1
|
Kalampalikis A, Panagiotopoulos M, Ivanidou S, Mavrogianni D, Drakakis P, Michala L. The association of the FOXE1 polyalanine tract length with the occurrence of premature ovarian insufficiency in the Greek population: A pilot, case-control study. Turk J Obstet Gynecol 2025; 22:41-45. [PMID: 40062677 PMCID: PMC11894772 DOI: 10.4274/tjod.galenos.2025.94024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/19/2025] [Indexed: 03/14/2025] Open
Abstract
Objective To investigate the relationship between the FOXE1 gene polyalanine tract length and premature ovarian insufficiency (POI) in the Greek population. Materials and Methods Peripheral blood was collected from 28 women with POI and 29 healthy controls. DNA was extracted and the gene was amplified using the polymerase chain reaction (PCR) technique. The PCR product was sequenced and the number of alanine tracts and the genotypes was recorded. Statistical analysis examined differences in allele and genotype frequencies between the groups. Results The patients' group mean age was 31.68 years with a mean age of POI diagnosis of 25.18 years. Five alleles (8, 12, 14, 16, 17 comprising alanine residues) and seven genotypes (14/14, 14/16, 16/16, 14/17, 16/17, 8/16, 12/14) were identified. The 8-alanine allele was exclusive to patients, while the 12-alanine allele appeared only in controls. The most common genotype in the study group was 14/16 (64.29%), whereas the most common genotype in the control group was 14/14 (41.4%). No differences of statistical significance were observed in the prevalences of the allele with 14 (p=0.590) and 16 (p=0.594) residues or the genotype prevalences between the two groups (p=0.066). Conclusion Our preliminary findings suggest no correlation between FOXE1 polyalanine tract length and POI, but given the study's small sample size, they should be interpreted with caution. Further research is deemed necessary.
Collapse
Affiliation(s)
- Andreas Kalampalikis
- National and Kapodistrian University of Athens Faculty of Medicine, Department of Obstetrics and Gynecology, Athens, Greece
| | - Michail Panagiotopoulos
- National and Kapodistrian University of Athens Faculty of Medicine, Department of Obstetrics and Gynecology, Athens, Greece
| | - Sophia Ivanidou
- National and Kapodistrian University of Athens Faculty of Medicine, Department of Obstetrics and Gynecology, Athens, Greece
| | - Depy Mavrogianni
- National and Kapodistrian University of Athens Faculty of Medicine, Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Athens, Greece
| | - Peter Drakakis
- National and Kapodistrian University of Athens Faculty of Medicine, Department of Obstetrics and Gynecology, Athens, Greece
| | - Lina Michala
- National and Kapodistrian University of Athens Faculty of Medicine, Department of Obstetrics and Gynecology, Athens, Greece
| |
Collapse
|
2
|
贾 思, 张 思, 尤 玥, 孙 嘉, 段 世, 石 冰, 贾 仲. Association analysis between forkhead box E1 gene and non-syndromic cleft lip with or without cleft palate in Han Chinese population. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2025; 43:28-36. [PMID: 39840623 PMCID: PMC11917515 DOI: 10.7518/hxkq.2024.2024110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/23/2024] [Indexed: 01/23/2025]
Abstract
OBJECTIVES This study aims to explore the association between single nucleotide polymorphisms (SNPs) loci near the haplotype region hg19 chr9:100560865-100660865 of the forkhead box E1 (FOXE1) gene and the occurrence of non-syndromic cleft lip with or without cleft palate (NSCL/P) in western Han Chinese population. METHODS In the first stage, our study recruited 159 NSCL/P patients and performed targeted region sequencing to screen SNPs loci near the haplotype region of the FOXE1 gene associated with NSCL/P. In the second stage, we selected 21 common SNPs and re-enrolled 1 000 non-syndromic cleft lip only (NSCLO) patients, 1 000 non-syndromic cleft palate only (NSCPO) patients, and 1 000 normal controls to verify the association. PLINK software was used to perform Hardy-Weinberg equilibrium (HWE) test. Association analysis for common variants, gene burden analysis for rare mutations, and function prediction of SNPs with non-synonymous mutations were performed using Mutation Taster and other software programs. RESULTS In the first stage, 126 variants, including 76 single nucleotide variants and 50 insertion-deletions were identified. All the included SNPs confirmed to HWE, and the results of gene burden analysis and prediction of functional harmfulness for rare variants were not statistically significant. Association analysis showed that rs13292899 of the FOXE1 gene was significantly associated with NSCL/P (P=1.85E-27) and was also correlated with NSCLO (P=6.41E-23) and non-syndromic cleft lip with cleft palate (NSCLP) (P=2.36E-15) subtypes. In the validation phase, rs79268293 (P=0.013, P=0.022), rs10983951 (P=0.009 2, P=0.007 6), rs117227387 (P=0.009 2, P=0.007 6), rs3758250 (P=0.009 2, P=0.007 6), and rs116899397 (P=0.009 2, P=0.007 6) were significantly associated with NSCLO and NSCPO; rs13292899 (P=0.008 5), rs74606599 (P=0.008 3), rs143226042 (P=0.008 3), and rs117236550 (P=0.01) were associated with the occurrence of NSCLO; and rs12343182 (P=0.008 7), rs10119760 (P=0.012), rs10113907 (P=0.012), and rs13299924 (P=0.012) were associated with the occurrence of NSCPO. CONCLUSIONS This study found a new susceptible SNP rs13292899 of the FOXE1 gene that is closely associated with NSCL/P and NSCLO subtype and 13 other SNPs associated with NSCLO or NSCPO.
Collapse
|
3
|
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
|
4
|
Diba Lahmidi M, Le Noc M, Dali O, Kernanec PY, Merret PE, Jaulin C, Smagulova F. Sex-specific transgenerational effects on murine thyroid gland imposed by ancestral exposure to neonicotinoid thiacloprid. Sci Rep 2024; 14:13047. [PMID: 38844538 PMCID: PMC11156953 DOI: 10.1038/s41598-024-63986-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Neonicotinoids, a relatively new widely used class of insecticide is used in agriculture to control insect populations. We examined the capacity of ancestral exposure to the neonicotinoid thiacloprid (thia) to induce transgenerational effects on thyroid tissue. Pregnant outbred Swiss female mice were exposed to thia at embryonic days E6.5 to E15.5 using 0, 0.6, and 6 mg/kg/day doses. Thyroid paraffin sections were prepared for morphology analysis. We apply ELISA method to measure T4 and TSH levels, RT-qPCR for gene expression analysis, ChIP-qPCR techniques for sperm histone H3K4me3 analysis, and immunofluorescence microscopy and western blots for protein detection. We observed an alteration in the morphology of thyroids in both males and females in the F3 generation. We observed an increase in T4 hormone in F1 females and a significant T4 level decrease in F3 males. T4 changes in F1 females were associated with a TSH increase. We found that the amount of Iodothyronine Deiodinase 1 (DIO1) (an enzyme converting T4 to T3) was decreased in both F1 and F3 generations in female thyroids. GNAS protein which is important for thyroid function has increased in female thyroids. Gene expression analysis showed that the expression of genes encoding thyroid gland development, chromatin, biosynthesis and transport factors were affected in the thyroid gland in both sexes in F1 and F3. The analysis of sperm histone H3K4me3 showed that H3K4me3 occupancy at the Dio1 locus has decreased while Thyroglobulin (Tg) and Matrix Metallopeptidase 2 (Mmp2) genes have increased H3K4me3 occupancy in the sperm of F3 mice. Besides, DNA methylation analysis of our previously published datasets showed that, in the sperm of F1 and F3 thia-derived mice, several genes related to thyroid function show consistent alterations. Our data suggest that ancestral exposure to thiacloprid affects thyroid function not only in exposed but also in indirectly exposed F3 generation.
Collapse
Affiliation(s)
- Mariam Diba Lahmidi
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Morgane Le Noc
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Ouzna Dali
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Pierre-Yves Kernanec
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Pierre-Etienne Merret
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Christian Jaulin
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Fatima Smagulova
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France.
- Irset-Inserm UMR 1085, 9 Avenue du Prof. Léon Bernard, 35000, Rennes, France.
| |
Collapse
|
5
|
Grasberger H, Dumitrescu AM, Liao XH, Swanson EG, Weiss RE, Srichomkwun P, Pappa T, Chen J, Yoshimura T, Hoffmann P, França MM, Tagett R, Onigata K, Costagliola S, Ranchalis J, Vollger MR, Stergachis AB, Chong JX, Bamshad MJ, Smits G, Vassart G, Refetoff S. STR mutations on chromosome 15q cause thyrotropin resistance by activating a primate-specific enhancer of MIR7-2/MIR1179. Nat Genet 2024; 56:877-888. [PMID: 38714869 PMCID: PMC11472772 DOI: 10.1038/s41588-024-01717-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 03/14/2024] [Indexed: 05/22/2024]
Abstract
Thyrotropin (TSH) is the master regulator of thyroid gland growth and function. Resistance to TSH (RTSH) describes conditions with reduced sensitivity to TSH. Dominantly inherited RTSH has been linked to a locus on chromosome 15q, but its genetic basis has remained elusive. Here we show that non-coding mutations in a (TTTG)4 short tandem repeat (STR) underlie dominantly inherited RTSH in all 82 affected participants from 12 unrelated families. The STR is contained in a primate-specific Alu retrotransposon with thyroid-specific cis-regulatory chromatin features. Fiber-seq and RNA-seq studies revealed that the mutant STR activates a thyroid-specific enhancer cluster, leading to haplotype-specific upregulation of the bicistronic MIR7-2/MIR1179 locus 35 kb downstream and overexpression of its microRNA products in the participants' thyrocytes. An imbalance in signaling pathways targeted by these micro-RNAs provides a working model for this cause of RTSH. This finding broadens our current knowledge of genetic defects altering pituitary-thyroid feedback regulation.
Collapse
Affiliation(s)
- Helmut Grasberger
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Alexandra M Dumitrescu
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, USA
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Elliott G Swanson
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Roy E Weiss
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Theodora Pappa
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Junfeng Chen
- Institute of Transformative Bio-Molecules (WPI-ITbM) and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takashi Yoshimura
- Institute of Transformative Bio-Molecules (WPI-ITbM) and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Phillip Hoffmann
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Rebecca Tagett
- Michigan Medicine BRCF Bioinformatics Core, University of Michigan, Ann Arbor, MI, USA
| | | | - Sabine Costagliola
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Jane Ranchalis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mitchell R Vollger
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Andrew B Stergachis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Jessica X Chong
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Michael J Bamshad
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Guillaume Smits
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, Brussels, Belgium
- Center of Human Genetics, Hôpital Erasme, Hôpital Universitaire de Bruxelles, and Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Gilbert Vassart
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, IL, USA.
- Committee on Genetics, The University of Chicago, Chicago, IL, USA.
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
6
|
Kang HS, Grimm SA, Liao XH, Jetten AM. GLIS3 expression in the thyroid gland in relation to TSH signaling and regulation of gene expression. Cell Mol Life Sci 2024; 81:65. [PMID: 38281222 PMCID: PMC10822819 DOI: 10.1007/s00018-024-05113-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 12/01/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024]
Abstract
Loss of GLI-Similar 3 (GLIS3) function in mice and humans causes congenital hypothyroidism (CH). In this study, we demonstrate that GLIS3 protein is first detectable at E15.5 of murine thyroid development, a time at which GLIS3 target genes, such as Slc5a5 (Nis), become expressed. This, together with observations showing that ubiquitous Glis3KO mice do not display major changes in prenatal thyroid gland morphology, indicated that CH in Glis3KO mice is due to dyshormonogenesis rather than thyroid dysgenesis. Analysis of GLIS3 in postnatal thyroid suggested a link between GLIS3 protein expression and blood TSH levels. This was supported by data showing that treatment with TSH, cAMP, or adenylyl cyclase activators or expression of constitutively active PKA enhanced GLIS3 protein stability and transcriptional activity, indicating that GLIS3 activity is regulated at least in part by TSH/TSHR-mediated activation of PKA. The TSH-dependent increase in GLIS3 transcriptional activity would be critical for the induction of GLIS3 target gene expression, including several thyroid hormone (TH) biosynthetic genes, in thyroid follicular cells of mice fed a low iodine diet (LID) when blood TSH levels are highly elevated. Like TH biosynthetic genes, the expression of cell cycle genes is suppressed in ubiquitous Glis3KO mice fed a LID; however, in thyroid-specific Glis3 knockout mice, the expression of cell cycle genes was not repressed, in contrast to TH biosynthetic genes. This indicated that the inhibition of cell cycle genes in ubiquitous Glis3KO mice is dependent on changes in gene expression in GLIS3 target tissues other than the thyroid.
Collapse
Affiliation(s)
- Hong Soon Kang
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, Research Triangle Park, NC, 27709, USA
| | - Sara A Grimm
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Anton M Jetten
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
7
|
Radak M, Ghamari N, Fallahi H. Identification of common factors among fibrosarcoma, rhabdomyosarcoma, and osteosarcoma by network analysis. Biosystems 2024; 235:105093. [PMID: 38052344 DOI: 10.1016/j.biosystems.2023.105093] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/13/2023] [Accepted: 11/23/2023] [Indexed: 12/07/2023]
Abstract
Sarcoma cancers are uncommon malignant tumors, and there are many subgroups, including fibrosarcoma (FS), which mainly affects middle-aged and older adults in deep soft tissues. Rhabdomyosarcoma (RMS), on the other hand, is the most common soft-tissue sarcoma in children and is located in the head and neck area. Osteosarcomas (OS) is the predominant form of primary bone cancer among young adults, primarily resulting from sporadically random mutations. This frequently results in the dissemination of cancer cells to the lungs, commonly known as metastasis. Mesodermal cells are the origin of sarcoma cancers. In this study, a rather radical approach has been applied. Instead of comparing homogenous cancer types, we focus on three main subtypes of sarcoma: fibrosarcoma, rhabdomyosarcoma, and osteosarcoma, and compare their gene expression with normal cell groups to identify the differentially expressed genes (DEGs). Next, by applying protein-protein interaction (PPI) network analysis, we determine the hub genes and crucial factors, such as transcription factors (TFs), affected by these types of cancer. Our findings indicate a modification in a range of pathways associated with cell cycle, extracellular matrix, and DNA repair in these three malignancies. Results showed that fibrosarcoma (FS), rhabdomyosarcoma (RMS), and osteosarcoma (OS) had 653, 1270, and 2823 differentially expressed genes (DEGs), respectively. Interestingly, there were 24 DEGs common to all three types. Network analysis showed that the fibrosarcoma network had two sub-networks identified in FS that contributed to the catabolic process of collagen via the G-protein coupled receptor signaling pathway. The rhabdomyosarcoma network included nine sub-networks associated with cell division, extracellular matrix organization, mRNA splicing via spliceosome, and others. The osteosarcoma network has 13 sub-networks, including mRNA splicing, sister chromatid cohesion, DNA repair, etc. In conclusion, the common DEGs identified in this study have been shown to play significant and multiple roles in various other cancers based on the literature review, indicating their significance.
Collapse
Affiliation(s)
- Mehran Radak
- Department of Biology, School of Sciences, Razi University, Baq-e-Abrisham, Kermanshah, 6714967346, Iran.
| | - Nakisa Ghamari
- Department of Biology, School of Sciences, Razi University, Baq-e-Abrisham, Kermanshah, 6714967346, Iran.
| | - Hossein Fallahi
- Department of Biology, School of Sciences, Razi University, Baq-e-Abrisham, Kermanshah, 6714967346, Iran.
| |
Collapse
|
8
|
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
|
9
|
Kearns NA, Lobo M, Genga RMJ, Abramowitz RG, Parsi KM, Min J, Kernfeld EM, Huey JD, Kady J, Hennessy E, Brehm MA, Ziller MJ, Maehr R. Generation and molecular characterization of human pluripotent stem cell-derived pharyngeal foregut endoderm. Dev Cell 2023; 58:1801-1818.e15. [PMID: 37751684 PMCID: PMC10637111 DOI: 10.1016/j.devcel.2023.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/15/2023] [Accepted: 08/18/2023] [Indexed: 09/28/2023]
Abstract
Approaches to study human pharyngeal foregut endoderm-a developmental intermediate that is linked to various human syndromes involving pharynx development and organogenesis of tissues such as thymus, parathyroid, and thyroid-have been hampered by scarcity of tissue access and cellular models. We present an efficient stepwise differentiation method to generate human pharyngeal foregut endoderm from pluripotent stem cells. We determine dose and temporal requirements of signaling pathway engagement for optimized differentiation and characterize the differentiation products on cellular and integrated molecular level. We present a computational classification tool, "CellMatch," and transcriptomic classification of differentiation products on an integrated mouse scRNA-seq developmental roadmap confirms cellular maturation. Integrated transcriptomic and chromatin analyses infer differentiation stage-specific gene regulatory networks. Our work provides the method and integrated multiomic resource for the investigation of disease-relevant loci and gene regulatory networks and their role in developmental defects affecting the pharyngeal endoderm and its derivatives.
Collapse
Affiliation(s)
- Nicola A Kearns
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Macrina Lobo
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ryan M J Genga
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ryan G Abramowitz
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Krishna M Parsi
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jiang Min
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Eric M Kernfeld
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jack D Huey
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jamie Kady
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Erica Hennessy
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Michael J Ziller
- Department of Psychiatry, University of Münster, Münster, Germany
| | - René Maehr
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
10
|
Grassi ES, Rurale G, de Filippis T, Gentilini D, Carbone E, Coscia F, Uraghi S, Bullock M, Clifton-Bligh RJ, Gupta AK, Persani L. The length of FOXE1 polyalanine tract in congenital hypothyroidism: Evidence for a pathogenic role from familial, molecular and cohort studies. Front Endocrinol (Lausanne) 2023; 14:1127312. [PMID: 37008944 PMCID: PMC10060985 DOI: 10.3389/fendo.2023.1127312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
INTRODUCTION FOXE1 is required for thyroid function and its homozygous mutations cause a rare syndromic form of congenital hypothyroidism (CH). FOXE1 has a polymorphic polyalanine tract whose involvement in thyroid pathology is controversial. Starting from genetic studies in a CH family, we explored the functional role and involvement of FOXE1 variations in a large CH population. METHODS We applied NGS screening to a large CH family and a cohort of 1752 individuals and validated these results by in silico modeling and in vitro experiments. RESULTS A new heterozygous FOXE1 variant segregated with 14-Alanine tract homozygosity in 5 CH siblings with athyreosis. The p.L107V variant demonstrated to significantly reduce the FOXE1 transcriptional activity. The 14-Alanine-FOXE1 displayed altered subcellular localization and significantly impaired synergy with other transcription factors, when compared with the more common 16-Alanine-FOXE1. The CH group with thyroid dysgenesis was largely and significantly enriched with the 14-Alanine-FOXE1 homozygosity. DISCUSSION We provide new evidence that disentangle the pathophysiological role of FOXE1 polyalanine tract, thereby significantly broadening the perspective on the role of FOXE1 in the complex pathogenesis of CH. FOXE1 should be therefore added to the group of polyalanine disease-associated transcription factors.
Collapse
Affiliation(s)
- Elisa Stellaria Grassi
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giuditta Rurale
- Laboratory of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
| | - Tiziana de Filippis
- Laboratory of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Bioinformatics and Statistical Genomics Unit, Milano, Italy
| | - Erika Carbone
- Laboratory of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
| | | | - Sarah Uraghi
- Department of Health Science, University of Milan, Milan, Italy
| | - Martyn Bullock
- Cancer Genetics Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Roderick J. Clifton-Bligh
- Cancer Genetics Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Endocrinology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Abhinav K. Gupta
- Department of Diabetes and Endocrine Sciences, CK Birla Hospitals, Jaipur, Rajasthan, India
| | - Luca Persani
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Laboratory of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
- *Correspondence: Luca Persani,
| |
Collapse
|
11
|
Siewert A, Reiz B, Krug C, Heggemann J, Mangold E, Dickten H, Ludwig KU. Analysis of candidate genes for cleft lip ± cleft palate using murine single-cell expression data. Front Cell Dev Biol 2023; 11:1091666. [PMID: 37169019 PMCID: PMC10165499 DOI: 10.3389/fcell.2023.1091666] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/03/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction: Cleft lip ± cleft palate (CL/P) is one of the most common birth defects. Although research has identified multiple genetic risk loci for different types of CL/P (i.e., syndromic or non-syndromic forms), determining the respective causal genes and understanding the relevant functional networks remain challenging. The recent introduction of single-cell RNA sequencing (scRNA-seq) has provided novel opportunities to study gene expression patterns at cellular resolution. The aims of our study were to: (i) aggregate available scRNA-seq data from embryonic mice and provide this as a resource for the craniofacial community; and (ii) demonstrate the value of these data in terms of the investigation of the gene expression patterns of CL/P candidate genes. Methods and Results: First, two published scRNA-seq data sets from embryonic mice were re-processed, i.e., data representing the murine time period of craniofacial development: (i) facial data from embryonic day (E) E11.5; and (ii) whole embryo data from E9.5-E13.5 from the Mouse Organogenesis Cell Atlas (MOCA). Marker gene expression analyses demonstrated that at E11.5, the facial data were a high-resolution representation of the MOCA data. Using CL/P candidate gene lists, distinct groups of genes with specific expression patterns were identified. Among others we identified that a co-expression network including Irf6, Grhl3 and Tfap2a in the periderm, while it was limited to Irf6 and Tfap2a in palatal epithelia, cells of the ectodermal surface, and basal cells at the fusion zone. The analyses also demonstrated that additional CL/P candidate genes (e.g., Tpm1, Arid3b, Ctnnd1, and Wnt3) were exclusively expressed in Irf6+ facial epithelial cells (i.e., as opposed to Irf6- epithelial cells). The MOCA data set was finally used to investigate differences in expression profiles for candidate genes underlying different types of CL/P. These analyses showed that syndromic CL/P genes (syCL/P) were expressed in significantly more cell types than non-syndromic CL/P candidate genes (nsCL/P). Discussion: The present study illustrates how scRNA-seq data can empower research on craniofacial development and disease.
Collapse
Affiliation(s)
- Anna Siewert
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | | | - Carina Krug
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Julia Heggemann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Elisabeth Mangold
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | | | - Kerstin U. Ludwig
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- *Correspondence: Kerstin U. Ludwig,
| |
Collapse
|
12
|
Zamboni M, Strimpakos G, Poggiogalle E, Donini LM, Civitareale D. Adipocyte signaling affects thyroid-specific gene expression via down-regulation of TTF-2/FOXE1. J Mol Endocrinol 2023; 70:e220129. [PMID: 36347053 DOI: 10.1530/jme-22-0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/08/2022] [Indexed: 11/09/2022]
Abstract
Obesity affects thyroid gland function. Hypothyroidism, thyroid nodules, goiter, and thyroid cancer are more frequent in patients with higher BMI values. Although these data are supported by many clinical and epidemiological studies, our knowledge is very scarce at the molecular level. In this study, we present the first experimental evidence that adipocyte signaling downregulates the expression of thyroid-specific transcription factor 2 (TTF-2/FoxE1). It plays a crucial role in thyroid development and thyroid homeostasis and it is strictly connected to thyroid cancer as well. We provide in vivo and in vitro evidence that inhibition of TTF-2/FoxE1 gene expression is mediated by adipocyte signaling.
Collapse
Affiliation(s)
- Michela Zamboni
- Institute of Biochemistry and Cell Biology, National Council of Research, Monterotondo, Rome, Italy
| | - Georgios Strimpakos
- Institute of Biochemistry and Cell Biology, National Council of Research, Monterotondo, Rome, Italy
| | - Eleonora Poggiogalle
- Department of Experimental Medicine - Medical Pathophysiology, Food Science and Endocrinology Section, Sapienza University of Rome, Rome, Italy
| | - Lorenzo M Donini
- Department of Experimental Medicine - Medical Pathophysiology, Food Science and Endocrinology Section, Sapienza University of Rome, Rome, Italy
| | - Donato Civitareale
- Institute of Biochemistry and Cell Biology, National Council of Research, Monterotondo, Rome, Italy
| |
Collapse
|
13
|
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
|
14
|
A new FOXE1 homozygous frameshift variant expands the genotypic and phenotypic spectrum of Bamforth-Lazarus syndrome. Eur J Med Genet 2022; 65:104591. [PMID: 35963604 DOI: 10.1016/j.ejmg.2022.104591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/21/2022] [Accepted: 08/07/2022] [Indexed: 11/22/2022]
Abstract
Bamforth-Lazarus syndrome is a rare autosomal recessive disease caused by biallelic loss-of-function variants in the FOXE1 gene. The condition is characterized by congenital hypothyroidism due to thyroid agenesis or thyroid hypoplasia, cleft palate, spiky hair, with or without choanal atresia, and bifid epiglottis. To date, seven pathogenic variants have been reported in the FOXE1 gene causing Bamforth-Lazarus syndrome. Here we report a novel homozygous loss-of-function variant in the FOXE1 gene NM_004473.4:c.141dupC:p.(Leu49Profs*75) leading to congenital hypothyroidism due to thyroid agenesis, scalp hair abnormalities, cleft palate, small areola, cafe-au-lait spots, mild bilateral hearing loss, skin abnormalities, and facial dysmorphism. We describe the evolving phenotype in the patient with age and review previous variants reported in FOXE1. This report further expands the clinical and molecular spectrum of Bamforth-Lazarus syndrome.
Collapse
|
15
|
Hayashi T, Eto K, Kadoya Y. Downregulation of ten-eleven translocation-2 triggers epithelial differentiation during organogenesis. Differentiation 2022; 125:45-53. [DOI: 10.1016/j.diff.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/25/2022] [Accepted: 05/02/2022] [Indexed: 11/26/2022]
|
16
|
Deficiency of Thyroid Hormone Reduces Voltage-Gated Na + Currents as Well as Expression of Na +/K +-ATPase in the Mouse Hippocampus. Int J Mol Sci 2022; 23:ijms23084133. [PMID: 35456949 PMCID: PMC9031557 DOI: 10.3390/ijms23084133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Mice lacking functional thyroid follicular cells, Pax8−/− mice, die early postnatally, making them suitable models for extreme hypothyroidism. We have previously obtained evidence in postnatal rat neurons, that a down-regulation of Na+-current density could explain the reduced excitability of the nervous system in hypothyroidism. If such a mechanism underlies the development of coma and death in severe hypothyroidism, Pax8−/− mice should show deficits in the expression of Na+ currents and potentially also in the expression of Na+/K+-ATPases, which are necessary to maintain low intracellular Na+ levels. We thus compared Na+ current densities in postnatal mice using the patch-clamp technique in the whole-cell configuration as well as the expression of three alpha and two beta-subunits of the Na+/K+-ATPase in wild type versus Pax8−/− mice. Whereas the Na+ current density in hippocampal neurons from wild type mice was upregulated within the first postnatal week, the Na+ current density remained at a very low level in hippocampal neurons from Pax8−/− mice. Pax8−/− mice also showed significantly decreased protein expression levels of the catalytic α1 and α3 subunits of the Na+/K+-ATPase as well as decreased levels of the β2 isoform, with no changes in the α2 and β1 subunits.
Collapse
|
17
|
Sox9 is involved in the thyroid differentiation program and is regulated by crosstalk between TSH, TGFβ and thyroid transcription factors. Sci Rep 2022; 12:2144. [PMID: 35140269 PMCID: PMC8828901 DOI: 10.1038/s41598-022-06004-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/21/2022] [Indexed: 11/09/2022] Open
Abstract
While the signaling pathways and transcription factors involved in the differentiation of thyroid follicular cells, both in embryonic and adult life, are increasingly well understood, the underlying mechanisms and potential crosstalk between the thyroid transcription factors Nkx2.1, Foxe1 and Pax8 and inductive signals remain unclear. Here, we focused on the transcription factor Sox9, which is expressed in Nkx2.1-positive embryonic thyroid precursor cells and is maintained from embryonic development to adulthood, but its function and control are unknown. We show that two of the main signals regulating thyroid differentiation, TSH and TGFβ, modulate Sox9 expression. Specifically, TSH stimulates the cAMP/PKA pathway to transcriptionally upregulate Sox9 mRNA and protein expression, a mechanism that is mediated by the binding of CREB to a CRE site within the Sox9 promoter. Contrastingly, TGFβ signals through Smad proteins to inhibit TSH-induced Sox9 transcription. Our data also reveal that Sox9 transcription is regulated by the thyroid transcription factors, particularly Pax8. Interestingly, Sox9 significantly increased the transcriptional activation of Pax8 and Foxe1 promoters and, consequently, their expression, but had no effect on Nkx2.1. Our study establishes the involvement of Sox9 in thyroid follicular cell differentiation and broadens our understanding of transcription factor regulation of thyroid function.
Collapse
|
18
|
Chan M, Yuan H, Soifer I, Maile TM, Wang RY, Ireland A, O'Brien JJ, Goudeau J, Chan LJ, Vijay T, Freund A, Kenyon C, Bennett BD, McAllister FE, Kelley DR, Roy M, Cohen RL, Levinson AD, Botstein D, Hendrickson DG. Novel insights from a multiomics dissection of the hayflick limit. eLife 2022; 11:70283. [PMID: 35119359 PMCID: PMC8933007 DOI: 10.7554/elife.70283] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 01/31/2022] [Indexed: 01/10/2023] Open
Abstract
The process wherein dividing cells exhaust proliferative capacity and enter into replicative senescence has become a prominent model for cellular aging in vitro. Despite decades of study, this cellular state is not fully understood in culture and even much less so during aging. Here, we revisit Leonard Hayflick’s original observation of replicative senescence in WI-38 human lung fibroblasts equipped with a battery of modern techniques including RNA-seq, single-cell RNA-seq, proteomics, metabolomics, and ATAC-seq. We find evidence that the transition to a senescent state manifests early, increases gradually, and corresponds to a concomitant global increase in DNA accessibility in nucleolar and lamin associated domains. Furthermore, we demonstrate that senescent WI-38 cells acquire a striking resemblance to myofibroblasts in a process similar to the epithelial to mesenchymal transition (EMT) that is regulated by t YAP1/TEAD1 and TGF-β2. Lastly, we show that verteporfin inhibition of YAP1/TEAD1 activity in aged WI-38 cells robustly attenuates this gene expression program.
Collapse
Affiliation(s)
- Michelle Chan
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Han Yuan
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Ilya Soifer
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Tobias M Maile
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Rebecca Y Wang
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Andrea Ireland
- Calico Life Sciences, LLC, South San Francisco, United States
| | | | - Jérôme Goudeau
- Calico Life Sciences LLC, South San Francisco, United States
| | - Leanne Jg Chan
- Calico Life Sciences LLC, South San Francisco, United States
| | - Twaritha Vijay
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Adam Freund
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Cynthia Kenyon
- Calico Life Sciences LLC, South San Francisco, United States
| | | | | | - David R Kelley
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Margaret Roy
- Calico Life Sciences LLC, South San Francisco, United States
| | - Robert L Cohen
- Calico Life Sciences, LLC, South San Francisco, United States
| | | | - David Botstein
- Calico Life Sciences, LLC, South San Francisco, United States
| | | |
Collapse
|
19
|
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
|
20
|
Kostopoulou E, Miliordos K, Spiliotis B. Genetics of primary congenital hypothyroidism-a review. Hormones (Athens) 2021; 20:225-236. [PMID: 33400193 DOI: 10.1007/s42000-020-00267-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Congenital primary hypothyroidism (CH) is a state of inadequate thyroid hormone production detected at birth, caused either by absent, underdeveloped or ectopic thyroid gland (dysgenesis), or by defected thyroid hormone biosynthesis (dyshormonogenesis). A genetic component has been identified in many cases of CH. This review summarizes the clinical and biochemical features of the genetic causes of primary CH. METHODS A literature review was conducted of gene defects causing congenital hypothyroidism. RESULTS Mutations in five genes have predominantly been implicated in thyroid dysgenesis (TSHR, FOXE1, NKX2-1, PAX8, and NKX2-5), the primary cause of CH (85%), and mutations in seven genes in thyroid dyshormonogenesis (SLC5A5, TPO, DUOX2, DUOXA2, SLC6A4, Tg, and DEHAL1). These genes encode for proteins that regulate genes expressed during the differentiation of the thyroid, such as TPO and Tg genes, or genes that regulate iodide organification, thyroglobulin synthesis, iodide transport, and iodotyrosine deiodination. Besides thyroid dysgenesis and dyshormonogenesis, additional causes of congenital hypothyroidism, such as iodothyronine transporter defects and resistance to thyroid hormones, have also been associated with genetic mutations. CONCLUSION The identification of the underlying genetic defects of CH is important for genetic counseling of families with an affected member, for identifying additional clinical characteristics or the risk for thyroid neoplasia and for diagnostic and management purposes.
Collapse
Affiliation(s)
- Eirini Kostopoulou
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics|, University of Patras School of Medicine, Patras, Greece.
| | - Konstantinos Miliordos
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics|, University of Patras School of Medicine, Patras, Greece
| | - Bessie Spiliotis
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics|, University of Patras School of Medicine, Patras, Greece
| |
Collapse
|
21
|
Romitti M, Eski SE, Fonseca BF, Gillotay P, Singh SP, Costagliola S. Single-Cell Trajectory Inference Guided Enhancement of Thyroid Maturation In Vitro Using TGF-Beta Inhibition. Front Endocrinol (Lausanne) 2021; 12:657195. [PMID: 34135860 PMCID: PMC8202408 DOI: 10.3389/fendo.2021.657195] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/10/2021] [Indexed: 12/30/2022] Open
Abstract
The thyroid gland regulates metabolism and growth via secretion of thyroid hormones by thyroid follicular cells (TFCs). Loss of TFCs, by cellular dysfunction, autoimmune destruction or surgical resection, underlies hypothyroidism. Recovery of thyroid hormone levels by transplantation of mature TFCs derived from stem cells in vitro holds great therapeutic promise. However, the utilization of in vitro derived tissue for regenerative medicine is restricted by the efficiency of differentiation protocols to generate mature organoids. Here, to improve the differentiation efficiency for thyroid organoids, we utilized single-cell RNA-Seq to chart the molecular steps undertaken by individual cells during the in vitro transformation of mouse embryonic stem cells to TFCs. Our single-cell atlas of mouse organoid systematically and comprehensively identifies, for the first time, the cell types generated during production of thyroid organoids. Using pseudotime analysis, we identify TGF-beta as a negative regulator of thyroid maturation in vitro. Using pharmacological inhibition of TGF-beta pathway, we improve the level of thyroid maturation, in particular the induction of Nis expression. This in turn, leads to an enhancement of iodide organification in vitro, suggesting functional improvement of the thyroid organoid. Our study highlights the potential of single-cell molecular characterization in understanding and improving thyroid maturation and paves the way for identification of therapeutic targets against thyroid disorders.
Collapse
Affiliation(s)
- Mírian Romitti
- *Correspondence: Mírian Romitti, ; Sumeet Pal Singh, ; Sabine Costagliola,
| | | | | | | | - Sumeet Pal Singh
- *Correspondence: Mírian Romitti, ; Sumeet Pal Singh, ; Sabine Costagliola,
| | - Sabine Costagliola
- *Correspondence: Mírian Romitti, ; Sumeet Pal Singh, ; Sabine Costagliola,
| |
Collapse
|
22
|
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: 20] [Impact Index Per Article: 5.0] [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
|
23
|
Iwahashi-Odano M, Fujisawa Y, Ogata T, Nakashima S, Muramatsu M, Narumi S. Identification and functional characterization of a novel PAX8 mutation (p.His39Pro) causing familial thyroid hypoplasia. Clin Pediatr Endocrinol 2020; 29:173-178. [PMID: 33088016 PMCID: PMC7534521 DOI: 10.1297/cpe.29.173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/04/2020] [Indexed: 11/24/2022] Open
Abstract
Mutations in PAX8, the gene for a thyroid-specific transcription factor,
causes congenital hypothyroidism (CH) with autosomal dominant inheritance. All previously
detected PAX8 mutations except one are located in the DNA-binding paired
domain. The proband, a 1-yr-old boy, was diagnosed with CH in the frame
of newborn screening. He had high serum TSH level (180 mU/L) and low serum free
T4 level (0.4 ng/dL). Ultrasonography revealed that the proband had thyroid
hypoplasia. Importantly, he had a family history of CH, i.e., his mother
also had CH and hypoplasia. Next generation sequencing-based mutation screening revealed a
novel heterozygous PAX8 mutation (c.116A>C, p.His39Pro) that was
transmitted to the proband from the mother. Expression experiments with HeLa cells
confirmed that His39Pro-PAX8 exhibited defective transactivation of the
TG promoter–luciferase reporter. In conclusion, we identified and
described a novel loss-of-function PAX8 mutation in a family with thyroid
hypoplasia. Patients with dominantly inherited CH and no extrathyroidal abnormalities
could have PAX8 mutations.
Collapse
Affiliation(s)
- Megumi Iwahashi-Odano
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yasuko Fujisawa
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shinichi Nakashima
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mayumi Muramatsu
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
24
|
Xiao WL, Jia KN, Yu G, Zhao N. Association between forkhead box E1 polymorphisms and risk of non-syndromic cleft lip with or without cleft palate: A meta-analysis. Orthod Craniofac Res 2020; 23:151-159. [PMID: 31944555 DOI: 10.1111/ocr.12366] [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: 09/26/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVE The purpose of the present work was to investigate the association between forkhead box E1 (FOXE1) and the risk of non-syndromic cleft lip with or without cleft palate (NSCL/P). MATERIALS AND METHODS Relevant studies were searched in several professional databases up to 31 July 2019. The pooled odds ratios (ORs) and 95% confidence intervals (95% CIs) were calculated using a fixed-effect model or a random-effect model to analyse the relationship between FOXE1 polymorphisms and NSCL/P. RESULTS A total of four single nucleotide polymorphisms (SNPs), including rs3758249, rs4460498, rs1443434 and rs10217225, were analysed. The overall findings showed that FOXE1 rs4460498 was statistically associated with NSCL/P (including cleft lip with or without cleft palate (CL/P) and cleft palate only (CPO)). Genotypes CC and CT of rs4460498 were significantly more closely correlated with NSCL/P (including CL/P and CPO) than genotype TT (NSCL/P: TT vs CC, OR = 0.630, P = .000; TT vs TC + CC, OR = 0.775, P = .020; CL/P: TT vs CC, OR = 0.664, P = .000; TT vs TC + CC, OR = 0.738, P = .006. CPO: TT vs CC, OR = 0.761, P = .027; TT vs TC + CC, OR = 0.792, P = .045). For rs10217225, only the TT genotype might have contributed to the elevated risk of CL/P (TT vs CC OR = 2.236, P = .000). The other FOXE1 polymorphisms were not associated with NSCLP, CL/P or CPO. CONCLUSION The meta-analysis provided confirmation that the polymorphism of FOXE1 rs10217225 was correlated with an increased risk of CL/P, and the polymorphism of FOXE1 rs4460498 was a protective factor for NSCL/P, including CLP and CPO.
Collapse
Affiliation(s)
- Wen-Lin Xiao
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kai-Ning Jia
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guo Yu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ning Zhao
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Jang D, Morgan SJ, Klubo-Gwiezdzinska J, Banga JP, Neumann S, Gershengorn MC. Thyrotropin, but Not Thyroid-Stimulating Antibodies, Induces Biphasic Regulation of Gene Expression in Human Thyrocytes. Thyroid 2020; 30:270-276. [PMID: 31805824 PMCID: PMC7047096 DOI: 10.1089/thy.2019.0418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background: Thyrotropin (TSH) and thyroid-stimulating antibodies (TSAbs) activate TSH receptor (TSHR) signaling by binding to its extracellular domain. TSHR signaling has been studied extensively in animal thyrocytes and in engineered cell lines, and differences in signaling have been observed in different cell systems. We, therefore, decided to characterize and compare TSHR signaling mediated by TSH and monoclonal TSAbs in human thyrocytes in primary culture. Methods: We used quantitative reverse transcription-polymerase chain reaction to measure mRNA levels of thyroid-specific genes thyroglobulin (TG), thyroperoxidase (TPO), iodothyronine deiodinase type 2 (DIO2), sodium-iodide symporter (NIS), and TSHR after stimulation by TSH or two monoclonal TSAbs, KSAb1 and M22. We also compared secreted TG protein after TSHR activation by TSH and TSAbs using an enzyme-linked immunosorbent assay. TSHR cell surface expression was determined using fluorescence activated cell sorting (FACS). Results: We found that TSH at low doses increases and at high doses (>1 mU/mL) decreases levels of gene expression for TSHR, TG, TPO, NIS, and DIO2. The biphasic effect of TSH on signaling was not caused by downregulation of cell surface TSHRs. This bell-shaped biphasic dose-response curve has been termed an inverted U-shaped dose-response curve (IUDRC). An IUDRC was also found for TSH-induced regulation of TG secretion. In contrast, KSAb1- and M22-induced regulation of TSHR, TG, TPO, NIS, and DIO2 gene expression, and secreted TG followed a monotonic dose-response curve that plateaus at high doses of activating antibody. Conclusions: Our data demonstrate that the physiological activation of TSHRs by TSH in primary cultures of human thyrocytes is characterized by a regulatory mechanism that may inhibit thyrocyte overstimulation. In contrast, TSAbs do not exhibit biphasic regulation. Although KSAb1 and M22 may not be representative of all TSAbs found in patients with Graves' disease, we suggest that persistent robust stimulation of TSHRs by TSAbs, unrelieved by a decrease at high TSAb levels, fosters chronic stimulation of thyrocytes in Graves' hyperthyroidism.
Collapse
Affiliation(s)
- Daesong Jang
- Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, Maryland
| | - Sarah J. Morgan
- Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, Maryland
| | - Joanna Klubo-Gwiezdzinska
- Metabolic Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - J. Paul Banga
- Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Susanne Neumann
- Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, Maryland
| | - Marvin C. Gershengorn
- Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
28
|
Mio C, Grani G, Durante C, Damante G. Molecular defects in thyroid dysgenesis. Clin Genet 2019; 97:222-231. [PMID: 31432505 DOI: 10.1111/cge.13627] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 12/12/2022]
Abstract
Congenital hypothyroidism (CH) is a neonatal endocrine disorder that might occur as itself or be associated to congenital extra-thyroidal defects. About 85% of affected subjects experience thyroid dysgenesis (TD), characterized by defect in thyroid gland development. In vivo experiments on null mice paved the way for the identification of genes involved thyroid morphogenesis and development, whose mutation has been strongly associated to TD. Most of them are thyroid-specific transcription factors expressed during early thyroid development. Despite the arduous effort in unraveling the genetics of TD in animal models, up to now these data have been discontinuously confirmed in humans and only 5% of TD have associated with known null mice-related mutations (mainly PAX8 and TSHR). Notwithstanding, the advance in genetic testing represented by the next-generation sequencing (NGS) approach is steadily increasing the list of genes whose highly penetrant mutation predisposes to TD. In this review we intend to outline the molecular bases of TD, summarizing the current knowledge on thyroid development in both mice and humans and delineating the genetic features of its monogenetic forms. We will also highlight current strategies to enhance the insight into the non-Mendelian mechanisms of abnormal thyroid development.
Collapse
Affiliation(s)
- Catia Mio
- Department of Medicine, University of Udine, Udine, Italy
| | - Giorgio Grani
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Cosimo Durante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Damante
- Department of Medicine, University of Udine, Udine, Italy.,Institute of Medical Genetics, Academic Hospital "Azienda Sanitaria Universitaria Integrata di Udine", Udine, Italy
| |
Collapse
|
29
|
Ding Z, Ke R, Zhang Y, Fan Y, Fan J. FOXE1 inhibits cell proliferation, migration and invasion of papillary thyroid cancer by regulating PDGFA. Mol Cell Endocrinol 2019; 493:110420. [PMID: 31129275 DOI: 10.1016/j.mce.2019.03.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/30/2019] [Accepted: 03/31/2019] [Indexed: 01/14/2023]
Abstract
PURPOSE Forkhead box E1 (FOXE1) plays an important role in the development, proliferation and differentiation of thyroid cells. However, the biological functions of FOXE1 in papillary thyroid cancer (PTC) remain unclear. MATERIALS AND METHODS In this study, the level of FOXE1 expression was examined in human PTC tissues and cells. Then, the high expression of FOXE1 was specifically silenced by RNA interference in vitro. Subsequently, FOXE1-shRNA was transfected into PTC cells (TPC-1 and K1). The effects on cell proliferation, migration and invasion were evaluated. In addition, FOXE1 targets were screened by cDNA microarray assays. The correlation between the expression of target gene platelet-derived growth factor A (PDGFA) and clinicopathological features of PTC patients was analysed. RESULTS FOXE1 is highly expressed in PTC tissues and PTC cell lines. The silencing of FOXE1 significantly promotes PTC cell proliferation, migration and invasion in vitro. The cDNA microarray analyses show that PDGFA is a critical downstream target gene of FOXE1 in PTC cells. It was also observed that PDGFA is negatively regulated by FOXE1 in PTC. The clinical data indicate that the low expression level of PDGFA is correlated with the small size of PTC. CONCLUSION Collectively, the results indicate for the first time that high expression of FOXE1 may function as a tumour suppressor in the early stage of PTC and restrain the proliferation, migration and invasion of PTC by negatively regulating PDGFA expression. Thus, FOXE1 could serve as a prognostic biomarker for PTC.
Collapse
Affiliation(s)
- Zheng Ding
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Ronghu Ke
- Department of Plastic and Reconstructive Surgery, Huashan Hospital, Fudan University School of Medicine, Shanghai, 200040, PR China
| | - Yong Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Youben Fan
- Center of Thyroid and Parathyroid, Department of General Surgery, Shanghai Jiao Tong University Affiliated the Sixth People's Hospital, Shanghai, 200233, PR China.
| | - Jianxia Fan
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China.
| |
Collapse
|
30
|
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: 9] [Impact Index Per Article: 1.5] [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
|
31
|
Iwahashi M, Narumi S. Systematic alanine scanning of PAX8 paired domain reveals functional importance of the N-subdomain. J Mol Endocrinol 2019; 62:129-135. [PMID: 30730849 DOI: 10.1530/jme-18-0207] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/07/2019] [Indexed: 12/17/2022]
Abstract
Thyroid-specific transcription factor PAX8 has an indispensable role in the thyroid gland development, which is evidenced by the facts that PAX8/Pax8 mutations cause congenital hypothyroidism in humans and mice. More than 90% of known PAX8 mutations were located in the paired domain, suggesting the central role of the domain in exerting the molecular function. Structure-function relationships of PAX8, as well as other PAX family transcription factors, have never been investigated in a systematic manner. Here, we conducted the first alanine scanning mutagenesis study, in which 132 alanine variants located in the paired domain of PAX8 were created and systematically evaluated in vitro. We found that 76 alanine variants (55%) were loss of function (LOF) variants (defined by <30% activity as compared with wild type PAX8). Importantly, the distribution of LOF variants were skewed, with more frequently observed in the N-subdomain (65% of the alanine variants in the N-subdomain) than in the C-subdomain (45%). Twelve out of 13 alanine variants in residues that have been affected in patients with congenital hypothyroidism were actually LOF, suggesting that the alanine scanning data can be used to evaluate the functional importance of mutated residues. Using our in vitro data, we tested the accuracy of seven computational algorithms for pathogenicity prediction, showing that they are sensitive but not specific to evaluate on the paired domain alanine variants. Collectively, our experiment-based data would help better understand the structure-function relationships of the paired domain, and would provide a unique resource for pathogenicity prediction of future PAX8 variants.
Collapse
Affiliation(s)
- Megumi Iwahashi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
32
|
Zhao Y, Zhong L, Yi H. A review on the mechanism of iodide metabolic dysfunction in differentiated thyroid cancer. Mol Cell Endocrinol 2019; 479:71-77. [PMID: 30287400 DOI: 10.1016/j.mce.2018.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 12/27/2022]
Abstract
The incidence of differentiated thyroid cancer (DTC) has been increasing rapidly worldwide, and the risk factors remain unclear. With the growing number of patients with DTC, the related issues have been gradually highlighted. 131Iodide (131I) is an important treatment for DTC and has the potential to reduce the risk of recurrence. 131I is also an effective treatment for distant metastases of thyroid carcinoma. However, iodide metabolism dysfunction in metastatic foci causes patients to lose the opportunity of 131I treatment. This article reviews the related mechanisms of iodide metabolism dysfunction in DTC cells and summarizes the clinical transformation progression.
Collapse
Affiliation(s)
- Yinlong Zhao
- Department of Nuclear Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, 130041, PR China.
| | - Lili Zhong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, 130041, PR China.
| | - Heqing Yi
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang, 310021, PR China.
| |
Collapse
|
33
|
Does the Polymorphism in the Length of the Polyalanine Tract of FOXE1 Gene Influence the Risk of Thyroid Dysgenesis Occurrence? J Thyroid Res 2018; 2017:2793205. [PMID: 29479488 PMCID: PMC5727785 DOI: 10.1155/2017/2793205] [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: 09/07/2017] [Accepted: 11/06/2017] [Indexed: 11/17/2022] Open
Abstract
Background. Recent data have suggested that polymorphisms in the length of the polyalanine tract (polyA) of FOXE1 gene may act as a susceptibility factor for thyroid dysgenesis. The main purpose of this study was to investigate the influence of polyA of FOXE1 gene on the risk of thyroid dysgenesis. Method. A case-control study was conducted in a sample of 90 Brazilian patients with thyroid dysgenesis and 131 controls without family history of thyroid disease. Genomic DNA was isolated from peripheral blood samples and the genotype of each individual was determined by automated sequencing. Results. More than 90% of genotypes found in the group of patients with thyroid dysgenesis and in controls subjects were represented by sizes 14 and 16 polymorphisms in the following combinations: 14/14, 14/16, and 16/16. Genotypes 14/16 and 16/16 were more frequent in the control group, while genotype 14/14 was more frequent in the group of patients with thyroid dysgenesis. There was no difference between agenesis group and control group. Genotype 14/14 when compared to genotypes 14/16 and 16/16A showed an association with thyroid dysgenesis. Conclusion. PolyA of FOXE1 gene alters the risk of thyroid dysgenesis, which may explain in part the etiology of this disease.
Collapse
|
34
|
Bullock M, Lim G, Li C, Choi IH, Kochhar S, Liddle C, Zhang L, Clifton-Bligh RJ. Thyroid transcription factor FOXE1 interacts with ETS factor ELK1 to co-regulate TERT. Oncotarget 2018; 7:85948-85962. [PMID: 27852061 PMCID: PMC5349888 DOI: 10.18632/oncotarget.13288] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 11/06/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Although FOXE1 was initially recognized for its role in thyroid organogenesis, more recently a strong association has been identified between the FOXE1 locus and thyroid cancer. The role of FOXE1 in adult thyroid, and in particular regarding cancer risk, has not been well established. We hypothesised that discovering key FOXE1 transcriptional partners would in turn identify regulatory pathways relevant to its role in oncogenesis. RESULTS In a transcription factor-binding array, ELK1 was identified to bind FOXE1. We confirmed this physical association in heterologously transfected cells by IP and mammalian two-hybrid assays. In thyroid tissue, endogenous FOXE1 was shown to bind ELK1, and using ChIP assays these factors bound thyroid-relevant gene promoters TPO and TERT in close proximity to each other. Using a combination of electromobility shift assays, TERT promoter assays and siRNA-silencing, we found that FOXE1 positively regulated TERT expression in a manner dependent upon its association with ELK1. Treating heterologously transfected thyroid cells with MEK inhibitor U0126 inhibited FOXE1-ELK1 interaction, and reduced TERT and TPO promoter activity. METHODOLOGY We investigated FOXE1 interactions within in vitro thyroid cell models and human thyroid tissue using a combination of immunoprecipitation (IP), chromatin IP (ChIP) and gene reporter assays. CONCLUSIONS FOXE1 interacts with ELK1 on thyroid relevant gene promoters, establishing a new regulatory pathway for its role in adult thyroid function. Co-regulation of TERT suggests a mechanism by which allelic variants in/near FOXE1 are associated with thyroid cancer risk.
Collapse
Affiliation(s)
- Martyn Bullock
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia
| | - Grace Lim
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia
| | - Cheng Li
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia
| | - In Ho Choi
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia
| | - Shivansh Kochhar
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia
| | - Chris Liddle
- University of Sydney, Sydney, Australia.,Storr Liver Centre, Westmead Millennium Institute for Medical Research, Westmead Hospital, Sydney, Australia
| | - Lei Zhang
- Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff, UK
| | - Roderick J Clifton-Bligh
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia.,Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
35
|
Wang F, Yan D, Ji X, Han J, Chen M, Qiao H, Zhang S. rs965513 polymorphism as a common risk marker is associated with papillary thyroid cancer. Oncotarget 2018; 7:41336-41345. [PMID: 27191655 PMCID: PMC5173063 DOI: 10.18632/oncotarget.9324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 04/16/2016] [Indexed: 02/06/2023] Open
Abstract
Papillary thyroid cancer (PTC) is the most common type of thyroid cancer. With the rapid development of genome-wide association studies (GWAS), many genome variants associated with susceptibility to PTC have been identified, including the single nucleotide polymorphism rs965513 (9q22.33) near FOXE1. To evaluate the association between rs965513 and PTC in different ethnicities and countries, we conducted a meta-analysis using relatively large-scale samples from 23 studies (N = 163,136; 20,736 cases and 142,400 controls) by searching the PubMed and Google Scholar databases. Significant heterogeneity caused by different populations among the selected studies was observed. The A allele of rs965513 polymorphism was shown to be highly associated with risk of thyroid cancer, with odds ratios of 1.58 (95% CI 1.32–1.90) in all populations, 1.65 (95% CI 1.31–2.07)) in Caucasian populations and 1.49 in Asian populations. Compared to the dominant and recessive models, we observed the highest odds ratio (OR = 2.80, 95% CI 2.12–3.69) in the homozygous model. These results revealed that the rs965513 polymorphism is a risk factor for thyroid cancer
Collapse
Affiliation(s)
- Fang Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Dehui Yan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xu Ji
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Jun Han
- Department of Endemic Disease, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150086, China
| | - Meijun Chen
- Department of Endemic Disease, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150086, China
| | - Hong Qiao
- Department of Endemic Disease, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150086, China
| | - Shaojun Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
36
|
Son HY, Hwangbo Y, Yoo SK, Im SW, Yang SD, Kwak SJ, Park MS, Kwak SH, Cho SW, Ryu JS, Kim J, Jung YS, Kim TH, Kim SJ, Lee KE, Park DJ, Cho NH, Sung J, Seo JS, Lee EK, Park YJ, Kim JI. Genome-wide association and expression quantitative trait loci studies identify multiple susceptibility loci for thyroid cancer. Nat Commun 2017; 8:15966. [PMID: 28703219 PMCID: PMC5511346 DOI: 10.1038/ncomms15966] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/16/2017] [Indexed: 01/12/2023] Open
Abstract
Thyroid cancer is the most common cancer in Korea. Several susceptibility loci of differentiated thyroid cancer (DTC) were identified by previous genome-wide association studies (GWASs) in Europeans only. Here we conducted a GWAS and a replication study in Koreans using a total of 1,085 DTC cases and 8,884 controls, and validated these results using expression quantitative trait loci (eQTL) analysis and clinical phenotypes. The most robust associations were observed in the NRG1 gene (rs6996585, P=1.08 × 10-10) and this SNP was also associated with NRG1 expression in thyroid tissues. In addition, we confirmed three previously reported loci (FOXE1, NKX2-1 and DIRC3) and identified seven novel susceptibility loci (VAV3, PCNXL2, INSR, MRSB3, FHIT, SEPT11 and SLC24A6) associated with DTC. Furthermore, we identified specific variants of DTC that have different effects according to cancer type or ethnicity. Our findings provide deeper insight into the genetic contribution to thyroid cancer in different populations.
Collapse
Affiliation(s)
- Ho-Young Son
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Yul Hwangbo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Center for Thyroid Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Seong-Keun Yoo
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Sun-Wha Im
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - San Duk Yang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea
| | - Soo-Jung Kwak
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea
| | - Min Seon Park
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Graduate Program in Genetic Counseling, Northwestern University, Chicago, Illinois 60637, USA
| | - Soo Heon Kwak
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sun Wook Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jun Sun Ryu
- Center for Thyroid Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Jeongseon Kim
- Molecular Epidemiology Branch, Division of Cancer Epidemiology and Prevention, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Yuh-Seog Jung
- Center for Thyroid Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Tae Hyun Kim
- Center for Thyroid Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Su-jin Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Kyu Eun Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Do Joon Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Nam Han Cho
- Department of Preventive Medicine Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Joohon Sung
- Department of Epidemiology and Institute of Environment and Health, School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Sun Seo
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Eun Kyung Lee
- Center for Thyroid Cancer, National Cancer Center, Goyang 10408, Republic of Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Jong-Il Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
37
|
Abstract
Congenital hypothyroidism (CH) is the most common inborn endocrine disorder and causes significant morbidity. To date, we are only aware of the molecular basis responsible for the defects in a small portion of patients with CH. A better understanding of the pathophysiology of these cases at the genetic and molecular basis provides useful information for proper counseling to patients and their families a well as for the development of better targeted therapies. This article provides a succinct outline of the pathophysiology and genetics of the known causes of thyroid dysgenesis, dyshormonogenesis, and syndrome of impaired sensitivity to thyroid hormone.
Collapse
Affiliation(s)
- Zeina C Hannoush
- Department of Medicine, University of Miami Miller School of Medicine, 1120 NW 14th Street, Suite 310F, Miami, FL 33136, USA
| | - Roy E Weiss
- Department of Medicine, University of Miami Miller School of Medicine, 1120 NW 14th Street, Suite 310F, Miami, FL 33136, USA.
| |
Collapse
|
38
|
Abstract
Developmental anomalies of the thyroid gland, defined as thyroid dysgenesis, underlie the majority of cases of congenital hypothyroidism. Thyroid dysgenesis is predominantly a sporadic disorder although a reported familial enrichment, variation of incidence by ethnicity and the monogenic defects associated mainly with athyreosis or orthotopic thyroid hypoplasia, suggest a genetic contribution. Of note, the most common developmental anomaly, thyroid ectopy, remains unexplained. Ectopy may result from multiple genetic or epigenetic variants in the germline and/or at the somatic level. This review provides a brief overview of the monogenic defects in candidate genes that have been identified so far and of the syndromes which are known to be associated with thyroid dysgenesis.
Collapse
Affiliation(s)
- Rasha Abu-Khudir
- Endocrinology Service and Research Center, Sainte-Justine Hospital and Department of Pediatrics, University of Montreal, Montreal, H3T 1C5, Quebec, Canada; Chemistry Department, Biochemistry Division, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Stéphanie Larrivée-Vanier
- Endocrinology Service and Research Center, Sainte-Justine Hospital and Department of Pediatrics, University of Montreal, Montreal, H3T 1C5, Quebec, Canada.
| | - Jonathan D Wasserman
- Division of Endocrinology, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada.
| | - Johnny Deladoëy
- Endocrinology Service and Research Center, Sainte-Justine Hospital and Department of Pediatrics, University of Montreal, Montreal, H3T 1C5, Quebec, Canada.
| |
Collapse
|
39
|
Raimundo J, Alvelos MI, Azevedo T, Martins T, Rodrigues FJ, Lemos MC. Association of FOXE1 polyalanine repeat region with thyroid cancer is dependent on tumour size. Clin Endocrinol (Oxf) 2017; 86:243-246. [PMID: 27474100 DOI: 10.1111/cen.13166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/23/2016] [Accepted: 07/27/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Polymorphisms in the thyroid transcription factor forkhead factor E1 (FOXE1) gene have been implicated in the genetic susceptibility to differentiated thyroid cancer, but little is known about their effect on tumour characteristics. The objective of this study was to determine the contribution of the FOXE1 polyalanine repeat region to the susceptibility to thyroid cancer and to its clinical characteristics. DESIGN, PATIENTS AND MEASUREMENTS A total of 500 patients with sporadic thyroid cancer (440 papillary and 60 follicular thyroid carcinoma) and 502 healthy controls were included in this case-control association study. The number of FOXE1 alanine repeats in each subject was determined by PCR and multiplex fragment analysis by capillary electrophoresis. FOXE1 genotype and allele frequencies among groups were compared by logistic regression and adjusted for sex and age at diagnosis. Data were analysed according to cancer subtype, tumour size and the presence of lymph node or distant metastasis. RESULTS FOXE1 alleles with 16 or more alanine repeats were more frequent in patients with tumour size > 1 cm compared to tumour size ≤ 1 cm (adjusted OR 1·44; 95% CI 1·05-1·88; P = 0·019). Genotypes containing at least one allele with 16 or more alanine repeats were associated with larger tumour size (adjusted OR 1·71; 95% CI 1·15-2·57; P = 0·009). No significant differences were observed between cancer subtypes or the presence/absence of metastasis. CONCLUSIONS FOXE1 polyalanine repeat polymorphisms are associated with thyroid cancer, but only for tumours larger than 1 cm, suggesting a role in disease progression.
Collapse
Affiliation(s)
- Joana Raimundo
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Maria I Alvelos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Teresa Azevedo
- Endocrinology Service, Portuguese Institute of Oncology of Coimbra, Coimbra, Portugal
| | - Teresa Martins
- Endocrinology Service, Portuguese Institute of Oncology of Coimbra, Coimbra, Portugal
| | - Fernando J Rodrigues
- Endocrinology Service, Portuguese Institute of Oncology of Coimbra, Coimbra, Portugal
| | - Manuel C Lemos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
40
|
Nikitski AV, Rogounovitch TI, Bychkov A, Takahashi M, Yoshiura KI, Mitsutake N, Kawaguchi T, Matsuse M, Drozd VM, Demidchik Y, Nishihara E, Hirokawa M, Miyauchi A, Rubanovich AV, Matsuda F, Yamashita S, Saenko VA. Genotype Analyses in the Japanese and Belarusian Populations Reveal Independent Effects of rs965513 and rs1867277 but Do Not Support the Role of FOXE1 Polyalanine Tract Length in Conferring Risk for Papillary Thyroid Carcinoma. Thyroid 2017; 27:224-235. [PMID: 27824288 DOI: 10.1089/thy.2015.0541] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Several functional single-nucleotide polymorphisms (SNPs) at the FOXE1 locus on chromosome 9q22.33 have been associated with the risk for papillary thyroid carcinoma (PTC). This study set out to elucidate whether their effects are independent, using genotyping results in populations of Asian and European descent. METHODS SNPs rs965513 and rs1867277 and a polymorphic region determining the length of the FOXE1 polyalanine (poly-Ala) tract were genotyped in 501 patients with PTC and 748 healthy individuals from Japan, and in 660 patients and 820 population controls from Belarus. Functional analysis of transactivation activities of FOXE1 isoforms with varying number of alanine repeats was performed by a Dual-Luciferase® Assay. RESULTS All three polymorphisms were significantly associated with PTC in both populations on univariate analysis. However, conditional analysis revealed independent effects of rs965513 and rs1867277 SNPs but not of the FOXE1 poly-Ala polymorphism. The independent effect of the lead rs965513 SNP was observed in both populations, while that of rs1867277 was only identified in the Japanese population, in which linkage disequilibrium between the three polymorphisms is markedly weaker. Despite the strong decrease in transcriptional activity with increasing FOXE1 poly-Ala tract length, no difference in transactivation potential of the FOXE1 poly-Ala isoforms could be seen after adjustment for the minimal promoter activity in the reporter vectors. Plasmids encoding FOXE1 isoforms of increasing poly-Ala tract length were also found to produce less FOXE1 protein after cell transfection. CONCLUSIONS The functional variants rs965513 and rs1867277 independently contribute to genetic predisposition to PTC, while a contributing role of the FOXE1 poly-Ala polymorphism could not be confirmed.
Collapse
Affiliation(s)
- Alyaksandr V Nikitski
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
| | - Tatiana I Rogounovitch
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
| | - Andrey Bychkov
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
| | - Meiko Takahashi
- 2 Center for the Promotion of Interdisciplinary Education and Research, Kyoto University , Kyoto, Japan
| | - Koh-Ichiro Yoshiura
- 3 Department of Human Genetics, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
| | - Norisato Mitsutake
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
- 4 Nagasaki University Research Center for Genomic Instability and Carcinogenesis , Nagasaki, Japan
| | - Takahisa Kawaguchi
- 5 Center for Genomic Medicine, Kyoto University Graduate School of Medicine , Kyoto, Japan
| | - Michiko Matsuse
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
| | - Valentina M Drozd
- 6 Department of Endocrinology, Belarusian Academy for Postgraduate Education , Minsk, Belarus
| | - Yuri Demidchik
- 7 Department of Oncology, Belarusian Academy for Postgraduate Education , Minsk, Belarus
| | | | | | | | - Alexander V Rubanovich
- 9 Ecological Genetics Laboratory, Vavilov Institute of General Genetics, Russian Academy of Sciences , Moscow, Russia
- 10 Department of Radiation Molecular Epidemiology, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
| | - Fumihiko Matsuda
- 5 Center for Genomic Medicine, Kyoto University Graduate School of Medicine , Kyoto, Japan
| | - Shunichi Yamashita
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
- 10 Department of Radiation Molecular Epidemiology, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
| | - Vladimir A Saenko
- 10 Department of Radiation Molecular Epidemiology, Atomic Bomb Disease Institute, Nagasaki University , Nagasaki, Japan
| |
Collapse
|
41
|
MYH9 binds to lncRNA gene PTCSC2 and regulates FOXE1 in the 9q22 thyroid cancer risk locus. Proc Natl Acad Sci U S A 2017; 114:474-479. [PMID: 28049826 DOI: 10.1073/pnas.1619917114] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A locus on chromosome 9q22 harbors a SNP (rs965513) firmly associated with risk of papillary thyroid carcinoma (PTC). The locus also comprises the forkhead box E1 (FOXE1) gene, which is implicated in thyroid development, and a long noncoding RNA (lncRNA) gene, papillary thyroid cancer susceptibility candidate 2 (PTCSC2). How these might interact is not known. Here we report that PTCSC2 binds myosin-9 (MYH9). In a bidirectional promoter shared by FOXE1 and PTCSC2, MYH9 inhibits the promoter activity in both directions. This inhibition can be reversed by PTCSC2, which acts as a suppressor. RNA knockdown of FOXE1 in primary thyroid cells profoundly interferes with the p53 pathway. We propose that the interaction between the lncRNA, its binding protein MYH9, and the coding gene FOXE1 underlies the predisposition to PTC triggered by rs965513.
Collapse
|
42
|
Arauchi A, Matsuura K, Shimizu T, Okano T. Functional Thyroid Follicular Cells Differentiation from Human-Induced Pluripotent Stem Cells in Suspension Culture. Front Endocrinol (Lausanne) 2017; 8:103. [PMID: 28588551 PMCID: PMC5439004 DOI: 10.3389/fendo.2017.00103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/01/2017] [Indexed: 11/13/2022] Open
Abstract
The replacement of regenerated thyroid follicular cells (TFCs) is a promising therapeutic strategy for patients with hypothyroidism. Here, we have succeeded in inducing functional TFCs from human-induced pluripotent stem cells (iPSCs) in scalable suspension culture. Differentiation of iPSCs with Activin A treatment produced Sox17- and FoxA2-expressing definitive endodermal cells that also expressed thyroid transcription factors Pax8 and Nkx2-1. Further treatment with thyroid-stimulating hormone (TSH) induced TFCs expressing various types of thyroid proteins including TSH receptor, sodium-iodide symporter, thyroglobulin, and thyroid peroxidase. Interestingly, differentiated cells secreted free thyroxine in vitro. These results indicate successful differentiation of human iPSCs to functional TFCs that may enable us to fabricate thyroid tissues for regenerative medicine and disease models.
Collapse
Affiliation(s)
- Ayumi Arauchi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
- Department of Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
- *Correspondence: Katsuhisa Matsuura,
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
| |
Collapse
|
43
|
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
|
44
|
Ren Y, Lence-Anta JJ, Pereda CM, Chappe M, Velasco M, Infante I, Bustillo M, Turcios S, Leufroy A, Guérin T, Noël L, Lesueur F, Maillard S, Cléro E, Xhaard C, Allodji RS, Rubino C, Rodriguez R, Ortiz RM, de Vathaire F. FOXE1 Polymorphism Interacts with Dietary Iodine Intake in Differentiated Thyroid Cancer Risk in the Cuban Population. Thyroid 2016; 26:1752-1760. [PMID: 27610545 DOI: 10.1089/thy.2015.0594] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND The incidence of differentiated thyroid cancer (DTC) is low in Cuba, and the contribution of dietary factors to DTC in this population has not been investigated so far. The aim of this study was to evaluate the relationship between dietary iodine intake and DTC with regard to the interaction with environmental factors or some common single nucleotide polymorphisms (SNPs), based on a case-control study carried out in Cuba. METHODS A total of 203 cases and 212 controls from the general population were interviewed face-to-face using the dietary intake questionnaire and the photo booklet from the E3N cohort. A specific food composition table was constructed for this study. For each parameter studied, the odds ratio (OR) was stratified on age group and sex, and further adjusted for dietary energy, smoking status, ethnic group, level of education, number of pregnancies, and body surface area. RESULTS The risk of DTC was significantly reduced with increasing consumption of fish (p = 0.04), but no association between total dietary iodine intake and DTC risk was evident (p = 0.7). This lack of significant association was true whatever the age, the smoking status, the dietary selenium intake, and the ethnicity (p > 0.05). DTC risk was positively and strongly associated with the number of copies in the minor allele (A) for SNP rs965513 near FOXE1 among people who consumed less iodine than the median (p = 0.005). CONCLUSION Overall, the majority of the studied population had an optimal dietary iodine intake. DTC risk was inversely associated with high fish consumption. Furthermore, DTC risk was positively associated with the number of copies in the minor allele (A) of rs965513 among people who consumed less iodine than the median. Because these findings are based on post-diagnostic measures, studies with pre-diagnostic dietary iodine are needed for confirmation.
Collapse
Affiliation(s)
- Yan Ren
- 1 Cancer and Radiations, Center for Research in Epidemiology and Population Health (CESP) - U1018, French National Institute of Health and Medical Research (INSERM), Villejuif, France
- 2 Department of Research, Gustave Roussy Institute , Villejuif, France
- 3 Faculty of Medicine, University of Paris XI , Le Kremlin-Bicêtre, France
| | - Juan J Lence-Anta
- 4 Department of Clinical Research, Institute of Oncology and Radiobiology , Havana, Cuba
| | - Celia M Pereda
- 4 Department of Clinical Research, Institute of Oncology and Radiobiology , Havana, Cuba
| | - Mae Chappe
- 4 Department of Clinical Research, Institute of Oncology and Radiobiology , Havana, Cuba
| | - Milagros Velasco
- 4 Department of Clinical Research, Institute of Oncology and Radiobiology , Havana, Cuba
| | - Idalmis Infante
- 4 Department of Clinical Research, Institute of Oncology and Radiobiology , Havana, Cuba
| | - Marlene Bustillo
- 4 Department of Clinical Research, Institute of Oncology and Radiobiology , Havana, Cuba
| | - Silvia Turcios
- 5 Medical Care, National Institute of Endocrinology , Havana, Cuba
| | - Axelle Leufroy
- 6 Laboratory for Food Safety, Department of Chemical Contaminants in Food, Metallic Trace Elements and Minerals Unit, University of Paris-Est , Anses, Maisons-Alfort, France
| | - Thierry Guérin
- 6 Laboratory for Food Safety, Department of Chemical Contaminants in Food, Metallic Trace Elements and Minerals Unit, University of Paris-Est , Anses, Maisons-Alfort, France
| | - Laurent Noël
- 7 The French Directorate General for Food, Ministry of Agriculture , Agro-16 Food and Forestry, Paris, France
| | - Fabienne Lesueur
- 8 Institut Curie, Mines ParisTech, U900, French National Institute of Health and Medical Research (INSERM), Paris, France
| | - Stéphane Maillard
- 1 Cancer and Radiations, Center for Research in Epidemiology and Population Health (CESP) - U1018, French National Institute of Health and Medical Research (INSERM), Villejuif, France
- 2 Department of Research, Gustave Roussy Institute , Villejuif, France
- 3 Faculty of Medicine, University of Paris XI , Le Kremlin-Bicêtre, France
| | - Enora Cléro
- 1 Cancer and Radiations, Center for Research in Epidemiology and Population Health (CESP) - U1018, French National Institute of Health and Medical Research (INSERM), Villejuif, France
- 2 Department of Research, Gustave Roussy Institute , Villejuif, France
- 3 Faculty of Medicine, University of Paris XI , Le Kremlin-Bicêtre, France
| | - Constance Xhaard
- 1 Cancer and Radiations, Center for Research in Epidemiology and Population Health (CESP) - U1018, French National Institute of Health and Medical Research (INSERM), Villejuif, France
- 2 Department of Research, Gustave Roussy Institute , Villejuif, France
- 3 Faculty of Medicine, University of Paris XI , Le Kremlin-Bicêtre, France
| | - Rodrigue S Allodji
- 1 Cancer and Radiations, Center for Research in Epidemiology and Population Health (CESP) - U1018, French National Institute of Health and Medical Research (INSERM), Villejuif, France
- 2 Department of Research, Gustave Roussy Institute , Villejuif, France
- 3 Faculty of Medicine, University of Paris XI , Le Kremlin-Bicêtre, France
| | - Carole Rubino
- 1 Cancer and Radiations, Center for Research in Epidemiology and Population Health (CESP) - U1018, French National Institute of Health and Medical Research (INSERM), Villejuif, France
- 2 Department of Research, Gustave Roussy Institute , Villejuif, France
- 3 Faculty of Medicine, University of Paris XI , Le Kremlin-Bicêtre, France
| | - Regla Rodriguez
- 9 Department of Foreign Affairs, Public Health Ministry, Havana, Cuba
| | - Rosa M Ortiz
- 4 Department of Clinical Research, Institute of Oncology and Radiobiology , Havana, Cuba
| | - Florent de Vathaire
- 1 Cancer and Radiations, Center for Research in Epidemiology and Population Health (CESP) - U1018, French National Institute of Health and Medical Research (INSERM), Villejuif, France
- 2 Department of Research, Gustave Roussy Institute , Villejuif, France
- 3 Faculty of Medicine, University of Paris XI , Le Kremlin-Bicêtre, France
| |
Collapse
|
45
|
Niwa O, Barcellos-Hoff MH, Globus RK, Harrison JD, Hendry JH, Jacob P, Martin MT, Seed TM, Shay JW, Story MD, Suzuki K, Yamashita S. ICRP Publication 131: Stem Cell Biology with Respect to Carcinogenesis Aspects of Radiological Protection. Ann ICRP 2016; 44:7-357. [PMID: 26637346 DOI: 10.1177/0146645315595585] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This report provides a review of stem cells/progenitor cells and their responses to ionising radiation in relation to issues relevant to stochastic effects of radiation that form a major part of the International Commission on Radiological Protection's system of radiological protection. Current information on stem cell characteristics, maintenance and renewal, evolution with age, location in stem cell 'niches', and radiosensitivity to acute and protracted exposures is presented in a series of substantial reviews as annexes concerning haematopoietic tissue, mammary gland, thyroid, digestive tract, lung, skin, and bone. This foundation of knowledge of stem cells is used in the main text of the report to provide a biological insight into issues such as the linear-no-threshold (LNT) model, cancer risk among tissues, dose-rate effects, and changes in the risk of radiation carcinogenesis by age at exposure and attained age. Knowledge of the biology and associated radiation biology of stem cells and progenitor cells is more developed in tissues that renew fairly rapidly, such as haematopoietic tissue, intestinal mucosa, and epidermis, although all the tissues considered here possess stem cell populations. Important features of stem cell maintenance, renewal, and response are the microenvironmental signals operating in the niche residence, for which a well-defined spatial location has been identified in some tissues. The identity of the target cell for carcinogenesis continues to point to the more primitive stem cell population that is mostly quiescent, and hence able to accumulate the protracted sequence of mutations necessary to result in malignancy. In addition, there is some potential for daughter progenitor cells to be target cells in particular cases, such as in haematopoietic tissue and in skin. Several biological processes could contribute to protecting stem cells from mutation accumulation: (a) accurate DNA repair; (b) rapidly induced death of injured stem cells; (c) retention of the DNA parental template strand during divisions in some tissue systems, so that mutations are passed to the daughter differentiating cells and not retained in the parental cell; and (d) stem cell competition, whereby undamaged stem cells outcompete damaged stem cells for residence in the niche. DNA repair mainly occurs within a few days of irradiation, while stem cell competition requires weeks or many months depending on the tissue type. The aforementioned processes may contribute to the differences in carcinogenic radiation risk values between tissues, and may help to explain why a rapidly replicating tissue such as small intestine is less prone to such risk. The processes also provide a mechanistic insight relevant to the LNT model, and the relative and absolute risk models. The radiobiological knowledge also provides a scientific insight into discussions of the dose and dose-rate effectiveness factor currently used in radiological protection guidelines. In addition, the biological information contributes potential reasons for the age-dependent sensitivity to radiation carcinogenesis, including the effects of in-utero exposure.
Collapse
|
46
|
Wen G, Eder K, Ringseis R. Sterol regulatory element-binding proteins are transcriptional regulators of the thyroglobulin gene in thyroid cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:994-1003. [PMID: 27321819 DOI: 10.1016/j.bbagrm.2016.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/24/2016] [Accepted: 06/15/2016] [Indexed: 01/13/2023]
Abstract
The genes encoding sodium/iodide symporter (NIS) and thyroid peroxidase (TPO), both of which are essential for thyroid hormone (TH) synthesis, were shown to be regulated by sterol regulatory element-binding proteins (SREBP)-1c and -2. In the present study we tested the hypothesis that transcription of a further gene essential for TH synthesis, the thyroglobulin (TG) gene, is under the control of SREBP. To test this hypothesis, we studied the influence of inhibition of SREBP maturation and SREBP knockdown on TG expression in FRTL-5 thyrocytes and explored transcriptional regulation of the TG promoter by reporter gene experiments in FRTL-5 and HepG2 cells, gel shift assays and chromatin immunoprecipitation. Inhibition of SREBP maturation by 25-hydroxycholesterol and siRNA-mediated knockdown of either SREBP-1c or SREBP-2 decreased mRNA and protein levels of TG in FRTL-5 thyrocytes. Reporter gene assays with wild-type and mutated TG promoter reporter truncation constructs revealed that the rat TG promoter is transcriptionally activated by nSREBP-1c and nSREBP-2. DNA-binding assays and chromatin immunoprecipitation assays showed that both nSREBP-1c and nSREBP-2 bind to a SREBP binding motif with characteristics of an E-box SRE at position -63 in the rat TG promoter. In connection with recent findings that NIS and TPO are regulated by SREBP in thyrocytes the present findings support the view that SREBP are regulators of essential steps of TH synthesis in the thyroid gland such as iodide uptake, iodide oxidation and iodination of tyrosyl residues of TG. This moreover suggests that SREBP may be molecular targets for pharmacological modulation of TH synthesis.
Collapse
Affiliation(s)
- Gaiping Wen
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Gießen, 35392 Gießen, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Gießen, 35392 Gießen, Germany
| | - Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Gießen, 35392 Gießen, Germany.
| |
Collapse
|
47
|
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
|
48
|
Batista FA, Ward LS, Marcello MA, Martins MB, Peres KC, Torricelli C, Bufalo NE, Soares FA, da Silva MJ, Assumpção LVM. Gene expression of thyroid-specific transcription factors may help diagnose thyroid lesions but are not determinants of tumor progression. J Endocrinol Invest 2016; 39:423-9. [PMID: 26370671 DOI: 10.1007/s40618-015-0386-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/24/2015] [Indexed: 01/06/2023]
Abstract
PURPOSE The role of thyroid-specific transcription factors in thyroid malignancy is still poorly understood, so we investigate thyroid-specific transcription factors gene expression both in benign and in malignant thyroid nodules, aiming to study a possible clinical utility of these molecules. METHODS We quantified TTF-1, FOXE1 and PAX8 mRNA levels, relating their expression to diagnostic and prognostic features of thyroid tumors. RNA was extracted from 4 normal thyroid tissues, 101 malignant [99 papillary thyroid carcinomas (PTC) and 2 anaplastic thyroid carcinomas] and 99 benign thyroid lesion tissues [49 goiter and 50 follicular adenomas (FA)]. RESULTS Levels of mRNA of both FOXE1 (P < 0.0001) and PAX8 (P < 0.0001) genes, but not TTF-1 (P = 0.7056), were higher in benign than in malignant thyroid lesions. FOXE1 was able to identify malignant nodules with 75.8 % sensitivity, 76.1 % specificity, 75.8 % positive predictive value, 76.1 % negative predictive value and 75.9 % accuracy. PAX8 was able to identify malignancy with 60.6 % sensitivity, 81.1 % specificity, 76.9 % positive predictive value, 66.4 % negative predictive value and 70.6 % accuracy. Both FOXE1 and PAX8 gene expression patterns were also able to differentiate FA from the follicular variant of PTC-FVPTC. However, the investigated gene expression was neither associated with any clinical feature of tumor aggressiveness nor associated with recurrence or survival. CONCLUSIONS We suggest that FOXE1 and PAX8 gene expression patterns may help to diagnose thyroid nodules, identifying malignancy and characterizing follicular-patterned thyroid lesions, but are not determinants of thyroid tumor progression.
Collapse
Affiliation(s)
- F A Batista
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences (FCM), School of Medical Sciences, University of Campinas (Unicamp), Rua Tessalia Vieira de Camargo, 126, Cidade Universitaria Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil.
| | - L S Ward
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences (FCM), School of Medical Sciences, University of Campinas (Unicamp), Rua Tessalia Vieira de Camargo, 126, Cidade Universitaria Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil
| | - M A Marcello
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences (FCM), School of Medical Sciences, University of Campinas (Unicamp), Rua Tessalia Vieira de Camargo, 126, Cidade Universitaria Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil
| | - M B Martins
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences (FCM), School of Medical Sciences, University of Campinas (Unicamp), Rua Tessalia Vieira de Camargo, 126, Cidade Universitaria Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil
| | - K C Peres
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences (FCM), School of Medical Sciences, University of Campinas (Unicamp), Rua Tessalia Vieira de Camargo, 126, Cidade Universitaria Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil
| | - C Torricelli
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences (FCM), School of Medical Sciences, University of Campinas (Unicamp), Rua Tessalia Vieira de Camargo, 126, Cidade Universitaria Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil
| | - N E Bufalo
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences (FCM), School of Medical Sciences, University of Campinas (Unicamp), Rua Tessalia Vieira de Camargo, 126, Cidade Universitaria Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil
| | - F A Soares
- Department of Pathology, AC Camargo Hospital - Antonio Prudente Foundation, Rua Professor Antônio Prudente, 211, Liberdade, São Paulo, São Paulo, 01509-010, Brazil
| | - M J da Silva
- Molecular Biology and Genetic Engineering Center (CBMEG), University of Campinas (Unicamp), Avenida Cândido Rondon, Cidade Universitaria Zeferino Vaz, Campinas, São Paulo, 13083-875, Brazil
| | - L V M Assumpção
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences (FCM), School of Medical Sciences, University of Campinas (Unicamp), Rua Tessalia Vieira de Camargo, 126, Cidade Universitaria Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil
| |
Collapse
|
49
|
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
|
50
|
Lidral AC, Liu H, Bullard SA, Bonde G, Machida J, Visel A, Uribe LMM, Li X, Amendt B, Cornell RA. A single nucleotide polymorphism associated with isolated cleft lip and palate, thyroid cancer and hypothyroidism alters the activity of an oral epithelium and thyroid enhancer near FOXE1. Hum Mol Genet 2015; 24:3895-907. [PMID: 25652407 PMCID: PMC4476440 DOI: 10.1093/hmg/ddv047] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 02/02/2015] [Indexed: 12/11/2022] Open
Abstract
Three common diseases, isolated cleft lip and cleft palate (CLP), hypothyroidism and thyroid cancer all map to the FOXE1 locus, but causative variants have yet to be identified. In patients with CLP, the frequency of coding mutations in FOXE1 fails to account for the risk attributable to this locus, suggesting that the common risk alleles reside in nearby regulatory elements. Using a combination of zebrafish and mouse transgenesis, we screened 15 conserved non-coding sequences for enhancer activity, identifying three that regulate expression in a tissue specific pattern consistent with endogenous foxe1 expression. These three, located -82.4, -67.7 and +22.6 kb from the FOXE1 start codon, are all active in the oral epithelium or branchial arches. The -67.7 and +22.6 kb elements are also active in the developing heart, and the -67.7 kb element uniquely directs expression in the developing thyroid. Within the -67.7 kb element is the SNP rs7850258 that is associated with all three diseases. Quantitative reporter assays in oral epithelial and thyroid cell lines show that the rs7850258 allele (G) associated with CLP and hypothyroidism has significantly greater enhancer activity than the allele associated with thyroid cancer (A). Moreover, consistent with predicted transcription factor binding differences, the -67.7 kb element containing rs7850258 allele G is significantly more responsive to both MYC and ARNT than allele A. By demonstrating that this common non-coding variant alters FOXE1 expression, we have identified at least in part the functional basis for the genetic risk of these seemingly disparate disorders.
Collapse
Affiliation(s)
| | - Huan Liu
- Dows Research Institute, State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | | | - Greg Bonde
- Department of Anatomy, University of Iowa, Iowa City, IA, USA
| | - Junichiro Machida
- Department of Oral and Maxillofacial Surgery, Toyota Memorial Hospital, Toyota City, Aichi, Japan
| | - Axel Visel
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA and Department of Energy Joint Genome Institute, Walnut Creek, CA, USA
| | | | - Xiao Li
- Department of Anatomy, University of Iowa, Iowa City, IA, USA
| | - Brad Amendt
- Department of Anatomy, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|