Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Drug Dev Res ; 84(5): 861-887, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37070554

RESUMEN

Papillary thyroid carcinoma contributes to about 80% of the total thyroid cancer cases. BRAFV600E is a frequently occurring mutation in PTCs. Although several BRAF inhibitors are available, many thyroid cancer patients acquire resistance to BRAF inhibitors. Therefore, new targets and drugs need to be identified as therapies. Ferroptosis is a recently discovered type of cell death, and inhibiting glutathione peroxidase 4 (GPX4) using small molecules was found to trigger ferroptosis. But it is unknown whether inhibiting GPX4 renders thyroid cancer cells susceptible to ferroptosis. To identify novel GPX4 inhibitors, we focused on our previously reported cohort of diaryl ether and dibenzoxepine molecules. In this study, we asked whether diaryl ether and dibenzoxepine derivatives trigger ferroptosis in thyroid cancer cells. To answer this question, we screened diaryl ether and dibenzoxepine derivatives in cell-based assays and performed mechanism of action studies. We found that a diaryl ether derivative, 16 decreased thyroid cell proliferation and triggered ferroptosis by inhibiting GPX4 expression levels. Molecular modeling and dynamics simulations showed that 16 binds to the active site of GPX4. Upon deciphering the mode of 16-induced ferroptosis, we found that 16 treatments decrease mitochondrial polarization and reduce mitochondrial respiration similar to a ferroptosis inducer, RSL3. We conclude that the diaryl ether derivative, 16 inhibits GPX4 expression levels to induce ferroptosis in thyroid cancer cells. Based on our observations, we suggest that 16 can be lead-optimized and developed as a ferroptosis-inducing agent to treat thyroid cancers.


Asunto(s)
Ferroptosis , Neoplasias de la Tiroides , Humanos , Éter , Proteínas Proto-Oncogénicas B-raf , Éteres de Etila , Neoplasias de la Tiroides/tratamiento farmacológico , Éteres
2.
Free Radic Biol Med ; 145: 428-441, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31614178

RESUMEN

In recent years there has been an upsurge in research focusing on reprogramming cancer cells through understanding of their metabolic signatures. Alterations in mitochondrial bioenergetics and impaired mitochondrial function may serve as effective targeting strategies especially in triple-negative breast cancers (TNBCs) where hormone receptors and endocrine therapy are absent. Glucose starvation (GS) of MDA-MB-231 and MCF-7 breast cancer cells showed decrease in mitochondrial Oxygen Consumption Rate (OCR), which was rescuable to control level through addition of exogenous antioxidant N-Acetyl Cysteine (NAC). Mechanistically, GS led to increase in mitochondrial ROS and upregulation of the pleiotropic protein, Prohibitin 1 (PHB1), leading to its dissociation from Dynamin-related protein 1 (DRP1), perturbance of mitochondrial membrane potential (MMP) and triggering of the apoptosis cascade. PHB1 also reduced the invasive and migratory potential of both cell lines. We emphasize that glucose starvation remarkably sensitized the highly glycolytic metastatic TNBC cell line, MDA-MB-231 to apoptosis and decreased its migratory potential. Based on our findings, additional TNBC cell lines can be evaluated and a nutritional paradigm be proposed for anticancer therapy.


Asunto(s)
Neoplasias de la Mama/genética , Glucosa/metabolismo , Estrés Oxidativo/genética , Proteínas Represoras/genética , Animales , Apoptosis/genética , Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Supervivencia Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glucosa/farmacología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Prohibitinas , Especies Reactivas de Oxígeno/metabolismo , Inanición/complicaciones , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Res ; 68(18): 7570-8, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18794145

RESUMEN

Radiotherapy combined with chemotherapy is the treatment of choice for glioblastoma and locally advanced lung cancer, but radioresistance of these two types of cancer remains a significant therapeutic hindrance. To identify molecular target(s) for radiosensitization, we screened a small interfering RNA (siRNA) library targeting all protein kinases and E3 ubiquitin ligases in the human genome and identified tumor necrosis factor receptor-associated factor 2 (TRAF2). Silencing of TRAF2 using siRNA caused a significant growth suppression of glioblastoma U251 cells and moderately sensitized these radioresistant cells to radiation. Overexpression of a really interesting new gene (RING)-deleted dominant-negative TRAF2 mutant also conferred radiosensitivity, whereas overexpression of wild-type (WT) TRAF2 significantly protected cells from radiation-induced killing. Likewise, siRNA silencing of TRAF2 in radioresistant lung cancer H1299 cells caused growth suppression and radiosensitization, whereas overexpression of WT TRAF2 enhanced radioresistance in a RING ligase-dependent manner. Moreover, siRNA silencing of TRAF2 in UM-SCC-1 head and neck cancer cells also conferred radiosensitization. Further support for the role of TRAF2 in cancer comes from the observations that TRAF2 is overexpressed in both lung adenocarcinoma tissues and multiple lung cancer cell lines. Importantly, TRAF2 expression was very low in normal bronchial epithelial NL20 cells, and TRAF2 silencing had a minimal effect on NL20 growth and radiation sensitivity. Mechanistically, TRAF2 silencing blocks the activation of the nuclear factor-kappaB signaling pathway and down-regulates several G(2)-M cell cycle control proteins, resulting in enhanced G(2)-M arrest, growth suppression, and radiosensitization. Our studies suggest that TRAF2 is an attractive drug target for anticancer therapy and radiosensitization.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/terapia , Glioblastoma/terapia , Neoplasias Pulmonares/terapia , ARN Interferente Pequeño/genética , Factor 2 Asociado a Receptor de TNF/genética , Animales , Apoptosis/genética , Apoptosis/efectos de la radiación , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Proteínas de Ciclo Celular/metabolismo , División Celular/genética , División Celular/efectos de la radiación , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Fase G2/genética , Fase G2/efectos de la radiación , Silenciador del Gen , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Humanos , Proteínas I-kappa B/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Ratones , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Tolerancia a Radiación/genética
4.
Cancer Biol Ther ; 6(3): 360-6, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17312389

RESUMEN

The ellipticinium and its derivatives have been studied as anti-cancer agents with preferentially cyto-toxicity to the brain tumor cell lines. During the course of our study to determine whether an ellipticine derivative, API59-Cl would sensitize radio-resistant U87 glioblastoma cells to radiation, we found that it reduced the level of p27, a cyclin-dependent kinase inhibitor. API59-Cl induced a dose and time dependent p27 reduction in U87 cells. The compound-induced p27 reduction was also seen in three additional glioblastoma lines, T98G, U251 and U118 as well as in mouse embryonic fibroblasts. Mechanistic study of API59-Cl mediated p27 reduction revealed that it was not due to an altered p27 transcription, rather due to a shortened protein half-life as a result of enhanced p27 degradation. Indeed, API59-Cl induced p27 degradation was dependent on ubiquitin-proteasome pathway, particularly E3 ubiquitin ligase component, Skp2, but not Cullin-4A/4B, and can be largely blocked by proteasome inhibitors MG132 or PS341. Finally, we demonstrated that API59-Cl inhibited U87 cell growth with an IC50 of 1.7 muM, which is independent of its p27 degrading activity. This is the first report, to our knowledge, that the ellipticinium class of small molecule compounds promotes p27 degradation via ubiquitin-proteasome pathway. The finding could provide a new tool to further understand the mechanism of p27 degradation.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias Encefálicas/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Elipticinas/farmacología , Glioblastoma/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Ácidos Borónicos/farmacología , Bortezomib , Proliferación Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Leupeptinas/farmacología , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Pirazinas/farmacología , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Transcripción Genética , Células Tumorales Cultivadas , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
5.
Mol Cell Biochem ; 295(1-2): 179-88, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16874460

RESUMEN

Sensitive to Apoptosis Gene (SAG), a RING component of SCF E3 ubiquitin ligase, was shown to be phosphorylated by protein kinase CK2 at the Thr10 residue. It is, however, unknown whether this phosphorylation is stress-responsive or whether the phosphorylation changes its E3 ubiquitin ligase activity. To address these, we made a specific antibody against the phosphor-SAG(Thr10). Transient transfection experiment showed that SAG was phosphorylated at Thr10 which can be significantly inhibited by TBB, a relatively specific inhibitor of protein kinase CK2. To determine whether this SAG phosphorylation is stress-responsive, we defined a chemical-hypoxia condition in which SAG and CK2 were both induced. Under this condition, we failed to detect SAG phosphorylation at Thr10, which was readily detected, however, in the presence of MG132, a proteasome inhibitor, suggesting that the phosphorylated SAG has undergone a rapid degradation. To further define this, we made two SAG mutants, SAG-T10A which abolishes the SAG phosphorylation and SAG-T10E, which mimics the constitutive SAG phosphorylation. The half-life study revealed that indeed, SAG-T10E has a much shorter protein half-life (2 h), as compared to wild-type SAG (10 h). Again, rapid degradation of SAG-T10E in cells can be blocked by MG132. Thus, it appears that CK2-induced SAG phosphorylation at Thr10 regulates its stability through a proteasome-dependent pathway. Immunocytochemistry study showed that SAG as well as its phosphorylation mutants, was mainly localized in nucleus and lightly in cytoplasm. Hypoxia condition did not change their sub-cellular localization. Finally, an in vitro ubiqutination assay showed that SAG mutation at Thr10 did not change its E3 ligase activity when complexed with cullin-1. These studies suggested that CK2 might regulate SAG-SCF E3 ligase activity through modulating SAG's stability, rather than its enzymatic activity directly.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Fosfotreonina/metabolismo , Proteínas de Unión al ARN/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Anticuerpos Fosfo-Específicos/inmunología , Quinasa de la Caseína II/antagonistas & inhibidores , Hipoxia de la Célula/efectos de los fármacos , Cobalto/farmacología , Activación Enzimática/efectos de los fármacos , Estabilidad de Enzimas , Semivida , Células HeLa , Humanos , Fosfoproteínas/inmunología , Fosforilación/efectos de los fármacos , Inhibidores de Proteasoma , Transporte de Proteínas/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos , Termodinámica , Triazoles/farmacología
6.
Neoplasia ; 7(4): 312-23, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15967108

RESUMEN

Chip profiling of a p53 temperature-sensitive tumor model identified SAK (Snk/Plk-akin kinase), encoding a new member of polo-like kinases (PLKs), as a gene strongly repressed by wild-type p53. Further characterization revealed that SAK expression was downregulated by wild-type p53 in several tumor cell models. Computer search of a 1.7-kb SAK promoter sequence revealed three putative p53 binding sites, but p53 failed to bind to any of these sites, indicating that SAK repression by p53 was not through a direct p53 binding to the promoter. Transcriptional analysis with luciferase reporters driven by SAK promoter deletion fragments identified SP-1 and CREB binding sites, which together conferred a two-fold SAK repression by p53. However, the repression was not reversed by cotransfection of SP-1 or CREB, suggesting a lack of interference between p53 and SP-1 or CREB. Significantly, p53-mediated SAK repression was largely reversed in a dose-dependent manner by Trichostatin A, a potent histone deacetylase (HDAC) inhibitor, suggesting an involvement of HDAC transcription repressors in SAK repression by p53. Biologically, SAK RNA interference (RNAi) silencing induced apoptosis, whereas SAK overexpression attenuated p53-induced apoptosis. Thus, SAK repression by p53 is likely mediated through the recruitment of HDAC repressors, and SAK repression contributes to p53-induced apoptosis.


Asunto(s)
Apoptosis , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Interferencia de ARN , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Northern Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Clonación Molecular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Fragmentación del ADN , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Etopósido/farmacología , Eliminación de Gen , Genes Reporteros , Células HeLa , Humanos , Luciferasas/metabolismo , Modelos Genéticos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Programas Informáticos , Factor de Transcripción Sp1/metabolismo , Temperatura , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...