Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Clin Epigenetics ; 14(1): 156, 2022 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-36443814

RESUMO

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.


Assuntos
Neoplasias Pulmonares , Melanoma , Humanos , Metilação de DNA , Melanoma/genética , Transdução de Sinais , Éxons , Neoplasias Pulmonares/genética
2.
Elife ; 112022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36125262

RESUMO

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.


Assuntos
Melanoma , Neoplasias Cutâneas , Animais , Cromossomos , Ilhas de CpG , Citosina , Metilação de DNA , Epigênese Genética , Epigenoma , Regulação Neoplásica da Expressão Gênica , Guanina , Humanos , Melanoma/genética , Melanoma/patologia , Camundongos , Fosfatos , Ratos , Neoplasias Cutâneas/genética , Melanoma Maligno Cutâneo
3.
Clin Epigenetics ; 11(1): 9, 2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30651148

RESUMO

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.


Assuntos
Metilação de DNA/efeitos dos fármacos , Decitabina/farmacologia , Neoplasias Pulmonares/secundário , Melanoma/genética , MicroRNAs/genética , Esferoides Celulares/citologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Melanoma/tratamento farmacológico , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Regiões Promotoras Genéticas , Análise de Sequência de RNA , Esferoides Celulares/efeitos dos fármacos , Regulação para Cima
4.
Biochem Pharmacol ; 129: 1-13, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-27956110

RESUMO

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.


Assuntos
DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Inibidores Enzimáticos/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , DNA Metiltransferase 3A , Inibidores Enzimáticos/farmacologia , Epigênese Genética , Humanos , Neoplasias/enzimologia
5.
Planta Med ; 77(5): 461-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20945277

RESUMO

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.


Assuntos
Antineoplásicos Fitogênicos/toxicidade , Centrossomo/efeitos dos fármacos , Gleiquênias/química , Flavonoides/toxicidade , Extratos Vegetais/toxicidade , Tubulina (Proteína)/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cicloexanonas/farmacologia , Flavonas/farmacologia , Células HeLa , Humanos , Concentração Inibidora 50 , Mitose/efeitos dos fármacos , Fenótipo
6.
Fundam Clin Pharmacol ; 23(4): 473-82, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19709324

RESUMO

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.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Briófitas/química , Extratos Vegetais/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Centrossomo/efeitos dos fármacos , Centrossomo/metabolismo , DNA/efeitos dos fármacos , DNA/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Células HeLa , Humanos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Fenótipo , Neoplasias do Colo do Útero/patologia
7.
Mol Cancer ; 8: 10, 2009 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-19243593

RESUMO

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.


Assuntos
Ciclo Celular/fisiologia , Centrossomo/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Fase G1/efeitos dos fármacos , Fase G1/genética , Humanos , Linagliptina , Proteínas Associadas aos Microtúbulos/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Purinas/farmacologia , Quinazolinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/genética , Proteína Supressora de Tumor p53/metabolismo , Quinase 1 Polo-Like
8.
Oncogene ; 24(33): 5165-72, 2005 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-15897881

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Tubulina (Proteína)/metabolismo , Animais , Células CHO , Ciclo Celular , Cricetinae , Cricetulus , Reparo do DNA/fisiologia , Células HeLa , Humanos , Imunoprecipitação , Complexos Multiproteicos/metabolismo , Rad51 Recombinase , Fase S/fisiologia
10.
Bioorg Med Chem Lett ; 13(8): 1459-62, 2003 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-12668012

RESUMO

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.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Fosfatos de Poli-Isoprenil/química , Fosfatos de Poli-Isoprenil/farmacologia , Alquil e Aril Transferases/metabolismo , Animais , Ligação Competitiva , Bovinos , Humanos , Concentração Inibidora 50 , Proteínas de Membrana/química , Modelos Moleculares , Prenilação de Proteína/efeitos dos fármacos , Sesquiterpenos , Relação Estrutura-Atividade , Células Tumorais Cultivadas , Proteínas ras/efeitos dos fármacos , Proteínas ras/metabolismo
11.
Biochem Pharmacol ; 65(5): 755-63, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12628489

RESUMO

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.


Assuntos
Apoptose , Naftalenos/farmacologia , Piranos/farmacologia , Inibidores da Topoisomerase I , Inibidores da Topoisomerase II , Animais , Caspases/metabolismo , Domínio Catalítico , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Leucemia P388/patologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Células Tumorais Cultivadas , Proteína X Associada a bcl-2
12.
Cancer Chemother Pharmacol ; 49(6): 479-86, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12107553

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Naftalenos/farmacologia , Neoplasias/tratamento farmacológico , Piranos/farmacologia , Inibidores da Topoisomerase I , Inibidores da Topoisomerase II , Células Tumorais Cultivadas/efeitos dos fármacos , Animais , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Humanos , Masculino , Camundongos , Neoplasias/enzimologia , Células Tumorais Cultivadas/enzimologia
13.
Anticancer Res ; 22(1A): 187-92, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12017285

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Dano ao DNA , Inibidores Enzimáticos/farmacologia , Naftalenos/farmacologia , Piranos/farmacologia , Inibidores da Topoisomerase I , Inibidores da Topoisomerase II , Animais , Células CHO/efeitos dos fármacos , Camptotecina/farmacologia , Ensaio Cometa , Cricetinae , DNA/efeitos dos fármacos , DNA/metabolismo , Reparo do DNA/fisiologia , Etoposídeo/farmacologia , Concentração Inibidora 50
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA