diminished protein expression was also detected in some DTC cell lines and in some tumor samples, suggesting the involvement of post-transcriptional regulatory mechanisms. CPGisland2 was proved to be an enhancer. TCGA data analysis showed low FOXE1 mRNA expression in tumors with a negative correlation with methylation status and a positive correlation with the expression of most of its target genes. Reduced FOXE1 expression, accompanied by a high methylation level, was associated with PTC aggressiveness (tall cell variant, advanced extra thyroid extension, T4 American Joint Committee on Cancer (AJCC) classification), age at diagnosis (over 45 years old), and presence of a BRAFV600E mutation. Conclusion FOXE1 mRNA was downregulated in DTC compared with non-tumors, followed by high CpGisland methylation. A coupling of low mRNA expression and high methylation status was related to characteristics of aggressiveness in DTC tumors.
Collapse
Affiliation(s)
- Erika Urbano De Lima
- Laboratório de Ciências Moleculares da Tireoide (LCMT) e Laboratório de Biologia Molecular do Câncer (LBMC), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Filipe Ferreira Dos Santos
- Centro de Oncologia Molecular (MOC), Hospital Sírio-Libanês - Instituto de Ensino e Pesquisa (HSL-IEP), São Paulo, Brazil
- Department of Biochemistry, Chemistry Institute (IQ), Universidade de São Paulo (USP), São Paulo, Brazil
| | - Igor Campos Da Silva
- Departamento de Cirurgia de Cabeça e Pescoço, Monte Tabor – Hospital São Rafael, Salvador, Brazil
| | | | - Vitoria Sousa Frutuoso
- Laboratório de Ciências Moleculares da Tireoide (LCMT) e Laboratório de Biologia Molecular do Câncer (LBMC), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Gustavo Felisola Caso
- Laboratório de Ciências Moleculares da Tireoide (LCMT) e Laboratório de Biologia Molecular do Câncer (LBMC), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Paloma Ramos De Oliveira
- Laboratório de Ciências Moleculares da Tireoide (LCMT) e Laboratório de Biologia Molecular do Câncer (LBMC), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | | | - Janete Maria Cerutti
- Laboratório de Bases Genéticas dos Tumores da Tiroide, Departamento de Morfologia e Genética Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Rodrigo Esaki Tamura
- Laboratório de Ciências Moleculares da Tireoide (LCMT) e Laboratório de Biologia Molecular do Câncer (LBMC), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Helton Estrela Ramos
- Laboratório de Estudos da Tireoide, Departamento de Bioregulação, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Ileana Gabriela Sanchez de Rubio
- Laboratório de Ciências Moleculares da Tireoide (LCMT) e Laboratório de Biologia Molecular do Câncer (LBMC), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
2
|
Li W, Xia M, Zeng H, Lin H, Teschendorff AE, Gao X, Wang S. Longitudinal analysis of epigenome-wide DNA methylation reveals novel loci associated with BMI change in East Asians. Clin Epigenetics 2024; 16:70. [PMID: 38802969 PMCID: PMC11131215 DOI: 10.1186/s13148-024-01679-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/11/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Obesity is a global public health concern linked to chronic diseases such as cardiovascular disease and type 2 diabetes (T2D). Emerging evidence suggests that epigenetic modifications, particularly DNA methylation, may contribute to obesity. However, the molecular mechanism underlying the longitudinal change of BMI has not been well-explored, especially in East Asian populations. METHODS This study performed a longitudinal epigenome-wide association analysis of DNA methylation to uncover novel loci associated with BMI change in 533 individuals across two Chinese cohorts with repeated DNA methylation and BMI measurements over four years. RESULTS We identified three novel CpG sites (cg14671384, cg25540824, and cg10848724) significantly associated with BMI change. Two of the identified CpG sites were located in regions previously associated with body shape and basal metabolic rate. Annotation of the top 20 BMI change-associated CpGs revealed strong connections to obesity and T2D. Notably, these CpGs exhibited active regulatory roles and located in genes with high expression in the liver and digestive tract, suggesting a potential regulatory pathway from genome to phenotypes of energy metabolism and absorption via DNA methylation. Cross-sectional and longitudinal EWAS comparisons indicated different mechanisms between CpGs related to BMI and BMI change. CONCLUSION This study enhances our understanding of the epigenetic dynamics underlying BMI change and emphasizes the value of longitudinal analyses in deciphering the complex interplay between epigenetics and obesity.
Collapse
Affiliation(s)
- Wenran Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
- Department of Endocrinology and Metabolism, Wusong Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hailuan Zeng
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
- Human Phenome Institute, Fudan University, Shanghai, China.
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- Taizhou Institute of Health Sciences, Fudan University, Taizhou, Jiangsu, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
3
|
Liu M, Zhang G, Wang Z, Liu X, He K, Luo R, Duan Q, Bai R, Wang Y, Du W, Zheng Y, Shao Y. FOXE1 Contributes to the Development of Psoriasis by Regulating WNT5A. J Invest Dermatol 2023; 143:2366-2377.e7. [PMID: 37394057 DOI: 10.1016/j.jid.2023.04.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/04/2023] [Accepted: 04/25/2023] [Indexed: 07/04/2023]
Abstract
Psoriasis is a common, chronic, and relapsing inflammatory skin disease characterized by hyperproliferation of keratinocytes (KCs) and infiltration of immune cells. The pathogenesis of psoriasis is complex, and the exact mechanism remains partially understood. In this study, we showed that the forkhead box family protein, FOXE1, had increased expression in lesional skins compared with nonlesional skin from patients with psoriasis. FOXE1 expression was also increased in an imiquimod-induced psoriatic mouse model as well as in M5-stimulated KCs. Using combinational approaches of knockdown and overexpression of FOXE1, we demonstrated that FOXE1 may promote the proliferation of KCs by facilitating G1/S transition and activating extracellular signal-regulated kinase 1/2 signaling pathway. In addition, knockdown of FOXE1 reduced the production of IL-1β, IL-6, and TNF-α by KCs. RNA-sequencing profiling identified WNT5A as a potential downstream effector of FOXE1. Knockdown of WNT5A inhibited the proliferation of KCs; reduced the production of IL-1β, IL-6, and TNF-α by KCs; and mitigated the growth-promoting effect of FOXE1 in FOXE1-overexpressed KCs. Finally, depletion of FOXE1 by lentiviral delivery of small hairpin RNAs or genetic approach ameliorated dermatitis symptoms in imiquimod-induced psoriasis-like mouse models. Taken together, our results indicated that FOXE1 participates in the pathogenesis of psoriasis and can serve as a target of psoriasis treatment.
Collapse
Affiliation(s)
- Meng Liu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guanfei Zhang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Ziyang Wang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyi Liu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ke He
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruiting Luo
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiqi Duan
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruimin Bai
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuqian Wang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wenqian Du
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zheng
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Yongping Shao
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China; Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
4
|
Mégier C, Dumery G, Luton D. Iodine and Thyroid Maternal and Fetal Metabolism during Pregnancy. Metabolites 2023; 13:metabo13050633. [PMID: 37233673 DOI: 10.3390/metabo13050633] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Thyroid hormones and iodine are required to increase basal metabolic rate and to regulate protein synthesis, long bone growth and neuronal maturation. They are also essential for protein, fat and carbohydrate metabolism regulation. Imbalances in thyroid and iodine metabolism can negatively affect these vital functions. Pregnant women are at risk of hypo or hyperthyroidism, in relation to or regardless of their medical history, with potential dramatic outcomes. Fetal development highly relies on thyroid and iodine metabolism and can be compromised if they malfunction. As the interface between the fetus and the mother, the placenta plays a crucial role in thyroid and iodine metabolism during pregnancy. This narrative review aims to provide an update on current knowledge of thyroid and iodine metabolism in normal and pathological pregnancies. After a brief description of general thyroid and iodine metabolism, their main modifications during normal pregnancies and the placental molecular actors are described. We then discuss the most frequent pathologies to illustrate the upmost importance of iodine and thyroid for both the mother and the fetus.
Collapse
Affiliation(s)
- Charles Mégier
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| | - Grégoire Dumery
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| | - Dominique Luton
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| |
Collapse
|
5
|
Kang HS, Grimm SA, Jothi R, Santisteban P, Jetten AM. GLIS3 regulates transcription of thyroid hormone biosynthetic genes in coordination with other thyroid transcription factors. Cell Biosci 2023; 13:32. [PMID: 36793061 PMCID: PMC9930322 DOI: 10.1186/s13578-023-00979-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Loss of the transcription factor GLI-Similar 3 (GLIS3) function causes congenital hypothyroidism (CH) in both humans and mice due to decreased expression of several thyroid hormone (TH) biosynthetic genes in thyroid follicular cells. Whether and to what extent, GLIS3 regulates thyroid gene transcription in coordination with other thyroid transcriptional factors (TFs), such as PAX8, NKX2.1 and FOXE1, is poorly understood. METHODS PAX8, NKX2.1, and FOXE1 ChIP-Seq analysis with mouse thyroid glands and rat thyrocyte PCCl3 cells was performed and compared to that of GLIS3 to analyze the co-regulation of gene transcription in thyroid follicular cells by these TFs. RESULTS Analysis of the PAX8, NKX2.1, and FOXE1 cistromes identified extensive overlaps between these TF binding loci and those of GLIS3 indicating that GLIS3 shares many of the same regulatory regions with PAX8, NKX2.1, and FOXE1, particularly in genes associated with TH biosynthesis, induced by thyroid stimulating hormone (TSH), and suppressed in Glis3KO thyroid glands, including Slc5a5 (Nis), Slc26a4, Cdh16, and Adm2. ChIP-QPCR analysis showed that loss of GLIS3 did not significantly affect PAX8 or NKX2.1 binding and did not cause major alterations in H3K4me3 and H3K27me3 epigenetic signals. CONCLUSIONS Our study indicates that GLIS3 regulates transcription of TH biosynthetic and TSH-inducible genes in thyroid follicular cells in coordination with PAX8, NKX2.1, and FOXE1 by binding within the same regulatory hub. GLIS3 does not cause major changes in chromatin structure at these common regulatory regions. GLIS3 may induce transcriptional activation by enhancing the interaction of these regulatory regions with other enhancers and/or RNA Polymerase II (Pol II) complexes.
Collapse
Affiliation(s)
- Hong Soon Kang
- grid.280664.e0000 0001 2110 5790Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC 27709 USA
| | - Sara A. Grimm
- grid.280664.e0000 0001 2110 5790Integrative Bioinformatics, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC 27709 USA
| | - Raja Jothi
- grid.280664.e0000 0001 2110 5790Epigenetics & Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC 27709 USA
| | - Pilar Santisteban
- grid.5515.40000000119578126Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Anton M. Jetten
- grid.280664.e0000 0001 2110 5790Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC 27709 USA
| |
Collapse
|
6
|
Lim G, Widiapradja A, Levick SP, McKelvey KJ, Liao XH, Refetoff S, Bullock M, Clifton-Bligh RJ. Foxe1 Deletion in the Adult Mouse Is Associated With Increased Thyroidal Mast Cells and Hypothyroidism. Endocrinology 2022; 163:bqac158. [PMID: 36156081 PMCID: PMC9618408 DOI: 10.1210/endocr/bqac158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Indexed: 11/29/2022]
Abstract
CONTEXT Foxe1 is a key thyroid developmental transcription factor. Germline deletion results in athyreosis and congenital hypothyroidism. Some data suggest an ongoing role for maintaining thyroid differentiation. OBJECTIVE We created a mouse model to directly examine the role of Foxe1 in the adult thyroid. METHODS A model of tamoxifen-inducible Cre-mediated ubiquitous deletion of Foxe1 was generated in mice of C57BL/6J background (Foxe1flox/flox/Cre-TAM). Tamoxifen or vehicle was administered to Foxe1flox/flox/Cre mice aged 6-8 weeks. Blood was collected at 4, 12, and 20 weeks, and tissues after 12 or 20 weeks for molecular and histological analyses. Plasma total thyroxine (T4), triiodothyronine, and thyrotropin (TSH) were measured. Transcriptomics was performed using microarray or RNA-seq and validated by reverse transcription quantitative polymerase chain reaction. RESULTS Foxe1 was decreased by approximately 80% in Foxe1flox/flox/Cre-TAM mice and confirmed by immunohistochemistry. Foxe1 deletion was associated with abnormal follicular architecture and smaller follicle size at 12 and 20 weeks. Plasma TSH was elevated in Foxe1flox/flox/Cre-TAM mice as early as 4 weeks and T4 was lower in pooled samples from 12 and 20 weeks. Foxe1 deletion was also associated with an increase in thyroidal mast cells. Transcriptomic analyses found decreased Tpo and Tg and upregulated mast cell markers Mcpt4 and Ctsg in Foxe1flox/flox/Cre-TAM mice. CONCLUSION Foxe1 deletion in adult mice was associated with disruption in thyroid follicular architecture accompanied by biochemical hypothyroidism, confirming its role in maintenance of thyroid differentiation. An unanticipated finding was an increase in thyroidal mast cells. These data suggest a possible explanation for previous human genetic studies associating alleles in/near FOXE1 with hypothyroidism and/or autoimmune thyroiditis.
Collapse
Affiliation(s)
- Grace Lim
- Cancer Genetics Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia
| | - Alexander Widiapradja
- Cardiac Biology and Heart Failure Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia
| | - Scott P Levick
- Cardiac Biology and Heart Failure Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Samuel Refetoff
- Department of Medicine, Pediatrics and Committee on Genetics, The University of Chicago, Chicago, Illinois 60637, USA
| | - Martyn Bullock
- Cancer Genetics Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia
| | - Roderick J Clifton-Bligh
- Cancer Genetics Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia
- Department of Endocrinology, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| |
Collapse
|
7
|
Jing L, Zhang Q. Intrathyroidal feedforward and feedback network regulating thyroid hormone synthesis and secretion. Front Endocrinol (Lausanne) 2022; 13:992883. [PMID: 36187113 PMCID: PMC9519864 DOI: 10.3389/fendo.2022.992883] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Thyroid hormones (THs), including T4 and T3, are produced and released by the thyroid gland under the stimulation of thyroid-stimulating hormone (TSH). The homeostasis of THs is regulated via the coordination of the hypothalamic-pituitary-thyroid axis, plasma binding proteins, and local metabolism in tissues. TH synthesis and secretion in the thyrocytes-containing thyroid follicles are exquisitely regulated by an elaborate molecular network comprising enzymes, transporters, signal transduction machineries, and transcription factors. In this article, we synthesized the relevant literature, organized and dissected the complex intrathyroidal regulatory network into structures amenable to functional interpretation and systems-level modeling. Multiple intertwined feedforward and feedback motifs were identified and described, centering around the transcriptional and posttranslational regulations involved in TH synthesis and secretion, including those underpinning the Wolff-Chaikoff and Plummer effects and thyroglobulin-mediated feedback regulation. A more thorough characterization of the intrathyroidal network from a systems biology perspective, including its topology, constituent network motifs, and nonlinear quantitative properties, can help us to better understand and predict the thyroidal dynamics in response to physiological signals, therapeutic interventions, and environmental disruptions.
Collapse
Affiliation(s)
- Li Jing
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| |
Collapse
|
8
|
Genetic Variants Associated with Thyroid Cancer Risk: Comprehensive Research Synopsis, Meta-Analysis, and Cumulative Epidemiological Evidence. JOURNAL OF ONCOLOGY 2021; 2021:9967599. [PMID: 34950210 PMCID: PMC8691982 DOI: 10.1155/2021/9967599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/09/2021] [Accepted: 11/20/2021] [Indexed: 11/28/2022]
Abstract
Purpose With the increasing incidence of thyroid cancer (TC), associations between genetic polymorphisms and TC risk have attracted a lot of attention. Considering that the results of associations of genetic variants with TC were usually inconsistent based on publications until now, we attempted to comprehensively evaluate the real evidence of associations between single nucleotide polymorphisms (SNPs) and TC risk. Method We performed meta-analyses on 36 SNPs in 23 genes associated with TC susceptibility based on the data from 99 articles and comprehensively valued the epidemiological evidence of significant associations through the Venice criteria and false-positive report probability (FPRP) test. OR and P value were also calculated for 19 SNPs in 13 genes based on the insufficient data from 22 articles. Results 19 SNPs were found significantly associated with TC susceptibility. Of these, strong epidemiological evidence of associations was identified for the following seven SNPs: POU5F1B rs6983267, FOXE1 rs966423, TERT rs2736100, NKX2-1 rs944289, FOXE1 rs1867277, FOXE1 rs2439302, and RET rs1799939, in which moderate associations were found in four SNPs and weak associations were found in eight SNPs. In addition, probable significant associations with TC were found in nine SNPs. Conclusion Our study systematically evaluated associations between SNPs and TC risk and offered reference information for further understanding of polymorphisms and TC susceptibility.
Collapse
|
9
|
Genetic predisposition of SNPs in miRNA-149 (rs2292832) and FOXE1 (rs3758249) in thyroid Cancer. Mol Biol Rep 2021; 48:7801-7809. [PMID: 34643920 DOI: 10.1007/s11033-021-06795-y] [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: 07/29/2021] [Accepted: 09/10/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Many efforts have been made in recent years to investigate the alterations in protein-coding genes as well as non-coding RNAs that are playing an emerging role in the development and progression of cancers. These miRNAs are short non-coding functional RNAs that are involved in the regulation of transcriptome. In different studies, it was found that human miRNA-149 is an important microRNA that is functioning either as onco-miRNAs or acting as tumor suppressors, in different conditions. RATIONALE Many of the miRNAs are regulating different SNPs of FOXE1 in different studies which are causing low-to-moderate penetrance of genes that initiates the development of thyroid cancer. The involvement of SNPs in miRNA-149 gene rs2292832 and FOXE1 rs3758249 with PTC for better disease prognosis and management was determined in this study and the relation between these SNPs at the genotypic level was also evaluated. MATERIALS AND METHODS: PTC patients with age and gender-matched controls were recruited in the present study. Blood samples were collected in EDTA vacutainer followed by DNA extraction by the organic method. Genotyping of rs2292832 and rs3758249 was done by ARMS-PCR and PCR- RFLP respectively. Statistical analyses were carried out by using SPSS software (version 20). RESULTS The mutation T>C in miRNA-149 rs2292832 was significantly associated with thyroid cancer (p-value 0.0004, < 0.05) while rs3758249 G>C did not show significant association with the disease (p-value 0.124244, > 0.05). Moreover, no correlation of rs2292832 at the genotype level was observed with rs3758249. CONCLUSIONS miRNA-149 gene SNP rs2292832 was observed in strong association with thyroid cancer. Lack of genetic association of rs3758249 of FOXE1 gene has been ruled for the disease. The statistically significant association of rs2292832 with thyroid cancer depicts its mechanistic involvement at the cellular level in Papillary Thyroid Carcinoma.
Collapse
|
10
|
Pilmane M, Jain N, Vitenberga-Verza Z. Expression Analysis of FGF/FGFR and FOX Family Proteins in Mucosal Tissue Obtained from Orofacial Cleft-Affected Children. BIOLOGY 2021; 10:423. [PMID: 34068496 PMCID: PMC8151933 DOI: 10.3390/biology10050423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 01/02/2023]
Abstract
Orofacial clefts affect hundreds of thousands of children worldwide annually and are usually corrected by a series of surgeries extending to childhood. The underlying mechanisms that lead to clefts are still unknown, mainly because of the multifactorial etiology and the myriad of interactions between genes and environmental factors. In the present study, we investigated the role and expression of candidate genes belonging to the FGF/FGFR signaling pathway and FOX family in tissue material obtained from 12 pediatric patients undergoing cleft correction surgery. The expression was investigated using immunohistochemistry (IHC) and chromogenic in-situ hybridization (CISH) in three cell/tissue types-epithelial cells, connective tissue, and endothelial cells. We found elevated expression of FGFR1 in epithelial cells while no expression was observed in endothelial cells. Further, our results elucidate the potential pathogenetic role of FGFR1 in cellular proliferation, local site inflammation, and fibrosis in cleft patients. Along with bFGF (also called FGF2), FGFR1 could play a pro-inflammatory role in clefts. Over-amplification of FGFR2 in some patients, along with bFGF, could potentially suggest roles for these genes in angiogenesis. Additionally, increased expression of FOXE1 (also called TTF2) contributes to local site inflammation. Finally, zero to low amplification of FOXO1 could suggest its potential role in inducing oxidative stress in the endothelium along with reduced epithelial apoptosis.
Collapse
Affiliation(s)
| | - Nityanand Jain
- Department of Morphology, Institute of Anatomy and Anthropology, Riga Stradinš University, LV-1007 Riga, Latvia; (M.P.); (Z.V.-V.)
| | | |
Collapse
|
11
|
Lafontaine N, Campbell PJ, Castillo-Fernandez JE, Mullin S, Lim EM, Kendrew P, Lewer M, Brown SJ, Huang RC, Melton PE, Mori TA, Beilin LJ, Dudbridge F, Spector TD, Wright MJ, Martin NG, McRae AF, Panicker V, Zhu G, Walsh JP, Bell JT, Wilson SG. Epigenome-Wide Association Study of Thyroid Function Traits Identifies Novel Associations of fT3 With KLF9 and DOT1L. J Clin Endocrinol Metab 2021; 106:e2191-e2202. [PMID: 33484127 PMCID: PMC8063248 DOI: 10.1210/clinem/dgaa975] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Indexed: 12/12/2022]
Abstract
CONTEXT Circulating concentrations of free triiodothyronine (fT3), free thyroxine (fT4), and thyrotropin (TSH) are partly heritable traits. Recent studies have advanced knowledge of their genetic architecture. Epigenetic modifications, such as DNA methylation (DNAm), may be important in pituitary-thyroid axis regulation and action, but data are limited. OBJECTIVE To identify novel associations between fT3, fT4, and TSH and differentially methylated positions (DMPs) in the genome in subjects from 2 Australian cohorts. METHOD We performed an epigenome-wide association study (EWAS) of thyroid function parameters and DNAm using participants from: Brisbane Systems Genetics Study (median age 14.2 years, n = 563) and the Raine Study (median age 17.0 years, n = 863). Plasma fT3, fT4, and TSH were measured by immunoassay. DNAm levels in blood were assessed using Illumina HumanMethylation450 BeadChip arrays. Analyses employed generalized linear mixed models to test association between DNAm and thyroid function parameters. Data from the 2 cohorts were meta-analyzed. RESULTS We identified 2 DMPs with epigenome-wide significant (P < 2.4E-7) associations with TSH and 6 with fT3, including cg00049440 in KLF9 (P = 2.88E-10) and cg04173586 in DOT1L (P = 2.09E-16), both genes known to be induced by fT3. All DMPs had a positive association between DNAm and TSH and a negative association between DNAm and fT3. There were no DMPs significantly associated with fT4. We identified 23 differentially methylated regions associated with fT3, fT4, or TSH. CONCLUSIONS This study has demonstrated associations between blood-based DNAm and both fT3 and TSH. This may provide insight into mechanisms underlying thyroid hormone action and/or pituitary-thyroid axis function.
Collapse
Affiliation(s)
- Nicole Lafontaine
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
- Correspondence: Nicole Lafontaine, MBBS, BMedSci, RACP, Department of Endocrinology & Diabetes, Level 1, Building C, QEII Medical Centre, Sir Charles Gairdner Hospital, Hospital Ave, Nedlands, WA 6009, Australia.
| | - Purdey J Campbell
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | | | - Shelby Mullin
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Ee Mun Lim
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Pathwest Laboratory Medicine, Nedlands, WA, Australia
| | | | | | - Suzanne J Brown
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Rae-Chi Huang
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Phillip E Melton
- School of Biomedical Sciences, University of Western Australia, Perth, Australia
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Trevor A Mori
- Medical School, Royal Perth Hospital Unit, University of Western Australia, Perth, WA, Australia
| | - Lawrence J Beilin
- Medical School, Royal Perth Hospital Unit, University of Western Australia, Perth, WA, Australia
| | - Frank Dudbridge
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Tim D Spector
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, UK
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | | | - Allan F McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Vijay Panicker
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Gu Zhu
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - John P Walsh
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
| | - Jordana T Bell
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, UK
| | - Scott G Wilson
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, UK
- School of Biomedical Sciences, University of Western Australia, Perth, Australia
| |
Collapse
|
12
|
López-Márquez A, Carrasco-López C, Fernández-Méndez C, Santisteban P. Unraveling the Complex Interplay Between Transcription Factors and Signaling Molecules in Thyroid Differentiation and Function, From Embryos to Adults. Front Endocrinol (Lausanne) 2021; 12:654569. [PMID: 33959098 PMCID: PMC8095082 DOI: 10.3389/fendo.2021.654569] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/29/2021] [Indexed: 12/29/2022] Open
Abstract
Thyroid differentiation of progenitor cells occurs during embryonic development and in the adult thyroid gland, and the molecular bases of these complex and finely regulated processes are becoming ever more clear. In this Review, we describe the most recent advances in the study of transcription factors, signaling molecules and regulatory pathways controlling thyroid differentiation and development in the mammalian embryo. We also discuss the maintenance of the adult differentiated phenotype to ensure the biosynthesis of thyroid hormones. We will focus on endoderm-derived thyroid epithelial cells, which are responsible for the formation of the thyroid follicle, the functional unit of the thyroid gland. The use of animal models and pluripotent stem cells has greatly aided in providing clues to the complicated puzzle of thyroid development and function in adults. The so-called thyroid transcription factors - Nkx2-1, Foxe1, Pax8 and Hhex - were the first pieces of the puzzle identified in mice. Other transcription factors, either acting upstream of or directly with the thyroid transcription factors, were subsequently identified to, almost, complete the puzzle. Among them, the transcription factors Glis3, Sox9 and the cofactor of the Hippo pathway Taz, have emerged as important players in thyroid differentiation and development. The involvement of signaling molecules increases the complexity of the puzzle. In this context, the importance of Bmps, Fgfs and Shh signaling at the onset of development, and of TSH, IGF1 and TGFβ both at the end of terminal differentiation in embryos and in the adult thyroid, are well recognized. All of these aspects are covered herein. Thus, readers will be able to visualize the puzzle of thyroid differentiation with most - if not all - of the pieces in place.
Collapse
Affiliation(s)
- Arístides López-Márquez
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Laboratorio de Investigación Aplicada en Enfermedades Neuromusculares, Unidad de Patología Neuromuscular, Servicio de Neuropediatría, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Carlos Carrasco-López
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Celia Fernández-Méndez
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Pilar Santisteban,
| |
Collapse
|
13
|
Saqcena M, Leandro-Garcia LJ, Maag JLV, Tchekmedyian V, Krishnamoorthy GP, Tamarapu PP, Tiedje V, Reuter V, Knauf JA, de Stanchina E, Xu B, Liao XH, Refetoff S, Ghossein R, Chi P, Ho AL, Koche RP, Fagin JA. SWI/SNF Complex Mutations Promote Thyroid Tumor Progression and Insensitivity to Redifferentiation Therapies. Cancer Discov 2020; 11:1158-1175. [PMID: 33318036 DOI: 10.1158/2159-8290.cd-20-0735] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/16/2020] [Accepted: 12/09/2020] [Indexed: 12/21/2022]
Abstract
Mutations of subunits of the SWI/SNF chromatin remodeling complexes occur commonly in cancers of different lineages, including advanced thyroid cancers. Here we show that thyroid-specific loss of Arid1a, Arid2, or Smarcb1 in mouse BRAFV600E-mutant tumors promotes disease progression and decreased survival, associated with lesion-specific effects on chromatin accessibility and differentiation. As compared with normal thyrocytes, BRAFV600E-mutant mouse papillary thyroid cancers have decreased lineage transcription factor expression and accessibility to their target DNA binding sites, leading to impairment of thyroid-differentiated gene expression and radioiodine incorporation, which is rescued by MAPK inhibition. Loss of individual SWI/SNF subunits in BRAF tumors leads to a repressive chromatin state that cannot be reversed by MAPK pathway blockade, rendering them insensitive to its redifferentiation effects. Our results show that SWI/SNF complexes are central to the maintenance of differentiated function in thyroid cancers, and their loss confers radioiodine refractoriness and resistance to MAPK inhibitor-based redifferentiation therapies. SIGNIFICANCE: Reprogramming cancer differentiation confers therapeutic benefit in various disease contexts. Oncogenic BRAF silences genes required for radioiodine responsiveness in thyroid cancer. Mutations in SWI/SNF genes result in loss of chromatin accessibility at thyroid lineage specification genes in BRAF-mutant thyroid tumors, rendering them insensitive to the redifferentiation effects of MAPK blockade.This article is highlighted in the In This Issue feature, p. 995.
Collapse
Affiliation(s)
- Mahesh Saqcena
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Jesper L V Maag
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vatche Tchekmedyian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gnana P Krishnamoorthy
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Prasanna P Tamarapu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vera Tiedje
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vincent Reuter
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeffrey A Knauf
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bin Xu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Samuel Refetoff
- Departments of Medicine and Pediatrics and the Committee on Genetics, The University of Chicago, Chicago, Illinois
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ping Chi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James A Fagin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
14
|
Jang D, Marcus-Samuels B, Morgan SJ, Klubo-Gwiezdzinska J, Neumann S, Gershengorn MC. Thyrotropin regulation of differentiated gene transcription in adult human thyrocytes in primary culture. Mol Cell Endocrinol 2020; 518:111032. [PMID: 32941925 PMCID: PMC7606794 DOI: 10.1016/j.mce.2020.111032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/09/2020] [Accepted: 09/09/2020] [Indexed: 01/05/2023]
Abstract
Thyroid transcription factors (TTFs) - NKX2-1, FOXE1, PAX8 and HHEX - regulate multiple genes involved in thyroid development in mice but little is known about TTF regulation of thyroid-specific genes - thyroglobulin (TG), thyroid peroxidase (TPO), deiodinase type 2 (DIO2), sodium/iodide symporter (NIS) and TSH receptor (TSHR) - in adult, human thyrocytes. Thyrotropin (thyroid-stimulating hormone, TSH) regulation of thyroid-specific gene expression in primary cultures of human thyrocytes is biphasic yielding an inverted U-shaped dose-response curve (IUDRC) with upregulation at low doses and decreases at high doses. Herein we show that NKX2-1, FOXE1 and PAX8 are required for TSH-induced upregulation of the mRNA levels of TG, TPO, DIO2, NIS, and TSHR whereas HHEX has little effect on the levels of these thyroid-specific gene mRNAs. We show that TSH-induced upregulation is mediated by changes in their transcription and not by changes in the degradation of their mRNAs. In contrast to the IUDRC of thyroid-specific genes, TSH effects on the levels of the mRNAs for NKX2-1, FOXE1 and PAX8 exhibit monophasic decreases at high doses of TSH whereas TSH regulation of HHEX mRNA levels exhibits an IUDRC that overlaps the IUDRC of thyroid-specific genes. In contrast to findings during mouse development, TTFs do not have major effects on the levels of other TTF mRNAs in adult, human thyrocytes. Thus, we found similarities and important differences in the regulation of thyroid-specific genes in mouse development and TSH regulation of these genes in adult, human thyrocytes.
Collapse
Affiliation(s)
- Daesong Jang
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Bethesda, MD, USA
| | - Bernice Marcus-Samuels
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Bethesda, MD, USA
| | - Sarah J Morgan
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Bethesda, MD, USA
| | - Joanna Klubo-Gwiezdzinska
- Metabolic Disease Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Bethesda, MD, USA
| | - Susanne Neumann
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Bethesda, MD, USA
| | - Marvin C Gershengorn
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Bethesda, MD, USA.
| |
Collapse
|
15
|
Mehrazin A, Safarpour H, Davoudi ST, Parsamanesh N, Saeedi F, Miri-Moghaddam E. Network-Based Analysis Reveals Association of FOXE1 Gene Polymorphisms in Thyroid Cancer Patients; A Case-Control Study in Southeast of Iran. Asian Pac J Cancer Prev 2020; 21:2771-2776. [PMID: 32986379 PMCID: PMC7779442 DOI: 10.31557/apjcp.2020.21.9.2771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 09/20/2020] [Indexed: 12/24/2022] Open
Abstract
Thyroid cancer (TC) is the mainly frequent endocrine cancer by different incidence rate in worldwide. However, early prediction of this cancer is still challenging due to the unclear pathogenicity. In this study with the aid of systems biology approach, performed a holistic study on GSE65144 dataset containing anaplastic thyroid carcinoma tissues. Co-expression network analysis by WGCNA suggested that highly preserved turquoise module with 1,480 genes was significantly correlated to TC. Most of the top 54 hub-genes of this module are functionality correlated to thyroid hormone generation (GO:0006590). Of these 54 hub-genes, FOXE1 has been reported previously to contain mutation asosiated to TC and chosen for experimental validation step. To this end, we conducted a case-control study including 81 TC patients and 165 controls individuals to evaluate the effects of FOXE1 functional polymorphisms (rs1867277) on the development of TC in Sistan and Balouchestan province of Iran. The polymorphisms of FOXE1 gene (rs1867277) assessed by tetra-ARMS PCR technique. Homozygous (GG) and (AA) variant of rs1867277 polymorphism were detected in 26 (32.1%) and 15 (18.5 %) of TC patients, and 66 (40.0%), and 15 (9.1%) in controls, respectively (p-value= 0.03, OR= 2.53). The A allele frequency was 70 (43.2%) in TC patients and 114 (34.5%) in controls (p-value= 0.06, OR= 1.44). Overall, our results suggested that FOXE1 gene could be used as a prognostic marker in TC and also provides information related to FOXE1 functional polymorphisms (rs1867277) in Sistan and Balouchestan province of Iran. .
Collapse
Affiliation(s)
- Ahmad Mehrazin
- Clinical Immunology Research Center, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences (BUMS), Birjand, Iran.
| | | | - Negin Parsamanesh
- Student Research Committee and Dep. of Molecular Medicine, School of Medicine, (BUMS), Birjand, Iran.
| | - Farhad Saeedi
- Student Research Committee, School of Medicine, (BUMS), Birjand, Iran.
| | - Ebrahim Miri-Moghaddam
- Cardiovascular Diseases Research Center and Department of Molecular Medicine, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
16
|
Morillo-Bernal J, Fernández LP, Santisteban P. FOXE1 regulates migration and invasion in thyroid cancer cells and targets ZEB1. Endocr Relat Cancer 2020; 27:137-151. [PMID: 31846430 PMCID: PMC6993207 DOI: 10.1530/erc-19-0156] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
Abstract
FOXE1 is a thyroid-specific transcription factor essential for thyroid gland development and maintenance of the differentiated state. Interestingly, a strong association has been recently described between FOXE1 expression and susceptibility to thyroid cancer, but little is known about the mechanisms underlying FOXE1-induced thyroid tumorigenesis. Here, we used a panel of human thyroid cancer-derived cell lines covering the spectrum of thyroid cancer phenotypes to examine FOXE1 expression and to test for correlations between FOXE1 expression, the allele frequency of two SNPs and a length polymorphism in or near the FOXE1 locus associated with cancer susceptibility, and the migration ability of thyroid cancer cell lines. Results showed that FOXE1 expression correlated with differentiation status according to histological sub-type, but not with SNP genotype or cell migration ability. However, loss-and-gain-of-function experiments revealed that FOXE1 modulates cell migration, suggesting a role in epithelial-to-mesenchymal transition (EMT). Our previous genome-wide expression analysis identified Zeb1, a major EMT inducer, as a putative Foxe1 target gene. Indeed, gene silencing of FOXE1 decreased ZEB1 expression, whereas its overexpression increased ZEB1 transcriptional activity. FOXE1 was found to directly interact with the ZEB1 promoter. Lastly, ZEB1 silencing decreased the ability of thyroid tumoral cells to migrate and invade, pointing to its importance in thyroid tumor mestastases. In conclusion, we have identified ZEB1 as a bona fide target of FOXE1 in thyroid cancer cells, which provides new insights into the role of FOXE1 in regulating cell migration and invasion in thyroid cancer.
Collapse
Affiliation(s)
- Jesús Morillo-Bernal
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’, Consejo Superior Investigaciones Científicas, and Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Lara P Fernández
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’, Consejo Superior Investigaciones Científicas, and Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM-CSIC, 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 should be addressed to P Santisteban:
| |
Collapse
|
17
|
Evdokiou A, Kanisicak O, Gierek S, Barry A, Ivey MJ, Zhang X, Bodnar RJ, Satish L. Characterization of Burn Eschar Pericytes. J Clin Med 2020; 9:jcm9020606. [PMID: 32102389 PMCID: PMC7074206 DOI: 10.3390/jcm9020606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022] Open
Abstract
Pericytes are cells that reside adjacent to microvasculature and regulate vascular function. Pericytes gained great interest in the field of wound healing and regenerative medicine due to their multipotential fate and ability to enhance angiogenesis. In burn wounds, scarring and scar contractures are the major pathologic feature and cause loss of mobility. The present study investigated the influence of burn wound environment on pericytes during wound healing. Pericytes isolated from normal skin and tangentially excised burn eschar tissues were analyzed for differences in gene and protein expression using RNA-seq., immunocytochemistry, and ELISA analyses. RNA-seq identified 443 differentially expressed genes between normal- and burn eschar-derived pericytes. Whereas, comparing normal skin pericytes to normal skin fibroblasts identified 1021 distinct genes and comparing burn eschar pericytes to normal skin fibroblasts identified 2449 differential genes. Altogether, forkhead box E1 (FOXE1), a transcription factor, was identified as a unique marker for skin pericytes. Interestingly, FOXE1 levels were significantly elevated in burn eschar pericytes compared to normal. Additionally, burn wound pericytes showed increased expression of profibrotic genes periostin, fibronectin, and endosialin and a gain in contractile function, suggesting a contribution to scarring and fibrosis. Our findings suggest that the burn wound environment promotes pericytes to differentiate into a myofibroblast-like phenotype promoting scar formation and fibrosis.
Collapse
Affiliation(s)
- Alexander Evdokiou
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA; (O.K.); (M.J.I.)
| | - Stephanie Gierek
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
| | - Amanda Barry
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
| | - Malina J. Ivey
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA; (O.K.); (M.J.I.)
| | - Xiang Zhang
- Genomics, Epigenomics and Sequencing Core, University of Cincinnati, Cincinnati, OH 45267, USA;
| | - Richard J. Bodnar
- Veterans Affairs Medical Center, University Dr. C, Pittsburgh, PA 15240, USA;
| | - Latha Satish
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA; (O.K.); (M.J.I.)
- Correspondence: or ; Tel.: +1-513-872-6278
| |
Collapse
|
18
|
Wang F, Zang Y, Li M, Liu W, Wang Y, Yu X, Li H, Wang F, Liu S. DUOX2 and DUOXA2 Variants Confer Susceptibility to Thyroid Dysgenesis and Gland- in-situ With Congenital Hypothyroidism. Front Endocrinol (Lausanne) 2020; 11:237. [PMID: 32425884 PMCID: PMC7212429 DOI: 10.3389/fendo.2020.00237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/31/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Thyroid dysgenesis (TD), which is caused by gland developmental abnormalities, is the most common cause of congenital hypothyroidism (CH). In addition, advances in diagnostic techniques have facilitated the identification of mild CH patients with a gland-in-situ (GIS) with normal thyroid morphology. Therefore, TD and GIS account for the vast majority of CH cases. Methods: Sixteen known genes to be related to CH were sequenced and screened for variations by next-generation sequencing (NGS) in a cohort of 377 CH cases, including 288 TD cases and 89 GIS cases. Results: In our CH cohort, we found that DUOX2 (21.22%) was the most commonly variant pathogenic gene, while DUOXA2 was prominent in TD (18.75%) and DUOX2 was prominent in GIS (34.83%). Both biallelic and triple variants of DUOX2 were found to be most common in children with TD and children with GIS. The most frequent combination was DUOX2 with DUOXA1 among the 61 patients who carried digenic variants. We also found for the first time that biallelic TG, DUOXA2, and DUOXA1 variants participate in the pathogenesis of TD. In addition, the variant p.Y246X in DUOXA2 was the most common variant hotspot, with 58 novel variants identified in our study. Conclusion: We meticulously described the types and characteristics of variants from sixteen known gene in children with TD and GIS in the Chinese population, suggesting that DUOXA2 and DUOX2 variants may confer susceptibility to TD and GIS via polygenic inheritance and multiple factors, which further expands the genotype-phenotype spectrum of CH in China.
Collapse
Affiliation(s)
- Fengqi Wang
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yucui Zang
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Miaomiao Li
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenmiao Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangang Wang
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaolong Yu
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hua Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fang Wang
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Fang Wang
| | - Shiguo Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
- Shiguo Liu
| |
Collapse
|
19
|
Luzón-Toro B, Fernández RM, Villalba-Benito L, Torroglosa A, Antiñolo G, Borrego S. Influencers on Thyroid Cancer Onset: Molecular Genetic Basis. Genes (Basel) 2019; 10:E913. [PMID: 31717449 PMCID: PMC6895808 DOI: 10.3390/genes10110913] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/25/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023] Open
Abstract
Thyroid cancer, a cancerous tumor or growth located within the thyroid gland, is the most common endocrine cancer. It is one of the few cancers whereby incidence rates have increased in recent years. It occurs in all age groups, from children through to seniors. Most studies are focused on dissecting its genetic basis, since our current knowledge of the genetic background of the different forms of thyroid cancer is far from complete, which poses a challenge for diagnosis and prognosis of the disease. In this review, we describe prevailing advances and update our understanding of the molecular genetics of thyroid cancer, focusing on the main genes related with the pathology, including the different noncoding RNAs associated with the disease.
Collapse
Affiliation(s)
- Berta Luzón-Toro
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.L.-T.); (R.M.F.); (L.V.-B.); (A.T.); (G.A.)
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 41013 Seville, Spain
| | - Raquel María Fernández
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.L.-T.); (R.M.F.); (L.V.-B.); (A.T.); (G.A.)
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 41013 Seville, Spain
| | - Leticia Villalba-Benito
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.L.-T.); (R.M.F.); (L.V.-B.); (A.T.); (G.A.)
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 41013 Seville, Spain
| | - Ana Torroglosa
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.L.-T.); (R.M.F.); (L.V.-B.); (A.T.); (G.A.)
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 41013 Seville, Spain
| | - Guillermo Antiñolo
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.L.-T.); (R.M.F.); (L.V.-B.); (A.T.); (G.A.)
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 41013 Seville, Spain
| | - Salud Borrego
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.L.-T.); (R.M.F.); (L.V.-B.); (A.T.); (G.A.)
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 41013 Seville, Spain
| |
Collapse
|
20
|
Bann DV, Jin Q, Sheldon KE, Houser KR, Nguyen L, Warrick JI, Baker MJ, Broach JR, Gerhard GS, Goldenberg D. Genetic Variants Implicate Dual Oxidase-2 in Familial and Sporadic Nonmedullary Thyroid Cancer. Cancer Res 2019; 79:5490-5499. [PMID: 31501191 DOI: 10.1158/0008-5472.can-19-0721] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/03/2019] [Accepted: 09/03/2019] [Indexed: 11/16/2022]
Abstract
Highly penetrant hereditary thyroid cancer manifests as familial nonmedullary thyroid cancer (FNMTC), whereas low-penetrance hereditary thyroid cancer manifests as sporadic disease and is associated with common polymorphisms, including rs965513[A]. Whole-exome sequencing of an FNMTC kindred identified a novel Y1203H germline dual oxidase-2 (DUOX2) mutation. DUOX2Y1203H is enzymatically active, with increased production of reactive oxygen species. Furthermore, patients with sporadic thyroid cancer homozygous for rs965513[A] demonstrated higher DUOX2 expression than heterozygous rs965513[A/G] or homozygous rs965513[A]-negative patients. These data suggest that dysregulated hydrogen peroxide metabolism is a common mechanism by which high- and low-penetrance genetic factors increase thyroid cancer risk. SIGNIFICANCE: This study provides novel insights into the genetic and molecular mechanisms underlying familial and sporadic thyroid cancers.
Collapse
Affiliation(s)
- Darrin V Bann
- Department of Otolaryngology-Head & Neck Surgery, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania.,Institute for Personalized Medicine, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania
| | - Qunyan Jin
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kathryn E Sheldon
- Institute for Personalized Medicine, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania
| | - Kenneth R Houser
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania
| | - Lan Nguyen
- Institute for Personalized Medicine, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania
| | - Joshua I Warrick
- Department of Pathology, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania
| | - Maria J Baker
- Department of Medicine, Division of Hematology/Oncology, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania
| | - James R Broach
- Institute for Personalized Medicine, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania.,Department of Biochemistry and Molecular Biology, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania
| | - Glenn S Gerhard
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - David Goldenberg
- Department of Otolaryngology-Head & Neck Surgery, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania.
| |
Collapse
|
21
|
López-Márquez A, Fernández-Méndez C, Recacha P, Santisteban P. Regulation of Foxe1 by Thyrotropin and Transforming Growth Factor Beta Depends on the Interplay Between Thyroid-Specific, CREB and SMAD Transcription Factors. Thyroid 2019; 29:714-725. [PMID: 30652527 DOI: 10.1089/thy.2018.0136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: Thyroid follicular cells are characterized by the expression of a specific set of genes necessary for the synthesis and secretion of thyroid hormones, which are in turn regulated by the transcription factors Nkx2-1, Pax8, and Foxe1. Thyroid differentiation is finely tuned by the balance between positive regulatory signals, including thyrotropin (TSH), and by negative regulatory signals, such as transforming growth factor beta (TGF-β), which counteracts the action of TSH. A role for Foxe1 as a mediator of hormonal and growth-factor control of thyroid differentiation has been previously suggested. Therefore, the aim of this work was to study the mechanisms governing Foxe1 expression to define the ligands and signals that regulate one of the important factors in thyroid differentiation. Methods: Expression of Foxe1 was evaluated in rat PCCl3 thyroid follicular cells under different treatments. The mouse Foxe1 promoter was cloned, and site-directed mutagenesis was undertaken to study its transcriptional regulation and to identify response elements. Protein/DNA binding assays were performed to evaluate the binding of different transcription factors, and gene-silencing approaches were used to elucidate their functional roles. Results:In silico analysis of the Foxe1 promoter identified binding sites for Nkx2-1, Pax8, Foxe1, and Smad proteins, as well as cAMP-response element (CRE) sites. It was found that both CRE-binding protein and CRE modulator were necessary for the TSH-mediated induction of Foxe1 expression via the cAMP/PKA signaling pathway. Moreover, transcription of Foxe1 was regulated by Nkx2-1 and Pax8 and by itself, suggesting an autoregulatory mechanism of activation and an important role for thyroid transcription factors. Finally, TGF-β, through Smad proteins, inhibited the TSH-induced Foxe1 expression. Conclusions: This study shows that Foxe1 is the final target of TSH/cAMP and TGF-β regulation that mediates expression of thyroid differentiation genes, and provides evidence of an interplay between CRE-binding proteins, thyroid transcription factors, and Smad proteins in its regulation. Thus, Foxe1 plays an important role in the complex transcriptional network that regulates thyroid follicular cell differentiation.
Collapse
Affiliation(s)
- Arístides López-Márquez
- 1 Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Celia Fernández-Méndez
- 1 Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Pablo Recacha
- 1 Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Pilar Santisteban
- 1 Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
- 2 CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
22
|
Ock S, Ahn J, Lee SH, Kim HM, Kang H, Kim YK, Kook H, Park WJ, Kim S, Kimura S, Jung CK, Shong M, Holzenberger M, Abel ED, Lee TJ, Cho BY, Kim HS, Kim J. Thyrocyte-specific deletion of insulin and IGF-1 receptors induces papillary thyroid carcinoma-like lesions through EGFR pathway activation. Int J Cancer 2018; 143:2458-2469. [PMID: 30070361 DOI: 10.1002/ijc.31779] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/16/2018] [Accepted: 07/26/2018] [Indexed: 01/25/2023]
Abstract
Insulin and insulin-like growth factor (IGF)-1 signaling in the thyroid are thought to be permissive for the coordinated regulation by thyroid-stimulating hormone (TSH) of thyrocyte proliferation and hormone production. However, the integrated role of insulin receptor (IR) and IGF-1 receptor (IGF-1R) in thyroid development and function has not been explored. Here, we generated thyrocyte-specific IR and IGF-1R double knockout (DTIRKO) mice to precisely evaluate the coordinated functions of these receptors in the thyroid of neonates and adults. Neonatal DTIRKO mice displayed smaller thyroids, paralleling defective folliculogenesis associated with repression of the thyroid-specific transcription factor Foxe1. By contrast, at postnatal day 14, absence of IR and IGF-1R paradoxically induced thyrocyte proliferation, which was mediated by mTOR-dependent signaling pathways. Furthermore, we found elevated production of TSH during the development of follicular hyperplasia at 8 weeks of age. By 50 weeks, all DTIRKO mice developed papillary thyroid carcinoma (PTC)-like lesions that correlated with induction of the ErbB pathway. Taken together, these data define a critical role for IR and IGF-1R in neonatal thyroid folliculogenesis. They also reveal an important reciprocal relationship between IR/IGF-1R and TSH/ErbB signaling in the pathogenesis of thyroid follicular hyperplasia and, possibly, of papillary carcinoma.
Collapse
Affiliation(s)
- Sangmi Ock
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Jihyun Ahn
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Seok Hong Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Hyun Min Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Hyun Kang
- Department of Anesthesiology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Hyun Kook
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, Gwangju, Korea
| | - Woo Jin Park
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Shin Kim
- Department of Immunology, Keimyung University School of Medicine, Daegu, Korea
| | - Shioko Kimura
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Department of Internal Medicine, Chungnam National University, Daejeon, Korea
| | - Martin Holzenberger
- INSERM and Sorbonne University, Saint-Antoine Research Center, Paris, France
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Tae Jin Lee
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Bo Youn Cho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Ho-Shik Kim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| |
Collapse
|
23
|
Chen YH, Zhang YQ. Exploration of the association between FOXE1 gene polymorphism and differentiated thyroid cancer: a meta-analysis. BMC MEDICAL GENETICS 2018; 19:83. [PMID: 29788924 PMCID: PMC5964894 DOI: 10.1186/s12881-018-0604-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Several association analyses and linkage researches indicated that inherited genetic variations effectively influence differentiated thyroid carcinogenesis. METHODS The results from 15 published studies on differentiated thyroid carcinoma (DTC) were combined. The genetic model included rs965513, rs944289 and rs1867277. Meta-analyses were performed and cochran's χ2 based Q-statistic and I2 test were performed to assess heterogeneity using STATA software. RESULTS Significant results were noticed for rs965513(Odds Ratio(OR) = 1.162(1.117, 1.208)), rs944289(OR = 1.082(1.035, 1.131)) and rs1867277(OR = 1.415(1.324, 1.512)). In the subgroup analysis by ethnicity, rs965513 polymorphism conferred that risk of Caucasians (OR = 1.168(1.122, 1.215)) was more than that of East Asians of 1.35 (OR = 0.897(0.680, 1.193)). CONCLUSION This meta-analysis revealed that common variations of FOXE1 (rs965513, rs944289 and rs1867277) were risk factors associated with increased DTC susceptibility.
Collapse
Affiliation(s)
- Yong-Hui Chen
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China. .,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China.
| | - Ying-Qiang Zhang
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| |
Collapse
|
24
|
Camargo RY, Kanamura CT, Friguglietti CU, Nogueira CR, Iorcansky S, Tincani AJ, Bezerra AK, Brust E, Koyama FC, Camargo AA, Rego FOR, Galante PAF, Medeiros-Neto G, Rubio IGS. Histopathological Characterization and Whole Exome Sequencing of Ectopic Thyroid: Fetal Architecture in a Functional Ectopic Gland from Adult Patient. Int J Endocrinol 2018; 2018:4682876. [PMID: 29593791 PMCID: PMC5822907 DOI: 10.1155/2018/4682876] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/16/2017] [Indexed: 12/17/2022] Open
Abstract
Ectopic thyroid results from a migration defect of the developing gland during embryogenesis causing congenital hypothyroidism. But it has also been detected in asymptomatic individuals. This study aimed to investigate the histopathological, functional, and genetic features of human ectopic thyroids. Six samples were histologically examined, and the expression of the specific thyroid proteins was assessed by immunohistochemistry. Two samples were submitted to whole exome sequencing. An oropharynx sample showed immature fetal architecture tissue with clusters or cords of oval thyrocytes and small follicles; one sample exhibited a normal thyroid pattern while four showed colloid goiter. All ectopic thyroids expressed the specific thyroid genes and T4 at similar locations to those observed in normal thyroid. No somatic mutations associated with ectopic thyroid were found. This is the first immature thyroid fetal tissue observed in an ectopic thyroid due to the arrest of structural differentiation early in the colloid stage of development that proved able to synthesize thyroid hormone but not to respond to TSH. Despite the ability of all ectopic thyroids to synthetize specific thyroid proteins and T4, at some point in life, it may be insufficient to support body growth leading to hypothyroidism, as observed in some of the patients.
Collapse
Affiliation(s)
- Rosalinda Yasato Camargo
- Thyroid Unit, Cellular and Molecular Endocrine Laboratory, LIM-25, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Avenida Doutor Arnaldo 455, Cerqueira César, 01246-904 São Paulo, SP, Brazil
| | - Cristina Takami Kanamura
- Adolfo Lutz Institute, São Paulo Public Health Service, Av. Dr. Arnaldo 355, Cerqueira César, 01246-000 São Paulo, SP, Brazil
| | | | - Célia Regina Nogueira
- Department of Internal Medicine, Botucatu School of Medicine, UNESP, Av. Prof. Montenegro, s/n Distrito de Rubião Junior, 18618-687 Botucatu, SP, Brazil
| | - Sonia Iorcansky
- Servicio de Endocrinología, Hospital de Pediatría Dr. Juan Garrahan, Combate de los Pozos 1881, C1245AAM Buenos Aires, Argentina
| | - Alfio José Tincani
- Departamento de Cirurgia na Disciplina de Cirurgia de Cabeça e Pescoço da Faculdade de Ciências Médicas da UNICAMP, R. Tessália Vieira de Camargo 126, 13083-887 Campinas, SP, Brazil
| | - Ana Karina Bezerra
- Medicine School, Universidade de Fortaleza (Unifor), Av. Washington Soares 1321, Edson Queiroz, 60811-905 Fortaleza, CE, Brazil
| | - Ester Brust
- Postgraduate Program in Biotechnology, Universidade Federal de São Paulo (UNIFESP), Pedro de Toledo 669, 040399-032 São Paulo, SP, Brazil
- Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo, Departamento de Ciências Biológicas, Postgraduation Programs in Biotechnology and Structural and Functional Biology, UNIFESP, Pedro de Toledo 669, 040399-032 São Paulo, SP, Brazil
| | | | - Anamaria Aranha Camargo
- Molecular Oncology Center, Hospital Sírio-Libanés, Rua Prof. Daher Cutait 69, 01308-060 São Paulo, SP, Brazil
| | - Fernanda Orpinelli R. Rego
- Molecular Oncology Center, Hospital Sírio-Libanés, Rua Prof. Daher Cutait 69, 01308-060 São Paulo, SP, Brazil
| | | | - Geraldo Medeiros-Neto
- Thyroid Unit, Cellular and Molecular Endocrine Laboratory, LIM-25, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Avenida Doutor Arnaldo 455, Cerqueira César, 01246-904 São Paulo, SP, Brazil
| | - Ileana Gabriela Sanchez Rubio
- Postgraduate Program in Biotechnology, Universidade Federal de São Paulo (UNIFESP), Pedro de Toledo 669, 040399-032 São Paulo, SP, Brazil
- Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo, Departamento de Ciências Biológicas, Postgraduation Programs in Biotechnology and Structural and Functional Biology, UNIFESP, Pedro de Toledo 669, 040399-032 São Paulo, SP, Brazil
| |
Collapse
|
25
|
Kizys MML, Louzada RA, Mitne-Neto M, Jara JR, Furuzawa GK, de Carvalho DP, Dias-da-Silva MR, Nesi-França S, Dupuy C, Maciel RMB. DUOX2 Mutations Are Associated With Congenital Hypothyroidism With Ectopic Thyroid Gland. J Clin Endocrinol Metab 2017; 102:4060-4071. [PMID: 28666341 DOI: 10.1210/jc.2017-00832] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/22/2017] [Indexed: 12/11/2022]
Abstract
CONTEXT Thyroid dysgenesis (TD) is the leading cause of congenital hypothyroidism (CH). The etiology of TD remains unknown in ∼90% of cases, the most common form being thyroid ectopia (TE) (48% to 61%). OBJECTIVE To search for candidate genes in hypothyroid children with TE. DESIGN, SETTING, AND PARTICIPANTS We followed a cohort of 268 children with TD and performed whole-exome sequencing (WES) in three children with CH with TE (CHTE) and compared them with 18 thyroid-healthy controls. We then screened an additional 41 children with CHTE by Sanger sequencing and correlated the WES and Sanger molecular findings with in vitro functional analysis. MAIN OUTCOME MEASURES Genotyping, mutation prediction analysis, and in vitro functional analysis. RESULTS We identified seven variants in the DUOX2 gene, namely G201E, L264CfsX57, P609S, M650T, E810X, M822V, and E1017G, and eight known variations. All children carrying DUOX2 variations had high thyroid-stimulating hormone levels at neonatal diagnosis. All mutations were localized in the N-terminal segment, and three of them led to effects on cell surface targeting and reactive oxygen species generation. The DUOX2 mutants also altered the interaction with the maturation factor DUOXA2 and the formation of a stable DUOX2/DUOXA2 complex at the cell surface, thereby impairing functional enzymatic activity. We observed no mutations in the classic genes related to TD or in the DUOX1 gene. CONCLUSION Our findings suggest that, in addition to thyroid hormonogenesis, the DUOX2 N-terminal domain may play a role in thyroid development.
Collapse
Affiliation(s)
- Marina M L Kizys
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Ruy A Louzada
- UMR 8200 CNRS, Villejuif, 94800, France
- Institut Gustave Roussy, Villejuif, 94800, France
- Université Paris-Saclay, Orsay, 91405, France
- Laboratory of Endocrine Physiology Doris Rosenthal, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Miguel Mitne-Neto
- Fleury Group, São Paulo 04344-070, Brazil
- Human Genome and Stem Cell Research Center, Biosciences Institute, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Jessica R Jara
- Department of Pediatrics, Universidade Federal do Paraná, Curitiba 80060-240, Brazil
| | - Gilberto K Furuzawa
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Denise P de Carvalho
- Laboratory of Endocrine Physiology Doris Rosenthal, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Magnus R Dias-da-Silva
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Suzana Nesi-França
- Department of Pediatrics, Universidade Federal do Paraná, Curitiba 80060-240, Brazil
| | - Corinne Dupuy
- UMR 8200 CNRS, Villejuif, 94800, France
- Institut Gustave Roussy, Villejuif, 94800, France
- Université Paris-Saclay, Orsay, 91405, France
| | - Rui M B Maciel
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
- Fleury Group, São Paulo 04344-070, Brazil
| |
Collapse
|
26
|
de Conti A, Beland FA, Pogribny IP. The role of epigenomic alterations in furan-induced hepatobiliary pathologies. Food Chem Toxicol 2017; 109:677-682. [DOI: 10.1016/j.fct.2017.07.049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/24/2017] [Indexed: 01/05/2023]
|
27
|
Abstract
Thyroid hormones are crucial for organismal development and homeostasis. In humans, untreated congenital hypothyroidism due to thyroid agenesis inevitably leads to cretinism, which comprises irreversible brain dysfunction and dwarfism. Elucidating how the thyroid gland - the only source of thyroid hormones in the body - develops is thus key for understanding and treating thyroid dysgenesis, and for generating thyroid cells in vitro that might be used for cell-based therapies. Here, we review the principal mechanisms involved in thyroid organogenesis and functional differentiation, highlighting how the thyroid forerunner evolved from the endostyle in protochordates to the endocrine gland found in vertebrates. New findings on the specification and fate decisions of thyroid progenitors, and the morphogenesis of precursor cells into hormone-producing follicular units, are also discussed.
Collapse
Affiliation(s)
- Mikael Nilsson
- Sahlgrenska Cancer Center, Institute of Biomedicine, University of Gothenburg, Göteborg SE-40530, Sweden
| | - Henrik Fagman
- Sahlgrenska Cancer Center, Institute of Biomedicine, University of Gothenburg, Göteborg SE-40530, Sweden.,Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Göteborg SE-41345, Sweden
| |
Collapse
|
28
|
FOXE1 Mutation Screening in a Case with Cleft Lip, Hypothyroidism, and Thyroid Carcinoma: A New Syndrome? Case Rep Genet 2017; 2017:6390545. [PMID: 28928994 PMCID: PMC5591984 DOI: 10.1155/2017/6390545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/19/2017] [Accepted: 07/30/2017] [Indexed: 01/30/2023] Open
Abstract
A 26-year-old woman is referred to the Internal Medicine consultation due to increases in laboratory studies associated with Papillary Thyroid Carcinoma (PTC) that was confirmed by histopathological studies. Her clinical history revealed that, at 3 months of age, she was successfully treated with surgery for cleft lip (CL) and at the age of 24 years was diagnosed with hypothyroidism. Single nucleotide polymorphisms (SNPs) in FOXE1 and its promoter regions have been associated with various etiologies related to the thyroid, including orofacial clefting, specially cleft palate (CP) and CL, hypothyroidism (HT), and thyroid cancer. The association of CL, HT, and PTC might be component of a new syndrome; however FOXE1 coding region, which has been involved with these entities, has not exhibited mutations or SNPs. Further study of other genes may help in better characterization of the possible syndrome.
Collapse
|
29
|
Kelkar MG, Thakur B, Derle A, Chatterjee S, Ray P, De A. Tumor suppressor protein p53 exerts negative transcriptional regulation on human sodium iodide symporter gene expression in breast cancer. Breast Cancer Res Treat 2017; 164:603-615. [PMID: 28528452 DOI: 10.1007/s10549-017-4297-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/14/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE Aberrant expression of human sodium iodide symporter (NIS) in breast cancer (BC) is well documented but the transcription factors (TF) regulating its aberrant expression is poorly known. We identify the presence of three p53 binding sites on the human NIS promoter sequence by conducting genome-wide TF analysis, and further investigate their regulatory role. METHODS The differences in transcription and translation were measured by real-time PCR, luciferase reporter assay, site-directed mutagenesis, in vivo optical imaging, and chromatin immunoprecipitation. The relation of NIS and p53 in clinical samples was judged by TCGA data analysis and immunohistochemistry. RESULTS Overexpression of wild-type p53 as a transgene or pharmacological activation by doxorubicin drug treatment shows significant suppression of NIS transcription in multiple BC cell types which also results in lowered NIS protein content and cellular iodide intake. NIS repression by activated p53 is further confirmed by non-invasive bioluminescence imaging in live cell and orthotropic tumor model. Abrogation of p53-binding sites by directional mutagenesis confirms reversal of transcriptional activity in wild-type p53-positive BC cells. We also observe direct binding of p53 to these sites on the human NIS promoter. Importantly, TCGA data analysis of NIS and p53 co-expression registers an inverse relationship between the two candidates. CONCLUSION Our data for the first time highlight the role of p53 as a negative regulator of functional NIS expression in BC, where the latter is a potential targeted radioiodine therapy candidate. Thus, the study provides an important insight into prospective clinical application of this approach that may significantly impact the patient with mutant versus wild-type p53 profile.
Collapse
Affiliation(s)
- Madhura G Kelkar
- Molecular Functional Imaging Lab, Tata Memorial Centre, ACTREC, Sector 22, Kharghar, Navi Mumbai, 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Bhushan Thakur
- Imaging Cell Signaling and Therapeutics Lab, Tata Memorial Centre, ACTREC, Navi Mumbai, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Abhishek Derle
- Molecular Functional Imaging Lab, Tata Memorial Centre, ACTREC, Sector 22, Kharghar, Navi Mumbai, 410210, India
| | - Sushmita Chatterjee
- Molecular Functional Imaging Lab, Tata Memorial Centre, ACTREC, Sector 22, Kharghar, Navi Mumbai, 410210, India
| | - Pritha Ray
- Imaging Cell Signaling and Therapeutics Lab, Tata Memorial Centre, ACTREC, Navi Mumbai, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Abhijit De
- Molecular Functional Imaging Lab, Tata Memorial Centre, ACTREC, Sector 22, Kharghar, Navi Mumbai, 410210, India. .,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India.
| |
Collapse
|
30
|
Murata T, Iwadate M, Takizawa Y, Miyakoshi M, Hayase S, Yang W, Cai Y, Yokoyama S, Nagashima K, Wakabayashi Y, Zhu J, Kimura S. An Adult Mouse Thyroid Side Population Cell Line that Exhibits Enriched Epithelial-Mesenchymal Transition. Thyroid 2017; 27:460-474. [PMID: 28125936 PMCID: PMC5346910 DOI: 10.1089/thy.2016.0130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Studies of thyroid stem/progenitor cells have been hampered due to the small organ size and lack of tissue, which limits the yield of these cells. A continuous source that allows the study and characterization of thyroid stem/progenitor cells is desired to push the field forward. METHOD A cell line was established from Hoechst-resistant side population cells derived from mouse thyroid that were previously shown to contain stem/progenitor-like cells. Characterization of these cells were carried out by using in vitro two- and three-dimensional cultures and in vivo reconstitution of mice after orthotopic or intravenous injection, in conjunction with quantitative reverse transcription polymerase chain reaction, Western blotting, immunohisto(cyto)chemistry/immunofluorescence, and RNA seq analysis. RESULTS These cells were named SPTL (side population cell-derived thyroid cell line). Under low serum culturing conditions, SPTL cells expressed the thyroid differentiation marker NKX2-1, a transcription factor critical for thyroid differentiation and function, while no expression of other thyroid differentiation marker genes were observed. SPTL cells formed follicle-like structures in Matrigel® cultures, which did not express thyroid differentiation marker genes. In mouse models of orthotopic and intravenous injection, the latter following partial thyroidectomy, a few SPTL cells were found in part of the follicles, most of which expressed NKX2-1. SPTL cells highly express genes involved in epithelial-mesenchymal transition, as demonstrated by RNA seq analysis, and exhibit a gene-expression pattern similar to anaplastic thyroid carcinoma. CONCLUSION These results demonstrate that SPTL cells have the capacity to differentiate into thyroid to a limited degree. SPTL cells may provide an excellent tool to study stem cells, including cancer stem cells of the thyroid.
Collapse
Affiliation(s)
- Tsubasa Murata
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Manabu Iwadate
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Yoshinori Takizawa
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Masaaki Miyakoshi
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Suguru Hayase
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wenjing Yang
- DNA Sequencing and Genomics Core, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Yan Cai
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Shigetoshi Yokoyama
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kunio Nagashima
- Electron Microscope Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Yoshiyuki Wakabayashi
- DNA Sequencing and Genomics Core, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Jun Zhu
- Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Shioko Kimura
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
31
|
Alotaibi H, Tuzlakoğlu-Öztürk M, Tazebay UH. The Thyroid Na+/I- Symporter: Molecular Characterization and Genomic Regulation. Mol Imaging Radionucl Ther 2017; 26:92-101. [PMID: 28117294 PMCID: PMC5283716 DOI: 10.4274/2017.26.suppl.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Iodide (I-) is an essential constituent of the thyroid hormones triiodothyronine (T3) and thyroxine (T4), and the iodide concentrating mechanism of the thyroid gland is essential for the synthesis of these hormones. In addition, differential uptake of iodine isotopes (radioiodine) is a key modality for the diagnosis and therapy of thyroid cancer. The sodium dependent iodide transport activity of the thyroid gland is mainly attributed to the functional expression of the Na+/I- Symporter (NIS) localized at the basolateral membrane of thyrocytes. In this paper, we review and summarize current data on molecular characterization, on structure and function of NIS protein, as well as on the transcriptional regulation of NIS encoding gene in the thyroid gland. We also propose that a better and more precise understanding of NIS gene regulation at the molecular level in both healthy and malignant thyroid cells may lead to the identification of small molecule candidates. These could then be translated into clinical practice for better induction and more effective modulation of radioiodine uptake in dedifferentiated thyroid cancer cells and in their distant metastatic lesions.
Collapse
Affiliation(s)
| | | | - Uygar Halis Tazebay
- Gebze Technical University, Department of Molecular Biology and Genetics, Kocaeli, Turkey, Phone: +90 262 605 25 22, E-mail:
| |
Collapse
|
32
|
Rossich LE, Thomasz L, Nicola JP, Nazar M, Salvarredi LA, Pisarev M, Masini-Repiso AM, Christophe-Hobertus C, Christophe D, Juvenal GJ. Effects of 2-iodohexadecanal in the physiology of thyroid cells. Mol Cell Endocrinol 2016; 437:292-301. [PMID: 27568464 DOI: 10.1016/j.mce.2016.08.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/22/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
Abstract
Iodide has direct effects on thyroid function. Several iodinated lipids are biosynthesized by the thyroid and they were postulated as intermediaries in the action of iodide. Among them, 2-iodohexadecanal (2-IHDA) has been identified and proposed to play a role in thyroid autoregulation. The aim of this study was to compare the effect of iodide and 2-IHDA on thyroid cell physiology. For this purpose, FRTL-5 thyroid cells were incubated with the two compounds during 24 or 48 h and several thyroid parameters were evaluated such as: iodide uptake, intracellular calcium and H2O2 levels. To further explore the molecular mechanism involved in 2-IHDA action, transcript and protein levels of genes involved in thyroid hormone biosynthesis, as well as the transcriptional expression of these genes were evaluated in the presence of iodide and 2-IHDA. The results obtained indicate that 2-IHDA reproduces the action of excess iodide on the "Wolff-Chaikoff" effect as well as on thyroid specific genes transcription supporting its role in thyroid autoregulation.
Collapse
Affiliation(s)
- Luciano E Rossich
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, CONICET, Buenos Aires, Argentina
| | - Lisa Thomasz
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, CONICET, Buenos Aires, Argentina
| | - Juan P Nicola
- Department of Clinical Biochemistry, School of Chemical Sciences, National University of Cordoba, CONICET, Buenos Aires, Argentina
| | - Magali Nazar
- Department of Clinical Biochemistry, School of Chemical Sciences, National University of Cordoba, CONICET, Buenos Aires, Argentina
| | - Leonardo A Salvarredi
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, CONICET, Buenos Aires, Argentina
| | - Mario Pisarev
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, CONICET, Buenos Aires, Argentina; Department of Human Biochemistry, University of Buenos Aires School of Medicine, CONICET, Buenos Aires, Argentina
| | - Ana M Masini-Repiso
- Department of Clinical Biochemistry, School of Chemical Sciences, National University of Cordoba, CONICET, Buenos Aires, Argentina
| | | | | | - Guillermo J Juvenal
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
33
|
Tryndyak V, de Conti A, Doerge DR, Olson GR, Beland FA, Pogribny IP. Furan-induced transcriptomic and gene-specific DNA methylation changes in the livers of Fischer 344 rats in a 2-year carcinogenicity study. Arch Toxicol 2016; 91:1233-1243. [PMID: 27387713 DOI: 10.1007/s00204-016-1786-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/22/2016] [Indexed: 01/10/2023]
Abstract
Furan is a significant food contaminant and a potent hepatotoxicant and rodent liver carcinogen. The carcinogenic effect of furan has been attributed to genotoxic and non-genotoxic, including epigenetic, changes in the liver; however, the mechanisms of the furan-induced liver tumorigenicity are still unclear. The goal of the present study was to investigate the role of transcriptomic and epigenetic events in the development of hepatic lesions in Fischer (F344) rats induced by furan treatment in a classic 2-year rodent tumorigenicity bioassay. High-throughput whole-genome transcriptomic analysis demonstrated distinct alterations in gene expression in liver lesions induced in male F344 rats treated with 0.92 or 2.0 mg furan/kg body weight (bw)/day for 104 weeks. Compared to normal liver tissue, 1336 and 1541 genes were found to be differentially expressed in liver lesions in rats treated with 0.92 and 2.0 mg furan/kg bw/day, respectively, among which 1001 transcripts were differentially expressed at both doses. Pairing transcriptomic and next-generation bisulfite sequencing analyses of the common differentially expressed genes identified 42 CpG island-containing genes in which the methylation level was correlated inversely with gene expression. Forty-eight percent of these genes (20 genes, including Areg, Jag1, and Foxe1) that exhibited the most significant methylation and gene expression changes were involved in key pathways associated with different aspects of liver pathology. Our findings illustrate that gene-specific DNA methylation changes have functional consequences and may be an important component of furan hepatotoxicity and hepatocarcinogenicity.
Collapse
Affiliation(s)
- Volodymyr Tryndyak
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), 3900 NCTR Rd., Jefferson, AR, 72079, USA
| | - Aline de Conti
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), 3900 NCTR Rd., Jefferson, AR, 72079, USA
| | - Daniel R Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), 3900 NCTR Rd., Jefferson, AR, 72079, USA
| | - Greg R Olson
- Toxicologic Pathology Associates, National Center for Toxicological Research (NCTR), Jefferson, AR, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), 3900 NCTR Rd., Jefferson, AR, 72079, USA
| | - Igor P Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), 3900 NCTR Rd., Jefferson, AR, 72079, USA.
| |
Collapse
|
34
|
Sugimachi K, Matsumura T, Shimamura T, Hirata H, Uchi R, Ueda M, Sakimura S, Iguchi T, Eguchi H, Masuda T, Morita K, Takenaka K, Maehara Y, Mori M, Mimori K. Aberrant Methylation of FOXE1 Contributes to a Poor Prognosis for Patients with Colorectal Cancer. Ann Surg Oncol 2016; 23:3948-3955. [PMID: 27271927 DOI: 10.1245/s10434-016-5289-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND Hypermethylation of DNA silences gene expression and is an important event in colorectal cancer (CRC). This study aimed to identify aberrantly methylated genes that contribute to a poor prognosis for patients with CRC. METHODS The study comprehensively explored DNA methylation microarray profiles from 396 CRC samples and 45 normal control samples in a database and selected aberrantly methylated transcription factors associated with prognosis and metastasis. Using quantitative reverse transcription polymerase chain reaction, the identified genes in 140 patients with CRC were validated to assess the relationship between expression of methylated genes and prognosis. RESULTS In the study, FOXE1 was newly identified as a gene associated with prognosis and metastasis in CRC. Expression of FOXE1 in CRC tissues was significantly lower than in normal colorectal tissues (p = 0.01). The survival rate for the patients with low expression of FOXE1 was significantly lower than that for patients with high expression of FOXE1 in uni- and multivariate analyses. Inhibition of DNA methylation recovered FOXE1 expression in CRC cells. CONCLUSIONS Methylation-mediated silencing of FOXE1 expression was shown to be a potential prognostic factor in CRC.
Collapse
Affiliation(s)
- Keishi Sugimachi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.,Department of Surgery, Fukuoka City Hospital, Fukuoka, Japan
| | - Tae Matsumura
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.,Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Teppei Shimamura
- Department of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidenari Hirata
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Ryutaro Uchi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Masami Ueda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Shotaro Sakimura
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Tomohiro Iguchi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Hidetoshi Eguchi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Kazutoyo Morita
- Department of Surgery, Fukuoka City Hospital, Fukuoka, Japan
| | - Kenji Takenaka
- Department of Surgery, Fukuoka City Hospital, Fukuoka, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.
| |
Collapse
|
35
|
Abstract
Thyroid dysgenesis (TD) is the most common cause of congenital hypothyroidism in iodine-sufficient regions and includes a spectrum of developmental anomalies. The genetic components of TD are complex. Although a sporadic disease, advances in developmental biology have revealed monogenetic forms of TD. Inheritance is not based on a simple Mendelian pattern and additional genetic elements might contribute to the phenotypic spectrum. This article summarizes the key steps of normal thyroid development and provides an update on responsible genes and underlying mechanisms of TD. Up-to-date technologies in genetics and biology will allow us to advance in our knowledge of TD.
Collapse
Affiliation(s)
- Athanasia Stoupa
- Pediatric Endocrinology, Diabetology and Gynecology Department, Necker Enfants-Malades University Hospital, Assistance Publique Hôpitaux de Paris, 149 rue de Sèvres, 75015, Paris, France; Imagine Institute, Inserm U1163, 24 boulevard du Montparnasse, 75015, Paris, France
| | - Dulanjalee Kariyawasam
- Pediatric Endocrinology, Diabetology and Gynecology Department, Necker Enfants-Malades University Hospital, Assistance Publique Hôpitaux de Paris, 149 rue de Sèvres, 75015, Paris, France; Imagine Institute, Inserm U1163, 24 boulevard du Montparnasse, 75015, Paris, France; Cochin Institute, Inserm U1016, 22 rue Mechain, 75014, Paris, France
| | - Aurore Carré
- Imagine Institute, Inserm U1163, 24 boulevard du Montparnasse, 75015, Paris, France; Cochin Institute, Inserm U1016, 22 rue Mechain, 75014, Paris, France
| | - Michel Polak
- Pediatric Endocrinology, Diabetology and Gynecology Department, Necker Enfants-Malades University Hospital, Assistance Publique Hôpitaux de Paris, 149 rue de Sèvres, 75015, Paris, France; Imagine Institute, Inserm U1163, 24 boulevard du Montparnasse, 75015, Paris, France; Cochin Institute, Inserm U1016, 22 rue Mechain, 75014, Paris, France; Paris Descartes University, Sorbonne Paris Cité, 12 rue de l'École de Médecine, 75006, Paris, France.
| |
Collapse
|
36
|
Serrano-Nascimento C, Nicola JP, Teixeira SDS, Poyares LL, Lellis-Santos C, Bordin S, Masini-Repiso AM, Nunes MT. Excess iodide downregulates Na(+)/I(-) symporter gene transcription through activation of PI3K/Akt pathway. Mol Cell Endocrinol 2016; 426:73-90. [PMID: 26872612 DOI: 10.1016/j.mce.2016.02.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 02/08/2016] [Accepted: 02/08/2016] [Indexed: 12/19/2022]
Abstract
Transcriptional mechanisms associated with iodide-induced downregulation of NIS expression remain uncertain. Here, we further analyzed the transcriptional regulation of NIS gene expression by excess iodide using PCCl3 cells. NIS promoter activity was reduced in cells treated for 12-24 h with 10(-5) to 10(-3) M NaI. Site-directed mutagenesis of Pax8 and NF-κB cis-acting elements abrogated the iodide-induced NIS transcription repression. Indeed, excess iodide (10(-3) M) excluded Pax8 from the nucleus, decreased p65 total expression and reduced their transcriptional activity. Importantly, p65-Pax8 physical interaction and binding to NIS upstream enhancer were reduced upon iodide treatment. PI3K/Akt pathway activation by iodide-induced ROS production is involved in the transcriptional repression of NIS expression. In conclusion, the results indicated that excess iodide transcriptionally represses NIS gene expression through the impairment of Pax8 and p65 transcriptional activity. Furthermore, the data presented herein described novel roles for PI3K/Akt signaling pathway and oxidative status in the thyroid autoregulatory phenomenon.
Collapse
Affiliation(s)
- Caroline Serrano-Nascimento
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Juan Pablo Nicola
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - Silvania da Silva Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Leonice Lourenço Poyares
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Camilo Lellis-Santos
- Department of Biological Sciences, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Brazil.
| | - Silvana Bordin
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Ana Maria Masini-Repiso
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - Maria Tereza Nunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
37
|
Nikitski A, Saenko V, Shimamura M, Nakashima M, Matsuse M, Suzuki K, Rogounovitch T, Bogdanova T, Shibusawa N, Yamada M, Nagayama Y, Yamashita S, Mitsutake N. Targeted Foxe1 Overexpression in Mouse Thyroid Causes the Development of Multinodular Goiter But Does Not Promote Carcinogenesis. Endocrinology 2016; 157:2182-95. [PMID: 26982637 DOI: 10.1210/en.2015-2066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent genome-wide association studies have identified several single nucleotide polymorphisms in the forkhead box E1 gene (FOXE1) locus, which are strongly associated with the risk for thyroid cancer. In addition, our recent work has demonstrated FOXE1 overexpression in papillary thyroid carcinomas. To assess possible contribution of Foxe1 to thyroid carcinogenesis, transgenic mice overexpressing Foxe1 in their thyroids under thyroglobulin promoter (Tg-Foxe1) were generated. Additionally, Tg-Foxe1 mice were exposed to x-rays at the age of 5 weeks or crossed with Pten(+/-) mice to examine the combined effect of Foxe1 overexpression with radiation or activated phosphatidylinositol-3-kinase/Akt pathway, respectively. In 5- to 8-week-old Tg-Foxe1 mice, severe hypothyroidism was observed, and mouse thyroids exhibited hypoplasia of the parenchyma. Adult 48-week-old mice were almost recovered from hypothyroidism, their thyroids were enlarged, and featured colloid microcysts and multiple benign nodules of macrofollicular-papilloid growth pattern, but no malignancy was found. Exposure of transgenic mice to 1 or 8 Gy of x-rays and Pten haploinsufficiency promoted hyperplastic nodule formation also without carcinogenic effect. These results indicate that Foxe1 overexpression is not directly involved in the development of thyroid cancer and that proper Foxe1 dosage is essential for achieving normal structure and function of the thyroid.
Collapse
Affiliation(s)
- Alyaksandr Nikitski
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Vladimir Saenko
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Mika Shimamura
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Masahiro Nakashima
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Michiko Matsuse
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Keiji Suzuki
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Tatiana Rogounovitch
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Tetiana Bogdanova
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Nobuyuki Shibusawa
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Masanobu Yamada
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Yuji Nagayama
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Shunichi Yamashita
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Norisato Mitsutake
- Departments of Radiation Medical Sciences (A.N., M.M., K.S., S.Y., N.M.), Radiation Molecular Epidemiology (V.S., S.Y.), Molecular Medicine (M.S., Y.N.), Global Health, Medicine and Welfare (T.R.), and Department of Tumor and Diagnostic Pathology (M.N.), Atomic Bomb Disease Institute, Nagasaki University; Nagasaki University Graduate School of Biomedical Sciences (A.N.); and Nagasaki University Research Centre for Genomic Instability and Carcinogenesis (N.M.), Nagasaki 852-8523, Japan; Laboratory of Morphology of Endocrine System (T.B.), State Institution V.P. Komisarenko Institute of Endocrinology and Metabolism of Academy of Medical Sciences of Ukraine, Kyiv 254114, Ukraine; and Department of Medicine and Molecular Science (N.S., M.Y.), Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
38
|
Montesinos MDM, Nicola JP, Nazar M, Peyret V, Lucero AM, Pellizas CG, Masini-Repiso AM. Nitric oxide-repressed Forkhead factor FoxE1 expression is involved in the inhibition of TSH-induced thyroid peroxidase levels. Mol Cell Endocrinol 2016; 420:105-15. [PMID: 26610751 DOI: 10.1016/j.mce.2015.11.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 11/02/2015] [Accepted: 11/13/2015] [Indexed: 01/07/2023]
Abstract
Thyroid peroxidase (TPO) is essential for thyroid hormone synthesis mediating the covalent incorporation of iodine into tyrosine residues of thyroglobulin process known as organification. Thyroid-stimulating hormone (TSH) via cAMP signaling is the main hormonal regulator of TPO gene expression. In thyroid cells, TSH-stimulated nitric oxide (NO) production inhibits TSH-induced thyroid-specific gene expression, suggesting a potential autocrine role of NO in modulating thyroid function. Indeed, NO donors downregulate TSH-induced iodide accumulation and organification in thyroid cells. Here, using FRTL-5 thyroid cells as model, we obtained insights into the molecular mechanism underlying the inhibitory effects of NO on iodide organification. We demonstrated that NO donors inhibited TSH-stimulated TPO expression by inducing a cyclic guanosine monophosphate-dependent protein kinase-mediated transcriptional repression of the TPO gene. Moreover, we characterized the FoxE1 binding site Z as mediator of the NO-inhibited TPO expression. Mechanistically, we demonstrated that NO decreases TSH-induced FoxE1 expression, thus repressing the transcripcional activation of TPO gene. Taken together, we provide novel evidence reinforcing the inhibitory role of NO on thyroid cell function, an observation of potential pathophysiological relevance associated with human thyroid pathologies that come along with changes in the NO production.
Collapse
Affiliation(s)
- María del Mar Montesinos
- 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, Córdoba, Argentina
| | - 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, Córdoba, Argentina
| | - Magalí Nazar
- 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, Córdoba, Argentina
| | - Victoria Peyret
- 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, Córdoba, Argentina
| | - Ariel Maximiliano Lucero
- 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, Córdoba, Argentina
| | - Claudia Gabriela Pellizas
- 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, Córdoba, Argentina
| | - Ana María Masini-Repiso
- 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, Córdoba, Argentina.
| |
Collapse
|
39
|
Somuncu E, Karatas A, Ferahman S, Saygili N, Yilmaz E, Ozturk O, Kapan M. The investigation of foxe1 variations in papillary thyroid carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:13458-13464. [PMID: 26722557 PMCID: PMC4680502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/25/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Recent reports indicated that incidence of thyroid carcinoma is increasing throughout the worldwide. The aim of our study was to determine a possible relationship between Forkhead box E1 (FOXE1) gene variants and histopathological features of papillary thyroid carcinoma. METHODS FOXE1 gene variations; rs894673, rs1867277 and rs3758249 were analyzed in 57 Papillary thyroid carcinoma patients and 51 age matched healthy control subjects. Restriction fragment length polymorphism (RFLP) technique was used to specifically detect the variations. RESULTS There was a significant difference in the distribution of rs894673 genotypes in Papillary thyroid carcinoma cases (P=0.01). AA genotype presence of rs1867277 was more significantly associated with several histopathological parameters such as focal and diffuse capsular invasion, lymphatic invasion, P3 with P4 tumor grade and surgical margins. AA genotype presence in rs1867277 variation was significantly associated with the classical variant which is subtype of papillary thyroid carcinoma. Furthermore, the presence of the allel A was found to be related with lymph node invasion risk by 2.46 fold, capsular invasion risk by 2.97 fold, and pT3 with pT4 pathological stage risk by 4.13 fold and the presence of allele A in rs1867277 was significantly associated with classic variants. The presence of allele A in rs1867277 was more significantly associated with several histopathological parameters in classic variant in papillary thyroid carcinoma cases such as, the presence of the A allele was found relationship with lymph node invasion risk by 2.0 fold, capsular invasion risk by 2.39 fold , and pT3 with pT4 pathological stage risk by 3.57 fold. In addition, AATT, AAAA and GATT haplotypes (rs1867277 and rs894673) were evaluated for association with papillary thyroid carcinoma cases. Our results indicate that the significant difference according to two-allele haplotype distribution between papillary thyroid carcinoma cases and control groups. CONCLUSION Our findings suggest that FOXE1 variations generate a higher risk for poor histopathological features of papillary thyroid carcinoma.
Collapse
Affiliation(s)
- Erkan Somuncu
- General Surgery, Erzincan University, Mengucek Gazi Training Research HospitalErzincan, Turkey
| | - Adem Karatas
- General Surgery, Istanbul University, Cerrahpasa Medicine FacultyIstanbul, Turkey
| | - Sina Ferahman
- General Surgery, Istanbul University, Cerrahpasa Medicine FacultyIstanbul, Turkey
| | - Neslihan Saygili
- Molecular Biology, Istanbul University, The Institute of Experimental MedicineIstanbul, Turkey
| | - Eren Yilmaz
- Molecular Biology, Istanbul University, The Institute of Experimental MedicineIstanbul, Turkey
| | - Oguz Ozturk
- Molecular Biology, Istanbul University, The Institute of Experimental MedicineIstanbul, Turkey
| | - Metin Kapan
- General Surgery, Istanbul University, Cerrahpasa Medicine FacultyIstanbul, Turkey
| |
Collapse
|
40
|
Mond M, Bullock M, Yao Y, Clifton-Bligh RJ, Gilfillan C, Fuller PJ. Somatic Mutations of FOXE1 in Papillary Thyroid Cancer. Thyroid 2015; 25:904-10. [PMID: 25950909 DOI: 10.1089/thy.2015.0030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Population-based studies have demonstrated an association of single nucleotide polymorphisms close to the thyroid transcription factor forkhead box E1 (FOXE1) gene with thyroid cancer. The dysregulation of forkhead proteins is increasingly recognized to play a role in the development and progression of cancer. The objective of the study was to seek to identify novel mutations in FOXE1 in papillary thyroid cancer (PTC) and to assess the effect of these mutations on protein expression and transcriptional function on FOXE1 responsive promoters. METHODS The study was conducted at two tertiary referral hospitals. The coding region of FOXE1 was sequenced in tissue-derived DNA or RNA from 120 patients with PTC and 110 patients with multinodular goiter (MNG). In vitro studies were performed to examine the protein expression and transcriptional function of FOXE1 mutants. A molecular model of the forkhead domain (FHD) of FOXE1 was generated using the SWISS-MODEL online server with the three-dimensional structure of FOXD3 as a template. RESULTS Three somatic missense mutations were detected in PTC resulting in the amino acid substitutions P54Q, K95Q, and L112F. One additional mutation was detected in a MNG (G140R). In vitro studies demonstrated marked impairment in transcriptional activation by all four FOXE1 mutants, which was not explained by differences in protein expression. Molecular modeling localized three of the mutations to highly conserved regions of the FHD. CONCLUSIONS We have identified novel somatic mutations of FOXE1 in PTC. Mutational inactivation of FOXE1 is an uncommon event in thyroid tumors but may contribute to thyroid carcinogenesis and dedifferentiation in concert with other oncogenic drivers.
Collapse
Affiliation(s)
- Michael Mond
- 1 MIMR-PHI Institute of Medical Research , Clayton, Victoria, Australia
- 2 Eastern Clinical School and Eastern Clinical Research Unit, Monash University , Box Hill Hospital, Box Hill, Victoria, Australia
| | - Martyn Bullock
- 3 Cancer Genetics Unit, Hormones and Cancer Group, Kolling Institute of Medical Research , Royal North Shore Hospital, Sydney, Australia
| | - Yizhou Yao
- 1 MIMR-PHI Institute of Medical Research , Clayton, Victoria, Australia
| | - Roderick J Clifton-Bligh
- 3 Cancer Genetics Unit, Hormones and Cancer Group, Kolling Institute of Medical Research , Royal North Shore Hospital, Sydney, Australia
| | - Christopher Gilfillan
- 2 Eastern Clinical School and Eastern Clinical Research Unit, Monash University , Box Hill Hospital, Box Hill, Victoria, Australia
| | - Peter J Fuller
- 1 MIMR-PHI Institute of Medical Research , Clayton, Victoria, Australia
| |
Collapse
|
41
|
Medici M, Visser WE, Visser TJ, Peeters RP. Genetic determination of the hypothalamic-pituitary-thyroid axis: where do we stand? Endocr Rev 2015; 36:214-44. [PMID: 25751422 DOI: 10.1210/er.2014-1081] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
For a long time it has been known that both hypo- and hyperthyroidism are associated with an increased risk of morbidity and mortality. In recent years, it has also become clear that minor variations in thyroid function, including subclinical dysfunction and variation in thyroid function within the reference range, can have important effects on clinical endpoints, such as bone mineral density, depression, metabolic syndrome, and cardiovascular mortality. Serum thyroid parameters show substantial interindividual variability, whereas the intraindividual variability lies within a narrow range. This suggests that every individual has a unique hypothalamus-pituitary-thyroid axis setpoint that is mainly determined by genetic factors, and this heritability has been estimated to be 40-60%. Various mutations in thyroid hormone pathway genes have been identified in persons with thyroid dysfunction or altered thyroid function tests. Because these causes are rare, many candidate gene and linkage studies have been performed over the years to identify more common variants (polymorphisms) associated with thyroid (dys)function, but only a limited number of consistent associations have been found. However, in the past 5 years, advances in genetic research have led to the identification of a large number of new candidate genes. In this review, we provide an overview of the current knowledge about the polygenic basis of thyroid (dys)function. This includes new candidate genes identified by genome-wide approaches, what insights these genes provide into the genetic basis of thyroid (dys)function, and which new techniques will help to further decipher the genetic basis of thyroid (dys)function in the near future.
Collapse
Affiliation(s)
- Marco Medici
- Rotterdam Thyroid Center, Department of Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
42
|
Cardoso-Weide L, Cardoso-Penha R, Costa M, Ferreira A, Carvalho D, Santisteban P. DuOx2 Promoter Regulation by Hormones, Transcriptional Factors and the Coactivator TAZ. Eur Thyroid J 2015; 4:6-13. [PMID: 25960956 PMCID: PMC4404926 DOI: 10.1159/000379749] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 02/06/2015] [Indexed: 01/13/2023] Open
Abstract
The production of H2O2, which is essential to thyroid hormone synthesis, involves two NADPH oxidases: dual oxidases 1 and 2 (DuOx1 and DuOx2). A functional study with human DuOx genes and their 5'-flanking regions showed that DuOx1 and -2 promoters are different from thyroid-specific gene promoters. Furthermore, their transcriptional activities are not restricted to thyroid cells. While regulation of Tg (thyroglobulin) and TPO (thyroperoxidase) expression have been extensively studied, DuOx2 promoter regulation by hormones and transcriptional factors need to be more explored. Herein we investigated the role of TSH, insulin and insulin-like growth factor 1 (IGF-1), as well as the cAMP effect on DuOx2 promoter (ptx41) activity in transfected rat thyroid cell lines (PCCL3). We also assessed DuOx2 promoter activity in the presence of transcriptional factors crucial to thyroid development such as TTF-1 (thyroid transcription factor 1), PAX8, CREB, DREAM, Nkx2.5 and the coactivator TAZ in HeLa and HEK 293T-transfected cells. Our results show that TSH and forskolin, which increase cAMP in thyroid cells, stimulated DuOx2 promoter activity. IGF-1 led to pronounced stimulation, while insulin induction was not statistically different from DuOx2 promoter basal activity. All transcriptional factors selected for this work and coactivator TAZ, except DREAM, stimulated DuOx2 promoter activity. Moreover, Nkx2.5 and TAZ synergistically increased DuOx2 promoter activity. In conclusion, we show that DuOx2 expression is regulated by hormones and transcription factors involved in thyroid organogenesis and carcinogenesis, reinforcing the importance of the control of H2O2 generation in the thyroid.
Collapse
Affiliation(s)
- L.C. Cardoso-Weide
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal Fluminense (UFF), Niterói, Brazil
- *L.C. Cardoso-Weide, Departamento de Patologia, 4° andar, sala 4, Faculdade de Medicina, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense (UFF), Rua Marques do Paraná, 303, Niterói, RJ 24033-900 (Brazil), E-Mail
| | - R.C. Cardoso-Penha
- Laboratório de Fisiologia Endócrina Doris Rosenthal, IBCCF, Rio de Janeiro, Brazil
| | - M.W. Costa
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Vic., Australia
| | - A.C.F. Ferreira
- NUMPEX, Polo de Xerém, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - D.P. Carvalho
- Laboratório de Fisiologia Endócrina Doris Rosenthal, IBCCF, Rio de Janeiro, Brazil
| | - P.S. Santisteban
- Instituto de Investigaciones Biomédicas Alberto Sols, Spanish Council of Research-Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
43
|
Fernández LP, López-Márquez A, Santisteban P. Thyroid transcription factors in development, differentiation and disease. Nat Rev Endocrinol 2015; 11:29-42. [PMID: 25350068 DOI: 10.1038/nrendo.2014.186] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Identification of the thyroid transcription factors (TTFs), NKX2-1, FOXE1, PAX8 and HHEX, has considerably advanced our understanding of thyroid development, congenital thyroid disorders and thyroid cancer. The TTFs are fundamental to proper formation of the thyroid gland and for maintaining the functional differentiated state of the adult thyroid; however, they are not individually required for precursor cell commitment to a thyroid fate. Although knowledge of the mechanisms involved in thyroid development has increased, the full complement of genes involved in thyroid gland specification and the signals that trigger expression of the genes that encode the TTFs remain unknown. The mechanisms involved in thyroid organogenesis and differentiation have provided clues to identifying the genes that are involved in human congenital thyroid disorders and thyroid cancer. Mutations in the genes that encode the TTFs, as well as polymorphisms and epigenetic modifications, have been associated with thyroid pathologies. Here, we summarize the roles of the TTFs in thyroid development and the mechanisms by which they regulate expression of the genes involved in thyroid differentiation. We also address the implications of mutations in TTFs in thyroid diseases and in diseases not related to the thyroid gland.
Collapse
Affiliation(s)
- Lara P Fernández
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas, and Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid 28029, Spain
| | - Arístides López-Márquez
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas, and Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid 28029, Spain
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas, and Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid 28029, Spain
| |
Collapse
|
44
|
He H, Li W, Liyanarachchi S, Jendrzejewski J, Srinivas M, Davuluri RV, Nagy R, de la Chapelle A. Genetic predisposition to papillary thyroid carcinoma: involvement of FOXE1, TSHR, and a novel lincRNA gene, PTCSC2. J Clin Endocrinol Metab 2015; 100:E164-72. [PMID: 25303483 PMCID: PMC4283026 DOI: 10.1210/jc.2014-2147] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
CONTEXT By genome-wide association studies, the risk allele [A] of SNP rs965513 predisposes strongly to papillary thyroid carcinoma (PTC). It is located in a gene-poor region of 9q22, some 60 kb from the FOXE1 gene. The underlying mechanisms remain to be discovered. OBJECTIVE Our objective was to identify novel transcripts in the 9q22 locus and correlate gene expression levels with the genotypes of rs965513. DESIGN We performed 3' and 5' rapid amplification of cDNA ends and RT-PCR to detect novel transcripts. One novel transcript was forcibly expressed in a cell line followed by gene expression array analysis. We genotyped rs965513 from PTC patients and measured gene expression levels by real-time RT-PCR in unaffected thyroid tissue and matched tumor. SETTING This was a laboratory-based study using cells from clinical tissue samples and a cancer cell line. MAIN OUTCOME MEASURES We detected previously uncharacterized transcripts and evaluated the gene expression levels and the correlation with the risk allele of rs965513, age, gender, chronic lymphocyte thyroiditis (CLT), and TSH levels. RESULTS We found a novel long intergenic noncoding RNA gene and named it papillary thyroid cancer susceptibility candidate 2 (PTCSC2). Transcripts of PTCSC2 are down-regulated in PTC tumors. The risk allele [A] of rs965513 was significantly associated with low expression of unspliced PTCSC2, FOXE1, and TSHR in unaffected thyroid tissue. We also observed a significant association of age and CLT with PTCSC2 unspliced transcript levels. The correlation between the rs965513 genotype and the PTCSC2 unspliced transcript levels remained significant after adjusting for age, gender, and CLT. Forced expression of PTCSC2 in the BCPAP cell line affected the expression of a subset of noncoding and coding transcripts with enrichment of genes functionally involved in cell cycle and cancer. CONCLUSIONS Our data suggest a role for PTCSC2, FOXE1, and TSHR in the predisposition to PTC.
Collapse
Affiliation(s)
- Huiling He
- Human Cancer Genetics Program and Department of Molecular Virology, Immunology, and Medical Genetics (H.H., W.L., S.L., J.J., M.S., R.N., A.d.l.C), and Department of Internal Medicine (R.N.), Ohio State University Comprehensive Cancer Center, the Ohio State University, Columbus, Ohio 43210; and Division of Health and Biomedical Informatics, Department of Preventive Medicine, Robert H. Lurie Comprehensive Cancer Center (R.V.D.), Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Association of three SNPs in the PARP-1 gene with Hashimoto's thyroiditis. Hum Genome Var 2014; 1:14016. [PMID: 27081507 PMCID: PMC4785522 DOI: 10.1038/hgv.2014.16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 08/12/2014] [Accepted: 08/23/2014] [Indexed: 12/20/2022] Open
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) has a vital role in the progression of the inflammatory response, and its inhibition confers protection in various models of inflammatory disorders. Therefore, we investigated the effect of promoter and exon variations of the PARP-1 gene on the risk for the inflammatory disease Hashimoto's thyroiditis (HT). This case-control association study was comprised of 141 HT patients and 150 controls from a group of women in a Turkish population. Two polymorphisms in the promoter region of the PARP-1 gene, rs2793378 and rs7527192, were analyzed using the PCR-RFLP method. In addition, single nucleotide polymorphism (SNP) rs1136410, which is located at codon 762, was analyzed using bidirectional sequencing. The combined genotype and haplotype analyses of these polymorphisms were performed using SPSS 18 and Haploview 4.2. The results were statistically analyzed by calculating the odds ratios and 95% confidence interval using Pearson's χ (2)-test and Fisher's exact test (two-sided). Although we had a number of significant results, the associations became nonsignificant following a Bonferroni correction for multiple comparisons. Nonetheless, a protective factor against HT was still observed for the heterozygous genotype (TC) of SNP rs1136410 (P=0.001), even following Bonferroni correction, and according to the rs2793378-rs7527192 combined analysis, the occurrence of the TT/GA combined genotype was significantly higher in the controls (P=0.007). These results prove that the heterozygosity of SNP rs1136410 provides a protective effect against HT disease in a group of women in a Turkish population.
Collapse
|
46
|
Lakshmanan A, Scarberry D, Shen DH, Jhiang SM. Modulation of sodium iodide symporter in thyroid cancer. Discov Oncol 2014; 5:363-73. [PMID: 25234361 DOI: 10.1007/s12672-014-0203-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/05/2014] [Indexed: 11/29/2022] Open
Abstract
Radioactive iodine (RAI) is a key therapeutic modality for thyroid cancer. Loss of RAI uptake in thyroid cancer inversely correlates with patient's survival. In this review, we focus on the challenges encountered in delivering sufficient doses of I-131 to eradicate metastatic lesions without increasing the risk of unwanted side effects. Sodium iodide symporter (NIS) mediates iodide influx, and NIS expression and function can be selectively enhanced in thyroid cells by thyroid-stimulating hormone. We summarize our current knowledge of NIS modulation in normal and cancer thyroid cells, and we propose that several reagents evaluated in clinical trials for other diseases can be used to restore or further increase RAI accumulation in thyroid cancer. Once validated in preclinical mouse models and clinical trials, these reagents, mostly small-molecule inhibitors, can be readily translated into clinical practice. We review available genetically engineered mouse models of thyroid cancer in terms of their tumor development and progression as well as their thyroid function. These mice will not only provide important insights into the mechanisms underlying the loss of RAI uptake in thyroid tumors but will also serve as preclinical animal models to evaluate the efficacy of candidate reagents to selectively increase RAI uptake in thyroid cancers. Taken together, we anticipate that the optimal use of RAI in the clinical management of thyroid cancer is yet to come in the near future.
Collapse
Affiliation(s)
- Aparna Lakshmanan
- Department of Physiology and Cell Biology, The Ohio State University, 1645 Neil Avenue, 304 Hamilton Hall, Columbus, OH, 43210, USA
| | | | | | | |
Collapse
|
47
|
Martinez VD, Vucic EA, Pikor LA, Thu KL, Hubaux R, Lam WL. Frequent concerted genetic mechanisms disrupt multiple components of the NRF2 inhibitor KEAP1/CUL3/RBX1 E3-ubiquitin ligase complex in thyroid cancer. Mol Cancer 2013; 12:124. [PMID: 24138990 PMCID: PMC4016213 DOI: 10.1186/1476-4598-12-124] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 10/02/2013] [Indexed: 01/12/2023] Open
Abstract
Background Reactive oxygen species contribute to normal thyroid function. The NRF2 oxidative response pathway is frequently and constitutively activated in multiple tumor types, including papillary thyroid carcinoma (PTC). Genetic mechanisms underlying NRF2 pathway activation in PTC are not fully understood. Thus, we aimed to determine whether inactivating patterns of DNA-level alterations affect genes encoding for individual NRF2 inhibitor complex components (CUL3/KEAP1/RBX1) occur in PTC. Findings Combined patterns of epi/genetic alterations for KEAP1/CUL3/RBX1 E3 ubiquitin-ligase complex components were simultaneously interrogated for a panel of 310 PTC cases and 40 adjacent non-malignant tissues. Data were obtained from The Cancer Genome Atlas project. Enrichment of NRF2 pathway activation was assessed by gene-set enrichment analysis using transcriptome data. Our analyses revealed that PTC sustain a strikingly high frequency (80.6%) of disruption to multiple component genes of the NRF2 inhibitor complex. Hypermethylation is the predominant inactivating mechanism primarily affecting KEAP1 (70.6%) and CUL3 (20%), while copy number loss mostly affects RBX1 (16.8%). Concordantly, NRF2-associated gene expression signatures are positively and significantly enriched in PTC. Conclusions The KEAP1/CUL3/RBX1 E3-ubiquitin ligase complex is almost ubiquitously affected by multiple DNA-level mechanisms and downstream NRF2 pathway targets are activated in PTC. Given the importance of this pathway to normal thyroid function as well as to cancer; targeted inhibition of NRF2 regulators may impact strategies for therapeutic intervention involving this pathway.
Collapse
Affiliation(s)
- Victor D Martinez
- BC Cancer Research Centre, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z1L3, Canada.
| | | | | | | | | | | |
Collapse
|