Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
J Pharmacokinet Pharmacodyn ; 49(2): 243-256, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34604941

ABSTRACT

The current approach to selection of a population PK/PD model is inherently flawed as it fails to account for interactions between structural, covariate, and statistical parameters. Further, the current approach requires significant manual and redundant model modifications that heavily lend themselves to automation. Within the discipline of numerical optimization it falls into the "local search" category. Genetic algorithms are a class of algorithms inspired by the mathematics of evolution. GAs are general, powerful, robust algorithms and can be used to find global optimal solutions for difficult problems even in the presence of non-differentiable functions, as is the case in the discrete nature of including/excluding model components in search of the best performing mixed-effects PK/PD model. A genetic algorithm implemented in an R-based NONMEM workbench for identification of near optimal models is presented. In addition to the GA capabilities, the workbench supports modeling efforts by: (1) Organizing and displaying models in tabular format, allowing the user to sort, filter, edit, create, and delete models seamlessly, (2) displaying run results, parameter estimates and precisions, (3) integrating xpose4 and PsN to facilitate generation of model diagnostic plots and run PsN scripts, (4) running regression models between post-hoc parameter estimates and covariates. This approach will further facilitate the scientist to shift efforts to focus on model evaluation, hypotheses generation, and interpretation and applications of resulting models.


Subject(s)
Algorithms , Pharmacokinetics , Computer Simulation , Models, Biological
2.
Proteomics ; 16(17): 2419-31, 2016 09.
Article in English | MEDLINE | ID: mdl-27357730

ABSTRACT

Prostate cancer is one of the leading cancers in men. Taking dietary supplements, such as fish oil (FO), which is rich in n-3 polyunsaturated fatty acids (PUFAs), has been employed as a strategy to lower prostate cancer risk and control disease progression. In this study, we investigated the global phosphoproteomic changes induced by FO using a combination of phosphoprotein-enrichment strategy and high-resolution tandem mass spectrometry. We found that FO induces many more phosphorylation changes than oleic acid when they both are compared to control group. Quantitative comparison between untreated group and FO- or oleic acid-treated groups uncovered a number of important protein phosphorylation changes induced by n-3PUFAs. This phosphoproteomic discovery study and the follow-up Western Blot validation study elucidate that phosphorylation levels of the two regulatory serine residues in pyruvate dehydrogenase alpha 1 (PDHA1), serine-232 and serine-300, are significantly decreased upon FO treatment. As expected, increased pyruvate dehydrogenase activity was also observed. This study suggests that FO-induced phosphorylation changes in PDHA1 is more likely related to the glucose metabolism pathway, and n-3 PUFAs may have a role in controlling the balance between lipid and glucose oxidation.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , Fish Oils/therapeutic use , Phosphoproteins/metabolism , Prostatic Neoplasms/diet therapy , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Dietary Supplements/analysis , Humans , Male , Oleic Acids/therapeutic use , Phosphopeptides/analysis , Phosphopeptides/metabolism , Phosphoproteins/analysis , Phosphorylation , Prostate/metabolism , Prostatic Neoplasms/metabolism , Proteome/analysis , Proteome/metabolism , Pyruvate Dehydrogenase (Lipoamide)/chemistry , Tandem Mass Spectrometry
3.
Proteomics Clin Appl ; 10(3): 267-79, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26929070

ABSTRACT

PURPOSE: The aim of this study is to investigate the role of n-3 and n-9 fatty acids in crucial processes involved in prostate cancer cell growth through a large-scale proteomic analysis. EXPERIMENTAL DESIGN: We used a label-free protein quantification method to profile global protein expression of fish oil and oleic acid treated PCa cells and validated a panel of differentially expressed proteins by either Western blot or multiple reaction monitoring. Bioinformatic analysis was also performed to uncover the pathways involved in fatty acid metabolism. RESULTS: Fish oil, not oleic acid, suppresses prostate cancer cell viability. Assessment of fatty acid synthesis pathway activity also shows that oleic acid is a more potent inhibitor than fish oil on de novo fatty acid synthesis. Although fatty acid synthase activity decreases with fish oil treatment, the inhibition of its activity occurs over time while reduction in viability occurs within 24 h. Bioinformatic analysis revealed the pathways altered by these fatty acid treatments. CONCLUSIONS AND CLINICAL RELEVANCE: This study suggests that suppression of cell viability by fish oil is independent of fatty acid synthase and fish oil regulates prostate cancer cells through activation of other pathways depending upon length of exposure to fish oil.


Subject(s)
Dietary Supplements , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Computational Biology , Humans , Male , Prostatic Neoplasms/pathology , Structure-Activity Relationship
4.
J Med Chem ; 58(2): 778-84, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25513712

ABSTRACT

Fatty acid synthase (FASN), the enzyme responsible for de novo synthesis of free fatty acids, is up-regulated in many cancers. FASN is essential for cancer cell survival and contributes to drug resistance and poor prognosis. However, it is not expressed in most nonlipogenic normal tissues. Thus, FASN is a desirable target for drug discovery. Although different FASN inhibitors have been identified, none has successfully moved into clinical use. In this study, using in silico screening of an FDA-approved drug database, we identified proton pump inhibitors (PPIs) as effective inhibitors of the thioesterase activity of human FASN. Further investigation showed that PPIs inhibited proliferation and induced apoptosis of cancer cells. Supplementation of palmitate, the end product of FASN catalysis, rescued cancer cells from PPI-induced cell death. These findings provide new evidence for the mechanism by which this FDA-approved class of compounds may be acting on cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Repositioning , Fatty Acid Synthases/antagonists & inhibitors , Palmitoyl-CoA Hydrolase/antagonists & inhibitors , Proton Pump Inhibitors/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Hydrogen-Ion Concentration , Lansoprazole/pharmacology , Palmitic Acid/pharmacology , Protein Structure, Tertiary
5.
Endocrinology ; 151(12): 5721-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20962047

ABSTRACT

The androgen-androgen receptor signaling pathway plays an important role in the pathogenesis of prostate cancer. Accordingly, androgen deprivation has been the most effective endocrine therapy for hormone-dependent prostate cancer. Here, we report a novel pregnane X receptor (PXR)-mediated and metabolism-based mechanism to reduce androgenic tone. PXR is a nuclear receptor previously known as a xenobiotic receptor regulating the expression of drug metabolizing enzymes and transporters. We showed that genetic (using a PXR transgene) or pharmacological (using a PXR agonist) activation of PXR lowered androgenic activity and inhibited androgen-dependent prostate regeneration in castrated male mice that received daily injections of testosterone propionate by inducing the expression of cytochrome P450 (CYP)3As and hydroxysteroid sulfotransferase (SULT)2A1, which are enzymes important for the metabolic deactivation of androgens. In human prostate cancer cells, treatment with the PXR agonist rifampicin (RIF) inhibited androgen-dependent proliferation of LAPC-4 cells but had little effect on the growth of the androgen-independent isogenic LA99 cells. Down-regulation of PXR or SULT2A1 in LAPC-4 cells by short hairpin RNA or small interfering RNA abolished the RIF effect, indicating that the inhibitory effect of RIF on androgens was PXR and SULT2A1 dependent. In summary, we have uncovered a novel function of PXR in androgen homeostasis. PXR may represent a novel therapeutic target to lower androgen activity and may aid in the treatment and prevention of hormone-dependent prostate cancer.


Subject(s)
Receptors, Steroid/metabolism , Testosterone/blood , Animals , Cell Line, Tumor , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Gene Expression , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orchiectomy , Pregnane X Receptor , Pregnenolone Carbonitrile/pharmacology , Prostate/physiology , Prostatic Neoplasms , Receptors, Androgen/metabolism , Receptors, Steroid/agonists , Receptors, Steroid/antagonists & inhibitors , Receptors, Steroid/genetics , Regeneration , Sulfotransferases/genetics , Sulfotransferases/metabolism , Testosterone/administration & dosage , Testosterone/metabolism , Testosterone/pharmacology , Testosterone Propionate/pharmacology
6.
Clin Cancer Res ; 15(19): 6177-84, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19789311

ABSTRACT

PURPOSE: The transmembrane molecule, translocator protein (TSPO), has been implicated in the progression of epithelial tumors. TSPO gene expression is high in tissues involved in steroid biosynthesis, neurodegenerative disease, and in cancer, and overexpression has been shown to contribute to pathologic conditions including cancer progression in several different models. The goal of our study was to examine the expression and biological relevance of TSPO in prostate cancer and show that the commonly prescribed benzodiazepine lorazepam, a ligand for TSPO, exhibits anticancer properties. EXPERIMENTAL DESIGN: Immunohistochemical analysis using tissue microarrays was used to determine the expression profile of TSPO in human prostate cancer tissues. To show the effect of TSPO ligands (lorazepam and PK11195) in prostate cancer, we used cell proliferation assays, apoptosis ELISA, prostate cancer xenograft study, and immunohistochemistry. RESULTS: TSPO expression is increased in prostatic intraepithelial neoplasia, primary prostate cancer, and metastases compared with normal prostate tissue and benign prostatic hyperplasia. Furthermore, TSPO expression correlates with disease progression, as TSPO levels increased with increasing Gleason sum and stage with prostate cancer metastases demonstrating the highest level of expression among all tissues examined. Functionally, we have shown that lorazepam has antiproliferative and proapoptotic properties in vitro and in vivo. Additionally, we have shown that TSPO overexpression in nontumorigenic cells conferred susceptibility to lorazepam-induced growth inhibition. CONCLUSION: These data suggest that blocking TSPO function in tumor cells induces cell death and denotes a survival role for TSPO in prostate cancer and provides the first evidence for the use of benzodiazepines in prostate cancer therapeutics.


Subject(s)
Carcinoma/pathology , Cell Proliferation/drug effects , Isoquinolines/pharmacology , Lorazepam/pharmacology , Prostatic Neoplasms/pathology , Receptors, GABA , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Carcinoma/metabolism , Cell Death/drug effects , Cells, Cultured , Down-Regulation/drug effects , HeLa Cells , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , Receptors, GABA/genetics , Receptors, GABA/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Urol Oncol ; 26(4): 378-85, 2008.
Article in English | MEDLINE | ID: mdl-18367102

ABSTRACT

(Z)-1-1-Dichloro-2,3-diphenylcyclopropane (A(II)) and (Z)-1,1-dichloro-2-(4-methoxyphenyl)-3-phenylcyclopropane [2-(4-methoxyphenyl)-A(II)] inhibit tubulin polymerization, PSA production, and the proliferation of human prostate cancer cells. The actions of the agents were studied in three transgenic adenocarcinomas of the mouse prostate (TRAMP) cell lines. Antiproliferative potencies were determined and cells treated with the more potent 2-(4-methoxyphenyl)-A(II) were examined for induction of apoptosis. Microarray analyses were conducted to determine the apoptosis-related genes up- and down-regulated by the agent. 2-(4-Methoxyphenyl)-A(II) concentration-dependently inhibited growth of all three cell lines. Fifty percent and 100% growth inhibitory and 50% lethal concentrations were determined to be 0.3, 1.5, and 5 muM, respectively. Minimum detectable apoptosis-inducing concentrations by ELISA were 0.10 to 0.14 muM. PARP cleavage and two-color flow cytometry assays verified apoptosis induction. Microarray analyses showed Bok and Siva-pending to be up-regulated and that Birc, Dad1, and Atf5 were down-regulated. 2-(4-methoxyphenyl)-A(II) inhibits proliferation and induces apoptosis in the in vivo-adaptable TRAMP cells, suggesting the compound should be further examined in preclinical models.


Subject(s)
Anisoles/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cyclopropanes/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Male , Mice , Oligonucleotide Array Sequence Analysis , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/pathology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
8.
Urology ; 71(2): 341-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18308116

ABSTRACT

OBJECTIVES: We investigated the effects of suppression of endothelin-A (ET(A)) receptors on bladder function and ET-1 levels in the bladder in rats with chronic spinal cord injury (SCI). METHODS: We transected the spinal cord of female Sprague-Dawley rats at the level of Th 8-9. Awake cystometrograms were performed 4 weeks after spinal cord transection. We evaluated cystometric parameters such as mean amplitudes of nonvoiding contractions (NVCs), the number of NVCs, voided volume, voiding efficiency, and micturition pressure before and after intravenous (i.v.) injection of ABT-627, an ET(A) antagonist, or A-19261, an ET(B) antagonist, in SCI animals. Four weeks after spinalization, we also measured the protein and mRNA levels of ET-1 in the bladder using enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS: ABT-627 (1 mg/kg, i.v.) but not A-192621 (10 mg/kg, i.v.) significantly decreased the amplitude of NVCs and the number of NVCs in SCI rats. There were no significant changes in pressure threshold, maximum voiding pressure, voided volume, or voiding efficiency. ELISA analysis for ET-1 showed significantly elevated protein concentrations in SCI rats compared with spinal cord intact rats. Significant upregulation of the ET-1 mRNA was also noted in SCI bladders. CONCLUSIONS: These results suggest that upregulation of ET-1 is involved in the mechanism inducing bladder overactivity in chronic SCI rats, and that an ET(A) receptor antagonist can suppress SCI-induced bladder overactivity as indicated by a reduction in NVCs. Thus, ET(A) receptor inhibition could be an effective treatment for neurogenic bladder overactivity in pathological conditions such as SCI.


Subject(s)
Endothelin A Receptor Antagonists , Pyrrolidines/therapeutic use , Spinal Cord Injuries/complications , Urinary Bladder, Overactive/drug therapy , Urinary Bladder, Overactive/etiology , Animals , Atrasentan , Chronic Disease , Female , Rats , Rats, Sprague-Dawley
9.
Prostate ; 68(7): 698-714, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18302219

ABSTRACT

BACKGROUND: Following androgen ablation therapy, the majority of prostate cancer patients develop treatment resistance with a median time of 18-24 months to disease progression. METHODS: To identify molecular targets that promote prostate cancer cell survival and contribute to androgen independence, we evaluated changes in LNCaP cell gene expression during 12 months of androgen deprivation. At time points reflecting critical growth and phenotypic changes, we performed Affymetrix expression array analysis to examine the effects of androgen deprivation during the acute response, during the period of apparent quiescence, and following the emergence of a highly proliferative, androgen-independent prostate cancer cell phenotype (LNCaP-AI). RESULTS: We discovered alterations in gene expression for molecules associated with promoting prostate cancer cell growth and survival, and regulating cell cycle progression and apoptosis. Additionally, expression of AR co-regulators, adrenal androgen metabolizing enzymes, and markers of neuroendocrine disease were significantly altered. CONCLUSIONS: These findings contribute greatly to our understanding of androgen-independent prostate cancer. The value of this longitudinal approach lies in the ability to examine gene expression changes throughout the adaptive response to androgen deprivation; it provides a more dynamic illustration of genes which contribute to disease progression in addition to specific genes which constitute an androgen-independent phenotype.


Subject(s)
Androgen Antagonists/pharmacology , Androgens/physiology , Antineoplastic Agents, Hormonal/pharmacology , Gene Expression Regulation, Neoplastic/physiology , Prostatic Neoplasms/genetics , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Endothelin-1/genetics , Endothelin-1/metabolism , Gene Expression Profiling , Humans , Male , Prostatic Neoplasms/metabolism , Receptor, Endothelin A/genetics , Receptor, Endothelin A/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
10.
Urology ; 70(1): 209-15, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17656249

ABSTRACT

OBJECTIVES: Emerging evidence supports a role for endothelin-1 (ET-1), endothelin A and B receptors (ET(A) and ET(B), respectively), and neutral endopeptidase (NEP) in the progression of prostate carcinoma. In clinical trials for advanced prostate cancer, ET axis blockade significantly delayed the time to disease progression in a subset of patients. We examined ET axis expression in prostate cancer, prostatic intraepithelial neoplasia, and normal adjacent tissue and then analyzed the relationship of the protein levels with disease progression. METHODS: The expression levels of ET(A), ET(B), and NEP were determined in 120 prostate cancer specimens obtained at surgery or biopsy by immunohistochemistry. In situ hybridization on a subset of the specimens was used to confirm the immunohistochemistry findings. RESULTS: In regions of adenocarcinoma, immunohistochemistry analysis demonstrated high ET(A) expression in 72% of the specimens. ET(A) expression was significantly elevated with increased pathologic stage and grade. ET(B) and NEP levels were significantly decreased in adenocarcinoma compared with normal adjacent tissue and prostatic intraepithelial neoplasia; however, reduced expression did not correlate with tumor grade or stage. Patients with prostate-specific antigen recurrence had significantly greater ET(A) levels in their primary tumors than did patients who were disease free 5 years after prostatectomy. Patients with high ET(A) expression in the adenocarcinoma regions with low ET(B) and NEP had a significantly decreased interval to prostate-specific antigen progression compared with patients with low ET(A) or high ET(B)/NEP expression. CONCLUSIONS: These data suggest two patterns of ET(A) expression in primary prostate cancer, with increased expression correlating with more advanced disease. The use of these expression patterns to identify patients more likely to respond to ET axis blockade might enhance treatment outcomes.


Subject(s)
Endothelin-1/biosynthesis , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/metabolism , Receptor, Endothelin A/biosynthesis , Receptor, Endothelin B/biosynthesis , Aged , Aged, 80 and over , Endothelin-1/analysis , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/chemistry , Neoplasm Recurrence, Local/metabolism , Prostatic Intraepithelial Neoplasia/chemistry , Prostatic Intraepithelial Neoplasia/surgery , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/surgery , Receptor, Endothelin A/analysis , Receptor, Endothelin B/analysis
11.
Cancer Lett ; 246(1-2): 139-48, 2007 Feb 08.
Article in English | MEDLINE | ID: mdl-16581180

ABSTRACT

Endothelin-1 (ET-1) is a potent vasoconstrictor that has been shown to significantly impact many benign and malignant tissues by signaling through its two cognate receptors: ET(A) and ET(B). As ET-1 has a role in both normal and diseased kidney, we initiated studies to investigate endothelin axis expression and function in renal cell carcinoma (RCC). In this study, relatively high levels of ET-1 were detected in all six human RCC cell lines investigated. RT-PCR and Southern analyses revealed that all six RCC cell lines expressed ET(A) receptor mRNA, while 3/6 cell lines also expressed ET(B) mRNA. High affinity ET-1 binding occurred in all but one RCC cell line and quantitative RT-PCR demonstrated ET(A) mRNA expression in all six cell lines. Methylation of the ET(B) promoter (EDNRB) in 4/6 RCC cell lines was observed, suggesting a mechanism for repressed ET(B) expression. Moreover, methylation occurred in 32/48 of renal tumors and in 27/55 of histologically normal adjacent tissue samples studied, while no methylation was evident in any normal tissue isolated from nephrectomy or at autopsy. Functionally, ET-1 significantly inhibited paclitaxel-induced apoptosis in RCC cells through binding ET(A) with the ET-1 signaling mediated via the PI3-kinase/Akt pathway. Collectively, these data support the therapeutic targeting of the ET(A) receptor as a novel treatment strategy for RCC.


Subject(s)
Endothelin-1/metabolism , Gene Expression Regulation, Neoplastic , Receptor, Endothelin A/genetics , Androstadienes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , Chromones/pharmacology , DNA Methylation , Endothelin A Receptor Antagonists , Endothelin-1/pharmacology , Endothelin-1/physiology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Morpholines/pharmacology , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Endothelin A/agonists , Receptor, Endothelin B/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Wortmannin
12.
Neoplasia ; 8(9): 725-32, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16984730

ABSTRACT

Endothelin (ET) 1 is important in the growth of prostate cancer cells through the activation of the endothelin A (ET(A)) receptor. ET receptor blockade is a new therapeutic target in treating advanced prostate cancer. This study investigates the impact of the combination of the ET(A) antagonist atrasentan (ABT-627) and taxane chemotherapy on prostate cancer cell survival in vitro and on the delay of prostate cancer in a xenograft mouse model. In vitro, PPC-1 cells transfected with an ET(A)-overexpressing vector were treated with ABT-627, paclitaxel/docetaxel, or both. Clonogenic viability and cell death assays were used to determine cell survival and apoptosis, respectively. ABT-627 and docetaxel combination treatment was used in vivo to treat mice with established ET(A)-overexpressing PPC-1 xenograft tumors, and tumor growth rates were assessed. Cell proliferation and vascularity were determined with Ki-67 and CD31 staining, respectively. Cells treated with combination therapy had significantly fewer viable cells and more programmed cell death than cells given monotherapy. Xenograft tumor growth rates were significantly lower in mice treated with combination therapy than in animals given a single agent. Ki-67 immunostaining demonstrated significantly fewer proliferative cells following combination therapy than following monotherapy. This study demonstrates ABT-627 to have additive antitumor effects when used in combination with taxane drugs both in vitro and in vivo.


Subject(s)
Bridged-Ring Compounds/pharmacology , Endothelin A Receptor Antagonists , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Taxoids/pharmacology , Animals , Apoptosis , Atrasentan , Cell Survival , Humans , Ki-67 Antigen/biosynthesis , Male , Mice , Neoplasm Transplantation , Neovascularization, Pathologic , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Pyrrolidines/pharmacology
13.
Hum Pathol ; 37(4): 401-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16564913

ABSTRACT

Cancer cells frequently exhibit a significant increase in overexpression and activity of fatty acid synthase (FASN). Elevated FASN pathway activity also occurs in prostate cancer, the second leading cause of cancer-related death in men in the United States. Studies show that genes associated with an increase in protein expression, such as HER2/neu in breast cancer, are associated with an increase in gene copy number as well as an increase in transcription. In the present study, we evaluated whether FASN follows a similar paradigm in prostate cancer. To date, elevated FASN expression in prostate cancer has not been correlated with gene copy number alterations. Using immunohistochemistry and fluorescence in situ hybridization analysis in paraffin-embedded tissue microarrays, we observed gene copy gain in 24% of all prostate adenocarcinoma specimens examined with concurrent increased FASN protein expression. Immunohistochemistry alone showed 59% of prostate cancer specimens in the same tissue microarray with high FASN expression. Increased FASN gene was observed in 53% of all prostate tissues expressing elevated FASN protein levels and in 2 of 5 prostate tumor cell lines tested. These findings suggest that FASN gene copy number increases may be involved in the resultant increase in FASN protein expression observed in prostatic disease.


Subject(s)
Adenocarcinoma/genetics , Fatty Acid Synthases/genetics , Gene Dosage , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Adenocarcinoma/enzymology , Adenocarcinoma/secondary , DNA, Neoplasm/analysis , Fatty Acid Synthases/metabolism , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Neoplasm Staging , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Tissue Array Analysis
14.
Neoplasia ; 7(7): 631-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16026642

ABSTRACT

Endothelin-1 (ET-1), produced by the prostate epithelia, likely plays an important role in the progression of prostate cancer. ET-1 can bind two receptor subtypes; generally, binding of the endothelin receptor A (ET(A)) induces a survival pathway, whereas binding of the endothelin receptor B (ET(B)) mediates clearance of circulating ET-1 as well as promotes apoptosis. In prostate carcinoma, hypermethylation of the ET(B) promoter results in repression of ET(B) expression, thereby eliminating the negative growth response that ET-1 binding elicits through this receptor. Therefore, activation of ET(A) exclusively provides a pathway for survival advantage. Our current studies examine the mechanisms by which activation of the ET(A) may allow growth/survival. ET-1 treatment of prostate tumor cells significantly decreased paclitaxel-induced apoptosis through activation of the ET(A) subtype. The anti-apoptotic effects of ET-1 are mediated, at least in part, through the Bcl-2 family. Although no significant changes in Bcl-2 expression occurred with ET-1 treatment, the pro-apoptotic family members Bad, Bax, and Bak all decreased significantly. Further analysis of the survival pathway demonstrated that phosphorylation of Akt occurs with ET-1 treatment in a time- and dose-dependent manner through phosphatidyinositol 3-kinase activation. These data support the combination of ET(A) antagonists and apoptosis-inducing therapies for prostate cancer treatment.


Subject(s)
Apoptosis , Endothelin-1/metabolism , Endothelin-1/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Annexin A5/pharmacology , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Survival , DNA Methylation , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Immunoblotting , Male , Membrane Proteins/metabolism , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Endothelin/metabolism , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein , bcl-Associated Death Protein
15.
Prostate ; 65(1): 27-34, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-15800935

ABSTRACT

BACKGROUND: Mitogenic and anti-apoptotic actions of endothelin-1 (ET-1) are mediated through endothelin A (ET(A)) receptors. We investigated endothelin receptor expression in increasingly aggressive phenotype and in vivo effects of combination therapy using ET(A) antagonist with paclitaxel. METHODS: Dunning prostate cancer cells ranged in aggressiveness from non-tumorigenic G, to tumorigenic, non-metastatic AT-1, and to tumorigenic and metastatic MLL. Binding assays were performed alongside Q-PCR to assess receptor density. MLL xenografts were treated with vehicle, atrasentan, paclitaxel, and paclitaxel+atrasentan. RESULTS: Saturation binding assays demonstrated endothelin receptor density of MLL and AT-1 cells seven- and threefold higher than G cells, respectively. Q-PCR showed 9- and 4.5-fold greater ET(A) mRNA expression in MLL and AT-1 than G cells, respectively and no endothelin receptor B (ET(B)) expression. Combination therapy had significant effect on reduction of tumor volume than paclitaxel or atrasentan alone. CONCLUSIONS: ET(A) expression increases in aggressive prostate carcinoma. ET(A) blockade combined with paclitaxel may reduce tumor growth in advanced prostate carcinoma.


Subject(s)
Adenocarcinoma/pathology , Endothelin-1/physiology , Prostatic Neoplasms/pathology , Animals , Disease Progression , Endothelin A Receptor Antagonists , Humans , Male , Mice , Phenotype , Prostatic Neoplasms/drug therapy , Receptor, Endothelin A/analysis , Receptor, Endothelin B/analysis
16.
J Urol ; 172(4 Pt 1): 1533-7, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15371886

ABSTRACT

PURPOSE: We investigated the effects of endothelin (ET) receptor activation in the bladder and the spinal cord on the micturition reflex in urethane anesthetized rats. MATERIALS AND METHODS: The effects of ET receptor activation on bladder activity were examined during continuous infusion cystometrograms. ET-1 was administered intrathecally or intravesically in normal rats or rats pretreated with capsaicin. The effects of intravenous injection of the selective ETA receptor antagonist ABT-627, or selective ETB receptor antagonist A-192621 (Abbott Laboratories, Abbott Park, Illinois) intrathecal injection of the opioid receptor antagonist naloxone hydrochloride on changes in bladder activity induced by intravesical or intrathecal ET-1 administration were investigated. RESULTS: Intravesical injection of ET-1 (0.1 to 10 microM) induced detrusor overactivity, as evidenced by a decrease in intercontraction intervals, in a dose dependent manner. ET-1 induced detrusor overactivity was suppressed by intravenous application of ABT-627 (0.1 mg/kg) as well as capsaicin pretreatment but not by A-192621. In contrast, intrathecal injection of ET-1 (0.5 to 50 fmol) increased intercontraction intervals dose dependently and ET-1 at a higher dose (50 fmol) induced urinary retention. These inhibitory effects were antagonized by ABT-627 (10 mg/kg) and also by intrathecal application of naloxone but not by A-192621. CONCLUSIONS: These results indicate that the activation of ETA receptors in capsaicin sensitive C-fiber afferents in the bladder can induce detrusor overactivity, while ETA receptor activation in the spinal cord can inhibit the micturition reflex via activation of a spinal opioid mechanism. Thus, targeting peripheral ETA receptors could be an effective treatment for bladder overactivity and/or painful conditions.


Subject(s)
Peripheral Nerves/physiology , Receptors, Endothelin/physiology , Reflex/physiology , Spinal Cord/physiology , Urinary Bladder/innervation , Urination/physiology , Afferent Pathways/drug effects , Afferent Pathways/physiopathology , Animals , Atrasentan , Capsaicin/toxicity , Dose-Response Relationship, Drug , Endothelin A Receptor Antagonists , Endothelin B Receptor Antagonists , Endothelin-1/pharmacology , Female , Injections, Spinal , Muscle Hypertonia/chemically induced , Muscle Hypertonia/physiopathology , Naloxone/pharmacology , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Pyrrolidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid/drug effects , Receptors, Opioid/physiology , Urodynamics/drug effects , Urodynamics/physiology
17.
Prostate ; 60(3): 175-7, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15176046

ABSTRACT

BACKGROUND: Some men treated with atrasentan (ABT-627), an endothelin A (ETA) receptor inhibitor, had declines in their serum PSA levels. It is our hypothesis that this decrease is due to anti-tumoral activity and not a reduction in PSA secretion at the cellular level. METHODS: Two PSA secreting prostate cancer cell lines (LAPC4 and LNCaP) were treated with atrasentan and an ETB receptor antagonist (A192621) in varying concentrations (10(-6)-10(-10) M) and PSA levels were measured in the culture media. RESULTS: LNCaP and LAPC4 cells both express ETA receptors. Neither the ETA or ETB antagonist altered PSA secretion, while addition of DHT, a positive control, produced a marked increase in PSA secretion. CONCLUSIONS: Blockade of the ETA receptor does not affect the secretion of PSA in prostate cancer cell lines.


Subject(s)
Endothelin A Receptor Antagonists , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/pathology , Receptor, Endothelin A/physiology , Atrasentan , Endothelin-1/antagonists & inhibitors , Humans , Male , Pyrrolidines/pharmacology , Tumor Cells, Cultured
18.
Blood ; 104(7): 2107-15, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15213100

ABSTRACT

The biologic effects of endothelin-1 (ET-1) are not limited to its potent vasoconstricting activity. The endothelin receptors, ETA and ETB, have differential tissue and functional distributions. Here we showed that dendritic cells (DCs), the major antigen-presenting cells in the adaptive limb of the immune system, produce large amounts of ET-1 and significantly increase the expression of endothelin receptors upon maturation. Selective blockade of the ETA receptor significantly reduced expression of the mature DC marker CD83, decreased the production of the immunostimulatory cytokine interleukin-12, down-regulated DC ability to stimulate T cells, and promoted DC apoptosis. Selective ETB receptor blockade, on the other hand, resulted in increased expression of CD83 and improved DC survival. Therefore, ET-1/ETA/ETB autocrine/paracrine loops on DCs appear to be essential for the normal maturation and function of human DCs, presenting a unique target for immunomodulatory therapies.


Subject(s)
Dendritic Cells/cytology , Endothelin-1/physiology , Immunotherapy/methods , Receptors, Endothelin/physiology , Annexin A5/pharmacology , Antigens, CD , Apoptosis , Cell Death , Cell Division , Cell Survival , Dendritic Cells/metabolism , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulins/biosynthesis , Immunoglobulins/chemistry , Immunohistochemistry , Interleukin-12/biosynthesis , Interleukin-12/metabolism , Leukocytes/cytology , Leukocytes, Mononuclear/immunology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/chemistry , Oligonucleotide Array Sequence Analysis , Pyrrolidines/pharmacology , Receptor, Endothelin B/metabolism , Receptors, Endothelin/chemistry , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , CD83 Antigen
19.
Prostate ; 57(3): 245-54, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14518031

ABSTRACT

BACKGROUND: Fatty acid synthase (FAS) is the major enzyme required to convert carbohydrates to fatty acids. Recent evidence suggests that FAS activity is essential for prostate cancer growth and survival, since blocking the enzyme activity results in cell death. In this study, the role of FAS up-regulation during prostate tumor progression in the transgenic adenocarcinoma of mouse prostate (TRAMP) model was investigated. Sensitivity to FAS anti-metabolites was also analyzed in TRAMP prostate tumor cells and tissue to determine therapeutic potential of FAS inhibition in the treatment of prostate cancer. METHODS: FAS expression was evaluated by immunohistochemistry of TRAMP tissues, including primary and metastatic lesions in mice of varying ages. FAS pathway activity was studied in vitro using TRAMP-derived cell lines and in vivo in TRAMP tissues. The sensitivity of TRAMP cell lines and tissues to the antimetabolite drugs (2R,3S)-2,3-epoxy-4-oxo-7,10-trans, transdodecadienamide (cerulenin) and C-75, which target FAS, was determined by FAS antimetabolite inhibition of 14C-acetate conversion to fatty acids, cell growth inhibition, and apoptosis analyses. RESULTS: High FAS expression and activity in the TRAMP mouse prostate was evident at 12 weeks of age compared with nontransgenic littermates and further increased with age, tumor progression, and in metastatic lesions. FAS pathway inhibition resulted in a dose-dependent reduction in cell survival and decreased enzyme activity in these models. CONCLUSIONS: These data suggest that the up-regulation of FAS expression play a role in tumorigenesis of the prostate in the TRAMP model and hence can provide valuable insight into human prostate cancer. Given the response of tumor cells to FAS antimetabolites, FAS may serve as a novel target for prostate cancer therapy.


Subject(s)
Adenocarcinoma/enzymology , Fatty Acid Synthases/biosynthesis , Prostatic Neoplasms/enzymology , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Blotting, Western , Cerulenin/pharmacology , Disease Models, Animal , Disease Progression , Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostatic Neoplasms/pathology
20.
Prostate ; 52(2): 123-9, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12111703

ABSTRACT

BACKGROUND: Prostate cancer frequently progresses from an initial androgen dependence to androgen independence, rendering the only effective androgen ablation therapy useless. The mechanism underlying the androgen-independent progression is unknown. Stat3, a member of the family of signal transducers and activators of transcription, is activated in numerous cancers, including prostate. This study is to investigate the role of Stat3 activation in the growth of prostate cancer cells. METHODS: A constitutively active Stat3 was ectopically expressed in androgen-sensitive LNCaP prostate cancer cells and resulting stable clones expressing activated Stat3 were isolated. The effect of Stat3 activation on LNCaP cell growth in response to androgen in vitro and in vivo was examined. RESULTS: We show that the levels of activated Stat3 are associated with the progression of androgen-independent prostate cancer. Activation of Stat3 in androgen-sensitive LNCaP prostate cancer cells results in enhancement of tumor growth in both intact and castrated male nude mice and enhances androgen receptor-mediated prostate specific antigen expression. CONCLUSIONS: These findings demonstrate that intracellular signaling mediated by Stat3 can enhance the growth of androgen-sensitive human LNCaP prostate cancer cells in both intact and castrated male nude mice.


Subject(s)
DNA-Binding Proteins/physiology , Orchiectomy , Prostatic Neoplasms/pathology , Trans-Activators/physiology , Androgens/metabolism , Animals , Cell Division , DNA-Binding Proteins/metabolism , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , STAT3 Transcription Factor , Trans-Activators/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...