Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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.

4.
Cancer Res ; 73(7): 2271-80, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23554447

ABSTRACT

Non-small cell lung carcinoma patients are frequently treated with cisplatin (CDDP), most often yielding temporary clinical responses. Here, we show that PARP1 is highly expressed and constitutively hyperactivated in a majority of human CDDP-resistant cancer cells of distinct histologic origin. Cells manifesting elevated intracellular levels of poly(ADP-ribosyl)ated proteins (PAR(high)) responded to pharmacologic PARP inhibitors as well as to PARP1-targeting siRNAs by initiating a DNA damage response that translated into cell death following the activation of the intrinsic pathway of apoptosis. Moreover, PARP1-overexpressing tumor cells and xenografts displayed elevated levels of PAR, which predicted the response to PARP inhibitors in vitro and in vivo more accurately than PARP1 expression itself. Thus, a majority of CDDP-resistant cancer cells appear to develop a dependency to PARP1, becoming susceptible to PARP inhibitor-induced apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Lung Neoplasms/pathology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/drug effects , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Nude , Phenanthrenes/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Science ; 337(6102): 1678-84, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-23019653

ABSTRACT

Cancer cells accommodate multiple genetic and epigenetic alterations that initially activate intrinsic (cell-autonomous) and extrinsic (immune-mediated) oncosuppressive mechanisms. Only once these barriers to oncogenesis have been overcome can malignant growth proceed unrestrained. Tetraploidization can contribute to oncogenesis because hyperploid cells are genomically unstable. We report that hyperploid cancer cells become immunogenic because of a constitutive endoplasmic reticulum stress response resulting in the aberrant cell surface exposure of calreticulin. Hyperploid, calreticulin-exposing cancer cells readily proliferated in immunodeficient mice and conserved their increased DNA content. In contrast, hyperploid cells injected into immunocompetent mice generated tumors only after a delay, and such tumors exhibited reduced DNA content, endoplasmic reticulum stress, and calreticulin exposure. Our results unveil an immunosurveillance system that imposes immunoselection against hyperploidy in carcinogen- and oncogene-induced cancers.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Immunologic Surveillance , Neoplasms/genetics , Neoplasms/immunology , Ploidies , Animals , Calreticulin/immunology , Cell Line, Tumor , Common Variable Immunodeficiency/genetics , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Eukaryotic Initiation Factor-2/metabolism , Humans , Immunocompetence , Mice , Mice, Inbred BALB C , Neoplasms/chemically induced , Phosphorylation
6.
PLoS One ; 6(7): e22280, 2011.
Article in English | MEDLINE | ID: mdl-21814573

ABSTRACT

The Wnt/ß-catenin signaling pathway is involved in the normal development of thyroid gland, but its disregulation provokes the appearance of several types of cancers, including papillary thyroid carcinomas (PTC) which are the most common thyroid tumours. The follow-up of PTC patients is based on the monitoring of serum thyroglobulin levels which is regulated by the thyroid transcription factor 1 (TTF-1): a tissue-specific transcription factor essential for the differentiation of the thyroid. We investigated whether the Wnt/ß-catenin pathway might regulate TTF-1 expression in a human PTC model and examined the molecular mechanisms underlying this regulation. Immunofluorescence analysis, real time RT-PCR and Western blot studies revealed that TTF-1 as well as the major Wnt pathway components are co-expressed in TPC-1 cells and human PTC tumours. Knocking-down the Wnt/ß-catenin components by siRNAs inhibited both TTF-1 transcript and protein expression, while mimicking the activation of Wnt signaling by lithium chloride induced TTF-1 gene and protein expression. Functional promoter studies and ChIP analysis showed that the Wnt/ß-catenin pathway exerts its effect by means of the binding of ß-catenin to TCF/LEF transcription factors on the level of an active TCF/LEF response element at [-798, -792 bp] in TTF-1 promoter. In conclusion, we demonstrated that the Wnt/ß-catenin pathway is a direct and forward driver of the TTF-1 expression. The localization of TCF-4 and TTF-1 in the same area of PTC tissues might be of clinical relevance, and justifies further examination of these factors in the papillary thyroid cancers follow-up.


Subject(s)
Carcinoma, Papillary/metabolism , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Thyroid Neoplasms/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Blotting, Western , Carcinoma, Papillary/genetics , Cell Differentiation , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Humans , Immunoenzyme Techniques , Lithium Chloride/pharmacology , Luciferases/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Thyroid Neoplasms/genetics , Transcription Factors , Tumor Cells, Cultured , Wnt Proteins/genetics , beta Catenin/genetics
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.
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
9.
Biochimie ; 93(3): 457-68, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21075166

ABSTRACT

Nox4, a member of Nox family of NADPH oxidase expressed in nonphagocytic cells, is a major source of reactive oxygen species in many cell types. But understanding of the role of Nox4 in the production of ROS and of regulation mechanism of oxidase activity is largely unknown. This study reports for the first time the generation and characterization of 5 mAbs against a recombinant Nox4 protein (AA: 206-578). Among 5 novel mAbs, 3 mAbs (8E9, 5F9, 6B11) specifically recognized Nox4 protein in HEK293 transfected cells or human kidney cortex by western blot analysis; mAb 8E9 reacted with intact tet-induced T-REx™ Nox4 cells in FACS studies. The other 2 mAbs 10B4 and 7C9 were shown to have a very weak reactivity after purification. Immunofluorescence confocal microscopy showed that Nox4 localized not only in the perinuclear and endoplasmic reticulum regions but also at the plasma membrane of the cells which was further confirmed by TIRF-microscopy. Epitope determination showed that mAb 8E9 recognizes a region on the last extracellular loop of Nox4, while mAbs 6B11 and 5F9 are directed to its cytosolic tail. Contrary to mAb 6B11, mAb 5F9 failed to detect Nox4 at the plasma membrane. Cell-free oxidase assays demonstrated a moderate but significant inhibition of constitutive Nox4 activity by mAbs 5F9 and 6B11. In conclusion, 5 mAbs raised against Nox4 were generated for the first time. 3 of them will provide powerful tools for a structure/function relationship of Nox4 and for physiopathological investigations in humans.


Subject(s)
Antibodies, Monoclonal/immunology , Intracellular Space/metabolism , NADPH Oxidases/immunology , NADPH Oxidases/metabolism , Animals , Epitope Mapping , HEK293 Cells , Humans , Intracellular Space/enzymology , Mice , Mice, Inbred BALB C , NADPH Oxidase 4 , NADPH Oxidases/genetics , Peptide Library , Protein Transport , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Deletion
10.
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
11.
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
12.
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
13.
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
14.
Am J Clin Oncol ; 32(1): 1-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19194115

ABSTRACT

OBJECTIVE: Radiation exposure during childhood is the only well-established risk factor for papillary thyroid carcinoma (PTC). To better define the biologic profile of radiation-induced and sporadic PTC, we compared in these two groups of PTC the expression of cell cycle regulatory proteins and telomere length. METHODS: Cell cycle markers (cyclin A, B1, D1, E, and Ki67) were evaluated on 100 PTC specimens (26 radiation-induced and 74 sporadic PTCs). The expression of cell cycle regulators was studied using immunohistochemistry; telomere length heterogeneity was studied using in situ hybridization in a subset of 16 formalin-fixed samples (8 radiation-induced and 8 sporadic PTCs). RESULTS: At multivariate analysis, only cytoplasmic cyclin E staining was overexpressed in sporadic cases (P = 0.006). The other cell cycle markers and telomere length did not differ significantly between sporadic PTC and radiation-induced PTC. CONCLUSIONS: These markers cannot be used to differentiate radiation-induced from sporadic PTCs.


Subject(s)
Carcinoma, Papillary/metabolism , Cell Cycle Proteins/metabolism , Neoplasms, Radiation-Induced/metabolism , Telomere/metabolism , Thyroid Neoplasms/metabolism , Adenocarcinoma, Follicular/metabolism , Adenocarcinoma, Follicular/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Papillary/pathology , Case-Control Studies , Child , Child, Preschool , Female , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Infant , Male , Middle Aged , Neoplasms, Radiation-Induced/pathology , Prognosis , Thyroid Neoplasms/pathology , Tissue Array Analysis , Young Adult
15.
Thyroid ; 17(7): 639-46, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17696834

ABSTRACT

OBJECTIVE: Poorly differentiated follicular thyroid carcinoma (PDFC) is a tumor of follicular cell origin with attributes intermediate between well-differentiated carcinomas and anaplastic carcinomas, but neither a clear histological description nor an established definition of prognostic indicators are available. DESIGN: This study correlates the clinical outcome and survival of 40 PDFC patients with histological architecture, cytological characteristics, and expression of various markers of cell proliferation and differentiation (cyclin A, cyclin B1, cyclin D1, cyclin E, Ki67, thyroperoxidase, galectin 3, dual oxidase [Duox], vascular endothelial growth factor, epidermal growth factor receptor, and p53). MAIN OUTCOME: At 5 years, the overall survival rate was 63% and the metastasis-free survival rate was 57%. An older age at the time of diagnosis and a larger tumor size were associated with an increased risk of distant metastases and of cancer-related death. Polymorph architecture was associated with a reduced risk of metastases, whereas a high expression of Duox was associated with a reduced risk of death. In these patients with PDFC, no other histological features or expression of any other marker had a prognostic significance. CONCLUSION: PDFC has a more aggressive behavior than well-differentiated carcinomas; prognosis is related to indicators that are also relevant in patients with well-differentiated carcinomas.


Subject(s)
Adenocarcinoma, Follicular/pathology , Thyroid Neoplasms/pathology , Adenocarcinoma, Follicular/classification , Adenocarcinoma, Follicular/mortality , Adult , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Software , Survival Analysis , Survivors , Thyroid Neoplasms/classification , Thyroid Neoplasms/mortality
16.
Thyroid ; 17(3): 203-11, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17381352

ABSTRACT

Immunohistochemistry provides insights in the expression of functional proteins and of their localization in normal thyroid tissue and in thyroid diseases. In hyperfunctional thyroid tissues, staining for sodium/iodide symporter (NIS), pendrin, thyroid peroxidase (TPO), and thyroglobulin (Tg) is increased. In hypofunctioning thyroid tissues, NIS staining is markedly decreased; in benign hypofunctioning adenomas, the expression of the other functional proteins is unmodified or slightly decreased, whereas their expression is profoundly decreased or absent in differentiated thyroid carcinoma.


Subject(s)
Immunohistochemistry/methods , Iodide Peroxidase/metabolism , Membrane Transport Proteins/metabolism , Symporters/metabolism , Thyroglobulin/metabolism , Thyroid Gland/pathology , Biomarkers/chemistry , Carcinoma/metabolism , Epitopes/chemistry , Humans , Neoplasm Metastasis , Prognosis , Sulfate Transporters , Thyroid Neoplasms/metabolism
17.
J Clin Endocrinol Metab ; 92(1): 70-6, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17077129

ABSTRACT

CONTEXT: Terminal differentiation of the human thyroid is characterized by the onset of follicle formation and thyroid hormone synthesis at 11 gestational weeks (GW). OBJECTIVE: This study aimed to investigate the ontogeny of thyroglobulin (Tg), thyroid peroxidase (TPO), sodium/iodide symporter (NIS), pendrin (PDS), dual oxidase 2 (DUOX2), thyroid-stimulating hormone receptor (TSHR), and thyroid transcription factor 1 (TITF1), forkhead box E1 (FOXE1), and paired box gene 8 (PAX8) in the developing human thyroid. DESIGN: Thyroid tissues from human embryos and fetuses (7-33 GW; n = 45) were analyzed by quantitative PCR to monitor mRNA expression for each gene and by immunohistochemistry to determine the cellular distribution of TITF1, TSHR, Tg, TPO, NIS, and the onset of T4 production. A broken line regression model was fitted for each gene to compare the loglinear increase in expression before and after the onset of T4 synthesis. RESULTS: TITF1, FOXE1, PAX8, TSHR, and DUOX2 were stably expressed from 7 to 33 GW. Tg, TPO, and PDS expression was detectable as early as 7 GW and was correlated with gestational age (all, P < 0.01), and the slope of the regression line was significantly different before and after the onset of T4 synthesis at 11 GW (all, P < 0.01). NIS expression appeared last and showed the highest fit by the broken line regression model of all genes (correlation age P < 0.0001, broken line regression P < 0.0001). Immunohistochemical studies detected TITF1, TSHR, and Tg in unpolarized thyrocytes before follicle formation. T(4) and NIS labeling were only found in developing follicles from 11 GW on. CONCLUSION: These results imply a key role of NIS for the onset of human thyroid function.


Subject(s)
Fetus/metabolism , Symporters/genetics , Thyroid Gland/embryology , Female , Gestational Age , Humans , Immunohistochemistry , Iodide Peroxidase/analysis , Iodide Peroxidase/genetics , Pregnancy , RNA, Messenger/analysis , Receptors, Thyrotropin/analysis , Symporters/analysis , Thyroglobulin/analysis , Thyroglobulin/genetics
18.
Am J Pathol ; 167(1): 223-31, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15972966

ABSTRACT

Follicular thyroid carcinomas (FTC) arise through oncogenic pathways distinct from those involved in the papillary histotype. Recently, a t(2;3)(q13;p25) rearrangement, which juxtaposes the thyroid transcription factor PAX8 to the peroxisome proliferator-activated receptor (PPAR) gamma1, was described in FTCs. In this report, we describe gene expression in 11 normal tissues, 4 adenomas, and 8 FTCs, with or without the PAX8-PPARgamma1 translocation, using custom 60-mer oligonucleotide microarrays. Results were confirmed by quantitative real-time polymerase chain reaction of 65 thyroid tissues and by immunohistochemistry. Statistical analysis revealed a pattern of 93 genes discriminating FTCs, with or without the translocation, that were morphologically undistinguishable. Although the expression of thyroid-specific genes was detectable, none appeared to be differentially regulated between tumors with or without the translocation. Differentially expressed genes included genes related to lipid/glucose/amino acid metabolism, tumorigenesis, and angiogenesis. Surprisingly, several PPARgamma target genes were up-regulated in PAX8-PPARgamma-positive FTCs such as angiopoietin-like 4 and aquaporin 7. Moreover many genes involved in PAX8-PPARgamma expression profile presented a putative PPARgamma-promoter site, compatible with a direct activity of the fusion product. These data identify several differentially expressed genes, such as FGD3, that may serve as potential targets of PPARgamma and as members of novel molecular pathways involved in the development of thyroid carcinomas.


Subject(s)
Adenocarcinoma, Follicular/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , PPAR gamma/genetics , Thyroid Neoplasms/genetics , Trans-Activators/genetics , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , PAX8 Transcription Factor , Paired Box Transcription Factors , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic
19.
Am J Physiol Gastrointest Liver Physiol ; 288(5): G933-42, 2005 May.
Article in English | MEDLINE | ID: mdl-15591162

ABSTRACT

The dual oxidase (Duox)2 flavoprotein is strongly expressed in the thyroid gland, where it plays a critical role in the synthesis of thyroid hormones by providing thyroperoxidase with H2O2. DUOX2 mRNA was recently detected by RT-PCR and in-situ hybridization experiments in other tissues, such as rat colon and rat and human epithelial cells from the salivary excretory ducts and rectal glands. We examined Duox2 expression at the protein level throughout the porcine digestive tract and in human colon. Western blot analysis identified Duox2 as the same two molecular species (M(r) 165 and 175 kDa) as detected in the thyroid. It was expressed in all the tissues tested, but the highest levels were found in the cecum and sigmoidal colon. Immunohistochemical studies showed that Duox2 protein is mainly present in these parts of the gut and located at the apical membrane of the enterocytes in the brush border, indicating that it is expressed only in highly differentiated cells. A Ca2+/NADPH-dependent H2O2-generating system was associated with Duox2 protein expression, which had the same biochemical characteristics as the NADPH oxidase in the thyroid. Indeed, treatment of the thyroid and cecum particulate fractions with phenylarsine oxide resulted in complete calcium desensitization of both enzymes. A marked increase in DUOX2 expression was also found during spontaneous differentiation of postconfluent Caco-2 cells. The discovery of Duox2 as a novel source of H2O2 in the digestive tract, particularly in the cecum and colon, makes it a new candidate mediator of physiopathological processes.


Subject(s)
Flavoproteins/biosynthesis , Gastrointestinal Tract/enzymology , Animals , Caco-2 Cells , Colon/enzymology , Dual Oxidases , Duodenum/enzymology , Gene Expression , Humans , Hydrogen Peroxide/metabolism , Intestine, Small/enzymology , NADPH Oxidases , Swine , Thyroid Gland/enzymology
20.
Eur J Endocrinol ; 151(3): 367-74, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15362967

ABSTRACT

OBJECTIVE: Genetic alterations involving the thyroid transcription factor PAX8 and the peroxisome proliferator-activated receptor gamma 1 (PPARgamma1) genes have been described in thyroid neoplasms. We investigated in a series of thyroid samples, including 14 normal, 13 hyperfunctioning tissues, 26 follicular adenomas, 21 follicular and 41 papillary carcinomas, both the frequency of the PAX8-PPARgamma1 rearrangement and the expression of the PAX8 and PPARgamma transcripts. METHODS: Using RT-PCR followed by sequencing PCR products, PAX8-PPARgamma1 translocation was not detected in benign tissues nor in papillary carcinomas and was detected in 4 (19%) of 21 follicular carcinomas and in one (4%) of 26 follicular adenomas. RESULTS: Specific real-time quantitative RT-PCR (Q RT-PCR) methods detected high levels of PPARgamma transcripts in follicular carcinomas presenting the rearrangement. Interestingly, the level of PPARgamma transcripts was significantly decreased in papillary carcinomas in comparison with those found in benign adenomas and follicular carcinomas. Finally, PAX8 gene expression was decreased in both papillary and follicular thyroid carcinomas, and in these tumors to the same extent in the presence or absence of the rearrangement. These alterations in both PPARgamma and PAX8 gene expression may explain the poorly differentiated histotype of follicular carcinomas harboring the translocation. Immunohistochemistry showed that nuclear PPARgamma staining was weak in normal tissues, adenomas, papillary carcinomas and in some follicular carcinomas, and strong in the follicular carcinomas positive for the PAX8-PPARgamma1 translocation, but also in some follicular tumors in which no translocation could be evidenced. CONCLUSION: These observations confirm that the PAX8-PPARgamma1 translocation characterizes a subset of thyroid follicular carcinomas but is not a specific marker of carcinoma and that its frequency is lower than that initially reported. Finally, immunohistochemistry is not a reliable method for the specific detection of the translocation, that can be specifically evidenced by Q RT-PCR.


Subject(s)
Adenocarcinoma, Follicular/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Thyroid Neoplasms/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Translocation, Genetic , Adenocarcinoma, Follicular/physiopathology , Adult , Aged , Biomarkers, Tumor , Carcinoma, Papillary/genetics , Carcinoma, Papillary/physiopathology , Female , Humans , Male , Middle Aged , Neoplasms/genetics , Neoplasms/physiopathology , PAX8 Transcription Factor , Paired Box Transcription Factors , Polymorphism, Genetic , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...