Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Endocrinology ; 164(10)2023 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-37702560

RESUMO

Thyroid hormone receptor beta (TRß) is a recognized tumor suppressor in numerous solid cancers. The molecular signaling of TRß has been elucidated in several cancer types through re-expression models. Remarkably, the potential impact of selective activation of endogenous TRß on tumor progression remains largely unexplored. We used cell-based and in vivo assays to evaluate the effects of the TRß 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 TRß 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 TRß 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.


Assuntos
Carcinoma Anaplásico da Tireoide , Neoplasias da Glândula Tireoide , Feminino , Humanos , Animais , Camundongos , Carcinoma Anaplásico da Tireoide/tratamento farmacológico , Receptores beta dos Hormônios Tireóideos , Agressão , Neoplasias da Glândula Tireoide/tratamento farmacológico
2.
Mol Carcinog ; 60(12): 874-885, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34534367

RESUMO

The thyroid hormone receptor beta (TRß) 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 TRß abrogates tumorigenesis will aid in understanding the core tumor-suppressive functions of TRß. Here, we restored TRß expression in the MDA-MB-468 basal-like breast cancer cell line and perform RNA-sequencing to determine the TRß-mediated changes in gene expression and associated signaling pathways. The TRß 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 TRß expressing thyroid cancer cell line SW1736 to determine which genes are TRß 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 TRß suppresses breast tumorigenesis.


Assuntos
Neoplasias da Mama/genética , Perfilação da Expressão Gênica/métodos , Receptores beta dos Hormônios Tireóideos/genética , Neoplasias da Glândula Tireoide/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metabolismo dos Lipídeos , Análise de Componente Principal , Análise de Sequência de RNA , Transdução de Sinais , Receptores beta dos Hormônios Tireóideos/metabolismo , Neoplasias da Glândula Tireoide/metabolismo
3.
J Endocr Soc ; 5(8): bvab102, 2021 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-34258492

RESUMO

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 (TRß) is strongly associated with ATC. TRß is known to suppress PI3K in follicular thyroid cancer and breast cancer by binding to the PI3K regulatory subunit p85α. However, the role of TRß in suppressing PI3K signaling in ATC is not completely delineated. Here we report that TRß 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 TRß 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 TRß but shed light on the implication of TRß status and activation on inhibitor efficacy in ATC tumors.

4.
Mol Cancer Res ; 18(10): 1443-1452, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32554601

RESUMO

The thyroid hormone receptor beta (TRß), a key regulator of cellular growth and differentiation, is frequently dysregulated in cancers. Diminished expression of TRß is noted in thyroid, breast, and other solid tumors and is correlated with more aggressive disease. Restoration of TRß levels decreased tumor growth supporting the concept that TRß could function as a tumor suppressor. Yet, the TRß tumor suppression transcriptome is not well delineated and the impact of TRß is unknown in aggressive anaplastic thyroid cancer (ATC). Here, we establish that restoration of TRß expression in the human ATC cell line SW1736 (SW-TRß) reduces the aggressive phenotype, decreases cancer stem cell populations and induces cell death in a T3-dependent manner. Transcriptomic analysis of SW-TRß cells via RNA sequencing revealed distinctive expression patterns induced by ligand-bound TRß and revealed novel molecular signaling pathways. Of note, liganded TRß repressed multiple nodes in the PI3K/AKT pathway, induced expression of thyroid differentiation markers, and promoted proapoptotic pathways. Our results further revealed the JAK1-STAT1 pathway as a novel, T3-mediated, antitumorigenic pathway that can be activated in additional ATC lines. These findings elucidate a TRß-driven tumor suppression transcriptomic signature, highlight unexplored therapeutic options for ATC, and support TRß activation as a promising therapeutic option in cancers. IMPLICATIONS: TRß-T3 induced a less aggressive phenotype and tumor suppression program in anaplastic thyroid cancer cells revealing new potential therapeutic targets.


Assuntos
Carcinoma Anaplásico da Tireoide/genética , Receptores beta dos Hormônios Tireóideos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Genes Supressores de Tumor , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...