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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1833(5): 1212-21, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23403278

RESUMEN

The pp32 (ANP32A) gene acts as a tumor suppressor while its closely related homologue pp32r1 (ANP32C) is oncogenic and is overexpressed in breast, prostate and pancreatic tumors. The transduction of p53wt cell lines (ACHN and HeLa) with pp32r1 or pp32r1Y140H lentivirus increased the proliferation of p53wt cell lines compared to the untransduced control cells while transduction of the p53(R248W) MiaPaCa2 cell line had no effect. Cell cycle analysis of transduced ACHN cells by PI staining and BrdU incorporation illustrated a pronounced shift toward the S-phase of the cell cycle in cells overexpressing the pp32r1 and pp32r1Y140H proteins. Confocal microscopy and western blotting demonstrated that pp32r1 and the pp32r1Y140H mutant protein reside predominantly in the cytoplasm in constrast to pp32 which is a nuclear/cytoplasmic shuttling protein. To determine the effects of pp32r1 or pp32r1Y140H overexpression at the proteomic level we performed a comprehensive proteome analysis on ACHN, ACHN-pp32r1 and ACHN-pp32r1Y140H cell lysates using the isotope-coded protein label (ICPL) method. Among those proteins with >40% regulation were Macrophage Capping protein (CAPG) and Chromodomain Helicase DNA binding protein 4 (CHD4) proteins which were significantly upregulated by pp32r1 and pp32r1Y140H overexpression. This increase in CHD4 also appears to influence a number of cell cycle regulator genes including; p53, p21 and cyclinD1 as judged by western blotting. Silencing of CHD4 in ACHN-pp32r1Y140H cells using specific shRNA reverted the cell cycle dysregulation caused by pp32r1Y140H expression to that of the untransduced ACHN cell line, suggesting that CHD4 is the prominent effector of the pp32r1/pp32r1Y140H phenotype.


Asunto(s)
Autoantígenos , Puntos de Control del Ciclo Celular/genética , Péptidos y Proteínas de Señalización Intracelular , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2 , Proteínas Nucleares , Fosfoproteínas , Autoantígenos/genética , Autoantígenos/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Ciclina D1/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oncogenes , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN , Proteína p53 Supresora de Tumor/metabolismo
2.
Commun Biol ; 4(1): 1333, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34824367

RESUMEN

Cancer cell plasticity due to the dynamic architecture of interactome networks provides a vexing outlet for therapy evasion. Here, through chemical biology approaches for systems level exploration of protein connectivity changes applied to pancreatic cancer cell lines, patient biospecimens, and cell- and patient-derived xenografts in mice, we demonstrate interactomes can be re-engineered for vulnerability. By manipulating epichaperomes pharmacologically, we control and anticipate how thousands of proteins interact in real-time within tumours. Further, we can essentially force tumours into interactome hyperconnectivity and maximal protein-protein interaction capacity, a state whereby no rebound pathways can be deployed and where alternative signalling is supressed. This approach therefore primes interactomes to enhance vulnerability and improve treatment efficacy, enabling therapeutics with traditionally poor performance to become highly efficacious. These findings provide proof-of-principle for a paradigm to overcome drug resistance through pharmacologic manipulation of proteome-wide protein-protein interaction networks.


Asunto(s)
Epigénesis Genética , Genoma , Chaperonas Moleculares/genética , Neoplasias/genética , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Animales , Femenino , Xenoinjertos , Humanos , Ratones , Transducción de Señal
3.
J Cell Biol ; 219(4)2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32328634

RESUMEN

Diverse stresses, including reactive oxygen species (ROS), ionizing radiation, and chemotherapies, activate acid sphingomyelinase (ASMase) and generate the second messenger ceramide at plasma membranes, triggering apoptosis in specific cells, such as hematopoietic cells and endothelium. Ceramide elevation drives local bilayer reorganization into ceramide-rich platforms, macrodomains (0.5-5-µm diameter) that transmit apoptotic signals. An unresolved issue is how ASMase residing within lysosomes is released extracellularly within seconds to hydrolyze sphingomyelin preferentially enriched in outer plasma membranes. Here we show that physical damage by ionizing radiation and ROS induces full-thickness membrane disruption that allows local calcium influx, membrane lysosome fusion, and ASMase release. Further, electron microscopy reveals that plasma membrane "nanopore-like" structures (∼100-nm diameter) form rapidly due to lipid peroxidation, allowing calcium entry to initiate lysosome fusion. We posit that the extent of upstream damage to mammalian plasma membranes, calibrated by severity of nanopore-mediated local calcium influx for lysosome fusion, represents a biophysical mechanism for cell death induction.


Asunto(s)
Apoptosis , Membrana Celular/metabolismo , Lisosomas/metabolismo , Radioisótopos de Carbono , Humanos , Células Jurkat , Especies Reactivas de Oxígeno/metabolismo , Esfingomielinas/química
4.
Cancer Biol Ther ; 15(3): 289-96, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24335183

RESUMEN

pp32r1 (ANP32C) is oncogenic and has been shown to be overexpressed in tumors of the breast, prostate, and pancreas. In this work we show that pp32 family proteins are able to bind to the sphingosine analog FTY720 (Finguimod). Molecular docking studies highlight that a conserved residue F136 is likely to be a key determinant of the FTY720 binding site on the pp32 leucine-rich repeat domain. Transduction of the renal carcinoma cell line ACHN or cervical cancer cell line HeLa with lentivirus expressing the oncogenic family member pp32r1 or a pp32r1Y140H functional mutant illustrated an enhanced resistance to FTY720 induced apoptosis. These findings highlight that certain cancers overexpressing pp32r1 or pp32r1 mutants are likely to demonstrate enhanced resistance to FTY720 treatment.


Asunto(s)
Resistencia a Antineoplásicos/genética , Inmunosupresores/farmacología , Proteínas Nucleares/genética , Fosfoproteínas/genética , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Clorhidrato de Fingolimod , Vectores Genéticos , Humanos , Inmunosupresores/química , Lentivirus/genética , Simulación del Acoplamiento Molecular , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Oncogenes , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Glicoles de Propileno/química , Estructura Terciaria de Proteína , Secuencias Repetitivas de Aminoácido , Esfingosina/química , Esfingosina/farmacología , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA