Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Wound Repair Regen ; 21(2): 300-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23438188

RESUMO

In recent years, interest in the topical use of platelet gel (PG) to stimulate wound healing has rapidly extended into various clinical applications and specialized fields. Many recent in vitro and in vivo studies have attempted to explain the biological mechanisms involved in PG-induced tissue regeneration/reparation. However, it remains unclear which parameters should be used in clinical applications to obtain satisfactory results in the healing of wounds. Toward this end, the present study focused on understanding the relationship between platelet concentrations and the cellular parameters of the cell types, i.e., fibroblasts, involved in wound healing. Normal human dermal fibroblasts were treated with PG-released supernatant at various concentrations in different assays (proliferation, migration, invasion, and in vitro scratch wound closure) to identify the most effective concentration to promote the fibroblasts' activities. Different concentrations of platelets per microliter in PG have different levels of efficacy in inducing fibroblast activity. The most effective concentration was obtained from PG at a concentration of approximately 0.5-1.5 × 10(6) plt/µL; higher concentrations were less effective. This study shows that excessively high concentrations of platelets per microliter have an inhibitory effect on the wound healing processes and are, therefore, counterproductive.


Assuntos
Plaquetas/metabolismo , Proteínas de Transporte/metabolismo , Fibroblastos/metabolismo , Lipoproteínas/metabolismo , Metaloproteinases da Matriz/metabolismo , Transativadores/metabolismo , Cicatrização , Ferimentos e Lesões/metabolismo , Western Blotting , Proteínas de Transporte/farmacologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Géis/farmacologia , Humanos , Lipoproteínas/farmacologia , Regeneração , Transativadores/farmacologia , Ferimentos e Lesões/tratamento farmacológico
2.
Gynecol Oncol ; 119(3): 557-63, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20825984

RESUMO

OBJECTIVES: The purpose of this study was to determine whether the addition of the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) to paclitaxel (PTX) can sensitize PTX-resistant human ovarian cancer cell lines (CABA-PTX and IGROV-PTX) in vitro. METHODS: SAHA was studied in combination with paclitaxel in PTX-sensitive and PTX-resistant human ovarian cancer cell lines. Using cell proliferation analysis, immunofluorescence, and flow cytometric assays, we can determine whether the resistance was partly removed when the cells were treated with a combination of SAHA and PTX. Cells were also assayed for cytochrome c release. The levels of acetylated tubulin, ß-tubulin, and HDAC6 were quantified by Western blots. RESULTS: SAHA in combination with PTX led to a more pronounced inhibition of cell growth compared with PTX alone. In addition, the combined exposure to PTX and SAHA resulted in a marked arrest in the G2/M phase of the cell cycle and in a significant increase in the percentage of apoptotic cells. The expression of acetylated tubulin was dramatically increased by exposure to the combination of PTX and SAHA. These data paralleled the findings of an increased expression of HDAC6 in the presence of PTX in PTX-resistant cell lines. CONCLUSIONS: The results of this study suggest the existence of a novel resistance mechanism based upon the upregulation of HDAC6 and that the histone deacetylase inhibitor SAHA holds promise to overcome PTX resistance in ovarian cancer cell lines.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/farmacologia , Acetilação , Divisão Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Feminino , Fase G2/efeitos dos fármacos , Inibidores de Histona Desacetilases/administração & dosagem , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Paclitaxel/administração & dosagem , Tubulina (Proteína)/metabolismo , Vorinostat
3.
Cancer Res ; 70(15): 6150-60, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20631064

RESUMO

Breast cancer shows a strong predilection to metastasize to bone. Cell surface glycoprotein extracellular matrix metalloproteinase inducer (EMMPRIN)/CD147 induces metalloproteinases (MMP) and vascular endothelial growth factor (VEGF), which may support osteoclastic activity and increased incidence of breast cancer bone metastases. In support of this hypothesis, we observed that MDA-MB-231 human breast tumor cells engineered to overexpress EMMPRIN strongly induced osteolytic lesions in immunodeficient mice, which was blunted by in vivo treatment with an EMMPRIN blocking antibody. Similarly, these cells exhibited increased expression of MMP-9 and VEGF relative to control cells. Treatment of MDA-MB-231 cells with the osteoclastogenic cytokine receptor activator of NF-kappaB ligand (RANKL) upregulated EMMPRIN expression with a parallel increase of MMP-9 and VEGF. Conditioned medium from osteoblasts similarly increased EMMPRIN, MMP-9, and VEGF expression in cells. Osteoblast treatment with the RANKL decoy receptor osteoprotegerin abolished this effect. EMMPRIN overexpression stimulated MDA-MB-231 cell invasion but not proliferation. Conversely, small interfering RNA-mediated knockdown of EMMPRIN downregulated MMP-9 and VEGF basal expression and RANKL-stimulated expression, and reduced cell invasion. Our results argue that EMMPRIN drives breast cancer-induced osteolytic lesions and that activation of the RANKL pathway increases EMMPRIN in osteotropic tumor cells, in turn enhancing tumor-induced bone resorption.


Assuntos
Basigina/biossíntese , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Ligante RANK/metabolismo , Animais , Basigina/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes , Humanos , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ligante RANK/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transfecção , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
4.
Int J Oncol ; 35(6): 1423-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19885565

RESUMO

The term vasculogenic mimicry (VM) indicates the process by which aggressive tumor cells are able to generate in vitro non-endothelial cell-lined channels delimited by extracellular matrix. Although VM has been described in several human malignancies, the molecular basis of this phenomenon is not entirely understood. In the present study, we examined VM in two ovarian cancer cell lines with different invasion capability (CABA I, low invasion activity; SKOV3, high invasion activity). Specifically, we focused on the potential role played by CD147/extracellular MMP inducer, a membrane spanning molecule highly expressed in tumor cells, in VM. Previous studies have shown that CD147 may be involved in the progression of malignancies by regulating the expression of metalloproteinases in peritumoral stromal cells. In this study, we found significant correlations between expression of CD147 in ovarian cancer cell lines and tumor invasiveness, the activity of the proteases and the ability to form vascular channels. The treatment of SKOV3 cells with small interfering RNA against CD147 suppressed the ability of these cells to generate non-endothelial cell-lined channels. In contrast, transfection of CD147 cDNA into the CABA I cell line resulted in an increased tumor invasiveness and enabled the formation of vascular channels. Altogether, our data suggest that CD147 may play a critical role in VM of CABA I and SKOV3, human ovarian cancer cell lines.


Assuntos
Basigina/metabolismo , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/patologia , Western Blotting , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Ovarianas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Urology ; 74(2): 452-7, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19285710

RESUMO

OBJECTIVES: To evaluate the role of Her2/neu as a molecular marker predictive of the treatment response to bicalutamide in prostate cancer (PCa). METHODS: Four PCa cell lines with graded Her2/neu expression levels and 63 primary tumor cultures derived from men with PCa and selected according to Her2/neu tumor levels were used. Primary tumor cultures and PCa cell lines were treated with bicalutamide (0.1-10 microM) in the presence of dehydrotestosterone (10(-12) M) for 4 days. The presence of a significant correlation between Her2/nue expression and drug efficacy was used to define the role of Her2/neu as molecular predictor of bicalutamide effectiveness. As an indicator of drug effectiveness we used the concentration that inhibits 50% values determined after bicalutamide treatment. RESULTS: After bicalutamide treatment, no significant differences in the concentration that inhibits 50% were found among the different tumor cell lines (P = .081). In this experimental model, the correlation analysis suggested that the effectiveness of this drug was not influenced by Her2/neu levels (r = 0.053, P = .823). In primary cultures with high Her2/neu levels (43 tumor cultures), the mean value of the concentration that inhibits 50% for bicalutamide was 0.43 +/- 0.13 microM, and in cultures with low Her2/neu levels (20 tumor cultures), the same parameter was 0.5 +/- 0.16 microM (P = .14). The correlation analysis suggested that the effectiveness of this drug was not influenced by Her2/neu levels (r = 0.33, P = .101). CONCLUSIONS: Our biologic data seem to indicate that the antitumor effect of bicalutamide is independent of Her2/neu levels in preclinical models of PCa. Bicalutamide could be configured as a pharmacologic option to treat patients with high or low levels of Her2/neu.


Assuntos
Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Antineoplásicos/farmacologia , Nitrilas/farmacologia , Neoplasias da Próstata/patologia , Receptor ErbB-2/metabolismo , Compostos de Tosil/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Fator de Crescimento Epidérmico/farmacologia , Citometria de Fluxo , Humanos , Masculino , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Células Tumorais Cultivadas
6.
Endocr Relat Cancer ; 16(2): 401-13, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19153211

RESUMO

One of the major obstacles in the treatment of hormone-refractory prostate cancer (HRPC) is the development of chemoresistant tumors. The aim of this study is to evaluate the role of azacitidine as chemosensitizing agent in association with docetaxel (DTX) and cisplatin using two models of aggressive prostate cancer, the 22rv1, and PC3 cell lines. Azacitidine shows antiproliferative effects associated with increased proportion of cells in G0/G1 and evident apoptosis in 22rv1 cells and increased proportion of cells in G2/M phase with the absence of acute cell killing in PC3 cells. In vivo, azacitidine (0.8 mg/kg i.p.) reduced tumor proliferation and induced apoptosis in both xenografts upmodulating the expression of p16INKA, Bax, Bak, p21/WAF1, and p27/KIP1, and inhibiting the activation of Akt activity and the expression of cyclin D1, Bcl-2, and Bcl-XL. In vitro treatments with azacitidine lead to upregulation of cleaved caspase 3 and PARP. BCl2 antagonists, such as HA-14-1, enhanced the effects of azacitidine in these two prostate cancer models. In addition, azacitidine showed synergistic effects with both DTX and cisplatin. In vivo this agent caused tumor growth delay without complete regression in xenograft systems. Azacitidine sensitized PC3 and 22rv1 xenografts to DTX and cisplatin treatments. These combinations were also tolerable in mice and superior to either agent alone. As DTX is the standard first-line chemotherapy for HRPC, the development of DTX-based combination therapies is of great interest in this disease stage. Our results provide a rationale for clinical trials on combination treatments with azacitidine in patients with hormone-refractory and chemoresistant prostate tumors.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Azacitidina/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Neoplasias da Próstata/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/metabolismo , Cisplatino/administração & dosagem , Ciclina D1/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Docetaxel , Sinergismo Farmacológico , Quimioterapia Combinada , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Taxoides/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
7.
Transfusion ; 49(4): 771-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19170984

RESUMO

BACKGROUND: Numerous studies have supported the use of topical blood components to improve wound healing and tissue regeneration. Platelet gel (PG), a hemocomponent obtained from mix of activated platelets (PLTs) and cryoprecipitate, is currently being used clinically in an attempt to improve tissue healing. The present study sought to define the most effective PG concentration to promote angiogenesis in vitro. STUDY DESIGN AND METHODS: The effects of PG-released supernatant at different concentrations on human endothelial cells were studied using different in vitro assays (proliferation, migration, invasion, cord formation, and wound healing). RESULTS: The concentration of PG-released supernatant had a significant influence on the angiogenic potential of endothelial cells. The optimal concentration for the stimulation of angiogenesis was 1.5 x 10(6) PLTs per microL in most of the in vitro experiments used in this study. Lower or higher concentrations of PG displayed a lower angiogenic potential. CONCLUSION: An optimal concentration of PG to promote angiogenesis in human endothelial cells was identified. Excessively high PG concentrations may inhibit the angiogenic process, thereby being counterproductive for wound healing in a clinical setting.


Assuntos
Plaquetas/fisiologia , Células Endoteliais/efeitos dos fármacos , Géis/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Plaquetas/química , Plaquetas/metabolismo , Adesão Celular/efeitos dos fármacos , Técnicas de Cultura de Células , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/fisiologia , Humanos , Masculino , Camundongos , Células NIH 3T3 , Concentração Osmolar
8.
Prostate ; 69(4): 337-45, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19016477

RESUMO

BACKGROUND: To date, no effective therapeutic treatment prevents prostate cancer (PCa) progression to more advanced and invasive disease forms. It has been demonstrated that the simultaneous high expression of p185(HER2) and TrkA might confer a proliferative advantage to PCa cells. METHODS: In this work we verified the crosstalk between TrkA and Her2 signaling pathways and the effects of a combined treatment with Her2 and TrkA inhibitors. RESULTS: NGF induced TrkA activation and stimulated cell proliferation of PCa cells. NGF induced also tyrosine phosphorylation of p185(HER2). This event was only partially inhibited by the pan Trk inhibitor, CEP-701 but was strongly blocked by pertuzumab, a humanized antibody blocking Her2 heterodimerization. In presence of NGF, TrkA and Her2 co-precipitated and this was dependent to the relative high cellular levels of TrkA since when cell lysates were immunoprecipitated with an antibody against Her2 the amount of TrkA were proportional to the cellular levels of this receptor. On the contrary when we immunoprecipitated using an antibody against TrkA the amount of Her2 seemed independent to cellular levels of Her2. So, combined treatment between CEP-701 and pertuzumab showed supra-additive effects in cells with higher levels of TrkA and Her2 suggesting once again that this was indicative of a higher response to treatment. CONCLUSIONS: Our data suggest that the dual inhibition of TrkA and Her2 may be useful in a subset of patients in which TrkA and Her2 are overexpressed and in which the possibility of TrkA and Her2 protein-binding is elevated.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Carbazóis/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Receptor Cross-Talk/fisiologia , Receptor ErbB-2/metabolismo , Receptor trkA/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Carbazóis/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimioterapia Combinada , Furanos , Masculino , Camundongos , Fator de Crescimento Neural/farmacologia , Neoplasias da Próstata/patologia , Receptor Cross-Talk/efeitos dos fármacos , Receptor ErbB-2/efeitos dos fármacos , Receptor trkA/efeitos dos fármacos , Receptores Androgênicos/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Int J Oncol ; 33(2): 381-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18636160

RESUMO

One of the major obstacles in curing prostate cancer is the development of drug resistance. It is not only imperative to discover the molecular basis of resistance but also to find therapeutic agents that can disrupt the resistant pathways. Tumor necrosis factor TNF-related apoptosis-inducing ligand TRAIL-like ligands or agonist TRAIL-receptor monoclonal antibodies have entered phase I and II clinical trials with a very limited cytotoxic profile when used systemically in a variety of cancers. Therefore, TRAIL-receptor agonists are new proapoptotic pharmaceutical agents with great potential as new cancer therapeutic agents. Although many cancer cells undergo TRAIL-mediated apoptosis, some are resistant to TRAIL. Therefore, we have been investigating mechanisms to overcome TRAIL resistance in cancer cells so that TRAIL-associated compounds can be used effectively in clinical trials. Epigenetic inactivation of proapoptotic genes, or activation of survival signaling, can cause cross-resistance to several anti-tumor therapies and to immune cytotoxic lymphocytes. We hypothesize that 5-aza-2 deoxycytidine aza-dCR, decitabine may render TRAIL-resistant prostate cancer cells sensitive to caspase-8-mediated apoptosis and may, therefore, be therapeutically efficient. We evaluated the antiproliferative effects of decitabine on the following four prostate cancer cell lines: well-differentiated AR positive LnCaP p53(+), PTEN- and 22rv1 p53(+) and PTEN(+)]; poorly-differentiated AR negative PC3 p53-, PTEN- and DU145 p53 mutant, PTEN(+). Here, we provide evidence that treatment with sub-optimal concentrations of decitabine are additive to TRAIL effects in well-differentiated PCa cells whereas the same treatment shows synergistic effects in poorly-differentiated PCa cells through increased caspase-8 expression, down-modulation of Akt activation and through the expression of certain anti-apoptotic molecules including FLIP, PED/PEA-15, survivin and c-IAP-1. Our findings demonstrate that decitabine at relatively low concentrations restores caspase-8 expression and sensitises resistant PCa cells to TRAIL-induced apoptosis leading to important implications in novel therapeutic strategies targeting defective apoptosis pathways in advanced prostate tumors.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias da Próstata/enzimologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Azacitidina/farmacologia , Western Blotting , Caspase 8/efeitos dos fármacos , Caspase 8/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Decitabina , Relação Dose-Resposta a Droga , Regulação para Baixo , Citometria de Fluxo , Humanos , Masculino , Neoplasias da Próstata/patologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
10.
Neoplasia ; 10(5): 481-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18472965

RESUMO

Vesicles shed by cancer cells are known to mediate several tumor-host interactions. Tumor microenvironment may, in turn, influence the release and the activity of tumor-shed microvesicles. In this study, we investigated the molecular mediators of the pH-dependent proinvasive activity of tumor-shed vesicles. Gelatinase zymography showed increased microvesicle activity of matrix metalloproteinases 9 and 2 as a result of acid exposure (pH 5.6) compared to pH 7.4. Thus, we reasoned that the cysteine protease cathepsin B might play a role in mediating the pH-dependent activation of gelatinases. Cathepsin B expression in tumor-shed microvesicles was confirmed by Western blot analysis and zymography. The activity of vesicle-associated cathepsin B measured using Z-Arg-Arg-pNA as substrate was significantly increased at acidic pH values. Inhibition of protease activity by the cysteine protease inhibitor, E-64, and treatment of ovarian cancer cells with small interfering RNA against cathepsin B suppressed the ability of tumor-shed microvesicles to stimulate both gelatinase activation and the invasiveness of endothelial cells observed at low pH values. We conclude that microvesicle shedding is a major secretory pathway for cathepsin B release from tumor cells. Hence, the acidic microenvironment found in most solid tumors may contribute to cathepsin B-mediated proinvasive capabilities of tumor-shed vesicles.


Assuntos
Catepsina B/metabolismo , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/patologia , Neoplasias Ovarianas/enzimologia , Catepsina B/genética , Linhagem Celular Tumoral , Movimento Celular , Meios de Cultivo Condicionados/farmacologia , Ativação Enzimática , Feminino , Humanos , Concentração de Íons de Hidrogênio , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Invasividade Neoplásica , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Prostate ; 68(9): 965-74, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18361408

RESUMO

BACKGROUND: PTEN is a well-characterized tumor suppressor that negatively regulates cell growth and survival through the modulation of PI3K/Akt pathway. METHODS: In this paper, we investigated the effects of an PI3K/Akt inhibitor, perifosine, in human prostate cancer (PCa) cells analyzing cell proliferation, apoptosis, and the synergy with EGFR inhibitors. RESULTS: Clinically achievable concentrations of perifosine, as well as Akt gene knockdown, induced a G0/G1 arrest and apoptosis in PTEN defective PCa cells. Although PTEN introduction was able to restore the control of Akt activity and to reduce cell proliferation, the manipulation of PTEN gene was not able alone to influence apoptosis. Perifosine induced apoptotic program also in PTEN positive cells when Akt activity was augmented by EGF suggesting the possibility that this drug could be used in combination with EGFR inhibitors. The combination treatment between erlotinib and pharmacological or molecular Akt knockdown, indeed, showed synergistic effects. This is the first demonstration that a pharmacological compound against Akt activity can restore the efficacy against EGFR inhibitors in PCa and has important therapeutic fallout since EGFR inhibitors have demonstrated very low effectiveness in PCa patients. CONCLUSIONS: Taken together our data have an important clinical relevance in the treatment of advanced prostate tumors. However, further studies in the setting of combination therapies in advanced PCas are necessary.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Hormônio-Dependentes/patologia , Fosforilcolina/análogos & derivados , Neoplasias da Próstata/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Apoptose/fisiologia , Western Blotting , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromonas/farmacologia , Sinergismo Farmacológico , Ativação Enzimática , Fator de Crescimento Epidérmico/imunologia , Cloridrato de Erlotinib , Citometria de Fluxo , Humanos , Masculino , Morfolinas/farmacologia , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/enzimologia , PTEN Fosfo-Hidrolase/metabolismo , Fosforilcolina/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Quinazolinas/farmacologia
12.
Prostate ; 68(7): 793-801, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18324645

RESUMO

BACKGROUND: About 20-30% of hormone-independent PCa are characterized by the extensive loss of AR expression that appears to occur at the transcriptional level. Treatment of AR-negative PCa cells with demethylating agents (Aza-CR) leads to expression of AR mRNA and protein. Here, we investigate the effect of Aza-CR administered both acutely and chronically on AR expression, PSA expression, cell survival, and proliferation in androgen-independent/AR-negative PCa cells. We also studied whether epigenetically reactivated AR is a target for bicalutamide therapy. METHODS: The in vitro effect of Aza-CR as single agent and its ability to induce AR expression and to augment the efficacy to bicalutamide were assessed using two androgen-independent and AR-negative cell lines (PC3 and DU145). RESULTS: Our results show that acute treatment (4 days) with Aza-CR results in a relatively low decrease in cell proliferation with G2 cell cycle arrest and no significant evidence of apoptosis or AR expression. Interestingly, when Aza-CR was chronically administered (20 days), this treatment resulted in marked decrease in tumor cell proliferation with significant increase in AR and PSA protein levels. Furthermore, following Aza-CR chronic treatment the formerly androgen-independent PC3 and DU145 cells increase their susceptibility to the apoptotic effects of bicalutamide. CONCLUSIONS: Aza-CR acute treatment has modest effects on androgen-independent and AR-negative PCa cell survival and proliferation, but chronic administration results in profound decrease in proliferation and in sensitization to antiandrogen agents. All these effects seem, in some measure, dependent on a partial restoration of androgen regulation.


Assuntos
Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Antineoplásicos/farmacologia , Azacitidina/farmacologia , Nitrilas/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Compostos de Tosil/farmacologia , Androgênios/metabolismo , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Inativação Gênica , Humanos , Masculino , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
13.
Prostate ; 68(6): 588-98, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18288684

RESUMO

BACKGROUND: Prostate cancer (PCa) is the most commonly diagnosed cancer in men in the Western Countries. When prostatectomy fails to eradicate the primary tumor, PCa is generally refractory to all therapeutic approaches. Valproic acid (VPA) is a promising anticancer agent recently assigned to the class of histone deacetylase (HDAC) inhibitors. However molecular mechanisms underlying VPA action in PCa cells are largely unknown and further experimental validation to prove its potential application in clinic practice is needed. RESULTS: In our study we show that VPA is a potent inducer of neuro-endocrine transdifferentiation (NET) in androgen receptor null PCa cells, both in vitro and in vivo. NET was an early event detectable through the expression of neuro-endocrine (NE) markers within 72 hr after VPA treatment and it was associated to a reduction in the overall cell proliferation. When we interrupted VPA treatment we observed the recovery in residual cells of the basal proliferation rate both in vitro and in a xenograft model. The NET process was related to Bcl-2 over-expression in non-NE PCa cells and to the activation of PPARgamma in NE cells. The use of specific PPARgamma antagonist was able to reduce significantly the expression of NE markers induced by VPA. CONCLUSIONS: Our data indicate that the use of VPA as monotherapy in PCa has to be considered with extreme caution, since it may induce an unfavorable NET. In order to counteract the VPA-induced NET, the inhibition of PPARgamma may represent a suitable adjuvant treatment strategy and awaits further experimental validation.


Assuntos
Adenocarcinoma/tratamento farmacológico , Transdiferenciação Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Sistemas Neurossecretores/efeitos dos fármacos , PPAR gama/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Ácido Valproico/farmacologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Anilidas/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transdiferenciação Celular/fisiologia , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Combinação de Medicamentos , Inibidores de Histona Desacetilases , Humanos , Masculino , Camundongos , Camundongos Nus , Sistemas Neurossecretores/patologia , PPAR gama/antagonistas & inibidores , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Oncol Rep ; 18(2): 503-11, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17611677

RESUMO

The clinical efficacy of ErbB1 kinase inhibitors is limited by the development of acquired autoresistance. The activation of alternative signaling pathways can contribute to gefitinib resistance. In this study, we demonstrate that the continuous in vitro exposure of the phosphatase and tensin homologue (deleted from chromosome 10)-negative prostate cancer (PC)3 cell line to gefitinib resulted in a sustained growth inhibition of 50% for about 2 months, but afterwards the surviving cells resumed their usual proliferation rate. During chronic treatment, gefitinib-treated cells developed drug resistance undergoing a G0/G1 cell cycle arrest, with a corresponding reduction in the G2/M cells without evident cell apoptosis, and thus a tyrosine kinase inhibitor-resistant (TKI-R) PC3 cell subline was isolated. TKI-R cells show i) an increment in basal ERK activation, ii) an epidermal growth factor-mediated and gefitinib insensitive ERK phosporylation, iii) increased levels of Her2/Neu, iv) a significant decrement in epidermal growth factor receptor (EGFR) expression, v) a very low sensitivity against EGFR TKIs and blocking antibodies, vi) a moderate increase in the sensitivity to growth inhibition by the Her2 inhibitor, AG825 or by 2C4, the humanized monoclonal antibody which blocks Her2 heterodymerization, vii) an increased expression of the neutrophine receptors, TrkA and TrkB, and viii) a significantly increased sensitivity to growth inhibition by the TrkA inhibitor, CEP701. Treatment with a mitogen-activated-protein kinase inhibitor abolished gefitinib resistance completely. Therefore, the ability of tumor cells to maintain high ERK activity under EGFR inhibition could represent a potential mechanism of the resistance to gefitinib.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Gefitinibe , Humanos , Masculino , Fator de Crescimento Neural/farmacologia , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Fatores de Tempo
15.
Neoplasia ; 9(4): 349-57, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17460779

RESUMO

Matrix metalloproteinase (MMP) degradation of extracellular matrix is thought to play an important role in invasion, angiogenesis, tumor growth, and metastasis. Several studies have demonstrated that CD147/extracellular MMP inducer, a membrane-spanning molecule highly expressed in tumor cells, may be involved in the progression of malignancies by regulating expression of MMP in peritumoral stromal cells. In the present study we show that CD147 is expressed in microvesicles derived from epithelial ovarian cancer cells and that CD147-positive vesicles may promote an angiogenic phenotype in endothelial cells in vitro. Vesicles shed by human ovarian carcinoma cell lines OVCAR3, SKOV3, and A2780 expressed different levels of CD147 and stimulated proangiogenic activities of human umbilical vein endothelial cells (HUVECs) in a CD147-dependent fashion (OVCAR3 > SKOV3 > A2780). Moreover, vesicles shed by ovarian carcinoma cell line CABA I with low CD147 expression had no significant effect on the development of angiogenic phenotype in HUVECs. The treatment of OVCAR3 cells with small interfering RNA against CD147 suppressed the angiogenic potential of OVCAR3-derived microvesicles. However, transfection of CD147 cDNA into the CABA I cell line enabled CABA I-derived vesicles to induce angiogenesis and to promote MMP genes expression in HUVECs. We therefore conclude that vesicles shed by ovarian cancer cells may induce proangiogenic activities of HUVECs by a CD147-mediated mechanism.


Assuntos
Basigina/fisiologia , Vesículas Citoplasmáticas/patologia , Células Endoteliais/patologia , Endotélio Vascular/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/patologia , Basigina/biossíntese , Basigina/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Vesículas Citoplasmáticas/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Feminino , Humanos , Neoplasias Epiteliais e Glandulares/irrigação sanguínea , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neovascularização Patológica/genética , Neoplasias Ovarianas/metabolismo
16.
Toxicol Lett ; 169(2): 129-36, 2007 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-17267145

RESUMO

The aim of this study is to investigate the effects in vitro induced by androgenic anabolic steroids (AAS) (testosterone, nandrolone, androstenedione, norandrostenedione, and norandrostenediol) used illicitly in sport competitions, on the proliferation ability, apoptosis and the intracellular calcium concentration ([Ca2+]i) in human umbilical vein endothelial cells (HUVECs), selected as a prototype of a biological target system whose structure and function can be affected by steroids. For this purpose, we evaluated the proliferation inhibition by cytotoxic assay expressed as the concentration of drug inducing a 50% decrease in growth (IC50). The IC50 was reached for testosterone at 100 microM, androstenedione at 375 microM, nandrolone at 9 microM, norandrostenedione at 500 microM. The IC50 value for norandrostenediol was not reached until a concentration of 6000 microM. The apoptotic effect was evaluated by flow cytometry at IC50 for each drug. We observed that testosterone induced 31% of apoptotic cells, norandrostenedione 25%, androstenedione 15% and nandrolone 18%. We have analyzed the effects of these drugs on [Ca2+]i both in the immediate and long-term continuous presence of each compound. Our data show a statistically significant increase of [Ca2+]i in the acute condition and in long-term treated cultures, suggesting that androgen steroids modulate intracellular levels of calcium independent of incubation time or compound identity. As a whole, this study demonstrates that AAS might alter endothelial homeostasis, predisposing to the early endothelial cell activation that is responsible for vascular complications observed frequently in AAS users.


Assuntos
Anabolizantes/toxicidade , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Esteroides/toxicidade , Androstenodiona/toxicidade , Cálcio/metabolismo , Dopagem Esportivo , Endotélio Vascular/metabolismo , Citometria de Fluxo , Fluorometria , Humanos , Concentração Inibidora 50 , Nandrolona/toxicidade , Testosterona/toxicidade
17.
Int J Oncol ; 30(1): 193-200, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17143529

RESUMO

In the prostate, cellular growth and differentiation are finely regulated by a complex interaction between stromal and epithelial cells under the control of both autocrine and paracrine regulatory factors such as the nerve growth factor (NGF). However, the role of NGF and its receptors including the high-affinity p-140 TrkA and the low-affinity p75 NTR receptors remains controversial. Moreover prostate tissues stored other neutrophins such as NT3, NT4 and brain derived neutrophic factor (BDNF) as well as the corresponding receptors (NTRs). Different members of NTRs are expressed during prostate cancer (PCa) progression, suggesting their involvement in cell proliferation, anoikis protection and malignancy. Therefore, we analyzed the expression of NTRs including NTRK1 (TrkA), NTRK2 (TrkB), NTRK3 (TrkC) and p75 NGFR in a panel of 7 well-characterized PCa cell lines and 12 cell derivatives from PC3 (4), DU145 (2), CWR22R (4) and LnCap (2) cell lines possessing different proliferative/invasive capabilities. We evaluated also the role of NGF, BDNF and NT3 in the modulation of cell migration and invasion and, finally, the effects of a pan Trk inhibitor, CEP-701 which has been included in some clinical trials for the treatment of PCa. We observed the following: i) TrkA and TrkB expression was significantly higher in AR-negative compared to AR-positive cells; ii) TrkA and TrkB expression was related to the invasive capacity/malignancy of PCa cells; iii) p75 NGFR could be considered a tumor suppressor gene which is present at high levels only in AR-positive cells; and iv) that NGF and BDNF (targeting TrkA/p75 NTR and TrKB, respectively) induced cell migration and this was inhibited by the CEP-701 treatment. In conclusion, the malignancy of PCa seems to be accompanied by increased TrkA and TrkB signaling (with a reduction of p75 NGFR expression) and CEP-701 could be used to reduce the metastasis formation in advanced PCa. CEP-701 is a trademark of Cephalon Inc., West Chester, PA, USA.


Assuntos
Carbazóis/farmacologia , Indóis/farmacologia , Neoplasias da Próstata/patologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Receptor trkA/antagonistas & inibidores , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Furanos , Humanos , Masculino , Invasividade Neoplásica , Fatores de Crescimento Neural/farmacologia , Receptor trkA/genética , Receptores Androgênicos/análise
18.
Anticancer Drugs ; 17(10): 1141-50, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17075313

RESUMO

Valproic acid is a well-known antiepileptic drug that was recently discovered to have a wide-spectrum antitumoral action in several tumors. In our work, we tested the proapoptotic activity of valproic acid in prostate cancer. Valproic acid-induced apoptosis was described by several in-vitro assays in three prostate cancer cell lines: two representing the prototype of advanced, clinically untreatable stages of prostate progression, PC3 and DU145, and one resembling a more differentiated androgen-sensitive tumor, LNCaP. We observed that valproic acid was a potent and early apoptotic inducer, mainly in less-differentiated prostate cancer cell lines. The molecular analysis of the apoptotic machinery involved in valproic acid action revealed a central role in Bcl-2 downmodulation. When prostate cancer cells were treated for a longer time with valproic acid, we detected an enhancement of Fas-dependent apoptosis associated with an overexpression in Fas and Fas ligand. Our data indicate that the use of valproic acid may be a suitable therapeutic agent in the control of prostate cancer progression and its action appears particularly relevant in the control of refractory stages of prostate cancer.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma/patologia , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos , Ácido Valproico/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteína Ligante Fas/metabolismo , Humanos , Masculino , Mitocôndrias/efeitos dos fármacos , Transcrição Gênica/fisiologia , Receptor fas/metabolismo
19.
Neoplasia ; 8(2): 96-103, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16611402

RESUMO

Tumor angiogenesis is regulated by a dynamic cross-talk between tumor cells and the host microenvironment. Because membrane vesicles shed by tumor cells are known to mediate several tumor-host interactions, we determined whether vesicles might also stimulate angiogenesis. Vesicles shed by human ovarian carcinoma cell lines CABA I and A2780 stimulated the motility and invasiveness of endothelial cells in vitro. Enzyme-linked immunosorbent assay and Western blot analysis revealed relevant amounts of vascular endothelial growth factor (VEGF) and the two matrix metalloproteinases MMP-2 and MMP-9, but not fibroblast growth factor-2, contained in shed vesicles. An A2780 cell-derived clone transfected to overexpress VEGF shed the same amount of vesicles as did a control clone, but contained significantly more VEGF within the vesicles. Despite a greater amount of VEGF in vesicles of the overexpressing clone, vesicles of both clones stimulated endothelial cell motility to comparable levels, suggesting that VEGF was stored within the vesicle and was unavailable. Only following vesicle burst induced by acidic pH (a characteristic of the tumor microenvironment) was VEGF released, leading to significantly higher stimulation of cell motility. Thus, tumor-shed membrane vesicles carry VEGF and release it in a bioactive form in conditions typical of the tumor microenvironment.


Assuntos
Endotélio Vascular/patologia , Concentração de Íons de Hidrogênio , Neovascularização Patológica/patologia , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/patologia , Fator A de Crescimento do Endotélio Vascular/farmacocinética , Animais , Disponibilidade Biológica , Bovinos , Linhagem Celular Tumoral , Movimento Celular , Células Cultivadas , Meios de Cultura , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Endotélio Vascular/fisiopatologia , Feminino , Humanos , Invasividade Neoplásica , Veias Umbilicais , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Endocr Relat Cancer ; 13(1): 197-210, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16601288

RESUMO

The activation of epidermal growth factor receptor (EGF-R) plays a key role in the promotion of proliferation and invasion in prostatic carcinoma (PCa). Gefitinib (Iressa; ZD1839), an orally active EGF-R tyrosine kinase inhibitor, has shown an important anti-proliferative activity in tumors expressing EGF-R both in vitro and in vivo. Our aim was to elucidate the role of gefitinib in the modulation of the metastatic spread of PCa cells. The therapeutic role of gefitinib was investigated by evaluating the proliferative and invasive ability of the PCa cell line PC3 and of its high metastatic sub-line, PCb2, by in vitro assays and intracardiac injection in nude mice. The inhibitory effect of gefitinib was tested in vivo by injecting PCa cells subcutaneously or in the left ventricle of nude mice and by administering daily 150 mg/kg of gefitinib. While xenograft growth was equally reduced in all PCa lines (about 50%), the bone metastasis formation was inhibited especially for the high metastatic PCb2 sub-line (81%) in comparison to PC3 cells (47%). The comparative in vitro analysis among PCa cell lines showed that PCb2 cells were more sensitive to the inhibitory effect of gefitinib in their invasive ability compared to parental PC3 cells but not in their proliferation rate. Moreover, PCb2 cells demonstrated an increased invasive ability in vitro in response to bone stromal cell conditioned medium (BCM). The simultaneous presence of 0.1 ng/ml gefitinib was sufficient to reduce the number of invaded cells in the presence of both EGF and BCM. The molecular characterization of the highly aggressive PCa sub-lines demonstrated that this phenomenon was associated with an increment in uPA/uPAR axis but not in EGF-R expression. In conclusion, our data suggest that the use of gefitinib as a therapeutic agent may be indicated in the control of PCa spreading to bone.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Ósseas/prevenção & controle , Receptores ErbB/antagonistas & inibidores , Neoplasias da Próstata/prevenção & controle , Quinazolinas/uso terapêutico , Animais , Western Blotting , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Receptores ErbB/metabolismo , Gefitinibe , Humanos , Imunoprecipitação , Incidência , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Nus , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...