Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Cell Sci ; 128(9): 1787-99, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25788698

RESUMEN

A common feature of non-coding repeat expansion disorders is the accumulation of RNA repeats as RNA foci in the nucleus and/or cytoplasm of affected cells. These RNA foci can be toxic because they sequester RNA-binding proteins, thus affecting various steps of post-transcriptional gene regulation. However, the precise step that is affected by C9orf72 GGGGCC (G4C2) repeat expansion, the major genetic cause of amyotrophic lateral sclerosis (ALS), is still poorly defined. In this work, we set out to characterise these mechanisms by identifying proteins that bind to C9orf72 RNA. Sequestration of some of these factors into RNA foci was observed when a (G4C2)31 repeat was expressed in NSC34 and HeLa cells. Most notably, (G4C2)31 repeats widely affected the distribution of Pur-alpha and its binding partner fragile X mental retardation protein 1 (FMRP, also known as FMR1), which accumulate in intra-cytosolic granules that are positive for stress granules markers. Accordingly, translational repression is induced. Interestingly, this effect is associated with a marked accumulation of poly(A) mRNAs in cell nuclei. Thus, defective trafficking of mRNA, as a consequence of impaired nuclear mRNA export, might affect translation efficiency and contribute to the pathogenesis of C9orf72 ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Núcleo Celular/metabolismo , Modelos Biológicos , Biosíntesis de Proteínas , Proteínas/metabolismo , Expansión de Repetición de Trinucleótido , Esclerosis Amiotrófica Lateral/patología , Animales , Proteína C9orf72 , Proteínas de Unión al ADN , Factor 2 Eucariótico de Iniciación/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Células HeLa , Humanos , Espacio Intracelular/metabolismo , Ratones , Neuronas Motoras/metabolismo , Fosforilación , Proteínas de Unión a Poli(A)/metabolismo , Unión Proteica , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción
2.
Mol Carcinog ; 51(10): 783-95, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21882253

RESUMEN

Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, has been reported to exert chemopreventive and antitumor effects on colon cancer, one of the most common solid epithelial malignancy worldwide. The aim of this study was to elucidate whether celecoxib may be able to affect epithelial-mesenchymal transition (EMT), a critical process involved in cancer cell invasiveness and metastasis and then proposed to be relevant for cancer progression. Human HT-29 colon cancer cells were exposed to carefully controlled hypoxic conditions and/or epidermal growth factor (EGF) and then investigated for EMT changes and signal transduction pathways involved by using morphological, molecular, and cell biology techniques. Celecoxib inhibited basal and EGF-stimulated proliferation, hypoxia-related HIF-1α recruitment/stabilization as well as hypoxia- and EGF-dependent activation of ERK and PI3K. Interestingly, celecoxib prevented EMT-related changes, as shown by modifications of ß-catenin intracellular localization or vimentin and E-cadherin levels, as well as HT-29 invasiveness induced by hypoxia, EGF, or hypoxia plus EGF. Finally, experiments performed on SW-480 colon cancer cells (i.e., cells lacking COX-2) exposed to hypoxia, used here as a stimulus able to induce EMT and invasiveness, revealed that in these cells celecoxib was ineffective. Results of the present study indicate that celecoxib has the potential to negatively affect induction of EMT and increased invasiveness of colon cancer cells as elicited by different signals originating from tumor microenvironment (i.e., hypoxia and EGF). Moreover, these effects are likely be related to the pharmacological inhibitory effect exerted on COX-2 activity.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Factor de Crecimiento Epidérmico/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Pirazoles/farmacología , Sulfonamidas/farmacología , Cadherinas/metabolismo , Celecoxib , Hipoxia de la Célula , Proliferación Celular , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HT29 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Vimentina/metabolismo , beta Catenina/metabolismo
3.
Mol Cell Biochem ; 361(1-2): 105-10, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22002318

RESUMEN

Rofecoxib is a specific COX-2 inhibitor able to exert antiproliferative activity against colorectal cancer cells. It was withdrawn from the market after the demonstration of an increased risk of cardiovascular complications after prolonged use. Nevertheless, it remains an interesting compound for laboratory research as an experimental COX-2 inhibitor. In this study, the antiproliferative activity of a novel dinitro-oxy-substituted analogue of rofecoxib (NO-rofe), potentially less cardiotoxic, has been investigated in vitro on human colon cancer cells and compared with the action of the parent drug. Due to the fact that COX-2 inhibition is the main characteristic of coxibs, we performed all experiments in COX-2-overexpressing (HT-29) and COX-2-negative (SW-480) human colon cancer cells, to elucidate whether the observed effects were dependent on COX-2 inhibition. Moreover, experiments were performed in order to evaluate whether COX-2 pharmacological inhibition may affect beta-catenin/E-cadherin signaling pathway. NO-rofe exerted a significant antiproliferative activity on COX-2 positive HT-29 human colon cancer cells, being less effective on the COX-2 negative SW-480 human colon cancer cell line. In particular, the rofecoxib analogue retained similar potencies with respect to COX-2 inhibition but was much more active than rofecoxib in inhibiting the growth of human colon cancer cells in vitro. In addition, this novel compound resulted in the induction of membrane ß-catenin/E-cadherin expression, a feature that may significantly contribute to its antiproliferative activity.


Asunto(s)
4-Butirolactona/análogos & derivados , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Nitratos/farmacología , 4-Butirolactona/farmacología , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
4.
Mol Cell Biochem ; 350(1-2): 59-70, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21140284

RESUMEN

The inducible COX-2 enzyme is over-expressed in human breast cancer and its over-expression generally correlates with angiogenesis, deregulation of apoptosis and worse prognosis. This observation may explain the beneficial effect of nonsteroidal anti-inflammatory drugs and COX-2 inhibitors on breast cancer treatment. Here, we evaluated the antiproliferative activity of celecoxib, a selective COX-2 inhibitor, and its nitro-oxy derivative on human breast cancer cells characterized by low and high COX-2 expression, respectively. In ERα(+) MCF-7 cells celecoxib and its derivative induce a strong inhibition of cell growth, inhibition that is associated with the reduction of ERα expression and activation. These effects may be directly associated with ERK and Akt suppression and with PP2A and PTEN induction. In this cell line the drugs exert only weak effect on COX-2 level while they are able to reduce aromatase expression. On the contrary, in ERα(-) MDA-MB-231 cells, both drugs induce a marked inhibition of COX-2, inhibition that is associated with the reduction of aromatase expression and of cell proliferation. In both cell lines the effects of the drugs are associated with the suppression of cell invasion.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma/patología , Proliferación Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , Antineoplásicos/farmacología , Celecoxib , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Modelos Biológicos , Invasividad Neoplásica
5.
Carcinogenesis ; 29(12): 2267-78, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18791199

RESUMEN

Epithelial-mesenchymal transition (EMT) and hypoxia are considered as crucial events favouring invasion and metastasis of many cancer cells. In this study, different human neoplastic cell lines of epithelial origin were exposed to hypoxic conditions in order to investigate whether hypoxia per se may trigger EMT programme as well as to mechanistically elucidate signal transduction mechanisms involved. The following human cancer cell lines were used: HepG2 (from human hepatoblastoma), PANC-1 (from pancreatic carcinoma), HT-29 (from colon carcinoma) and MCF-7 (from breast carcinoma). Cancer cells were exposed to carefully controlled hypoxic conditions and investigated for EMT changes and signal transduction by using morphological, cell and molecular biology techniques. All cancer cells responded to hypoxia within 72 h by classic EMT changes (fibroblastoid phenotype, SNAIL and beta-catenin nuclear translocation and changes in E-cadherin) and by increased migration and invasiveness. This was involving very early inhibition of glycogen synthase kinase-3beta (GSK-3beta), early SNAIL translocation as well as later and long-lasting activation of Wnt/beta-catenin-signalling machinery. Experimental manipulation, including silencing of hypoxia-inducible factor (HIF)-1alpha and the specific inhibition of mitochondrial generation of reactive oxygen species (ROS), revealed that early EMT-related events induced by hypoxia (GSK-3beta inhibition and SNAIL translocation) were dependent on transient intracellular increased generation of ROS whereas late migration and invasiveness were sustained by HIF-1alpha- and vascular endothelial growth factor (VEGF)-dependent mechanisms. These findings indicate that in cancer cells, early redox mechanisms can switch on hypoxia-dependent EMT programme whereas increased invasiveness is sustained by late and HIF-1alpha-dependent release of VEGF.


Asunto(s)
Hipoxia de la Célula/fisiología , Transformación Celular Neoplásica/metabolismo , Transducción de Señal/fisiología , Western Blotting , Diferenciación Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Epitelio/metabolismo , Epitelio/patología , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mesodermo/metabolismo , Mesodermo/patología , Invasividad Neoplásica/genética , Invasividad Neoplásica/fisiopatología , Oxidación-Reducción , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
Br J Nutr ; 100(4): 739-50, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18304389

RESUMEN

Epidemiological studies suggest that dietary PUFA may influence breast cancer progression. n-3 PUFA are generally known to exert antitumour effects, whereas reports relative to n-6 PUFA anti-carcinogen effects are controversial. Arachidonic acid (AA; 20:4n-6) and its metabolites have been shown to inhibit the growth of human breast cancer cell lines, even if the downstream mechanisms by which AA may influence carcinogenesis remain unresolved. We explored the molecular basis for AA influence on proliferation, signal transduction and apoptosis in two human breast cancer cell lines, MCF-7 and MDA-MB-231. In both cell lines AA inhibited cell growth in a dose-dependent manner, even if MDA-MB-231 was somewhat more growth-inhibited than MCF-7. AA decreased extracellular signal-regulated protein kinase 1/2 phosphorylation level, and positively modulated PPARgamma and PPARalpha expression, with only a slight effect against PPARbeta/delta. In addition, AA increased Bak (an apoptosis-regulating protein) expression and reduced procaspase-3 and -9 levels only in MDA-MB-231 cells, thus indicating that the growth inhibitory effect can be correlated with apoptosis induction. In both cell lines the use of a specific antagonist made it possible to establish a relationship between AA growth inhibitory effect and PPARalpha involvement. AA decreases cell proliferation most likely by inducing apoptosis in MDA-MB-231 cells, while in the MCF-7 cell line the growth inhibitory activity can be attributed to the inhibition of the signal transduction pathway involved in cell proliferation. In both cases, the results here presented suggest PPARalpha as a possible contributor to the growth inhibitory effect of AA.


Asunto(s)
Ácido Araquidónico/farmacología , Neoplasias de la Mama/patología , PPAR alfa/farmacología , Análisis de Varianza , Apoptosis/efectos de los fármacos , Western Blotting/métodos , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , PPAR alfa/genética , PPAR alfa/metabolismo , Fosforilación/efectos de los fármacos , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
7.
J Nutr Biochem ; 18(5): 332-40, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-16963252

RESUMEN

Conjugated linoleic acid (CLA) is a naturally occurring compound found in dairy and beef products. In recent years, it has received considerable attention because several studies showed a lower incidence of certain cancers in animals fed CLA-supplemented diets. In vitro studies further showed growth inhibitory activity on tumor cell proliferation, the CLA being effective above all against colon cancer cells. The aim of the present work was to investigate the growth inhibitory effect of CLA on Caco-2 cell line. Under our experimental conditions, CLA repressed Caco-2 cell proliferation, and the growth-inhibitory action increased by repeating treatments. However, in Caco-2 cells, CLA was unable to induce apoptosis, as revealed by cell-cycle analysis and Western blot studies. To determine the mechanism by which CLA inhibits cell growth, we studied its effect on extracellular-regulated kinase signaling. Conjugated linoleic acid reduced expression levels of Raf-1 and phosphorylation of ERK1/2, which was accompanied by a decrease in the expression of the downstream transcription factor c-myc. Our data suggest that CLA is dependent, at least in part, on the ERK kinase pathway for its ability to inhibit the growth of Caco-2 cancer cells.


Asunto(s)
Células CACO-2/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Ácidos Linoleicos Conjugados/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , L-Lactato Deshidrogenasa/efectos de los fármacos , L-Lactato Deshidrogenasa/metabolismo , Proteínas Proto-Oncogénicas c-raf/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal , Proteína Destructora del Antagonista Homólogo bcl-2/efectos de los fármacos , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo
8.
Chem Biol Interact ; 169(2): 110-21, 2007 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-17632092

RESUMEN

Conjugated linoleic acid (CLA), a naturally occurring substance in food sources, occurs as mixtures of positional and geometrical isomers of octadecadienoate (18:2), and may inhibit colon tumorigenesis. It has been hypothesized that CLA can modulate cell proliferation and differentiation through the activation of peroxisome proliferator-activated receptors (PPARs), among which PPARgamma is involved in growth inhibition of transformed cells. The aim of the present study was to investigate whether the antiproliferative effects of CLA are mediated by its interaction with PPARgamma and APC/beta-catenin signalling pathway in human colon cancer cells. In CLA-treated caco-2 cells we found a remarkable increase in the expression of PPARgamma, which translocated into the nucleus, while PPARalpha and beta/delta protein levels were not affected. GW259662, a well known PPARgamma antagonist, blocked the increase in PPARgamma protein rate and abrogated some biological effects of CLA, as it restored the proliferative capability of the cells and ERK1/2 phosphorylation level. We demonstrated that CLA treatment determined the down-regulation of APC and c-myc proteins, but in this case the administration of the antagonist was not able to revert CLA effects. Furthermore, CLA induced a reorganization of E-cadherin and beta-catenin, as well as a redistribution of actin and tubulin filaments. Our data suggest that CLA may regulate PPARgamma expression by selectively acting as an agonist; however, the discrepancies in PPARgamma antagonist efficacy suggest the involvement of other pathways, independent of PPARgamma, in CLA antiproliferative activity.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Genes APC , Ácido Linoleico/farmacología , PPAR gamma/fisiología , beta Catenina/fisiología , Western Blotting , Células CACO-2 , Cadherinas/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-myc/metabolismo , beta Catenina/metabolismo
9.
Cancer Lett ; 234(2): 149-57, 2006 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-15885890

RESUMEN

We investigated the molecular mechanisms involved in the anti-proliferative activity exerted by conjugated linoleic acid (CLA) on the estrogen unresponsive MDA-MB-231 human breast cancer cell line. The effects on cell proliferation, cell cycle progression and induction of apoptosis were examined. CLA caused the reduction of cell proliferation along with the accumulation of cells in the S phase of the cycle. The occurrence of apoptosis in these cells was indicated by flow cytometry data and further confirmed by the onset of cells with morphological features typical of apoptosis. ERK1/2 reduction and upregulation of pro-apoptotic protein Bak were induced. These events were associated with: (a) reduced levels of the anti-apoptotic protein Bcl-x(L), (b) the translocation of cytochrome c from the mitochondria to the cytosol, (c) the cleavage of pro-caspase-9 and pro-caspase-3. From the above data, we are induced to think that CLA may trigger apoptosis in the estrogen unresponsive MDA-MB-231 cell line via mechanisms involving above all the mitochondrial pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Ácidos Linoleicos Conjugados/farmacología , Mitocondrias/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Western Blotting , Neoplasias de la Mama , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Citometría de Flujo , Humanos , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/fisiología
10.
Oncotarget ; 7(22): 33257-71, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27119353

RESUMEN

The c-MYC oncoprotein is a DNA binding transcription factor that enhances the expression of many active genes. c-MYC transcriptional signatures vary according to the transcriptional program defined in each cell type during differentiation. Little is known on the involvement of c-MYC in regulation of gene expression programs that are induced by extracellular cues such as a changing microenvironment. Here we demonstrate that inhibition of c-MYC in glioblastoma multiforme cells blunts hypoxia-dependent glycolytic reprogramming and mitochondria fragmentation in hypoxia. This happens because c-MYC inhibition alters the cell transcriptional response to hypoxia and finely tunes the expression of a subset of Hypoxia Inducible Factor 1-regulated genes. We also show that genes whose expression in hypoxia is affected by c-MYC inhibition are able to distinguish the Proneural subtype of glioblastoma multiforme, thus potentially providing a molecular signature for this class of tumors that are the least tractable among glioblastomas.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/farmacología , Hipoxia Tumoral , Microambiente Tumoral , Sitios de Unión , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Glucólisis/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
11.
Front Cell Neurosci ; 9: 41, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25741238

RESUMEN

It is well known that mitochondrial damage (MD) is both the major contributor to oxidative stress (OS) (the condition arising from unbalance between production and removal of reactive oxygen species) and one of the major consequences of OS, because of the high dependance of mitochondrial function on redox-sensitive targets such as intact membranes. Conditions in which neuronal cells are not able to cope with MD and OS seem to lead or contribute to several neurodegenerative diseases including Amyotrophic Lateral Sclerosis (ALS), at least in the most studied superoxide dismutase 1 (SOD1)-linked genetic variant. As summarized in this review, new evidence indicates that MD and OS play a role also in non-SOD1 ALS and thus they may represent a target for therapy despite previous failures in clinical trials.

12.
Cancer Lett ; 209(2): 139-45, 2004 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-15159015

RESUMEN

Insulin regulates metabolism through homologous receptor tyrosine kinases, and plays a role in proliferation of breast cancer cells. Our research studied whether insulin, administered separately or in combination with paclitaxel, interferes with paclitaxel-mediated biological activity in human breast cancer cells. Not only did insulin influence paclitaxel-mediated cell microtubule reorganization, but it also influenced MCF-7 cell sensitivity to paclitaxel. Furthermore, combined administrations of insulin and paclitaxel affected MAPK pathway, Raf-1 activation and p53 expression levels. Our findings indicate that insulin seems to modulate MCF-7 cell response to paclitaxel; consequently, elevated levels of insulin could influence tumor cell resistance.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Hipoglucemiantes/farmacología , Insulina/farmacología , Microtúbulos/efectos de los fármacos , Paclitaxel/farmacología , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Quimioterapia Combinada , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Femenino , Flavonoides , Humanos , Sistema de Señalización de MAP Quinasas , Fosforilación , Proteínas Proto-Oncogénicas c-raf/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
13.
Chem Biol Interact ; 147(1): 79-86, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-14726154

RESUMEN

Costunolide is an active sesquiterpene lactone of medicinal herbs with anti-inflammatory and potential anti-cancer activity. Nevertheless, the pharmacological pathways of costunolide have not yet been fully elucidated. In this study we showed that costunolide exerts a dose-dependent antiproliferative activity in the human breast cancer MCF-7 cells. In addition, light microscopy observations indicated that costunolide affected nuclear organization and reorganized microtubule architecture. The antiproliferative and antimicrotubular effects of costunolide were not influenced by paclitaxel, well-known microtubule-stabilizing anticancer agent. The microtubule-interacting activity of costunolide was confirmed by in vitro studies on purified microtubular protein. In fact, costunolide demonstrated polymerizing ability, by inducing the formation of well organized microtubule polymers. Our data suggest an interaction of costunolide with microtubules, which may represent a new intracellular target for this drug.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteínas de Microtúbulos/efectos de los fármacos , Sesquiterpenos/farmacología , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/toxicidad , División Celular/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Humanos , Proteínas de Microtúbulos/ultraestructura , Paclitaxel/farmacología , Sesquiterpenos/química , Sesquiterpenos/toxicidad , Factores de Tiempo
14.
Chem Biol Interact ; 149(2-3): 165-73, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15501437

RESUMEN

Parthenolide is an active sesquiterpene lactone present in a variety of medicinal herbs, well known as anti-inflammatory drug. It has recently been proposed as a chemotherapeutic drug, but the pharmacological pathways of its action have not yet been fully elucidated. Firstly, we explored whether the anticancer properties of parthenolide may be related to a tubulin/microtubule-interfering activity. We additionally compared bioactivities of parthenolide with those checked after combined treatments with paclitaxel in human breast cancer MCF-7 cells. Parthenolide exerted in vitro stimulatory activity on tubulin assembly, by inducing the formation of well-organized microtubule polymers. Light microscopy detections showed that parthenolide-induced alterations of either microtubule network and nuclear morphology happened only after combined exposures to paclitaxel. In addition, the growth of MCF-7 cells was significantly inhibited by parthenolide, which enhanced paclitaxel effectiveness. In conclusion, the antimicrotubular and antiproliferative effects of parthenolide, well known microtubule-stabilizing anticancer agent, may influence paclitaxel activity. The tubulin/microtubule system may represent a novel molecular target for parthenolide, to be utilized in developing new combinational anticancer strategies.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Microtúbulos/efectos de los fármacos , Sesquiterpenos/farmacología , Tanacetum parthenium/química , Tubulina (Proteína)/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/ultraestructura , Línea Celular Tumoral , Colchicina/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Microscopía Electrónica , Microscopía Fluorescente , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Paclitaxel/farmacología , Extractos Vegetales/farmacología
15.
Anticancer Res ; 34(4): 1793-800, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24692712

RESUMEN

BACKGROUND: Signals from the tumor microenvironment (hypoxia, growth factors) are known to induce an invasive phenotype. Cyclooxygenase-2 (COX2) overexpression, involved in colorectal carcinoma (CRC) progression, is also associated with epidermal growth factor receptor (EGFR) up-regulation. The present study investigated whether inhibition of COX2 may affect, under normoxia and hypoxia, EGF-induced cell proliferation and invasiveness by using immunoblotting, trypan blue assay, Boyden chamber assay and zymography. RESULTS: The proliferative and invasive activity of HT-29 cells was enhanced under hypoxia. COX2 expression was increased after epidermal growth factor (EGF) stimulation under both hypoxia and normoxia, expression that was efficiently reduced by the COX2 inhibitor NS398. Under normoxia, NS398 reduced signalling pathways induced by EGF [phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT), extracellular-signal-regulated kinases (ERKs)], while under hypoxia, EGF stimulation and NS398 treatment was associated with HIF-1α expression. Under both conditions, NS398 was able to inhibit cell invasiveness and matrix-metalloproteinase-2 release. CONCLUSION: COX2 inhibition can contribute to reducing cell aggressiveness through interfering with EGF- and hypoxia-mediated signaling.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/farmacología , Receptores ErbB/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Nitrobencenos/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Transición Epitelial-Mesenquimal , Células HT29 , Humanos
16.
Expert Opin Ther Targets ; 18(2): 121-35, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24325753

RESUMEN

INTRODUCTION: Cyclooxygenase-2 (COX-2) is overexpressed in several malignancies and is implicated in breast cancer progression. OBJECTIVES: We investigated whether changes in COX-2 expression may affect epithelial-to-mesenchymal transition (EMT) and then invasive potential of human breast cancer cells, in relationship with hypoxia. COX-2-null MCF-7 human breast cancer cells, MCF-7 cells transiently expressing COX-2 and COX-2-expressing MDA-MB-231 cells were employed. RESULTS: COX-2 overexpression resulted in downregulation of E-cadherin and ß-catenin, upregulation of vimentin, N-cadherin and SNAI1, suggesting EMT occurrence. COX-2-overexpressing MCF-7 cells were also characterized by increased invasiveness and release of matrix-metalloproteinase-9. The above-mentioned characteristics, homologous to those detected in highly invasive MDA-MB-231 cells, were reverted by treatment of COX-2-overexpressing MCF-7 cells with celecoxib, a COX-2-specific inhibitor, partly through the inhibition of COX-2-related intracellular generation of reactive oxygen species. Hypoxia further exacerbated COX-2 expression, EMT changes and invasive ability in both COX-2-overexpressing MCF-7 cells and MDA-MB-231 cells. Finally, immunohistochemistry performed on samples from normal and neoplastic human breast tissues revealed that COX-2-positive malignant cells were also positive for EMT-related antigens, hypoxia-inducible factor (HIF)-2α and the oxidative stress marker heme oxygenase. CONCLUSIONS: These findings support the existence of a direct link between COX-2 overexpression, EMT and invasiveness in human breast cancer cells, emphasizing the role of hypoxic microenvironment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ciclooxigenasa 2/metabolismo , Transición Epitelial-Mesenquimal , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Ciclooxigenasa 2/genética , ADN Complementario/genética , Dinoprostona/metabolismo , Femenino , Hemo-Oxigenasa 1/metabolismo , Humanos , Hipoxia/metabolismo , Células MCF-7 , Invasividad Neoplásica , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
17.
Dis Markers ; 34(4): 257-67, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23396293

RESUMEN

The human cancer antigen 125 (CA125) is over-expressed in epithelial ovarian cancer cells and it plays a role in the pathogenesis of ovarian cancer. This protein presents a repeat region containing up to sixty tandem repeat units. The anti-CA125 monoclonal antibodies have been previously classified into three groups: two major families, the OC125-like antibodies and M11-like antibodies, and a third group, the OV197-like antibodies. A model in which a single repeat unit contains all the epitopes for these antibodies has been also proposed, even if their exact position is still undetermined. In the present work, the affinities of the monoclonal antibodies, representative of the three families, have been investigated for different CA125-recombinant repeats through Western blot analysis. Different patterns of antibody recognition for the recombinant repeats show that CA125 epitopes are not uniformly distributed in the tandem repeat region of the protein. The minimal region for the recognition of these antibodies has been also individuated in the SEA domain through the subcloning of deleted sequences of the highly recognized repeat-25 (R-25), their expression as recombinant fragments in E. coli and Western blot analysis. Obtained data have been further confirmed by ELISA using the entire R-25 as coating antigen.


