Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
BMC Cancer ; 13: 440, 2013 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-24073922

RESUMEN

BACKGROUND: The t(6;9)(p23;q34) chromosomal translocation is found in 1% of acute myeloid leukemia and encodes the fusion protein DEK-NUP214 (formerly DEK-CAN) with largely uncharacterized functions. METHODS: We expressed DEK-NUP214 in the myeloid cell lines U937 and PL-21 and studied the effects on cellular functions. RESULTS: In this study, we demonstrate that expression of DEK-NUP214 increases cellular proliferation. Western blot analysis revealed elevated levels of one of the key proteins regulating proliferation, the mechanistic target of rapamycin, mTOR. This conferred increased mTORC1 but not mTORC2 activity, as determined by the phosphorylation of their substrates, p70 S6 kinase and Akt. The functional importance of the mTOR upregulation was determined by assaying the downstream cellular processes; protein synthesis and glucose metabolism. A global translation assay revealed a substantial increase in the translation rate and a metabolic assay detected a shift from glycolysis to oxidative phosphorylation, as determined by a reduction in lactate production without a concomitant decrease in glucose consumption. Both these effects are in concordance with increased mTORC1 activity. Treatment with the mTORC1 inhibitor everolimus (RAD001) selectively reversed the DEK-NUP214-induced proliferation, demonstrating that the effect is mTOR-dependent. CONCLUSIONS: Our study shows that the DEK-NUP214 fusion gene increases proliferation by upregulation of mTOR, suggesting that patients with leukemias carrying DEK-NUP214 may benefit from treatment with mTOR inhibitors.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Expresión Génica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Oncogénicas/genética , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteínas Cromosómicas no Histona/metabolismo , Glucólisis , Humanos , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Fosforilación Oxidativa , Proteínas de Unión a Poli-ADP-Ribosa , Biosíntesis de Proteínas , Transducción de Señal , Células U937
2.
Biol Cell ; 104(8): 462-75, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22509910

RESUMEN

BACKGROUND INFORMATION: The interferon (IFN)-inducible protein TRIM22 (Staf50) is a member of the tripartite motif protein family and has been suggested a role in the regulation of viral replication as well as of protein ubiquitylation. In addition, we have previously shown that TRIM22 is a direct target gene for the tumour suppressor p53. Consistently, over-expression of TRIM22 inhibits the clonogenic growth of monoblastic U937 cells, suggesting anti-proliferative or cell death-inducing effects. RESULTS: Here, we demonstrate that TRIM22 directly or indirectly interacts with the eukaryotic translation initiation factor (eIF)4E, and inhibits the binding of eIF4E to eIF4G, thus disturbing the assembly of the eIF4F complex, which is necessary for cap-dependent translation. Furthermore, TRIM22 exerts a repressive effect on luciferase reporter protein levels and to some extent on radiolabelled methionine incorporation. Even though all nuclear mRNAs are capped, some are more dependent on eIF4F than others for translation. The translation of one of these mRNAs, IRF-7C, was indeed found to be repressed in the presence of TRIM22. CONCLUSIONS: Our data suggest TRIM22 to repress protein translation preferably of some specific mRNAs. Taken together, we show that TRIM22 represses translation by inhibiting the binding of eIF4E to eIF4G, suggesting a mechanism for regulation of protein translation, which may be of importance in response to p53 and/or IFN signalling.


Asunto(s)
Factor 4E Eucariótico de Iniciación , Factor 4G Eucariótico de Iniciación , Biosíntesis de Proteínas , Proteínas Represoras , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Factor 4G Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Factor 7 Regulador del Interferón/antagonistas & inhibidores , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferones/metabolismo , Antígenos de Histocompatibilidad Menor , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas de Motivos Tripartitos , Proteína p53 Supresora de Tumor/metabolismo
3.
Exp Cell Res ; 317(8): 1179-91, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21324313

RESUMEN

Prenylation is a post-translational hydrophobic modification of proteins, important for their membrane localization and biological function. The use of inhibitors of prenylation has proven to be a useful tool in the activation of apoptotic pathways in tumor cell lines. Rab geranylgeranyl transferase (Rab GGT) is responsible for the prenylation of the Rab family. Overexpression of Rab GGTbeta has been identified in CHOP refractory diffuse large B cell lymphoma (DLBCL). Using a cell line-based model for CHOP resistant DLBCL, we show that treatment with simvastatin, which inhibits protein farnesylation and geranylgeranylation, sensitizes DLBCL cells to cytotoxic treatment. Treatment with the farnesyl transferase inhibitor FTI-277 or the geranylgeranyl transferase I inhibitor GGTI-298 indicates that the reduction in cell viability was restricted to inhibition of geranylgeranylation. In addition, treatment with BMS1, a combined inhibitor of farnesyl transferase and Rab GGT, resulted in a high cytostatic effect in WSU-NHL cells, demonstrated by reduced cell viability and decreased proliferation. Co-treatment of BMS1 or GGTI-298 with CHOP showed synergistic effects with regard to markers of apoptosis. We propose that inhibition of protein geranylgeranylation together with conventional cytostatic therapy is a potential novel strategy for treating patients with CHOP refractory DLBCL.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/patología , Transferasas Alquil y Aril/antagonistas & inhibidores , Transferasas Alquil y Aril/metabolismo , Anticuerpos Monoclonales de Origen Murino/farmacología , Antineoplásicos/farmacología , Benzamidas/farmacología , Ciclo Celular/fisiología , Supervivencia Celular , Ciclofosfamida/farmacología , Ciclofosfamida/uso terapéutico , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Farnesiltransferasa/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Metionina/análogos & derivados , Metionina/farmacología , Prednisona/farmacología , Prednisona/uso terapéutico , Prenilación , Rituximab , Simvastatina/farmacología , Vincristina/farmacología , Vincristina/uso terapéutico
4.
Genes Chromosomes Cancer ; 47(4): 276-87, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18181180

RESUMEN

The t(6;9)(p22;q34) chromosomal translocation is found in a subset of patients with acute myeloid leukemia (AML). The translocation results in a fusion between the nuclear phosphoprotein DEK and the nucleoporin NUP214 (previously CAN). The mechanism by which the fusion protein DEK-NUP214 contributes to leukemia development has not been identified, and disruptions of normal cellular functions by DEK-NUP214 have previously not been described. In the present study, a novel effect of the DEK-NUP214 fusion protein is demonstrated. Our findings reveal a substantial increase in global protein synthesis in DEK-NUP214 expressing cells. Furthermore, we conclude that this effect is not the result of dysregulated transcription but merely due to increased translation. Consistent with the association with AML, the increased protein synthesis mediated by DEK-NUP214 is restricted to cells of the myeloid lineage. Analysis of potential mechanisms for regulating protein synthesis shows that expression of DEK-NUP214 correlates to the phosphorylation of the translation initiation protein, EIF4E. The present data provide evidence that increase of translational activity constitutes a mechanism by which the leukemogenic effect of DEK-NUP124 may be mediated.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteínas de Fusión Oncogénica/fisiología , Biosíntesis de Proteínas , Apoptosis , Western Blotting , Factor 4E Eucariótico de Iniciación/metabolismo , Citometría de Flujo , Regulación Leucémica de la Expresión Génica/fisiología , Genes Reporteros , Vectores Genéticos , Humanos , Leucemia Mieloide Aguda/genética , Luciferasas/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia , Transcripción Genética , Transfección , Células Tumorales Cultivadas
5.
Haematologica ; 91(2): 268-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16461319

RESUMEN

The role of the DEK protein, involved in the leukemia-associated fusion protein DEK-CAN, is not yet known. In this study, we show a higher expression of DEK mRNA in immature cells than in mature cells. Furthermore, a correlation between DEK expression and cell proliferation was demonstrated, suggesting that DEK plays a role in the proliferation of hematopoietic cells and raising the question of whether the DEK-CAN fusion protein might perturb regulation of proliferation in leukemic cells.


Asunto(s)
Proliferación Celular , Proteínas Cromosómicas no Histona/fisiología , Regulación de la Expresión Génica , Proteínas Oncogénicas/fisiología , Células Sanguíneas , Células de la Médula Ósea , Proteínas Cromosómicas no Histona/genética , Humanos , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/fisiología , ARN Mensajero/análisis
6.
Am J Transl Res ; 5(2): 170-83, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23573362

RESUMEN

Epigenetic code modifications by histone deacetylase inhibitors (HDACis) have recently been proposed as potential new therapies for hematological malignancies. Diffuse large B-cell lymphoma (DLBCL) is the most common form of aggressive lymphoma. At present, standard first line treatment for DLBCL patients is the antracycline-based chemotherapy regimen CHOP (cyclophosphamide, doxorubicin, vincristine and prednisone) combined with the monoclonal anti-CD20 antibody rituximab (R-CHOP). Since only 50-60% of patients reach a long-time cure by this treatment, there is an urgent need for novel treatment strategies to increase the response and long-term remission to initial R-CHOP therapy. In this study, we investigated the effect of the HDAC inhibitor valproic acid (VPA) on DLBCL cell lines. To elucidate the effects of VPA on chemo-sensitivity, we used a cell-line based model of CHOP-refractory DLBCL. All five DLBCL cell lines treated with VPA alone or in combination with CHOP showed decreased viability and proliferation. The VPA-induced sensitization of DLBCL cells to cytotoxic treatment resulted in increased number of apoptotic cell as judged by annexin V-positivity and the presence of cleaved caspase-3. In addition, pretreatment with VPA resulted in a significantly increased DNA-damage as compared to CHOP alone. In summary, HDAC inhibitors such as VPA, are promising therapeutic agents in combination with R-CHOP for patients with DLBCL.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA