Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Clin Epigenetics ; 14(1): 156, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-36443814

RESUMEN

The protocadherin proteins are cell adhesion molecules at the crossroad of signaling pathways playing a major role in neuronal development. It is now understood that their role as signaling hubs is not only important for the normal physiology of cells but also for the regulation of hallmarks of cancerogenesis. Importantly, protocadherins form a cluster of genes that are regulated by DNA methylation. We have identified for the first time that PCDHB15 gene is DNA-hypermethylated on its unique exon in the metastatic melanoma-derived cell lines and patients' metastases compared to primary tumors. This DNA hypermethylation silences the gene, and treatment with the DNA demethylating agent 5-aza-2'-deoxycytidine reinduces its expression. We explored the role of PCDHB15 in melanoma aggressiveness and showed that overexpression impairs invasiveness and aggregation of metastatic melanoma cells in vitro and formation of lung metastasis in vivo. These findings highlight important modifications of the methylation of the PCDHß genes in melanoma and support a functional role of PCDHB15 silencing in melanoma aggressiveness.


Asunto(s)
Neoplasias Pulmonares , Melanoma , Humanos , Metilación de ADN , Melanoma/genética , Transducción de Señal , Exones , Neoplasias Pulmonares/genética
2.
Elife ; 112022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36125262

RESUMEN

Aberrant DNA methylation is a well-known feature of tumours and has been associated with metastatic melanoma. However, since melanoma cells are highly heterogeneous, it has been challenging to use affected genes to predict tumour aggressiveness, metastatic evolution, and patients' outcomes. We hypothesized that common aggressive hypermethylation signatures should emerge early in tumorigenesis and should be shared in aggressive cells, independent of the physiological context under which this trait arises. We compared paired melanoma cell lines with the following properties: (i) each pair comprises one aggressive counterpart and its parental cell line and (ii) the aggressive cell lines were each obtained from different host and their environment (human, rat, and mouse), though starting from the same parent cell line. Next, we developed a multi-step genomic pipeline that combines the DNA methylome profile with a chromosome cluster-oriented analysis. A total of 229 differentially hypermethylated genes was commonly found in the aggressive cell lines. Genome localization analysis revealed hypermethylation peaks and clusters, identifying eight hypermethylated gene promoters for validation in tissues from melanoma patients. Five Cytosine-phosphate-Guanine (CpGs) identified in primary melanoma tissues were transformed into a DNA methylation score that can predict survival (log-rank test, p=0.0008). This strategy is potentially universally applicable to other diseases involving DNA methylation alterations.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Animales , Cromosomas , Islas de CpG , Citosina , Metilación de ADN , Epigénesis Genética , Epigenoma , Regulación Neoplásica de la Expresión Génica , Guanina , Humanos , Melanoma/genética , Melanoma/patología , Ratones , Fosfatos , Ratas , Neoplasias Cutáneas/genética , Melanoma Cutáneo Maligno
3.
Clin Epigenetics ; 11(1): 9, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30651148

RESUMEN

BACKGROUND: Efficient treatments against metastatic melanoma dissemination are still lacking. Here, we report that low-cytotoxic concentrations of 5-aza-2'-deoxycytidine, a DNA demethylating agent, prevent in vitro 3D invasiveness of metastatic melanoma cells and reduce lung metastasis formation in vivo. RESULTS: We unravelled that this beneficial effect is in part due to MIR-199A2 re-expression by promoter demethylation. Alone, this miR showed an anti-invasive and anti-metastatic effect. Throughout integration of micro-RNA target prediction databases with transcriptomic analysis after 5-aza-2'-deoxycytidine treatments, we found that miR-199a-3p downregulates set of genes significantly involved in invasion/migration processes. In addition, analysis of data from melanoma patients showed a stage- and tissue type-dependent modulation of MIR-199A2 expression by DNA methylation. CONCLUSIONS: Thus, our data suggest that epigenetic- and/or miR-based therapeutic strategies can be relevant to limit metastatic dissemination of melanoma.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Decitabina/farmacología , Neoplasias Pulmonares/secundario , Melanoma/genética , MicroARNs/genética , Esferoides Celulares/citología , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Melanoma/tratamiento farmacológico , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Análisis de Secuencia de ARN , Esferoides Celulares/efectos de los fármacos , Regulación hacia Arriba
4.
Biochem Pharmacol ; 129: 1-13, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27956110

RESUMEN

DNA methylation is a mammalian epigenetic mark that participates to define where and when genes are expressed, both in normal cells and in the context of diseases. Like other epigenetic marks, it is reversible and can be modulated by chemical agents. Because it plays an important role in cancer by silencing certain genes, such as tumour suppressor genes, it is a promising therapeutic target. Two compounds are already approved to treat haematological cancers, and many efforts have been carried out to discover new molecules that inhibit DNA methyltransferases, the enzymes responsible for DNA methylation. Here, we analyse the molecular mechanisms and cellular pharmacology of these inhibitors, pointing out the necessity for new pharmacological models and paradigms. The parameters of pharmacological responses need to be redefined: the aim is cellular reprogramming rather than general cytotoxicity. Thus, "epigenetic" rather than cytotoxic dosages are defined. Another issue is the delay of the response: cellular reprogramming can take several generations to produce observable phenotypes. Is this compatible with laboratory scale experiments? Finally, it is important to consider the specificity for cancer cells compared to normal cells and the appearance of resistance. We also discuss different techniques that are used and the selection of pharmacological models.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , ADN Metiltransferasa 3A , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Humanos , Neoplasias/enzimología
5.
Planta Med ; 77(5): 461-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20945277

RESUMEN

Six protoflavonoids, including two new compounds, were isolated during a large scale screening of fern extracts for original interaction with mitosis. The new compounds isolated from PHEGOPTERIS decursive-pinnata and EQUISETUM fluviatile were 2',3'-dihydroprotogenkwanone (1) and 2',3'-dihydro-2'-hydroxyprotoapigenone (2). Known compounds were: protoapigenone, protogenkwanone, protoapigenin, and 4'- O- ß-D-glucopyranosyl protoapigenin. They showed a cytotoxic activity against HeLa cells at a micromolar level. IC50 values were 2 µM for compound 1 > 10 µM for compound 2, and respectively 2.4, 0.6, > 10 µM for the known compounds. Their cytotoxic effects were associated with phenotypic changes never observed before and characterized by the loss of centrosomal γ-tubulin labelling in both mitotic and interphasic cells.


Asunto(s)
Antineoplásicos Fitogénicos/toxicidad , Centrosoma/efectos de los fármacos , Helechos/química , Flavonoides/toxicidad , Extractos Vegetales/toxicidad , Tubulina (Proteína)/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclohexanonas/farmacología , Flavonas/farmacología , Células HeLa , Humanos , Concentración 50 Inhibidora , Mitosis/efectos de los fármacos , Fenotipo
6.
Fundam Clin Pharmacol ; 23(4): 473-82, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19709324

RESUMEN

Antitumor activities of substances from natural sources apart from vascular plants and micro-organisms have been poorly investigated. Here we report on a pharmacological screening of a bryophyte extract library using a phenotypic cell-based assay revealing microtubules, centrosomes and DNA. Among the 219 moss extracts tested, we identified 41 extracts acting on cell division with various combinations of significant effects on interphasic and mitotic cells. Seven extracts were further studied using a cell viability assay, cell cycle analysis and the phenotypic assay. Three distinct pharmacological patterns were identified including two unusual phenotypes.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Briófitas/química , Extractos Vegetales/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , ADN/efectos de los fármacos , ADN/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Células HeLa , Humanos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Fenotipo , Neoplasias del Cuello Uterino/patología
7.
Mol Cancer ; 8: 10, 2009 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-19243593

RESUMEN

BACKGROUND: NEDD1 is a protein that binds to the gamma-tubulin ring complex, a multiprotein complex at the centrosome and at the mitotic spindle that mediates the nucleation of microtubules. RESULTS: We show that NEDD1 is expressed at comparable levels in a variety of tumor-derived cell lines and untransformed cells. We demonstrate that silencing of NEDD1 expression by treatment with siRNA has differential effects on cells, depending on their status of p53 expression: p53-positive cells arrest in G1, whereas p53-negative cells arrest in mitosis with predominantly aberrant monopolar spindles. However, both p53-positive and -negative cells arrest in mitosis if treated with low doses of siRNA against NEDD1 combined with low doses of the inhibitor BI2536 against the mitotic kinase Plk1. Simultaneous reduction of NEDD1 levels and inhibition of Plk1 act in a synergistic manner, by potentiating the anti-mitotic activity of each treatment. CONCLUSION: We propose that NEDD1 may be a promising target for controlling cell proliferation, in particular if targeted in combination with Plk1 inhibitors.


Asunto(s)
Ciclo Celular/fisiología , Centrosoma/metabolismo , Proteínas Asociadas a Microtúbulos/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Fase G1/efectos de los fármacos , Fase G1/genética , Humanos , Linagliptina , Proteínas Asociadas a Microtúbulos/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Purinas/farmacología , Quinazolinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Fase de Descanso del Ciclo Celular/genética , Proteína p53 Supresora de Tumor/metabolismo , Quinasa Tipo Polo 1
8.
Oncogene ; 24(33): 5165-72, 2005 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-15897881

RESUMEN

Rad51 protein plays an essential role in recombination repair of DNA double-strand breaks and DNA crosslinking adducts. It is part of complexes which can vary with the stage of the cell cycle and the nature of the DNA lesions. During a search for Rad51-associated proteins in CHO nuclear extracts of S-phase cells by mass spectrometry of proteins immunoprecipitated with Rad51 antibodies, we identified a centrosomal protein, gamma-tubulin. This association was confirmed by the reverse immunoprecipitation with gamma-tubulin antibodies. Both proteins copurified from HeLa cells nuclear extracts following a tandem affinity purification of double-tagged Rad51. Immunofluorescence analysis showed colocalization of both Rad51 and gamma-tubulin in discrete foci in mammalian cell nuclei. The number of colocalized foci and their overlapping area increased in the presence of DNA damage produced by genotoxic treatments either during S phase or in exponentially growing cells. These variations did not result from an overall stress because microtubule cytoskeleton poisons devoid of direct interactions with DNA, such as taxol or colcemid, did not lead to an increase of this association. The recruitment of Rad51 and gamma-tubulin in the same nuclear complex suggests a link between DNA recombination repair and the centrosome function during the cell cycle.


Asunto(s)
Núcleo Celular/metabolismo , Daño del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Células CHO , Ciclo Celular , Cricetinae , Cricetulus , Reparación del ADN/fisiología , Células HeLa , Humanos , Inmunoprecipitación , Complejos Multiproteicos/metabolismo , Recombinasa Rad51 , Fase S/fisiología
10.
Bioorg Med Chem Lett ; 13(8): 1459-62, 2003 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-12668012

RESUMEN

Starting from a FPP analogue with nanomolar inhibitory activity against isolated FPTase, yet lacking activity in cellular assays, structural modifications were performed to enhance cellular activity by removing all acidic functionalities. Overall, these changes resulted in the transformation of a pure FPP to a mixed FPP and CAAX competitive inhibitor with nanomolar activity on isolated FPTase and micromolar inhibitory activity in the farnesylation of H-Ras in cultured DLD-1 cells.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Fosfatos de Poliisoprenilo/química , Fosfatos de Poliisoprenilo/farmacología , Transferasas Alquil y Aril/metabolismo , Animales , Unión Competitiva , Bovinos , Humanos , Concentración 50 Inhibidora , Proteínas de la Membrana/química , Modelos Moleculares , Prenilación de Proteína/efectos de los fármacos , Sesquiterpenos , Relación Estructura-Actividad , Células Tumorales Cultivadas , Proteínas ras/efectos de los fármacos , Proteínas ras/metabolismo
11.
Biochem Pharmacol ; 65(5): 755-63, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12628489

RESUMEN

F 11782 (2",3"-bis-pentafluorophenoxyacetyl-4",6"ethylidene-beta-D-glucoside of 4'-phosphate-4'-dimethylepipodophyllotoxin-2N-methyl glucamine salt), is a novel dual catalytic inhibitor of topoisomerases I and II characterised by marked in vivo antitumour activity, which also proved cytotoxic and exhibited DNA damaging properties in vitro. Mechanisms associated with this cell killing by F 11782 have been examined in P388 leukaemia cells. Treatment with F 11782 resulted in a dose-dependent DNA fragmentation coupled with the characteristic morphological features of apoptosis. Apoptosis-inducing concentrations of F 11782 induced caspases-3/7 activation accompanied by proteolytic cleavage of poly(ADP-ribose)-polymerase, which could be inhibited by the caspase inhibitor acetyl-Asp-Glu-Val-Asp-aldehyde. In addition, F 11782-induced apoptosis in P388 cells was associated with an increased expression of the pro-apototic Bax protein, without significant changes in the level of the anti-apoptotic Bcl-2 protein, and with modification at the mitochondrial membrane function. These results indicate that F 11782 leads to apoptosis through a caspase-3/7 dependent mechanism and suggest that the so-called "mitochondrial pathway" is implicated in F 11782-induced apoptosis in P388 cells.


Asunto(s)
Apoptosis , Naftalenos/farmacología , Piranos/farmacología , Inhibidores de Topoisomerasa I , Inhibidores de Topoisomerasa II , Animales , Caspasas/metabolismo , Dominio Catalítico , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Leucemia P388/patología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Células Tumorales Cultivadas , Proteína X Asociada a bcl-2
12.
Cancer Chemother Pharmacol ; 49(6): 479-86, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12107553

RESUMEN

PURPOSE: F 11782, or 2',3'-bis-pentafluorophenoxyacetyl-4',6'-ethylidine-beta- D-glucoside of 4'-phosphate-4'-dimethylepipodophyllotoxin 2 N-methyl glucamine salt, a novel dual catalytic inhibitor of topoisomerases I and II, characterized by marked antitumour activity in vivo in a series of experimental murine and human tumours, has been selected for further development. This preclinical study was undertaken to investigate its potential for inclusion in combination chemotherapy regimens. The in vitro cytotoxicity of F 11782 incubated simultaneously with the following drugs was investigated: aclarubicin, cisplatin, doxorubicin, etoposide, 5-fluorouracil, mitomycin C, paclitaxel, topotecan or vinorelbine. METHODS: The combinations were first evaluated in vitro against the GCT27 human testicular teratoma cell line and then against the A549 human non-small cell lung cancer cell line using median effect analysis. RESULTS: F 11782 in combination with cisplatin, mitomycin C, etoposide or doxorubicin showed synergistic cytotoxicity against both cell lines. Moreover, F 11782 combined with cisplatin or mitomycin C showed antitumour activity in vivo against P388 murine leukaemia grafted intravenously. Such synergy might have resulted from the identified nucleotide excision repair inhibitory activity of F 11782. CONCLUSIONS: F 11782 appears to be a promising candidate for combination chemotherapy, especially with DNA-damaging agents.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Naftalenos/farmacología , Neoplasias/tratamiento farmacológico , Piranos/farmacología , Inhibidores de Topoisomerasa I , Inhibidores de Topoisomerasa II , Células Tumorales Cultivadas/efectos de los fármacos , Animales , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Humanos , Masculino , Ratones , Neoplasias/enzimología , Células Tumorales Cultivadas/enzimología
13.
Anticancer Res ; 22(1A): 187-92, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12017285

RESUMEN

F 11782, or 2'', 3''-bispentafluorophenoxyacetyl-4, 6'-ethylidene-beta-D glucoside of 4'-phosphate-4'-dimethylepipodopliyllotoxin 2N-methyl glucamine salt, is a novel fluorinated lipophylic epipodophylloid which has shown marked antitumour activity in vivo. In vitro studies have demonstrated a dual catalytic inhibitory activity of F 11782 against topoisomerases and I and II by an original mechanism involving interference with the DNA binding activity of these enzymes, without DNA intercalating properties. Nevertheless, the precise mechanism(s) of cytotoxicity of F 11782 remains unclear and recent studies have suggested that this cytotoxicity might result, at least in part, from an induction of DNA-strand breaks without stabilisation of cleavable complex. In this study, DNA damage induced by F 11782 and its repair by non-homologous recombination was investigated in CHO-K1 cells. The results suggest that the nature of such damage differs from that induced by etoposide, a structurally-related topoisomerase II poison and identify a high level of stability of the damage induced which may account, at least in part, for the superior preclinical anti-tumour activity of F 11782.


Asunto(s)
Antineoplásicos/farmacología , Daño del ADN , Inhibidores Enzimáticos/farmacología , Naftalenos/farmacología , Piranos/farmacología , Inhibidores de Topoisomerasa I , Inhibidores de Topoisomerasa II , Animales , Células CHO/efectos de los fármacos , Camptotecina/farmacología , Ensayo Cometa , Cricetinae , ADN/efectos de los fármacos , ADN/metabolismo , Reparación del ADN/fisiología , Etopósido/farmacología , Concentración 50 Inhibidora
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA