Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Toxicol ; 5: 1294760, 2023.
Article in English | MEDLINE | ID: mdl-38098751

ABSTRACT

Several ecological studies suggest that ambient air pollution is associated with the occurrence of thyroid cancer. In this study, we used certified diesel particulate matter as a proxy for fine particulate matter. Human thyroid cancer cell lines 8505C and TPC-1 were incubated with different concentrations of NIST1650b for 5 days and subjected to functional assays. We found that NIST1650b treatment did not affect short-term cell growth but reduced colony formation at high concentrations. Notably, NIST1650b-treated cells showed altered morphology toward cluster coalescence following treatment. Wound healing assays revealed that leading-edge cells formed protruding tips while maintaining cell-cell adhesion, and a significantly higher ratio of wound closure following treatment at 10 µg/mL was seen in both cell lines. A weak stimulatory effect on transwell cell migration was observed in 8505C cells. Taken together, our results suggest that fine particulate matter induced a coherent phenotype accompanied by augmented collective cell migration in thyroid cancer cells.

2.
Anticancer Res ; 42(5): 2487-2493, 2022 May.
Article in English | MEDLINE | ID: mdl-35489723

ABSTRACT

BACKGROUND/AIM: Expression of sterol regulatory element-binding protein 1 (SREBP1) is upregulated in thyroid cancer and associated with shorter disease-specific survival. The molecular regulatory mechanisms governing SREBP1 over-expression in thyroid cancer are still unclear. MATERIALS AND METHODS: Thyroid cancer cell lines BHT-101 (with the BRAF V600E mutation) and FTC-131 (wild-type for BRAF) were treated with specific inhibitors. The expression of SREBP1 was determined at the mRNA level using quantitative real-time PCR and at the protein level using immunoblotting. RESULTS: Lenvatinib and a MEK inhibitor, selumetinib, suppressed SREBP1 expression in BHT-101 but not FTC-133 cells. Olitigaltin, a galectin-3 inhibitor, decreased SREBP1 expression in a time- and dose-dependent manner in both cells. MK2206, an allosteric AKT inhibitor, did not change SREBP1 expression in either cell line. CONCLUSION: The galectin-3 inhibitor attenuates SREBP1 expression in thyroid cancer cells, likely independent of AKT phosphorylation. Lenvatinib and selumetinib decreases SREBP1 expression in the BRAF-mutant cell line BHT-101.


Subject(s)
Proto-Oncogene Proteins B-raf , Sterol Regulatory Element Binding Protein 1 , Thyroid Neoplasms , Humans , Cell Proliferation , Galectin 3 , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism
3.
Endocr Relat Cancer ; 29(2): 47-58, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34821220

ABSTRACT

Aberrant lipid metabolism provides bioenergetic, biosynthetic, and redox supplies to cancer cells. Previous studies have reported differential lipid profiling in thyroid malignancies. Sterol regulatory element-binding protein 1 (SREBP1), encoded by the SREBF1 gene, is a master regulator of cellular lipid homeostasis. The clinical and functional significance of SREBP1 in thyroid cancer is not well understood. Here, we showed that SREBP1 expression is significantly upregulated in invasive thyroid cancer than in normal thyroid tissue or benign thyroid nodules. High tumoral SREBP1 expression was associated with extrathyroidal extension, advanced disease stage, and shorter disease-specific survival in patients with differentiated thyroid cancer. SREBP1 overexpression significantly increased the oxygen consumption rate, filopodia formation, and migratory and invasive capacities of thyroid cancer cells. Knockdown of SREBF1 or treatment with an SREBP1 activation inhibitor fatostatin had the opposite effect. RNA-Seq analysis showed that modulation of SREBP1 expression was accompanied by corresponding changes in the expression of epithelial-mesenchymal transition markers and CYR61/CTGF. SREBP1-facilitated cell invasion could be abrogated by treatment with a YAP inhibitor such as verteporfin or genetic silencing of CYR61 or CTGF. In summary, SREBP1 upregulation can be used as a prognostic indicator for thyroid cancer and SREBP1 overexpression is involved in cancer invasiveness, at least partly, through upregulation of CYR61/CTGF via the Hippo-YAP pathway.


Subject(s)
Epithelial-Mesenchymal Transition , Sterol Regulatory Element Binding Protein 1/metabolism , Thyroid Neoplasms , Cell Line, Tumor , Humans , Lipids/therapeutic use , Phenotype , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics , Thyroid Neoplasms/pathology
4.
Front Psychol ; 11: 573432, 2020.
Article in English | MEDLINE | ID: mdl-33192871

ABSTRACT

The study examines how the remote associates test (RAT) has been used to examine theories of creativity through a review of recent studies on creativity. Creativity-related studies published between 2000 and 2019 were retrieved from the SCOPUS database. A total of 172 papers were chosen for further analysis. Content analysis shows that research on creativity using RAT mainly concerns remote association, insight problem-solving, general creative process, test development, individual difference, effect of treatment, clinical case, social interaction effect, and predictor or criterion. The study constructs a theoretical framework based on the 4P (Product-Person-Process-Place) model and demonstrates how empirical studies using the RAT explore the individual differences, internal processes, and external influences of creative thinking. In addition, the most commonly used version of the RAT is the Compound Remote Associates Problems (Bowden and Jung-Beeman, 2003a). Current research shows a trend whereby the creative thinking process has been receiving greater attention. In particular, a growing number of studies in this field have been carried out using cognitive neuroscience technologies. These findings suggest that the RAT provides researchers with a way to deepen their understanding of different levels of creativity.

5.
Mol Cell Endocrinol ; 499: 110595, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31563469

ABSTRACT

The pentose phosphate pathway (PPP) plays an important role in the biosynthesis of ribonucleotide precursor and NADPH. Cancer cells frequently increase the flux of glucose into the PPP to support the anabolic demands and regulate oxidative stress. Consistently, metabolomic analyses indicate an upregulation of the PPP in thyroid cancer. In the present study, we found that the combination of glucose-6-phosphate dehydrogenase (G6PD) and transketolase inhibitors (6-aminonicotinamide and oxythiamine) exerted an additive or synergistic effect on cell growth inhibition in thyroid cancer cells. Targeting PPP significantly increased cellular reactive oxygen species (ROS) and induced endoplasmic reticulum (ER) stress and apoptosis. Suppressed cell viability could be partially rescued with treatment with the ROS scavenger or apoptosis inhibitor but not ER-stress inhibitor. Taken together, dual PPP blockade leads to pharmacologic additivity or synergism and causes ROS-mediated apoptosis in thyroid cancer cells.


Subject(s)
6-Aminonicotinamide/pharmacology , Oxythiamine/pharmacology , Pentose Phosphate Pathway/drug effects , Reactive Oxygen Species/metabolism , Thyroid Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucosephosphate Dehydrogenase , Humans , NADP/metabolism , Oxidative Stress/drug effects , Thyroid Neoplasms/drug therapy
6.
J Cancer ; 10(16): 3819-3829, 2019.
Article in English | MEDLINE | ID: mdl-31333799

ABSTRACT

Increased aerobic glycolysis portends an unfavorable prognosis in thyroid cancer. The metabolic reprogramming likely results from altered mitochondrial activity and may promote cancer progression. Peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) plays a pivotal role in mitochondrial biogenesis and function. In the present study, we aimed to evaluate the clinicopathological significance of PGC1α expression and the potential effects of PGC1α modulation. Firstly, the expression of PGC1α in thyroid cancer samples was evaluated using western blot analysis and immunohistochemical staining. Compared with normal thyroid tissue, PGC1α expression was downregulated in thyroid cancer. PGC1α-negative papillary cancer was associated with BRAF V600E mutation, large tumor size, extrathyroidal or lymphovascular invasion, lymph node metastasis, and advanced stage. The results were consistent with the analysis of The Cancer Genome Atlas data. PGC1α expression correlated with oxygen consumption in thyroid cancer cells and was inversely related to AKT activity. The biologic relevance of PGC1α was further investigated by gain- and loss-of-function experimental studies. PGC1α overexpression led to augmented oxidative metabolism and accelerated tumor growth, whereas PGC1α knockdown induced a glycolytic phenotype but reduced tumor growth in vivo. In conclusion, PGC1α downregulation is associated with glycolytic metabolism and advanced disease in thyroid cancer. Nonetheless, manipulating PGC1α expression and metabolic phenotype does not necessarily translate into beneficial effects. It suggests that the metabolic phenotype is likely the consequence rather than the cause of disease progression in thyroid cancer.

7.
Int J Mol Sci ; 19(9)2018 Aug 24.
Article in English | MEDLINE | ID: mdl-30149527

ABSTRACT

Heme oxygenase-1 (HO-1) is induced by a variety of stimuli and plays a multifaceted role in cellular protection. We have shown that HO-1 is overexpressed in thyroid cancer and is associated with tumor aggressiveness. Therefore, we set out to assess the effects of HO-1 inhibitors on the biology of thyroid cancer cells. Two different classes of HO-1 inhibitors were used, including a metalloporphyrin, zinc protoporphyrin-IX (ZnPP), and an azole antifungal agent, ketoconazole. The viability and colony formation of thyroid cancer cells decreased in a concentration- and time-dependent fashion following treatment with HO-1 inhibitors. Cancer cells exhibited a higher sensitivity to HO-1 inhibitors than non-malignant cells. HO-1 inhibitors induced a G0/G1 arrest accompanied by decreased cyclin D1 and CDK4 expressions and an increase in levels of p21 and p27. HO-1 inhibitors significantly increased intracellular ROS levels and suppressed cell migration and invasion. Oxygen consumption rate and mitochondrial mass were increased with ZnPP treatment. Mice treated with ZnPP had a reduced xenograft growth and diminished cyclin D1 and Ki-67 staining in tumor sections. Taken together, HO-1 inhibitors might have therapeutic potential for inducing cell cycle arrest and promoting growth suppression of thyroid cancer cells in vitro and in vivo.


Subject(s)
Cell Cycle Checkpoints/drug effects , Enzyme Inhibitors/pharmacology , Heme Oxygenase-1/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Female , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Heterografts , Humans , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Neoplastic Stem Cells , Oxygen Consumption/drug effects , Reactive Oxygen Species/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism
8.
Cancer Lett ; 370(1): 56-65, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26454215

ABSTRACT

Cyproheptadine, a serotonin antagonist, has recently been reported to function as a novel therapeutic agent by inhibiting PI3K/AKT signaling in several human cancers. However, the therapeutic effect of cyproheptadine in urothelial carcinoma (UC) has never been explored. In this study, we determined the effect of cyproheptadine on the growth of five human UC cell lines and an in vivo xenograft model. The results showed that cyproheptadine exerted an inhibitory effect on the proliferation of UC cells both in vitro and in vivo. Cyproheptadine also induced cell cycle arrest in the G1 phase, subsequently followed by apoptosis and necrosis. The underlying mechanisms of cell cycle arrest were associated with the reduction of c-Myc, induction of p21 and p27, and the stabilization of Rb expression. In addition, the suppression of the GSK3ß/TSC2/mTOR pathway and deregulation of the GSK3ß/ß-catenin signaling were observed in cyproheptadine-treated UC cells. Furthermore, cyproheptadine-induced apoptosis was associated with ANGPTL4 expression followed by activation of caspase3 and PARP in UC cells. Our experimental results provide evidence that cyproheptadine is a suitable therapeutic agent for the treatment of UC.


Subject(s)
Antineoplastic Agents/pharmacology , Cyproheptadine/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , beta Catenin/antagonists & inhibitors , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Animals , Apoptosis/drug effects , Cell Line, Tumor , G1 Phase Cell Cycle Checkpoints/drug effects , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Inbred BALB C , Receptors, Serotonin/analysis , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/physiology , Urinary Bladder Neoplasms/pathology , beta Catenin/physiology
9.
BMC Bioinformatics ; 15: 381, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25431302

ABSTRACT

BACKGROUND: Next generation sequencing technology has allowed efficient production of draft genomes for many organisms of interest. However, most draft genomes are just collections of independent contigs, whose relative positions and orientations along the genome being sequenced are unknown. Although several tools have been developed to order and orient the contigs of draft genomes, more accurate tools are still needed. RESULTS: In this study, we present a novel reference-based contig assembly (or scaffolding) tool, named as CAR, that can efficiently and more accurately order and orient the contigs of a prokaryotic draft genome based on a reference genome of a related organism. Given a set of contigs in multi-FASTA format and a reference genome in FASTA format, CAR can output a list of scaffolds, each of which is a set of ordered and oriented contigs. For validation, we have tested CAR on a real dataset composed of several prokaryotic genomes and also compared its performance with several other reference-based contig assembly tools. Consequently, our experimental results have shown that CAR indeed performs better than all these other reference-based contig assembly tools in terms of sensitivity, precision and genome coverage. CONCLUSIONS: CAR serves as an efficient tool that can more accurately order and orient the contigs of a prokaryotic draft genome based on a reference genome. The web server of CAR is freely available at http://genome.cs.nthu.edu.tw/CAR/ and its stand-alone program can also be downloaded from the same website.


Subject(s)
Chromosome Mapping/methods , Computational Biology/methods , Contig Mapping/methods , Gene Rearrangement , Genome , Sequence Analysis, DNA/methods , Software , Algorithms , Prokaryotic Cells
10.
PLoS One ; 9(2): e89563, 2014.
Article in English | MEDLINE | ID: mdl-24586874

ABSTRACT

Local anesthetics are frequently used in fine-needle aspiration of thyroid lesions and locoregional control of persistent or recurrent thyroid cancer. Recent evidence suggests that local anesthetics have a broad spectrum of effects including inhibition of cell proliferation and induction of apoptosis in neuronal and other types of cells. In this study, we demonstrated that treatment with lidocaine and bupivacaine resulted in decreased cell viability and colony formation of both 8505C and K1 cells in a dose-dependent manner. Lidocaine and bupivacaine induced apoptosis, and necrosis in high concentrations, as determined by flow cytometry. Lidocaine and bupivacaine caused disruption of mitochondrial membrane potential and release of cytochrome c, accompanied by activation of caspase 3 and 7, PARP cleavage, and induction of a higher ratio of Bax/Bcl-2. Based on microarray and pathway analysis, apoptosis is the prominent transcriptional change common to lidocaine and bupivacaine treatment. Furthermore, lidocaine and bupivacaine attenuated extracellular signal-regulated kinase 1/2 (ERK1/2) activity and induced activation of p38 mitogen-activated protein kinase (MAPK) and c-jun N-terminal kinase. Pharmacological inhibitors of MAPK/ERK kinase and p38 MAPK suppressed caspase 3 activation and PARP cleavage. Taken together, our results for the first time demonstrate the cytotoxic effects of local anesthetics on thyroid cancer cells and implicate the MAPK pathways as an important mechanism. Our findings have potential clinical relevance in that the use of local anesthetics may confer previously unrecognized benefits in the management of patients with thyroid cancer.


Subject(s)
Anesthetics, Local/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bupivacaine/pharmacology , Lidocaine/pharmacology , MAP Kinase Signaling System/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Proliferation/drug effects , Cytochromes c/metabolism , Drug Screening Assays, Antitumor , Enzyme Activation , Gene Expression/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Thyroid Neoplasms
11.
Cell Biochem Biophys ; 65(2): 203-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22907586

ABSTRACT

Obesity is associated with a higher incidence of thyroid cancer. Adiponectin is one of the most abundant adipokines with a pleiotropic role in metabolism and in the development and progression of cancer. It has been shown that circulating adiponectin level is inversely associated with the risk of thyroid cancer. This study aimed to investigate the possible association between the expression of adiponectin receptors (AdipoR1 and AdipoR2) and clinicopathological variables in papillary thyroid cancer. We found that protein levels of AdipoR1 and AdipoR2 were increased in some thyroid cancer specimens compared with adjacent normal thyroid tissues. Thyroid cancer cells expressed AdipoR1 and AdipoR2, which were attenuated by histone deacetylase inhibitors valproic acid and trichostatin A. Adiponectin stimulated AMP-activated protein kinase phosphorylation in thyroid cancer cells. We further determined the expression of AdipoR1 and AdipoR2 by immunohistochemical staining in primary tumor samples and metastatic lymph nodes. AdipoR1 was expressed in 27 % of primary tumors and AdipoR2 in 47 %. Negative expression of both adiponectin receptors was significantly associated with extrathyroidal invasion, multicentricity, and higher TNM stage. There was a trend toward decreased disease-free survival in patients with negative tumor expression of AdipoR1 and AdipoR2 (log-rank P = 0.051). Collectively, overexpression of adiponectin receptors was observed in some tumor tissues of papillary thyroid cancer and was associated with a better prognosis.


Subject(s)
Carcinoma, Papillary/metabolism , Receptors, Adiponectin/metabolism , Thyroid Neoplasms/metabolism , AMP-Activated Protein Kinases/metabolism , Adiponectin/pharmacology , Adult , Blotting, Western , Carcinoma, Papillary/pathology , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Immunohistochemistry , Leptin/metabolism , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Phosphorylation/drug effects , Receptors, Leptin/metabolism , Thyroid Neoplasms/pathology
12.
Biomed Pharmacother ; 66(6): 469-73, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22560341

ABSTRACT

Epidemiological studies suggest an important link between obesity and thyroid cancer. The adipose tissue-derived polypeptide leptin acting via leptin receptor may modulate cell migration of thyroid cancer cells. Previously we have demonstrated that leptin receptor is overexpressed in papillary thyroid cancer and is associated with tumor aggressiveness. The present study was undertaken to explore the possible regulatory factors which would influence leptin receptor expression in papillary thyroid cancer cells. We found that DNA methyltransferase inhibitor (5-Aza-2'-deoxycytidine) and histone deacetylase inhibitor (trichostatin A) reduced leptin receptor expression. Conversely, insulin upregulated leptin receptor expression in a time- and dose-dependent manner. Hypoxia-mimicking agent (cobalt chloride) had no effect on leptin receptor expression. Taken together, our study provides evidence that epigenetic events and insulin stimulation take part in regulation of leptin receptor expression in papillary thyroid cancer cells.


Subject(s)
Carcinoma/metabolism , DNA Methylation , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Receptors, Leptin/metabolism , Thyroid Neoplasms/metabolism , Blotting, Western , Carcinoma/genetics , Carcinoma, Papillary , Cell Hypoxia , Cell Line, Tumor , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Osmolar Concentration , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Up-Regulation/drug effects
13.
Oncol Rep ; 26(5): 1265-71, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21750869

ABSTRACT

The incidence of thyroid cancer has remarkably increased in recent years. Epidemiologic data suggest that obesity is associated with an increased incidence of several types of malignancies, including thyroid cancer. Leptin, an adipocyte-derived cytokine, has been shown to be involved in cancer development and progression. We previously demonstrated that papillary thyroid cancer expressing leptin receptor and/or leptin has a higher incidence of lymph node metastasis. In this study, we investigated the effects of leptin on cell migration in K1 and B-CPAP papillary thyroid cancer cells. Expression of leptin receptor was observed in both cell lines. Leptin enhanced the migratory activity significantly in a dose-dependent manner. We showed that leptin induced AKT and extracellular signal-regulated kinase (ERK) phosphorylation. Inhibition of phosphatidylinositol 3-kinase and ERK activation using pharmacological inhibitors effectively blocked leptin-induced migration of K1 and B-CPAP cells. Taken together, this study provides new mechanistic evidence for a role of leptin in the regulation of papillary thyroid cancer progression by stimulating tumor cell migration.


Subject(s)
Carcinoma, Papillary/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Leptin/pharmacology , MAP Kinase Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thyroid Neoplasms/enzymology , Carcinoma , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Humans , Immunohistochemistry , Leptin/metabolism , MAP Kinase Signaling System , Phosphorylation , Receptors, Leptin/biosynthesis , Signal Transduction/drug effects , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
14.
Oncol Rep ; 23(6): 1721-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20428831

ABSTRACT

Excess body weight is associated with a moderately increased risk of thyroid cancer. Adipocyte-derived hormone, leptin, has been shown to enhance cell growth and migration in many cancer types. Limited evidence suggests that leptin has direct actions on the thyroid gland, but there are no data available on the effect of leptin on thyroid cancer cells. We evaluated the action of leptin on gene expression, cell growth, cell cycle, and cell migration in anaplastic (ARO), follicular (WRO) and papillary (CGTH-W3) thyroid carcinoma cell lines. Expression of long-form leptin receptors was observed in all thyroid cancer cell lines. Leptin stimulation did not alter the expression levels of leptin, leptin receptor and sodium-iodide symporter. Cell growth and cell cycle were not changed after leptin treatment. However, leptin was able to promote cell migration of papillary thyroid cancer cells, but inhibited migration of anaplastic and follicular cancer cells. In summary, our study suggests that leptin modulates cell migration of thyroid cancer cells in a cell type-specific manner.


Subject(s)
Adenocarcinoma, Follicular/metabolism , Carcinoma, Papillary/metabolism , Carcinoma/metabolism , Cell Movement , Leptin/metabolism , Thyroid Neoplasms/metabolism , Adenocarcinoma, Follicular/pathology , Apoptosis , Blotting, Western , Carcinoma/pathology , Carcinoma, Papillary/pathology , Cell Cycle , Cell Proliferation , Humans , RNA, Messenger/genetics , Receptors, Leptin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...