Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Prostate ; 76(13): 1203-17, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27273171

RESUMO

BACKGROUND: Prostate-specific antigen (PSA) is an important prostate cancer biomarker. It is also a protease expressed at high concentrations by the normal and malignant prostate. PSA is secreted as a zymogen (proPSA) with an inhibitory prodomain that must be removed for full activity. ProPSA variants, assumed to be inactive, are found in the blood of prostate cancer patients, and are indicative of poor clinical outcome. Despite the abundance of clinical reports, our understanding of PSA's enzymology is limited, in part due to a lack of appropriate experimental systems. We sought to develop a series of PSA-derived mutants that would help to enhance our understanding of the gene. METHODS: Sixteen rPSA variants were generated and characterized by a variety of biochemical methods. RESULTS: The wildtype cDNA (WT) provided the template for generating a panel of recombinants. These included variants that abolished removal of the prodomain (R24A), disabled its enzymatic activity (S213A), and/or facilitated a cell-based conversion to the active conformation (FR). The purified variants' proteolytic activity was examined using a fluorogenic substrate, known PSA-cleavable proteins, and physiologically relevant inhibitors. Upon demonstrating our successful generation and purification of the PSA variants, we characterized proPSA activity, describing cleavage of synthetic and biologic substrates, but not serum protease inhibitors. This finding was exploited in the development of a self-activating mutant (PSA_QY) that exhibited the greatest enzymatic activity of all the variants. CONCLUSIONS: The system described herein will prove useful for varied applications. ProPSA is partially functional with relatively high activity compared to the mature enzyme. In demonstrating the zymogen's intrinsic activity, we suggest that the proPSA in prostate cancer patient serum is not inert. This may have implications for our understanding of the disease. Prostate 76:1203-1217, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Variação Genética/fisiologia , Mutação/fisiologia , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Humanos , Masculino , Proteólise , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
2.
J Immunol ; 190(6): 2567-74, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23401592

RESUMO

Prostate-specific Ag (PSA) is a serine protease that is expressed exclusively by normal and malignant prostate epithelial cells. The continued high-level expression of PSA by the majority of men with both high- and low-grade prostate cancer throughout the course of disease progression, even in the androgen-ablated state, suggests that PSA has a role in the pathogenesis of disease. Current experimental and clinical evidence suggests that chronic inflammation, regardless of the cause, may predispose men to prostate cancer. The responsibility of the immune system in immune surveillance and eventually tumor progression is well appreciated but not completely understood. In this study, we used a mass spectrometry-based evaluation of prostatic fluid obtained from diseased prostates after removal by radical prostatectomy to identify potential immunoregulatory proteins. This analysis revealed the presence of Igs and the complement system proteins C3, factor B, and clusterin. Verification of these findings by Western blot confirmed the high-level expression of C3 in the prostatic fluid and the presence of a previously uncharacterized C-terminal C3 cleavage product. Biochemical analysis of this C3 cleavage fragment revealed a putative PSA cleavage site after tyrosine-1348. Purified PSA was able to cleave iC3b and the related complement protein C5. These results suggest a previously uncharacterized function of PSA as an immunoregulatory protease that could help to create an environment hospitable to malignancy through proteolysis of the complement system.


Assuntos
Biomarcadores Tumorais/imunologia , Complemento C3b/metabolismo , Complemento C5/metabolismo , Antígeno Prostático Específico/fisiologia , Próstata/imunologia , Proteólise , Sêmen/imunologia , Serina Proteases/fisiologia , Animais , Líquidos Corporais/enzimologia , Líquidos Corporais/imunologia , Linhagem Celular , Humanos , Masculino , Próstata/metabolismo , Próstata/patologia , Antígeno Prostático Específico/imunologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/imunologia , Sêmen/enzimologia , Ovinos
3.
Prostate ; 72(11): 1233-8, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22213008

RESUMO

BACKGROUND: Prostate-Specific Antigen (PSA) is a serine protease whose expression is maintained in all stages of prostate cancer. A role for PSA in the pathobiology for prostate cancer has not been firmly established. Experimental studies to date support a role for PSA through mechanisms such as release or processing of growth factors and degradation of the extracellular matrix. Exposure of prostate cancer cells to exogenous PSA also results in gene expression changes. These in vitro and biochemical assays rely on the use of commercially available PSA. Contamination of these commercial preparations can significantly impact the results of these in vitro studies. METHODS: We characterized PSA and trypsin-like activity of PSA preparations obtained from three commercial sources: Calbiochem, Fitzgerald, and AbD Serotec. Silver stained gels were used to compare the purity of each preparation and mass spectrometry was performed to characterize contaminating proteases. RESULTS: PSA activity varied between PSA preparations with AbD Serotec PSA having highest degree of activity. Significant trypsin-like activity, which was inhibited by aprotinin, was observed in PSA preparations from Calbiochem and Fitzgerald, but not AbD Serotec. These former two PSA preparations also contained the greatest degree of non-PSA contaminants by silver stain and mass spectrometry. CONCLUSIONS: Commercially available preparations of PSA contain contaminating proteins, including trypsin-like protease activity, that could potentially complicate the interpretation of results obtained from in vitro studies assessing PSA proteolysis of potential protein substrates and effects of PSA on gene expression.


Assuntos
Contaminação de Medicamentos , Antígeno Prostático Específico/análise , Sêmen/química , Tripsina/análise , Humanos , Espectrometria de Massas , Peptídeo Hidrolases/análise
4.
Prostate ; 71(15): 1595-607, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21394741

RESUMO

BACKGROUND: Prostate specific antigen (PSA) is the best-known member of the kallikrein-related peptidase family, with an established role as a prostatic disease biomarker. Although it is produced at high levels by all stages of prostate cancer, it is uncertain if PSA plays a role in prostate cancer initiation and progression. We decided to investigate the impact of PSA and its enzymatic activity on tumor cell growth rates. METHODS: A gene-specific shRNA lentiviral construct reduced endogenous PSA expression in the LNCaP human prostate cancer cell line. Resulting changes in growth rates in vitro and in vivo were determined. Using a mass spectroscopy-based approach, alterations to the LNCaP proteome due to reduced PSA were measured. Finally, to evaluate the importance of PSA's proteolytic activity, the PSA-null Du145 human prostate cancer cell line was engineered to express either enzymatically inactive pro-PSA (WT) or a furin-activated variant (FR) with high enzymatic activity. The resulting clones were evaluated for PSA-induced changes in growth rates in vivo and in vitro. RESULTS: Lowered PSA levels dramatically reduced LNCaP growth rates. Expressing active PSA (FR), but not the inactive WT variant, conferred a growth advantage on Du145 cells. Proteomics analysis revealed global changes to the LNCaP proteome as a result of reduced PSA expression. CONCLUSIONS: These studies demonstrate the importance of PSA to prostate cancer cell growth. We also show that the enzymatic activity of PSA confers an enhanced growth rate to human prostate cancer cells, suggesting a causal role in prostate cancer progression.


Assuntos
Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/enzimologia , Animais , Bioensaio , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Clonagem Molecular , Humanos , Imuno-Histoquímica , Masculino , Espectrometria de Massas , Camundongos , Camundongos Nus , Antígeno Prostático Específico/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteômica/métodos , RNA Interferente Pequeno/farmacologia
5.
Prostate ; 70(7): 788-96, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20058238

RESUMO

BACKGROUND: Prostate-specific antigen (PSA) is a serine protease secreted as a zymogen. Previously, cell-free biochemical studies have identified various kallikreins (KLK) as candidate activating proteases. In this study, KLK2-mediated activation of PSA in cell-based in vitro, xenograft, and transgenic models was evaluated. METHODS: Du145-derived PSA- or KLK2-expressing clones were coincubated in vitro and in vivo to evaluate KLK2-induced PSA activity. While mice possess orthologs of KLK4-15, they do not have functional orthologs of PSA or KLK2. Therefore, transgenic animals expressing PSA or both PSA and KLK2 were generated to assess orthotopic PSA activation. RESULTS: PSA is activated by KLK2 when the cells are physically in contact, and through co-conditioned media. In vivo, the free (inactive PSA) to total (active + inactive PSA) ratio in the blood is decreased when PSA and KLK2-expressing cells are co-inoculated subcutaneously, suggesting increased active PSA. Additionally, double-transgenic mice expressing both genes in the prostate produce more active PSA compared to single transgenic animals. A longitudinal evaluation over a 2-year period demonstrated no morphologic changes (i.e., no PIN or prostate cancer) due to PSA or PSA/KLK2 double transgene expression relative to non-transgenic mice. CONCLUSIONS: These data demonstrate, with biologically relevant models, that KLK2 is the protease responsible for activating PSA. While PSA is involved in the processing and release of a number of important growth factors, our results suggest that active PSA is not sufficient to induce the development of prostate cancer or prostate cancer precursors in aging PSA transgenic mice.


Assuntos
Antígeno Prostático Específico/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Calicreínas Teciduais/metabolismo , Animais , Western Blotting , Células Cultivadas , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Transplante de Neoplasias , Próstata/citologia , Antígeno Prostático Específico/genética , Neoplasias da Próstata/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Calicreínas Teciduais/genética
6.
Luminescence ; 24(1): 35-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18780328

RESUMO

Bioluminescence has gained favour in the last decade as an approach for observing tumours in vivo in a non-destructive manner. This very sensitive technique is based on light emission by the reaction of luciferin with the enzyme luciferase, as measured by a photodetector. Ever since the development of recombinant tumour cell lines that have been engineered to produce luciferase, a vast number of experiments have been carried out examining tumour growth, tumour metastasis and the effect of therapeutic regimens in such cases. A primary stumbling block, however, is the relatively short circulatory half-life of luciferin. In this paper, we propose the PEGylation of 6-amino-D-luciferin to extend its in vivo circulatory half-life, thus making the possibility of long-term observations in animals possible. The covalent attachment was through a carbamate linker that is known to hydrolyse in vivo, releasing the parent compound. Based on our studies, longer emission of the PEGylated luciferin was observed, as compared to free luciferin in mice bearing PC3 prostate tumours expressing luciferase. This result suggests that this reagent can be used in applications requiring extended monitoring of luciferase activation in vivo.


Assuntos
Vaga-Lumes/enzimologia , Luciferases/metabolismo , Polietilenoglicóis/química , Tiazóis/análise , Tiazóis/química , Animais , Linhagem Celular Tumoral , Vaga-Lumes/genética , Humanos , Luciferases/genética , Camundongos , Camundongos Nus , Estrutura Molecular , Transplante de Neoplasias , Tiazóis/metabolismo
7.
Proteins ; 70(4): 1416-28, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17894328

RESUMO

Prostate Specific Antigen (PSA) is a biomarker used in the diagnosis of prostate cancer and to monitor therapeutic response. However, its precise role in prostate carcinogenesis and metastasis remains largely unknown. A number of studies arguing in the favor of an active role of PSA in prostate cancer development and progression have implicated this serine protease in the release and activation of growth factors such as insulin-like growth factor 1 (IGF1) through cleavage of insulin like growth factor binding protein 3 and Transforming Growth Factor beta (TGF-beta) through cleavage of Latent TGF-beta. In contrast, other studies suggest that PSA activity might hinder tumor development and progression. In light of these contradictory findings, efficient inhibitors of PSA are needed for exploring its biological role in tumor development and metastasis. Towards the goal of developing potent inhibitors of PSA, we have explored the molecular mechanism of a series of beta-lactam based compounds on binding to PSA using activity assays, matrix assisted laser desorption ionization with a time-of-flight mass spectrometry, and GOLD docking methodology. The mass spectrometry experiments and the activity assays confirmed the time-dependent and covalent nature of beta-lactam binding. To gain insights on the reaction intermediates at the molecular level, we docked beta-lactam inhibitors to a homology modeled PSA using the GOLD docking program in noncovalent and covalent binding modes. The docking studies elucidated the molecular details of the early noncovalent Michaelis complex, the acyl-enzyme covalent complex, and the nature of conformational reorganization required for the long term stability of the covalent complex. Additionally, the molecular basis for the effect of stereochemistry of the lactam ring on the inhibitory potency was elucidated through docking of beta-lactam enantiomers. As a validation of our docking methodology, two novel enantiomers were synthesized and evaluated for their inhibitory potency using fluorogenic substrate based activity assays. Additionally, cis enantiomers of eight beta-lactam compounds reported in a previous study were docked and their GOLD scores and binding modes were analyzed in order to assess the general applicability of our docking results. The close agreement of our docking results with the experimental data validates the mechanistic insights revealed through the docking studies and paves the way for the design and development of potent and specific inhibitors of PSA.


Assuntos
Antígeno Prostático Específico/antagonistas & inibidores , beta-Lactamas , Simulação por Computador , Desenho de Fármacos , Humanos , Masculino , Métodos , Modelos Moleculares , Ligação Proteica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
9.
Cancer Res ; 65(11): 4902-8, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15930312

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the tumor necrosis factor family of cytokines that induces apoptosis in some tumor cells but not in normal cells. Unfortunately, many human cancer cell lines are refractory to TRAIL-induced cell death, and the molecular mechanisms underlying resistance are unclear. Here we report that TRAIL resistance was reversed in human bladder and prostate cancer cell lines by the proteasome inhibitor bortezomib (PS-341, Velcade). Synergistic induction of apoptosis occurred within 4 to 6 hours in cells treated with TRAIL plus bortezomib and was associated with accumulation of p21(WAF-1/Cip-1) (p21) and inhibition of cyclin-dependent kinase (cdk) activity. Roscovitine, a specific cdk1/2 inhibitor, also sensitized cells to TRAIL. Silencing p21 expression reduced levels of DNA fragmentation by 50% in cells treated with bortezomib and TRAIL, confirming that p21 was required for the response. Analysis of the TRAIL pathway revealed that caspase-8 processing was enhanced in a p21-dependent fashion in cells exposed to TRAIL and bortezomib as compared with cells treated with TRAIL alone. Thus, all downstream components of the pathway (Bid cleavage, cytochrome c release, and caspase-3 activation) were amplified. These data strongly suggest that p21-mediated cdk inhibition promotes TRAIL sensitivity via caspase-8 activation and that TRAIL and bortezomib should be combined in appropriate in vivo models as a possible approach to solid tumor therapy.


Assuntos
Ácidos Borônicos/farmacologia , Proteínas de Ciclo Celular/fisiologia , Glicoproteínas de Membrana/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Pirazinas/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Bortezomib , Caspase 3 , Caspase 8 , Caspases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21 , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Humanos , Masculino , Neoplasias da Próstata/patologia , Ligante Indutor de Apoptose Relacionado a TNF , Neoplasias da Bexiga Urinária/patologia
10.
Mol Cancer Ther ; 5(8): 2043-50, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16928825

RESUMO

Bortezomib (PS-341, Velcade) is a peptide boronate inhibitor of the 20S proteasome that is currently being combined with taxanes in several clinical trials in patients with prostate cancer. Here, we report that bortezomib inhibited docetaxel-induced M-phase arrest and apoptosis in androgen-dependent LNCaP-Pro5 cells. Direct analysis of kinase activity in immune complex kinase assays revealed that docetaxel activated cyclin-dependent kinase (CDK) 1 (CDC2) and that bortezomib blocked this activation. The effects of bortezomib were associated with accumulation of p21 and mimicked by chemical CDK inhibitors or by transfecting cells with a small interfering RNA construct specific for CDK1. Transient transfection with p21 also inhibited docetaxel-induced apoptosis; conversely, p21 silencing reversed the antagonistic effects of bortezomib on docetaxel-induced apoptosis. Together, our data show that bortezomib interferes with docetaxel-induced apoptosis via a p21-dependent mechanism that is associated with CDK1 inhibition. These observations may have important implications for the ongoing bortezomib-docetaxel combination trials as well as trials using bortezomib and other cell cycle-sensitive agents.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Pirazinas/farmacologia , Taxoides/farmacologia , Apoptose/fisiologia , Bortezomib , Proteína Quinase CDC2/efeitos dos fármacos , Proteína Quinase CDC2/metabolismo , Divisão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/genética , Docetaxel , Humanos , Masculino , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas
11.
Br J Psychol ; 108(1): 43-72, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26856707

RESUMO

Inhibitory control (IC), an ability to suppress irrelevant and/or conflicting information, has been found to underlie performance on a variety of cognitive tasks, including bilingual language processing. This study examines the relationship between IC and the speech patterns of second language (L2) users from the perspective of individual differences. While the majority of studies have supported the role of IC in bilingual language processing using single-word production paradigms, this work looks at inhibitory processes in the context of extended speech, with a particular emphasis on disfluencies. We hypothesized that the speech of individuals with poorer IC would be characterized by reduced fluency. A series of regression analyses, in which we controlled for age and L2 proficiency, revealed that IC (in terms of accuracy on the Stroop task) could reliably predict the occurrence of reformulations and the frequency and duration of silent pauses in L2 speech. No statistically significant relationship was found between IC and other L2 spoken output measures, such as repetitions, filled pauses, and performance errors. Conclusions focus on IC as one out of a number of cognitive functions in the service of spoken language production. A more qualitative approach towards the question of whether L2 speakers rely on IC is advocated.


Assuntos
Aprendizagem , Multilinguismo , Fala , Adulto , Feminino , Humanos , Individualidade , Testes de Linguagem , Masculino , Pessoa de Meia-Idade , Análise de Regressão , Adulto Jovem
12.
Cancer Res ; 63(21): 7338-44, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14612532

RESUMO

Advanced prostate cancer is resistant to current therapeutic strategies. Bortezomib (Velcade; previously called PS-341) is a potent and specific inhibitor of the 26S proteasome that is currently in clinical trials for treatment of various malignancies, including prostate cancer. We investigated the effects of bortezomib on p53 in the LNCaP-Pro5 prostate cancer cell line. Bortezomib induced strong stabilization of p53, but it did not promote phosphorylation on serines 15 and 20, and p53 remained bound to its inhibitor, mdm2. Nonetheless, bortezomib stimulated p53 translocation to the nucleus (not mitochondria) and enhanced p53 DNA binding, accumulation of p53-dependent transcripts, and activation of a p53-responsive reporter gene. Furthermore, stable LNCaP-Pro5 transfectants of LNCaP-Pro5 expressing the p53 inhibitor human papillomavirus-E6 displayed reduced bortezomib-induced p53 activation and cell death. Together, our data demonstrate that bortezomib stimulates p53 activation via a novel mechanism.


Assuntos
Ácidos Borônicos/farmacologia , Proteínas Nucleares , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Complexo de Endopeptidases do Proteassoma , Pirazinas/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Bortezomib , Linhagem Celular Tumoral , DNA de Neoplasias/metabolismo , Etoposídeo/farmacologia , Humanos , Masculino , Proteínas Oncogênicas Virais/genética , Papillomaviridae/genética , Peptídeo Hidrolases/metabolismo , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2 , Frações Subcelulares/metabolismo , Ativação Transcricional , Transfecção , Proteína Supressora de Tumor p53/genética
13.
Cancer Prev Res (Phila) ; 5(2): 229-39, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22139053

RESUMO

Insufficient dose of dietary methyl groups are associated with a host of conditions ranging from neural tube defects to cancer. On the other hand, it is not certain what effect excess dietary methyl groups could have on cancer. This is especially true for prostate cancer, a disease that is characterized by increasing DNA methylation changes with increasing grade of the cancer. In this three-part study in animals, we look at (i) the effect of excess methyl donors on the growth rate of prostate cancer in vivo, (ii) the ability of 5-aza-2'-deoxycytidine (AdC), a demethylating agent, to demethylate in the presence of excess dietary methyl donors, and (iii) the effect of in utero feeding of excess methyl donors to the later onset of prostate cancer. The results show that when mice are fed a dietary excess of methyl donors, we do not see (i) an increase in the growth rate of DU-145 and PC-3 xenografts in vivo, or (ii) interference in the ability of AdC to demethylate the promoters of androgen receptor or Reprimo of prostate cancer xenografts but (iii) a protective effect on the development of higher grades of prostate cancer in the "Hi-myc" mouse model of prostate cancer which were fed the increased methyl donors in utero. We conclude that the impact of dietary methyl donors on prostate cancer progression depends upon the timing of exposure to the dietary agents. When fed before the onset of cancer, that is, in utero, excess methyl donors can have a protective effect on the progression of cancer.


Assuntos
Azacitidina/análogos & derivados , Transformação Celular Neoplásica , Metilação de DNA , Dieta , Neoplasias da Próstata/patologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/farmacologia , Colina/administração & dosagem , Decitabina , Progressão da Doença , Ácido Fólico/administração & dosagem , Homocisteína/administração & dosagem , Humanos , Masculino , Metionina/administração & dosagem , Camundongos , Camundongos Nus , Neoplasias da Próstata/etiologia , Células Tumorais Cultivadas , Vitamina B 6/administração & dosagem
14.
Prostate ; 67(3): 312-29, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17143882

RESUMO

Prostate cancer cells, like normal prostate epithelial cells, produce high levels of the differentiation marker and serine protease prostate-specific antigen (PSA). PSA is used extensively as a biomarker to screen for prostate cancer, to detect recurrence following local therapies, and to follow response to systemic therapies for metastatic disease. While much is known about PSA's role as a biomarker, only a relatively few studies address the role played by PSA in the pathobiology of prostate cancer. Autopsy studies have documented that not only do prostate cancer cells maintain production of high amounts of PSA but they also maintain the enzymatic machinery required to process PSA to an enzymatically active form. A variety studies performed over the last 10 years have hinted at a role for PSA in growth, progression, and metastases of prostate cancer. A fuller understanding of PSA's functional role in prostate cancer biology, however, has been hampered by the lack of appropriate models and tools. Therefore, the purpose of this review is not to address issues related to PSA as a biomarker. Instead, by reviewing what is known about the genetics, biochemistry, and biology of PSA in normal and malignant prostate tissue, insights may be gained into the role PSA may be playing in the pathobiology of prostate cancer that can connect measurement of this biomarker to an understanding of the underlying etiology and progression of the disease.


Assuntos
Neoplasias Hormônio-Dependentes/enzimologia , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Adulto , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Ativação Enzimática , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Neoplasias Hormônio-Dependentes/patologia , Antígeno Prostático Específico/sangue
15.
Bioorg Med Chem ; 15(14): 4973-84, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17502149

RESUMO

Four novel water-soluble peptide-paclitaxel conjugates were designed and synthesized as prostate-specific antigen (PSA)-activated prodrugs for prostate cancer therapy. These prodrugs were composed of a peptide, HSSKLQ or SSKYQ, each of which is selectively cleavable by PSA; a self-immolative linker, either para-aminobenzyl alcohol (PABS) or ethylene diamine (EDA); and the parent drug, paclitaxel. Introduction of a PABA or EDA linker between the peptide and paclitaxel in prodrugs 2-5 resulted in products with an increased rate of hydrolysis by PSA. The stability of prodrugs 2 and 3, with the PABA linker, was poor in the serum-containing medium because of the weak carbonate bond between the PABA and paclitaxel; however, this disadvantage was overcome by introducing a carbamate bond using an EDA linker in prodrugs 4 and 5. Thus, the incorporation of an EDA linker increased both the stability and PSA-mediated activation of these prodrugs. The cytotoxicity of each prodrug, as compared to paclitaxel, was determined against a variety of cell lines, including the PSA-secreting CWR22Rv1 prostate cancer cell line. The EDA-derived prodrug of paclitaxel 5 was stable and capable of being efficiently converted to an active drug that killed cells specifically in the presence of PSA, suggesting that this prodrug and similarly designed PSA-cleavable prodrugs may have potential as prostate cancer-specific therapeutic agents.


Assuntos
Paclitaxel/química , Paclitaxel/farmacologia , Neoplasias da Próstata/patologia , Soluções Tampão , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Meios de Cultura Livres de Soro , Humanos , Masculino , Estrutura Molecular , Paclitaxel/síntese química , Pró-Fármacos/síntese química , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Soluções , Relação Estrutura-Atividade
16.
J Natl Cancer Inst ; 99(5): 376-85, 2007 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-17341729

RESUMO

BACKGROUND: Most men will develop prostatic abnormalities, such as benign prostatic hyperplasia (BPH) or prostate cancer, as they age. Prostate-specific antigen (PSA) is a serine protease that is secreted at high levels by the normal and diseased prostate. Therapies that are activated by PSA may prove effective in treating prostatic malignancies. METHODS: We modified proaerolysin (PA), the inactive precursor of a bacterial cytolytic pore-forming protein, to produce a PSA-activated protoxin (PRX302). The viability of the prostate adenocarcinoma cell lines LNCaP, PC-3, CWR22H, and DU145 and the bladder cancer cell line TSU after treatment with PA or PRX302 in the presence or absence of purified PSA was assayed. Mice carrying xenograft tumors derived from LNCaP, CWR22H, or TSU cells were treated with intratumoral injection of PA or PRX302, and tumor size was monitored. To test the safety of PRX302, we administered it into the PSA-secreting prostate glands of cynomolgus monkeys. All statistical tests were two-sided. RESULTS: Native PA was highly toxic in vitro but had no tumor-specific effects in vitro or in vivo. Picomolar concentrations of PRX302 led to PSA-dependent decreases in cell viability in vitro (PRX302 versus PRX302 + PSA: DU145 cells, mean viability = 78.7% versus mean = 1.6%, difference = 77.1%, 95% confidence interval [CI] = 70.6% to 86.1%; P<.001; TSU cells, mean = 100.2% versus mean = 1.4%, difference = 98.8%, 95% CI = 96.4% to 104.0%; P<.001). Single intratumoral injections of PRX302 produced substantial and often complete regression of PSA-secreting human prostate cancer xenografts (5 microg dose, complete regression in 6 of 26 mice bearing LNCap or CWR22H xenografts [23%]; 10 microg dose, complete regression in 10 of 26 mice [38.5%]) but not PSA-null bladder cancer xenografts. The prostates of cynomolgus monkeys injected with a single dose of PRX302 displayed extensive but organ-confined damage, with no toxicity to neighboring organs or general morbidity. CONCLUSIONS: Our observations demonstrate the potential safe and effective intraprostatic application of this engineered protoxin.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/farmacologia , Toxinas Bacterianas/farmacologia , Proteínas Citotóxicas Formadoras de Poros/farmacologia , Antígeno Prostático Específico/metabolismo , Hiperplasia Prostática/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Linhagem Celular Tumoral , Humanos , Imuno-Histoquímica , Injeções Intralesionais , Macaca fascicularis , Masculino , Camundongos , Proteínas Citotóxicas Formadoras de Poros/uso terapêutico , Pró-Fármacos/farmacologia , Projetos de Pesquisa , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Anticancer Drugs ; 18(7): 809-16, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17581303

RESUMO

Native proaerolysin is a channel-forming bacterial protoxin that binds to cell-surface receptors and then is activated by furin or furin-like proteases. We genetically engineered proaerolysin by replacing the furin-cleavage sequence with a prostate-specific antigen-selective sequence. The recombinant modified proaerolysin was expressed and purified from Aeromonas salmonicida in good yields and purity. Recombinant modified proaerolysin had no furin sensitivity and markedly increased prostate-specific antigen sensitivity relative to wild-type proaerolysin. Human prostate cancer cells were significantly more sensitive to recombinant modified proaerolysin in the presence of active prostate-specific antigen when compared with the absence of prostate-specific antigen or the presence of potent prostate-specific antigen inhibitors. Most normal human cells with the exception of prostate and renal epithelial cells showed very low sensitivity to recombinant modified proaerolysin. Our results suggest that recombinant modified proaerolysin is a potent prostate-specific antigen-sensitive protoxin that deserves further development for regional therapy of benign and malignant prostate growths.


Assuntos
Antineoplásicos/farmacologia , Toxinas Bacterianas/farmacologia , Proteínas Citotóxicas Formadoras de Poros/farmacologia , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Aeromonas salmonicida/química , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/química , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Eletroforese em Gel de Poliacrilamida , Engenharia Genética , Humanos , Masculino , Peptídeo Hidrolases/metabolismo , Proteínas Citotóxicas Formadoras de Poros/administração & dosagem , Proteínas Citotóxicas Formadoras de Poros/química , Pró-Fármacos
18.
Prostate ; 66(9): 903-10, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16496413

RESUMO

BACKGROUND: Intraprostatic PSMA targeted prodrugs/protoxins are under development in our laboratory. Future toxicologic studies of these therapies require identification of animal models that express PSMA within the prostate. METHOD: PSMA enzymatic activity and protein expression was determined. PSMA expression in the prostates of mouse, dog, and monkey were compared to humans by real-time PCR analysis. RESULTS: No substrate hydrolysis was observed in dog or monkey prostate homogenates. Monkey prostate was negative for PSMA protein expression. No significant PSMA mRNA levels were detected by real time PCR in mouse, dog, or monkey prostate tissue compared to PSMA negative tissues. CONCLUSIONS: PSMA is not expressed in any significant amount in the prostates of mouse, beagle dog, or macaque monkeys in this study but is expressed in high levels by human prostate. These non-human species, therefore, are not suitable toxicologic models to assess prostate damage from PSMA-activated intraprostatic prodrug/protoxin therapies.


Assuntos
Antígenos de Superfície/análise , Antígenos de Superfície/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Glutamato Carboxipeptidase II/análise , Glutamato Carboxipeptidase II/genética , Próstata/química , Animais , Western Blotting , Modelos Animais de Doenças , Cães , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Imuno-Histoquímica , Macaca mulatta , Masculino , Camundongos , RNA Mensageiro/análise , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testes de Toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA