Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros




Base de datos
Asunto de la revista
Intervalo de año de publicación
1.
J Mol Biol ; 426(4): 830-42, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24295899

RESUMEN

Whereas moderately increased cellular oxidative stress is supportive for cancerous growth of cells, excessive levels of reactive oxygen species (ROS) are detrimental to their growth and survival. We demonstrated that high ROS levels, via increased oxidized glutathione (GSSG), induce isoform-specific S-glutathionylation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) at residue Cys206, which is located near the entrance to the 6-phosphofructo-2-kinase catalytic pocket. Upon this ROS-dependent, reversible, covalent modification, a marked decrease in its catalytic ability to synthesize fructose-2,6-bisphosphate (Fru-2,6-P2), the key glycolysis allosteric activator, was observed. This event was coupled to a decrease in glycolytic flux and an increase in glucose metabolic flux into the pentose phosphate pathway. This shift, in turn, caused an increase in reduced glutathione (GSH) and, ultimately, resulted in ROS detoxification inside HeLa cells. The ability of PFKFB3 to control the Fru-2,6-P2 levels in an ROS-dependent manner allows the PFKFB3-expressing cancer cells to continue energy metabolism with a reduced risk of excessive oxidative stress and, thereby, to support their cell survival and proliferation. This study provides a new insight into the roles of PFKFB3 as switch that senses and controls redox homeostasis in cancer in addition to its role in cancer glycolysis.


Asunto(s)
Neoplasias/metabolismo , Estrés Oxidativo , Fosfofructoquinasa-2/química , Fosfofructoquinasa-2/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Cisteína/química , Cisteína/metabolismo , Metabolismo Energético , Fructosadifosfatos/metabolismo , Glucosa/metabolismo , Disulfuro de Glutatión/metabolismo , Glucólisis/fisiología , Células HeLa , Homeostasis , Humanos , Datos de Secuencia Molecular , Fosfofructoquinasa-2/genética , Conformación Proteica , Especies Reactivas de Oxígeno/metabolismo , Azufre/química , Azufre/metabolismo
2.
PLoS One ; 6(9): e24179, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21957443

RESUMEN

Cancer cells adopt glycolysis as the major source of metabolic energy production for fast cell growth. The HIF-1-induced PFKFB3 plays a key role in this adaptation by elevating the concentration of Fru-2,6-BP, the most potent glycolysis stimulator. As this metabolic conversion has been suggested to be a hallmark of cancer, PFKFB3 has emerged as a novel target for cancer chemotherapy. Here, we report that a small molecular inhibitor, N4A, was identified as an initial lead compound for PFKFB3 inhibitor with therapeutic potential. In an attempt to improve its potency, we determined the crystal structure of the PFKFB3•N4A complex to 2.4 Šresolution and, exploiting the resulting molecular information, attained the more potent YN1. When tested on cultured cancer cells, both N4A and YN1 inhibited PFKFB3, suppressing the Fru-2,6-BP level, which in turn suppressed glycolysis and, ultimately, led to cell death. This study validates PFKFB3 as a target for new cancer therapies and provides a framework for future development efforts.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glucólisis/efectos de los fármacos , Fosfofructoquinasa-2/antagonistas & inhibidores , Antineoplásicos/metabolismo , Benzopiranos/química , Benzopiranos/metabolismo , Benzopiranos/farmacología , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Fructosafosfatos/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Fosfofructoquinasa-2/química , Fosfofructoquinasa-2/metabolismo , Conformación Proteica
3.
Anal Biochem ; 418(1): 143-8, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21771574

RESUMEN

Efforts toward improving the predictiveness in tier-based approaches to virtual screening (VS) have mainly focused on protein kinases. Despite their significance as drug targets, small molecule kinases have been rarely tested with these approaches. In this paper, we investigate the efficacy of a pharmacophore screening-combined structure-based docking approach on the human inducible 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase, an emerging target for cancer chemotherapy. Six out of a total 1364 compounds from NCI's Diversity Set II were selected as true actives via throughput screening. Using a database constructed from these compounds, five programs were tested for structure-based docking (SBD) performance, the MOE of which showed the highest enrichments and second highest screening rates. Separately, using the same database, pharmacophore screening was performed, reducing 1364 compounds to 287 with no loss in true actives, yielding an enrichment of 4.75. When SBD was retested with the pharmacophore filtered database, 4 of the 5 SBD programs showed significant improvements to enrichment rates at only 2.5% of the database, with a 7-fold decrease in an average VS time. Our results altogether suggest that combinatorial approaches of VS technologies are easily applicable to small molecule kinases and, moreover, that such methods can decrease the variability associated with single-method SBD approaches.


Asunto(s)
Bases de Datos Factuales , Ensayos Analíticos de Alto Rendimiento , Modelos Moleculares , Fosfofructoquinasa-2/antagonistas & inhibidores , Fosfofructoquinasa-2/química , Diseño de Fármacos , Humanos , Ligandos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA