Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 55: 128441, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34767912

ABSTRACT

The combination of androgen receptor antagonists with histone deacetylase inhibitors (HDACi) has been shown to be more effective than antiandrogens alone in halting growth of prostate cancer cell lines. Here we have designed, synthesized and assessed a series of antiandrogen/HDACi hybrids by combining structural features of enzalutamide with either SAHA or entinostat. The hybrids are demonstrated to maintain bifunctionality using a fluorometric HDAC assay and a bioluminescence resonance energy transfer (BRET) antiandrogen assay. Antiproliferative assays showed that hybrids bearing o-aminoanilide-based HDACi motifs outperformed hydroxamic acid based HDACi's. The hybrids demonstrated selectivity for epithelial cell lines vs. stromal cell lines, suggesting a potentially useful therapeutic window.


Subject(s)
Androgen Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Nitriles/pharmacology , Phenylthiohydantoin/pharmacology , Pyridines/pharmacology , Androgen Antagonists/chemical synthesis , Androgen Antagonists/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzamides/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fluorescence Resonance Energy Transfer , Fluorometry , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Molecular Structure , Nitriles/chemistry , Phenylthiohydantoin/chemistry , Pyridines/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
2.
Dis Model Mech ; 12(7)2019 07 25.
Article in English | MEDLINE | ID: mdl-31350272

ABSTRACT

Prostate development is controlled by androgens, the eandrogen receptor (AR) and mesenchymal-epithelial signalling. We used chromatin immunoprecipitation sequencing (ChIP-seq) to define AR genomic binding in the male and female mesenchyme. Tissue- and single-cell-based transcriptional profiling was used to define mesenchymal AR target genes. We observed significant AR genomic binding in females and a strong enrichment at proximal promoters in both sexes. In males, there was greater AR binding to introns and intergenic regions as well as to classical AR binding motifs. In females, there was increased proximal promoter binding and involvement of cofactors. Comparison of AR-bound genes with transcriptomic data enabled the identification of novel sexually dimorphic AR target genes. We validated the dimorphic expression of AR target genes using published datasets and confirmed regulation by androgens using ex vivo organ cultures. AR targets showed variable expression in patients with androgen insensitivity syndrome. We examined AR function at single-cell resolution using single-cell RNA sequencing (scRNA-seq) in male and female mesenchyme. Surprisingly, both AR and target genes were distributed throughout cell subsets, with few positive cells within each subset. AR binding was weakly correlated with target gene expression.


Subject(s)
Prostate/growth & development , Receptors, Androgen/metabolism , Transcriptome , Animals , Chromatin Immunoprecipitation , Female , Humans , Male , Mesoderm/metabolism , Organogenesis , Promoter Regions, Genetic , Protein Binding , Rats , Real-Time Polymerase Chain Reaction , Receptors, Androgen/genetics
3.
J Pathol ; 249(4): 411-424, 2019 12.
Article in English | MEDLINE | ID: mdl-31206668

ABSTRACT

Prostate cancer is heterogeneous in both cellular composition and patient outcome, and development of biomarker signatures to distinguish indolent from aggressive tumours is a high priority. Stroma plays an important role during prostate cancer progression and undergoes histological and transcriptional changes associated with disease. However, identification and validation of stromal markers is limited by a lack of datasets with defined stromal/tumour ratio. We have developed a prostate-selective signature to estimate the stromal content in cancer samples of mixed cellular composition. We identified stromal-specific markers from transcriptomic datasets of developmental prostate mesenchyme and prostate cancer stroma. These were experimentally validated in cell lines, datasets of known stromal content, and by immunohistochemistry in tissue samples to verify stromal-specific expression. Linear models based upon six transcripts were able to infer the stromal content and estimate stromal composition in mixed tissues. The best model had a coefficient of determination R2 of 0.67. Application of our stromal content estimation model in various prostate cancer datasets led to improved performance of stromal predictive signatures for disease progression and metastasis. The stromal content of prostate tumours varies considerably; consequently, deconvolution of stromal proportion may yield better results than tumour cell deconvolution. We suggest that adjusting expression data for cell composition will improve stromal signature performance and lead to better prognosis and stratification of men with prostate cancer. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Models, Genetic , Prostatic Neoplasms/genetics , Stromal Cells/metabolism , Transcriptome , Biomarkers, Tumor/metabolism , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Male , PC-3 Cells , Predictive Value of Tests , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Registries , Reproducibility of Results , Retrospective Studies , Stromal Cells/pathology
4.
Mol Cell Endocrinol ; 471: 1-14, 2018 08 15.
Article in English | MEDLINE | ID: mdl-28483704

ABSTRACT

The androgen receptor (AR) is a transcription factor, and key regulator of prostate development and cancer, which has discrete functions in stromal versus epithelial cells. AR expressed in mesenchyme is necessary and sufficient for prostate development while loss of stromal AR is predictive of prostate cancer progression. Many studies have characterized genome-wide binding of AR in prostate tumour cells but none have used primary mesenchyme or stroma. We applied ChIPseq to identify genomic AR binding sites in primary human fetal prostate fibroblasts and patient derived cancer associated fibroblasts, as well as the WPMY1 cell line overexpressing AR. We identified AR binding sites that were specific to fetal prostate fibroblasts (7534), cancer fibroblasts (629), WPMY1-AR (2561) as well as those common among all (783). Primary fibroblasts had a distinct AR binding profile versus prostate cancer cell lines and tissue, and showed a localisation to gene promoter binding sites 1 kb upstream of the transcriptional start site, as well as non-classical AR binding sequence motifs. We used RNAseq to define transcribed genes associated with AR binding sites and derived cistromes for embryonic and cancer fibroblasts as well as a cistrome common to both. These were compared to several in vivo ChIPseq and transcript expression datasets; which identified subsets of AR targets that were expressed in vivo and regulated by androgens. This analysis enabled us to deconvolute stromal AR targets active in stroma within tumour samples. Taken together, our data suggest that the AR shows significantly different genomic binding site locations in primary prostate fibroblasts compared to that observed in tumour cells. Validation of our AR binding site data with transcript expression in vitro and in vivo suggests that the AR target genes we have identified in primary fibroblasts may contribute to clinically significant and biologically important AR-regulated changes in prostate tissue.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Fetus/cytology , Fibroblasts/metabolism , Genome, Human , Prostate/pathology , Receptors, Androgen/metabolism , Base Sequence , Cells, Cultured , Humans , Male , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Sci Rep ; 7(1): 16385, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180763

ABSTRACT

Prostate organogenesis involves epithelial growth controlled by inductive signalling from specialised mesenchymal subsets. To identify pathways active in mesenchyme we used tissue and single cell transcriptomics to define mesenchymal subsets and subset-specific transcript expression. We documented transcript expression using Tag-seq and RNA-seq in female rat Ventral Mesenchymal Pad (VMP) as well as adjacent urethra comprised of smooth muscle and peri-urethral mesenchyme. Transcripts enriched in female VMP were identified with Tag-seq of microdissected tissue, RNA-seq of cell populations, and single cells. We identified 400 transcripts as enriched in the VMP using bio-informatic comparisons of Tag-seq and RNA-seq data, and 44 were confirmed by single cell RNA-seq. Cell subset analysis showed that VMP and adjacent mesenchyme were composed of distinct cell types and that each tissue contained two subgroups. Markers for these subgroups were highly subset specific. Thirteen transcripts were validated by qPCR to confirm cell specific expression in microdissected tissues, as well as expression in neonatal prostate. Immunohistochemical staining demonstrated that Ebf3 and Meis2 showed a restricted expression pattern in female VMP and prostate mesenchyme. We conclude that prostate inductive mesenchyme shows limited cellular heterogeneity and that transcriptomic analysis identified new mesenchymal subset transcripts associated with prostate organogenesis.


Subject(s)
Gene Expression Profiling , Mesoderm/embryology , Mesoderm/metabolism , Organogenesis/genetics , Prostate/enzymology , Prostate/metabolism , Transcriptome , Animals , Computational Biology/methods , Gene Ontology , High-Throughput Nucleotide Sequencing , Male , Rats , Single-Cell Analysis
6.
Br J Cancer ; 116(6): 775-784, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28152543

ABSTRACT

BACKGROUND: Prostate cancer shows considerable heterogeneity in disease progression and we propose that markers expressed in tumour stroma may be reliable predictors of aggressive tumour subtypes. METHODS: We have used Kaplan-Meier, univariate and multivariate analysis to correlate the expression of Asporin (ASPN) mRNA and protein with prostate cancer progression in independent cohorts. We used immunohistochemistry and H scoring to document stromal localisation of ASPN in a tissue microarray and mouse prostate cancer model, and correlated expression with reactive stroma, defined using Masson Trichrome staining. We used cell cultures of primary prostate cancer fibroblasts treated with serum-free conditioned media from prostate cancer cell lines to examine regulation of ASPN mRNA in tumour stromal cells. RESULTS: We observed increased expression of ASPN mRNA in a data set derived from benign vs tumour microdissected tissue, and a correlation with biochemical recurrence using Kaplan-Meier and Cox proportional hazard analysis. ASPN protein localised to tumour stroma and elevated expression of ASPN was correlated with decreased time to biochemical recurrence, in a cohort of 326 patients with a median follow up of 9.6 years. Univariate and multivariate analysis demonstrated that ASPN was correlated with progression, as were Gleason score, and clinical stage. Additionally, ASPN expression correlated with the presence of reactive stroma, suggesting that it may be a stromal marker expressed in response to the presence of tumour cells and particularly with aggressive tumour subtypes. We observed expression of ASPN in the stroma of tumours induced by p53 inhibition in a mouse model of prostate cancer, and correlation with neuroendocrine marker expression. Finally, we demonstrated that ASPN transcript expression in normal and cancer fibroblasts was regulated by conditioned media derived from the PC3, but not LNCaP, prostate cancer cell lines. CONCLUSIONS: Our results suggest that ASPN is a stromally expressed biomarker that correlates with disease progression, and is observed in reactive stroma. ASPN expression in stroma may be part of a stromal response to aggressive tumour subtypes.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fetus/pathology , Fibroblasts/pathology , Neoplasm Recurrence, Local/pathology , Prostate/pathology , Prostatic Neoplasms/pathology , Stromal Cells/pathology , Adult , Aged , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cells, Cultured , Cohort Studies , Culture Media, Conditioned/pharmacology , Extracellular Matrix Proteins/genetics , Fetus/metabolism , Fibroblasts/metabolism , Follow-Up Studies , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Knockout , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Staging , Prognosis , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinoblastoma Protein/physiology , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Survival Rate , Tumor Microenvironment , Tumor Suppressor Protein p53/physiology
7.
Mol Cell Endocrinol ; 439: 261-272, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27634452

ABSTRACT

Androgen receptor (AR) signalling in fibroblasts is important in prostate development and carcinogenesis, and is inversely related to prostate cancer mortality. However, the molecular mechanisms of AR action in fibroblasts and other non-epithelial cell types are largely unknown. The genome-wide DNA binding profile of AR in human prostate fibroblasts was identified by chromatin immunoprecipitation sequencing (ChIP-Seq), and found to be common to other fibroblast lines but disparate from AR cistromes of prostate cancer cells and tissue. Although AR binding sites specific to fibroblasts were less well conserved evolutionarily than those shared with cancer epithelia, they were likewise correlated with androgen regulation of fibroblast gene expression. Whereas FOXA1 is the key pioneer factor of AR in cancer epithelia, our data indicated that AP-1 likely plays a more important role in the AR cistrome in fibroblasts. The specificity of AP-1 and FOXA1 to binding in these cells is demonstrated using immunoblot and immunohistochemistry. Importantly, we find the fibroblast cistrome is represented in whole tissue/in vivo ChIP-seq studies at both genomic and resulting protein levels, highlighting the importance of the stroma in whole tissue -omic studies. This is the first nuclear receptor ChIP-seq study in prostatic fibroblasts, and provides novel insight into the action of fibroblast AR in prostate cancer.


Subject(s)
Cell Lineage , Fibroblasts/metabolism , Prostate/cytology , Receptors, Androgen/genetics , Transcription Factor AP-1/metabolism , Base Sequence , Cell Line , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Male , Protein Binding , Receptors, Androgen/metabolism , Reproducibility of Results , Telomerase/metabolism
8.
Cancers (Basel) ; 8(9)2016 Aug 24.
Article in English | MEDLINE | ID: mdl-27563925

ABSTRACT

BACKGROUND: Prostate cancer-associated fibroblasts (CAF) can stimulate malignant progression and invasion of prostatic tumour cells via several mechanisms including those active in extracellular matrix; METHODS: We isolated CAF from prostate cancer patients of Gleason Score 6-10 and confirmed their cancer-promoting activity using an in vivo tumour reconstitution assay comprised of CAF and BPH1 cells. We tested the effects of heat shock protein 90 (HSP90) inhibitors upon reconstituted tumour growth in vivo. Additionally, CAF contractility was measured in a 3D collagen contraction assay and migration was measured by scratch assay; RESULTS: HSP90 inhibitors dipalmitoyl-radicicol and 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG) reduced tumour size and proliferation in CAF/BPH1 reconstituted tumours in vivo. We observed that the most contractile CAF were derived from patients with lower Gleason Score and of younger age compared with the least contractile CAF. HSP90 inhibitors radicicol and 17-DMAG inhibited contractility and reduced the migration of CAF in scratch assays. Intracellular levels of HSP70 and HSP90 were upregulated upon treatment with HSP90 inhibitors. Inhibition of HSP90 also led to a specific increase in transforming growth factor beta 2 (TGFß2) levels in CAF; CONCLUSIONS: We suggest that HSP90 inhibitors act not only upon tumour cells, but also on CAF in the tumour microenvironment.

9.
Dis Model Mech ; 6(2): 530-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23136397

ABSTRACT

Human prostatic cancer-associated fibroblasts (CAFs) can elicit malignant changes in initiated but non-tumorigenic human prostate epithelium, demonstrating that they possess pro-tumorigenic properties. We set out to reduce the pro-tumorigenic activity of patient CAFs using the Dlk1 and SCUBE1 molecules that we had previously identified in prostate development. Our hypothesis was that mesenchymally expressed molecules might reduce CAF pro-tumorigenic activity, either directly or indirectly. We isolated primary prostatic CAFs and characterised their expression of CAF markers, expression of Notch2, Dlk1 and SCUBE1 transcripts, and confirmed their ability to stimulate BPH1 epithelial cell proliferation. Next, we expressed Dlk1 or SCUBE1 in CAFs and determined their effects upon tumorigenesis in vivo following recombination with BPH1 epithelia and xenografting in SCID mice. Tumour size was reduced by about 75% and BPH1 proliferation was reduced by about 50% after expression of Dlk1 or SCUBE1 in CAFs, and there was also a reduction in invasion of BPH1 epithelia into the host kidney. Inhibition of Notch signalling, using inhibitor XIX, led to a reduction in BPH1 cell proliferation in CAF-BPH1 co-cultures, whereas inhibition of Dlk1 in NIH3T3-conditioned media led to an increase in BPH1 growth. Our results suggest that pro-tumorigenic CAF activity can be reduced by the expression of developmental pathways.


Subject(s)
Cell Transformation, Neoplastic/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Prostatic Neoplasms/pathology , Animals , Biomarkers, Tumor/metabolism , Calcium-Binding Proteins , Cell Proliferation , Cell Separation , Cell Transformation, Neoplastic/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Mice , Mice, SCID , NIH 3T3 Cells , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Signal Transduction
10.
PLoS One ; 7(8): e42516, 2012.
Article in English | MEDLINE | ID: mdl-22880013

ABSTRACT

BACKGROUND AND AIM: During prostate development, mesenchymal-epithelial interactions regulate organ growth and differentiation. In adult prostate, stromal-epithelial interactions are important for tissue homeostasis and also play a significant role in prostate cancer. In this study we have identified molecules that show a mesenchymal expression pattern in the developing prostate, and one of these showed reduced expression in prostate cancer stroma. METHODOLOGY AND PRINCIPAL FINDINGS: Five candidate molecules identified by transcript profiling of developmental prostate mesenchyme were selected using a wholemount in situ hybridisation screen and studied Decorin (Dcn), Semaphorin6D (Sema6D), SPARC/Osteonectin (SPARC), Sprouty1 (Spry-1) and Tsukushi (Tsku). Expression in rat tissues was evaluated using wholemount in situ hybridisation (postnatal day (P) 0.5) and immunohistochemistry (embryonic day (E) E17.5, E19.5; P0.5; P6; 28 & adult). Four candidates (Decorin, SPARC, Spry-1, Tsukushi) were immunolocalised in human foetal prostate (weeks 14, 16, 19) and expression of Decorin was evaluated on a human prostate cancer tissue microarray. In embryonic and perinatal rats Decorin, Semaphorin6D, SPARC, Spry-1 and Tsukushi were expressed with varying distribution patterns throughout the mesenchyme at E17.5, E19.5, P0.5 and P6.5. In P28 and adult prostates there was either a decrease in the expression (Semaphorin6D) or a switch to epithelial expression of SPARC, and Spry-1, whereas Decorin and Tsukushi were specific to mesenchyme/stroma at all ages. Expression of Decorin, SPARC, Spry-1 and Tsukushi in human foetal prostates paralleled that in rat. Decorin showed mesenchymal and stromal-specific expression at all ages and was further examined in prostate cancer, where stromal expression was significantly reduced compared with non-malignant prostate. CONCLUSION AND SIGNIFICANCE: We describe the spatio-temporal expression of Decorin, Semaphorin6D, SPARC, Spry-1 and Tsukushi in developing prostate and observed similar mesenchymal expression patterns in rat and human. Additionally, Decorin showed reduced expression in prostate cancer stroma compared to non-malignant prostate stroma.


Subject(s)
Decorin/metabolism , Membrane Proteins/metabolism , Osteonectin/metabolism , Prostate/embryology , Prostatic Neoplasms/metabolism , Proteoglycans/metabolism , Semaphorins/metabolism , Animals , Decorin/genetics , Fetus/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Humans , Male , Membrane Proteins/genetics , Mesoderm/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Osteonectin/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proteoglycans/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Semaphorins/genetics , Stromal Cells/metabolism
11.
Prostate ; 71(3): 305-17, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-20812209

ABSTRACT

BACKGROUND: Androgens and paracrine signaling from mesenchyme/stroma regulate development and disease of the prostate, and gene profiling studies of inductive prostate mesenchyme have identified candidate molecules such as pleiotrophin (Ptn). METHODS: Ptn transcripts and protein were localized by in situ and immunohistochemistry and Ptn mRNA was quantitated by Northern blot and qRT-PCR. Ptn function was examined by addition of hPTN protein to rat ventral prostate organ cultures, primary human fetal prostate fibroblasts, prostate cancer associated fibroblasts, and BPH1 epithelia. RESULTS: During development, Ptn transcripts and protein were expressed in ventral mesenchymal pad (VMP) and prostatic mesenchyme. Ptn was localized to mesenchyme surrounding ductal epithelial tips undergoing branching morphogenesis, and was located on the surface of epithelia. hPTN protein stimulated branching morphogenesis and stromal and epithelial proliferation, when added to rat VP cultures, and also stimulated growth of fetal human prostate fibroblasts, prostate cancer associated fibroblasts, and BPH1 epithelia. PTN mRNA was enriched in patient-matched normal prostate fibroblasts versus prostate cancer associated fibroblasts. PTN also showed male enriched expression in fetal human male urethra versus female, and between wt male and ARKO male mice. Transcripts for PTN were upregulated by testosterone in fetal human prostate fibroblasts and organ cultures of female rat VMP. Ptn protein was increased by testosterone in organ cultures of female rat VMP and in rat male urethra compared to female. CONCLUSIONS: Our data suggest that in the prostate Ptn functions as a regulator of both mesenchymal and epithelial proliferation, and that androgens regulate Ptn levels.


Subject(s)
Carrier Proteins/physiology , Cytokines/physiology , Epithelial Cells/physiology , Fibroblasts/physiology , Mesoderm/cytology , Prostate/growth & development , Prostatic Neoplasms/pathology , Testosterone/pharmacology , Animals , Carrier Proteins/genetics , Cell Differentiation , Cytokines/genetics , Female , Humans , Male , Mice , Paracrine Communication , Prostate/chemistry , RNA, Messenger/analysis , Receptors, Androgen/physiology , Urethra/chemistry
12.
Differentiation ; 80(2-3): 89-98, 2010.
Article in English | MEDLINE | ID: mdl-20633976

ABSTRACT

Paracrine signalling from mesenchyme to epithelium plays a key role in regulating prostate organogenesis and it is important to identify the mesenchymally expressed molecules that regulate organ growth, though currently few such molecules are known. Tyrosine kinase signalling via EphB receptors has been characterised in many developmental processes, and EphB3 mRNA expression was detected in prostate inductive mesenchyme in previous gene profiling studies. This led us to examine the expression and function of EphrinB signalling in prostate development, to determine if EphrinB ligands might function as mesenchymal paracrine regulators of prostate growth. Using PCR, wholemount in situ hybridisation, and immunohistochemistry we examined the expression of EphB receptors and EphrinB ligands in rat prostate during development to determine which showed mesenchymal expression. EphB3 and EphrinB1 transcripts and proteins were expressed in the mesenchyme of developing prostate and in female urogenital mesenchyme and smooth muscle. The function of EphrinB signalling was examined using in vitro organ culture assays of ventral prostate (VP), which were treated with EphB3-Fc and EphrinB1-Fc proteins to inhibit or augment Ephrin signalling. Addition of recombinant EphB3-Fc resulted in a significant decrease in VP organ size, while recombinant EphrinB1-Fc resulted in a significant increase in VP organ size and epithelial proliferation. Additionally, EphrinB1-Fc reduced the degree of epithelial branching in VP organs and increased ductal tip size, though without disrupting normal differentiation. We have identified expression of EphrinB1 in prostatic mesenchyme and suggest that the EphrinB signalling system acts as a regulator of prostate growth. EphrinB-EphB signalling may function as an autocrine regulator of mesenchyme and/or as a paracrine regulator of epithelia.


Subject(s)
Ephrin-B1/genetics , Mesoderm/embryology , Prostate/embryology , Receptors, Eph Family/metabolism , Signal Transduction , Animals , Base Sequence , DNA Primers , Ephrin-B1/metabolism , Immunohistochemistry , In Situ Hybridization , Ligands , Male , Mesoderm/metabolism , Paraffin Embedding , Polymerase Chain Reaction , Prostate/metabolism , Prostate/pathology , RNA Probes , Rats , Rats, Wistar
13.
Reproduction ; 139(2): 395-407, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19900988

ABSTRACT

In humans and domestic mammals, pivotal processes in ovary development, including primordial follicle assembly, occur prenatally. These events are essential for determining fertility in adult life; however, they remain poorly understood at the mechanistic level. In mammals, the SLITs (SLIT1, SLIT2 and SLIT3) and their ROBO (ROBO1, ROBO2, ROBO3/RIG-1 and ROBO4/MAGIC ROBO) receptors regulate neural, leukocyte, vascular smooth muscle cell and endothelial cell migration. In addition, the SLIT/ROBO pathway has functional roles in embryonic development and in the adult ovary by inhibiting cell migration and promoting apoptosis. We therefore characterised follicle formation and investigated the expression and localisation of the ROBO/SLIT pathway in the ovine fetal ovary. Using RT-PCR, we identified SLIT2, SLIT3, ROBO1, ROBO2 and ROBO4 in sheep ovaries harvested across gestation. The real-time quantitative PCR results implied that ROBO2 expression and ROBO4 expression were elevated during the early stages of follicle formation and stayed abundant during primordial follicle maturation (P<0.05). Immunohistochemistry examination demonstrated that ROBO1 was localised to the pre-granulosa cells, while ROBO2, ROBO4 and SLIT2 were expressed in the oocytes of the developing primordial follicle. This indicates that in the fetal ovary, SLIT-ROBO signalling may require an autocrine and paracrine interaction. Furthermore, at the time of increased SLIT-ROBO expression, there was a significant reduction in the number of proliferating oocytes in the developing ovary (P<0.0001). Overall, these results suggest, for the first time, that the SLIT-ROBO pathway is expressed at the time of follicle formation during fetal ovary development.


Subject(s)
Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Ovarian Follicle/metabolism , Ovary/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Animals , Autocrine Communication , Cell Proliferation , Female , Fertility , Gene Expression Regulation, Developmental , Gestational Age , Glycoproteins/genetics , Immunohistochemistry , Nerve Tissue Proteins/genetics , Ovarian Follicle/embryology , Ovary/embryology , Paracrine Communication , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Signal Transduction/genetics , Roundabout Proteins
14.
Endocrinology ; 150(1): 463-72, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18801907

ABSTRACT

Notch1 signaling is involved in epithelial growth and differentiation of prostate epithelia, and we have examined the role that notch signaling plays in the stroma of the developing prostate. We initially observed expression of delta-like 1 (Dlk1) and Notch2 in gene profiling studies of prostatic mesenchyme, and anticipated that they might be expressed in a key subset of inductive mesenchyme. Using quantitative RT-PCR, Northern blotting, and whole mount in situ hybridization, we confirmed that both Dlk1 and Notch2 mRNAs showed a restricted expression pattern within subsets of the stroma during prostate development. Localization of Dlk1 and Notch2 proteins mirrored the transcript expression, and showed both distinct and overlapping expression patterns within the stroma. Dlk1 and Notch2 were coexpressed in condensed inductive mesenchyme of the ventral mesenchymal pad (VMP), and were partially colocalized in the smooth muscle (SM) layer of the urethral stroma. In addition, Dlk1 was not expressed in SM adjacent to the VMP in female urethra. The function of notch signaling was examined using organ cultures of prostate rudiments and a small molecule inhibitor of notch receptor activity. Inhibition of notch signaling led to a loss of stromal tissue in both prostate and female VMP cultures, suggesting that this pathway was required for stromal survival. Inhibition of notch signaling also led to changes in both epithelial and stromal differentiation, which was evident in altered distributions of SM alpha-actin and p63 in prostates grown in vitro. The effects of notch signaling upon the stroma were only evident in the presence of testosterone, in contrast to effects upon epithelial differentiation.


Subject(s)
Prostate/growth & development , Receptor, Notch2/genetics , Stromal Cells/cytology , Animals , Cell Differentiation , Female , Gene Expression Regulation , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Organ Culture Techniques , Prostate/cytology , Prostate/physiology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Urethra/growth & development , Urethra/physiology , Urinary Bladder/growth & development , Urinary Bladder/physiology
16.
Differentiation ; 76(6): 587-98, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18752494

ABSTRACT

The development of the prostate is dependent upon androgens and stromal-epithelial interactions. Understanding the molecules and mechanisms by which androgens control prostate organogenesis has been a considerable challenge over the past few decades. Similarly, identifying the molecular signals passing between stromal and epithelial cells has been difficult, and consequently understanding how androgens and stromal-epithelial signalling interact is poorly understood. There remains significant uncertainty regarding how androgens control the growth of the prostate, although several pathways have been identified that are required for prostate development or which alter prostate growth. This review will summarize past findings relating to the pathways that might mediate the effects of androgens as well as molecules that act as stromal to epithelial signals in the prostate. It will also examine the approaches used to identify pathways of importance and the historical concepts that have informed these studies. In particular, the question of which mechanisms might be involved in early prostate organogenesis as well as anatomic aspects of organ induction will be described. Finally, models of prostatic development will be proposed and discussed.


Subject(s)
Mesoderm/physiology , Organogenesis , Prostate/embryology , Prostate/growth & development , Androgens/physiology , Female , Humans , Male , Muscle, Smooth/physiology , Signal Transduction
17.
Genome Biol ; 8(10): R213, 2007.
Article in English | MEDLINE | ID: mdl-17922897

ABSTRACT

BACKGROUND: The mesenchymal compartment plays a key role in organogenesis, and cells within the mesenchyme/stroma are a source of potent molecules that control epithelia during development and tumorigenesis. We used serial analysis of gene expression (SAGE) to profile a key subset of prostatic mesenchyme that regulates prostate development and is enriched for growth-regulatory molecules. RESULTS: SAGE libraries were constructed from prostatic inductive mesenchyme and from the complete prostatic rudiment (including inductive mesenchyme, epithelium, and smooth muscle). By comparing these two SAGE libraries, we generated a list of 219 transcripts that were enriched or specific to inductive mesenchyme and that may act as mesenchymal regulators of organogenesis and tumorigenesis. We identified Scube1 as enriched in inductive mesenchyme from the list of 219 transcripts; also, quantitative RT-PCR and whole-mount in situ hybridization revealed Scube1 to exhibit a highly restricted expression pattern. The expression of Scube1 in a subset of mesenchymal cells suggests a role in prostatic induction and branching morphogenesis. Additionally, Scube1 transcripts were expressed in prostate cancer stromal cells, and were less abundant in cancer associated fibroblasts relative to matched normal prostate fibroblasts. CONCLUSION: The use of a precisely defined subset of cells and a back-comparison approach allowed us to identify rare mRNAs that could be overlooked using other approaches. We propose that Scube1 encodes a novel stromal molecule that is involved in prostate development and tumorigenesis.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Mesoderm/metabolism , Prostate/growth & development , Animals , Carrier Proteins/genetics , Female , Gene Library , In Situ Hybridization , Male , Membrane Proteins/genetics , Prostate/cytology , Prostate/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
18.
Differentiation ; 74(7): 382-92, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16916376

ABSTRACT

The prostate gland and seminal vesicles are the major exocrine glands in the male reproductive tracts of mammals. Although the morphology of these organs varies widely among species, epithelial branching morphogenesis is a key feature of organ development in most mammals including rodents and humans. Insight into the mechanisms that control prostatic and seminal vesicle branching morphogenesis has come from experimental embryological work as well as from the study of mice and humans harboring mutations that alter branching morphogenesis. These studies have demonstrated a requirement for androgens to initiate branching morphogenesis as well as a role for androgens in sustaining the normal rate and extent of branching. In addition, these studies have revealed a series of reciprocal paracrine signals between the developing prostatic epithelium and prostatic mesenchyme that are essential for regulating branching morphogenesis. Key growth factors that participate in these signaling events include members of the fibroblast growth factor, Hedgehog, and transforming growth factor-beta families. Additional genes including several homeobox-containing transcription factors have also been implicated as key regulators of prostatic and seminal vesicle branching morphogenesis. While research in recent years has greatly enhanced our understanding of the molecular control of prostatic and seminal vesicle development, known genes cannot yet explain in molecular terms the complex biological interactions that descriptive and experimental embryological studies have elucidated in the control of branching morphogenesis in these organs.


Subject(s)
Morphogenesis , Prostate/growth & development , Seminal Vesicles/growth & development , Animals , Humans , Male , Steroids/physiology
19.
Prostate ; 66(7): 696-707, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16425200

ABSTRACT

BACKGROUND: Smooth muscle (SM) has been proposed to play an important role in controlling prostate organogenesis by regulating signaling between inductive mesenchyme and developing epithelial prostatic buds. METHODS: We have examined the effects of testosterone and estrogen upon SM patterning in the embryonic rat urogenital tract (UGT) using in vitro organ cultures, immunohistochemistry, and Western blotting. RESULTS: We observed that testosterone elicited a sexually dimorphic difference in SM structure of embryonic UGTs, in cultures grown with testosterone. The addition of estrogen led to an increase in the rate of SM closure, in both males and females. To quantify the effects of steroids upon SM we used Western blotting of SM actin, which showed that estrogen stimulated SM content, while testosterone reduced SM content. Finally, we examined the expression of ERalpha, ERbeta, PR, and SM actin under different hormonal treatments of UGTs grown in vitro. The expression patterns of ERalpha and ERbeta were largely unchanged by hormonal treatment, while PR showed a much broader expression pattern in response to estradiol. CONCLUSIONS: Our results indicate that testosterone can directly regulate SM patterning and content in the UGT, and that SM is sensitive to both androgens and estrogens.


Subject(s)
Estradiol/physiology , Muscle, Smooth/embryology , Prostate/embryology , Testosterone/physiology , Animals , Blotting, Western , Humans , Immunohistochemistry , Male , Rats , Rats, Wistar , Receptors, Estrogen/physiology , Sex Characteristics
20.
Neuroendocrinology ; 84(5): 285-300, 2006.
Article in English | MEDLINE | ID: mdl-17202804

ABSTRACT

Gonadotropin-releasing hormone (GnRH) analogs constitute the most widely employed medical treatment for prostatic cancer. The predominant mechanism of action is presumed to be via the inhibition of gonadotropins and resultant decrease in androgen. However, GnRH analogs have also been shown to directly inhibit prostate cancer cells both in vitro and in vivo through antiproliferative cell cycle arrest and stimulation of apoptosis. Since the GnRH receptor has been shown to affect sex steroid hormone receptor function, we considered that part of GnRH analog actions on prostate cells may be mediated through modulation of the human androgen receptor. Using a model HEK293 cell line expressing the GnRH receptor, we demonstrated a novel signalling pathway of the GnRH receptor that induces nuclear translocation of the androgen receptor that renders it transcriptionally inactive. This mechanism involves the calcium-dependent tyrosine kinase Pyk2, the non-receptor tyrosine kinase c-Src and the focal adhesion protein/steroid receptor co-factor, Hic-5. In this setting there is a GnRH-induced association and nuclear translocation of the androgen receptor with Hic-5. GnRH-induced Pyk2 activation opposed the association of Hic-5 with androgen receptor as overexpression of a dominant negative Pyk2 enhanced the GnRH-induced nuclear translocation of a green fluorescent protein-tagged human androgen receptor. GnRH-induced c-Src activation resulted in the phosphorylation of expressed Hic-5 and promoted its association with the human androgen receptor. In contrast to testosterone, GnRH-induced nuclear translocation did not transcriptionally activate the androgen receptor. We then demonstrated that GnRH can also stimulate androgen receptor mobilization in human prostate PC3, BPH-1 and LNCaP cells, and in cultured rat ventral prostate cells through the same mechanism. To determine if GnRH could antagonize androgen effects in normal tissue, we examined the effect of GnRH on rat ventral prostate organ cultures and demonstrated that GnRH can functionally antagonize the actions of testosterone on prostate cell proliferation and tissue growth. This antagonism of testosterone action by GnRH may underlie in part the capacity of GnRH receptor activation to inhibit prostate tumor growth.


Subject(s)
Androgen Receptor Antagonists , Focal Adhesion Protein-Tyrosine Kinases/physiology , Gonadotropin-Releasing Hormone/pharmacology , Intracellular Signaling Peptides and Proteins/physiology , Prostate/drug effects , Testosterone/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Animals, Newborn , CSK Tyrosine-Protein Kinase , Cell Nucleus/metabolism , Cells, Cultured , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/chemistry , Focal Adhesions/physiology , Green Fluorescent Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins , Male , Prostate/growth & development , Prostate/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Wistar , Receptors, Androgen/metabolism , Transcriptional Activation/drug effects , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...