Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Sci Rep ; 5: 16255, 2015 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-26542881

RESUMO

Telomeres are nucleoprotein structures at the end of chromosomes which stabilize and protect them from nucleotidic degradation and end-to-end fusions. The G-rich telomeric single-stranded DNA overhang can adopt a four-stranded G-quadruplex DNA structure (G4). Stabilization of the G4 structure by binding of small molecule ligands enhances radiosensitivity of tumor cells, and this combined treatment represents a novel anticancer approach. We studied the effect of the platinum-derived G4-ligand, Pt-ctpy, in association with radiation on human glioblastoma (SF763 and SF767) and non-small cell lung cancer (A549 and H1299) cells in vitro and in vivo. Treatments with submicromolar concentrations of Pt-ctpy inhibited tumor proliferation in vitro with cell cycle alterations and induction of apoptosis. Non-toxic concentrations of the ligand were then combined with ionizing radiation. Pt-ctpy radiosensitized all cell lines with dose-enhancement factors between 1.32 and 1.77. The combined treatment led to increased DNA breaks. Furthermore, a significant radiosensitizing effect of Pt-ctpy in mice xenografted with glioblastoma SF763 cells was shown by delayed tumor growth and improved survival. Pt-ctpy can act in synergy with radiation for efficient killing of cancer cells at concentrations at which it has no obvious toxicity per se, opening perspectives for future therapeutic applications.


Assuntos
Neoplasias Encefálicas/terapia , Carcinoma Pulmonar de Células não Pequenas/terapia , DNA de Neoplasias/efeitos dos fármacos , Quadruplex G/efeitos dos fármacos , Glioblastoma/terapia , Neoplasias Pulmonares/terapia , Radiossensibilizantes/administração & dosagem , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/radioterapia , Xenoenxertos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/radioterapia , Camundongos , Telomerase/genética , Proteína 1 de Ligação a Repetições Teloméricas/genética
2.
Int J Cancer ; 133(5): 1042-53, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23404099

RESUMO

The development of alternative therapies for melanoma treatment is of great interest as long-term tumour regression is not achieved with new targeted chemotherapies on selected patients. We previously demonstrated that radioiodinated heteroarylcarboxamide ([131I]ICF01012) induced a strong anti-tumoural effect by inhibiting both primary tumour growth and dissemination process in a B16BL6 melanoma model. In our study, we show that a single injection of [131I]ICF01012 (ranging from 14.8 to 22.2 MBq) was effective and associated with low and transient haematological toxicity. Concerning pigmented organs, cutaneous melanocytes and skin were undamaged. In 30% of treated animals, no histological alteration of retina was observed, and in the remaining 70%, damages were restricted to the optic nerve area. Using the Medical Internal Radiation Dose methodology, we determined that the absorbed dose in major organs is very low (<4 Gy) and that a delivery of 30 Gy to the tumour is sufficient for an effective anti-tumoural response. Molecular analyses of treated tumours showed a strong radiobiological effect with a decrease in proliferation, survival and pro-angiogenic-related markers and an increase in tumour suppressor gene expression, melanogenesis and anti-angiogenic markers. All these features are in accordance with a tumour cell death mechanism that mainly occurs by mitotic catastrophe and provide a better understanding of in vivo anti-tumoural effects of [131I] radionuclide. Our findings raise [131I]ICF01012 a good candidate for disseminated melanoma treatment and strongly support transfer of [131I]ICF01012 to clinical trial.


Assuntos
Radioisótopos do Iodo/uso terapêutico , Melaninas/antagonistas & inibidores , Melanoma Experimental/radioterapia , Quinoxalinas/uso terapêutico , Animais , Ciclo Celular/efeitos da radiação , Humanos , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL
3.
Invest New Drugs ; 30(4): 1782-90, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21499733

RESUMO

Cartilage tumours present ongoing therapeutic challenges due to their chondrogenic extracellular matrix that potentially hampers drug delivery, their low percentage of dividing cells, and their poor vascularity. In this context, and based on the affinity of the quaternary ammonium moiety for proteoglycans (PG), we developed a strategy that uses the quaternary ammonium function to selectively deliver DNA alkylating agents to the cartilage tumour tissue. We engineered the quaternary ammonium derivative of melphalan (Mel-AQ) and assessed its antitumoural activity in vitro and in vivo. In vitro, micromolar concentrations of Mel-AQ inhibited the proliferation of human HEMC-SS chondrosarcoma and Saos-2 osteosarcoma cell lines. Moreover, 24-h incubation with 20 µM Mel-AQ induced a 2.5-fold increase in S population and a 1.5-fold increase in subG0G1 population compared to controls. In vivo, Mel-AQ demonstrated antitumour activity in the orthotopic model of primary Swarm rat chondrosarcoma. When given to chondrosarcoma-bearing rats (three doses of 16 µmol/kg at days 8, 12 and 16 post-implant), Mel-AQ demonstrated an optimal antitumour effect at day 43, when tumour cell growth inhibition peaked at 69%. Interestingly, the treatment protocol was proved well tolerated, since the animals showed no weight loss over the course of the study. This antitumoural effect was assessed in vivo by scintigraphic imaging using (99m)Tc-NTP 15-5 developed in our lab as a PG-targeting radiotracer, and tumour tissue was analyzed at study-end by biochemical PG assay with Alcian blue staining. Mel-AQ treatment led to a significant decrease in the PG content of tumoural tissue. These experimental results highlighted the promising antitumour potential of Mel-AQ as a PG-targeting strategy for therapeutic management of chondrosarcoma.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Condrossarcoma/tratamento farmacológico , Melfalan/análogos & derivados , Compostos de Amônio Quaternário/uso terapêutico , Animais , Antineoplásicos Alquilantes/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Condrossarcoma/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Compostos Heterocíclicos com 1 Anel , Humanos , Masculino , Melfalan/química , Melfalan/farmacologia , Melfalan/uso terapêutico , Proteoglicanas/metabolismo , Compostos de Amônio Quaternário/química , Compostos de Amônio Quaternário/farmacologia , Ratos , Ratos Sprague-Dawley
4.
Biochem Pharmacol ; 81(9): 1116-23, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21371445

RESUMO

The cyclization of anticancer drugs into active intermediates has been reported mainly for DNA alkylating molecules including nitrosoureas. We previously defined the original cytotoxic mechanism of anticancerous N-phenyl-N'-(2-chloroethyl)ureas (CEUs) that involves their reactivity towards cellular proteins and not against DNA; two CEU subsets have been shown to alkylate ß-tubulin and prohibitin leading to inhibition of cell proliferation by G2/M or G1/S cell cycle arrest. In this study, we demonstrated that cyclic derivatives of CEUs, N-phenyl-4,5-dihydrooxazol-2-amines (Oxas) are two- to threefold more active than CEUs and share the same cytotoxic properties in B16F0 melanoma cells. Moreover, the CEU original covalent binding by an ester linkage on ß-tubulin Glu198 and prohibitin Asp40 was maintained with Oxas. Surprisingly, we observed that Oxas were spontaneously formed from CEUs in the cell culture medium and were also detected within the cells. Our results suggest that the intramolecular cyclization of CEUs leads to active Oxas that should then be considered as the key intermediates for protein alkylation. These results will be useful for the design of new prodrugs for cancer chemotherapy.


Assuntos
Alquilantes/farmacologia , Aminas/metabolismo , Ácido Aspártico/metabolismo , Ácido Glutâmico/metabolismo , Proteínas Repressoras/metabolismo , Tubulina (Proteína)/metabolismo , Ureia/metabolismo , Aminas/química , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Ciclização , Eletroforese em Gel Bidimensional , Imunofluorescência , Melanoma Experimental/patologia , Camundongos , Proibitinas , Proteínas Repressoras/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Tubulina (Proteína)/química , Ureia/química
5.
Anticancer Res ; 30(6): 2049-54, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20651350

RESUMO

BRCA1 acts as a tumour suppressor and germ-line mutations within this gene are found in a large proportion of families with breast cancer. The aim of our study was to unravel the mechanism of action of genistein, the major soy phytoestrogen, in BRCA1-mutant human breast cancer cell lines. Four breast cancer cell lines were studied for their response to genistein, three of them harbouring different mutations within the BRCA1 gene (HCC1937, SUM149 and SUM1315 cells) and the MDA-MB-231 cell line, which expresses a functional BRCA1 protein. We showed that genistein inhibits proliferation and induces apoptosis more efficiently in BRCA1-mutant cells than in cells expressing wild-type BRCA1 protein. Increased AKT and decreased p21(WAF1/CIP1) protein levels could explain the relative resistance to genistein elicited by cells with wild-type BRCA1. BRCA1-mutant breast cancer cells are highly sensitive to genistein treatment and p21(WAF1/CIP1) and AKT could be genistein targets in these cells.


Assuntos
Anticarcinógenos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Genes BRCA1 , Genisteína/farmacologia , Mutação , Proteínas Proto-Oncogênicas c-akt/fisiologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/análise , Inibidor de Quinase Dependente de Ciclina p21/genética , Feminino , Genes BRCA1/fisiologia , Humanos , Proteínas Proto-Oncogênicas c-akt/análise , Proteínas Proto-Oncogênicas c-akt/genética , RNA Mensageiro/análise
6.
Biochem Biophys Res Commun ; 379(3): 785-9, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19126406

RESUMO

Soy phytoestrogens, among which genistein, seem to protect from breast cancer development. In order to study the role of the breast tumour suppressor BRCA1 in response to genistein, we used a new breast cancer cell model: the SUM1315MO2 cell line carrying the 185delAG BRCA1 mutation, which we stably transfected with a plasmid encoding wild-type BRCA1. We showed that growth of BRCA1 mutant cells was strongly inhibited by genistein whereas it only had a weak effect in cells expressing wild-type BRCA1 protein. BRCA1 mutant cells hypersensitivity could be linked to higher expression of ERbeta gene, which suggests that genistein may be an efficient inhibitor of cancer development in BRCA1 mutant breast cancer cells.


Assuntos
Anticarcinógenos/farmacologia , Proteína BRCA1/genética , Neoplasias da Mama/prevenção & controle , Transformação Celular Neoplásica/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Genisteína/farmacologia , Apoptose , Neoplasias da Mama/genética , Ciclo Celular/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Receptor beta de Estrogênio/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Mutação
7.
J Mol Endocrinol ; 39(2): 151-62, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17693613

RESUMO

In the male, androgens promote growth and differentiation of sex reproductive organs through ligand activation of the androgen receptor (AR). Here, we show that androgens are not major actors of the cell cycle arrest associated with the differentiation process, and that the epidermal growth factor (EGF)-mediated signalling interferes with AR activities to regulate androgen response when epithelial cells are differentiated. Higher AR expression and enhanced androgen responsiveness correlate with reduction of phosphorylated ERK1/2 over differentiation. These modifications are associated with recruitment of cells in phase G(0)/G(1), up-regulation of p27(kip1), down-regulation of p21(Cip1) and p53 proteins, and accumulation of hypo-phosphorylated Rb. Exposure to EGF reduces AR expression levels and blocks androgen-dependent transcription in differentiated cells. It also restores p53 and p21(Cip1) levels, Rb hyper-phosphorylation, ERK1/2 activation and promotes cell cycle re-entry as p27(kip1) protein levels are decreased. Treatment with a MEK inhibitor reverses the EGF-mediated AR down-regulation in differentiated cells, thus suggesting the existence of an inverse correlation between EGF and androgen signalling in non-tumoural epithelia. Interestingly, when androgen signalling is set in differentiated cells, dihydrotestosterone exerts an inhibitory effect on ERK activity but paradoxically does not modify EGFR (ErbB1) phosphorylation, indicating that androgens are able to disrupt the EGFR-ERK cascade. Overall, our data demonstrate the existence of a balance between AR and mitogen-activated protein kinase activities that favours either the maintenance of differentiated conditions or the enhancement of cell proliferation capacities.


Assuntos
Receptores ErbB/metabolismo , Receptor Cross-Talk/fisiologia , Receptores Androgênicos/metabolismo , Transdução de Sinais/fisiologia , Androgênios , Animais , Northern Blotting , Western Blotting , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Di-Hidrotestosterona/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Receptores ErbB/agonistas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Imunoprecipitação , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , Receptor Cross-Talk/efeitos dos fármacos , Receptores Androgênicos/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Ducto Deferente/citologia
8.
J Biol Chem ; 279(15): 14579-86, 2004 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-14668339

RESUMO

Androgens are known to modulate many cellular processes such as cell growth and survival by binding to the androgen receptor (AR) and activating the transcription of target genes. Recent data suggested that AR can also mediate non-transcriptional actions outside the nucleus in addition to its ligand-inducible transcription factor function. Here, we describe a transcription-independent activation of the phosphatidylinositol 3-OH kinase (PI3-K) signaling pathway by androgens. Using non-transformed androgen-sensitive epithelial cells, we show that androgens enhance the PI3-K activity by promoting accumulation of phosphoinositide-3-P phospholipids in vitro. This activation is found in conjunction with an increased time-dependent phosphorylation of the downstream kinase AKT/protein kinase B on both Ser(473) and Thr(308) residues. Hormone-stimulated phosphorylation of AKT requires AR since incubation with the anti-androgen bicalutamide completely abolishes the androgen-stimulated AKT phosphorylation. Accordingly, we show that androgens increase AKT phosphorylation level in prostatic carcinoma PC3 cells only once they have been transfected with AR. Downstream, androgens enhance phosphorylation of transcription factor FKHR (Forkhead in rhabdomyosarcoma)-L1 and proapoptotic Bad protein and promote cell survival as they can counteract an apoptotic process. We also report that non-genomic effects of androgens are based on direct interaction between AR and the p85alpha regulatory subunit of class I(A) PI3-K. Together, these novel findings point out an important and physiologically relevant link between androgens and the PI3-K/AKT signaling pathway in governing cell survival.


Assuntos
Células Epiteliais/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Receptores Androgênicos/fisiologia , Anilidas/farmacologia , Animais , Apoptose , Western Blotting , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática , Células Epiteliais/metabolismo , Escherichia coli/metabolismo , Glutationa Transferase/metabolismo , Humanos , Ligantes , Camundongos , Modelos Genéticos , Nitrilas , Fosforilação , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Serina/química , Transdução de Sinais , Treonina/química , Fatores de Tempo , Compostos de Tosil , Transcrição Gênica , Transfecção , Proteína de Morte Celular Associada a bcl
9.
Clin Chim Acta ; 320(1-2): 101-10, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11983207

RESUMO

BACKGROUND: BRCA1 and BRCA2 are breast cancer susceptibility genes. Recent studies suggest that BRCA1 interacts with a great variety of proteins, including BRCA2, cell-cycle regulators, transcriptional activators and repressors. We investigated the expression of both BRCA1 and BRCA2 during the progression of the cell cycle of human tumor cell lines from different origins (MCF7, MDA-MB231, PA1 and CCL221) in two growth status (60% and 100% of confluency). METHODS: First, the growth status was characterized by determination of the cell cycle by flow cytometry analysis. At the same time, immunohistochemistry was performed to follow BRCA1 and BRCA2 protein expression and then, quantification of BRCA1 and BRCA2 transcripts was realized using real-time quantitative RT-PCR. RESULTS: We reported in studied tumor cell lines with 60% of confluency by comparison with 100% of confluency, an increase in the BRCA1 and BRCA2 expression at the level of proteins and transcripts. CONCLUSION: Therefore, the expression of both BRCA1 and BRCA2 genes at the protein and mRNA levels appear to be up-regulated after cell proliferation in human tumor cell lines from different origins.


Assuntos
Proteína BRCA1/genética , Proteína BRCA2/genética , Regulação Neoplásica da Expressão Gênica , Genes BRCA1 , Genes BRCA2 , Proteína BRCA1/análise , Proteína BRCA1/biossíntese , Proteína BRCA2/análise , Proteína BRCA2/biossíntese , Éxons/genética , Humanos , Imuno-Histoquímica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA