Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Eur J Cancer Prev ; 26(1): 71-77, 2017 01.
Article in English | MEDLINE | ID: mdl-26886237

ABSTRACT

9-cis-Retinoic acid (9cRA), which binds to both retinoic acid receptors and retinoic X receptors, inhibits prostate cancer induction in rats and reduces growth of prostate cancer cells. However, the nature of this growth inhibition and the interactive influence of androgens are not well defined and are the subject of this report. LNCaP and PC-3 cells were cultured and treated with a range of 9cRA concentrations for 3-6 days in the absence or presence of 5α-dehydrotestosterone. 9cRA inhibited cell proliferation in a dose-dependent manner, plateauing at 10 mol/l. Treatment of cells with 10 mol/l 9cRA inhibited 5α-dihydroxytestosterone (DHT)-stimulated proliferation, the effect of which was maximal at 10 mol/l DHT. Treatment of DHT (10 mol/l)-exposed cells with 9cRA caused a dose-dependent increase in prostate-specific antigen in the medium after 6 days, but not 3 days. 9cRA caused a dose-dependent increase in apoptotic cells stained with H33258 after 3 days, but not 6 days; however, on using flow cytometry, apoptosis was apparent at both 3 and 6 days. Flow cytometry also revealed interference of G0/G1 to S phase transition by 9cRA. Inhibition by 9cRA of anchorage-independent growth of PC-3 cells was also found; LNCaP cells did not grow colonies in soft agar. 9cRA inhibited growth and induced differentiation of human LNCaP prostate cancer cells in vitro and inhibited anchorage-independent growth of PC-3 cells. Because 9cRA and 13-cis-retinoic acid, which is retinoic acid receptor-selective, prevent prostate carcinogenesis in rats, and 13-cis-retinoic acid also inhibits growth of human prostate cancer cells, the RAR is a potential molecular target for prostate cancer prevention and therapy.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Prostatic Neoplasms/metabolism , Testosterone/metabolism , Tretinoin/metabolism , Alitretinoin , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Drug Interactions/physiology , Humans , Male , Prostatic Neoplasms/pathology , Testosterone/pharmacology , Tretinoin/analogs & derivatives , Tretinoin/pharmacology
2.
Cancer Res ; 66(14): 7075-82, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16849553

ABSTRACT

The B-cell translocation gene-2 (BTG2) is present in the nuclei of epithelial cells in many tissues, including the mammary gland where its expression is regulated during glandular proliferation and differentiation in pregnancy. In immortalized mammary epithelial cells and breast cancer cells, BTG2 protein localized predominantly to the nucleus and cytoplasm, respectively. The highly conserved domains (BTG boxes A, B, and C) were required for regulating localization, suppression of cyclin D1 and growth inhibitory function of BTG2. Expression analysis of BTG2 protein in human breast carcinoma (n = 148) revealed the loss of nuclear expression in 46% of tumors, whereas it was readily detectable in the nuclei of adjacent normal glands. Loss of nuclear BTG2 expression in estrogen receptor-alpha (ERalpha)-positive breast tumors correlated significantly with increased histologic grade and tumor size. Consistent with its ability to suppress cyclin D1 transcription, loss of nuclear BTG2 expression in ER-positive breast carcinomas showed a significant correlation with cyclin D1 protein overexpression, suggesting that loss of BTG2 may be a factor involved in deregulating cyclin D1 expression in human breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cyclin D1/biosynthesis , Estrogen Receptor alpha/biosynthesis , Immediate-Early Proteins/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Female , Genes, Tumor Suppressor , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/deficiency , Immediate-Early Proteins/metabolism , Middle Aged , Protein Structure, Tertiary , Tumor Suppressor Proteins
3.
J Lipid Res ; 47(7): 1449-56, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16651661

ABSTRACT

Mechanisms that function to regulate the rate of de novo phosphatidylinositol (PtdIns) synthesis in mammalian cells have not been elucidated. In this study, we characterize the effect of phorbol ester treatment on de novo PtdIns synthesis in C3A human hepatoma cells. Incubation of cells with 12-O-tetradecanoyl phorbol 13-acetate (TPA) initially (1-6 h) results in a decrease in precursor incorporation into PtdIns; however, at later times (18-24 h), a marked increase is observed. TPA-induced glucose uptake from the medium is not required for observation of the stimulation of PtdIns synthesis, because the effect is apparent in glucose-free medium. Inhibition of the activation of arachidonic acid substantially blocks the synthesis of PtdIns but has no effect on the synthesis of phosphatidylcholine (PtdCho). Increasing the concentration of cellular phosphatidic acid by blocking its conversion to diacylglycerol, on the other hand, enhances the synthesis of PtdIns and inhibits the synthesis of PtdCho. The TPA-induced stimulation of PtdIns synthesis is not the result of the concomitant TPA-induced G1 arrest, because G1 arrest induced by mevastatin has no effect on PtdIns synthesis. Inhibition of protein kinase C activity blocks the stimulatory action of TPA on de novo synthesis of PtdIns but has no effect on TPA-induced inhibition. Potential sites of enzymatic regulation are discussed.


Subject(s)
Phosphatidylinositols/biosynthesis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , G1 Phase , Glucose/metabolism , Humans , Kinetics , Protein Kinase C/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
4.
Mol Endocrinol ; 20(2): 426-36, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16223973

ABSTRACT

Somatostatin analogs (SAs) treat acromegaly by lowering pituitary GH secretion, which, in turn, lowers systemic IGF-I. The profound systemic effect is often greater than expected in the face of only partial GH suppression. Here we report that the SA SOM230 can also act by a nonpituitary-mediated inhibition of IGF-I action. SOM230 inhibited mammary development in intact and hypophysectomized female rats, a process requiring IGF-I. IGF-I overcame this inhibition. SOM230 also inhibited other actions of IGF-I (inhibition of apoptosis, phosphorylation of insulin receptor substrate-1, and cell division). SOM230 did not reduce IGF-I mRNA abundance in mammary gland but did stimulate IGF binding protein 5 (IGFBP5). IGFBP5 was 3.75 times higher in mammary epithelium of SOM230 than in placebo animals (P < 0.001). Administration of IGFBP-5 also inhibited GH-induced mammary development (P < 0.001). Measurement of sstr(1-5) (somatostatin subtype receptor) by real-time RT-PCR revealed that the mammary glands had an abundance of sstr(3) and lower amounts of sstr(4) and sstr(5) but no sstr(1) or sstr(2.) That mammary development was also inhibited to a lesser degree than SOM230 by octreotide, whose main action is through sstr(2), strongly suggests that sstr(3) is at least in part mediating the effects of the SAs. We conclude that 1) SAs inhibit IGF-I action in the mammary gland through a novel nonpituitary mechanism; 2) IGFBP-5, here shown to inhibit pubertal mammary development, might mediate the effect; and 3) Measurement of available sstr receptors in the mammary gland suggests that sstr(3) mediates the SA activity, but sstr(5) is also a possible mediator.


Subject(s)
Insulin-Like Growth Factor I/antagonists & inhibitors , Mammary Glands, Animal/growth & development , Receptors, Somatostatin/agonists , Somatostatin/analogs & derivatives , Animals , Apoptosis , Cell Division/drug effects , Female , Growth Hormone/antagonists & inhibitors , Growth Hormone/pharmacology , Insulin-Like Growth Factor Binding Protein 5/metabolism , Insulin-Like Growth Factor Binding Protein 5/pharmacology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Octreotide/pharmacology , Phosphorylation , Rats , Rats, Inbred Strains , Somatostatin/pharmacology
5.
Am J Physiol Renal Physiol ; 290(2): F428-37, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16159897

ABSTRACT

Recent studies have shown that accessory proteins that interact with the apical Na(+)/H+ exchanger NHE3 are a vital part of the dynamic nature of the Na(+)/H+ exchanger regulation. We have identified MAST205, a microtubule-associated serine/threonine kinase with a molecular mass of 205 kDa that interacts with NHE3. MAST205 contains a S/T kinase domain and a PDZ domain that mediates interaction with NHE3. Northern blot analysis showed that MAST205 is highly expressed in human and rat kidney. Expression in opossum kidney (OK) cells showed that MAST205 is predominantly expressed in the apical membrane of the cells. Immunohistochemical studies demonstrated the presence of MAST205 at the apical region of the renal proximal tubules. Heterologous expression of MAST205 in OK cells inhibited endogenous NHE3 activity, and this inhibition required the presence of the kinase domain of MAST205, since deletion of the kinase domain or a dominant-negative mutant of MAST205 did not affect the activity of NHE3. Consistent with these results, we found that MAST205 phosphorylated NHE3 under in vitro conditions. However, overexpression of MAST205 did not affect expression of NHE3 proteins, suggesting that the effect of MAST205 was not mediated by a decrease in NHE3 expression. These findings suggest that MAST205 regulates NHE3 activity and, although the precise mechanism is yet to be determined, MAST205 appears to inhibit NHE3 activity through a phosphorylation-dependent mechanism.


Subject(s)
Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Cell Line , Humans , Kidney Tubules/cytology , Male , Mice , Microtubule-Associated Proteins/genetics , Opossums , Organ Specificity , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Hydrogen Exchanger 3 , Transfection
6.
Dev Biol ; 288(2): 334-47, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16324690

ABSTRACT

The mouse prostate gland develops by branching morphogenesis from the urogenital epithelium and mesenchyme. Androgens and developmental factors, including FGF10 and SHH, promote prostate growth (Berman, D.M., Desai, N., Wang, X., Karhadkar, S.S., Reynon, M., Abate-Shen, C., Beachy, P.A., Shen, M.M., 2004. Roles for Hedgehog signaling in androgen production and prostate ductal morphogenesis. Dev. Biol. 267, 387-398; Donjacour, A.A., Thomson, A.A., Cunha, G.R., 2003. FGF-10 plays an essential role in the growth of the fetal prostate. Dev. Biol. 261, 39-54), while BMP4 signaling from the mesenchyme has been shown to suppresses prostate branching (Lamm, M.L., Podlasek, C.A., Barnett, D.H., Lee, J., Clemens, J.Q., Hebner, C.M., Bushman, W., 2001. Mesenchymal factor bone morphogenetic protein 4 restricts ductal budding and branching morphogenesis in the developing prostate. Dev. Biol. 232, 301-314). Here, we show that Bone Morphogenetic Protein 7 (BMP7) restricts branching of the prostate epithelium. BMP7 is expressed in the periurethral urogenital mesenchyme prior to formation of the prostate buds and, subsequently, in the prostate epithelium. We show that BMP7(lacZ/lacZ) null prostates show a two-fold increase in prostate branching, while recombinant BMP7 inhibits prostate morphogenesis in organ culture in a concentration-dependent manner. We further explore the mechanisms by which the developmental signals may be interpreted in the urogenital epithelium to regulate branching morphogenesis. We show that Notch1 activity is associated with the formation of the prostate buds, and that Notch1 signaling is derepressed in BMP7 null urogenital epithelium. Based on our studies, we propose a model that BMP7 inhibits branching morphogenesis in the prostate and limits the number of domains with high Notch1/Hes1 activity.


Subject(s)
Bone Morphogenetic Proteins/physiology , Morphogenesis , Prostate/embryology , Receptor, Notch1/physiology , Transforming Growth Factor beta/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/genetics , Epithelium/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Male , Mesoderm/physiology , Mice , Mice, Knockout , Organ Culture Techniques , Prostate/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Transcription Factor HES-1 , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics
7.
Prostate ; 63(4): 358-68, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15611996

ABSTRACT

BACKGROUND: Sensory peptide neurotransmitters have been implicated as significant regulators of prostate growth. This study was designed to evaluate the role of neurokinins in proliferation, differentiation, and contraction of canine prostate cells in culture. METHODS: NK1, NK2, and NK3 receptor subtypes were localized in canine prostate tissue by immunocytochemistry and ligand binding studies. Functional effects of neurokinin agonists were tested on cell differentiation (expression of smooth muscle actin (SMA)), proliferation (MTS assay), and contraction of canine prostate cells in culture. RESULTS: Immunocytochemical staining of canine prostate sections revealed strong stromal staining for NK1 together with weak stromal staining for NK2 and even weaker staining for NK3. Furthermore, there was overlapping localization of NK1 receptors, substance P (SP), and calcitonin gene-regulated peptide (CGRP) in prostate tissue sections. SP caused concentration-dependent increase in SMA expression that was attenuated in a concentration-dependent manner by YM-44778, a non-selective antagonist for neurokinin receptors, but not by either the NK2 antagonist (SR-48968) nor by the NK3 antagonist (SB-223412). SP and neurokinin A (NKA) also caused a modest contraction of stromal cells in collagen gels. NKA stimulated proliferation of prostate epithelial cells without any apoptotic effect, which was attenuated by SR-48968. Surprisingly, in binding studies NK3 appeared to be the most abundant neurokinin receptor subtype, although functional studies failed to reveal significant coupling of this receptor. CONCLUSIONS: Our results suggest that, at least in vitro, neurokinins have modest effects on canine prostate epithelial cell proliferation, stromal differentiation, and contraction.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Prostate/cytology , Prostate/metabolism , Receptors, Tachykinin/metabolism , Substance P/metabolism , Animals , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Dogs , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Immunohistochemistry , Male , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Neurokinin A/pharmacology , Quinolines/metabolism , Quinolines/pharmacology , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-2/metabolism , Receptors, Neurokinin-3/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Substance P/pharmacology , Tritium
8.
Oncogene ; 23(50): 8310-9, 2004 Oct 28.
Article in English | MEDLINE | ID: mdl-15378000

ABSTRACT

BTG2, a p53-inducible antiproliferative gene, is stimulated in breast cancer cells by activation of nuclear factor kappa B (NF-kappaB). In rat mammary glands, BTG2 is expressed in epithelial cells and levels decreased during pregnancy and lactation but recovered during involution. Estrogen and progestin suppress BTG2 expression, suggesting that these steroids, which stimulate proliferation and lobuloalveolar development of mammary epithelial cells, may downregulate BTG2 in the mammary gland during pregnancy. Consistent with the report that BTG2 inhibits cyclin D1 expression, suppression of BTG2 mRNA in the mammary gland during gestation, and by estrogen and progestin, correlated with stimulation of cyclin D1. Ectopic expression of BTG2 inhibited breast cancer cell growth by arresting cells in the G1 phase, an effect reversed by cyclin D1. BTG2 expression was very low or undetectable in human breast cancer cell lines compared with nontumorigenic mammary epithelial cells, and nuclear expression of BTG2 was absent in 65% of human breast tumors compared with adjacent matched normal glands. Spontaneous mammary tumors arising in a mouse model with targeted expression of the early region of the SV40 large tumor Ag demonstrated loss of BTG2 protein very early during the tumorigenic process. Thus deregulation of BTG2 may be an important step in the development of mammary tumors.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Immediate-Early Proteins/genetics , NF-kappa B/physiology , Animals , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Tumor Suppressor Proteins
9.
Clin Cancer Res ; 10(14): 4614-21, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15269132

ABSTRACT

PURPOSE: The Cooperative Prostate Cancer Tissue Resource (CPCTR) is a National Cancer Institute-supported tissue bank that provides large numbers of clinically annotated prostate cancer specimens to investigators. This communication describes the CPCTR to investigators interested in obtaining prostate cancer tissue samples. EXPERIMENTAL DESIGN: The CPCTR, through its four participating institutions, has collected specimens and clinical data for prostate cancer cases diagnosed from 1989 onward. These specimens include paraffin blocks and frozen tissue from radical prostatectomy specimens and paraffin blocks from prostate needle biopsies. Standardized histopathological characterization and clinical data extraction are performed for all cases. Information on histopathology, demography (including ethnicity), laboratory data (prostate-specific antigen values), and clinical outcome related to prostate cancer are entered into the CPCTR database for all cases. Materials in the CPCTR are available in multiple tissue formats, including tissue microarray sections, paraffin-embedded tissue sections, serum, and frozen tissue specimens. These are available for research purposes following an application process that is described on the CPCTR web site (www.prostatetissues.org). RESULTS: The CPCTR currently (as of October 2003) contains 5135 prostate cancer cases including 4723 radical prostatectomy cases. Frozen tissues, in some instances including patient serum samples, are available for 1226 cases. Biochemical recurrence data allow identification of cases with residual disease, cases with recurrence, and recurrence-free cases. CONCLUSIONS: The CPCTR offers large numbers of highly characterized prostate cancer tissue specimens, including tissue microarrays, with associated clinical data for biomarker studies. Interested investigators are encouraged to apply for use of this material (www.prostatetissues.org).


Subject(s)
Prostatic Neoplasms/pathology , Tissue Banks/organization & administration , Adult , Aged , Aged, 80 and over , Biomedical Research/methods , Biomedical Research/statistics & numerical data , Humans , Male , Middle Aged , Prostatectomy , Prostatic Neoplasms/surgery , Prostatic Neoplasms/therapy , Tissue Banks/trends , United States
10.
Urol Res ; 32(4): 261-5, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15221243

ABSTRACT

The quinazoline family of alpha1-blockers (prazosin, doxazosin, and terazosin) induce apoptosis of prostate cells through an alpha1-adrenoceptor-independent mechanism. The objective of this study was to gain insight into the non-adrenergic, apoptotic mechanism of action of doxazosin in the prostate and the induction of anoikis by doxazosin. Primary cultures of benign prostate stromal and epithelial cells and the LNCaP (androgen sensitive) and PC-3 (androgen insensitive) prostate carcinoma cell lines were treated with doxazosin (0-50 microM). The effects of doxazosin on cell morphology, caspase-3 activity, and the expression levels of focal adhesion kinase (FAK) and integrin-linked kinase (ILK) were examined. Doxazosin induced changes in morphology consistent with anoikis in both benign and cancerous prostatic cells and increased caspase-3 activity. The effects were similar comparing benign cells (which express alpha1-adrenoceptors) and cancer cells (which do not express alpha1-adrenoceptors), but were more robust in benign cells. Norepinephrine had no effect on doxazosin-induced cell morphology or caspase-3 activity. Treatment of PC-3 cells with doxazosin significantly reduced the protein levels of FAK but did not significantly affect the levels of ILK. These findings suggest that doxazosin induces apoptosis and anoikis of prostate cancer cells by a mechanism of action that is alpha1-adrenoceptor independent. The apoptosis of cancer cells induced by doxazosin counteracts cell proliferation and may have the potential of retarding or reversing prostate cancer cell growth.


Subject(s)
Anoikis/drug effects , Caspases/metabolism , Doxazosin/pharmacology , Prostatic Neoplasms/drug therapy , Caspase 3 , Caspases/drug effects , Focal Adhesion Kinase 2 , Humans , Immunoblotting , Male , Prostatic Neoplasms/enzymology , Protein-Tyrosine Kinases , Sensitivity and Specificity , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
11.
Biochem Biophys Res Commun ; 317(2): 444-50, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-15063778

ABSTRACT

Mice that are homozygous for the vibrator mutation express 65-85% less phosphatidylinositol transfer protein alpha (PITPalpha) than their wild type litter mates. By postnatal day 10-12 (P10-12) they exhibit signs of neurodegeneration and die prematurely by P40. In the present study, we examine the lipid content of brain, liver, and mammary glands from these animals. Lipid-mediated signal transduction is evaluated in primary fibroblast cultures. With respect to the lipid make-up of brain and liver, we report that there is a significant increase (2- to 4-fold) in the neutral lipids present in the livers of vb/vb animals when compared with wild type (+/+) litter mates. No significant changes are observed in the brains of these animals. The mammary glands of vb/vb mice are underdeveloped with respect to ductal and alveolar structures, and the fat pad is composed of predominantly brown adipose tissue rather than the white adipose tissue characteristic of age-matched wild type litter mates. No differences are observed in any aspect of lipid-mediated signal transduction.


Subject(s)
Adipose Tissue/metabolism , Brain/metabolism , Lipid Metabolism , Liver/metabolism , Mammary Glands, Animal/metabolism , Membrane Proteins/deficiency , Phosphatidylcholines/metabolism , Phosphatidylinositols/metabolism , Animals , Carrier Proteins , Cells, Cultured , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Phospholipid Transfer Proteins , Signal Transduction/drug effects , Signal Transduction/physiology , Tissue Distribution , Vasopressins/pharmacology
12.
J Immunol ; 172(4): 2559-68, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14764729

ABSTRACT

Stimulation of murine macrophages with LPS results in the coordinated activation of a set of proinflammatory cytokines and costimulatory molecules, including TNF-alpha, IL-6, IL-1, IL-8, IL-12, and CD80. Macrophage LPS-induced synthesis of IL-12 is inhibited following FcgammaR ligation; TNF-alpha secretion is unchanged. We report that microtubule-associated serine/threonine kinase-205 kDa (MAST205) is required for LPS-induced IL-12 synthesis. RNA interference-mediated suppression of MAST205 results in the inhibition of LPS-stimulated IL-12 promoter activity and IL-12 secretion, from both J774 cells and bone marrow-derived macrophages. Similarly, dominant-negative MAST205 mutants inhibit LPS-stimulated IL-12 synthesis and NF-kappaB activation, but do not affect IL-1 or TNF-alpha signaling. Finally, macrophage FcgammaR ligation regulates MAST205 by inducing the rapid ubiquitination and proteasomal degradation of the protein.


Subject(s)
Interleukin-12/biosynthesis , Microtubule-Associated Proteins/physiology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/physiology , Protein Subunits/biosynthesis , Receptors, IgG/physiology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Cell Line, Tumor , Cells, Cultured , Interleukin-12/antagonists & inhibitors , Interleukin-12 Subunit p40 , Leukemia P388 , Ligands , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/isolation & purification , NF-kappa B/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/isolation & purification , Protein Subunits/antagonists & inhibitors , RNA Interference/immunology , Receptors, IgG/immunology , Receptors, IgG/metabolism
13.
J Biol Chem ; 279(14): 13944-52, 2004 Apr 02.
Article in English | MEDLINE | ID: mdl-14711828

ABSTRACT

Androgen receptor trapped clone-27 (ART-27) is a newly described transcriptional coactivator that binds to the N terminus of the androgen receptor (AR). Given the vital importance of AR signaling in prostate growth and differentiation, we investigated the role of ART-27 in these processes. Immunohistochemical studies indicate that ART-27 protein is expressed in differentiated epithelial cells of adult human prostate and breast tissue. In prostate, ART-27 is abundant in AR-positive prostate luminal epithelial cells, in contrast to the stroma, where cells express AR but not ART-27. The use of a rat model of androgen depletion/reconstitution indicates that ART-27 expression is associated with the elaboration of differentiated prostate epithelial cells. Interestingly, regulated expression of ART-27 in the androgen-sensitive LNCaP prostate cancer cell line inhibits androgen-mediated cellular proliferation and enhances androgen-mediated transcription of the prostate-specific antigen (PSA) gene. Consistent with a growth suppressive function, we show that ART-27 expression levels are negligible in human prostate cancer. Importantly, examination of ART-27 protein expression in early fetal prostate development demonstrates that ART-27 is detected only when the developing prostate gland has proceeded from a solid mass of undifferentiated cells to a stage in which differentiated luminal epithelial cells are evident. Thus, ART-27 is an AR cofactor shown to be subject to both cell type and developmental regulation in humans. Overall, the results suggest that decreased levels of ART-27 protein in prostate cancer tissue may occur as a result of de-differentiation, and indicate that ART-27 is likely to regulate a subset of AR-responsive genes important to prostate growth suppression and differentiation.


Subject(s)
Prostate/embryology , Prostate/physiology , Prostatic Neoplasms/physiopathology , Trans-Activators/genetics , Trans-Activators/metabolism , Androgens/pharmacology , Animals , Breast/cytology , Cell Cycle Proteins , Cell Division/drug effects , Cell Division/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Male , Molecular Chaperones , Neoplasm Proteins , Prostate/cytology , Rats , Rats, Sprague-Dawley
14.
Cancer Res ; 63(9): 2067-71, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12727821

ABSTRACT

Chronic lymphocytic leukemia (CLL) is characterized by an accumulation of monoclonal B lymphocytes in the hematopoietic organs. Rarely, CLL cells accumulate in a single atypical site. The mechanism underlying this unusual distribution of CLL cells has not been studied previously. We obtained peripheral blood from five patients having early stage CLL with heavy prostate infiltration. These patients' circulating CLL cells bound strongly in vitro to cultured prostate cell lines PC3, LNCaP, and DU145 and to short-term cultures of fresh prostate cells but not to colon, breast, or bladder cells. CLL cells from patients without prostate infiltration did not bind in vitro to any cell line. Peripheral blood CLL cells from one patient with CLL with heavy prostate infiltration were fused with a mouse-human heteromyeloma line to make hybridomas expressing the same monoclonal IgM as the patient's CLL cells. The hybridoma cells bound specifically to prostate cells. IgM secreted by the hybridoma blocked binding of the patient's CLL cells to prostate cells. Flow cytometry and immunohistochemistry demonstrated that the secreted IgM bound specifically to prostate cells. These results indicate that CLL with atypical prostate infiltration can be mediated by specific surface-bound IgM against an antigen expressed specifically by prostate cells and suggest that a similar mechanism might also apply to cases of CLL with atypical infiltration into other organs.


Subject(s)
Immunoglobulin M/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemic Infiltration/pathology , Prostate/pathology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Membrane/immunology , Cell Membrane/metabolism , Flow Cytometry , Humans , Hybridomas , Immunoglobulin M/immunology , Immunohistochemistry , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemic Infiltration/immunology , Leukemic Infiltration/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Prostate/immunology , Prostate/metabolism , Tumor Cells, Cultured
15.
Prostate ; 54(3): 230-7, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12518328

ABSTRACT

BACKGROUND: The norepinephrine (NE) analog phenylephrine has previously been shown to induce atypical prostate hyperplasia in rats. The objective of the present study was to provide further insight into the mechanism of phenylephrine-induced prostate growth. METHODS: Adult male C57/BL6 mice were given daily subcutaneous injection of phenylephrine, isoproterenol, or phenylephrine in combination with BMY7378, cyclazosin, RS100329, or yohimbine, and the effects on ventral prostate histology, and proliferative and apoptotic indices determined. Phenylephrine was also administered in combination with testosterone in castrated mice. RESULTS: Atypical prostatic hyperplasia characterized by piling up and/or papillary infolding of epithelial cells with concomitant stromal smooth muscle hyperplasia was seen in adult mice given subcutaneous injection of phenylephrine daily for 26 days. Phenylephrine induced hyperplasia was more severe proximally and was associated with significantly reduced rates of apoptosis (but no change in cell proliferation) in both stromal and epithelial compartments. Only the alpha(1A)-adrenoceptor selective subtype antagonist RS100329 abrogated the phenylephrine-induced hyperplasia. Using selective antibodies, the alpha(1A-1)-adrenoceptor subtype was predominantly localized to the stromal compartments of the mouse and rat ventral prostates. The effects of phenylephrine were mediated independent of testicular androgens. CONCLUSIONS: Prostatic hyperplasia in mice occurs as a consequence of subchronic administration of the sympathomimetic phenylephrine. Response to phenylephrine is mediated by the alpha(1A)-adrenoceptor, which predominates in the stroma of the rodent ventral prostate. Conceivably, therefore, phenylephrine could directly modulate prostate stromal growth, and indirectly modulate epithelial growth in a paracrine fashion. We cannot, however, rule out the contribution of other indirect effects such as hypoxia/reperfusion or effects on intermediary metabolism.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Phenylephrine/pharmacology , Prostatic Hyperplasia/chemically induced , Prostatic Hyperplasia/pathology , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Stromal Cells/pathology , Sympathomimetics/pharmacology , Testis/metabolism , Testosterone/biosynthesis
16.
J Cell Physiol ; 194(2): 151-61, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12494453

ABSTRACT

Agonist-stimulated phosphoinositide turnover is accompanied by compensatory resynthesis of these lipids. Several lines of evidence suggest that resynthesis of phosphatidylinositol (PtdIns) involves phosphorylation of diacylglycerol (DG) (salvage pathway) rather than acylation of glycerol phosphate (de novo pathway), although a contribution from the de novo pathway has not been ruled out. To determine the relative contribution of the de novo and salvage pathways in stimulated PtdIns resynthesis, an inhibitor of de novo synthesis (Triacsin C) was incubated simultaneously with the hormone agonist. Results indicate that at early times (90 min), hormone-stimulated PtdIns synthesis proceeds predominantly via the salvage pathway, although some de novo synthesis is also taking place. At later times (24 h), stimulated synthesis is solely via the de novo pathway. Increasing cellular DG content by either adding exogenous DG or treating cells with bacterial phospholipase C (bPLC) results in deacylation of the DG rather than phosphorylation; however, inhibition of this deacylation fails to stimulate phosphorylation by DG kinase (DGK), suggesting channeling of the DG substrate between PLC and DG kinase. Receptor activation is not required for activation of DGK, since treatment with a calcium ionophore induces the same Triacsin C-insensitive PtdIns synthesis. Depletion of the polyphosphoinositide pools by treatment with wortmannin prevents both hormone and A23187-induced polyphosphoinositide hydrolysis; however, A23187 is still able to induce hydrolysis of PtdIns and subsequent compensatory resynthesis. The inability of R59949 to inhibit either hormone-induced or ionophore-induced PtdIns resynthesis suggests that the alpha isoform is not involved; however, its possible that the channeling phenomenon prevents the inhibitor from gaining access to the diacylglycerol kinase enzyme. Further study will be required to determine which isoform catalyzes hormone-induced resynthesis of PtdIns.


Subject(s)
Hormones/pharmacology , Phosphatidylinositols/biosynthesis , Animals , Calcium/physiology , Diacylglycerol Kinase/metabolism , Enzyme Activation , Hydrolysis , Phosphatidylinositol Phosphates/metabolism , Phospholipids/biosynthesis , Rats , Receptors, Cell Surface/physiology , Triazenes/pharmacology , Tumor Cells, Cultured , Type C Phospholipases/metabolism
17.
Oncogene ; 21(44): 6772-78, 2002 Oct 03.
Article in English | MEDLINE | ID: mdl-12360398

ABSTRACT

The p53-transcriptional target, BTG2(TIS21/PC3), was previously identified as an antiproliferative gene. However, the precise biological functions of the protein product remain to be elucidated. BTG2(TIS21/PC3) expression is induced in vivo during neurogenesis, and the gene is transiently expressed in vitro in rat pheochromocytoma PC12 cells after induction of neuronal differentiation by addition of nerve growth factor (NGF). These observations suggest that BTG2(TIS21/PC3) is functionally significant during the neuronal differentiation process. To test this hypothesis, a vector that expressed BTG2(TIS21/PC3) under the control of an inducible promoter was introduced into PC12 cells. Growth arrest and differentiation in response to NGF were greatly enhanced by BTG2(TIS21/PC3) overexpression. Furthermore, an antisense oligonucleotide complementary to BTG2(TIS21/PC3) mRNA, which was able to inhibit endogenous BTG2(TIS21/PC3) expression, triggered programmed cell death in differentiated PC12 cells. These observations confirm that BTG2(TIS21/PC3) expression promotes neuronal differentiation and that it is required for survival of terminally differentiated cells.


Subject(s)
Apoptosis , Immediate-Early Proteins/physiology , Neurons/physiology , Animals , Cell Differentiation , Cell Division , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/physiology , Immediate-Early Proteins/genetics , Nerve Growth Factor/pharmacology , PC12 Cells , Rats
18.
Regul Toxicol Pharmacol ; 35(3): 338-46, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12202049

ABSTRACT

Studies conducted in our laboratories and by others found no consistent correlation between prostate size, prostate pathology, or the development of prostate cancer under a variety of experimental conditions. Furthermore, an evaluation of eight published studies that were conducted in mice and rats following in utero exposure by oral treatment of dams with low levels of bisphenol A (BPA) and that focused on the prostate identified several discrepancies that affect their adequacy for use in human risk assessment. For example, there was inadequate reporting of the purity of BPA and the animal supplier used, and housing of offspring was not the same among the studies. In addition, there were differences between studies with mice and rats in exposure regimen, route of exposure, and numbers of dams or pups used per BPA dose group. Poor inter- and intraspecies correlation (i.e., mouse to rat or between mouse or rat strains) further complicates the ability to use results from these studies to predict potential prostate effects in humans. Thus, we conclude that a finding of increased prostate weight in rodent studies with perinatal exposure in the absence of associated pathologic and/or functional changes is meaningless and not indicative of a potential adverse effect in humans.


Subject(s)
Air Pollutants/adverse effects , Estrogens, Non-Steroidal/adverse effects , Phenols/adverse effects , Prostate/drug effects , Prostatitis/chemically induced , Air Pollutants/toxicity , Animals , Benzhydryl Compounds , Dose-Response Relationship, Drug , Estrogens, Non-Steroidal/toxicity , Female , Humans , Hyperplasia/chemically induced , Hyperplasia/pathology , Male , Maternal Exposure , Mice , Organ Size/drug effects , Phenols/toxicity , Pregnancy , Prenatal Exposure Delayed Effects , Prostate/pathology , Prostatitis/pathology , Rats , Risk Assessment , Species Specificity
19.
Eur J Cancer Prev ; 11 Suppl 2: S18-27, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12570331

ABSTRACT

Prostate cancer is the most common male malignancy in western countries. Although primary prevention of prostate cancer is not possible, screening using prostate-specific antigen (PSA) may eliminate prostate cancers by definitive treatments. Prevention of clinically detectable prostate cancer requires earlier chemoprevention interventions. Because prostate cancer is histologically present in 30-50% of 30- to 50-year-old men, effective chemoprevention needs to inhibit not only prostate carcinogenesis but also growth and progression of these cancers. A prostate carcinogenesis animal model has been used to screen chemopreventive agents; inhibitory effects were found with 9-cis-retinoic acid, dehydroepiandrosterone, fluasterone, and the Bowman-Birk inhibitor and an isoflavone mixture which both occur in soy. Such results can be used to select agents for clinical trials. Besides large-scale long-duration prevention trials, trials of short/intermediate duration using smaller cohorts prior to or following radical prostatectomy may provide excellent and cost-effective approaches for chemopreventive agent efficacy testing. Intervention prior to surgery allows measurements of intervention agents and intermediate end-points in the prostate. These peri-surgical trials only assess inhibition of growth and progression of preexisting cancer, not real preventive effects, but they focus on clinically significant cancers. Such trials are an essential step in the development of antiprostate cancer chemoprevention agents.


Subject(s)
Adenocarcinoma/prevention & control , Chemoprevention/methods , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/prevention & control , Adenocarcinoma/epidemiology , Aged , Animals , Clinical Trials as Topic , Disease Models, Animal , Humans , Incidence , Male , Mass Screening/methods , Middle Aged , Primary Prevention/methods , Prognosis , Prostatic Neoplasms/epidemiology , Risk Assessment , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL