Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Clin Med ; 10(11)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070472

RESUMEN

Sarcomas are a heterogeneous group of mesenchymal tumours, with a great variability in their clinical behaviour. While our knowledge of sarcoma initiation has advanced rapidly in recent years, relatively little is known about mechanisms of sarcoma progression. JUN-murine fibrosarcoma progression series consists of four sarcoma cell lines, JUN-1, JUN-2, JUN-2fos-3, and JUN-3. JUN-1 and -2 were established from a single tumour initiated in a H2K/v-jun transgenic mouse, JUN-3 originates from a different tumour in the same animal, and JUN-2fos-3 results from a targeted in vitro transformation of the JUN-2 cell line. The JUN-1, -2, and -3 cell lines represent a linear progression from the least transformed JUN-2 to the most transformed JUN-3, with regard to all the transformation characteristics studied, while the JUN-2fos-3 cell line exhibits a unique transformation mode, with little deregulation of cell growth and proliferation, but pronounced motility and invasiveness. The invasive sarcoma sublines JUN-2fos-3 and JUN-3 show complex metabolic profiles, with activation of both mitochondrial oxidative phosphorylation and glycolysis and a significant increase in spared respiratory capacity. The specific transcriptomic profile of invasive sublines features very complex biological relationships across the identified genes and proteins, with accentuated autocrine control of motility and angiogenesis. Pharmacologic inhibition of one of the autocrine motility factors identified, Ccl8, significantly diminished both motility and invasiveness of the highly transformed fibrosarcoma cell. This progression series could be greatly valuable for deciphering crucial aspects of sarcoma progression and defining new prognostic markers and potential therapeutic targets.

2.
Int J Gynecol Cancer ; 25(2): 236-43, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25594141

RESUMEN

OBJECTIVE: This study aimed to evaluate the correlation between the expressions of lung resistance protein (LRP), P-glycoprotein (Pgp), multidrug resistance-associated protein (MRP)-1, MRP3, and MRP5 and histopathological parameters and clinical outcome, and to determine the predictive and prognostic value of these transport proteins in patients with ovarian cancer. METHODS: Tumor samples from 111 chemonaive patients with epithelial ovarian cancer who underwent primary surgery from 2006 to 2010 were immunohistochemically stained for LRP, Pgp, MRP1, MRP3, and MRP5 expressions. RESULTS: MRP1 expression was greater among patients with late disease than among patients with early stage ovarian cancer [International Federation of Gynecology and Obstetrics (FIGO) I + II, 71.6% (confidence interval, 60-100); FIGO III + IV, 83.6% (confidence interval, 100-100); P = 0.03]. The histological subtype correlated with the expressions of LRP, Pgp, MRP1, and MRP3. Relapse of disease during the next 24 months occurred more often among patients with higher Pgp and MRP1 than among patients with lower Pgp and MRP1 expressions. FIGO stage, histological type, debulking efficiency, strong Pgp expression, and strong MRP1 expression correlated significantly with shorter progression-free survival (log-rank test, P = 0.001, P = 0.004, P = 0.001, P = 0.051, and P = 0.046, respectively). FIGO stage, histological type, debulking efficiency, and strong MRP1 expression correlated with poor patient survival (log-rank test, P = 0.001, P = 0.042, P = 0.005, and P = 0.018, respectively). CONCLUSIONS: Pgp and MRP1 expressions were clinically significant in patients with ovarian cancer. Pgp and MRP1 may be reliable, independent predictive and prognostic factors regarding the clinical outcome of ovarian cancer. MRP3 is less important as a predictive and prognostic factor than MRP1 expression. MRP5 and LRP expressions were not applicable prognostic parameters regarding ovarian cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Resistencia a Antineoplásicos , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Epitelial de Ovario , Femenino , Humanos , Persona de Mediana Edad , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/mortalidad , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Pronóstico , Análisis de Supervivencia , Partículas Ribonucleoproteicas en Bóveda/metabolismo
3.
Toxicol Lett ; 231(1): 29-37, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25204997

RESUMEN

Effects of chronic exposure to supranutritional sodium selenite (Se) were investigated in colonic fibroblasts. Initially, Se did not produce any gross changes in exposed cells; however, basal levels of autophagy were transiently increased and p38 activity was stimulated. From the 3rd week onwards, Se decreased cell proliferation, with corrensponding changes in cell cycle distribution. Also, in exposed cells oxidative stress and DNA damage slowly but gradually increased along with decreasing mitochondrial function and upon continued elevated activity of p38 kinase. Towards the end of the experiment, premature senescence features became more prominent in treated cells. Pharmacological inhibition as well as gene knockdown of these processes confirmed the involvement of p38 in balancing autophagy and premature senescence in cells exposed to Se and suggests that this element may in a given time frame compromise selected cell populations in digestive system.


Asunto(s)
Autofagia/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Colon/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Selenito de Sodio/toxicidad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Colon/enzimología , Colon/patología , Daño del ADN , Metilación de ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Activación Enzimática , Fibroblastos/enzimología , Fibroblastos/patología , Humanos , Mitocondrias/enzimología , Mitocondrias/patología , Estrés Oxidativo/efectos de los fármacos , Fenotipo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección
4.
Anticancer Agents Med Chem ; 13(5): 811-20, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22721392

RESUMEN

The roles of autophagic cell death and apoptosis induced by topoisomerase inhibitor irinotecan in colon cancer cells with deleted p53 were investigated during 48 h. We report that irinotecan-dependent cytotoxicity and proapoptotic activity were reduced in the present model while autophagy levels significantly increased. Upon p53 transfection, cell demise rates increased, with cells bearing the features of apoptosis and autophagic cell death. The subsequent studies into mechanisms of cell death process revealed the important role of Bax in mediating mitochondrial and lysosomal leakage which might serve as leading signals for both apoptosis and autophagic cell death. These results suggest that different modes of cell death in p53 null colon cancer cells treated with cytostatics (irinotecan) may be activated simultaneously. Moreover, their interactions possibly occur at several stages and aren't mutually exclusive. This might thus lead to a potential synergism with interesting therapeutic ramifications.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Camptotecina/análogos & derivados , Neoplasias del Colon/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Antineoplásicos Fitogénicos/uso terapéutico , Apoptosis/fisiología , Autofagia/fisiología , Camptotecina/farmacología , Camptotecina/uso terapéutico , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Células HCT116 , Células HT29 , Humanos , Irinotecán , Proteína p53 Supresora de Tumor/genética
5.
Toxicol Lett ; 214(1): 1-8, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-22898888

RESUMEN

Irinotecan (CPT-11) is topoisomerase I inhibitor used in the treatment of disseminated colorectal cancer. In colon cancer cells it induces DNA damage which leads to cytotoxicity with ensuing apoptosis or premature senescence. Despite its clinical use and efficiency in malignant colonocytes, its effects in normal colonic cells are relatively underexplored. In this work we report that CPT-11 induces dose-dependent cytotoxicity which results in apoptosis and premature senescence whose occurrence nevertheless varies in relation to the type of exposed cells. In normal colonic epithelial cells (NCM) the prevailing type of response is apoptosis whereas in normal colonic fibroblasts (NCF) it is premature senescence. Further analyses showed that CPT-11 induced in both types of cells DNA damage and activated stress response pathways including p53 and p16 but with varying activity of stress kinase p38 and selected stress-associated microRNAs. Epithelial cells upregulated the expression of p53, which was subsequently specifically phosphorylated, massively activated p38 and initiated mitochondrial, caspase-dependent apoptosis. These events occurred in the presence of moderately increased expression of miR-34a only. Conversely, in colonic fibroblasts p38 was only moderately activated, p53 as well as p16 expressions were upregulated in the presence of increased expression of miR-34a, miR-128a and miR-449a. Caspase-dependent apoptosis was found only in a minority of treated cells and the premature senescence phenotype was prevailing. Specific inhibition further proved that p53-dependent as well as independent mechanisms might be responsible for these cell type-specific differences.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Camptotecina/análogos & derivados , Carcinoma/metabolismo , Colon/citología , Fibroblastos/efectos de los fármacos , Camptotecina/farmacología , Carcinoma/genética , Senescencia Celular/efectos de los fármacos , Neoplasias Colorrectales , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Daño del ADN , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células HCT116 , Humanos , Irinotecán , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
Toxicol In Vitro ; 26(2): 258-68, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22200533

RESUMEN

Sodium selenite (Se) is known to induce diverse stress responses in malignant cells which may lead to various types of cell death including apoptosis and/or autophagy. In colon cancer cells, Se activates several signaling pathways whose interactions and ultimate endpoints may vary in individual study models. In our previous work we showed differences in Se-dependent growth inhibition, cell cycle alterations and apoptosis in colon cancer cells with functional (HCT-116) and deleted (HCT-116-p53KO) p53. Moreover, detailed morphological and biochemical analyses revealed the presence of autophagy in Se-treated cells. Thus the aim of this study was to investigate in detail mechanisms, relationship and crosstalk between apoptosis and autophagy in Se-treated HCT-116 cancer cells differing in p53 status since p53 has been shown to play a well-known role in apoptosis but dichotomous role in autophagy. We report that the absence of p53 in malignant colonocytes changes patterns of response to Se-induced stress which include differential activation of MAP kinases (p38 - HCT-116 and JNK - HCT-116 p53KO) including their respective roles in the process of apoptosis and autophagy as well as the involvement of mTOR or PI3K signaling. Our results seem to suggest that deletion of p53 inevitably leads to a higher level of instability and delays in an individual cell decision in the face of stress whether to activate apoptosis or autophagy which may consequently occur simultaneously with mutual dichotomous relationship.


Asunto(s)
Anticarcinógenos/farmacología , Selenito de Sodio/farmacología , Proteína p53 Supresora de Tumor/deficiencia , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Daño del ADN , Técnicas de Silenciamiento del Gen , Células HCT116 , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
7.
Acta Medica (Hradec Kralove) ; 55(3): 116-24, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23297519

RESUMEN

The aim of our study was to determine the effect of selected cytostatics on a human ovarian cancer cell line A2780 as a model system for ovarian cancer treatment. This cell line is considered cisplatin-sensitive. Panel of tested cytostatics included cisplatin, paclitaxel, carboplatin, gemcitabine, topotecan and etoposide. These cytostatics have a different mechanism of action. To evaluate cytotoxic potential of the tested compounds, the methods measuring various toxicological endpoints were employed including morphological studies, MTT assay, dynamic monitoring of cell proliferation with xCELLigence, cell cycle analysis, caspase 3 activity and expression of proteins involved in cell cycle regulation and cell death. The A270 cell line showed different sensitivity towards the selected cytostatics, the highest cytotoxic effect was associated with paclitaxel and topotecan.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales , Neoplasias Ováricas/patología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos
8.
Toxicol Lett ; 204(2-3): 164-73, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21557991

RESUMEN

Zinc pyrithione is used as a topical agent in a range of medicinal and cosmetic applications. Despite its extensive use and reported beneficial effects in treatment of various dermal problems, its potential toxicity towards skin cells remains relatively underexplored. In this work we investigated effects of nM zinc pyrithione on cell stress response pathways of primary human skin fibroblasts during 24h of exposure. We demonstrate that zinc pyrithione-induced cytotoxity in dermal fibroblasts is dose-dependent and it associates with increased intracellular zinc concentrations and activated stress response pathways including p53 and stress kinase p38. Higher zinc pyrithione concentrations (500nM and above) stimulate oxidative stress and moderate DNA damage which occur in the presence of activated p38 kinase. Cells further upregulate the expression of p53 which increases its transcriptional activity while mitogenic signaling exemplified by mTOR (mammalian target of rapamycin) expression is suppressed and these steps lead to mitochondrial, caspase-dependent apoptosis. Conversely, lower zinc concentrations (125nM) fail to induce oxidative stress and significant DNA damage; however, treated cells still activate p38 and upregulate the expression and transcriptional activity of p53 and its target gene p21 as well as the expression of p16 in the presence of active mTOR pathway and a changed DNA methylation pattern. The end result is premature senescence phenotype. Specific pharmacological inhibitors as well as gene knockdown technology prove that an interaction between p38, p53 and mTOR might be responsible for these observed endpoints. Taken together, exposure of dermal fibroblasts to varying concentrations of zinc pyrithione may result in either cell death-apoptosis or cellular premature senescence which attests to the ability of this compound to affect this type of cells in an in vitro model system.


Asunto(s)
Fibroblastos/efectos de los fármacos , Queratolíticos/toxicidad , Compuestos Organometálicos/toxicidad , Piridinas/toxicidad , Piel/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , ADN/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Piel/citología , Proteína p53 Supresora de Tumor/fisiología
9.
Toxicol In Vitro ; 25(7): 1302-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21557998

RESUMEN

Mechanisms and pathways responsible for cytotoxicity of sulforaphane (SF) in colon cancer cells with deleted p53 were investigated during 48 h of exposure. SF showed dose-dependent cytotoxicity and proapoptotic activity in the present model. In addition, in HCT-116 p53KO cells SF induced DNA damage with the subsequent cellular response and signaling not including p53 and caspase-2 pathways. Conversely, in SF-treated cells JNK was activated which led to an early lysosomal membrane permeabilization, release of cathepsin B and D and activation of Bid by specific cleavage. Concomitantly, the expression of Bax increased in the presence of JNK-mediated Bcl-2 inhibition which was followed by mitochondrial release of cytochrome c and activation of apoptosis. These results suggest that SF may be useful as a chemopreventive agent in colon cancer with inactivated or lost p53.


Asunto(s)
Anticarcinógenos/toxicidad , Neoplasias del Colon/metabolismo , Lisosomas/metabolismo , Mitocondrias/metabolismo , Tiocianatos/toxicidad , Proteína p53 Supresora de Tumor/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/prevención & control , Daño del ADN/efectos de los fármacos , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Isotiocianatos , Lisosomas/efectos de los fármacos , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Mitocondrias/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Sulfóxidos , Factores de Tiempo , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
11.
Altern Lab Anim ; 38(2): 119-38, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20507184

RESUMEN

Morphology, motility, proliferation rate and markers of oxidative stress in primary human gingival fibroblasts (GF) and periodontal ligamental fibroblasts (PDL-F) grown in zinc-deficient cultivation medium (ZDM), were studied over a 5-week culture period. A low-zinc environment effectively reduced the total, as well as the free, intracellular zinc content in both cell types, over the course of the experiment. Decreased intracellular zinc content resulted in altered cellular morphology, reduced motility, and rearrangement of actin and tubulin in the cytoskeleton. In addition, fibroblasts with low zinc content exhibited decreased proliferation, accompanied by changes in cell cycle distribution, expression of specific biochemical markers, increased oxidative stress and the activation of caspase-3. Supplementation of ZDM with exogenous zinc prevented the loss of intracellular zinc, while also restoring the morphology, cell proliferation and mitogenic signalling of the cultured cells. Moreover, such supplemented cells were protected against oxidative stress and cell death. Of the two primary cell cultures examined, GF were more sensitive to decreased intracellular zinc content, when compared to PDL-F. The results obtained suggest that the human primary cell cultures can be useful for the longer-term evaluation of the effects of nutritional factors originating from the environment.


Asunto(s)
Fibroblastos/citología , Encía/citología , Ligamento Periodontal/citología , Zinc/toxicidad , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Encía/efectos de los fármacos , Encía/fisiología , Glutatión/metabolismo , Humanos , Ligamento Periodontal/efectos de los fármacos , Ligamento Periodontal/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Zinc/deficiencia , Zinc/farmacología
12.
Toxicol Lett ; 197(2): 143-50, 2010 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-20493934

RESUMEN

We investigated combined effects of hexavalent chromium and nickel on viability, intracellular signaling and cell death of primary human skin fibroblasts during 24 h of exposure. We show that nickel at non-toxic concentrations prevents hexavalent chromium-induced cell damage and apoptosis, mainly by overexpression of heat shock proteins (HSPs), in particular HSP27 and activation of nuclear factor kappa B (NFkappaB) as demonstrated by specific knockdown of HSPs or NFkappaB. Conversely, cytotoxic nickel concentrations which induce apoptosis in dermal fibroblasts by themselves act to enhance hexavalent chromium effects in the same cells by stimulating oxidative stress and depleting ATP leading to rapid necrosis as demonstrated by markedly increased LDH release in exposed cells. Using specific pharmacological inhibitors it was further demonstrated that oxidative stress and PARP-1 activity are responsible for rapid necrosis. In conclusion, exposure of dermal fibroblasts to high nickel concentrations in combination with hexavalent chromium may result in rapid cell damage leading to necrosis while low nickel concentrations may prevent hexavalent chromium-induced cell death with potential accumulation of damaged but otherwise viable cells.


Asunto(s)
Cromo/toxicidad , Dermis/citología , Fibroblastos/efectos de los fármacos , Níquel/toxicidad , Apoptosis/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos
13.
Biometals ; 23(2): 339-54, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20151177

RESUMEN

Increased intracellular free zinc concentrations are associated with activation of several stress signaling pathways, specific organelle injury and final cell death. In the present work we examined the involvement of mitochondria and lysosomes and their crosstalk in free zinc-induced cell demise. We report that treatment of cervical tumor Hep-2 cells with zinc pyrithione leads to an early appearance of cytoplasmic zinc-specific foci with corresponding accumulation of zinc first in mitochondria and later in lysosomes. Concomitant with these changes, upregulation of expression of metallothionein II A gene as well as the increased abundance of its protein occurs. Moreover, zinc activates p53 and its dependent genes including Puma and Bax and they contribute to an observed loss of mitochondrial membrane potential and activation of apoptosis. Conversely, lysosomal membrane permeabilization and its promoted cleavage of Bid occurs in a delayed manner in treated cells and their effect on decrease of mitochondrial membrane potential is limited. The use of specific inhibitors as well as siRNA technology suggest a crucial role of MT-IIA in trafficking of free zinc into mitochondria or lysosomes and regulation of apoptotic or necrotic cell demise.


Asunto(s)
Apoptosis/efectos de los fármacos , Queratolíticos/farmacología , Lisosomas/metabolismo , Mitocondrias/metabolismo , Compuestos Organometálicos/farmacología , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Neoplasias del Cuello Uterino/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Metaloproteinasas de la Matriz/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Neoplasias del Cuello Uterino/genética , Zinc/metabolismo
14.
Toxicol In Vitro ; 23(7): 1406-11, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19607906

RESUMEN

A number of cytotoxicity assays are currently available, each of them using specific approach to detect different aspects of cell viability, such as cell integrity, proliferation and metabolic functions. In this study we compared the potential of five commonly employed cytotoxicity assays (WST-1, XTT, MTT, Brilliant blue and Neutral red assay) to detect antiproliferative effects of three selenium compounds, sodium selenite, seleno-L-methionine (SeMet) and Se-(Methyl)selenocysteine (SeMCys) on three colorectal cancer cell lines in vitro. Cells were exposed to the selected selenium compounds in the concentration range of 0-256 microM during 48 h. WST-1 and XTT failed to detect cytotoxic effect, with the exception of the highest concentration of selenium compounds tested. Conversely, the metabolic activity of selenium treated cells measured by WST-1 and XTT significantly increased in comparison to untreated controls. MTT, Neutral red and Brilliant blue assays were more sensitive and yielded mutually comparable results, with significant decrease of measured parameters in a concentration-dependent manner. To a smaller extent, the results were affected by the different chemical nature of the selenium compounds tested as well as by the biological properties of individual cell lines.


Asunto(s)
Anticarcinógenos/toxicidad , Neoplasias del Colon/tratamiento farmacológico , Citotoxinas/toxicidad , Compuestos de Organoselenio/toxicidad , Selenito de Sodio/toxicidad , Pruebas de Toxicidad/métodos , Anticarcinógenos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/prevención & control , Cisteína/análogos & derivados , Cisteína/uso terapéutico , Cisteína/toxicidad , Citotoxinas/uso terapéutico , Humanos , Metionina/análogos & derivados , Metionina/uso terapéutico , Metionina/toxicidad , Compuestos de Organoselenio/uso terapéutico , Selenocisteína/análogos & derivados , Selenito de Sodio/uso terapéutico
15.
Toxicol In Vitro ; 23(8): 1497-503, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19602434

RESUMEN

Sodium selenite has been reported to interfere with cell growth and proliferation and to induce cell death. Despite of our current knowledge, details about its effects on growth and behavior of colonocytes with differing p53 status remain unknown. In our study, we evaluated the antiproliferative, cell cycle specific and proapoptotic potential of sodium selenite in HCT-116 colorectal cells with wild type p53 and its isogenic control HCT-116-p53KO cell line. Cell proliferation in selenite-treated cells was followed by computer-enhanced time-lapse videomicroscopy, by measuring protein content (Coomassie Brilliant Blue assay), metabolic activity (WST-1) and DNA synthesis (BrdU). Changes in cell cycle were determined by flow cytometry and Western blotting. Cell death was measured with the nuclear fragmentation assay and caspase-3 immunostaining. We show that sodium selenite inhibits the growth and proliferation of colon cancer cells in a time- and dose-dependent manner, with HCT-116 cells being more sensitive than HCT-116-p53KO cells. Moreover, upon sodium selenite treatment, there was a tendency for cells to accumulate at G2 phase which was accompanied by the increasing expression of cyclin B1, Cdc2 p34, p21 and the sub G1 fraction of the cell cycle. In addition, PARP and nuclear fragmentation and activation of caspase-3 were more profound in HCT-116 cells versus HCT-116-p53KO cells, thus indicating important role of p53 and dependent signaling in selenite-induced toxicity.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Selenito de Sodio/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/análisis , Células HCT116 , Humanos , Proteína p53 Supresora de Tumor/fisiología
16.
Cancer Invest ; 27(7): 704-17, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19488905

RESUMEN

Cytotoxicity and apoptosis induced by etoposide were studied during 72 hr in human melanoma cells. Etoposide initiated DNA-damage signaling via ATM kinase and activated p53 pathway and caspase-2. In response to treatment with etoposide, mitochondria of melanoma cells first increased their abundance and activity, and at later treatment intervals their dynamic behavior and functions became suppressed. Observed mitochondrial perturbation was not preceded by membrane potential loss but cytochrome c release was observed together with a rise in caspase-9 and caspase-3 activities. The pharmacological inhibition of relevant induced targets proved the importance of ATM and caspase-2 in etoposide-mediated cytotoxicity and apoptosis.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Etopósido/farmacología , Melanoma/patología , Adenosina Trifosfato/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Benzotiazoles/farmacología , Caspasa 2/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Inhibidores de Caspasas , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Ciclosporina/farmacología , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Daño del ADN , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Superóxidos/metabolismo , Tolueno/análogos & derivados , Tolueno/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
17.
Toxicol Lett ; 188(3): 236-42, 2009 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-19406221

RESUMEN

In this study we examined interactions between human dermal fibroblasts and chromium acetate hydroxide originating from environmental waste sediments. We show that initially exposure of fibroblasts to Cr (III) induced membrane-dependent signaling including activation of Rac1 GTPase, Src and apoptosis signal-regulating kinase 1 (ASK-1) kinases leading to increased activities of p38 and particularly Jun N-terminal kinase (JNK) and subsequent activation of caspase-3. At later treatment intervals (48-96 h), caspase-3 activity became suppressed and markedly increased lactate dehydrogenase (LDH) release was observed. Further experiments demonstrated that LDH release occurred in the presence of increased oxidative stress, extensive DNA damage, overactivation of poly(ADP-ribose)polymerase-1 (PARP-1) and depletion of ATP. Using specific inhibitors it was demonstrated that oxidative stress along with PARP-1 activity are responsible for cell death mode switch and upon their inhibition caspase-3 activity could be restored. In conclusion, Cr (III) seems to induce a biphasic response in dermal fibroblasts, with initial apoptosis switched to necrosis via increased DNA damage and resulting PARP-1 activity.


Asunto(s)
Acetatos/toxicidad , Apoptosis/efectos de los fármacos , Cromo/toxicidad , Contaminantes Ambientales/toxicidad , Fibroblastos/efectos de los fármacos , Piel/citología , Proteína de Unión al GTP rac1/metabolismo , Familia-src Quinasas/metabolismo , Adenosina Trifosfato/metabolismo , Caspasa 3/biosíntesis , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Daño del ADN , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/enzimología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Poli(ADP-Ribosa) Polimerasas/metabolismo , Eliminación de Residuos , Proteína de Unión al GTP rac1/biosíntesis , Familia-src Quinasas/biosíntesis
18.
Arch Toxicol ; 83(4): 363-72, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19020865

RESUMEN

The aim of the study was to evaluate time course and dose dependence of peroxidative damage induced by tert-butyl hydroperoxide (tBHP) in rat hepatocytes cultured in suspension and in monolayer. At the lowest (0.1 mM) concentration, decrease of cytosolic glutathione and discharge of mitochondrial membrane potential (MMP) could be detected. Significant increases in leakage of lactate dehydrogenase and in malondialdehyde concentrations together with decrease of pyruvate-dependent respiration were detected at higher tBHP concentrations (above 0.5 mM) and after longer periods of incubation. Changes in plasma membrane integrity were observed at 1 mM concentration of tBHP. Succinate-dependent oxidation was most resistant to peroxidative damages. Opening of the mitochondrial permeability transition pore was responsible for the discharge of mitochondria membrane potential. In the presence of cyclosporine A and succinate, the membrane potential could be restored. Our data showed that the most sensitive indicators of the peroxidative damage are changes of cytosolic glutathione concentration and MMP.


Asunto(s)
Citosol/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , terc-Butilhidroperóxido/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citosol/metabolismo , Relación Dosis-Respuesta a Droga , Glutatión/metabolismo , Hepatocitos/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Malondialdehído/metabolismo , Mitocondrias Hepáticas/enzimología , Mitocondrias Hepáticas/fisiología , Estrés Oxidativo/fisiología , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Wistar
19.
Acta Medica (Hradec Kralove) ; 51(1): 51-7, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18683670

RESUMEN

Zinc has been shown to have inhibitory effects on proliferation and metabolism of malignant colonocytes. Still, there is no information available concerning putative effects of zinc against motility and migration of colon cancer cells. Using fluorescence microscopy, immunoblotting and microflorimetry we show that treatment with zinc sulfate affected motility, invasiveness, cytoskeletal integrity and expression of selected markers (E-cadherin, catenin, vimentin, tubulin and actin) of invasive SW480 colon tumor cells. These results emphasize the possible multitudinous role of zinc in the process of colon cancer development and hint at the potential of this element in chemoprevention of advanced colorectal carcinoma.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/fisiopatología , Citoesqueleto/efectos de los fármacos , Zinc/farmacología , Actinas/metabolismo , Cadherinas/metabolismo , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Microtúbulos/efectos de los fármacos , Invasividad Neoplásica , Células Tumorales Cultivadas , Vimentina/metabolismo
20.
Anticancer Agents Med Chem ; 8(6): 598-602, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18690824

RESUMEN

Colorectal cancer is among the most common cancers worldwide, in terms of overall mortality. Environmental factors have been identified to play the most important roles in the development of this disease, in particular diet and its specific components. Selenium is an important micronutrient engaged in the protection of colonic cells against a wide range of external and internal stressors. In addition, selenium has been reported to actively inhibit growth of malignant colonic cells as well as to induce their demise. Furthermore, besides its promising chemopreventive role in the various stages of colorectal cancer development, selected chemical forms of selenium have shown interesting interaction patterns with some cytostatic chemicals or inducers of apoptosis. The advantages of selected selenium preparations thus might reach beyond chemoprevention since they may be used in conjunction with established antioneoplastic drugs, thereby establishing new treatment modality for colorectal cancer. In addition to summarizing our current knowledge about the mechanisms whereby selenium imparts its chemopreventive potential in colon carcinogenesis, the possibilities of a combined use of selenium with other cytostatics and chemicals are discussed.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/prevención & control , Selenio/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioprevención/tendencias , Citoprotección/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Humanos , Selenio/administración & dosificación , Selenio/química , Selenoproteínas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA