Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Arch Toxicol ; 92(1): 181-194, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28776197

RESUMEN

Arsenic is one of the most important human carcinogens and environmental pollutants. However, the evaluation of the underlying carcinogenic mechanisms is challenging due to the lack of suitable in vivo and in vitro models, as distinct interspecies differences in arsenic metabolism exist. Thus, it is of high interest to develop new experimental models of arsenic-induced skin tumorigenesis in humans. Consequently, aim of this study was to establish an advanced 3D model for the investigation of arsenic-induced skin derangements, namely skin equivalents, built from immortalized human keratinocytes (NHEK/SVTERT3-5). In contrast to spontaneously immortalized HACAT cells, NHEK/SVTERT3-5 cells more closely resembled the differentiation pattern of primary keratinocytes. With regard to arsenic, our results showed that while our new cell model was widely unaffected by short-time treatment (72 h) with low, non-toxic doses of ATO (0.05-0.25 µM), chronic exposure (6 months) resulted in distinct changes of several cell characteristics. Thus, we observed an increase in the G2 fraction of the cell cycle accompanied by increased nucleus size and uneven tubulin distribution. Moreover, cells showed strong signs of de-differentiation and upregulation of several epithelial-to-mesenchymal transition markers. In line with these effects, chronic contact to arsenic resulted in impaired skin-forming capacities as well as localization of ki67-positive (proliferating) cells at the upper layers of the epidermis; a condition termed Bowen's disease. Finally, chronically arsenic-exposed cells were characterized by an increased tumorigenicity in SCID mice. Taken together, our study presents a new model system for the investigation of mechanisms underlying the tumor-promoting effects of chronic arsenic exposure.


Asunto(s)
Arsénico/toxicidad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Piel/citología , Pruebas de Toxicidad Crónica/métodos , Animales , Trióxido de Arsénico/toxicidad , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Transformación Celular Neoplásica , Femenino , Humanos , Queratinocitos/patología , Ratones Endogámicos , Técnicas de Cultivo de Órganos , Piel/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Br J Cancer ; 116(4): 489-500, 2017 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-28095394

RESUMEN

BACKGROUND: Colorectal carcinoma (CRC) is the third most common cancer worldwide. Platinum-based anticancer compounds still constitute one mainstay of systemic CRC treatment despite limitations due to adverse effects and resistance development. Trabectedin has shown promising antitumor effects in CRC, however, again resistance development may occur. In this study, we aimed to develop strategies to circumvent or even exploit acquired trabectedin resistance in novel CRC treatment regimens. METHODS: Human HCT116 CRC cells were selected for acquired trabectedin resistance in vitro and characterised by cell biological as well as bioinformatic approaches. In vivo xenograft experiments were conducted. RESULTS: Selection of HCT116 cells for trabectedin resistance resulted in p53-independent hypersensitivity of the selected subline against cisplatin. Bioinformatic analyses of mRNA microarray data suggested deregulation of nucleotide excision repair and particularly loss of the ubiquitin ligase CUL4A in trabectedin-selected cells. Indeed, transient knockdown of CUL4A sensitised parental HCT116 cells towards cisplatin. Trabectedin selected but not parental HCT116 xenografts were significantly responsive towards cisplatin treatment. CONCLUSIONS: Trabectedin selection-mediated CUL4A loss generates an Achilles heel in CRC cancer cells enabling effective cisplatin treatment. Hence, inclusion of trabectedin in cisplatin-containing cancer treatment regimens might cause profound synergism based on reciprocal resistance prevention.


Asunto(s)
Cisplatino/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas Cullin/genética , Dioxoles/uso terapéutico , Resistencia a Antineoplásicos/genética , Tetrahidroisoquinolinas/uso terapéutico , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Proteínas Cullin/antagonistas & inhibidores , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Genes p53 , Células HCT116 , Humanos , ARN Interferente Pequeño/farmacología , Trabectedina
3.
Br J Cancer ; 112(5): 783-92, 2015 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-25633038

RESUMEN

BACKGROUND: Estimating the prognosis in malignant pleural mesothelioma (MPM) remains challenging. Thus, the prognostic relevance of Ki67 was studied in MPM. METHODS: Ki67 index was determined in a test cohort of 187 cases from three centres. The percentage of Ki67-positive tumour cells was correlated with clinical variables and overall survival (OS). The prognostic power of Ki67 index was compared with other prognostic factors and re-evaluated in an independent cohort (n=98). RESULTS: Patients with Ki67 higher than median (>15%) had significantly (P<0.001) shorter median OS (7.5 months) than those with low Ki67 (19.1 months). After multivariate survival analyses, Ki67 proved to be-beside histology and treatment-an independent prognostic marker in MPM (hazard ratio (HR): 2.1, P<0.001). Interestingly, Ki67 was prognostic exclusively in epithelioid (P<0.001) but not in non-epithelioid subtype. Furthermore, Ki67 index was significantly lower in post-chemotherapy samples when compared with chemo-naive cases. The prognostic power was comparable to other recently published prognostic factors (CRP, fibrinogen, neutrophil-to-leukocyte ratio (NLR) and nuclear grading score) and was recapitulated in the validation cohort (P=0.048). CONCLUSION: This multicentre study demonstrates that Ki67 is an independent and reproducible prognostic factor in epithelioid but not in non-epithelioid MPM and suggests that induction chemotherapy decreases the proliferative capacity of MPM.


Asunto(s)
Células Epitelioides/patología , Antígeno Ki-67/metabolismo , Neoplasias Pulmonares/mortalidad , Mesotelioma/mortalidad , Neoplasias Pleurales/mortalidad , Adulto , Anciano , Anciano de 80 o más Años , Células Epitelioides/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Mesotelioma/tratamiento farmacológico , Mesotelioma/metabolismo , Mesotelioma/patología , Mesotelioma Maligno , Persona de Mediana Edad , Neoplasias Pleurales/tratamiento farmacológico , Neoplasias Pleurales/metabolismo , Neoplasias Pleurales/patología , Análisis de Supervivencia , Resultado del Tratamiento
4.
Br J Cancer ; 110(4): 984-90, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24434429

RESUMEN

BACKGROUND: To investigate the clinical utility of pretreatment plasma fibrinogen levels in malignant pleural mesothelioma (MPM) patients. METHODS: A retrospective multicenter study was performed in histologically proven MPM patients. All fibrinogen levels were measured at the time of diagnosis and clinical data were retrospectively collected after approval of the corresponding ethics committees. RESULTS: In total, 176 MPM patients (mean age: 63.5 years ± 10.4 years, 38 females and 138 males) were analysed. Most patients (n=154, 87.5%) had elevated (≥ 390 mg dl(-1)) plasma fibrinogen levels. When patients were grouped by median fibrinogen, patients with low level (≤ 627 mg dl(-1)) had significantly longer overall survival (OS) (19.1 months, confidence interval (CI) 14.5-23.7 months) when compared with those with high level (OS 8.5; CI 6.2-10.7 months). In multivariate survival analyses, fibrinogen was found to be an independent prognostic factor (hazard ratio 1.81, CI 1.23-2.65). Most interestingly, fibrinogen (cutoff 75th percentile per 750 mg dl(-1)) proved to be a predictive biomarker indicating treatment benefit achieved by surgery within multimodality therapy (interaction term: P=0.034). Accordingly, only patients below the 75th percentile benefit from surgery within multimodality therapy (31.3 vs 5.3 months OS). CONCLUSIONS: Fibrinogen is a novel independent prognostic biomarker in MPM. Most importantly, fibrinogen predicted treatment benefit achieved by surgery within multimodality therapy.


Asunto(s)
Biomarcadores de Tumor/sangre , Fibrinógeno/análisis , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/cirugía , Mesotelioma/sangre , Mesotelioma/cirugía , Terapia Combinada , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Mesotelioma/tratamiento farmacológico , Mesotelioma Maligno , Persona de Mediana Edad , Neoplasias Pleurales/sangre , Neoplasias Pleurales/tratamiento farmacológico , Neoplasias Pleurales/cirugía , Pronóstico , Estudios Retrospectivos
5.
Br J Cancer ; 108(3): 570-8, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23299527

RESUMEN

BACKGROUND: As metastasis is the prime cause of death from malignancies, there is vibrant interest to discover options for the management of the different mechanistic steps of tumour spreading. Some approved pharmaceuticals exhibit activities against diseases they have not been developed for. In order to discover such activities that might attenuate lymph node metastasis, we investigated 225 drugs, which are approved by the US Food and Drug Administration. METHODS: A three-dimensional cell co-culture assay was utilised measuring tumour cell-induced disintegrations of the lymphendothelial wall through which tumour emboli can intravasate as a limiting step in lymph node metastasis of ductal breast cancer. The disintegrated areas in the lymphendothelial cell (LEC) monolayers were induced by 12(S)-HETE, which is secreted by MCF-7 tumour cell spheroids, and are called 'circular chemorepellent induced defects' (CCIDs). The putative mechanisms by which active drugs prevented the formation of entry gates were investigated by western blotting, NF-κB activity assay and by the determination of 12(S)-HETE synthesis. RESULTS: Acetohexamide, nifedipin, isoxsuprine and proadifen dose dependently inhibited the formation of CCIDs in LEC monolayers and inhibited markers of epithelial-to-mesenchymal-transition and migration. The migration of LECs is a prerequisite of CCID formation, and these drugs either repressed paxillin levels or the activities of myosin light chain 2, or myosin-binding subunit of myosin phosphatase. Isoxsuprine inhibited all three migration markers, and isoxsuprine and acetohexamide suppressed the synthesis of 12(S)-HETE, whereas proadifen and nifedipin inhibited NF-κB activation. Both the signalling pathways independently cause CCID formation. CONCLUSION: The targeting of different mechanisms was most likely the reason for synergistic effects of different drug combinations on the inhibition of CCID formation. Furthermore, the treatment with drug combinations allowed also a several-fold reduction in drug concentrations. These results encourage further screening of approved drugs and their in vivo testing.


Asunto(s)
Acetohexamida/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Endotelio Linfático/efectos de los fármacos , Isoxsuprina/farmacología , Vasos Linfáticos/efectos de los fármacos , Nifedipino/farmacología , Proadifeno/farmacología , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Adhesión Celular/efectos de los fármacos , Movimiento Celular , Quimiotaxis/efectos de los fármacos , Técnicas de Cocultivo , Sinergismo Farmacológico , Endotelio Linfático/citología , Endotelio Linfático/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Hipoglucemiantes/farmacología , Metástasis Linfática , Vasos Linfáticos/irrigación sanguínea , Vasos Linfáticos/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Esferoides Celulares/metabolismo , Células Tumorales Cultivadas , Vasodilatadores/farmacología
6.
Br J Cancer ; 108(3): 564-9, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23093227

RESUMEN

BACKGROUND: Many cancers spread through lymphatic routes, and mechanistic insights of tumour intravasation into the lymphatic vasculature and targets for intervention are limited. The major emphasis of research focuses currently on the molecular biology of tumour cells, while still little is known regarding the contribution of lymphatics. METHODS: Breast cancer cell spheroids attached to lymphendothelial cell (LEC) monolayers were used to investigate the process of intravasation by measuring the areas of 'circular chemorepellent-induced defects' (CCID), which can be considered as entry gates for bulky tumour intravasation. Aspects of tumour cell intravasation were furthermore studied by adhesion assay, and siRNA-mediated knockdown of intracellular adhesion molecule-1 (ICAM-1). Replacing cancer spheroids with the CCID-triggering compound 12(S)-hydroxyeicosatetraenoic acid (HETE) facilitated western blot analyses of Bay11-7082- and baicalein-treated LECs. RESULTS: Binding of LECs to MCF-7 spheroids, which is a prerequisite for CCID formation, was mediated by ICAM-1 expression, and this depended on NF-κB and correlated with the expression of the prometastatic factor S100A4. Simultaneous inhibition of NF-κB with Bay11-7082 and of arachidonate lipoxygenase (ALOX)-15 with baicalein prevented CCID formation additively. CONCLUSION: Two mechanisms contribute to CCID formation: ALOX15 via the generation of 12(S)-HETE by MCF-7 cells, which induces directional migration of LECs, and ICAM-1 in LECs under control of NF-κB, which facilitates adhesion of MCF-7 cells to LECs.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Adhesión Celular/efectos de los fármacos , Endotelio Linfático/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/química , FN-kappa B/antagonistas & inhibidores , Nitrilos/farmacología , Esferoides Celulares/efectos de los fármacos , Sulfonas/farmacología , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Quimiotaxis/efectos de los fármacos , Endotelio Linfático/citología , Endotelio Linfático/metabolismo , Femenino , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
7.
Br J Cancer ; 107(12): 1978-86, 2012 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-23169291

RESUMEN

BACKGROUND: Activins control the growth of several tumour types including thoracic malignancies. In the present study, we investigated their expression and function in malignant pleural mesothelioma (MPM). METHODS: The expression of activins and activin receptors was analysed by quantitative PCR in a panel of MPM cell lines. Activin A expression was further analysed by immunohistochemistry in MPM tissue specimens (N=53). Subsequently, MPM cells were treated with activin A, activin receptor inhibitors or activin-targeting siRNA and the impact on cell viability, proliferation, migration and signalling was assessed. RESULTS: Concomitant expression of activin subunits and receptors was found in all cell lines, and activin A was overexpressed in most cell lines compared with non-malignant mesothelial cells. Similarly, immunohistochemistry demonstrated intense staining of tumour cells for activin A in a subset of patients. Treatment with activin A induced SMAD2 phosphorylation and stimulated clonogenic growth of mesothelioma cells. In contrast, treatment with kinase inhibitors of activin receptors (SB-431542, A-8301) inhibited MPM cell viability, clonogenicity and migration. Silencing of activin A expression by siRNA oligonucleotides further confirmed these results and led to reduced cyclin D1/3 expression. CONCLUSION: Our study suggests that activin A contributes to the malignant phenotype of MPM cells via regulation of cyclin D and may represent a valuable candidate for therapeutic interference.


Asunto(s)
Activinas/antagonistas & inhibidores , Antineoplásicos/farmacología , Ciclina D/metabolismo , Mesotelioma/metabolismo , Mesotelioma/patología , Neoplasias Pleurales/metabolismo , Neoplasias Pleurales/patología , Western Blotting , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Ciclina D/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Inmunohistoquímica , Mesotelioma/tratamiento farmacológico , Fenotipo , Fosforilación/efectos de los fármacos , Neoplasias Pleurales/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba
8.
Br J Cancer ; 105(2): 263-71, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21629247

RESUMEN

BACKGROUND: The intravasation of breast cancer into the lymphendothelium is an early step of metastasis. Little is known about the mechanisms of bulky cancer invasion into lymph ducts. METHODS: To particularly address this issue, we developed a 3-dimensional co-culture model involving MCF-7 breast cancer cell spheroids and telomerase-immortalised human lymphendothelial cell (LEC) monolayers, which resembles intravasation in vivo and correlated the malignant phenotype with specific protein expression of LECs. RESULTS: We show that tumour spheroids generate 'circular chemorepellent-induced defects' (CCID) in LEC monolayers through retraction of LECs, which was induced by 12(S)-hydroxyeicosatetraenoic acid (HETE) secreted by MCF-7 spheroids. This 12(S)-HETE-regulated retraction of LECs during intravasation particularly allowed us to investigate the key regulators involved in the motility and plasticity of LECs. In all, 12(S)-HETE induced pro-metastatic protein expression patterns and showed NF-κB-dependent up-regulation of the mesenchymal marker protein S100A4 and of transcriptional repressor ZEB1 concomittant with down-regulation of the endothelial adherence junction component VE-cadherin. This was in accordance with ∼50% attenuation of CCID formation by treatment of cells with 10 µM Bay11-7082. Notably, 12(S)-HETE-induced VE-cadherin repression was regulated by either NF-κB or by ZEB1 since ZEB1 siRNA knockdown abrogated not only 12(S)-HETE-mediated VE-cadherin repression but inhibited VE-cadherin expression in general. INTERPRETATION: These data suggest an endothelial to mesenchymal transition-like process of LECs, which induces single cell motility during endothelial transmigration of breast carcinoma cells. In conclusion, this study demonstrates that the 12(S)-HETE-induced intravasation of MCF-7 spheroids through LECs require an NF-κB-dependent process of LECs triggering the disintegration of cell-cell contacts, migration, and the generation of CCID.


Asunto(s)
Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacología , Neoplasias de la Mama/patología , Carcinoma/patología , Transdiferenciación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , FN-kappa B/fisiología , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Línea Celular Transformada , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Células Endoteliales/fisiología , Femenino , Humanos , Mesodermo/efectos de los fármacos , Mesodermo/fisiología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Invasividad Neoplásica , Nitrilos/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonas/farmacología , Células Tumorales Cultivadas
9.
Br J Cancer ; 104(8): 1303-12, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21407220

RESUMEN

BACKGROUND: Activins and inhibins belong to the TGFß-superfamily, which controls cell proliferation and differentiation in many organs. Activin A, the dimer of inhibin ßA subunit, acts strongly anti-proliferative in hepatocytes. Little is known on the other activin/inhibin subunits in human liver and hepatocellular carcinoma (HCC). METHODS: We studied the expression of the complete inhibin family α, ßA, ßB, ßC, ßE in normal liver, tumour-adjacent and HCC tissue, 12 additional organs and rodent liver. A total of 16 HCC and 10 disease-free livers were analysed. Expression of inhibin subunits was determined by qRT-PCR, normalised to RNA input and by geNorm algorithm, and confirmed by immunohistochemistry. RESULTS: Remarkably, ßA expression was not decreased in HCC. Similarly, ßC and ßE exhibited no major changes. In contrast, inhibin α, barely detectable in normal liver, was strongly increased in tumour-adjacent liver and dramatically enhanced in HCC. ßB was strongly enhanced in some HCC. At variance with human liver, rodent liver showed higher inhibin α and ßC expression, but ßA was somewhat, and ßB dramatically lower. CONCLUSIONS: Upregulation of inhibin α - and possibly of ßB - may shield HCC cells from anti-proliferative effects of activin A. Dramatic variations between humans and rodents may reflect different functions of some inhibins/activins.


Asunto(s)
Activinas/genética , Carcinoma Hepatocelular/genética , Inhibinas/genética , Neoplasias Hepáticas/genética , Hígado/metabolismo , Roedores/genética , Activinas/metabolismo , Adulto , Anciano , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Estudios de Casos y Controles , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inhibinas/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Wistar , Roedores/metabolismo
10.
Br J Cancer ; 102(9): 1361-70, 2010 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-20424615

RESUMEN

BACKGROUND: Digalloyl-resveratrol (di-GA) is a synthetic compound aimed to combine the biological effects of the plant polyhydroxy phenols gallic acid and resveratrol, which are both radical scavengers and cyclooxygenase inhibitors exhibiting anticancer activity. Their broad spectrum of activities may probably be due to adjacent free hydroxyl groups. METHODS: Protein activation and expression were analysed by western blotting, deoxyribonucleoside triphosphate levels by HPLC, ribonucleotide reductase activity by (14)C-cytidine incorporation into nascent DNA and cell-cycle distribution by FACS. Apoptosis was measured by Hoechst 33258/propidium iodide double staining of nuclear chromatin and the formation of gaps into the lymphendothelial barrier in a three-dimensional co-culture model consisting of MCF-7 tumour cell spheroids and human lymphendothelial monolayers. RESULTS: In HL-60 leukaemia cells, di-GA activated caspase 3 and dose-dependently induced apoptosis. It further inhibited cell-cycle progression in the G1 phase by four different mechanisms: rapid downregulation of cyclin D1, induction of Chk2 with simultaneous downregulation of Cdc25A, induction of the Cdk-inhibitor p21(Cip/Waf) and inhibition of ribonucleotide reductase activity resulting in reduced dCTP and dTTP levels. Furthermore, di-GA inhibited the generation of lymphendothelial gaps by cancer cell spheroid-secreted lipoxygenase metabolites. Lymphendothelial gaps, adjacent to tumour bulks, can be considered as gates facilitating metastatic spread. CONCLUSION: These data show that di-GA exhibits three distinct anticancer activities: induction of apoptosis, cell-cycle arrest and disruption of cancer cell-induced lymphendothelial disintegration.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Ácido Gálico/análogos & derivados , Células HL-60/efectos de los fármacos , Estilbenos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Colorantes , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Citometría de Flujo , Ácido Gálico/farmacología , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/fisiología , Células HL-60/citología , Humanos , Pulmón/citología , Pulmón/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
11.
Br J Cancer ; 102(7): 1145-56, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20234367

RESUMEN

BACKGROUND: Deregulation of fibroblast growth factor receptor 3 (FGFR3) is involved in several malignancies. Its role in colorectal cancer has not been assessed before. METHODS: Expression of FGFR3 in human colorectal tumour specimens was analysed using splice variant-specific real-time reverse transcriptase PCR assays. To analyse the impact of FGFR3-IIIc expression on tumour cell biology, colon cancer cell models overexpressing wild-type (WT-3b and WT3c) or dominant-negative FGFR3 variants (KD3c and KD3b) were generated by either plasmid transfection or adenoviral transduction. RESULTS: Although FGFR3 mRNA expression is downregulated in colorectal cancer, alterations mainly affected the FGFR3-IIIb splice variant, resulting in an increased IIIc/IIIb ratio predominantly in a subgroup of advanced tumours. Overexpression of WT3c increased proliferation, survival and colony formation in all colon cancer cell models tested, whereas WT3b had little activity. In addition, it conferred sensitivity to autocrine FGF18-mediated growth and migration signals in SW480 cells with low endogenous FGFR3-IIIc expression. Disruption of FGFR3-IIIc-dependent signalling by dominant-negative FGFR3-IIIc or small interfering RNA-mediated FGFR3-IIIc knockdown resulted in inhibition of cell growth and induction of apoptosis, which could not be observed when FGFR3-IIIb was blocked. In addition, KD3c expression blocked colony formation and migration and distinctly attenuated tumour growth in SCID mouse xenograft models. CONCLUSION: Our data show that FGFR3-IIIc exerts oncogenic functions by mediating FGF18 effects in colorectal cancer and may constitute a promising new target for therapeutic interventions.


Asunto(s)
Movimiento Celular , Neoplasias Colorrectales/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Apoptosis , Células CACO-2 , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética
12.
Cell Mol Life Sci ; 66(1): 43-61, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18759128

RESUMEN

The unique and evolutionary highly conserved major vault protein (MVP) is the main component of ubiquitous, large cellular ribonucleoparticles termed vaults. The 100 kDa MVP represents more than 70% of the vault mass which contains two additional proteins, the vault poly (ADP-ribose) polymerase (vPARP) and the telomerase-associated protein 1 (TEP1), as well as several short untranslated RNAs (vRNA). Vaults are almost ubiquitously expressed and, besides chemotherapy resistance, have been implicated in the regulation of several cellular processes including transport mechanisms, signal transmissions and immune responses. Despite a growing amount of data from diverse species and systems, the definition of precise vault functions is still highly complex and challenging. Here we review the current knowledge on MVP and vaults with focus on regulatory functions in intracellular signal transduction and immune defence.


Asunto(s)
Poli(ADP-Ribosa) Polimerasas/fisiología , Transducción de Señal/fisiología , Partículas Ribonucleoproteicas en Bóveda/fisiología , Animales , Proteínas Portadoras/química , Proteínas Portadoras/fisiología , Resistencia a Antineoplásicos/genética , Humanos , Inmunidad Innata/fisiología , Ratones , Poli(ADP-Ribosa) Polimerasas/química , Estructura Terciaria de Proteína , Proteínas de Unión al ARN , Partículas Ribonucleoproteicas en Bóveda/química , Partículas Ribonucleoproteicas en Bóveda/inmunología
13.
Oncogene ; 27(30): 4180-90, 2008 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-18362893

RESUMEN

Fibroblast growth factor 5 (FGF5) is widely expressed in embryonic but scarcely in adult tissues. Here we report simultaneous overexpression of FGF5 and its predominant high-affinity receptor (FGFR1 IIIc) in astrocytic brain tumour specimens (N=49) and cell cultures (N=49). The levels of both ligand and receptor increased with enhanced malignancy in vivo and in vitro. Furthermore, secreted FGF5 protein was generally present in the supernatants of glioblastoma (GBM) cells. siRNA-mediated FGF5 downmodulation reduced moderately but significantly GBM cell proliferation while recombinant FGF5 (rFGF5) increased this parameter preferentially in cell lines with low endogenous expression levels. Apoptosis induction by prolonged serum starvation was significantly prevented by rFGF5. Moreover, tumour cell migration was distinctly stimulated by rFGF5 but attenuated by FGF5 siRNA. Blockade of FGFR1-mediated signals by pharmacological FGFR inhibitors or a dominant-negative FGFR1 IIIc protein inhibited GBM cell proliferation and/or induced apoptotic cell death. Moreover, rFGF5 and supernatants of highly FGF5-positive GBM cell lines specifically stimulated proliferation, migration and tube formation of human umbilical vein endothelial cells. In summary, we demonstrate for the first time that FGF5 contributes to the malignant progression of human astrocytic brain tumours by both autocrine and paracrine effects.


Asunto(s)
Comunicación Autocrina/fisiología , Neoplasias Encefálicas/genética , Factor 5 de Crecimiento de Fibroblastos/fisiología , Glioblastoma/genética , Oncogenes , Comunicación Paracrina/fisiología , Comunicación Autocrina/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Progresión de la Enfermedad , Factor 5 de Crecimiento de Fibroblastos/genética , Factor 5 de Crecimiento de Fibroblastos/farmacología , Genes Dominantes/fisiología , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/genética , Oncogenes/fisiología , Comunicación Paracrina/efectos de los fármacos , Proteínas Recombinantes/farmacología , Transfección , Células Tumorales Cultivadas
14.
Br J Cancer ; 94(11): 1718-25, 2006 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-16685273

RESUMEN

Upregulation of cyclooxygenase-2 (COX-2) and prostaglandin-dependent vascularisation in small adenomatous polyps is an essential part of colon carcinogenesis. To study the underlying cellular mechanisms, LT97 and Caco2 human colorectal tumour cells not expressing endogenous COX-2 were exposed to 1 microM prostaglandin E(2) (PGE(2)) in their medium. At 30 min after addition, expression of c-fos was stimulated 5-fold and 1.3-fold, respectively, depending on the activation of both extracellular signal-regulated kinase and p38. The amount of c-jun in nuclear extracts was increased 20% in LT97 cells. Expression of COX-2 was upregulated 1.7-fold in LT97 cells and 1.5-fold in Caco2 2 h after prostaglandin (PG) addition by a p38-mediated pathway. The known PGE(2) target gene vascular endothelial growth factor (VEGF) was not modulated. Effects of sustained PGE(2) production were studied in VACO235 cells that have high endogenous COX-2 and in LT97 cells infected with an adenovirus expressing COX-2. Prostaglandin E(2) secretion into the medium was 1-2 nM and 250 pM, respectively. Expression of both VEGF and c-fos was high in VACO235 cells. In LT97 cells, COX-2 upregulated c-fos expression and c-jun content in nuclear extracts 1.7- and 1.2-fold, respectively, in a PG-dependent way. This shows that exogenous PGE(2) as well as COX-2 overexpression affect signalling and gene expression in a way that enhances tumour progression.


Asunto(s)
Adenoma/genética , Neoplasias del Colon/genética , Dinoprostona/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Ciclooxigenasa 2/genética , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa , Prostaglandinas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal
15.
Apoptosis ; 11(3): 427-40, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16538382

RESUMEN

Chemoresistance is a biological response of cells to survive toxic stress. During cancer treatment the development of chemoresistance is a major problem. The mechanisms how cells become insensitive, and which downstream pathways are affected are not completely understood. Since it has not been well analysed which and how many regulative disorders are subsummised under the term "chemoresistance", we examined and measured arabinosylcytosine (AraC)-mediated desensitation of two mechanisms relevant for tissue homeostasis, cell cycle inhibition and apoptosis induction. MCF-7 cells harbouring ectopic mutated p53 were suitable for this investigation because they activated these mechanisms subsequently and became insensitive to AraC with regard to cell cycle inhibition and apoptosis induction. The major causal mechanism of acquired resistance against AraC was most likely through the inhibition of the first step of AraC phosphorylation within the cell, which is rate limiting for its activation. With regard to cell cycle inhibition AraC-resistant cells were also resistant against 5-fluorodeoxyuridine (5-FdUrd), but fully responsive to 5-FdUrd-induced apoptosis, evidencing that cell cycle and apoptosis are independent of each other. Apoptosis correlated with AIF-activation and was independent of Caspase 7, whereas cell cycle inhibition correlated with cyclinD1 expression but not with induction of p21 or p27. The phosphate conjugated 5-FdUrd-araC heterodimer (5-Fluoro-2'-desoxyuridylyl-(3'-->5')-Arabinocytidine), which is a prodrug of AraC-monophosphate, reactivated AIF and down-regulated cyclin D1 in AraC-resistant cells and circumvented resistance to apoptosis and to cell cycle inhibition. Also, cells which were resistant to 5-FdUrd or doxorubicin were sensitive to 5-FdUrd-araC. This investigation demonstrates that chemoresistance affects apoptosis induction and cell cycle inhibition independently and that detailed knowledge about the affected downstream pathways would enable the design of targeted intervention with small molecules to restore chemosensitivity.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Citarabina/farmacología , Resistencia a Antineoplásicos/fisiología , Floxuridina/farmacología , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular Tumoral , Citarabina/química , Citarabina/metabolismo , Femenino , Floxuridina/química , Floxuridina/metabolismo , Humanos , Estructura Molecular
16.
Nucleosides Nucleotides Nucleic Acids ; 23(8-9): 1507-11, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15571287

RESUMEN

In search for possible alternatives in the treatment of human malignancies we investigated several new heterodinucleoside phosphates consisting of 5-Fluorodeoxyuridine (5-FdUrd) and Arabinofuranosylcytosine (Ara-C). We show that all dimers tested inhibited the number of colonies of CCL228, CCL227, 5-FU resistant CCL227 and HT-29 human colon tumor cells with IC50 values ranging from 0.65 to 1 nM. Dimer # 2 inhibited the number of sensitive and Ara-C resistant H9 human lymphoma cells with IC50 values ranging from 200 to 230 nM. Since no significant difference in the cytotoxicity of the dimers could be observed between sensitive and resistant cells, these compounds might be used in the treatment of 5-FU and Ara-C resistant tumors.


Asunto(s)
Apoptosis , Citarabina/farmacología , Fosfatos de Dinucleósidos/química , Fluorouracilo/farmacología , Antimetabolitos Antineoplásicos/farmacología , Bisbenzimidazol/farmacología , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Colorantes/farmacología , Dimerización , Colorantes Fluorescentes/farmacología , Humanos , Concentración 50 Inhibidora , Linfoma/tratamiento farmacológico , Propidio/farmacología
17.
Cell Death Differ ; 11(1): 80-9, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12970676

RESUMEN

Cell division cycle 25A (Cdc25A) was shown to colocalise both with nuclear and cytoplasmic proteins. Recently, we have demonstrated that overexpressed Cdc25A promoted the survival of rat 423 cells through indirect activation of PKB-protein kinase B. Using a Cdc25A:ER fusion protein, which can be shuttled from the cytoplasm into the nucleus, the present investigation evidences that the antiapoptotic effect of Cdc25A was restricted to its cytoplasmic localisation in rat 423 cells. In contrast, nuclear Cdc25A overexpression caused dephosphorylation and nuclear retention of the proapoptotic transcription factor Forkhead in rhabdomyosarcoma-like 1 (FKHRL1) in human N.1 ovarian carcinoma cells. This resulted in the increased constitutive expression of the FKHRL1 targets Fas ligand and Bim, and promoted apoptosis. Thus, the Cdc25A oncogene, which was found to be frequently overexpressed in certain human cancers, can increase or decrease the susceptibility to apoptosis depending on the cell-type-specific subcellular distribution.


Asunto(s)
Apoptosis/fisiología , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Transcripción/metabolismo , Fosfatasas cdc25/metabolismo , Animales , Apoptosis/efectos de los fármacos , Fusión Artificial Génica , Compartimento Celular , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead , Vectores Genéticos , Humanos , Proteínas del Tejido Nervioso , Ratas , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes/genética , Transformación Genética , Factor de Necrosis Tumoral alfa/farmacología , Fosfatasas cdc25/genética
18.
Cell Death Differ ; 9(2): 169-78, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11840167

RESUMEN

A new synthetic drug, benzamide riboside (BR) exhibited strong oncolytic activity against leukemic cells in the 5-10 microM range. Higher BR-concentrations (20 microM) predominantly induced necrosis which correlated with DNA strand breaks and subsequent depletion of ATP- and dATP levels. Replenishment of the ATP pool by addition of adenosine prevented necrosis and favoured apoptosis. This effect was not a pecularity of BR-treatment, but was reproduced with high concentrations of all trans-retinoic acid (120 microM) and cyanide (20 mM). Glucose was also capable to suppress necrosis and to favour apoptosis of HL-60 cells, which had been treated with necrotic doses of BR and cyanide. Apoptosis eliminates unwanted cells without affecting the microenvironment, whereas necrosis causes severe inflammation of surrounding tissues due to spillage of cell fluids into the peri-cellular space. Thus, the monitoring and maintenance of cellular energy pools during therapeutic drug treatment may help to minimize nonspecific side effects and to improve attempted drug effects.


Asunto(s)
Adenosina Trifosfato/fisiología , Antineoplásicos/toxicidad , Apoptosis , Necrosis , Nucleósidos/toxicidad , Adenosina/farmacología , Adenosina Trifosfato/análisis , Benzamidas/farmacología , Ensayo Cometa , Daño del ADN , Nucleótidos de Desoxiadenina/análisis , Nucleótidos de Desoxicitosina/análisis , Desoxirribonucleótidos/análisis , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Células HL-60 , Humanos , IMP Deshidrogenasa/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Cianuro de Potasio/antagonistas & inhibidores , Tretinoina/antagonistas & inhibidores
19.
Oncogene ; 20(33): 4542-53, 2001 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-11494150

RESUMEN

The phosphatase Cdc25A was shown to be a target of the transcription factor c-Myc. Myc-induced apoptosis appeared dependent on Cdc25A expression and Cdc25A over-expression could substitute for Myc-triggered apoptosis. These findings suggested that an important downstream component of Myc-mediated apoptosis was identified. However and in contrast, we recently reported that during TNFalpha-induced apoptosis, which required c-Myc function, Cdc25A was down-regulated in a human carcinoma cell line. We now provide evidence that Cdc25A rendered the non-transformed rat embryonic cell line 423 refractory to apoptosis, which was induced by serum deprivation and in absence of detectable c-myc levels. The survival promoting activity of cdc25A was abolished upon infection of cells with a full-length cdc25A antisense construct. To identify the signaling proteins mediating the survival function of the phosphatase, cdc25A- and akt- over-expressing pooled clones were exposed to selected chemicals, which inhibit or activate key proteins in signaling pathways. Inhibition of apoptosis by SU4984, NF023 and Rapamycin placed Cdc25A and Akt function downstream of FGF.R, PDGF.R, and compensated G-protein- and PP2A- activity. Interestingly, upon treatment with LY-294002, cdc25A- and akt- over-expressing clones exhibited similar apoptotic patterns as control cells, which indicates that neither Akt- nor Cdc25A-mediated survival functions are dependent on PI.3 kinase activity in rat 423 cells. In cdc25A-overexpressing cells increased levels of serine 473 phosphorylated Akt were found, which co-precipitated with Cdc25A and Raf1. Since activation of proteins requires dephosphorylation of particular residues in addition to site-specific phosphorylation, the anti-apoptotic effect of Cdc25A might derive from its participation in a multimeric protein complex with phosphoAkt and Raf1, two prominent components of survival pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Arabidopsis , Medio de Cultivo Libre de Suero/farmacología , Fosfatasas cdc25/fisiología , Animales , Línea Celular/efectos de los fármacos , Cromonas/farmacología , Citocinas/farmacología , Depresión Química , Doxiciclina/farmacología , Embrión de Mamíferos/citología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica , Genes myc , Sustancias de Crecimiento/farmacología , Péptidos y Proteínas de Señalización Intercelular , Sustancias Macromoleculares , Morfolinas/farmacología , Péptidos/farmacología , Fosforilación , Piperazinas/farmacología , Proteínas de Plantas/fisiología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Canales de Potasio/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Ratas , Proteínas Recombinantes de Fusión/fisiología , Sirolimus/farmacología , Suramina/análogos & derivados , Suramina/farmacología , Transfección
20.
Exp Hematol ; 29(5): 623-32, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11376876

RESUMEN

OBJECTIVE: Amidox and didox are two polyhydroxy-substituted benzohydroxamic acid derivatives that belong to a new class of ribonucleotide reductase (RR) inhibitors. RR is the rate-limiting enzyme for de novo deoxyribonucleotide synthesis, and its activity is significantly increased in tumor cells in proportion to the proliferation rate. Therefore, RR is a target for antitumor therapy. MATERIALS AND METHODS: HL-60 and K562 leukemia cells were treated with increasing doses of amidox and didox. Thereafter, the mode of cytotoxic drug action was determined by Hoechst 33258/propidium iodide (HO/PI) double staining, annexin binding, DNA fragmentation, and caspase activation. This was correlated to the decrease in dNTP levels. Staining with HO/PI and binding of fluorescein isothiocyanate-conjugated annexin V to externalized phosphatidylserine were used to quantify apoptosis. RESULTS: Low doses of amidox or didox resulted in an increase of apoptotic HL-60 cells within 48 hours. Higher doses (50 microM amidox or 250 microM didox) led to rapid induction of apoptosis, which could be detected as early as 4 hours after treatment. After 48 hours with these concentrations, almost 100% of the HL-60 cells died by apoptosis without an increase in necrosis. K562 cells were found to be resistant to amidox but not to didox. In HL-60 cells, upstream caspase 8 is processed in response to didox, whereas caspases 8 and 9 are processed upon amidox treatment. Didox-induced apoptosis, but not amidox-induced apoptosis, can be correlated with the decrease in dNTP levels. The results suggests that amidox induces several apoptosis mechanisms in HL-60 cells. In contrast, only caspase 9 is activated by didox in K562 cells, and because amidox hardly induces apoptosis in this cell line, no caspase cleavage is observed. CONCLUSIONS: Didox triggers distinct apoptosis pathways in HL-60 and K562 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Caspasas/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ácidos Hidroxámicos/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Oximas/farmacología , Ribonucleótido Reductasas/antagonistas & inhibidores , Anexina A5/metabolismo , Caspasa 8 , Caspasa 9 , Fragmentación del ADN , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Gelsolina/metabolismo , Células HL-60/efectos de los fármacos , Células HL-60/enzimología , Humanos , Células K562/efectos de los fármacos , Células K562/enzimología , Fosfatidilserinas/metabolismo , Proyectos Piloto , Poli(ADP-Ribosa) Polimerasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA