Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Gen Subj ; 1864(8): 129625, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32335136

RESUMEN

BACKGROUND: N-myc downstream regulated gene 1 (NDRG1) is an established stress-response protein. This study investigated the effects of NDRG1 on autophagic degradation and how this can be therapeutically exploited. METHODS: Cell culture, western analysis, confocal microscopy, acridine orange staining, cholesterol determination, cellular proliferation assessment and combination index (CI) estimation. RESULTS: NDRG1 expression suppressed autophagic degradation and autolysosome formation, measured by increased p62 expression and reduced co-localization between the well-characterized, autophagosomal and lysosomal markers, LC3 and LAMP2, respectively. NDRG1 elicited autophagic suppression at the initiation stage of autophagy. The NDRG1-inducer and anti-cancer agent, di-2-pyridylketone 4,4,-dimethyl-3-thiosemicarbazone (Dp44mT), was able to induce lysosomal membrane permeabilization (LMP). Over-expression of NDRG1 further sensitized cells to LMP mediated by both Dp44mT, or the redox active Dp44mT­copper complex. This sensitization may be mediated via a decrease in cholesterol levels upon NDRG1 expression, as cholesterol stabilizes lysosomal membranes. However, the effect of NDRG1 on cholesterol appeared independent of the key energy homeostasis sensor, 5' AMP-activated protein kinase (AMPK), whose activation was significantly (p < 0.001) reduced by NDRG1. Finally, Dp44mT synergistically potentiated the anti-proliferative activity of Gemcitabine that activates autophagy. In fact, Dp44mT and Gemcitabine (Combination Index (CI): 0.38 ±â€¯0.07) demonstrated higher synergism versus the autophagy inhibitor, Bafilomycin A1 and Gemcitabine (CI: 0.64 ±â€¯0.19). CONCLUSIONS AND GENERAL SIGNIFICANCE: Collectively, this study demonstrated a dual-inhibitory mechanism of NDRG1 on autophagic activity, and that NDRG1 expression sensitized cells to Dp44mT-induced LMP. Considering the ability of Dp44mT to inhibit autophagy, studies demonstrated the potential of combination therapy for cancer treatment of Dp44mT with Gemcitabine.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Permeabilidad de la Membrana Celular , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pancreáticas/metabolismo , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Permeabilidad de la Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Tiosemicarbazonas/farmacología , Células Tumorales Cultivadas
2.
J Biol Chem ; 295(2): 481-503, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31744884

RESUMEN

Considering the role of proto-oncogene c-Met (c-Met) in oncogenesis, we examined the effects of the metastasis suppressor, N-myc downstream-regulated gene-1 (NDRG1), and two NDRG1-inducing thiosemicarbazone-based agents, Dp44mT and DpC, on c-Met expression in DU145 and Huh7 cells. NDRG1 silencing without Dp44mT and DpC up-regulated c-Met expression, demonstrating that NDRG1 modulates c-Met levels. Dp44mT and DpC up-regulated NDRG1 by an iron-dependent mechanism and decreased c-Met levels, c-Met phosphorylation, and phosphorylation of its downstream effector, GRB2-associated binding protein 1 (GAB1). However, incubation with Dp44mT and DpC after NDRG1 silencing or silencing of the receptor tyrosine kinase inhibitor, mitogen-inducible gene 6 (MIG6), decreased c-Met and its phosphorylation, suggesting NDRG1- and MIG6-independent mechanism(s). Lysosomal inhibitors rescued the Dp44mT- and DpC-mediated c-Met down-regulation in DU145 cells. Confocal microscopy revealed that lysosomotropic agents and the thiosemicarbazones significantly increased co-localization between c-Met and lysosomal-associated membrane protein 2 (LAMP2). Moreover, generation of c-Met C-terminal fragment (CTF) and its intracellular domain (ICD) suggested metalloprotease-mediated cleavage. In fact, Dp44mT increased c-Met CTF while decreasing the ICD. Dp44mT and a γ-secretase inhibitor increased cellular c-Met CTF levels, suggesting that Dp44mT induces c-Met CTF levels by increasing metalloprotease activity. The broad metalloprotease inhibitors, EDTA and batimastat, partially prevented Dp44mT-mediated down-regulation of c-Met. In contrast, the ADAM inhibitor, TIMP metallopeptidase inhibitor 3 (TIMP-3), had no such effect, suggesting c-Met cleavage by another metalloprotease. Notably, Dp44mT did not induce extracellular c-Met shedding that could decrease c-Met levels. In summary, the thiosemicarbazones Dp44mT and DpC effectively inhibit oncogenic c-Met through lysosomal degradation and metalloprotease-mediated cleavage.


Asunto(s)
Antineoplásicos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Lisosomas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/genética , Tiosemicarbazonas/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Proteolisis/efectos de los fármacos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA