Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Anticancer Res ; 38(3): 1531-1537, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29491082

RESUMO

BACKGROUND/AIM: Prostate-specific membrane antigen (PSMA) is emerging as a target for treatment of castration-resistant prostate cancer (CRPC) while its up-regulated in the majority of CRPC tumors. The most common approach is targeted radionuclide therapy. MATERIALS AND METHODS: The PSMA binding pharmacophore Glu-Urea-Lysine (GUL) and lysine were conjugated to oxidized dextran with reductive amination and subsequently labelled with fluorosceinisothiocyanate (FITC). Three prostate cancer cell lines were used for binding studies, 22Rv1 (PSMA positive), DU145 (PSMA negative) and PC3 (PSMA negative). Binding images were obtained by fluorescence microscopy. RESULTS: PDC binding was recorded on the 22Rv1 cell line while the negative cell lines showed no or slight background binding. PDC binding could be inhibited by pre-incubation with a molar excess of unlabelled PDC. CONCLUSION: This is a novel template for PSMA targeted CRPC therapy, either using cytostatics or radionuclides.


Assuntos
Antígenos de Superfície/metabolismo , Glutamato Carboxipeptidase II/antagonistas & inibidores , Glutamato Carboxipeptidase II/metabolismo , Compostos Radiofarmacêuticos/metabolismo , Compostos Radiofarmacêuticos/farmacologia , Linhagem Celular Tumoral , Dextranos/química , Fluoresceína-5-Isotiocianato/química , Ácido Glutâmico/química , Humanos , Lisina/química , Masculino , Microscopia de Fluorescência , Terapia de Alvo Molecular/métodos , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Ligação Proteica/efeitos dos fármacos , Compostos Radiofarmacêuticos/síntese química , Ureia/química
2.
Ups J Med Sci ; 111(3): 293-301, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17578796

RESUMO

The exocrine secretion of the acinar gland cells in the human prostate consists of, among other components, a serous secretion and prostasomes. The prostasomes are functionally associated with both reproduction and prostate cancer development and are capable to raise autoantibodies at various pathologies. Therefore, we are trying to characterize prostasome antigens by analysing prostasome-producing cell lines of prostate cancers with the cDNA microarray technique. To obtain one state with synthesis of prostasomes and another state without synthesis, we checked whether the prostasome differentiation was influenced by the mode of growing the cells, that is, whether the cells had been growing on a solid support or on a flexible one. We studied the expression of prostasomes in the cell lines PC3, DU145 and LNCaP. We grew the cells with the following methods: Monocellular layers on microbeads, multicellular spheroids, single cells in suspension cultures, and xenotransplants in nude rats. The presence of prostasomes was examined by ELISA, immunocytochemistry or electron microscopy. The results showed that growing the cells on microbeads (solid support) produced a differentiation of prostasomes, while growing the cells in multicellular spheroids (flexible support) did not. Thus it should be possible to apply cDNAmicroarray analyses for characterizing the genes which are active at the cellular expression of prostasomes and then deduce the prostasome antigens.


Assuntos
Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Vesículas Secretórias/genética , Vesículas Secretórias/metabolismo , Animais , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Masculino , Microscopia Eletrônica de Transmissão , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/patologia , Ratos , Ratos Nus , Vesículas Secretórias/patologia , Esferoides Celulares , Transplante Heterólogo
4.
Springerplus ; 5(1): 1861, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27822437

RESUMO

Docetaxel has been the standard first-line therapy in metastatic castration resistant prostate cancer. The survival benefit is, however, limited by either primary or acquired resistance. In this study, Du145 prostate cancer cells were converted to docetaxel-resistant cells Du145-R and Du145-RB by in vitro culturing. Next generation RNAseq was employed to analyze these cell lines. Forty-two genes were identified to have acquired mutations after the resistance development, of which thirty-four were found to have mutations in published sequencing studies using prostate cancer samples from patients. Fourteen novel and 2 previously known fusion genes were inferred from the RNA-seq data, and 13 of these were validated by RT-PCR and/or re-sequencing. Four in-frame fusion transcripts could be transcribed into fusion proteins in stably transfected HEK293 cells, including MYH9-EIF3D and LDLR-RPL31P11, which were specific identified or up-regulated in the docetaxel resistant DU145 cells. A panel of 615 gene transcripts was identified to have significantly changed expression profile in the docetaxel resistant cells. These transcriptional changes have potential for further study as predictive biomarkers and as targets of docetaxel treatment.

5.
Oncotarget ; 6(25): 21740-54, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25844599

RESUMO

Docetaxel is a cornerstone treatment for metastatic, castration resistant prostate cancer (CRPC) which remains a leading cause of cancer-related deaths, worldwide. The clinical usage of docetaxel has resulted in modest gains in survival, primarily due to the development of resistance. There are currently no clinical biomarkers available that predict whether a CRPC patient will respond or acquire resistance to this therapy. Comparative proteomics analysis of exosomes secreted from DU145 prostate cancer cells that are sensitive (DU145 Tax-Sen) or have acquired resistance (DU145 Tax-Res) to docetaxel, demonstrated significant differences in the amount of exosomes secreted and in their molecular composition. A panel of proteins was identified by proteomics to be differentially enriched in DU145 Tax-Res compared to DU145 Tax-Sen exosomes and was validated by western blotting. Importantly, we identified MDR-1, MDR-3, Endophilin-A2 and PABP4 that were enriched only in DU145 Tax-Res exosomes. We validated the presence of these proteins in the serum of a small cohort of patients. DU145 cells that have uptaken DU145 Tax-Res exosomes show properties of increased matrix degradation. In summary, exosomes derived from DU145 Tax-Res cells may be a valuable source of biomarkers for response to therapy.


Assuntos
Antineoplásicos/química , Resistencia a Medicamentos Antineoplásicos , Exossomos/efeitos dos fármacos , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Taxoides/química , Taxoides/uso terapêutico , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Biomarcadores Tumorais/metabolismo , Proteínas Sanguíneas/metabolismo , Morte Celular , Linhagem Celular Tumoral/efeitos dos fármacos , Estudos de Coortes , Biologia Computacional , Docetaxel , Exossomos/metabolismo , Matriz Extracelular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Nanopartículas/química , Proteínas de Ligação a Poli(A)/metabolismo , Neoplasias de Próstata Resistentes à Castração/metabolismo , Proteoma
6.
Oncotarget ; 6(35): 37066-82, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26416459

RESUMO

Autophagy is one of the main cytoprotective mechanisms that cancer cells deploy to withstand the cytotoxic stress and survive the lethal damage induced by anti-cancer drugs. However, under specific conditions, autophagy may, directly or indirectly, induce cell death. In our study, treatment of the Atg5-deficient DU145 prostate cancer cells, with the multi-tyrosine kinase inhibitor, sorafenib, induces mitochondrial damage, autophagy and cell death. Molecular inhibition of autophagy by silencing ULK1 and Beclin1 rescues DU145 cells from cell death indicating that, in this setting, autophagy promotes cell death. Re-expression of Atg5 restores the lipidation of LC3 and rescues DU145 and MEF atg5-/- cells from sorafenib-induced cell death. Despite the lack of Atg5 expression and LC3 lipidation, DU145 cells form autophagosomes as demonstrated by transmission and immuno-electron microscopy, and the formation of LC3 positive foci. However, the lack of cellular content in the autophagosomes, the accumulation of long-lived proteins, the presence of GFP-RFP-LC3 positive foci and the accumulated p62 protein levels indicate that these autophagosomes may not be fully functional. DU145 cells treated with sorafenib undergo a caspase-independent cell death that is inhibited by the RIPK1 inhibitor, necrostatin-1. Furthermore, treatment with sorafenib induces the interaction of RIPK1 with p62, as demonstrated by immunoprecipitation and a proximity ligation assay. Silencing of p62 decreases the RIPK1 protein levels and renders necrostatin-1 ineffective in blocking sorafenib-induced cell death. In summary, the formation of Atg5-deficient autophagosomes in response to sorafenib promotes the interaction of p62 with RIPK leading to cell death by necroptosis.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia , Embrião de Mamíferos/patologia , Fibroblastos/patologia , Niacinamida/análogos & derivados , Compostos de Fenilureia/farmacologia , Neoplasias da Próstata/patologia , Animais , Antineoplásicos/farmacologia , Proteína 5 Relacionada à Autofagia , Western Blotting , Células Cultivadas , Resistencia a Medicamentos Antineoplásicos , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Masculino , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Necrose , Niacinamida/farmacologia , Fagossomos/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Sorafenibe , Análise Serial de Tecidos
7.
Anticancer Res ; 24(3a): 1347-51, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15274294

RESUMO

BACKGROUND: Many previous studies show that cell surface sialylation of malignant cells is enhanced compared to normal tissue. The carboxyl group of the sialic acid yields, a negative surface charge of the tumor cells. This study investigates how tumor cell growth is affected when a cationic polymer is incubated with six different tumor cell lines. MATERIALS AND METHODS: Cationic dextran (CatDex) was prepared by periodate oxidation and subsequent coupling of cationic sidegroups by reductive amination. A fluorimetric cytotoxicity assay (FMCA) was used for the cell survival assay. Six different tumor cell lines (lung, breast, ovarian, prostate, colon, urinary bladder) were seeded into 96-well microtiter plates. CatDex was added at different microM concentrations and incubated for 72 h. Additionally, CatDex was fluorescence-labeled (FITC) and the interaction with the tumor cells was studied using fluorescence microscopy. The presence of sialic acid in the different cell lines was confirmed by using a FITC-labeled sialic acid binding lectin. RESULTS: CatDex showed a concentration-dependent growth inhibitory effect (i.e. the number of cationic side groups/dextran molecule and the molarity used). If the substitution was <20%, the growth inhibitory effect was small and difficult to reproduce. With 20-22% substitution, the growth inhibition varied between 20-95% depending on the molarity and the tumor type. Higher substitution resulted in complete cell death in all the cell lines. The fluorescent images showed intensive cell membrane interaction. CONCLUSION: Incubation with cationic dextran caused cell death in all six tumor cell lines. Our hypothesis is that CatDex binds to the anionic sialic acid residues and causes fatal disturbances in the cell membrane. However the exact mechanism remains to be elucidated. The results may indicate a new method of general interest for intra/local/regiolocal treatment of cancer. Clinical studies to explore this concept are pending.


Assuntos
Dextranos/farmacologia , Neoplasias/patologia , Cátions , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Microscopia de Fluorescência , Ácido N-Acetilneuramínico/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
8.
Anticancer Res ; 22(2A): 741-4, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12014645

RESUMO

BACKGROUND: In a previous study we reported on a new approach describing intravesical instillation of charged dextran in patients with superficial bladder carcinoma. The cationic derivative showed a strong tumor-selective accumulation. To develop this approach, the present study investigates the cytotoxic effect of cationic dextran derivatives on two urinary bladder cancer cell lines (J82 and 5637). METHODS: The dextran conjugates were prepared by periodate activation and subsequent coupling by reductive amination. A fluorimetric cytotoxicity assay (FMCA) was used for the cytotoxicity assay. The tumor cells were seeded into 96-well microtiter plates and different cationic dextran derivatives were added and incubated for 72 hours. RESULTS: The results showed that cationic epirubicin-dextran had a clear inhibitory effect on the growth in both cell lines (40-95% growth inhibition). The corresponding values for epirubicin (the reference) was 90-100% inhibition. Interestingly, cationic dextran had, by itself, a growth inhibitory effect. This cytotoxic effect could be strongly enhanced to be almost equal to the reference by changing the cationic sidegroup to aminohexane. Dextran alone showed no effect. CONCLUSION: The finding that cationic dextran by itself can be made cytotoxic, together with its capacity to accumulate in superficial bladder cancer, suggests possibilities for new therapeutic constructs. Cationic dextran with different cationic side-groups and in combination with cytotoxic drugs will be studied further. The cytotoxic mechanism needs to be elucidated.


Assuntos
Antineoplásicos/toxicidade , Carcinoma de Células de Transição/tratamento farmacológico , Dextranos/toxicidade , Neoplasias da Bexiga Urinária/tratamento farmacológico , Antineoplásicos/síntese química , Dextranos/síntese química , Epirubicina/análogos & derivados , Epirubicina/síntese química , Epirubicina/toxicidade , Fluorometria , Humanos , Lisina/análogos & derivados , Lisina/síntese química , Células Tumorais Cultivadas
9.
Int J Oncol ; 37(1): 15-20, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20514392

RESUMO

The multiple tyrosine kinase inhibitor sorafenib has recently demonstrated clinical effects in patients with androgen-independent prostate cancer. These observations provided the rational for investigating the anti-tumoral properties of this compound on prostate cancer cell lines at the molecular level. Two hormone refractory (PC3 and DU145) and one hormone responsive cell line (22Rv1) were used. By use of a panel of cell biology techniques such as immunoblotting, flow cytometry and immunocytochemistry, effects on the MAPK pathway and induction of apoptosis and autophagy were evaluated. We demonstrate that sorafenib reduced cell viability in a dose-dependent manner, induced apoptosis and inactivated the MAPK pathway. Moreover, we show for the first time, that sorafenib treatment of prostate cancer cells also induces cellular autophagy. This feature is in accordance with the anticancer potential of sorafenib and adds another important effector mechanism of this compound. These observations may open potentially interesting treatment combinations that may augment the effect of sorafenib, either by drugs that promote autophagy such as the rapalogues, or by combining sorafenib with compounds that specifically inhibit the autophagic process.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Benzenossulfonatos/farmacologia , Carcinoma/patologia , Neoplasias da Próstata/patologia , Piridinas/farmacologia , Antineoplásicos/farmacologia , Apoptose/genética , Carcinoma/genética , Carcinoma/metabolismo , Avaliação Pré-Clínica de Medicamentos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes bcl-2/fisiologia , Humanos , Masculino , Niacinamida/análogos & derivados , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Compostos de Fenilureia , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Sorafenibe , Transfecção , Células Tumorais Cultivadas
10.
Scand J Urol Nephrol ; 43(2): 98-103, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19101859

RESUMO

OBJECTIVE: To study and characterize the potential antitumoral effects of zoledronic acid (ZA) on renal cancer cell lines in vitro. MATERIAL AND METHODS: Three different and well-characterized renal cancer cell lines were studied, namely ACHN, A-498 and CAKI-2. The cytotoxic potential of ZA was evaluated using a fluorometric microculture cytotoxic assay. The degree of M30 induction following ZA treatment was measured using an M30 enzyme-linked immunosorbent assay (ELISA), and the blockage of this effect was studied using a pan-caspase inhibitor. Immunofluorescence of the M30 neoepitope was performed to visualize the M30-inducing properties of ZA. RESULTS: A significant reduction in viable cells was seen for all three cell lines following treatment with ZA, compared with untreated controls. This effect was most pronounced for the ACHN cells, as only 4% were viable following incubation with ZA for 72 h. A concomitant increase in the apoptosis significant caspase-dependent M30 antigen was demonstrated. This effect could be blocked by the pan-caspase inhibitor Z-VAD. CONCLUSIONS: ZA exerts cytotoxic effects on renal cancer cell lines in vitro. These include caspase-dependent induction of apoptosis, which can be quantified and visualized using M30 ELISA and immunofluorescence, respectively. The clinical relevance of this finding needs to be further investigated, but these results indicate that ZA may be a treatment alternative for selected patients with skeletal metastasized renal cell cancer, particularly for those with impaired performance status without other treatment options.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Difosfonatos/farmacologia , Imidazóis/farmacologia , Neoplasias Renais/tratamento farmacológico , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/patologia , Caspases/fisiologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Ensaio de Imunoadsorção Enzimática , Fluorometria , Humanos , Neoplasias Renais/patologia , Ácido Zoledrônico
11.
Acta Oncol ; 44(5): 490-5, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16118083

RESUMO

The potential overexpression of HER2 in prostate cancer cells has attended significant interest during the past few years, both as potential target for HER2 pathway focused therapy and as a mechanism involved in the progression to androgen independence. Conflicting results have been reported concerning HER2 status on clinical material, differences which generally have been attributed to methodological differences. Nevertheless, HER2 has been utilized for targeted therapy of prostate cancer in a number of preclinical studies and is still regarded as an exciting target molecule. In this study, the HER2 status of three widely used prostate cancer cell lines and corresponding xenografts has been analysed. By use of validated and FDA approved analytical staining techniques none of these cell lines or xenografts were shown to overexpress/amplify HER2, as demonstrated by immunohistochemistry and fluorescence in situ hybridization. These findings are important for the interpretation and understanding of the therapeutic effects when developing drugs targeting HER2 in prostate cancer cell lines and also emphasize the importance of using broad and validated analytical techniques.


Assuntos
Biomarcadores Tumorais/metabolismo , Genes erbB-2 , Neoplasias da Próstata/metabolismo , Receptor ErbB-2/biossíntese , Animais , Anticorpos Monoclonais , Neoplasias da Mama Masculina/genética , Neoplasias da Mama Masculina/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Masculino , Camundongos , Regulação para Cima
12.
Acta Oncol ; 44(6): 644-50, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16165924

RESUMO

Once bone metastasized and androgen independent, prostate cancer is often associated with skeletal morbidity and disability. New treatment modalities that can palliate symptoms from the skeleton and inhibit further progression are warranted. In this study, the antitumoral effects following treatment with a combination of docetaxel and the new generation bisphosphonate, zoledronic acid, were investigated on two hormone-refractory prostate cancer cell lines: PC3 and DU145. The prostate cancer cells were treated with increasing concentrations of zoledronic acid in the absence or presence of docetaxel. Toxicity was measured using fluorometric microculture cytotoxic assay technique. A concentration of 25 microM, zoledronic acid reduced the viable cell number to 68% and 98% for PC3 and DU145 cells respectively. Docetaxel, on the other hand, at a concentration of 0.1 ng/ml, had no effect on the viability. However, a combination of zoledronic acid and docetaxel reduced the cell number to 60% and 81% respectively. Furthermore, zoledronic acid in the concentration range 12.5 microM-50 microM enhanced the antitumoral effects of docetaxel (0.01-1 ng/ml) in an additive and/or synergistic manner for both cell lines. These data support the hypothesis that zoledronic acid, in addition to having bone resorption inhibiting properties, also exhibits anti-tumoral effects. It also appears that combined treatment with docetaxel causes additive and/or synergistic cytostatic effects on prostate cancer cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Conservadores da Densidade Óssea/administração & dosagem , Difosfonatos/administração & dosagem , Imidazóis/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Taxoides/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Conservadores da Densidade Óssea/farmacologia , Neoplasias Ósseas/prevenção & controle , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Difosfonatos/farmacologia , Docetaxel , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Imidazóis/farmacologia , Masculino , Mitose/efeitos dos fármacos , Taxoides/farmacologia , Ácido Zoledrônico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA