Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Mol Carcinog ; 57(1): 97-105, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28891089

ABSTRACT

The regulation of cancer-relevant genes by the thyroid hormones, 3, 5, 3'-Triiodo-L-thyronine (T3) and L-thyroxine (T4), was recently acknowledged. However, limited data exists on the hormonal effects on gene expression in ovarian cancer, a gynecological malignancy associated with a low cure rate. The expression of fifteen genes involved in DNA repair, cell cycle, apoptosis, and tumor suppression was evaluated in OVCAR-3 and A2780 cell lines, using real-time PCR following short incubation with T3 (1 nM) or T4 (100 nM). The thyroid hormones downregulated the expression of the majority of genes examined. Support for the involvement of the MAPK and PI3K in thyroid hormone-mediated gene expression was shown for a set of genes. FAS expression was inhibited in A2780 cells, while an unexpected induction was demonstrated in OVCAR-3 cells. An analogous effect on the protein levels of FAS receptor and its soluble form was demonstrated by Western blotting. We further established, using primer sets that discriminate between the different RNA isoforms, that the hormones increase the mRNA levels of both coding and non-coding FAS mRNAs. The prevalence of these isoforms, using The Cancer Genome Atlas (TCGA) analysis, was significantly more abundant in 17 cancer types, including ovarian cancer, compared to normal tissues. Our results highlight the role of thyroid hormones in the expression of cancer-relevant-genes in ovarian cancer and provide an important insight into the pathways by which mitogenic and anti-apoptotic effects are exerted.


Subject(s)
Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Thyroxine/pharmacology , Triiodothyronine/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Cell Line, Tumor , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , fas Receptor/genetics , fas Receptor/metabolism
2.
Mol Oncol ; 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38429887

ABSTRACT

The selenoenzyme type I iodothyronine deiodinase (DIO1) catalyzes removal of iodine atoms from thyroid hormones. Although DIO1 action is reported to be disturbed in several malignancies, no work has been conducted in high-grade serous ovarian carcinoma (HGSOC), the most lethal gynecologic cancer. We studied DIO1 expression in HGSOC patients [The Cancer Genome Atlas (TCGA) data and tumor tissues], human cell lines (ES-2 and Kuramochi), normal Chinese hamster ovarian cells (CHO-K1), and normal human fallopian tube cells (FT282 and FT109). To study its functional role, DIO1 was overexpressed, inhibited [by propylthiouracil (PTU)], or knocked down (KD), and cell count, proliferation, apoptosis, cell viability, and proteomics analysis were performed. Lower DIO1 levels were observed in HGSOC compared to normal cells and tissues. TCGA analyses confirmed that low DIO1 mRNA expression correlated with worse survival and therapy resistance in patients. Silencing or inhibiting the enzyme led to enhanced ovarian cancer proliferation, while an opposite effect was shown following DIO1 ectopic expression. Proteomics analysis in DIO1-KD cells revealed global changes in proteins that facilitate tumor metabolism and progression. In conclusion, DIO1 expression and ovarian cancer progression are inversely correlated, highlighting a tumor suppressive role for this enzyme and its potential use as a biomarker in this disease.

3.
Oncogene ; 40(44): 6248-6257, 2021 11.
Article in English | MEDLINE | ID: mdl-34556811

ABSTRACT

The enzyme iodothyronine deiodinase type 3 (DIO3) contributes to cancer proliferation by inactivating the tumor-suppressive actions of thyroid hormone (T3). We recently established DIO3 involvement in the progression of high-grade serous ovarian cancer (HGSOC). Here we provide a link between high DIO3 expression and lower survival in patients, similar to common disease markers such as Ki67, PAX8, CA-125, and CCNE1. These observations suggest that DIO3 is a logical target for inhibition. Using a DIO3 mimic, we developed original DIO3 inhibitors that contain a core of dibromomaleic anhydride (DBRMD) as scaffold. Two compounds, PBENZ-DBRMD and ITYR-DBRMD, demonstrated attenuated cell counts, induction in apoptosis, and a reduction in cell proliferation in DIO3-positive HGSOC cells (OVCAR3 and KURAMOCHI), but not in DIO3-negative normal ovary cells (CHOK1) and OVCAR3 depleted for DIO3 or its substrate, T3. Potent tumor inhibition with a high safety profile was further established in HGSOC xenograft model, with no effect in DIO3-depleted tumors. The antitumor effects are mediated by downregulation in an array of pro-cancerous proteins, the majority of which known to be repressed by T3. To conclude, using small molecules that specifically target the DIO3 enzyme we present a new treatment paradigm for ovarian cancer and potentially other DIO3-dependent malignancies.


Subject(s)
Carcinoma, Ovarian Epithelial/drug therapy , Cystadenocarcinoma, Serous/drug therapy , Enzyme Inhibitors/administration & dosage , Iodide Peroxidase/metabolism , Small Molecule Libraries/administration & dosage , Animals , Carcinoma, Ovarian Epithelial/enzymology , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Down-Regulation , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Iodide Peroxidase/antagonists & inhibitors , Iodide Peroxidase/genetics , Mice , Molecular Mimicry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays
4.
Cancer Lett ; 501: 224-233, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33221455

ABSTRACT

High grade serous ovarian cancer (HGSOC) is the most lethal gynecologic malignancy with a need for better understanding the disease pathogenesis. The biologically active thyroid hormone, T3, is considered a tumor suppressor by promoting cell differentiation and mitochondrial respiration. Tumors evolved a strategy to avoid these anticancer actions by expressing the T3 catabolizing enzyme, Deiodinase type 3 (DIO3). This stimulates cancer proliferation and aerobic glycolysis (Warburg effect). We identified DIO3 expression in HGSOC cell lines, tumor tissues from mice and human patients, fallopian tube (FT) premalignant lesion and secretory cells of normal FT, considered the disease site-of-origin. Stable DIO3 knockdown (DIO3-KD) in HGSOC cells led to increased T3 bioavailability and demonstrated induced apoptosis and attenuated proliferation, migration, colony formation, oncogenic signaling, Warburg effect and tumor growth in mice. Proteomics analysis further indicated alterations in an array of cancer-relevant proteins, the majority of which are involved in tumor suppression and metabolism. Collectively this study establishes the functional role of DIO3 in facilitating tumorigenesis and metabolic reprogramming, and proposes this enzyme as a promising target for inhibition in HGSOC.


Subject(s)
Cystadenocarcinoma, Serous/pathology , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Ovarian Neoplasms/pathology , Up-Regulation , Aerobiosis , Animals , Cell Line, Tumor , Cell Proliferation , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glycolysis , Humans , Mice , Neoplasm Grading , Neoplasm Transplantation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism
6.
Horm Cancer ; 9(1): 22-32, 2018 02.
Article in English | MEDLINE | ID: mdl-29260382

ABSTRACT

Ovarian cancer is a highly metastatic disease. The metastatic potential is enhanced by epithelial to mesenchymal transition (EMT) in which αvß3 integrin plays a role. Thyroid hormones (L-thyroxine, T4, and 3,5,3'-triiodo-L-thyronine, T3) bind this integrin, and we hypothesized that the thyroid hormone-αvß3 axis may be involved in EMT activity in ovarian cancer. The transcription (mRNA), protein abundance (westerns), and protein localization (fluorescence microscopy) of several EMT markers were studied in ovarian cancer cells (OVCAR-3, A2780, and SKOV-3) treated with 1 nM T3 or 100 nM T4 for 1-24 h. The protein levels of ß-catenin, and its downstream targets, zeb-1, slug, and vimentin, were significantly induced by both hormones, while the effect on transcription was limited. The pre-incubation of the cells overnight with two integrin inhibitors, RGD (0.1-10 µM) or αvß3 blocking antibody (1-100 ng/mL), prevented the induction of ß-catenin by T3 and zeb-1 by T4, indicating direct integrin involvement. The transcription of the mesenchymal markers, ß-catenin, zeb-1, slug/snail, vimentin, and n-cadherin was hardly affected by T3 and T4, while that of the epithelial markers, e-cadherin and zo-1, was inhibited. Our results suggest a novel role for the thyroid hormone-αvß3 axis in EMT, with possible implications for ovarian cancer metastasis.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Ovarian Neoplasms/genetics , Thyroid Hormones/genetics , Cadherins/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrin alphaVbeta3/genetics , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Snail Family Transcription Factors/genetics , Thyroid Hormones/pharmacology , Thyroxine/pharmacology , Triiodothyronine/pharmacology , Vimentin/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Zonula Occludens-1 Protein/genetics , beta Catenin/genetics
8.
Sci Rep ; 7(1): 16475, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29184090

ABSTRACT

Ovarian cancer is a highly aggressive disease and novel treatments are required. Thyroid hormones binding to αvß3 integrin produced growth-promoting activities in ovarian cancer and we hypothesized that natural thyroid hormone derivatives may antagonize these actions. The effect of three antagonists, tetraiodoacetic acid (tetrac), triiodothyroacetic acid (triac) and 3-iodothyronamine (T1AM), on cell proliferation, cell death and DNA damage was studied in two ovarian cancer cell lines (OVCAR3 and A2780), normal hamster ovary control cells (CHOK1) and αvß3-deficient or transfected HEK293 cells. A differential inhibition of cell proliferation was observed in ovarian cancer cells compared to CHOK1. In OVCAR3, an induction of cell cycle regulators was further shown. Apoptosis was confirmed (annexin-PI, SubG1/cell-cycle, apoptotic genes, caspase-3 and poly ADP ribose polymerase-1 (PARP-1) cleavage) and was reversed by a pan-caspase inhibitor. Induction in apoptosis inducing factor (AIF) was observed, suggesting a parallel caspase-independent mechanism. Integrin-involvement in triac/T1AM apoptotic action was shown in αvß3-transfected HEK293 cells. Lastly, in ovarian cancer models, key proteins that coordinate recognition of DNA damage, ataxia-telangiectasia mutated (ATM) and PARP-1, were induced. To conclude, the cytotoxic potential of thyroid hormone derivatives, tetrac, triac and T1AM, in ovarian cancer may provide a much-needed novel therapeutic approach.


Subject(s)
Cell Death/drug effects , DNA Damage/drug effects , Thyroid Hormones/pharmacology , Apoptosis/drug effects , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Flow Cytometry , Gene Expression , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Thyroid Hormones/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL