Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Med Rep ; 14(5): 4436-4444, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27633628

RESUMEN

Schiff base copper (II) complexes are known for their anticancer, antifungal, antiviral and anti­inflammatory activities. The aim of the current study was to investigate biological effects of Schiff base Cu (II) complexes (0.001­100 µmol/l)­[Cu2(sal­D, L­glu)2(isoquinoline)2]·2C2H5OH (1), [Cu(sal­5­met­L­glu)(H2O)].H2O (2), [Cu(ethanol)2(imidazole)4][Cu2(sal­D, L-glu)2(imidazole)2] (3), [Cu(sal­D,L­glu)(2­methylimidazole)] (4) on the human colon carcinoma cells HT­29, the mouse noncancerous cell line NIH­3T3 and the human noncancerous fibroblast cell line VH10. The results suggested that Cu (II) complexes exhibit cytotoxic effects against the HT­29 cell line, while complexes 3 and 4 were the most effective. Subsequent to 72 h of incubation, apoptosis was observed in the HT­29 cells induced by Cu (II) complexes 1 (0.1, 1, 10 and 50 µmol/l), 2 (1, 10, 50 and 100 µmol/l), 3 (0.01, 1, 10 and 50 µmol/l) and 4 (0.01, 0.1, 1 and 10 µmol/l). The apoptotic pathways activated by the Cu (II) complexes were identified. The results indicated that complexes 2, 3 and 4 were able to induce the mitochondria­dependent pathway of apoptosis in HT­29 cells, while complex 1 was obsered to activate the extrinsic pathway of apoptosis. The levels of the anti­apoptotic protein Bcl­2 were reduced and those of the pro­apoptotic protein Bax increased following treatment with complexes 2, 3 and 4. Complex 1 had no effect on Bax protein expression. Complexes 2 and 3 induced elevation of cytochrome c (cyt c), while complex 4 induced a time­dependent elevation of cyt c levels. No cyt c was detected in HT­29 cells exposed to complex 1, suggesting that Cu (II) complexes activated the extrinsic pathway of apoptosis. The results from the current study in addition to previous studies suggest that Schiff base Cu (II) complexes have potential as novel anticancer drugs.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Cobre/administración & dosificación , Mitocondrias/efectos de los fármacos , Bases de Schiff/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HT29 , Humanos , Ratones , Proteínas de Neoplasias/biosíntesis , Transducción de Señal/efectos de los fármacos
2.
Pharmacol Rep ; 68(6): 1221-1224, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27665074

RESUMEN

BACKGROUND: Autophagy plays an important role in cancer cells. Targeting autophagy in cancer can provide new opportunities for drug development. METHODS: In this study we tested four Schiff base Cu(II) complexes against human breast cancer cells (MCF-7) and human non-cancerous cells (HEK-293T). We have tested their cytotoxic effect by evaluating IC50 using MTT test. To detect morphological changes of the actin fibers we have used fluorescent microscopy. To determine the type of cell death we used electrophoretic analysis and western blot analysis (protein LC3). RESULTS: IC50 values of the complexes increased with time of their influence, indicating acquired resistance of MCF-7 to the complexes. Healthy cells HEK-293T were not sensitive to the Cu(II) complexes. Compared with the control cells (cells without Cu(II) complexes) which were without morphological changes of actin fibers, Cu(II) complexes induced condensation and asymmetric conformational changes in actin filaments. To examine the type of cell death induced by the Cu(II) complexes we treated MCF-7 cells with Cu(II) complexes (1, 10, 50 and 100µmol/L) during a 72h incubation period. By electrophoresis we have not detected any DNA fragmentation. To determine whether Cu(II) complexes induced autophagy or necrotic cell death we used the western blot analysis. MCF-7 cells influenced with tested Cu(II) complexes produced LC3 protein after their 72h incubation indicating autophagy in MCF-7 cancer cells. CONCLUSIONS: Tested Schiff base copper (II) complexes have antiproliferative activity against cancer cells but not against healthy cells. They have induced autophagy in the cancer cell line MCF-7.


Asunto(s)
Autofagia/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cobre/toxicidad , Animales , Autofagia/fisiología , Proliferación Celular/fisiología , Cobre/farmacología , Células HEK293 , Humanos , Células MCF-7 , Ratones , Bases de Schiff/farmacología , Bases de Schiff/toxicidad
3.
Cell Rep ; 10(3): 429-440, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25600876

RESUMEN

Cajal bodies (CBs) are evolutionarily conserved nuclear structures involved in the metabolism of spliceosomal small nuclear ribonucleoprotein particles (snRNPs). CBs are not present in all cell types, and the trigger for their formation is not yet known. Here, we depleted cells of factors required for the final steps of snRNP assembly and assayed for the presence of stalled intermediates in CBs. We show that depletion induces formation of CBs in cells that normally lack these nuclear compartments, suggesting that CB nucleation is triggered by an imbalance in snRNP assembly. Accumulation of stalled intermediates in CBs depends on the di-snRNP assembly factor SART3. SART3 is required for both the induction of CB formation as well as the tethering of incomplete snRNPs to coilin, the CB scaffolding protein. We propose a model wherein SART3 monitors tri-snRNP assembly and sequesters incomplete particles in CBs, thereby allowing cells to maintain a homeostatic balance of mature snRNPs in the nucleoplasm.

4.
Eur J Pharmacol ; 721(1-3): 178-84, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24113525

RESUMEN

The aim of our study was to estimate cytostatic/cytotoxic activity of the copper(II) Schiff base complex of the composition [Cu(N-salicylidene-l-glutamato)(H2O)2]·H2O, further Cu(SG-L)H2O, against human colon carcinoma cell line HT-29, as well as to determine type of cell death and to find out the molecular mechanism of apoptosis induced by this complex. Two highest concentrations (50, 100 µmol/l) of the complex showed a strong cytotoxic activity against human colon carcinoma cells HT-29 after 72 h of influence. Other concentrations had a cytostatic activity. Unchelated copper(II) ions and free ligands had no effect on the cell growth. Cu(SG-L)H2O preferentially reduced cancer cell viability compared to healthy cells (NIH-3T3). Cu(SG-L)H2O induced apoptosis of cells HT-29 at all concentrations used (1-100 µmol/l) after 48 h of influence. Apoptosis was carried out by the mitochondrial pathway with active caspases 3 and 9. By the spin-trapping technique combined with electron paramagnetic resonance we found that our complex is photochemically stable in aqueous systems and does not exhibit radical-scavenging activity when 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonate) cation radical was used as an oxidant. The complex exhibits a strong prooxidant property in the initial stages of thermal decomposition of K2S2O8 in water solutions leading to the massive production of (·)OH radicals. Therefore, this complex could strongly participate in anticancer action via a free radical mechanism.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Antioxidantes/química , Antioxidantes/farmacología , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células HT29 , Humanos , Enlace de Hidrógeno , Bases de Schiff/química , Relación Estructura-Actividad
5.
Lipids Health Dis ; 12: 126, 2013 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-23961716

RESUMEN

BACKGROUND: Statins (HMG-CoA reductase inhibitors) represent a major class of compounds for the treatment of hypercholesterolemia due to their ability to inhibit de novo cholesterol synthesis. In addition to their hypolipidemic effects, chemoprotective properties have been attributed to statins as well. These effects involve multiple mechanisms, which, however, are not known in detail. The aim of our study was to assess in non-malignant as well as cancer cells the impact of simvastatin on the amount of cytosolic lipid droplets (LDs) implicated in many biological processes including proliferation, inflammation, carcinogenesis, apoptosis, necrosis or growth arrest. METHODS: Human embryonic kidney cells HEK-293T and human pancreatic cancer cells MiaPaCa-2 were treated with simvastatin (6 and 12 µM) for 24 and 48 hours respectively. Neutral lipid probe Nile Red was used for detection of LDs by fluorescence microscopy. Cellular cholesterol content was determined by HPLC. Changes in expression of genes related to lipid metabolism in simvastatin-treated MiaPaCa-2 cells were examined by DNA microarray analysis. Validation of gene expression changes was performed using quantitative RT-PCR. RESULTS: The treatment of the cells with simvastatin increased their intracellular content of LDs in both non-malignant as well as cancer cells, partially due to the uptake of cholesterol and triacylglyceroles from medium; but in particular, due to enhanced synthesis of triacylglyceroles as proved by significant overexpression of genes related to de novo synthesis of triacylglyceroles and phospholipids. In addition, simvastatin also markedly influenced expression of genes directly affecting cell proliferation and signaling. CONCLUSIONS: Simvastatin treatment led to accumulation of cytosolic LDs within the examined cells, a phenomenon which might contribute to the antiproliferative effects of statins.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Gotas Lipídicas/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Simvastatina/farmacología , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Colesterol/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293/efectos de los fármacos , Humanos , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Neoplasias Pancreáticas/metabolismo
6.
Biochem Biophys Res Commun ; 436(2): 240-5, 2013 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-23726919

RESUMEN

UBL5 protein, a structural homologue of ubiquitin, was shown to be involved in pre-mRNA splicing and transcription regulation in yeast and Caenorhabditis elegans, respectively. However, role of the UBL5 human orthologue is still elusive. In our study, we observed that endogenous human UBL5 that was localized in the nucleus, partially associates with Cajal bodies (CBs), nuclear domains where spliceosomal components are assembled. Simultaneous expression of exogenous UBL5 and coilin resulted in their nuclear colocalization in HeLa cells. The ability of UBL5 to interact with coilin was proved by GST pull-down assay using coilin that was either in vitro translated or extracted from HEK293T cells. Further, our results showed that the UBL5-coilin interaction was not influenced by coilin phosphorylation. These results suggest that UBL5 could be targeted to CBs via its interaction with coilin. Relation between human UBL5 protein and CBs is in the agreement with current observations about yeast orthologue Hub1 playing important role in alternative splicing.


Asunto(s)
Cuerpos Enrollados/metabolismo , Proteínas del Ojo/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitinas/metabolismo , Proteínas del Ojo/genética , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Células HEK293 , Células HeLa , Humanos , Microscopía Fluorescente , Proteínas Nucleares/genética , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Ubiquitinas/genética
7.
Biochim Biophys Acta ; 1823(4): 911-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22306003

RESUMEN

Small ubiquitin-related modifiers 1, 2 and 3 (SUMO-1, -2, -3), members of the ubiquitin-like protein family, can be conjugated to various cellular proteins. Conjugates of SUMO-2 and SUMO-3 (SUMO-2/3) accumulate in cells exposed to various stress stimuli or to MG132 treatment. Although the proteins modified by SUMO-2/3 during heat shock or under MG132 treatment have been identified, the significance of this modification remains unclear. Our data show that the inhibition of translation by puromycin or cycloheximide blocks both the heat shock and MG132 induced accumulation of SUMO-2/3 conjugates in HEK 293T and U2OS cells. However, the heat shock induced accumulation of SUMO-2/3 conjugates was restored by proteasome inhibition, which suggests that the inhibition of translation did not abolish SUMOylation itself. Furthermore, we show that some of the proteins truncated due to the treatment by low concentration of puromycin are SUMOylated in HEK 293T cells. We suggest that the SUMO-2/3 conjugates accumulating under the heat shock or MG132 treatment result largely from new protein synthesis and that portion of them is incorrectly folded.


Asunto(s)
Respuesta al Choque Térmico/efectos de los fármacos , Leupeptinas/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Benzoquinonas/farmacología , Cicloheximida/farmacología , Células HEK293 , Células HeLa , Humanos , Lactamas Macrocíclicas/farmacología , Modelos Biológicos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Inhibidores de la Síntesis de la Proteína/farmacología , Puromicina/farmacología , Sumoilación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA