Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cancers (Basel) ; 12(9)2020 Sep 06.
Article in English | MEDLINE | ID: mdl-32899953

ABSTRACT

Although fine-needle aspiration cytology (FNAC) is helpful in determining whether thyroid nodules are benign or malignant, this distinction remains a cytological challenge in follicular neoplasms. Identification of genomic alterations in cytological specimens with direct and routine techniques would therefore have great clinical value. A series of 153 cases consisting of 72 and 81 histopathologically confirmed classic follicular adenomas (cFAs) and classic follicular thyroid carcinomas (cFTCs), respectively, was studied by means of different molecular techniques in three different cohorts of patients (pts). In the first cohort (training set) of 66 pts, three specific alterations characterized by array comparative genomic hybridization (aCGH) were exclusively found in half of cFTCs. These structural abnormalities corresponded to losses of 1p36.33-35.1 and 22q13.2-13.31, and gain of whole chromosome X. The second independent cohort (validation set) of 60 pts confirmed these data on touch preparations of frozen follicular neoplasms by triple DNA fluorescent in situ hybridization using selected commercially available probes. The third cohort, consisting of 27 archived cytological samples from an equal number of pts that had been obtained for preoperative FNAC and morphologically classified as and histologically verified to be follicular neoplasms, confirmed our previous findings and showed the feasibility of the DNA FISH (DNA fluorescent in situ hybridization) assay. All together, these data suggest that our triple DNA FISH diagnostic assay may detect 50% of cFTCs with a specificity higher than 98% and be useful as a low-cost adjunct to cytomorphology to help further classify follicular neoplasms on already routinely stained cytological specimens.

2.
Br J Cancer ; 115(5): 549-52, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27441499

ABSTRACT

BACKGROUND: We aimed to understand the dependence of MEK and m-TOR inhibition in EGFR(WT)/ALK(non-rearranged) NSCLC cell lines. METHODS: In a panel of KRAS(M) and KRAS(WT) NSCLC cell lines, we determined growth inhibition (GI) following maximal reduction in p-ERK and p-S6RP caused by trametinib (MEK inhibitor) and AZD2014 (m-TOR inhibitor), respectively. RESULTS: GI caused by maximal m-TOR inhibition was significantly greater than GI caused by maximal MEK inhibition in the cell line panel (52% vs 18%, P<10(-4)). There was no significant difference in GI caused by maximal m-TOR compared with maximal m-TOR+MEK inhibition. However, GI caused by the combination was significantly greater in the KRAS(M) cell lines (79% vs 61%, P=0.017). CONCLUSIONS: m-TOR inhibition was more critical to GI than MEK inhibition in EGFR(WT)/ALK(non-rearranged) NSCLC cells. The combination of MEK and m-TOR inhibition was most effective in KRAS(M) cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Genes, ras , Lung Neoplasms/metabolism , MAP Kinase Kinase Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Humans , Lung Neoplasms/genetics
3.
Proc Natl Acad Sci U S A ; 112(16): 5051-6, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25848056

ABSTRACT

Ionizing radiation (IR) causes not only acute tissue damage, but also late effects in several cell generations after the initial exposure. The thyroid gland is one of the most sensitive organs to the carcinogenic effects of IR, and we have recently highlighted that an oxidative stress is responsible for the chromosomal rearrangements found in radio-induced papillary thyroid carcinoma. Using both a human thyroid cell line and primary thyrocytes, we investigated the mechanism by which IR induces the generation of reactive oxygen species (ROS) several days after irradiation. We focused on NADPH oxidases, which are specialized ROS-generating enzymes known as NOX/DUOX. Our results show that IR induces delayed NADPH oxidase DUOX1-dependent H2O2 production in a dose-dependent manner, which is sustained for several days. We report that p38 MAPK, activated after IR, increased DUOX1 via IL-13 expression, leading to persistent DNA damage and growth arrest. Pretreatment of cells with catalase, a scavenger of H2O2, or DUOX1 down-regulation by siRNA abrogated IR-induced DNA damage. Analysis of human thyroid tissues showed that DUOX1 is elevated not only in human radio-induced thyroid tumors, but also in sporadic thyroid tumors. Taken together, our data reveal a key role of DUOX1-dependent H2O2 production in long-term persistent radio-induced DNA damage. Our data also show that DUOX1-dependent H2O2 production, which induces DNA double-strand breaks, can cause genomic instability and promote the generation of neoplastic cells through its mutagenic effect.


Subject(s)
Gamma Rays , NADPH Oxidases/metabolism , Oxidative Stress/radiation effects , Cell Line , DNA Damage , Dual Oxidases , Extracellular Space/metabolism , Extracellular Space/radiation effects , Gene Expression Regulation, Neoplastic , Humans , Hydrogen Peroxide/metabolism , Interleukin-13/genetics , Interleukin-13/metabolism , NADPH Oxidases/genetics , Thyroid Gland/enzymology , Thyroid Gland/pathology , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Mol Cell Endocrinol ; 392(1-2): 8-13, 2014 Jul 05.
Article in English | MEDLINE | ID: mdl-24813658

ABSTRACT

Epigenetic control of gene expression plays a major influence in the development and progression of many cancer types. Aim of the present study was to investigate the expression of epigenetic regulators in a large cohort of medullary thyroid carcinomas (MTC), correlating the data with the clinical outcome and mutational status of the patients. Taqman Low Density Arrays (TLDAs) were used to analyze expression levels of several genes involved in the epigenetic control of transcription in a series of 54 MTCs. The patients cohort included 13 familial MTCs and 41 sporadic forms; 33 hosted a RET mutation and 13 a RAS somatic mutation. The expression profiling revealed in the more aggressive diseases (i.e. occurrence of metastases; persistent disease; disease-related death) a significant increase of EZH2 and SMYD3 gene expression. The increased levels of EZH2 and SMYD3 did not correlate significantly with mutational status of RET or RAS genes. Thus, the histone methyltransferases EZH2 and SMYD3 mRNA expression may represent useful prognostic biomarkers tailoring the most appropriate follow-up and timing of therapeutic approaches.


Subject(s)
Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Polycomb Repressive Complex 2/genetics , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Neuroendocrine , Child , Enhancer of Zeste Homolog 2 Protein , Female , Histone-Lysine N-Methyltransferase/metabolism , Humans , Lymphatic Metastasis/genetics , Male , Middle Aged , Mutation/genetics , Neoplasm Invasiveness , Polycomb Repressive Complex 2/metabolism , Thyroid Neoplasms/pathology , Treatment Outcome , Young Adult
5.
Thyroid ; 24(1): 43-51, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24256343

ABSTRACT

BACKGROUND: Medullary thyroid carcinoma (MTC) is a rare tumor that is caused by activating mutations in the proto-oncogene RET. Vandetanib, a tyrosine-kinase inhibitor, has been recently approved to treat adult patients with metastatic MTC. The aim of this study was to investigate changes in signaling pathways induced by vandetanib treatment in preclinical MTC models, using the reverse-phase protein array method (RPPA). METHODS: The human TT cell line was used to assess in vitro and in vivo activity of vandetanib. Protein extracts from TT cells or TT xenografted mice, treated by increasing concentrations of vandetanib for different periods of time, were probed with a set of 12 antibodies representing major signaling pathways, using RPPA. Results were validated using two distinct protein detection methods: Western immunoblotting and immunohistochemistry. RESULTS: Vandetanib displays antiproliferative and antiangiogenic activities and inhibits RET autophosphorylation. The MAPK and AKT pathways were the two major signaling pathways inhibited by vandetanib. Interestingly, phosphorylated levels of NFκB-p65 were significantly increased by vandetanib. Comparable results were obtained in both the in vitro and in vivo approaches, as well as for the protein detection methods. However, some discrepancies were observed between RPPA and Western immunoblotting, possibly due to lack of specificity of the primary antibodies used. CONCLUSIONS: Overall, our results confirmed the interest of RPPA for screening global changes induced in signaling pathways by kinase inhibitors. MAPK and AKT were identified as the main pathways involved in vandetanib response in MTC models. Our results also suggest alternative routes for controlling the disease, and provide a rationale for the development of therapeutic combinations based on the comprehensive identification of molecular events induced by inhibitors.


Subject(s)
Piperidines/therapeutic use , Quinazolines/therapeutic use , Thyroid Neoplasms/drug therapy , Adult , Animals , Carcinoma, Neuroendocrine , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Mice , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects
6.
Thyroid ; 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23822199

ABSTRACT

BACKGROUND: Medullary thyroid carcinoma (MTC) is a rare tumor that is due to activating mutations in the proto-oncogene RET. Vandetanib, a tyrosine-kinase inhibitor, has been recently approved to treat adult patients with metastatic MTC. The aim of this study was to investigate changes in signaling pathways induced by vandetanib treatment in preclinical MTC models, using the reverse-phase protein array method (RPPA). METHODS: The human TT cell line was used to assess in vitro and in vivo activity of vandetanib. Protein extracts from TT cells or TT xenografted mice, treated by increasing concentrations of vandetanib for different periods of time, were probed with a set of 12 antibodies representing major signaling pathways, using RPPA. Results were validated using two distinct protein detection methods, western-immunoblotting and immunohistochemistry. RESULTS: Vandetanib displays antiproliferative and antiangiogenic activities and inhibits RET auto-phosphorylation. MAPK and AKT pathways were the two major signaling pathways inhibited by vandetanib. Interestingly, phosphorylated levels of NFκB-p65 were significantly increased by vandetanib. Comparable results were obtained in both the in vitro and in vivo approaches as well as for the protein detection methods, although some discrepancies were observed between RPPA and western-immunoblotting. CONCLUSIONS: Results confirmed the reliability and the utility of RPPA for screening global changes induced in signaling pathways by kinase inhibitors. MAPK and AKT were identified as the main pathways involved in vandetanib response in MTC models. Our results also suggest alternative routes for controlling the disease and provide a rationale for the development of therapeutic combinations based on the comprehensive identification of molecular events induced by inhibitors.

7.
PLoS One ; 6(7): e22567, 2011.
Article in English | MEDLINE | ID: mdl-21811634

ABSTRACT

BACKGROUND: A relationship between the increased density of tumor-associated macrophages (TAMs) and decreased survival was recently reported in thyroid cancer patients. Among these tumors, anaplastic thyroid cancer (ATC) is one of the most aggressive solid tumors in humans. TAMs (type M2) have been recognized as promoting tumor growth. The purpose of our study was to analyze with immunohistochemistry the presence of TAMs in a series of 27 ATC. METHODOLOGY/PRINCIPAL FINDINGS: Several macrophages markers such as NADPH oxidase complex NOX2-p22phox, CD163 and CD 68 were used. Immunostainings showed that TAMs represent more than 50% of nucleated cells in all ATCs. Moreover, these markers allowed the identification of elongated thin ramified cytoplasmic extensions, bestowing a "microglia-like" appearance on these cells which we termed "Ramified TAMs" (RTAMs). In contrast, cancer cells were totally negative. Cellular stroma was highly simplified since apart from cancer cells and blood vessels, RTAMs were the only other cellular component. RTAMs were evenly distributed and intermingled with cancer cells, and were in direct contact with other RTAMs via their ramifications. Moreover, RTAMs displayed strong immunostaining for connexin Cx43. Long chains of interconnected RTAMs arose from perivascular clusters and were dispersed within the tumor parenchyma. When expressed, the glucose transporter Glut1 was found in RTAMs and blood vessels, but rarely in cancer cells. CONCLUSION: ATCs display a very dense network of interconnected RTAMs in direct contact with intermingled cancer cells. To our knowledge this is the first time that such a network is described in a malignant tumor. This network was found in all our studied cases and appeared specific to ATC, since it was not found in differentiated thyroid cancers specimens. Taken together, these results suggest that RTAMs network is directly related to the aggressiveness of the disease via metabolic and trophic functions which remain to be determined.


Subject(s)
Macrophages/pathology , Thyroid Neoplasms/pathology , Aged , Aged, 80 and over , Blood Vessels/metabolism , Blood Vessels/pathology , CD3 Complex/metabolism , Cell Line, Tumor , Cell Shape , Connexin 43/metabolism , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Glucose Transporter Type 1/metabolism , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Macrophages/metabolism , Macrophages/ultrastructure , Middle Aged , NADPH Oxidases/metabolism , Neoplasm Proteins/metabolism , Positron-Emission Tomography , Staining and Labeling , Subcellular Fractions/metabolism , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/ultrastructure , Tomography, X-Ray Computed
8.
PLoS One ; 6(2): e16752, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21364918

ABSTRACT

Notch signalling plays an important role in endocrine development, through its target gene Hes1. Hes1, a bHLH transcriptional repressor, influences progenitor cell proliferation and differentiation. Recently, Hes1 was shown to be expressed in the thyroid and regulate expression of the sodium iodide symporter (Nis). To investigate the role of Hes1 for thyroid development, we studied thyroid morphology and function in mice lacking Hes1. During normal mouse thyroid development, Hes1 was detected from E9.5 onwards in the median anlage, and at E11.5 in the ultimobranchial bodies. Hes1(-/-) mouse embryos had a significantly lower number of Nkx2-1-positive progenitor cells (p<0.05) at E9.5 and at E11.5. Moreover, Hes1(-/-) mouse embryos showed a significantly smaller total thyroid surface area (-40 to -60%) compared to wild type mice at all study time points (E9.5-E16.5). In both Hes1(-/-) and wild type mouse embryos, most Nkx2-1-positive thyroid cells expressed the cell cycle inhibitor p57 at E9.5 in correlation with low proliferation index. In Hes1(-/-) mouse embryos, fusion of the median anlage with the ultimobranchial bodies was delayed by 3 days (E16.5 vs. E13.5 in wild type mice). After fusion of thyroid anlages, hypoplastic Hes1(-/-) thyroids revealed a significantly decreased labelling area for T4 (-78%) and calcitonin (-65%) normalized to Nkx2-1 positive cells. Decreased T4-synthesis might be due to reduced Nis labelling area (-69%). These findings suggest a dual role of Hes1 during thyroid development: first, control of the number of both thyrocyte and C-cell progenitors, via a p57-independent mechanism; second, adequate differentiation and endocrine function of thyrocytes and C-cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Differentiation/genetics , Homeodomain Proteins/physiology , Morphogenesis/genetics , Thyroid Gland/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Knockout , Models, Biological , Organ Size/genetics , Stem Cells/metabolism , Stem Cells/physiology , Thyroid Gland/abnormalities , Thyroid Gland/metabolism , Thyroid Gland/physiology , Transcription Factor HES-1
9.
Clin Cancer Res ; 17(7): 2044-54, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21325074

ABSTRACT

PURPOSE: Medullary thyroid carcinoma (MTC), an aggressive rare tumor due to activating mutations in the proto-oncogene RET, requires new therapeutic strategies. Sunitinib, a potent inhibitor of RET, VEGF receptor (VEGFR)-1, VEGFR-2, VEGFR-3, and platelet-derived growth factor receptor (PDGFR)α/ß, has been reported as clinically effective in some patients with advanced MTC. In this study, we examine molecular mechanisms of action of sunitinib and identify candidate soluble biomarkers of response. EXPERIMENTAL DESIGN: Both in vitro and in vivo assays, using the human TT RET(C634W) MTC cell line, were done to assess the activity of sunitinib. Kinetic microarray studies were used to analyze molecular pathways modified by sunitinib and to identify candidate biomarkers that were subsequently investigated in the serum of patients. RESULTS: Sunitinib displayed antiproliferative and antiangiogenic activities and inhibited RET autophosphorylation and activation of downstream signaling pathways. We showed that sunitinib treatment induced major changes in the expression of genes involved in tissue invasion and metastasis including vimentin (VIM), urokinase plasminogen (PLAU), tenascin-C (TN-C), SPARC, and CD44. Analyzing downregulated genes, we identified those encoding secreted proteins and, among them, interleukin (IL)-8 was found to be modulated in the serum of xenografted mice under sunitinib treatment. Furthermore, we demonstrated that metastatic MTC patients presented increased serum levels of IL-8 and TGF-ß2. CONCLUSIONS: Experimental models confirm the clinical efficacy of sunitinib observed in a few studies. Molecular pathways revealed by genomic signatures underline the impact of sunitinib on tissue invasion. Selected soluble candidate biomarkers could be of value for monitoring sunitinib response in metastatic MTC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Carcinoma, Medullary/metabolism , Indoles/pharmacology , Pyrroles/pharmacology , Thyroid Neoplasms/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Biomarkers, Tumor/genetics , Carcinoembryonic Antigen/blood , Carcinoma, Medullary/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/blood , Interleukin-8/blood , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , RNA Interference , Sunitinib , Tenascin/blood , Thyroid Neoplasms/pathology , Transforming Growth Factor beta2/blood , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
10.
Endocr Relat Cancer ; 18(1): 193-206, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21148326

ABSTRACT

Both external and internal exposure to ionizing radiation are strong risk factors for the development of thyroid tumors. Until now, the diagnosis of radiation-induced thyroid tumors has been deduced from a network of arguments taken together with the individual history of radiation exposure. Neither the histological features nor the genetic alterations observed in these tumors have been shown to be specific fingerprints of an exposure to radiation. The aim of our work is to define ionizing radiation-related molecular specificities in a series of secondary thyroid tumors developed in the radiation field of patients treated by radiotherapy. To identify molecular markers that could represent a radiation-induction signature, we compared 25K microarray transcriptome profiles of a learning set of 28 thyroid tumors, which comprised 14 follicular thyroid adenomas (FTA) and 14 papillary thyroid carcinomas (PTC), either sporadic or consecutive to external radiotherapy in childhood. We identified a signature composed of 322 genes which discriminates radiation-induced tumors (FTA and PTC) from their sporadic counterparts. The robustness of this signature was further confirmed by blind case-by-case classification of an independent set of 29 tumors (16 FTA and 13 PTC). After the histology code break by the clinicians, 26/29 tumors were well classified regarding tumor etiology, 1 was undetermined, and 2 were misclassified. Our results help shed light on radiation-induced thyroid carcinogenesis, since specific molecular pathways are deregulated in radiation-induced tumors.


Subject(s)
Adenoma/etiology , Carcinoma, Papillary/etiology , Gene Expression Profiling , Neoplasms, Radiation-Induced/etiology , Radiotherapy/adverse effects , Thyroid Neoplasms/etiology , Adenoma/diagnosis , Adenoma/genetics , Adolescent , Adult , Age Factors , Carcinoma, Papillary/diagnosis , Carcinoma, Papillary/genetics , Child , Child, Preschool , Diagnosis, Differential , Dose-Response Relationship, Radiation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Radiation-Induced/diagnosis , Neoplasms, Radiation-Induced/genetics , Oligonucleotide Array Sequence Analysis , Radiotherapy Dosage , Single-Blind Method , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/genetics , Young Adult
11.
J Clin Endocrinol Metab ; 95(12): 5403-11, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20826581

ABSTRACT

CONTEXT: Thyroperoxidase (TPO) and dual oxidase (DUOX) are present at the apical membrane of thyrocytes, where TPO catalyzes thyroid hormone biosynthesis in the presence of H2O2 produced by DUOX. Both enzymes are colocalized and associated, but the consequences of this interaction remain obscure. OBJECTIVE: The objective of this study was to evaluate the functional consequences of TPO-DUOX interaction at the plasma membrane. DESIGN: The functional consequences of DUOX-TPO interaction were studied by measuring extracellular H2O2 concentration and TPO activity in a heterologous system. For this purpose, HEK293 cells were transiently transfected with a combination of human TPO with human DUOX1 or DUOX2 in the presence of their respective maturation factors, DUOXA1 or DUOXA2. The effect of human DUOX2 mutants in which cysteine residues in the N-terminal domain were replaced by glycines was also analyzed. RESULTS: We observed that production of H2O2 decreases both TPO and DUOX activities. We show that TPO presents a catalase-like effect that protects DUOX from inhibition by H2O2. This catalase-like effect depends on the association between both enzymes, which probably occurs through the DUOX peroxidase-like domain because this effect was not observed with human DUOX2 mutants. CONCLUSION: The DUOX-TPO association at the plasma membrane is relevant for normal enzyme properties. Normally, TPO consumes H2O2 produced by DUOX, decreasing the availability of this substance at the apical membrane of thyrocytes and, in turn, probably decreasing the oxidative damage of macromolecules.


Subject(s)
Autoantigens/metabolism , Cell Membrane/enzymology , Iodide Peroxidase/metabolism , Iron-Binding Proteins/metabolism , NADPH Oxidases/metabolism , Oxidoreductases/metabolism , Autoantigens/genetics , Catalase/metabolism , Dual Oxidases , Flow Cytometry , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Iodide Peroxidase/genetics , Iron-Binding Proteins/genetics , Kidney/enzymology , NADPH Oxidases/genetics , Oligonucleotides, Antisense , Transfection
12.
Cancer Res ; 70(10): 4123-32, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20424115

ABSTRACT

During childhood, the thyroid gland is one of the most sensitive organs to the carcinogenetic effects of ionizing radiation that may lead to papillary thyroid carcinoma (PTC) associated with RET/PTC oncogene rearrangement. Exposure to ionizing radiation induces a transient "oxidative burst" through radiolysis of water, which can cause DNA damage and mediates part of the radiation effects. H(2)O(2) is a potent DNA-damaging agent that induces DNA double-strand breaks, and consequently, chromosomal aberrations. Irradiation by 5 Gy X-ray increased extracellular H(2)O(2). Therefore, we investigated the implication of H(2)O(2) in the generation of RET/PTC1 rearrangement after X-ray exposure. We developed a highly specific and sensitive nested reverse transcription-PCR method. By using the human thyroid cell line HTori-3, previously found to produce RET/PTC1 after gamma-irradiation, we showed that H(2)O(2), generated during a 5 Gy X-ray irradiation, causes DNA double-strand breaks and contributes to RET/PTC1 formation. Pretreatment of cells with catalase, a scavenger of H(2)O(2), significantly decreased RET/PTC1 rearrangement formation. Finally, RET/PTC chromosomal rearrangement was detected in HTori-3.1 cells after exposure of cells to H(2)O(2) (25 micromol/L), at a dose that did not affect the cell viability. This study shows for the first time that H(2)O(2) is able to cause RET/PTC1 rearrangement in thyroid cells and consequently highlights that oxidative stress could be responsible for the occurrence of RET/PTC1 rearrangement found in thyroid lesions even in the absence of radiation exposure.


Subject(s)
Carcinoma, Papillary/pathology , Gene Rearrangement/radiation effects , Hydrogen Peroxide/pharmacology , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/genetics , Thyroid Gland/radiation effects , Thyroid Neoplasms/genetics , Blotting, Western , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/radiation effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/radiation effects , Gene Rearrangement/drug effects , Humans , Lung/cytology , Lung/drug effects , Lung/radiation effects , Oncogene Proteins, Fusion/metabolism , Oxidants/pharmacology , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , X-Rays
13.
Endocr Relat Cancer ; 17(2): 361-71, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20228124

ABSTRACT

Non-functioning pituitary adenomas (NFPAs) may be locally invasive. Markers of invasiveness are needed to guide patient management and particularly the use of adjuvant radiotherapy. To examine whether invasive NFPAs display a specific gene expression profile relative to non-invasive tumors, we selected 40 NFPAs (38 of the gonadotroph type) and classified them as invasive (n=22) or non-invasive (n=18) on the basis of magnetic resonance imaging and surgical findings. We then performed pangenomic analysis with the 44k Agilent human whole genome expression oligonucleotide microarray in order to identify genes with differential expression between invasive and non-invasive NFPAs. Candidate genes were then tested in qRT-PCR. Prediction class analysis showed that the expression of 346 genes differed between invasive and non-invasive NFPAs (P<0.001), of which 233 genes were up-regulated and 113 genes were down-regulated in invasive tumors. On the basis of Ingenuity networks and the degree of up- or down-regulation in invasive versus non-invasive tumors, 35 genes were selected for expression quantification by qRT-PCR. Overexpression of only four genes was confirmed, namely IGFBP5 (P=0.02), MYO5A (P=0.04), FLT3 (P=0.01), and NFE2L1 (P=0.02). At the protein level, only myosin 5A (MYO5A) immunostaining was stronger in invasive than in non-invasive NFPAs. Molecular signature allows to differentiate 'grossly' invasive from non-invasive NFPAs. The product of one of these genes, MYO5A, may be a useful marker of tumor invasiveness.


Subject(s)
Adenoma/genetics , Neoplasm Recurrence, Local/genetics , Pituitary Neoplasms/genetics , Adenoma/metabolism , Adenoma/pathology , Adult , Aged , Aged, 80 and over , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Male , Microarray Analysis , Middle Aged , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Type V/genetics , Myosin Type V/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
14.
Endocr Relat Cancer ; 17(1): 27-37, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19779036

ABSTRACT

NADPH oxidase 4 (NOX4) belongs to the NOX family that generates reactive oxygen species (ROS). Function and tissue distribution of NOX4 have not yet been entirely clarified. To date, in the thyroid gland, only DUOX1/2 NOX systems have been described. NOX4 mRNA expression, as shown by real-time PCR, was present in normal thyroid tissue, regulated by TSH and significantly increased in differentiated cancer tissues. TSH increased the protein level of NOX4 in human thyroid primary culture and NOX4-dependent ROS generation. NOX4 immunostaining was detected in normal and pathologic thyroid tissues. In normal thyroid tissue, staining was heterogeneous and mostly found in activated columnar thyrocytes but absent in quiescent flat cells. Papillary and follicular thyroid carcinomas displayed more homogeneous staining. The p22(phox) protein that forms a heterodimeric enzyme complex with NOX4 displayed an identical cellular expression pattern and was also positively regulated by TSH. ROS may have various biological effects, depending on the site of production. Intracellular NOX4-p22(phox) localization suggests a role in cytoplasmic redox signaling, in contrast to the DUOX localization at the apical membrane that corresponds to an extracellular H(2)O(2) production. Increased NOX4-p22(phox) in cancer might be related to a higher proliferation rate and tumor progression but a role in the development of tumors has to be further studied and established in the future.


Subject(s)
Adenocarcinoma, Follicular/enzymology , Adenoma/enzymology , Carcinoma, Papillary/enzymology , Carcinoma/enzymology , NADPH Oxidases/biosynthesis , Neoplasm Proteins/biosynthesis , Reactive Oxygen Species/metabolism , Thyroid Gland/enzymology , Thyroid Neoplasms/enzymology , Adenocarcinoma, Follicular/pathology , Adenoma/pathology , Carcinoma/pathology , Carcinoma, Papillary/pathology , Cells, Cultured/enzymology , Cytoplasm/enzymology , Dual Oxidases , Enzyme Induction/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogen Peroxide/metabolism , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neoplasm Proteins/genetics , Oxidation-Reduction , RNA Interference , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , RNA, Small Interfering/pharmacology , Thyroid Gland/cytology , Thyroid Neoplasms/pathology , Thyrotropin/pharmacology
15.
Endocr Relat Cancer ; 16(4): 1261-72, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19675075

ABSTRACT

RET oncogene mutations are found in familial medullary thyroid carcinomas (MTC) and in one-third of sporadic cases. Oncogenic mechanisms involved in non-RET mutated sporadic MTC remain unclear. To study alterations associated with the development of both inherited and sporadic MTC, pangenomic DNA microarrays were used to analyze the transcriptome of 13 MTCs (four familial and nine sporadic). By using an ANOVA test, a list of 173 gene sequences with at least a twofold change expression was obtained. A subset of differentially expressed genes was controlled by real-time quantitative PCR and immunohistochemistry on a larger collection of MTCs. The expression pattern of those genes allowed us to distinguish two groups of sporadic tumors. The first group displays an expression profile similar to that expressed by inherited RET634 tumors. The second presents an expression profile close to that displayed by inherited RET918 tumors and includes tumors from patients with distant metastases. It is characterized by the overexpression of genes involved in proliferation and invasion (PTN, ESM1, and CEACAM6) or matrix remodeling (COL1A1, COL1A2, and FAP). Interestingly, RET918 tumors showed overexpression of the PTN gene, encoding pleiotrophin, a protein associated with metastasis. Using a MTC cell line, silencing of RET induced the inhibition of PTN gene expression. Overall, our results suggest that familial MTC and sporadic MTC could activate similar oncogenic pathways.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Medullary/genetics , Mutation/genetics , Oncogenes/genetics , Proto-Oncogene Proteins c-ret/genetics , Thyroid Neoplasms/genetics , Adolescent , Adult , Aged , Carcinoma, Medullary/metabolism , Carcinoma, Medullary/secondary , Child , Child, Preschool , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Prognosis , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Proto-Oncogene Proteins c-ret/metabolism , RNA, Small Interfering/pharmacology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Young Adult
16.
Clin Chem ; 55(8): 1484-91, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19541864

ABSTRACT

BACKGROUND: The 1st WHO International Reference Reagents (IRRs) for 6 human chorionic gonadotropin (hCG)-related molecular variants, highly purified and calibrated in substance concentrations by the IFCC Working Group for hCG, permit experimental elucidation of what commercially available hCG methods measure in molar terms and enable assessment of their fitness for clinical purposes. METHODS: Pools containing known amounts of the IRRs spiked into normal human serum were issued to participants through the UK National External Quality Assessment Service for hCG for a period of 7 years. Among 16 assays used, 4 recognized only hCG, whereas 6 recognized hCG and its free beta-subunit (hCGbeta), and 6 recognized hCG, hCGbeta, and the beta core fragment. RESULTS: Differences in calibration of current hCG assays are moderate. Mean recovery of the current International Standard (IS), hCG IS 75/589, was 107% (range 93% to 126%), whereas that of the IRR 99/688 for hCG was 139% (range 109%-164%). Between-method variation for the latter (CV 12.3%) was also greater than for IS 75/589 (CV 8.8%). Recognition of hCGbeta varied markedly (CV 37%). Most assays overestimated it, but 2 RIAs produced results that were slight underestimations. Recognition of the beta core fragment was even more variable (CV 57%) and was closest to equimolarity for the RIAs. CONCLUSIONS: Assays for hCG show considerable variation in their recognition of various forms of hCG, and this variability is the most important cause of method-related differences in hCG results in serum and an even more important cause of method-related differences in urine measurements. Equimolar recognition of the major hCG isoforms is essential if between-method comparability for hCG is to be improved.


Subject(s)
Chorionic Gonadotropin/blood , Chorionic Gonadotropin/urine , Immunoassay/standards , Calibration , Chorionic Gonadotropin, beta Subunit, Human/blood , Chorionic Gonadotropin, beta Subunit, Human/urine , Female , Glycoprotein Hormones, alpha Subunit/blood , Glycoprotein Hormones, alpha Subunit/urine , Humans , Peptide Fragments/blood , Peptide Fragments/urine , Protein Isoforms/blood , Protein Isoforms/urine , Reference Standards , World Health Organization
18.
Biochemistry ; 47(6): 1708-20, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18197704

ABSTRACT

Follicle-stimulating hormone (FSH) glycosylation is regulated by feedback from the gonads, resulting in an array of glycans associated with FSH preparations derived from pools of pituitary or urine extracts. FSH glycosylation varies due to inhibition of FSHbeta N-glycosylation, elaboration of 1-4 branches possessed by mature N-glycans, and the number and linkage of terminal sialic acid residues. To characterize FSH glycosylation, FSH isoforms in pituitary gland extracts and a variety of physiological fluids are commonly separated by chromatofocusing. Variations in the ratios of immunological and biological activities in the resulting FSH isoform preparations are generally attributed to changes in glycosylation, which are most often defined in terms of sialic acid content. Using Western blotting to assess human FSHbeta glycosylation inhibition revealed 30-47% nonglycosylated hFSHbeta associated with four of six hFSH isoform preparations derived by chromatofocusing. Glycopeptide mass spectrometry assessment of glycan branching in these isoforms extensively characterized two N-glycosylation sites, one at alphaAsn52, the critical glycan for FSH function, and the other at betaAsn24. With two to four N-glycans per FSH molecule, many combinations of charges distributed over these sites can provide the same isoelectric point. Indeed, several glycans were common to all isoform fractions that were analyzed. There was no trend showing predominantly monoantennary glycans associated with the high-pI fractions, nor were predominantly tri- and tetra-antennary glycans associated with low-pI fractions. Thus, differences in receptor binding activity could not be associated with any specific glycan type or location in the hormone. FSH aggregation was associated with reduced receptor binding activity but did not affect immunological activity. However, as gel filtration indicated sufficient heterodimer was present in each isoform preparation to generate complete inhibition curves, the near total loss of receptor binding activity in several preparations could not be explained by aggregation alone, and the mechanism remains unknown.


Subject(s)
Follicle Stimulating Hormone/isolation & purification , Polysaccharides/chemistry , Protein Isoforms/isolation & purification , Animals , Blotting, Western , CHO Cells , Carbohydrate Conformation , Cricetinae , Cricetulus , Follicle Stimulating Hormone/chemistry , Humans , Mass Spectrometry , Protein Isoforms/chemistry , Radioimmunoassay , Rats
19.
Nat Clin Pract Endocrinol Metab ; 4(1): 22-32, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18084343

ABSTRACT

Medullary thyroid carcinoma (MTC) accounts for up to 8% of all thyroid cancers. Although primary surgery is curative in the vast majority of patients treated at an early stage, disease can persist or recur with deleterious effects on quality of life. Local and distant metastases can occur and are the major causes of mortality. Reoperation, embolization, and perhaps radiotherapy can improve the outcome for some patients who are not cured by primary surgery, but there is a need for novel treatments. No comprehensive clinical trial data are available on conventional cytotoxic agents for the treatment of MTC. Patients with distant metastases, in particular, might benefit from several novel compounds directed against angiogenesis and molecular targets in tumor cells, such as products of the proto-oncogene RET and mutants of it, and other signaling components. Well-conducted clinical trials are needed to assess and optimize these treatment strategies, and this article outlines how such trials should be conducted. Although RET mutations are common in hereditary MTC and can occur in some cases of sporadic MTC, knowledge of other molecular defects associated with the development of MTC should reveal new targets for therapy.


Subject(s)
Carcinoma, Medullary/therapy , Thyroid Neoplasms/therapy , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Medullary/genetics , Combined Modality Therapy/trends , Humans , Proto-Oncogene Mas , Thyroid Neoplasms/genetics , Thyroidectomy
20.
Bull Cancer ; 94 Spec No: S254-9, 2007 Jul.
Article in French | MEDLINE | ID: mdl-17846012

ABSTRACT

Tumor vessel formation is largely dependent on the recruitment of endothelial cells. Rare in healthy individuals, circulating endothelial cells (CEC) are shed from vessel walls and enter the circulation reflecting endothelial damage or dysfunction. Increased numbers of CEC have been documented in different types of cancer. Recent studies have suggested the role for CEC in tumor angiogenesis, but whose presence could also reflect normal endothelium perturbation in cancer. Originating from the bone marrow rather than from vessel walls, endothelial progenitor cells (EPC) are mobilized following tissue ischemia and may be recruited to complement local angiogenesis supplied by existing endothelium. Recently, studies in mouse models suggest that the circulating fraction of endothelial progenitors (CEP) is involved in tumor angiogenesis but their contribution is less clear in humans. The detection of CEC and CEP is difficult and impeded by the rarity of these cells. They may have important clinical implication as novel biomarkers susceptible to predict more efficiently and rapidly the therapeutic response to anti-angiogenic treatments. However, a methodological consensus would be necessary in order to correctly evaluate the clinical interest of CEC and CEP in patients.


Subject(s)
Endothelial Cells/cytology , Endothelium, Vascular/cytology , Neoplasms/blood supply , Neovascularization, Pathologic/etiology , Stem Cells/cytology , Angiogenesis Inhibitors/therapeutic use , Biomarkers/metabolism , Endothelial Cells/metabolism , Humans , Neoplasms/blood , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/drug therapy , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...