Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anticancer Res ; 41(8): 3717-3729, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34281830

RESUMO

BACKGROUND/AIM: Interleukin 6 (IL6) is increased in patients with progressive prostate cancer and induces its transdifferentiation to neuroendocrine prostate cancer. Neuroendocrine prostate cancer has become one of the greatest challenges in treating castration-resistant disease and is linked to poor prognosis. It is necessary to understand better the cellular events associated with IL6-mediated neuroendocrine differentiation to prevent it and identify potential new therapeutic targets. MATERIALS AND METHODS: In the present study, an IL6-inducible neuroendocrine differentiation model established specifically for this purpose was applied using LNCaP cells. Proteomics and western blot analyses were used to identify proteins involved in neuroendocrine differentiation. Subsequently, the role of gelsolin (GSN) in the neuroendocrine differentiation model was characterized (knock-down analyses, microscopic co-localization analyses, apoptosis assay) and GSN expression levels in patient material were investigated. RESULTS: This study revealed that GSN is a crucial factor in the neuroendocrine differentiation process. CONCLUSION: It was shown that siRNA-mediated knock-down of GSN can inhibit neuroendocrine differentiation, making it a valid target for preventing IL6-mediated neuroendocrine differentiation.


Assuntos
Gelsolina/metabolismo , Neoplasias da Próstata/patologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Transdiferenciação Celular/efeitos dos fármacos , Transdiferenciação Celular/fisiologia , Gelsolina/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-6/metabolismo , Interleucina-6/farmacologia , Masculino , Células Neuroendócrinas/patologia , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/mortalidade , Neoplasias da Próstata/metabolismo , Canal de Ânion 1 Dependente de Voltagem/metabolismo
2.
Anticancer Res ; 39(10): 5427-5436, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31570437

RESUMO

BACKGROUND/AIM: Renal cell carcinoma (RCC) is one of the most common tumor diseases in adults, and new specific biomarkers are urgently needed to define diagnosis and prognosis of patients with RCC as well as monitor the outcome of therapeutic interventions. The enzyme nicotinamide N-methyltransferase (NNMT) is believed to represent such a marker molecule in RCC therapy. MATERIALS AND METHODS: NNMT expression was examined by western blotting in samples from patients with RCC and in RCC cell lines. Effects of NNMT on cell growth and metabolism were assessed using the Hoechst 33342 reagent assay and Vita-Orange cell viability assay. Incubation experiments were performed to study the influence of methionine and interleukin-6 (IL6) on expression of NNMT. RESULTS: In patient samples, NNMT was up-regulated depending on the stage of progression. Investigations in an RCC cell culture model showed that after modulation of NNMT expression, cellular metabolism, but not cell growth was affected. This regulatory function was also dependent on the presence of the NNMT precursor substrate methionine and IL6. CONCLUSION: The metabolism-regulatory activity of NNMT depends on the precursor substrate methionine and the presence of IL6. The function of methionine appears to be dependent on the stage of progression, since in individual RCC cell lines, opposing effects on metabolism were demonstrated. This, in turn, reflects the thoroughly complex situation in the clinic.


Assuntos
Carcinoma de Células Renais/metabolismo , Neoplasias Renais/metabolismo , Metionina/metabolismo , Nicotinamida N-Metiltransferase/metabolismo , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Células HEK293 , Humanos , Interleucina-6/metabolismo , Prognóstico , Regulação para Cima/fisiologia
3.
World J Urol ; 37(10): 2137-2145, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30603780

RESUMO

PURPOSE: Cabazitaxel, a semi-synthetic taxane of the third generation, inhibits prostate cancer (PC) cell growth by affecting the microtubule architecture. Since cabazitaxel has also been demonstrated to inhibit androgen receptor (AR) functionality, AR and AR-associated heat shock protein (HSP) expressions in the presence of cabazitaxel were characterized. METHODS: AR and HSP expressions were assessed via Western blotting utilizing a PC-cell-line in vitro system incubated with cabazitaxel. RESULTS: Incubation experiments with 0.3 nM cabazitaxel exhibited significantly reduced levels of AR and the AR-associated factors HSP90α, HSP40, and HSP70/HSP90 organising protein. Furthermore, expression of the anti-apoptotic factor HSP60 was suppressed. In contrast to other anticancer compounds, cabazitaxel did not alter the cytoprotective chemoresistance factor HSP27. CONCLUSIONS: Despite the deregulation of microtubule organisation, cabazitaxel has been shown to suppress the expression of HSP. Very notably, and may be as a result of down-regulated HSP, cabazitaxel additionally inhibits the expression of the AR in AR-positive PC cells. Thus, cabazitaxel bears an additional anti-proliferative activity which is at least in part specific for PC cells.


Assuntos
Antagonistas de Receptores de Andrógenos/farmacologia , Antagonistas de Receptores de Andrógenos/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Proteínas de Choque Térmico/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Taxoides/farmacologia , Taxoides/uso terapêutico , Proteínas de Choque Térmico/biossíntese , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/biossíntese , Células Tumorais Cultivadas
4.
Tumour Biol ; 39(5): 1010428317698382, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28466782

RESUMO

Our previous study showed that TPD52 overexpression could increase migration and proliferation of LNCaP cells contributing to the development of prostate cancer. However, mechanism of TPD52 in prostate cancer initiation and progression remains elusive. In this study, we investigated the possible underlying mechanism of TPD52 in prostate cancer progression. In LNCaP cells, TPD52 expression was altered by transfecting with either EGFP-TPD52 or specific short hairpin RNA. Overexpression of TPD52 protected LNCaP cells from apoptosis through elevated anti-apoptotic proteins XIAP, Bcl-2, and Cyclin D1, whereas Bax was downregulated. Mechanistically, we found that TPD52 confers transactivation of nuclear factor-κB, thereby enhancing its target gene expression in LNCaP cells. TPD52 promotes LNCaP cell invasion probably via increased matrix metalloproteinase 9 expression and its activity while tissue inhibitor of metalloproteinase expression is significantly downregulated. Notably, TPD52 might be involved in cell adhesion, promoting tumor metastasis by inducing loss of E-cadherin, expression of vimentin and vascular cell adhesion molecule, and additionally activation of focal adhesion kinase. Furthermore, TPD52 directly interacts with nuclear factor-κB p65 (RelA) and promotes accumulation of phosphorylated nuclear factor-κB (p65)S536 that is directly linked with nuclear factor-κB transactivation. Indeed, depletion of TPD52 or inhibition of nuclear factor-κB in TPD52-positive cells inhibited secretion of tumor-related cytokines and contributes to the activation of STAT3, nuclear factor-κB, and Akt. Interestingly, in TPD52 overexpressing LNCaP cells, nuclear factor-κB inhibition prevented the autocrine/paracrine activation of STAT3. TPD52 activates STAT3 through ascertaining a cross talk between the nuclear factor-κB and the STAT3 signaling systems. Collectively, these results reveal mechanism by which TPD52 is associated with prostate cancer progression and highlight the approach for therapeutic targeting of TPD52 in prostate cancer.


Assuntos
Proteínas de Neoplasias/biossíntese , Neoplasias da Próstata/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição RelA/genética , Apoptose/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteínas de Neoplasias/genética , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/metabolismo , Neoplasias da Próstata/patologia , Isoformas de Proteínas/genética , Proto-Oncogene Mas , RNA Interferente Pequeno/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição RelA/metabolismo , Transfecção
5.
PLoS One ; 11(3): e0150253, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26958841

RESUMO

Prostate cancer (PCa) is one amongst the most common cancersin western men. Incidence rate ofPCa is on the rise worldwide. The present study deals with theserum lipidome profiling of patients diagnosed with PCa to identify potential new biomarkers. We employed ESI-MS/MS and GC-MS for identification of significantly altered lipids in cancer patient's serum compared to controls. Lipidomic data revealed 24 lipids are significantly altered in cancer patinet's serum (n = 18) compared to normal (n = 18) with no history of PCa. By using hierarchical clustering and principal component analysis (PCA) we could clearly separate cancer patients from control group. Correlation and partition analysis along with Formal Concept Analysis (FCA) have identified that PC (39:6) and FA (22:3) could classify samples with higher certainty. Both the lipids, PC (39:6) and FA (22:3) could influence the cataloging of patients with 100% sensitivity (all 18 control samples are classified correctly) and 77.7% specificity (of 18 tumor samples 4 samples are misclassified) with p-value of 1.612×10-6 in Fischer's exact test. Further, we performed GC-MS to denote fatty acids altered in PCa patients and found that alpha-linolenic acid (ALA) levels are altered in PCa. We also performed an in vitro proliferation assay to determine the effect of ALA in survival of classical human PCa cell lines LNCaP and PC3. We hereby report that the altered lipids PC (39:6) and FA (22:3) offer a new set of biomarkers in addition to the existing diagnostic tests that could significantly improve sensitivity and specificity in PCa diagnosis.


Assuntos
Biomarcadores Tumorais/sangue , Lipídeos/sangue , Neoplasias da Próstata/sangue , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem , Idoso , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromatografia Gasosa , Análise por Conglomerados , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Componente Principal , Ácido alfa-Linolênico/farmacologia
6.
Urol Int ; 97(1): 112-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27007943

RESUMO

INTRODUCTION: Inhibition of androgen synthesis by abiraterone acetate (AA) entails enhanced overall survival rates and clinical benefit for patients with locally advanced and metastasized prostate cancer (PC). The expression of heat shock protein 27 (HSP27) is generally associated with cytoprotection and was demonstrated to mediate chemoresistance under cytostatic therapy, for instance, docetaxel treatment. In this study, we investigated the impact of AA treatment on HSP27 expression and PC cell growth. MATERIALS AND METHODS: HSP27 expression levels in docetaxel and AA-treated PC cell lines LNCaP and PC-3 were determined by SDS PAGE and Western blot analysis. Proliferation assays were performed using a CASY Cell Counter and Analyzer Model TT (Roche Applied Science). RESULTS: Despite significantly increased HSP27 expression in PC cells incubated with docetaxel, Western blot analysis implicated a significant reduction of the cytoprotective HSP27 in AA-treated PC cells. Notably, HSP27 stably overexpressed in PC-3-HSP27 cells did not appear as an HSP27-mediated proliferation benefit in the presence of AA as shown in docetaxel incubation studies. CONCLUSION: In contrast to repeatedly demonstrated HSP27-driven chemoresistance related to chemotherapeutics, our results may constitute a broader molecular mode of action of AA chemotherapy. AA efficacy may exert an HSP27 suppressive role that goes beyond the primarily assumed inhibition of androgen biosynthesis.


Assuntos
Acetato de Abiraterona/farmacologia , Inibidores das Enzimas do Citocromo P-450/farmacologia , Proteínas de Choque Térmico HSP27/antagonistas & inibidores , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Choque Térmico HSP27/biossíntese , Humanos , Masculino , Neoplasias da Próstata/metabolismo
7.
Proteomics ; 16(10): 1590-603, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27001414

RESUMO

Monanchocidin A (MonA) is a novel marine alkaloid with promising anti-cancer properties. We recently demonstrated its high efficacy in human urogenital cancers including germ cell tumors. Here, we applied a global proteome screening approach to investigate molecular targets and biological processes affected by MonA in the human cisplatin-resistant germ cell cancer cell line NCCIT-R. Bioinformatical analysis of the proteomics data predicted an effect of MonA on cancer cell migration. Thus, proteins known to be involved in cancer cell migration and invasion were chosen for further validation. The protein alterations identified by proteomics resulted from both, regulation of the total protein expression and post-transcriptional modifications. Among others, regulation of an isoform of vimentin, up-regulation of multiple apolipoprotein E isoforms, and inhibition of hypusination of eukaryotic translation initiation factor 5A-1 were found upon treatment with MonA. Further functional analyses were performed and revealed decreased cell migration and colony formation of cancer cells treated with MonA at non-cytotoxic and non-antiproliferative concentrations. This work provides further insights into the molecular mechanisms behind MonA bioactivity. Furthermore, our research is exemplary for the ability of proteomics to predict drug targets and mode of action of natural anti-cancer agents.


Assuntos
Antineoplásicos/farmacologia , Guanidina/análogos & derivados , Proteoma/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Expressão Gênica/efeitos dos fármacos , Guanidina/farmacologia , Humanos , Proteoma/genética , Proteômica
8.
Tumour Biol ; 37(8): 10435-46, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26846108

RESUMO

The tumour protein D52 isoform 1 (PC-1), a member of the tumour protein D52 (TPD52) protein family, is androgen-regulated and prostate-specific expressed. Previous studies confirmed that PC-1 contributes to malignant progression in prostate cancer with an important role in castration-resistant stage. In the present work, we identified its impact in mechanisms leading to neuroendocrine (NE) transdifferentiation. We established for long-term PC-1 overexpression an inducible expression system derived from the prostate carcinoma cell line LNCaP. We observed that PC-1 overexpression itself initiates characteristics of neuroendocrine cells, but the effect was much more pronounced in the presence of the cytokine interleukin-6 (IL-6). Moreover, to our knowledge, this is the first report that treatment with IL-6 leads to a significant upregulation of PC-1 in LNCaP cells. Other TPD52 isoforms were not affected. Proceeding from this result, we conclude that PC-1 overexpression enhances the IL-6-mediated differentiation of LNCaP cells into a NE-like phenotype, noticeable by morphological changes and increased expression of typical NE markers, like chromogranin A, synaptophysin or beta-3 tubulin. Immunofluorescent staining of IL-6-treated PC-1-overexpressing LNCaP cells indicates a considerable PC-1 accumulation at the end of the long-branched neuron-like cell processes, which are typically formed by NE cells. Additionally, the experimentally initiated NE transdifferentiation correlates with the androgen receptor status, which was upregulated additively. In summary, our data provide evidence for an involvement of PC-1 in NE transdifferentiation, frequently associated with castration resistance, which is a major therapeutic challenge in the treatment of advanced prostate cancer.


Assuntos
Adenocarcinoma/patologia , Antagonistas de Androgênios/uso terapêutico , Androgênios , Antineoplásicos Hormonais/uso terapêutico , Transdiferenciação Celular/fisiologia , Interleucina-6/farmacologia , Proteínas de Neoplasias/fisiologia , Neoplasias Hormônio-Dependentes/patologia , Células Neuroendócrinas/patologia , Neoplasias da Próstata/patologia , Biomarcadores , Linhagem Celular Tumoral , Transdiferenciação Celular/efeitos dos fármacos , Humanos , Interleucina-6/uso terapêutico , Masculino , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/genética , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Células Neuroendócrinas/química , Neoplasias da Próstata/tratamento farmacológico , Domínios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Receptores Androgênicos/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
9.
Prostate ; 76(4): 409-24, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26643154

RESUMO

BACKGROUND: Remodeling of the tumor environment and the modulation of tumor associated non-malignant cells are essential events in tumor progression. Exosomes are small membranous vesicles of 50-150 nm in diameter, which are secreted into the extracellular space and supposedly serve as vehicles for signal and effector molecules to modulate adjacent target cells. We characterized the mRNA and protein composition as well as cellular functions of prostate cancer cell-derived exosomes. METHODS: Exosomes were prepared from prostate cancer cell culture supernatant by ultracentrifugation and subsequently characterized by dynamic light scattering and electron microscopy. Exosomal mRNA and protein composition were analyzed by DNA microarrays and gel electrophoresis coupled with mass spectrometry. Physiological effects of exosomes were studied by means of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase release cell assays. Using a SILAC approach, putative uptake of exosomal human proteins in canine cells and canine de novo synthesis of proteins specified by exosome-transferred human mRNA was analyzed in MDCK cells via mass spectrometry. RESULTS: Preparations of exosomes revealed typical cup shaped particles of 150 nm in diameter. Analysis of mRNA and protein composition of exosomes exhibited a wide range of mRNA and protein species. Interestingly, the packaging of at least small proteins into exosomes was apparently unspecific, as shown with the example of two model proteins. In cell culture incubation experiments exosomal preparations of prostate cancer cells caused anti-proliferative effects. MS analysis revealed the uptake of exosomal human proteins into canine cells after 6 hr of incubation. CONCLUSIONS: The results reveal a distinct exosomal functionality in the modulation of the prostatic tumor adjacent environment. The multitude of translocated factors implies the induction of numerous effects in tumor-associated target cells, including impact on cellular growth.


Assuntos
Exossomos/fisiologia , Neoplasias da Próstata/ultraestrutura , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Animais , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Cães , Difusão Dinâmica da Luz , Exossomos/ultraestrutura , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Masculino , Microscopia Eletrônica de Transmissão , Tamanho da Partícula , Transporte Proteico/fisiologia , Proteínas/análise , Transporte de RNA/fisiologia , RNA Mensageiro/análise , Microambiente Tumoral
10.
Int J Cancer ; 138(10): 2450-65, 2016 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-26695519

RESUMO

Despite recent advances in the treatment of metastatic castration-resistant prostate cancer (CRPC), outcome of patients remains poor due to the development of drug resistance. Thus, new drugs are urgently needed. We investigated efficacy, toxicity and mechanism of action of marine triterpene glycoside frondoside A (FrA) using CRPC cell lines in vitro and in vivo. FrA revealed high efficacy in human prostate cancer cells, while non-malignant cells were less sensitive. Remarkably, proliferation and colony formation of cells resistant to enzalutamide and abiraterone (due to the androgen receptor splice variant AR-V7) were also significantly inhibited by FrA. The marine compound caused cell type specific cell cycle arrest and induction of caspase-dependent or -independent apoptosis. Up-regulation or induction of several pro-apoptotic proteins (Bax, Bad, PTEN), cleavage of PARP and caspase-3 and down-regulation of anti-apoptotic proteins (survivin and Bcl-2) were detected in treated cells. Global proteome analysis revealed regulation of proteins involved in formation of metastases, tumor cell invasion, and apoptosis, like keratin 81, CrkII, IL-1ß and cathepsin B. Inhibition of pro-survival autophagy was observed following FrA exposure. In vivo, FrA inhibited tumor growth of PC-3 and DU145 cells with a notable reduction of lung metastasis, as well as circulating tumor cells in the peripheral blood. Increased lymphocyte counts of treated animals might indicate an immune modulating effect of FrA. In conclusion, our results suggest that FrA is a promising new drug for the treatment of mCRPC. Induction of apoptosis, inhibition of pro-survival autophagy, and immune modulatory effects are suspected modes of actions.


Assuntos
Antineoplásicos/farmacologia , Glicosídeos/farmacologia , Triterpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Reprodutibilidade dos Testes , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Int J Oncol ; 48(2): 793-800, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26648519

RESUMO

Abiraterone provides significant survival advantages in prostate cancer (PC), however, the current understanding of the molecular mechanisms of abiraterone is still limited. Therefore, the abiraterone impact on androgen receptor (AR)-positive LNCaP and AR-negative PC-3 cells was assessed by cellular and molecular analyses. The present study demonstrated, that abiraterone treatment significantly decreased cell growth, AR expression, and AR activity of AR-positive LNCaP cells. Notably, AR-negative PC-3 cells exhibited comparable reductions in cellular proliferation, associated with DNA fragmentation and pro-apoptotic modulation of p21, caspase-3, survivin, and transforming growth factor ß (TGFß). Our observations suggest that the attenuation of AR signaling is not the only rationale to explain the abiraterone anticancer activity. Abiraterone efficacy may play a more global role in PC progression control than originally hypothesized. In this regard, abiraterone is not only a promising drug for treatment of AR-negative PC stages, even more, abiraterone may represent an alternative for treatment of other malignancies besides prostate cancer.


Assuntos
Androstenos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Receptores Androgênicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Masculino , Próstata/efeitos dos fármacos , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Survivina , Fator de Crescimento Transformador beta/metabolismo
12.
PLoS One ; 10(7): e0130350, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26132846

RESUMO

One of the promising possibilities of the clinical application of cold plasma, so-called cold atmospheric plasma (CAP), is its application on malignant cells and cancer tissue using its anti-neoplastic effects, primarily through the delivery of reactive oxygen and nitrogen species (ROS, RNS). In this study, we investigated the impact of CAP on cellular proliferation and consecutive molecular response mechanisms in established prostate cancer (PC) cell lines. PC cells showed a significantly reduced cell growth following CAP treatment as a result of both an immediate increase of intracellular peroxide levels and through the induction of apoptosis indicated by annexin V assay, TUNEL assay, and the evaluation of changes in nuclear morphology. Notably, co-administration of N-acetylcysteine (NAC) completely neutralized CAP effects by NAC uptake and rapid conversion to glutathione (GSH). Vitamin C could not counteract the CAP induced effects on cell growth. In summary, relatively short treatments with CAP of 10 seconds were sufficient to induce a significant inhibition of cancer proliferation, as observed for the first time in urogenital cancer. Therefore, it is important to understand the mode of CAP related cell death and clarify and optimize CAP as cancer therapy. Increased levels of peroxides can alter redox-regulated signaling pathways and can lead to growth arrest and apoptosis. We assume that the general intracellular redox homeostasis, especially the levels of cellular GSH and peroxidases such as peroxiredoxins affect the outcome of the CAP treatment.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Gases em Plasma/farmacologia , Neoplasias da Próstata/metabolismo , Linhagem Celular , Radicais Livres/metabolismo , Humanos , Masculino , Transdução de Sinais
13.
Oncotarget ; 6(19): 17328-41, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-26093146

RESUMO

Monanchocidin A (MonA) is a novel alkaloid recently isolated from the marine sponge Monanchora pulchra. The compound reveals cytotoxic activity in genitourinary cancers including cisplatin-sensitive and -resistant germ cell tumor (GCT) cell lines, hormone-sensitive and castration-resistant prostate carcinoma cell lines and different bladder carcinoma cell lines. In contrast, non-malignant cells were significantly less sensitive. MonA is highly synergistic with cisplatin in GCT cells. Induction of autophagy at lower and lysosomal membrane permeabilization (LMP) at higher concentrations were identified as the dominating modes of action. Cytotoxicity and protein degradation could be inhibited by 3-methyladenine, an inhibitor of autophagy. LMP was confirmed by loss of acridine orange staining of lysosoms and by release of cathepsin B. In conclusion, MonA exerts cytotoxic activity by mechanisms different from "classical" apoptosis, and could be a promising new compound to overcome resistance to standard therapies in genitourinary malignancies.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Guanidina/análogos & derivados , Lisossomos/efeitos dos fármacos , Neoplasias Urogenitais/patologia , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Citometria de Fluxo , Guanidina/farmacologia , Humanos , Membranas Intracelulares/efeitos dos fármacos , Espectrometria de Massas , Microscopia Eletrônica de Transmissão
14.
J Proteomics ; 119: 218-29, 2015 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-25724726

RESUMO

The limitations of the current prostate cancer (PCa) screening tests demands new biomarkers for early diagnosis of PCa. In this study, we aim to investigate serum autoantibody signatures as PCa specific biomarkers. PCa proteins were resolved by 2-DE and then transferred onto polyvinylidene difluoride membrane, which were subsequently incubated with either pooled serum from PCa patients or from normal controls. Mass spectrometry results have identified 18 antigens from 21 different 2-DE spots associated with PCa. Autoantibody response to antigens PRDX2, PRDX6 and ANXA11 in PCa patient's sera was confirmed using recombinant antigens. Further validation with an independent set of PCa patient's sera have shown relatively increased abundance of PRDX6 and ANXA11 antibodies in PCa patients. Formal concept analysis method was applied to assess whether the abundance of these autoantibodies could influence the classification of patients. However, sensitivity of the single antibody to discriminate prostate tumor and healthy controls varies from 70% to 80%, whereas combination of both PRDX6 and ANXA11 antibodies increased sensitivity to 90% for tumors and 100% for healthy controls. Therefore, we hereby report that the detection of these antibodies in PCa patient's serum in combination with the existing non-invasive diagnostic procedures may have significance in PCa diagnosis. BIOLOGICAL SIGNIFICANCE: The present study aimed to investigate serum autoantibody signatures as new biomarkers for early diagnosis of prostate cancer (PCa). To investigate serum autoantibodies in patients with PCa, we used proteomics approach based on two-dimensional gel electrophoresis (2-DE) and mass spectrometry. Total tissue proteins extracted from prostate were separated by 2-DE and then transferred onto polyvinylidene difluoride (PVDF) membrane, which were subsequently incubated with either pooled serum from PCa patients or from normal controls with no history for PCa. Proteomic analysis results have identified 18 antigens that showed antibody response specifically to cancer patient's serum. For validation experiments using recombinant antigens, confirmed autoantibody response to three antigens PRDX2, PRDX6 and ANXA11. Further validation using a second independent set of PCa patient's sera has shown relatively increased abundance of PRDX6 and ANXA11 antibodies specifically in PCa patients. Partition analysis of patients based on abundance of autoantibodies highlighted a combination of both PRDX6 and ANXA11 antibodies in serum with 90% sensitivity in case of tumors and 100% in case of healthy controls. Therefore, we hereby report that the detection of these antibodies in PCa patient's serum in combination with known markers may have significance in diagnosis of PCa with further validation in larger cohort of samples.


Assuntos
Anticorpos Antineoplásicos/sangue , Antígenos de Neoplasias/sangue , Autoanticorpos/sangue , Biomarcadores Tumorais/sangue , Neoplasias da Próstata/sangue , Humanos , Masculino , Neoplasias da Próstata/diagnóstico
15.
World J Urol ; 33(10): 1481-6, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25544376

RESUMO

PURPOSE: The development of a drug-resistant phenotype is the major challenge during treatment of castration-resistant prostate cancer (PC). In solid cancer entities, one of the major contributors to chemoresistance is the multidrug resistance 1 (MDR1) protein. Believed to be involved in the induction of MDR1 expression is the presence of anticancer drugs as well as the Y box binding protein 1 (YB-1). METHODS: Basal as well as drug-induced expression of MDR1 in established PC cell lines was assessed by Western blotting and mass spectrometry. Subsequently, the influence of YB-1 on MDR1 expression was examined via transient overexpression of YB-1. RESULTS: While LNCaP and PC-3 cells showed no detectable amounts of MDR1, the resistance factor was found to be expressed in 22Rv1 cells. Despite this difference, all three cell lines demonstrated similar growth behavior in the presence of the first-line chemotherapeutic agent docetaxel. Incubation of 22Rv1 cells with docetaxel, cabazitaxel, and abiraterone did not significantly alter MDR1 expression levels. Furthermore, overexpression of the MDR1 controlling factor YB-1 showed no impact on MDR1 expression levels. CONCLUSIONS: MDR1 was detectable in the PC cell line 22Rv1. However, this study suggests that MDR1 is of less importance for drug resistance in PC cells than in other types of solid cancer. Furthermore, in contrast to YB-1 properties in other malignancies, MDR1 regulation through YB-1 seems to be unlikely.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Próstata/metabolismo , Neoplasias da Próstata/genética , Proteína 1 de Ligação a Y-Box/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular , Cromatografia Líquida , Docetaxel , Resistência a Múltiplos Medicamentos , Humanos , Masculino , Próstata/patologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Espectrometria de Massas em Tandem , Taxoides , Proteína 1 de Ligação a Y-Box/biossíntese
16.
World J Urol ; 33(3): 323-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24798454

RESUMO

PURPOSE: Androgen receptor (AR) networks are predominantly involved in prostate cancer (PCa) progression; consequently, factors of AR regulation represent promising targets for PCa therapy. The ErbB3-binding protein 1 (Ebp1) is linked to AR suppression and chemoresistance by so far unknown mechanisms. In this study, an assumed regulation of Ebp1 by the newly identified AR controlling signaling axis heat-shock protein 27 (HSP27)-microRNA-1 (miR-1) was examined. METHODS: Transfection experiments were carried out overexpressing and knockdown HSP27 and miR-1, respectively, in LNCaP and PC-3 cells. Afterward, HSP27- and miR-1-triggered Ebp1 protein expression was monitored by Western blotting. RESULTS: AR-positive LNCaP cells and AR-negative PC-3 cells possessed diverse basal expression levels of Ebp1. However, subsequent studies revealed no differences in cellular Ebp1 concentrations after modulation of HSP27 and miR-1. Furthermore, docetaxel incubation experiments exhibited no effects on Ebp1 protein synthesis. CONCLUSION: In PCa, Ebp1 has been described as a regulator of AR functionality and as an effector of PCa therapy resistance. Our data suggest that Ebp1 functionality is independent from heat-shock-protein-regulated progression networks in PCa.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Adenocarcinoma/fisiopatologia , Proteínas de Choque Térmico HSP27/fisiologia , MicroRNAs/fisiologia , Neoplasias da Próstata/fisiopatologia , Proteínas de Ligação a RNA/fisiologia , Receptores Androgênicos/fisiologia , Transdução de Sinais/fisiologia , Adenocarcinoma/patologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Células Cultivadas , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Masculino , Neoplasias da Próstata/patologia , Receptores Androgênicos/efeitos dos fármacos , Transfecção
17.
Oncol Rep ; 32(6): 2380-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25231055

RESUMO

Drug resistance minimizes the effects of prostate cancer (PC) chemotherapy with docetaxel and is generally considered to be associated with the expression of heat shock protein (HSP) 27 including various cytoprotective pathways. In the present study, we investigated the effects of HSP27 phosphorylation on PC cell growth underlying docetaxel treatment. Cell counting revealed significantly reduced cell growth during docetaxel treatment as a result of both activation of mitogen-activated protein kinase p38 (MAPK p38) and protein kinase D1 (PKD1), and, most importantly, the overexpression of the phosphorylation-mimicking mutant HSP27-3D. Further analysis revealed a docetaxel-dependent induction of HSP27 accompanied by an initial phosphorylation and rapid dephosphorylation of the protein. Based on the data, we can conclude that phosphorylation of HSP27 protein is a crucial mechanism in the initiation of chemoresistance in PC. Moreover, the results indicate a key impact of HSP27 on viability and proliferation of PC cells underlying anticancer therapy. The protective function depends on the initial phosphorylation status of HSP27 and represents a putative co-therapeutic target to prevent chemoresistance during docetaxel therapy.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Choque Térmico HSP27/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Taxoides/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Docetaxel , Resistencia a Medicamentos Antineoplásicos , Expressão Gênica , Humanos , Masculino , Fosforilação , Neoplasias da Próstata
18.
Anticancer Res ; 34(7): 3475-80, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24982356

RESUMO

BACKGROUND: Heat-shock proteins (HSPs) as well as microRNAs have been identified to orchestrate crucial mechanisms in prostate cancer (PCa) progression and treatment resistance. Due to cytoprotective properties of HSPB1 we analyzed molecular mechanisms of drug resistance in PCa cell culture systems, and notably found HSPB1 functionality linked to microRNA miR-1 activities. MATERIALS AND METHODS: HSPB1 and miR-1 levels were genetically modified in PCa cell lines and alterations in molecular and cellular responses were assessed by quantitative reverse transcription/polymerase chain reaction, western blotting, and proliferation assays. RESULTS: Our data provided for the first time evidence that HSPB1 regulates miR-1 expression, and subsequently restores oncogenic signaling pathways of androgen receptor (AR) and transforming growth factor ß1 (TGFB1). CONCLUSION: Our data point towards HSPB1 and miR-1 involvement in development of castration-resistant PCa and therefore represent promising targets for anticancer therapy of advanced PCa.


Assuntos
Proteínas de Choque Térmico HSP27/metabolismo , MicroRNAs/biossíntese , Neoplasias de Próstata Resistentes à Castração/metabolismo , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Clonagem Molecular , Progressão da Doença , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico , Humanos , Masculino , MicroRNAs/genética , Chaperonas Moleculares , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo
19.
Dis Model Mech ; 7(8): 963-76, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24832488

RESUMO

The central importance of translational control by post-translational modification has spurred major interest in regulatory pathways that control translation. One such pathway uniquely adds hypusine to eukaryotic initiation factor 5A (eIF5A), and thereby affects protein synthesis and, subsequently, cellular proliferation through an unknown mechanism. Using a novel conditional knockout mouse model and a Caenorhabditis elegans knockout model, we found an evolutionarily conserved role for the DOHH-mediated second step of hypusine synthesis in early embryonic development. At the cellular level, we observed reduced proliferation and induction of senescence in 3T3 Dohh-/- cells as well as reduced capability for malignant transformation. Furthermore, mass spectrometry showed that deletion of DOHH results in an unexpected complete loss of hypusine modification. Our results provide new biological insight into the physiological roles of the second step of the hypusination of eIF5A. Moreover, the conditional mouse model presented here provides a powerful tool for manipulating hypusine modification in a temporal and spatial manner, to analyse both how this unique modification normally functions in vivo as well as how it contributes to different pathological conditions.


Assuntos
Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Desenvolvimento Embrionário , Lisina/análogos & derivados , Oxigenases de Função Mista/antagonistas & inibidores , Células 3T3 , Alelos , Animais , Caenorhabditis elegans , Proliferação de Células , Senescência Celular , Modelos Animais de Doenças , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Técnicas de Inativação de Genes , Hidroxilação , Lisina/metabolismo , Camundongos , Oxigenases de Função Mista/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Fenótipo , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas ras/metabolismo , Fator de Iniciação de Tradução Eucariótico 5A
20.
Integr Biol (Camb) ; 6(5): 540-4, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24658612

RESUMO

Extracellular recording of the glucose-induced electrical activity of mouse islets of Langerhans on microelectrode arrays (MEAs) is an innovative and powerful tool to address beta-cell (patho-)physiology. In a dual approach we tested whether this technique can detect concentration-dependent drug effects as well as characterize alterations in beta-cell activity during prolonged culture. First we established conditions that allow long-term investigation of beta-cell function by recording electrical activity. The results provide the first measurements of beta-cell membrane potential oscillations of individual murine islets during long-term culture. Oscillations were recorded for up to 34 days after islet isolation. Importantly, the glucose dependence of electrical activity did not change over a period of one month. Thus we can follow electrophysiological changes of individual islets induced by alterations in the beta-cell environment over weeks. Second, we used the MEA technique to assay beta-cell damage induced by oxidative stress and to evaluate appropriate protection mechanisms. Oxidative stress plays a key role in the development of type 2 diabetes mellitus (T2DM). Examination of the acute effects of H2O2 on electrical activity showed that the oxidant reduced the electrical activity in a concentration-dependent manner. The superoxide dismutase mimetic, tempol, protected against the detrimental effects of H2O2. In conclusion, we demonstrated that MEA recordings can be used to address disease-related mechanisms and protective interventions in beta-cells. In the future, this fundamental work should enable the monitoring of the electrical activity of islets of Langerhans under controlled ex vivo conditions including long-term exposure to oxidative stress, glucolipotoxicity, and other diabetes-inducing agents.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Glucose/farmacologia , Ilhotas Pancreáticas/metabolismo , Potenciais da Membrana/fisiologia , Estresse Oxidativo/fisiologia , Animais , Antioxidantes/farmacologia , Óxidos N-Cíclicos/farmacologia , Relação Dose-Resposta a Droga , Eletrofisiologia/métodos , Peróxido de Hidrogênio/antagonistas & inibidores , Peróxido de Hidrogênio/metabolismo , Camundongos Endogâmicos C57BL , Microeletrodos , Marcadores de Spin
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...