1
|
Cilleros-Rodríguez D, Lasa M. Analysis of Apoptosis by Thyroid Hormone Induction. Methods Mol Biol 2025; 2876:77-91. [PMID: 39579309 DOI: 10.1007/978-1-0716-4252-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Apoptosis is a type of programmed cell death which can be induced by thyroid hormone in pituitary and other cell models. This process is characterized by several biochemical changes, including modifications in plasma membrane lipid composition, mitochondrial membrane dysfunction, and DNA fragmentation, among others. These cellular alterations can be measured using western blotting and flow cytometry techniques. Here we describe the method to detect cleavage of poly-ADP ribose polymerase by western blotting, as well as different flow cytometry-based methods to quantify the sub-G1 hypodiploid cell population, the redistribution of phosphatidylserine to the outer leaflet of plasma membrane, and the loss of the mitochondrial inner membrane potential.
Collapse
Affiliation(s)
- Darío Cilleros-Rodríguez
- Departamento de Bioquímica-Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Marina Lasa
- Departamento de Bioquímica-Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, Madrid, Spain.
| |
Collapse
|
2
|
Zhu X, Cheng SY. Thyroid Hormone Receptors as Tumor Suppressors in Cancer. Endocrinology 2024; 165:bqae115. [PMID: 39226152 PMCID: PMC11406550 DOI: 10.1210/endocr/bqae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/05/2024]
Abstract
Accumulated research has revealed the multifaceted roles of thyroid hormone receptors (TRs) as potent tumor suppressors across various cancer types. This review explores the intricate mechanisms underlying TR-mediated tumor suppression, drawing insights from preclinical mouse models and cancer biology. This review examines the tumor-suppressive functions of TRs, particularly TRβ, in various cancers using preclinical models, revealing their ability to inhibit tumor initiation, progression, and metastasis. Molecular mechanisms underlying TR-mediated tumor suppression are discussed, including interactions with oncogenic signaling pathways like PI3K-AKT, JAK-STAT, and transforming growth factor β. Additionally, this paper examines TRs' effect on cancer stem cell activity and differentiation, showcasing their modulation of key cellular processes associated with tumor progression and therapeutic resistance. Insights from preclinical studies underscore the therapeutic potential of targeting TRs to impede cancer stemness and promote cancer cell differentiation, paving the way for precision medicine in cancer treatment and emphasizing the potential of TR-targeted therapies as promising approaches for treating cancers and improving patient outcomes.
Collapse
Affiliation(s)
- Xuguang Zhu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sheue-yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
3
|
Cherqaoui B, Crémazy F, Lauraine M, Shammas G, Said-Nahal R, Mambu Mambueni H, Costantino F, Fourmont M, Hulot A, Garchon HJ, Glatigny S, Araujo LM, Breban M. STAT1 deficiency underlies a proinflammatory imprint of naive CD4 + T cells in spondyloarthritis. Front Immunol 2023; 14:1227281. [PMID: 37920469 PMCID: PMC10619905 DOI: 10.3389/fimmu.2023.1227281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/07/2023] [Indexed: 11/04/2023] Open
Abstract
Introduction In spondyloarthritis (SpA), an increased type 3 immune response, including T helper cells (Th) 17 excess, is observed in both human and SpA animal models, such as the HLA-B27/human β2-microglobulin transgenic rat (B27-rat). Methods To investigate this unexplained Th17-biased differentiation, we focused on understanding the immunobiology of B27-rat naive CD4+ T cells (Tn). Results We observed that neutrally stimulated B27-rat Tn developed heightened Th17 profile even before disease onset, suggesting an intrinsic proinflammatory predisposition. In parallel with this observation, transcriptomic and epigenomic analyses showed that B27-rat Tn exhibited a decreased expression of Interferon/Th1- and increased expression of Th17-related genes. This molecular signature was predicted to be related to an imbalance of STAT1/STAT3 transcription factors activity. Stat1 mRNA and STAT1 protein expression were decreased before disease onset in Tn, even in their thymic precursors, whereas Stat3/STAT3 expression increased upon disease establishment. Confirming the relevance of these results, STAT1 mRNA expression was also decreased in Tn from SpA patients, as compared with healthy controls and rheumatoid arthritis patients. Finally, stimulation of B27-rat Tn with a selective STAT1 activator abolished this preferential IL-17A expression, suggesting that STAT1-altered activity in B27-rats allows Th17 differentiation. Discussion Altogether, B27-rat Tn harbor a STAT1 deficiency preceding disease onset, which may occur during their thymic differentiation, secondarily associated with a persistent Th17 bias, which is imprinted at the epigenomic level. This early molecular phenomenon might lead to the persistent proinflammatory skew of CD4+ T cells in SpA patients, thus offering new clues to better understand and treat SpA.
Collapse
Affiliation(s)
- Bilade Cherqaoui
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Frédéric Crémazy
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Marc Lauraine
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Ghazal Shammas
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Roula Said-Nahal
- Rheumatology Division, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Hendrick Mambu Mambueni
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
- Genomic Platform of Faculty of Health Simone Veil, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Félicie Costantino
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
- Rheumatology Division, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Marine Fourmont
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Audrey Hulot
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Henri-Jean Garchon
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
- Genomic Platform of Faculty of Health Simone Veil, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Simon Glatigny
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Luiza M. Araujo
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
| | - Maxime Breban
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d’Excellence Inflamex, Université Paris-Centre, Paris, France
- Rheumatology Division, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| |
Collapse
|
4
|
Gillis NE, Cozzens LM, Wilson ER, Smith NM, Tomczak JA, Bolf EL, Carr FE. TRβ Agonism Induces Tumor Suppression and Enhances Drug Efficacy in Anaplastic Thyroid Cancer in Female Mice. Endocrinology 2023; 164:bqad135. [PMID: 37702560 PMCID: PMC10506733 DOI: 10.1210/endocr/bqad135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 09/14/2023]
Abstract
Thyroid hormone receptor beta (TRβ) is a recognized tumor suppressor in numerous solid cancers. The molecular signaling of TRβ has been elucidated in several cancer types through re-expression models. Remarkably, the potential impact of selective activation of endogenous TRβ on tumor progression remains largely unexplored. We used cell-based and in vivo assays to evaluate the effects of the TRβ agonist sobetirome (GC-1) on a particularly aggressive and dedifferentiated cancer, anaplastic thyroid cancer (ATC). Here we report that GC-1 reduced the tumorigenic phenotype, decreased cancer stem-like cell populations, and induced redifferentiation of the ATC cell lines with different mutational backgrounds. Of note, this selective activation of TRβ amplified the effects of therapeutic agents in blunting the aggressive cell phenotype and stem cell growth. In xenograft assays, GC-1 alone inhibited tumor growth and was as effective as the kinase inhibitor, sorafenib. These results indicate that selective activation of TRβ not only induces a tumor suppression program de novo but enhances the effectiveness of anticancer agents, revealing potential novel combination therapies for ATC and other aggressive solid tumors.
Collapse
Affiliation(s)
- Noelle E Gillis
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
- University of Vermont Cancer Center, University of Vermont, Burlington, VT 05405, USA
| | - Lauren M Cozzens
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Emily R Wilson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Noah M Smith
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jennifer A Tomczak
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Eric L Bolf
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
- University of Vermont Cancer Center, University of Vermont, Burlington, VT 05405, USA
| | - Frances E Carr
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
- University of Vermont Cancer Center, University of Vermont, Burlington, VT 05405, USA
| |
Collapse
|
5
|
Thyroid Hormone Receptor β Knockdown Reduces Suppression of Progestins by Activating the mTOR Pathway in Endometrial Cancer Cells. Int J Mol Sci 2022; 23:ijms232012517. [PMID: 36293372 PMCID: PMC9604373 DOI: 10.3390/ijms232012517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Progestin resistance is a major obstacle to conservative therapy in patients with endometrial cancer (EC) and endometrial atypical hyperplasia (EAH). However, the related inducing factor is yet unclear. In this study, thyroid hormone and its receptor α (TRα) and β (TRβ) of patients were assayed. THRB-silenced RL95-2 and KLE EC cells were cultured to investigate the response of progestins. Transcriptomics and Western blotting were performed to investigate the changes in signaling pathways. We found that THRB, rather than THRA, knockdown promoted the viability and motilities of RL95-2 cells but not KLE cells. The suppressive effect of progestins on cell growth and motility significantly decreased in THRB-silenced RL95-2 cells. Multiple proliferation-related signaling pathways were enriched, and the activities of mammalian targets of rapamycin (mTOR)/4e-binding protein 1 (4EBP1)/eukaryotic translation initiation factor 4G (eIF4G) rather than phosphorylated protein kinase B (Akt) were remarkably boosted. Progestin treatment enhanced the effects, and the augmentation was partially abated on supplementation with T3. In THRB-knockdown KLE cells, the progestins-activated partial signaling pathway expression (either mTOR or eIF4G), and supplementation with T3 did not induce noticeable alterations. The serum levels of triiodothyronine (T3) were significantly lower in patients with EC compared with healthy women. A strong expression of TRβ was observed in most patients with EC and EAH sensitive to progestin treatment. In contrast, TRα positive expression was detected in less than half of the patients sensitive to progestin therapy. In conclusion, THRB knockdown enhanced the viability and motility of type I EC cells and attenuated the suppressive effects of progestins by activating the mTOR-4EBP1/eIF4G pathway. Lower expression of THRB is likely correlated with progesterone resistance.
Collapse
|
6
|
Li R, Zhang Q, Feng D, Jin F, Han S, Yu X. Case report: Lymph node metastases of breast cancer and thyroid cancer encountered in axilla. Front Oncol 2022; 12:983996. [PMID: 36248999 PMCID: PMC9561385 DOI: 10.3389/fonc.2022.983996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/13/2022] [Indexed: 11/26/2022] Open
Abstract
Occurrences of breast cancer and thyroid cancer metachronously or synchronously are common for women, but axillary lymph node metastasis from both cancers is rarely seen. We report a patient who had two metastatic lymph nodes from papillary thyroid carcinoma after axillary lymph node dissection with mastectomy. Papillary thyroid carcinoma diagnosis was ensured after thyroidectomy. A literature review revealed that even the co-occurrence of breast cancer and thyroid cancer is not rare, but the etiology behind this phenomenon is not elucidated well. Genetic disorders, thyroid dysfunction, and hormone receptors may be relevant. Considering the rareness of axillary lymph node metastasis of thyroid cancer, adjuvant therapy and surgery treatment for this kind of case should be considered elaborately.
Collapse
Affiliation(s)
- Rihan Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qingfu Zhang
- Department of Pathology, The First Hospital of China Medical University, Shenyang, China
| | - Dongdong Feng
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Feng Jin
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Siyuan Han
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Xinmiao Yu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Xinmiao Yu,
| |
Collapse
|
7
|
Dhingra M, Mahalanobis S, Das A. Thyroid receptor β might be responsible for breast cancer associated with Hashimoto's thyroiditis: a new insight into pathogenesis. Immunol Res 2022; 70:441-448. [PMID: 35562625 DOI: 10.1007/s12026-022-09288-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 05/04/2022] [Indexed: 11/30/2022]
Abstract
Breast cancer is the most common cancer affecting females worldwide. Often it is observed that women suffering from Hashimoto's thyroiditis exhibit a greater propensity towards development of breast cancer. The exact mechanism for the same is unknown. However, multiple experimental evidences suggest a significant role of thyroid receptor β (TR-β) in regulating cell growth and proliferation and thus play a potent role as a tumor suppressor in several cancers, including breast cancer. Thyroid receptor β shows anti-proliferative action through mediators like β-catenin, RUNX2, PI3K/AKT, and cyclin regulation. The present review explores the link between these pathways and how they may be dysregulated due to Hashimoto's thyroiditis. Further, we propose a new mechanism for cancer prognosis associated with Hashimoto's thyroiditis, which may lead to the development of TR-β targeting as a novel therapeutic approach.
Collapse
Affiliation(s)
- Muskaan Dhingra
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India
| | - Shayon Mahalanobis
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India.
| |
Collapse
|
8
|
Pourvali K, Shimi G, Ghorbani A, Shakery A, Shirazi FH, Zand H. Selective thyroid hormone receptor beta agonist, GC-1, is capable to reduce growth of colorectal tumor in syngeneic mouse models. J Recept Signal Transduct Res 2022; 42:495-502. [PMID: 35473566 DOI: 10.1080/10799893.2022.2032748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE The effect of thyroid hormone (TH) on cancers was proposed more than 100 years ago; however, conclusions are conflicting. THs are precisely regulated at tissue and cellular levels. It seems that this regulation is altered in cancers. Thyroid hormone receptor beta (TRβ) has anti-proliferative and tumor-suppressive effects in many cancer cells. Therefore, we decided to investigate thyroid hormone receptor beta (THRB) expression and activation by the selective agonist, GC-1, on tumor growth in a syngeneic mouse model of colorectal cancer (CRC) and colon cell lines. METHODS In vitro cell viability assay using MTT analysis, cell cycle analysis by PI staining, and FACS analysis were performed. In vivo tumor growth measurements were carried out by caliper and [18F] Fluoro-2-deoxy-2-D-glucose (FDG) - PET imaging. Gene expressions were determined using quantitative-PCR. RESULTS Some concentrations of GC-1 had a marked negative effect on the cell viability of colorectal cell lines. Cell cycle analysis showed that the anti-proliferative effect of GC-1 may not result from cell cycle arrest or apoptosis. Tumor growth analysis in mice harboring colorectal tumor showed that GC-1 treatment for 8 d profoundly inhibited tumor growth and 18FDG uptake. THRB expression was decreased in mice tumor; however, it was upregulated following GC-1 administration. CONCLUSIONS Our results showed that specific activation of TRβ by GC-1 had negative effect on tumor growth and restored its gene expression in tumors of CRC mice model.
Collapse
Affiliation(s)
- Katayoun Pourvali
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Shimi
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Ghorbani
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Shakery
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farshad Hosseini Shirazi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Zand
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Doolittle WKL, Zhu X, Park S, Zhu YJ, Zhao L, Meltzer P, Cheng SY. Regulation of cancer stem cell activity by thyroid hormone receptor β. Oncogene 2022; 41:2315-2325. [PMID: 35256781 PMCID: PMC9018601 DOI: 10.1038/s41388-022-02242-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/10/2022] [Indexed: 12/20/2022]
Abstract
Increasing numbers of cancer stem cell markers have been recently identified. It is not known, however, whether a member of the nuclear receptor superfamily, thyroid hormone receptor β (TRβ), can function to regulate cancer stem cell (CSC) activity. Using anaplastic thyroid cancer cells (ATC) as a model, we highlight the role of TRβ in CSC activity. ATC is one of the most aggressive solid cancers in humans and is resistant to currently available therapeutics. Recent studies provide evidence that CSC activity underlies aggressiveness and therapeutic resistance of ATC. Here we show that TRβ inhibits CSC activity by suppressing tumor-sphere formation of human ATC cells and their tumor-initiating capacity. TRβ suppresses the expression of CSC regulators, including ALDH, KLF2, SOX2, b-catenin, and ABCG2, in ATC cell-induced xenograft tumors. Single-cell transcriptomic analysis shows that TRβ reduces CSC population in ATC-induced xenograft tumors. Analysis of The Cancer Genome Atlas (TCGA) database demonstrates that the inhibition of CSC capacity by TRβ contributes to favorable clinical outcomes in human cancer. Our studies show that TRβ is a newly identified transcription regulator that acts to suppress CSC activity and that TRβ could be considered as a molecular target for therapeutic intervention of ATC.
Collapse
Affiliation(s)
- Woo Kyung Lee Doolittle
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuguang Zhu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sunmi Park
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yuelin Jack Zhu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Li Zhao
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Paul Meltzer
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
10
|
Ezh2 promotes TRβ lysine methylation-mediated degradation in hepatocellular carcinoma. Genes Genomics 2021; 44:369-377. [PMID: 34851506 DOI: 10.1007/s13258-021-01196-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/23/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Post-translational modification (PTM) of proteins controls various cellular functions of transcriptional regulators and participates in diverse signal transduction pathways in cancer. The thyroid hormone (triiodothyronine, T3) plays a critical role in metabolic homeostasis via its direct interaction with the thyroid hormone receptor beta (TRβ). TRβ is involved in physiological processes, such as cell growth, differentiation, apoptosis, and maintenance of metabolic homeostasis through transcriptional regulation of target genes. OBJECTIVE This study was performed to characterize the specific PTM of TRβ is an active control mechanism for the proteasomal degradation of TRβ in transcriptional signaling pathways in hepatocellular carcinoma cells. METHODS Based on a previous study, we predicted that the lysine methyltransferase and methylation sites of TRβ by comparing the amino acid sequences of histone H3 and TRβ. Methyl-acceptor site of TRβ was confirmed by point mutation. TRβ protein stability was evaluated by ubiquitination assay with MG132. For glucose starvation, HepG2 cells were incubated in media without D-glucose. Proliferation-related proteins were detected by western blotting. MicroRNA level and autophagy marker were measured by real-time qPCR. RESULTS The presence of enhancer of zeste homolog 2 (Ezh2), a methyltransferase of H3 lysine 27, as a methyltransferase of TRβ also revealed that direct lysine methylation and consequent stimulated protein degradation of TRβ underlies the negative correlation between Ezh2 and TRβ. Notably, glucose starvation significantly increased lysine methylation, and methylated TRβ showed further protein instability leading to an increase in the proliferation and growth of hepatocellular carcinoma cells. CONCLUSIONS TRβ functions as a tumor suppressor in various cancers; therefore, we evaluated the effect of TRβ degradation on oncogenesis during glucose starvation. These data clearly define a functional model and provide a link between metabolism and cancer by regulating methyl-dependent protein levels of tumor suppressors. Taken together, maintaining TRβ against methyl-dependent degradation is considered a possible therapeutic target for cancer progression.
Collapse
|
11
|
Villegas A, Satheeshkumar R, Ballesteros‐Casallas A, Paulino M, Castro A, Espinosa‐Bustos C, Salas CO. Convergent synthesis, drug target prediction, and docking studies of new 2,6,9‐trisubstituted purine derivatives. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alondra Villegas
- Departamento de Química Orgánica Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile Santiago de Chile Chile
| | - Rajendran Satheeshkumar
- Departamento de Química Orgánica Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile Santiago de Chile Chile
| | | | - Margot Paulino
- Departamento DETEMA Facultad de Química, Universidad de la República Montevideo Uruguay
| | - Alejandro Castro
- Laboratorio de Bioproductos Farmacéuticos y Cosméticos Centro de Excelencia en Medicina Traslacional, Facultad de Medicina, Universidad de La Frontera Temuco Chile
| | - Christian Espinosa‐Bustos
- Departamento de Farmacia Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile Santiago de Chile Chile
| | - Cristian O. Salas
- Departamento de Química Orgánica Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile Santiago de Chile Chile
| |
Collapse
|
12
|
Bolf EL, Gillis NE, Davidson CD, Cozzens LM, Kogut S, Tomczak JA, Frietze S, Carr FE. Common tumor-suppressive signaling of thyroid hormone receptor beta in breast and thyroid cancer cells. Mol Carcinog 2021; 60:874-885. [PMID: 34534367 DOI: 10.1002/mc.23352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 01/16/2023]
Abstract
The thyroid hormone receptor beta (TRβ) is a tumor suppressor in multiple types of solid tumors, most prominently in breast and thyroid cancer. An increased understanding of the molecular mechanisms by which TRβ abrogates tumorigenesis will aid in understanding the core tumor-suppressive functions of TRβ. Here, we restored TRβ expression in the MDA-MB-468 basal-like breast cancer cell line and perform RNA-sequencing to determine the TRβ-mediated changes in gene expression and associated signaling pathways. The TRβ expressing MDA-MB-468 cells exhibit a more epithelial character as determined by principle component analysis-based iterative PAM50 subtyping score and through reduced expression of mesenchymal cytokeratins. The epithelial to mesenchymal transition pathway is also significantly reduced. The MDA-MB-468 data set was further compared with RNA sequencing results from TRβ expressing thyroid cancer cell line SW1736 to determine which genes are TRβ correspondingly regulated across both cell types. Several pathways including lipid metabolism and chromatin remodeling processes were observed to be altered in the shared gene set. These data provide novel insights into the molecular mechanisms by which TRβ suppresses breast tumorigenesis.
Collapse
Affiliation(s)
- Eric L Bolf
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA
| | - Noelle E Gillis
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA
| | - Cole D Davidson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA
| | - Lauren M Cozzens
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Sophie Kogut
- Department of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, Vermont, USA
| | - Jennifer A Tomczak
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Seth Frietze
- University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA.,Department of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, Vermont, USA
| | - Frances E Carr
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,University of Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
13
|
Davidson CD, Gillis NE, Carr FE. Thyroid Hormone Receptor Beta as Tumor Suppressor: Untapped Potential in Treatment and Diagnostics in Solid Tumors. Cancers (Basel) 2021; 13:4254. [PMID: 34503062 PMCID: PMC8428233 DOI: 10.3390/cancers13174254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 01/07/2023] Open
Abstract
There is compelling evidence that the nuclear receptor TRβ, a member of the thyroid hormone receptor (TR) family, is a tumor suppressor in thyroid, breast, and other solid tumors. Cell-based and animal studies reveal that the liganded TRβ induces apoptosis, reduces an aggressive phenotype, decreases stem cell populations, and slows tumor growth through modulation of a complex interplay of transcriptional networks. TRβ-driven tumor suppressive transcriptomic signatures include repression of known drivers of proliferation such as PI3K/Akt pathway, activation of novel signaling such as JAK1/STAT1, and metabolic reprogramming in both thyroid and breast cancers. The presence of TRβ is also correlated with a positive prognosis and response to therapeutics in BRCA+ and triple-negative breast cancers, respectively. Ligand activation of TRβ enhances sensitivity to chemotherapeutics. TRβ co-regulators and bromodomain-containing chromatin remodeling proteins are emergent therapeutic targets. This review considers TRβ as a potential biomolecular diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Cole D. Davidson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; (C.D.D.); (N.E.G.)
- University of Vermont Cancer Center, Burlington, VT 05401, USA
| | - Noelle E. Gillis
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; (C.D.D.); (N.E.G.)
- University of Vermont Cancer Center, Burlington, VT 05401, USA
| | - Frances E. Carr
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; (C.D.D.); (N.E.G.)
- University of Vermont Cancer Center, Burlington, VT 05401, USA
| |
Collapse
|
14
|
Davidson CD, Bolf EL, Gillis NE, Cozzens LM, Tomczak JA, Carr FE. Thyroid Hormone Receptor Beta Inhibits PI3K-Akt-mTOR Signaling Axis in Anaplastic Thyroid Cancer via Genomic Mechanisms. J Endocr Soc 2021; 5:bvab102. [PMID: 34258492 PMCID: PMC8271203 DOI: 10.1210/jendso/bvab102] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Indexed: 01/21/2023] Open
Abstract
Thyroid cancer is the most common endocrine malignancy, and the global incidence has increased rapidly over the past few decades. Anaplastic thyroid cancer (ATC) is highly aggressive, dedifferentiated, and patients have a median survival of fewer than 6 months. Oncogenic alterations in ATC include aberrant phosphoinositide 3 kinase (PI3K) signaling through receptor tyrosine kinase (RTK) amplification, loss of phosphoinositide phosphatase expression and function, and protein kinase B (Akt) amplification. Furthermore, the loss of expression of the tumor suppressor thyroid hormone receptor beta (TRβ) is strongly associated with ATC. TRβ is known to suppress PI3K in follicular thyroid cancer and breast cancer by binding to the PI3K regulatory subunit p85α. However, the role of TRβ in suppressing PI3K signaling in ATC is not completely delineated. Here we report that TRβ indeed suppresses PI3K signaling in ATC cell lines through unreported genomic mechanisms, including a decrease in RTK expression and an increase in phosphoinositide and Akt phosphatase expression. Furthermore, the reintroduction and activation of TRβ in ATC cell lines enables an increase in the efficacy of the competitive PI3K inhibitors LY294002 and buparlisib on cell viability, migration, and suppression of PI3K signaling. These findings not only uncover additional tumor suppressor mechanisms of TRβ but shed light on the implication of TRβ status and activation on inhibitor efficacy in ATC tumors.
Collapse
Affiliation(s)
- Cole D Davidson
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
- University of Vermont Cancer Center, Burlington, Vermont 05401, USA
| | - Eric L Bolf
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
- University of Vermont Cancer Center, Burlington, Vermont 05401, USA
| | - Noelle E Gillis
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
- University of Vermont Cancer Center, Burlington, Vermont 05401, USA
| | - Lauren M Cozzens
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
| | - Jennifer A Tomczak
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
| | - Frances E Carr
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
- University of Vermont Cancer Center, Burlington, Vermont 05401, USA
| |
Collapse
|