Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Gene ; 506(2): 392-5, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22766399

ABSTRACT

BACKGROUND: Epispadias is the mildest phenotype of the human bladder exstrophy-epispadias complex (BEEC), and presents with varying degrees of severity. This urogenital birth defect results from a disturbance in the septation process, during which separate urogenital and anorectal components are formed through division of the cloaca. This process is reported to be influenced by androgen signaling. The human PARM1 gene encodes the prostate androgen-regulated mucin-like protein 1, which is expressed in heart, kidney, and placenta. METHODS: We performed whole mount in situ hybridization analysis of Parm1 expression in mouse embryos between gestational days (GD) 9.5 and 12.5, which are equivalent to human gestational weeks 4-6. Since the spatio-temporal localization of Parm1 corresponded to tissues which are affected in human epispadias, we sequenced PARM1 in 24 affected patients. RESULTS: We found Parm1 specifically expressed in the region of the developing cloaca, the umbilical cord, bladder anlage, and the urethral component of the genital tubercle. Additionally, Parm1 expression was detected in the muscle progenitor cells of the somites and head mesenchyme. PARM1 gene analysis revealed no alterations in the coding region of any of the investigated patients. CONCLUSIONS: These findings suggest that PARM1 does not play a major role in the development of human epispadias. However, we cannot rule out the possibility that a larger sample size would enable detection of rare mutations in this gene.


Subject(s)
Androgen-Binding Protein/biosynthesis , Androgen-Binding Protein/genetics , Epispadias/genetics , Epispadias/metabolism , Gene Expression Regulation, Developmental , Prostate/metabolism , Animals , DNA Mutational Analysis , Female , Humans , In Situ Hybridization , Male , Mice , Phenotype , Sequence Analysis, DNA , Time Factors , Tissue Distribution
2.
Mol Pharmacol ; 79(4): 710-23, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21252290

ABSTRACT

To identify novel factors or mechanisms that are important for the resistance of tissues to chemical toxicity, we have determined the mechanisms underlying the previously observed increases in resistance to acetaminophen (APAP) toxicity in the lateral nasal gland (LNG) of the male Cyp2g1-null/Cyp2a5-low mouse. Initial studies established that Cyp2a5-null mice, but not a newly generated strain of Cyp2g1-null mice, were resistant to APAP toxicity in the LNG; therefore, subsequent studies were focused on the Cyp2a5-null mice. Compared with the wild-type (WT) male mouse, the Cyp2a5-null male mouse had intact capability to metabolize APAP to reactive intermediates in the LNG, as well as unaltered circulating levels of APAP, APAP-GSH, APAP-glucuronide, and APAP-sulfate. However, it displayed reduced tissue levels of APAP and APAP-GSH and increased tissue levels of testosterone and salivary androgen-binding protein (ABP) in the LNG. Furthermore, we found that ABP was able to compete with GSH and cellular proteins for adduction with reactive metabolites of APAP in vitro. The amounts of APAP-ABP adducts formed in vivo were greater, whereas the amounts of APAP adducts formed with other cellular proteins were substantially lower, in the LNG of APAP-treated male Cyp2a5-null mice compared with the LNG of APAP-treated male WT mice. We propose that through its critical role in testosterone metabolism, CYP2A5 regulates 1) the bioavailability of APAP and APAP-GSH (presumably through modulation of the rates of xenobiotic excretion from the LNG) and 2) the expression of ABP, which can quench reactive APAP metabolites and thereby spare critical cellular proteins from inactivation.


Subject(s)
Acetaminophen/toxicity , Androgen-Binding Protein/biosynthesis , Aryl Hydrocarbon Hydroxylases/physiology , Nasal Mucosa/metabolism , Salivary Proteins and Peptides/biosynthesis , Testosterone/physiology , Acetaminophen/pharmacokinetics , Animals , Biological Availability , Cytochrome P-450 CYP2A6 , Cytochrome P450 Family 2 , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Homeostasis/drug effects , Homeostasis/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nasal Mucosa/drug effects , Nasal Mucosa/enzymology
3.
J Mol Biol ; 385(2): 446-56, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19013180

ABSTRACT

Androgen receptor (AR) is a ligand-dependent transcription factor and its activity is regulated by numerous AR coregulators. Aberrant expression of AR coregulators in prostate cancer cells has an important role in the development and progression of prostate cancer. We report here that CDC25A, a cell cycle-promoting phosphatase over-expressed in a number of cancers, functions as an AR coregulator suppressing the AR transcriptional activity. In this study, we found that CDC25A is upregulated in human prostate cancer and its expression level is positively associated with the Gleason score and disease metastasis. More importantly, we showed that CDC25A can physically interact with AR through its putative catalytic domain. In addition, ectopic expression of CDC25A in prostate cancer cell lines suppresses PSA and Probasin promoter activities significantly, indicating that CDC25A may function as an AR corepressor. This was further confirmed by knockdown of endogenous CDC25A expression using small interfering RNA (siRNA), which resulted in upregulation of PSA promoter activity. Moreover, a truncated mutant that does not interact with AR fails to suppress the PSA promoter activity, indicating that CDC25A downregulates androgen-responsive promoter by physically interacting with AR. Taken together, our results demonstrated a novel function of CDC25A in the regulation of androgen signaling in human prostate cancer cells.


Subject(s)
Androgen Receptor Antagonists , Prostatic Neoplasms/physiopathology , Repressor Proteins/metabolism , cdc25 Phosphatases/metabolism , Androgen-Binding Protein/biosynthesis , Cell Line , Gene Knockdown Techniques , Gene Silencing , Humans , Male , Prostate-Specific Antigen/biosynthesis , Protein Interaction Domains and Motifs , Protein Interaction Mapping , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
4.
Mol Cancer ; 6: 19, 2007 Mar 07.
Article in English | MEDLINE | ID: mdl-17343742

ABSTRACT

BACKGROUND: Robust Hedgehog (Hh) signaling has been implicated as a common feature of human prostate cancer and an important stimulus of tumor growth. The role of Hh signaling has been studied in several xenograft tumor models, however, the role of Hh in tumor development in a transgenic prostate cancer model has never been examined. RESULTS: We analyzed expression of Hh pathway components and conserved Hh target genes along with progenitor cell markers and selected markers of epithelial differentiation during tumor development in the LADY transgenic mouse model. Tumor development was associated with a selective increase in Ihh expression. In contrast Shh expression was decreased. Expression of the Hh target Patched (Ptc) was significantly decreased while Gli1 expression was not significantly altered. A survey of other relevant genes revealed significant increases in expression of Notch-1 and Nestin together with decreased expression of HNF3a/FoxA1, NPDC-1 and probasin. CONCLUSION: Our study shows no evidence for a generalized increase in Hh signaling during tumor development in the LADY mouse. It does reveal a selective increase in Ihh expression that is associated with increased expression of progenitor cell markers and decreased expression of terminal differentiation markers. These data suggest that Ihh expression may be a feature of a progenitor cell population that is involved in tumor development.


Subject(s)
Adenocarcinoma/metabolism , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Hedgehog Proteins/physiology , Neoplasm Proteins/physiology , Prostatic Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Androgen-Binding Protein/biosynthesis , Androgen-Binding Protein/genetics , Animals , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/physiology , Cell Differentiation , Cell Division , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/genetics , Hepatocyte Nuclear Factor 3-alpha/analysis , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/biosynthesis , Hepatocyte Nuclear Factor 3-beta/genetics , Intermediate Filament Proteins/biosynthesis , Intermediate Filament Proteins/genetics , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Transgenic , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nestin , Patched Receptors , Patched-1 Receptor , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/physiology , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Time Factors , Zinc Finger Protein GLI1
5.
Zhonghua Nan Ke Xue ; 12(2): 104-7, 111, 2006 Feb.
Article in Chinese | MEDLINE | ID: mdl-16519141

ABSTRACT

OBJECTIVE: To explore effects of p, p'DDE on the expression of androgen binding protein (ABP), transferrin (Tf) and inhibin B (INH B) mRNA in testis Sertoli cells of Sprague Dawley rats. METHODS: A method has been set up to obtain a large number of viable Sertoli cells from SD rats of 18-20 days of age. With a series of concentration p,p'-DDE (10, 30, and 50 micromol/L) co-incubating the Sertoli cells in vitro, the expression of ABP, Tf and INH B mRNA were determined by RT-PCR. RESULTS: a) With increase of the incubated p, p'-DDE, the expression of ABP mRNA in Sertoli cells went up while that of Tf and INH B dropped in a dose-dependent manner (P < 0. 05). b) The correlation analysis among ABP, Tf and INH B showed that negative relationships were found between ABP and Tf or INH B, respectively (r = - 0. 391 3, P = 0. 032 5; r = - 0.235 2, P = 0.0158), and that positive correlation was indicated between Tf and INH B (r =0.4516, P =0.0047). CONCLUSION: p,p'-DDE is a reproductive toxicant which disrupts the transcription of ABP, Tf and INH B in rat Sertoli cells so as to result in reproductive dysfunction.


Subject(s)
Androgen-Binding Protein/biosynthesis , Dichlorodiphenyl Dichloroethylene/toxicity , Inhibins/biosynthesis , Sertoli Cells/drug effects , Transferrin/biosynthesis , Androgen-Binding Protein/genetics , Animals , Inhibins/genetics , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/metabolism , Transferrin/genetics
6.
Reprod Biol Endocrinol ; 3: 70, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16336681

ABSTRACT

BACKGROUND: Spermatogenesis is an androgen-dependent process, yet the molecular mechanisms of androgens' actions in testis are poorly understood. Transgenic mice overexpressing rat androgen-binding protein (ABP) in their testes have reduced levels of intratesticular androgens and, as a result, show a progressive impairment of spermatogenesis. We used this model to characterize changes in global gene expression in testis in response to reduced bioavailability of androgens. METHODS: Total RNA was extracted from testes of 30-day old transgenic and wild-type control mice, converted to cRNA, labeled with biotin, and hybridized to oligonucleotide microarrays. Microarray results were confirmed by real-time reverse transcription polymerase chain reaction. RESULTS: Three-hundred-eighty-one genes (3.05% of all transcripts represented on the chips) were up-regulated and 198 genes (1.59%) were down-regulated by at least a factor of 2 in the androgen-deficient animals compared to controls. Genes encoding membrane proteins, intracellular signaling molecules, enzymes, proteins participating in the immune response, and those involved in cytoskeleton organization were significantly overrepresented in the up-regulated group. Among the down-regulated transcripts, those coding for extracellular proteins were overrepresented most dramatically, followed by those related to proteolysis, cell adhesion, immune response, and growth factor, cytokine, and ion channel activities. Transcripts with the greatest potential impact on cellular activities included several transcription factors, intracellular signal transducers, secreted signaling molecules and enzymes, and various cell surface molecules. Major nodes in the up-regulated network were IL-6, AGT, MYC, and A2M, those in the down-regulated network were IL-2, -4, and -10, MAPK8, SOCS1, and CREB1. CONCLUSION: Microarray analysis followed by gene ontology profiling and connectivity analysis identified several functional groups of genes and individual genes responding to sustained reduction of androgen levels in the mouse testis. These include genes whose products function as transcription factors, cell surface molecules including ion channels, extra- and intracellular signaling molecules, and secreted enzymes with the potential of regulating cell-to-cell attachment. The transcription factors CREB1 (down-regulated) and MYC (up-regulated) may mediate the most important initial phases of the testicular response to reduced levels of androgens. These results suggest specific avenues for further research that will lead to a better understanding of how androgens regulate spermatogenesis.


Subject(s)
Androgen-Binding Protein/genetics , Gene Expression Regulation , Spermatogenesis/physiology , Testis/metabolism , Androgen-Binding Protein/biosynthesis , Androgens/deficiency , Animals , Down-Regulation , Gene Expression Profiling , Male , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis/methods , Rats , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
7.
Clin Cancer Res ; 11(20): 7392-7, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16243812

ABSTRACT

PURPOSE: Raf-1 kinase inhibitor protein (RKIP) was originally identified as the first physiologic inhibitor of the Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (ERK) pathway. This pathway regulates fundamental cellular functions, including those that are subverted in cancer cells, such as proliferation, transformation, survival, and metastasis. Recently, RKIP has been recognized as a strong candidate for a metastasis suppressor gene in cell and animal model systems. Therefore, we investigated whether RKIP expression is altered in clinical specimens of human primary breast cancers and their lymph node metastases. EXPERIMENTAL DESIGN: Paraffin-embedded tumor samples from 103 breast cancer patients were examined immunohistochemically for the expression of RKIP, activated ERK, and apoptosis. The specificity of the antibodies used was validated by competition experiments with purified recombinant RKIP protein. RESULTS: RKIP expression was high in breast duct epithelia and retained to varying degrees in primary breast tumors. However, in lymph node metastases, RKIP expression was highly significantly reduced or lost (P = 0.000003). No significant correlations were observed between RKIP expression and histologic type, tumor differentiation grade, size, or estrogen receptor status. CONCLUSION: This is the first study of RKIP expression in a large clinical cohort. It confirms the results of cell culture and animal studies, suggesting that in human breast cancer, RKIP is a metastasis suppressor gene whose expression must be down-regulated for metastases to develop. RKIP expression is independent of other markers for breast cancer progression and prognosis.


Subject(s)
Androgen-Binding Protein/biosynthesis , Breast Neoplasms/pathology , Apoptosis , Breast Neoplasms/metabolism , Cell Line, Tumor , Cohort Studies , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Phosphatidylethanolamine Binding Protein , Phosphorylation
8.
Zhonghua Nan Ke Xue ; 10(10): 767-70, 2004 Oct.
Article in Chinese | MEDLINE | ID: mdl-15562793

ABSTRACT

Diabetes is a metabolic disease caused by complicated factors, and its damage to the male reproductive system is threatening men's health. This article reviews the pathophysiological changes in the diabetes-damaged male reproductive system and the mechanism of these changes. Oxidative stress induced by hyperglycemia plays an important role in working damage to the reproductive system of diabetic males, for which some anti-oxidative substances may prove to be an effective cure.


Subject(s)
Diabetes Mellitus/physiopathology , Testis/pathology , Androgen-Binding Protein/biosynthesis , Animals , Diabetes Mellitus/pathology , Free Radicals , Humans , Leydig Cells/metabolism , Male , Oxidative Stress , Rats , Sertoli Cells/metabolism , Testosterone/biosynthesis
9.
Cancer Res ; 64(19): 7117-26, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466208

ABSTRACT

Rituximab (Rituxan, IDEC-C2B8) has been shown to sensitize non-Hodgkin's lymphoma (NHL) cell lines to chemotherapeutic drug-induced apoptosis. Rituximab treatment of Bcl-2-deficient Ramos cells and Bcl-2-expressing Daudi cells selectively decreases Bcl-(xL) expression and sensitizes the cells to paclitaxel-induced apoptosis. This study delineates the signaling pathway involved in rituximab-mediated Bcl-(xL) down-regulation in Ramos and Daudi NHL B cells. We hypothesized that rituximab may interfere with the extracellular signal-regulated kinase (ERK) 1/2 pathway, leading to decreased Bcl-(xL) expression. Rituximab (20 microg/mL) inhibited the kinase activity of mitogen-activated protein kinase kinase (MEK) 1/2 and reduced the phosphorylation of the components of the ERK1/2 pathway (Raf-1, MEK1/2, and ERK1/2) and decreased activator protein-1 DNA binding activity and Bcl-(xL) gene expression. These events occurred with similar kinetics and were observed 3 to 6 hours after rituximab treatment. Rituximab-mediated effects were corroborated by using specific inhibitors of the ERK1/2 pathway, which also reduced Bcl-(xL) levels and sensitized the NHL B cells to paclitaxel-induced apoptosis. Previous findings implicated a negative regulatory role of the Raf-1 kinase inhibitor protein (RKIP) on the ERK1/2 pathway. Rituximab treatment of NHL B cells significantly up-regulated RKIP expression, thus interrupting the ERK1/2 signaling pathway through the physical association between Raf-1 and RKIP, which was concomitant with Bcl-(xL) down-regulation. These novel findings reveal a signaling pathway triggered by rituximab, whereby rituximab-mediated up-regulation of RKIP adversely regulates the activity of the ERK1/2 pathway, Bcl-(xL) expression, and subsequent chemosensitization of drug-refractory NHL B cells. The significance of these findings is discussed.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , B-Lymphocytes/drug effects , Lymphoma, Non-Hodgkin/drug therapy , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Amino Acid Sequence , Androgen-Binding Protein/biosynthesis , Androgen-Binding Protein/genetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Apoptosis/drug effects , B-Lymphocytes/enzymology , B-Lymphocytes/pathology , Cell Line, Tumor , Down-Regulation/drug effects , Drug Synergism , Humans , Lymphoma, Non-Hodgkin/enzymology , Lymphoma, Non-Hodgkin/pathology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Phosphatidylethanolamine Binding Protein , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-raf/metabolism , Rituximab , Up-Regulation/drug effects , bcl-X Protein
10.
Anticancer Drugs ; 15(7): 663-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15269597

ABSTRACT

The metastatic cascade is a complicated process that involves many steps from gain of the metastatic phenotype in the primary tumor cells through establishment of macroscopic tumor at the distant target organ. A group of genes, termed metastasis suppressor genes (MSG), encode for proteins that inhibit various steps of the metastatic cascade. Accordingly, loss of MSG promotes the metastatic phenotype. Although several MSG have been identified, the mechanisms through which they enhance metastasis are not clearly defined. Gene array analysis of a low metastatic LNCaP prostate cancer cell line compared to its highly metastatic derivative C4-2B prostate cancer cell line revealed decreased expression of raf kinase inhibitor protein (RKIP) in the C4-2B cell line. RKIP blocks the activation of several signaling pathways including MEK, G-proteins and NFkappaB. Immunohistochemical analysis of prostate cancer primary tumors and metastases revealed that RKIP protein expression was decreased in metastases. Restoration of RKIP expression in the C4-2B cell line diminished metastasis in a murine model. These results demonstrate that RKIP is a MSG. Loss of RKIP enhanced both angiogenesis and vascular invasion, and protected against apoptosis. These findings suggest that targeting the RKIP pathway may diminish the metastatic cascade. However, challenges exist as to the best method to target RKIP expression. Restoration of RKIP expression in all cancer cells in vivo is challenging. A plausible strategy is to use small molecules that target proteins in signaling pathways that are dysregulated due to loss of RKIP.


Subject(s)
Androgen-Binding Protein/genetics , Genes, Tumor Suppressor , Prostatic Neoplasms/genetics , Androgen-Binding Protein/biosynthesis , Animals , Humans , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphatidylethanolamine Binding Protein , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
11.
Prostate ; 59(4): 370-82, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15065085

ABSTRACT

BACKGROUND: Lentiviruses are retroviruses that can infect and stably integrate into the chromatin of non-dividing cells. The purpose of this study was to determine whether lentiviral vectors containing the probasin (PB) promoter displayed prostate-specific, androgen-regulated, and persistent gene expression. METHODS: Three lentiviral-PB promoter/enhanced green fluorescent protein (EGFP)-reporter vectors together with a control lentiviral-CMV-EGFP, were tested by microscopy and flowcytometry for expression of EGFP after infection of human prostate cancer cells (LNCaP, PC-3, PC-3(hAR), and Du145 cells) and non-prostate cells (COS-1, HeLa, HeLa(hAR), and MCF-7 cells). RESULTS: All cells infected in vitro with lentiviral-CMV vectors expressed EGFP, whereas with lentiviral-PB vectors (the most potent being Lv-ARR(2)PB), reporter expression was only observed in LNCaP cells with a small amount seen in androgen-independent PC-3 cells. Stable or transient transfection of androgen receptor only raised EGFP expression in prostate-derived cell lines, but did not change tumor specificity. With Lv-ARR(2)PB infected LNCaP cells, androgens regulated EGFP both in vitro and in vivo. After intra-tumor injection of this vector, EGFP expression was observed in LNCaP tumors, but not in A-549 lung or CaKi-2 kidney tumors. CONCLUSIONS: Lv-ARR(2)PB may be an ideal vector for prostate-tumor targeting and for persistent, hormone-enhanced expression of a therapeutic gene to treat slow growing prostate tumors.


Subject(s)
Androgen-Binding Protein/biosynthesis , Androgen-Binding Protein/genetics , Gene Expression Regulation, Neoplastic , Lentivirus/genetics , Prostatic Neoplasms/genetics , Genetic Therapy , Genetic Vectors , Green Fluorescent Proteins , Humans , Lentivirus/pathogenicity , Luminescent Proteins/biosynthesis , Male , Promoter Regions, Genetic , Tumor Cells, Cultured
12.
Prostate ; 58(1): 1-12, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14673947

ABSTRACT

BACKGROUND: Ectopic expression of fibroblast growth factor receptor 1 (FGFR1) tyrosine kinase in epithelial cells is associated with progression of prostate cancer. Ectopic expression by transfection of FGFR1 in premalignant epithelial cells from nonmalignant Dunning tumors accelerated time-dependent progression of epithelial cells to malignancy. This study was designed to test the effect of chronic androgen-dependent ectopic activity of FGFR1 in the normal adult mouse epithelium by gene targeting. MATERIALS AND METHODS: Constitutively active FGFR1 (caFGFR1) was targeted to prostate epithelial cells using the androgen-dependent probasin (PB) promoter. Prostate tissues of three strains were characterized over a period of 2 years by HE staining, immunohistochemical analyses for cytokeratin and alpha-actin, and rate of androgen-induced regeneration after castration. RESULTS: Relative to wildtype littermates, transgenic mice showed increased overall size, hyperplasia in epithelial, and, to a lesser extent, stromal compartments and nuclear atypia in epithelial cells of the prostate with increasing age. Androgen-induced regeneration after castration was enhanced at day 3 by two-fold in mice expressing ectopic caFGFR1. CONCLUSIONS: The ectopic presence and chronic activation of FGFR1 in mouse prostate epithelial cells induces progressive prostate intraepithelial neoplasia. These results confirm results suggested by the transplantable Dunning tumor and cell culture models that, in contrast to homeostasis-promoting resident FGFR2, chronic ectopic FGFR1 kinase activity in the epithelium disrupts homeostasis between stroma and epithelium. Although insufficient alone, it may cooperate with other oncogenic changes to promote epithelial cells down the path to malignancy.


Subject(s)
Prostatic Hyperplasia/enzymology , Prostatic Intraepithelial Neoplasia/enzymology , Prostatic Neoplasms/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Actins/biosynthesis , Androgen-Binding Protein/biosynthesis , Androgen-Binding Protein/metabolism , Animals , Epithelial Cells , Female , Immunohistochemistry , Keratins/metabolism , Male , Mice , Mice, Transgenic , Orchiectomy , Prostatic Hyperplasia/pathology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 1 , Testosterone/metabolism
13.
J Endocrinol Invest ; 26(8): 718-22, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14669824

ABSTRACT

Direct effects of PRL on Sertoli cell proliferation were investigated by using Sertoli cell primary cultures isolated from both prepubertal rat and porcine testes. PRL metabolic effects were analyzed in rat Sertoli cell primary cultures. Exposure to physiological doses of PRL resulted in a significant increase (+50-60%) of basal DNA synthesis, as reflected by the pattern of [3H] thymidine incorporation during culture; significant increases in lactate secretion (about 50%), androgen binding protein (ABP) production (about 30%) and basal protein synthesis (25-30%), as reflected in the augmented [14C] valine incorporation, were also evident. Taken together, our present findings, indicating significant effects of PRL on Sertoli cell proliferation and metabolism, demonstrate that Sertoli cells are a potential target for PRL action at testicular level during pre-pubertal development.


Subject(s)
Prolactin/pharmacology , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sexual Maturation/physiology , Androgen-Binding Protein/biosynthesis , Animals , Cell Division/drug effects , Cells, Cultured , DNA/biosynthesis , Dose-Response Relationship, Drug , Lactic Acid/metabolism , Male , Protein Biosynthesis , Rats , Swine , Thymidine/metabolism
14.
Reprod Biol Endocrinol ; 1: 48, 2003 Jun 12.
Article in English | MEDLINE | ID: mdl-12831397

ABSTRACT

The number and type of testicular germ cells undergoing apoptosis in different age groups of mice (from 7 to 360 days of age) was determined and compared in age-matched wild type (WT) control and in a transgenic (TG) mice homozygous to rat androgen binding protein (ABP) using flow cytometry. Flow cytometric quantification revealed that the total number of germ cells undergoing apoptosis did not differ significantly in WT and TG mice up to Day 14. From Day 21 to Day 60, the number of germ cells undergoing apoptosis was consistently higher in TG than in WT mice. Starting from Day 90, the number of germ cells undergoing apoptosis in TG mice was lower than controls until Day 360. In 21-60 days old TG mice, spermatogonia, S-Phase cells, and primary spermatocytes are the cell types undergoing apoptosis at significantly greater numbers than those in WT mice. However, starting from day 60, the total number of spermatids undergoing apoptosis was significantly lower in TG mice than in age-matched WT controls. TdT-mediated dUTP-biotin nick end labeling (TUNEL) in testicular sections from TG mice of 21 and 30 days of age confirmed the presence of increased numbers of apoptotic germ cells compared to their age matched controls. These data indicate that the continuous presence of greater than physiological concentrations of ABP in the mouse testis has a biphasic effect on the frequency of apoptosis in germ cells. The initial pre-pubertal increase in testicular germ cell apoptosis may result from direct or indirect actions of ABP and is likely to determine the subsequent life-death balance of germ cell populations in TG mice, whereas the subsequent reduction may result from maturation depletion. A wave of apoptosis during the pre-pubertal period is required for normal spermatogenesis to develop, and our data indicate that this apoptotic wave may be regulated by ABP and/or androgens.


Subject(s)
Androgen-Binding Protein/physiology , Apoptosis/physiology , Spermatogenesis/physiology , Spermatozoa/cytology , Testis/cytology , Acetates/pharmacology , Age Factors , Androgen-Binding Protein/biosynthesis , Androgen-Binding Protein/genetics , Animals , Apoptosis/drug effects , Cell Division/physiology , Gene Expression , Infertility, Male/genetics , Infertility, Male/physiopathology , Male , Mice , Mice, Transgenic , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/physiology , S Phase , Sexual Maturation/physiology , Spermatozoa/drug effects , Spermatozoa/metabolism , Testis/growth & development
15.
J Immunol ; 169(9): 4761-9, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391185

ABSTRACT

In addition to their overexpression in cancer cells, most of the tumor-associated Ags are expressed at low but detectable levels in normal tissues. It is not clear whether the repertoire of T cells specific for unmutated tumor Ags is shaped by negative selection during T cell development. The transgenic adenocarcinoma of mouse prostate (TRAMP) model is transgenic for the SV40 large T Ag (Tag) under the control of the rat probasin regulatory elements. Although it has been established that T lymphocytes from TRAMP mice are tolerant to SV40 Tag, the mechanism of the tolerance is largely unknown. To examine whether the T cell clonal deletion is responsible for the tolerance, we crossed the TRAMP mice with mice transgenic for a rearranged TCR specific for SV40 Tag presented by the H-2K(k). Double transgenic TRAMP/TCR mice showed profound thymic deletion of SV40 Tag-reactive T cells, including a 6- to 10-fold reduction in the total thymocyte numbers and a >50-fold reduction in phenotypically mature T cells. Consistent with this finding, we observed that the SV40 Tag and endogenous mouse probasin genes are expressed at low levels in the thymus. These results demonstrate that clonal deletion is a major mechanism for tolerance to Ags previously regarded as prostate-specific, and provide direct evidence that the T cell repertoire specific for an unmutated tumor Ag can be shaped by clonal deletion in the thymus.


Subject(s)
Adenocarcinoma/immunology , Antigens, Neoplasm/biosynthesis , Antigens, Polyomavirus Transforming/immunology , Clonal Deletion/genetics , Epitopes, T-Lymphocyte/immunology , Mice, Transgenic/immunology , Prostatic Neoplasms/immunology , T-Lymphocyte Subsets/immunology , Adenocarcinoma/genetics , Amino Acid Sequence , Androgen-Binding Protein/biosynthesis , Animals , Antigens, Neoplasm/genetics , Antigens, Polyomavirus Transforming/biosynthesis , Antigens, Polyomavirus Transforming/genetics , Cell Line , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/genetics , Female , Immunodominant Epitopes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Molecular Sequence Data , Prostatic Neoplasms/genetics , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/virology
16.
Toxicol Sci ; 69(1): 202-9, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12215675

ABSTRACT

A single maternal dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on gestation day (GD) 13 can greatly impair ventral prostate, dorsolateral prostate, anterior prostate, and seminal vesicle development in wild-type C57BL/6 mice. The developmental stages at which these organs are most sensitive to TCDD exposure have now been investigated. Pregnant mice were dosed orally with 5 micro g TCDD/kg or vehicle on GD 13, and their pups were fostered at birth to dams of the same treatment or cross-fostered to dams of the opposite treatment. Additional males had in utero and lactational TCDD exposure following maternal dosing on GD 16. Organ weights and secretory protein, cytokeratin 8, and cyclophilin mRNA expression were determined at 35 days of age. Effects of TCDD on ventral prostate development were due primarily to in utero exposure; the critical window was between GD 13 and birth. Dorsolateral prostate development was inhibited about equally by in utero or lactational exposure, and vulnerability did not begin until GD 16. Anterior prostate development was also affected by both in utero and lactational TCDD exposure, particularly the former. Vulnerability began before GD 16 and continued into postnatal life. Seminal vesicle development was essentially unaffected by in utero or lactational exposure alone but was significantly inhibited by combined exposure, regardless of whether dams were dosed on GD 13 or 16. In summary, the time during which each organ was most vulnerable to TCDD exposure varied from one organ to another. These findings provide insights into the developmental processes that TCDD inhibits in each organ, and demonstrate that TCDD inhibits ventral prostate development before this organ first appears, presumably via effects on the urogenital sinus. The observation that in utero TCDD exposure (alone) inhibited development of each prostate lobe is significant because previous studies have shown that little TCDD is transmitted to mice before birth.


Subject(s)
Environmental Pollutants/toxicity , Polychlorinated Dibenzodioxins/toxicity , Prostate/growth & development , Seminal Vesicles/growth & development , Androgen-Binding Protein/biosynthesis , Androgen-Binding Protein/genetics , Animals , Body Weight/drug effects , Female , Keratins/biosynthesis , Keratins/genetics , Lactation , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Pregnancy , Prostate/drug effects , Prostate/embryology , Receptors, Aryl Hydrocarbon/genetics , Seminal Vesicles/drug effects , Seminal Vesicles/embryology
17.
Prostate ; 47(3): 164-71, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11351345

ABSTRACT

BACKGROUND: The development and progression of prostate neoplasia may recapitulate the early developmental pattern of expression of genes in the prostate. The study of prostate development may, therefore, provide insights into the molecular mechanisms important in prostate neoplasia and reveal new markers. METHODS: We compared postnatal expression of four genes: neu and epidermal growth factor receptor genes (EGFR), androgen-upregulated in the ventral prostate of adult rats (C-3), and androgen-repressed (CK8) in Sprague-Dawley rats. In situ hybridization was performed on prostate frozen sections collected on postnatal days 1, 5, 10, 15, 20, 30, and 60 from five rats per day. Staining intensities for antisense probes specific for each gene were determined relative to day 1 intensity. RESULTS: Growth factor receptors including neu and EGFR may be coordinately regulated in the basal-cell population during prostate development. CK8 and C-3 show evidence of similar androgen regulation during development. CONCLUSIONS: CK8 and C-3 have distinct patterns of expression in the postnatal period of development and these genes may be good markers of differentiation. Both neu and EGFR may be involved in androgen-independent growth of basal cell population in prostate. Prostate 47:164-171, 2001.


Subject(s)
Androgen-Binding Protein/genetics , ErbB Receptors/genetics , Genes, erbB-2/physiology , Keratins/genetics , Prostate/physiology , Androgen-Binding Protein/biosynthesis , Androgen-Binding Protein/physiology , Animals , ErbB Receptors/biosynthesis , ErbB Receptors/physiology , Gene Expression Regulation , Genes, erbB-2/genetics , In Situ Hybridization , Keratins/blood , Keratins/physiology , Male , Prostate/growth & development , Prostate/metabolism , Prostatein , Rats , Rats, Sprague-Dawley , Secretoglobins , Uteroglobin
18.
Toxicology ; 163(1): 29-38, 2001 May 28.
Article in English | MEDLINE | ID: mdl-11376863

ABSTRACT

During the last decade, the possible effects of xenobiotics on male reproductive health have resulted in great concern. More recently, evidence of antiandrogen effect in vivo by certain chemicals has been reported. The classical Hershberger in vivo assay determining organ weight changes can be improved by measuring hormone levels as well as determining changes in gene expression of androgen-responsive genes. A real-time RT-PCR method using LightCycler technology (Roche) suitable for quantitative determination of gene expression is described. The technique combines rapid thermocycling with online fluorescence detection of PCR product formation. In this study, investigation of expression of prostate specific binding protein polypeptide C3 (PBP C3) and testosterone-repressed prostatic message 2 (TRPM-2) in the ventral prostate was performed in 60-days-old castrated Wistar rats treated daily with testosterone with or without addition of flutamide or vinclozolin for 7 days in total. We show that we can quantify the level of gene expression by use of LightCycler technology, supported by changes in reproductive organ weights as well as in hormone levels, and that analysis of gene expression levels is an even more sensitive endpoint.


Subject(s)
Androgen Antagonists/pharmacology , Prostate/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Androgen Antagonists/toxicity , Androgen-Binding Protein/biosynthesis , Androgen-Binding Protein/genetics , Animals , Clusterin , Flutamide/pharmacology , Fungicides, Industrial/pharmacology , Gene Expression/drug effects , Glycoproteins/biosynthesis , Glycoproteins/genetics , Luteinizing Hormone/metabolism , Male , Molecular Chaperones/biosynthesis , Molecular Chaperones/genetics , Organ Size/drug effects , Oxazoles/pharmacology , Prostate/anatomy & histology , Prostate/metabolism , Prostatein , Rats , Rats, Wistar , Secretoglobins , Testosterone/pharmacology , Toxicity Tests/methods , Uteroglobin
19.
Prostate ; 43(3): 195-204, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10797494

ABSTRACT

BACKGROUND: Although normal prostatic development is androgen-dependent, the prostate continues to grow in the neonate despite castration. However, the manner in which neonatal growth of the prostate occurs, in the absence of the testis, remains largely unknown. The purpose of this study was to examine the differentiation of prostatic epithelial cells after neonatal castration. METHODS: Immunohistochemistry was utilized to detect the expression of differentiation products: basal-cell cytokeratin (CK 5), luminal-cell cytokeratin (CK 18), and prostatic steroid-binding protein (PBP), a ventral prostate-specific marker indicative of secretory function in luminal cells. The reverse transcription-polymerase chain reaction was used to detect transcription products of the three polypeptide subunits of PBP, designated C1, C2, and C3. Rats were castrated on day 5 after birth, and ventral prostates were collected on day 14. Dihydrotestosterone was injected (100 microg/animal every 2 days) in castrated animals to determine if PBP expression could be initiated by androgen. RESULTS: Although no major effects of castration were detected on the differentiation of stromal or basal cells (which differentiate prior to day 5), castration had a pronounced effect on luminal-cell differentiation. Castration inhibited PBP protein expression, but did not affect the expression of luminal-cell cytokeratin (CK 18) protein. Furthermore, castration reduced C1, C2, and C3 transcription. Androgen replacement to castrated animals allowed for the initiation of PBP expression, although its onset was delayed. CONCLUSIONS: These observations indicate that the testis is not necessary for prostatic luminal-cell differentiation, but is necessary for full expression of luminal-cell secretory phenotype. Furthermore, our study suggests that factors of testicular origin, in addition to androgen, are needed for proper timing of PBP expression. This investigation establishes that the cytological and the physiological differentiation of the rat prostate are differentially regulated.


Subject(s)
Androgen-Binding Protein/genetics , Orchiectomy , Prostate/cytology , Androgen-Binding Protein/biosynthesis , Androgens/physiology , Animals , Cell Differentiation , Complement C1/biosynthesis , Complement C1/genetics , Complement C2/biosynthesis , Complement C2/genetics , Complement C3/biosynthesis , Complement C3/genetics , Dihydrotestosterone/pharmacology , Female , Gene Expression , Humans , Infant, Newborn , Keratins/metabolism , Male , Phosphatidylethanolamine Binding Protein , Prostatein , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Secretoglobins , Uteroglobin
20.
Endocrinology ; 141(3): 1168-77, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10698194

ABSTRACT

The fundamental role of androgen-binding protein (ABP) in spermatogenesis remains obscure after nearly 25 yr since its first characterization. In the present investigation, we used a transgenic mouse model that overexpresses rat ABP to examine the potential involvement of this protein in the regulation of processes occurring during spermatogenesis. Specifically, homozygous or heterozygous transgenic mice were analyzed in terms of spermatogenic progression, DNA fragmentation pattern, and germinal cell ploidy status. All animals homozygous for transgenic ABP exhibited an increased accumulation of primary spermatocytes and cells at metaphase with abnormal morphology and localization within the seminiferous epithelium. Analysis of DNA fragmentation by in situ techniques and agarose gel electrophoresis provided evidence for an increased occurrence of apoptosis in the transgenic animals, principally involving pachytene spermatocytes and cells at metaphase. Flow cytometric analysis of the DNA content of isolated germ cells revealed a reduction in the number of haploid cells, an increase in the number of tetraploid cells, and the appearance of a hypotetraploid cell population, consistent with degenerating primary spermatocytes. In mice heterozygous for the transgene, the effects were less prominent, and the degree to which spermatogenesis was compromised correlated with the levels of ABP messenger RNA in individual animals. The present results are interpreted to suggest that ABP can act as a modulator of spermatogenesis by regulating completion of the first meiotic division of primary spermatocytes.


Subject(s)
Androgen-Binding Protein/genetics , Apoptosis/physiology , Germ Cells/physiology , Meiosis/physiology , Androgen-Binding Protein/biosynthesis , Animals , Cell Separation , DNA Fragmentation , Electrophoresis, Agar Gel , Female , Fertility/physiology , Flow Cytometry , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Spermatogenesis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...