Asunto(s)
Antígeno Ca-125/química , Epítopos/análisis , Neoplasias Ováricas/inmunología , Secuencias Repetidas en Tándem , Secuencia de Aminoácidos , Secuencia de Bases , Western Blotting , Antígeno Ca-125/genética , Línea Celular Tumoral , Clonación Molecular , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
19.
Anticancer Res ; 30(7): 2659-66, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20682995

RESUMEN

It has been shown previously that a novel nitrooxy derivative of celecoxib exerts antiproliferative and pro-apoptotic effects in human colon cancer cells. The aim of this study was to elucidate whether these biological properties depend on COX-2 inhibition and/or NO release. Therefore, the derivative was decomposed into the parent compound celecoxib and the NO donor benzyl nitrate and the biological role of each was tested in COX-2-positive (HT-29) and -negative (SW-480) colon cancer cells. The main findings were that the nitro-oxy derivative behaved like celecoxib in HT-29 cells in terms of COX-2 and ERK/MAPK inhibition, as well as induction of apoptosis, while the benzyl nitrate had no such effects. Interestingly, the beta-catenin system was activated by the nitro-oxy derivative as well as by benzyl nitrate alone more potently than by the parent compound celecoxib, suggesting a possible regulatory role for NO. In SW480 cells, these activities were substantially less pronounced, suggesting the presence of COX-2-dependent mechanisms in the modulation of these parameters.


Asunto(s)
Compuestos de Bencilo/farmacología , Neoplasias del Colon/tratamiento farmacológico , Ciclooxigenasa 2/metabolismo , Nitratos/farmacología , Óxido Nítrico/metabolismo , Pirazoles/farmacología , Sulfonamidas/farmacología , Apoptosis/efectos de los fármacos , Celecoxib , Procesos de Crecimiento Celular/efectos de los fármacos , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Inhibidores de la Ciclooxigenasa 2/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HT29 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , beta Catenina/metabolismo
20.
Expert Opin Ther Targets ; 14(7): 655-64, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20536410

RESUMEN

OBJECTIVE: COX-2 is implicated in carcinogenesis and tumour progression in many cancers, including breast cancer. Recently, it has been reported that human breast carcinomas aberrantly express COX-2, and that raised tissue levels of COX-2 may have prognostic value. Patients expressing high levels of COX-2 can develop local recurrence, and have reduced disease-free and disease-related overall survival. The aim of this study was to investigate COX-2 expression in human ductal and lobular breast cancers and its possible association with clinicopathological features and prognostic molecular markers. RESEARCH DESIGN AND METHODS: Cytoplasmic COX-2 expression was detected by means of immunohistochemistry in a series of 91 breast carcinomas with ductal (n = 60) and lobular (n = 31) patterns. COX-2 expression was investigated by multivariate analyses and compared with clinicopathological features. RESULTS AND CONCLUSIONS: COX-2 immune positivity and percentage of positive cells correlated significantly with the size, grading, extent of primary tumour and vascular invasion of carcinoma but not with biological parameters (estrogen receptor, progesterone receptor and human EGF receptor 2). The findings of the present study suggest that COX-2 overexpression in lobular and ductal breast cancers, which correlates with traditional clinico-pathological parameters, may be considered as a negative prognostic marker.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Lobular/metabolismo , Ciclooxigenasa 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/diagnóstico , Carcinoma Ductal de Mama/patología , Carcinoma Ductal de Mama/secundario , Carcinoma Lobular/diagnóstico , Carcinoma Lobular/patología , Carcinoma Lobular/secundario , Femenino , Humanos , Inmunohistoquímica , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Metástasis Linfática , Persona de Mediana Edad , Estadificación de Neoplasias , Neovascularización Patológica , Pronóstico , Carga Tumoral , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA