Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Hum Pathol ; 63: 98-109, 2017 05.
Article En | MEDLINE | ID: mdl-28300578

Phospho-Akt (P-Akt1) promotes proliferation and increased survival in vitro and plays an important role in prostate cancer (PCa) progression as well as the prediction of the probability of recurrence. In this study, the goal was to demonstrate the involvement and impact of P-Akt1 on cellular interactions, biomechanisms, and pathways in PCa. Tissue microarrays from 640 PCa patients were immunostained with various antibodies. Ki-67 was used to measure proliferation index, and terminal deoxynucleotidyl transferase-mediated dUTP biotin nick end labeling was used for apoptotic index. Increased expression of P-Akt1 was associated with an increased proliferation but inversely correlated with apoptotic index. Higher levels of P-Akt1 are associated with both higher levels of cytoplasmic p27 and higher levels of nuclear p27, suggesting an involvement in both cytoplasmic entrapment and phosphorylation of p27. P-Akt1 expression significantly correlated with nuclear and cytoplasmic staining of FHKR and GSK. The strongest correlations were found with the P- forms of both, suggesting enzyme kinetics in the latter. Here, phosphorylation is the principal method of FHKR and GSK inactivation. P-Akt1 correlated with nuclear transcription factor kappa B, suggesting a role in the inhibition through phosphorylation of nuclear transcription factor kappa B. The results of the current study are unique because of the scope of the markers and the size of the population used. In vitro- and in vivo-derived information of P-Akt1 and its downstream effectors demonstrates significant involvement in PCa. Our data suggest that PCa uses multiple mechanisms to regulate this pathway and substantiate the concept of redundancy in cancer pathway regulation. Consequently, new hypothesis-driven studies can be derived from this information.


Biomarkers, Tumor/analysis , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/analysis , Signal Transduction , Adult , Aged , Aged, 80 and over , Apoptosis , Biopsy , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/analysis , Forkhead Box Protein O1/analysis , Glycogen Synthase Kinases/analysis , Humans , Immunohistochemistry , Male , Middle Aged , NF-kappa B/analysis , Neoplasm Invasiveness , Phosphorylation , Prostatectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Tissue Array Analysis
2.
Clin Cancer Res ; 22(15): 3937-49, 2016 08 01.
Article En | MEDLINE | ID: mdl-26968201

PURPOSE: Castration therapy in advanced prostate cancer eventually fails and leads to the development of castration-resistant prostate cancer (CRPC), which has no cure. Characteristic features of CRPC can be increased androgen receptor (AR) expression and altered transcriptional output. We investigated the expression of nuclear receptor corepressor 1 (NCOR1) in human prostate and prostate cancer and the role of NCOR1 in response to antiandrogens. EXPERIMENTAL DESIGN: NCOR1 protein levels were compared between matched normal prostate and prostate cancer in 409 patient samples. NCOR1 knockdown was used to investigate its effect on bicalutamide response in androgen-dependent prostate cancer cell lines and transcriptional changes associated with the loss of NCOR1. NCOR1 transcriptional signature was also examined in prostate cancer gene expression datasets. RESULTS: NCOR1 protein was detected in cytoplasm and nuclei of secretory epithelial cells in normal prostate. Both cytoplasmic and nuclear NCOR1 protein levels were lower in prostate cancer than in normal prostate. Prostate cancer metastases show significant decrease in NCOR1 transcriptional output. Inhibition of LNCaP cellular proliferation by bicalutamide requires NCOR1. NCOR1-regulated genes suppress cellular proliferation and mediate bicalutamide resistance. In the mouse, NCOR1 is required for bicalutamide-dependent regulation of a subset of the AR target genes. CONCLUSIONS: In summary, we demonstrated that NCOR1 function declines with prostate cancer progression. Reduction in NCOR1 levels causes bicalutamide resistance in LNCaP cells and compromises response to bicalutamide in mouse prostate in vivo Clin Cancer Res; 22(15); 3937-49. ©2016 AACR.


Gene Expression , Nuclear Receptor Co-Repressor 1/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Androgens/metabolism , Androgens/pharmacology , Anilides/pharmacology , Anilides/therapeutic use , Animals , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Gene Knockdown Techniques , Gene Silencing , Humans , Male , Mice , Mice, Transgenic , Neoplasm Metastasis , Nitriles/pharmacology , Nitriles/therapeutic use , Nuclear Receptor Co-Repressor 1/metabolism , Prostatic Neoplasms/therapy , RNA Interference , Tosyl Compounds/pharmacology , Tosyl Compounds/therapeutic use , Transcriptome
3.
J Pathol ; 231(1): 77-87, 2013 Sep.
Article En | MEDLINE | ID: mdl-23729330

Levels of caveolin-1 (Cav-1) in tumour epithelial cells increase during prostate cancer progression. Conversely, Cav-1 expression in the stroma can decline in advanced and metastatic prostate cancer. In a large cohort of 724 prostate cancers, we observed significantly decreased levels of stromal Cav-1 in concordance with increased Gleason score (p = 0.012). Importantly, reduced expression of Cav-1 in the stroma correlated with reduced relapse-free survival (p = 0.009), suggesting a role for stromal Cav-1 in inhibiting advanced disease. Silencing of Cav-1 by shRNA in WPMY-1 prostate fibroblasts resulted in up-regulation of Akt phosphorylation, and significantly altered expression of genes involved in angiogenesis, invasion, and metastasis, including a > 2.5-fold increase in TGF-ß1 and γ-synuclein (SNCG) gene expression. Moreover, silencing of Cav-1 induced migration of prostate cancer cells when stromal cells were used as attractants. Pharmacological inhibition of Akt caused down-regulation of TGF-ß1 and SNCG, suggesting that loss of Cav-1 in the stroma can influence Akt-mediated signalling in the tumour microenvironment. Cav-1-depleted stromal cells exhibited increased levels of intracellular cholesterol, a precursor for androgen biosynthesis, steroidogenic enzymes, and testosterone. These findings suggest that loss of Cav-1 in the tumour microenvironment contributes to the metastatic behaviour of tumour cells by a mechanism that involves up-regulation of TGF-ß1 and SNCG through Akt activation. They also suggest that intracrine production of androgens, a process relevant to castration resistance, may occur in the stroma.


Adenocarcinoma/mortality , Caveolin 1/metabolism , Prostatic Neoplasms/mortality , Stromal Cells/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Biomarkers/metabolism , Disease Progression , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Male , Neoplasm Recurrence, Local , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , Survival Rate , Transfection , Transforming Growth Factor beta1/metabolism , Tumor Cells, Cultured , Tumor Microenvironment , gamma-Synuclein/metabolism
4.
Clin Cancer Res ; 17(13): 4355-66, 2011 Jul 01.
Article En | MEDLINE | ID: mdl-21622724

PURPOSE: Increased expression of FGFR-4 and its ligands have been linked to lethal prostate cancer (PCa). Furthermore, a germ line polymorphism in the FGFR-4 gene, resulting in arginine at codon 388 (Arg³88) instead of glycine (Gly³88), is associated with aggressive disease. The FGFR-4 Arg³88 variant results in increased receptor stability, sustained receptor activation, and increased motility and invasion compared with Gly³88. However, the impact of sustained signaling on cellular signal transduction pathways is unknown. EXPERIMENTAL DESIGN: Expression microarray analysis of immortalized prostatic epithelial cells lines expressing FGFR-4 Arg³88 or Gly³88 was used to establish a gene signature associated with FGFR-4 Arg³88 expression. Transient transfection of reporters and inhibitors was used to establish the pathways activated by FGFR-4 Arg³88 expression. The impact of pathway knockdown in vitro and in an orthotopic model was assessed using inhibitors and/or short hairpin RNA (shRNA). RESULTS: Expression of the FGFR-4 Arg³88 protein leads to increased activity of the extracellular signal-related kinase (ERK) pathway, increased activity of serum response factor (SRF) and AP1, and transcription of multiple genes that are correlated with aggressive clinical behavior in PCa. Increased expression of SRF is associated with biochemical recurrence in men undergoing radical prostatectomy. Consistent with these observations, knockdown of FGFR-4 Arg³88 in PCa cells decreases proliferation and invasion in vitro and primary tumor growth and metastasis in vivo. CONCLUSIONS: These studies define a signal transduction pathway downstream of FGFR-4 Arg³88 that acts via ERK and SRF to promote PCa progression.


Extracellular Signal-Regulated MAP Kinases/metabolism , Prostatic Neoplasms/physiopathology , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Serum Response Factor/metabolism , Signal Transduction , Alleles , Cell Line, Tumor , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 4/genetics , Survival Analysis , Transcription Factor AP-1/metabolism
5.
Am J Pathol ; 178(1): 79-87, 2011 Jan.
Article En | MEDLINE | ID: mdl-21224046

We previously reported that reactive stroma grading in prostate cancer (PCa) is predictive of biochemical recurrence in prostatectomies and biopsies. In this study, we tested whether quantifying the percentage of reactive stromal grade 3 (RSG 3; stromogenic carcinoma pattern) in the entire tumor is predictive of PCa-specific death. Whole-mount prostatectomies operated by a single surgeon obtained between 1983 and 1998 were reviewed. Reactive stroma was evaluated as described previously, and areas of RSG 3 in the entire tumor were registered as percentages of total tumor. Statistical analysis was performed using Spearman, Kaplan-Meier, and Cox analyses. In all, 872 cases were evaluable. Quantification of RSG 3 percentage was an independent predictor of biochemical recurrence, analyzed as a continuous or grouped variable. Patients with higher RSG 3 percentages (larger tumor areas with RSG 3) had a significantly decreased biochemical recurrence-free survival than those with a lower RSG 3 percentage, even within the Gleason score 7 subset of patients. A nomogram introduced this new variable to the model. Furthermore, quantification of RSG 3 percentage was significantly predictive of PCa-specific death. Quantification of the RSG 3 (stromogenic carcinoma) area in PCa provides additional novel information on prognosis. These data substantiate the concept that the tumor microenvironment holds significant predictive information, as well as biological significance.


Carcinoma/mortality , Carcinoma/pathology , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Aged , Carcinoma/surgery , Cohort Studies , Disease-Free Survival , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Prostatectomy , Prostatic Neoplasms/surgery
6.
Cancer Res ; 71(2): 572-82, 2011 Jan 15.
Article En | MEDLINE | ID: mdl-21224358

Patients with metastatic prostate cancer who undergo androgen-ablation therapy invariably relapse and develop incurable castration-resistant disease. Activation of the prosurvival Akt pathway accompanies androgen ablation. We discovered that the androgen receptor induces the expression of the tumor suppressor inositol polyphosphate 4-phosphatase type II (INPP4B) but not PTEN in prostate cancer cells. Optimal induction of INPP4B by an androgen receptor required the expression of the transcriptional coactivator NCoR. INPP4B dephosphorylates phosphatidylinositol-3, 4-bisphosphate, which leads to reduced phosphorylation and activity of Akt. In support of a key role for INPP4B in Akt control, INPP4B depletion activated Akt and increased cellular proliferation. The clinical significance of INPP4B in androgen-dependent prostate cancers was determined in normal or primary tumor prostate tissues derived from radical prostatectomy specimens. In primary tumors, the expression of both INPP4B and PTEN was substantially reduced compared with normal tissue. Further, the decreased expression of INPP4B reduced the time to biochemical recurrence. Thus, androgen ablation can activate the Akt pathway via INPP4B downregulation, thereby mitigating the antitumor effects of androgen ablation. Our findings reinforce the concept that patients undergoing androgen ablation may benefit from Akt-targeting therapies.


Androgens/pharmacology , Genes, Tumor Suppressor , Phosphoric Monoester Hydrolases/genetics , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Androgens/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Recurrence, Local/enzymology , Neoplasm Recurrence, Local/metabolism , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphoric Monoester Hydrolases/biosynthesis , Phosphoric Monoester Hydrolases/deficiency , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/genetics , Transfection
7.
Cancer Res ; 71(4): 1325-33, 2011 Feb 15.
Article En | MEDLINE | ID: mdl-21169414

The TMPRSS2/ERG (T/E) fusion gene is present and thought to be an oncogenic driver of approximately half of all prostate cancers. Fusion of the androgen-regulated TMPRSS2 promoter to the ERG oncogene results in constitutive high level expression of ERG which promotes prostate cancer invasion and proliferation. Here, we report the characterization of multiple alternatively spliced T/E fusion gene isoforms which have differential effects on invasion and proliferation. We found that T/E fusion gene isoforms differentially increase NF-κB-mediated transcription, which may explain in part the differences in biological activities of the T/E fusion isoforms. This increased activity is due to phosphorylation of NF-κB p65 on Ser536. Tissue microarray immunochemistry revealed that p65 phospho-Ser536 is present in the majority of prostate cancers where it is associated with ERG protein expression. The T/E fusion gene isoforms differentially increase expression of a number of NF-κB associated genes including PAR1, CCL2, FOS, TLR3, and TLR4 (Toll-like receptor). TLR4 activation is known to promote p65 Ser536 phosphorylation and knockdown of TLR4 with shRNA decreases Ser536 phosphorylation in T/E fusion gene expressing cells. TLR4 can be activated by proteins in the tumor microenvironment and lipopolysacharide from Gram (-) bacteria. Our findings suggest that bacterial infection of the prostate and/or endogenous microenvironment proteins may promote progression of high-grade prostatic intraepithelial neoplasia and/or prostate cancers that express the T/E fusion gene, where the NF-κB pathway might be targeted as a rational therapeutic approach.


NF-kappa B/metabolism , Oncogene Proteins, Fusion/physiology , Toll-Like Receptor 4/physiology , Animals , COS Cells , Cell Proliferation/drug effects , Cells, Cultured , Chlorocebus aethiops , Gene Knockdown Techniques , Humans , Male , NF-kappa B/agonists , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/pharmacology , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/chemistry , Protein Isoforms/pharmacology , Protein Isoforms/physiology , RNA, Small Interfering/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology
8.
Clin Cancer Res ; 15(12): 3979-89, 2009 Jun 15.
Article En | MEDLINE | ID: mdl-19509179

PURPOSE: Marked reactive stroma formation, designated as grade 3 reactive stroma, is associated with poor outcome in clinically localized prostate cancer. To understand the biological processes and signaling mechanisms underlying the formation of such reactive stroma, we carried out microarray gene expression analysis of laser-captured reactive stroma and matched normal stroma. EXPERIMENTAL DESIGN: Seventeen cases of reactive stroma grade 3 cancer were used to laser-capture tumor and normal stroma. Expression analysis was carried out using Agilent 44K arrays. Up-regulation of selected genes was confirmed by quantitative reverse transcription-PCR. Expression data was analyzed to identify significantly up- and down-regulated genes, and gene ontology analysis was used to define pathways altered in reactive stroma. RESULTS: A total of 544 unique genes were significantly higher in the reactive stroma and 606 unique genes were lower. Gene ontology analysis revealed significant alterations in a number of novel processes in prostate cancer reactive stroma, including neurogenesis, axonogenesis, and the DNA damage/repair pathways, as well as evidence of increases in stem cells in prostate cancer reactive stroma. CONCLUSIONS: Formation of reactive stroma in prostate cancer is a dynamic process characterized by significant alterations in growth factor and signal transduction pathways and formation of new structures, including nerves and axons.


Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Breast Neoplasms/genetics , Down-Regulation/genetics , Down-Regulation/physiology , Female , Humans , Male , Prostatic Neoplasms/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Up-Regulation/genetics , Up-Regulation/physiology
9.
Anticancer Res ; 29(6): 2077-81, 2009 Jun.
Article En | MEDLINE | ID: mdl-19528467

BACKGROUND: Activation of glycogen synthase kinase-3 (GSK-3) is involved in the regulation of cell growth, differentiation, mobility, proliferation and survival. However, its clinicopathologic significance remains unclear in prostate cancer (PCa). MATERIALS AND METHODS: A tissue microarray was produced from 640 samples. Sections were immunostained with an antibody against the non-phosphorylated form of GSK-3(GSK-3beta) and were digitized. Spearman correlation test was processed for correlations between GSK-3beta and biological and clinicopathological variables. The prognostic value of GSK-3beta was analyzed by Cox Regression model. RESULTS: Cytoplasmic GSK-3beta was higher in PCa than in normal prostate (mean expression index 4.55 vs. 3.50, p<0.0001). Conversely, nuclear expression was higher in normal prostate than that in PCa (3.38 vs. 2.04, p<0.0001). Cytoplasmic levels of GSK-3beta were correlated with clinical stage (rho=0.095, p=0.0337), lymph node metastasis (rho=0.116, p=0.0096), extracapsular extension (rho=0.092, p=0.0392), and Gleason score (rho=0.167, p=0.0002). Increased cytoplasmic GSK-3beta expression was correlated with high Ki-67 labeling index (rho=0.319, p<0.0001), low apoptotic index by TUNEL (rho=-0.118, p=0.0134), high levels of androgen receptor (rho=0.292, p<0.0001) and p-Akt (rho=0.396, p<0.0001). Patients with higher cytoplasmic levels of GSK-3beta had a two-fold risk of biochemical recurrence-free survival compared to those with lower levels of GSK-3beta [HR 1.934 (1.020-3.667), p=0.043]. CONCLUSION: Cytoplasmic accumulation of GSK-3beta is potentially associated with a pro-survival mechanism that promotes PCa development and progression.


Cytoplasm/enzymology , Glycogen Synthase Kinase 3/metabolism , Prostate/enzymology , Prostatic Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Apoptosis , Glycogen Synthase Kinase 3 beta , Humans , Immunoenzyme Techniques , Male , Middle Aged , Prognosis , Prostatic Neoplasms/pathology , Survival Rate , Tissue Array Analysis
10.
Clin Cancer Res ; 15(10): 3568-73, 2009 May 15.
Article En | MEDLINE | ID: mdl-19417030

BACKGROUND: Akt/protein kinase B signaling pathway has been implicated in tumorigenesis and progression. Previous studies showed the predictive potential of p-Akt-1, but total Akt-1 could provide more reliable information. We used image deconvolution, nanotechnology (quantum dots), and image analysis to improve Akt-1 quantification. DESIGN: This tissue microarray study included 840 radical prostatectomy cases. Slides were incubated with primary antibody against nonphosphorylated Akt-1 (Akt-1) followed by biotinylated secondary antibody and then by Qdot655 streptavidin conjugate. Slides were imaged under fluorescence microscopy and spectral deconvolution (Nuance) and quantified using plug-in image analysis software. Average intensity of Akt-1 signal was measured and subject to statistical analysis. Multivariate analysis (Cox regression) was applied to assess the prognostic value of Akt-1 for biochemical recurrence and prostate cancer-specific death. Akt-1 expression was also examined for correlations with Ki-67 index and apoptotic index in our database. RESULT: Akt-1 was inversely correlated with apoptotic index (rho = -0.203; P = 0.004) but not with Ki-67 index. The correlation between Akt and p-Akt is significant but weak (P = 0.0496; R(2) = 0.118). On multivariate analysis Akt-1 was independently predictive of biochemical recurrence [hazard ratio, 2.863 (95% confidence interval, 1.127-7.271); P = 0.0270]. Akt-1 level is also predictive of prostate cancer-specific death (P = 0.0376). CONCLUSION: High levels of Akt-1, assessed by quantum dots, deconvolution imaging, and image analysis, are associated with a higher risk of biochemical recurrence and prostate cancer-specific death.


Image Processing, Computer-Assisted/methods , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Quantum Dots , Adult , Aged , Aged, 80 and over , Cohort Studies , Fluorescent Antibody Technique , Humans , Kaplan-Meier Estimate , Ki-67 Antigen/analysis , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local , Predictive Value of Tests , Prognosis , Prostatectomy/methods , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/surgery , Tissue Array Analysis
11.
Cancer Res ; 69(3): 819-27, 2009 Feb 01.
Article En | MEDLINE | ID: mdl-19176382

GGAP2/PIKE-A is a GTP-binding protein that can enhance Akt activity. Increased activation of the AKT and nuclear factor-kappaB (NF-kappaB) pathways have been identified as critical steps in cancer initiation and progression in a variety of human cancers. We have found significantly increased expression GGAP2 in the majority of human prostate cancers and GGAP2 expression increases Akt activation in prostate cancer cells. Thus, increased GGAP2 expression is a common mechanism for enhancing the activity of the Akt pathway in prostate cancers. In addition, we have found that activated Akt can bind and phosphorylate GGAP2 at serine 629, which enhances GTP binding by GGAP2. Phosphorylated GGAP2 can bind the p50 subunit of NF-kappaB and enhances NF-kappaB transcriptional activity. When expressed in prostate cancer cells, GGAP2 enhances proliferation, foci formation, and tumor progression in vivo. Thus, increased GGAP2 expression, which is present in three quarters of human prostate cancers, can activate two critical pathways that have been linked to prostate cancer initiation and progression.


GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , NF-kappa B p50 Subunit/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Enzyme Activation , GTP-Binding Proteins/biosynthesis , GTP-Binding Proteins/genetics , GTPase-Activating Proteins/biosynthesis , GTPase-Activating Proteins/genetics , Humans , Male , Mice , Mice, SCID , NF-kappa B p50 Subunit/biosynthesis , NF-kappa B p50 Subunit/genetics , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Transcriptional Activation , Transfection , Transplantation, Heterologous
12.
Clin Cancer Res ; 14(23): 7593-603, 2008 Dec 01.
Article En | MEDLINE | ID: mdl-19047084

PURPOSE: Perineural invasion is the only interaction between cancer cells and nerves studied to date. It is a symbiotic relationship between cancer and nerves that results in growth advantage for both. In this article, we present data on a novel biological phenomenon, cancer-related axonogenesis and neurogenesis. EXPERIMENTAL DESIGN: We identify spatial and temporal associations between increased nerve density and preneoplastic and neoplastic lesions of the human prostate. RESULTS: Nerve density was increased in cancer areas as well as in preneoplastic lesions compared with controls. Two- and three-dimensional reconstructions of entire prostates confirmed axonogenesis in human tumors. Furthermore, patients with prostate cancer had increased numbers of neurons in their prostatic ganglia compared with controls, corroborating neurogenesis. Finally, two in vitro models confirmed that cancer cells, particularly when interacting with nerves in perineural invasion, induce neurite outgrowth in prostate cancer. Neurogenesis is correlated with features of aggressive prostate cancer and with recurrence in prostate cancer. We also present a putative regulatory mechanism based on semaphorin 4F (S4F). S4F is overexpressed in cancers cells in the perineural in vitro model. Overexpression of S4F in prostate cancer cells induces neurogenesis in the N1E-115 neurogenesis assay and S4F inhibition by small interfering RNA blocks this effect. CONCLUSIONS: This is the first description of cancer-related neurogenesis and its putative regulatory mechanism.


Neoplasm Invasiveness/pathology , Neurogenesis/physiology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Humans , Male , Peripheral Nerves/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Prostate/innervation , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Semaphorins/metabolism , Tissue Array Analysis
13.
Clin Cancer Res ; 14(22): 7511-8, 2008 Nov 15.
Article En | MEDLINE | ID: mdl-19010869

PURPOSE: To assess the safety of administering bortezomib to patients undergoing a radical prostatectomy, to assess pathologic changes induced by bortezomib in prostate cancer specimen, and to verify alterations by the drug in proteasome protein targets. EXPERIMENTAL DESIGN: Bortezomib is a proteasome inhibitor that has shown activity in vitro and in vivo in prostate cancer. We performed a neoadjuvant clinical trial of bortezomib in men with prostate cancer at high risk of recurrence. The primary endpoints were to evaluate safety and biological activity. RESULTS: Bortezomib is generally safe in the preoperative setting. Antitumor activity was manifested by tumor cytopathic effect, drops in serum prostate-specific antigen in some patients, and increases in tumor apoptosis. This was associated with cytoplasmic entrapment of nuclear factor-kappaB. We found an unexpected increase in proliferation in treated tissues and in vitro. Bortezomib also increased SRC-3 levels and phosphorylated Akt, both in vitro and in treated prostate cancer tissues. Knockdown of SRC-3 blocked the increase in activated Akt in vitro. Combined treatment with bortezomib and the Akt inhibitor perifosine was more effective than either agent alone in vitro. CONCLUSION: These data suggest that combined therapies targeting the proteasome and the Akt pathway may have increased efficacy.


Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Histone Acetyltransferases/drug effects , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/drug effects , Pyrazines/therapeutic use , Trans-Activators/drug effects , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Blotting, Western , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Flow Cytometry , Histone Acetyltransferases/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , NF-kappa B/drug effects , Neoadjuvant Therapy , Nuclear Receptor Coactivator 3 , Phosphorylcholine/administration & dosage , Phosphorylcholine/analogs & derivatives , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/drug effects , Prostatectomy , Prostatic Neoplasms/surgery , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Trans-Activators/metabolism
14.
Cancer Res ; 68(13): 5460-8, 2008 Jul 01.
Article En | MEDLINE | ID: mdl-18593949

Steroid receptor coactivator-3 (SRC-3)/AIB1 is a member of the p160 nuclear receptor coactivator family involved in development and cell cycle progression. We previously showed that SRC-3/AIB1 is required for prostate cancer cell proliferation and survival. Here, we reported that the elevated SRC-3/AIB1 expression is significantly correlated with human prostate cancer seminal vesicle invasion and lymph node metastasis. Furthermore, SRC-3/AIB1 is associated with increased prostate cancer cell migration and invasion. SRC-3/AIB1 is required for focal adhesion turnover and focal adhesion kinase activation. In addition, SRC-3/AIB1 directly regulates transcription of matrix metalloproteinase (MMP)-2 and MMP-13 through its coactivation of AP-1 and PEA3. Taken together, these data suggest that SRC-3/AIB1 plays an essential role in prostate cancer cell invasion and metastasis.


Carcinoma/genetics , Cell Movement/genetics , Focal Adhesions/metabolism , Matrix Metalloproteinases/genetics , Prostatic Neoplasms/genetics , Transcription Factors/physiology , Carcinoma/metabolism , Carcinoma/pathology , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Male , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/physiology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Nuclear Receptor Coactivator 3 , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction/genetics , Transcription Factor AP-1/physiology , Transcription Factors/genetics , Tumor Cells, Cultured
15.
Hum Pathol ; 39(2): 282-91, 2008 Feb.
Article En | MEDLINE | ID: mdl-18206496

We previously reported that reactive stromal grading in radical prostatectomies is a predictor of recurrence and that reactive stromal grading 0 and 3 are associated with lower biochemical recurrence-free survival rates than reactive stromal grading 1 and 2. We explored the prognostic significance of reactive stromal grading in preoperative needle biopsies. At Baylor College of Medicine, 224 cases of prostatic carcinoma were diagnosed by needle biopsy. Reactive stromal grading was evaluated on hematoxylin-eosin (H&E)-stained sections on the basis of previously described criteria: grade 0, with 0% to 5% reactive stroma; grade 1, 6% to 15%; grade 2, 16% to 50%; grade 3, 51% to 100%, or at least a 1:1 ratio between glands and stroma. Kaplan-Meier and Cox proportional hazard analyses were used. Reactive stromal grading distribution was as follows: reactive stromal grading 0, 1 case (0.5%); reactive stromal grading 1, 149 cases (66.5%); reactive stromal grading 2, 59 cases (26.3%); reactive stromal grading 3, 15 cases (6.7%). Reactive stromal grading in biopsies was correlated with adverse clinicopathologic parameters in the prostatectomy. Patients with reactive stromal grading 1 and 2 had better survival than those with 0 and 3 (P = .0034). Reactive stromal grading was an independent predictor of recurrence (hazard ratio = 1.953; P = .0174). Reactive stromal grading is independent of Gleason 4 + 3 and 3 + 4 in patients with a Gleason score of 7. Quantitation of reactive stroma and recognition of the stromogenic carcinoma in H&E-stained biopsies is useful to predict biochemical recurrence in prostate carcinoma patients independent of Gleason grade and prostate-specific antigen.

16.
Cancer Cell ; 12(6): 559-71, 2007 Dec.
Article En | MEDLINE | ID: mdl-18068632

Fibroblast Growth Factor Receptor-1 (FGFR1) is commonly overexpressed in advanced prostate cancer (PCa). To investigate causality, we utilized an inducible FGFR1 (iFGFR1) prostate mouse model. Activation of iFGFR1 with chemical inducers of dimerization (CID) led to highly synchronous, step-wise progression to adenocarcinoma that is linked to an epithelial-to-mesenchymal transition (EMT). iFGFR1 inactivation by CID withdrawal led to full reversion of prostatic intraepithelial neoplasia, whereas PCa lesions became iFGFR1-independent. Gene expression profiling at distinct stages of tumor progression revealed an increase in EMT-associated Sox9 and changes in the Wnt signaling pathway, including Fzd4, which was validated in human PCa. The iFGFR1 model clearly implicates FGFR1 in PCa progression and demonstrates how CID-inducible models can help evaluate candidate molecules in tumor progression and maintenance.


Epithelial Cells/enzymology , Epithelial Cells/pathology , Mesoderm/enzymology , Mesoderm/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Animals , Dimerization , Disease Progression , Enzyme Activation , Enzyme Induction , Gene Expression Regulation, Neoplastic , High Mobility Group Proteins/metabolism , Humans , Immunohistochemistry , Male , Mice , Neoplasm Metastasis , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Neoplasms/genetics , Remission Induction , SOX9 Transcription Factor , Signal Transduction , Transcription Factors/metabolism
17.
Hum Pathol ; 38(11): 1611-20, 2007 Nov.
Article En | MEDLINE | ID: mdl-17868773

We previously reported that reactive stromal grading in radical prostatectomies is a predictor of recurrence and that reactive stromal grading 0 and 3 are associated with lower biochemical recurrence-free survival rates than reactive stromal grading 1 and 2. We explored the prognostic significance of reactive stromal grading in preoperative needle biopsies. At Baylor College of Medicine, 224 cases of prostatic carcinoma were diagnosed by needle biopsy. Reactive stromal grading was evaluated on hematoxylin-eosin (H&E)-stained sections on the basis of previously described criteria: grade 0, with 0% to 5% reactive stroma; grade 1, 6% to 15%; grade 2, 16% to 50%; grade 3, 51% to 100%, or at least a 1:1 ratio between glands and stroma. Kaplan-Meier and Cox proportional hazard analyses were used. Reactive stromal grading distribution was as follows: reactive stromal grading 0, 1 case (0.5%); reactive stromal grading 1, 149 cases (66.5%); reactive stromal grading 2, 59 cases (26.3%); reactive stromal grading 3, 15 cases (6.7%). Reactive stromal grading in biopsies was correlated with adverse clinicopathologic parameters in the prostatectomy. Patients with reactive stromal grading 1 and 2 had better survival than those with 0 and 3 (P = .0034). Reactive stromal grading was an independent predictor of recurrence (hazard ratio = 1.953; P = .0174). Reactive stromal grading is independent of Gleason 4 + 3 and 3 + 4 in patients with a Gleason score of 7. Quantitation of reactive stroma and recognition of the stromogenic carcinoma in H&E-stained biopsies is useful to predict biochemical recurrence in prostate carcinoma patients independent of Gleason grade and prostate-specific antigen.


Prostatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Biopsy, Needle , Cohort Studies , Disease-Free Survival , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Prostate/pathology , Prostatectomy , Prostatic Neoplasms/surgery , Recurrence , Stromal Cells/pathology
18.
Hum Pathol ; 38(10): 1501-7, 2007 Oct.
Article En | MEDLINE | ID: mdl-17597184

In vitro studies suggest that the proapoptotic function of forkhead protein FKHR is probably inactivated by means of phosphorylation through the protein kinase B pathway. However, the clinical significance of FKHR in prostate cancer remains unclear. Six hundred forty radical prostatectomies were used for building tissue microarrays. Slides were stained with antibodies against FKHR and phosphorylated FKHR (p-FKHR). Correlations with clinicopathologic parameters were analyzed by Spearman rank test. Cox regression test and Kaplan-Meier test were used to determine the probability of disease recurrence, which is defined as a serum prostate-specific antigen (PSA) level greater than 0.4 ng/mL after radical prostatectomy. Nuclear FKHR level was higher in normal prostate than in benign prostatic hyperplasia and prostate cancer (P = .0000). Nuclear expression of FKHR was correlated with preoperative PSA level (rho = 0.108, P = .029), extracapsular extension (rho = 0.137, P = .005), and seminal vesicle invasion (rho = 0.101, P = .039). FKHR expression was not a significant indicator of biochemical failure by either univariate or multivariate analysis. Nuclear p-FKHR expression correlated with patients' age (rho = 0.179, P = .0003), Gleason score (rho = 0.130, P = .0083), extracapsular extension (rho = 0.227, P = .0000), clinical stage (Union Internationale Contre le Cancer system) (rho = 0.166, P = .0007), and lymph node status (rho = 0.101, P = .0401). Cytoplasmic p-FKHR correlated with patients' age (rho = 0.146, P = .0030) and clinical stage (rho = 0.117, P = .0180). Cytoplasmic p-FKHR was a significant indicator of biochemical recurrence (P = .0164; hazard ratio, 1.114-2.929). Nuclear p-FKHR strongly correlated with phosphorylated protein kinase B (rho = 0.368, P = .0000), androgen receptor (rho = 0.385, P = .0000), and Skp-2 (rho = 0.170, P = .0036). Our data suggest that the proapoptotic role of FKHR is probably regulated by several signaling pathways in prostate cancer.


Biomarkers, Tumor/analysis , Forkhead Transcription Factors/metabolism , Prostatic Neoplasms/metabolism , Signal Transduction/physiology , 3-Phosphoinositide-Dependent Protein Kinases , Adult , Aged , Aged, 80 and over , Apoptosis/physiology , Cell Nucleus/metabolism , Cytoplasm/metabolism , Disease Progression , Forkhead Box Protein O1 , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Phosphorylation , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/metabolism , Tissue Array Analysis
19.
Clin Cancer Res ; 13(6): 1695-702, 2007 Mar 15.
Article En | MEDLINE | ID: mdl-17363522

PURPOSE: To understand the role of relaxin peptide in prostate cancer, we analyzed the expression of relaxin and its receptor in human prostate cancer samples, the effects of relaxin signaling on cancer cell phenotype in vitro, and the effects of increased serum relaxin concentrations on cancer progression in vivo. EXPERIMENTAL DESIGN: The relaxin and its receptor leucine-rich repeat containing G protein-coupled receptor 7 (LGR7) expression were studied by quantitative reverse transcription-PCR (11 benign and 44 cancer tissue samples) and by relaxin immunohistochemistry using tissue microarrays containing 10 normal and 69 cancer samples. The effects of relaxin treatment and endogenous relaxin/LGR7 suppression via short interfering RNA in PC-3 and LNCaP cells were analyzed in vitro. The effect of transgenic relaxin overexpression [Tg(Rln1)] on cancer growth and survival was evaluated in autochthonous transgenic adenocarcinoma of the mouse prostate (TRAMP). RESULTS: The relaxin mRNA expression was significantly higher in recurrent prostate cancer samples. In tissue microarrays of the 10 normal tissues, 8 had low staining in epithelial cells, whereas only 1 of 9 high-grade prostatic intraepithelial neoplasia lesions had low expression (P = 0.005) and only 29 of 65 cancers had low expression (P = 0.047). Stimulation with relaxin increased cell proliferation, invasiveness, and adhesion in vitro. The suppression of relaxin/LGR7 via short interfering RNAs decreased cell invasiveness by 90% to 95% and growth by 10% to 25% and increased cell apoptosis 0.6 to 2.2 times. The Tg(Rln1) TRAMP males had shorter median survival time, associated with the decreased apoptosis of tumor cells, compared with non-Tg(Rln1) TRAMP animals. CONCLUSIONS: Relaxin signaling plays a role in prostate cancer progression.


Adenocarcinoma/pathology , Prostatic Neoplasms/pathology , Relaxin/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide , Relaxin/genetics , Relaxin/metabolism , Relaxin/pharmacology , Survival Analysis
20.
Hum Pathol ; 38(2): 299-307, 2007 Feb.
Article En | MEDLINE | ID: mdl-17097719

Pancreatic cancer (PanCa) is characterized by perineural invasion (PNI), early lymph node and liver metastasis, and poor prognosis. PNI is one of the important causes of local recurrence. Little is known about the mechanism of PNI in PanCa. We presented a novel model system that may shed light on the mystery of PNI in PanCa. In this study, mouse dorsal root ganglia (DRGs) and human PanCa cell line (MIA PaCa-2) were cocultured in Matrigel matrix (BD Biosciences, San Jose, CA) to build this PNI model. MIA PaCa-2 cell line alone (control 1) or DRG alone (control 2) was cultured with Matrigel matrix as controls. Neurite outgrowth, cell colony growth, neurite-colony contact, and retrograde extension were observed under inverted microscopy and then were photographed and quantitated with the Optimas imaging system (Optimas Corp., Bothell, MA). At day 14, both the experimental and control 2 samples were harvested and subjected to total RNA isolation and fixed in paraffin-embedded blocks. Slides cut from paraffin blocks were studied with Ki-67 immunostaining and TUNEL assay. Gene profiling was performed using complementary DNA microarray. Overexpressed target genes were verified by quantitative reverse transcriptase polymerase chain reaction. The results showed that reciprocity was observed between neurites and MIA PaCa colonies with 24 hours of coculture. Neurite outgrowth was stimulated in the presence of pancreatic carcinoma cells, which showed 2-fold more area than did control 2. After 72 hours, MIA PaCa colonies cocultured with DRG exhibited 58% more colony area than did control 1. The Ki-67 index of the DRG/MIA PaCa cells (mean, 5.02%) was significantly higher than that in control 1 (mean, 1.18%) (P < .05); in contrast, the apoptotic index in the DRG/MIA PaCa cells was significantly lower (mean, 0.45%) than that in the control 1 (mean, 1.85%) (P < .001). Prosurvival genes MALT1 and TRAF were increased 2-fold in DRG/MIA PaCa compared with controls. We demonstrated that neural-epithelial interaction is a mutually beneficial process for the growth of nerves and PanCa cells. It is possible that oncogenes and growth factors might act synergistically in promoting proliferation and/or inhibiting apoptosis, a survival strategy crucial to the development of PNI in PanCa.


Cell Proliferation , Ganglia, Spinal/metabolism , Neurites/physiology , Animals , Apoptosis , Caspases/genetics , Cell Line, Tumor , Cell Survival , Coculture Techniques , Ganglia, Spinal/chemistry , Ganglia, Spinal/cytology , Gene Expression Profiling , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Immunohistochemistry , In Situ Nick-End Labeling , Ki-67 Antigen/analysis , Mice , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/genetics , Neurites/chemistry , Neurites/metabolism , Oligonucleotide Array Sequence Analysis/methods , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics
...