Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
2.
Oncogene ; 31(19): 2480-90, 2012 May 10.
Article in English | MEDLINE | ID: mdl-21996749

ABSTRACT

Hydrogen peroxide-inducible clone-5 (Hic-5, or androgen receptor-associated protein 55) is a transforming growth factor-ß-inducible LIM protein whose deregulation is implicated in the progression of prostate cancer. Here, we report that Hic-5 binds to Smads 1, 5 and 8, and represses bone morphogenetic protein (BMP) signaling responses. Myc-Hic-5 but not Myc-paxillin was specifically immunoprecipitated with anti-FLAG IgG1 from lysates of HEK293 co-transfected with either Myc-Hic-5 or Myc-paxillin and FLAG-tagged Smads 1, 5 or 8. We showed that such interactions require the LIM3 domain of Hic-5 and the MH2 domain of those Smads. Anti-Hic-5 antibody specifically pulled down endogenous Smad1 in both the PC3 human prostate cell line and primary cultures of rat prostate fibroblasts, supporting that Hic-5 binds to Smad1 at the endogenous level. Bacterially expressed glutathione S-transferase (GST)-Smads 1, 5 or 8, but not GST alone, pulled down in vitro transcribed and translated Hic-5, implicating that Hic-5 binds directly to Smads 1, 5 and 8. Significantly, using Hic-5 small hairpin RNA silencing and overexpression systems, we show that Hic-5 (at both the endogenous and exogenous levels) represses the ability of BMP4 to induce expression of the inhibitor of differentiation-1 (Id1; a downstream target gene of BMP), activate the Id1 gene promoter and induce apoptosis in human and rat prostate epithelial cells. Moreover, silencing of Hic-5 in PC3 cells as well as in the WPMY-1 human prostate stroma cell line greatly enhances the levels of endogenous phospho-Smad1/5/8. Finally, we provide fluorescent microscopic imaging to support that Smad1 and Hic-5 mutually interact also at the level of their nuclear export mechanisms. Collectively, these results provide the first evidence for a physical and mutual functional interaction between Hic-5 and the BMP signaling pathway.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Prostatic Neoplasms/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Inhibitor of Differentiation Protein 1/metabolism , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Male , Rats
3.
Cell Death Dis ; 1: e31, 2010.
Article in English | MEDLINE | ID: mdl-21364639

ABSTRACT

αA-crystallin is a molecular chaperone and an antiapoptotic protein. This study investigated the mechanism of inhibition of apoptosis by human αA-crystallin and determined if the chaperone activity of αA-crystallin is required for the antiapoptotic function. αA-crystallin inhibited chemical-induced apoptosis in Chinese hamster ovary (CHO) cells and HeLa cells by inhibiting activation of caspase-3 and -9. In CHO cells, it inhibited apoptosis induced by the overexpression of human proapoptotic proteins, Bim and Bax. αA-crystallin inhibited doxorubicin-mediated activation of human procaspase-3 in CHO cells and it activated the PI3K/Akt cell survival pathway by promoting the phosphorylation of PDK1, Akt and phosphatase tensin homologue in HeLa cells. The phosphoinositide 3 kinase (PI3K) activity was increased by αA-crystallin overexpression but the protein content was unaltered. Downregulation of PI3K by the expression of a dominant-negative mutant or inhibition by LY294002 abrogated the ability of αA-crystallin to phosphorylate Akt. These antiapoptotic functions of αA-crystallin were enhanced in a mutant protein (R21A) that shows increased chaperone activity than the wild-type (Wt) protein. Interestingly, a mutant protein (R49A) that shows decreased chaperone activity was far weaker than the Wt protein in its antiapoptotic functions. Together, our study results show that αA-crystallin inhibits apoptosis by enhancing PI3K activity and inactivating phosphatase tensin homologue and that the antiapoptotic function is directly related to its chaperone activity.


Subject(s)
Apoptosis/physiology , Crystallins/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , CHO Cells , Caspase 3/metabolism , Caspase 9/metabolism , Caspase Inhibitors , Cricetinae , Cricetulus , Crystallins/genetics , Cytochromes c/metabolism , Doxorubicin/pharmacology , Enzyme Activation/genetics , Enzyme Inhibitors/metabolism , HeLa Cells , Humans , Membrane Proteins/metabolism , Mitochondria/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , bcl-2-Associated X Protein/metabolism
4.
Cell Death Differ ; 16(5): 758-69, 2009 May.
Article in English | MEDLINE | ID: mdl-19247369

ABSTRACT

Earlier, we have reported that 70 kDa subunit of Ku protein heterodimer (Ku70) binds and inhibits Bax activity in the cytosol and that ubiquitin (Ub)-dependent proteolysis of cytosolic Ku70 facilitates Bax-mediated apoptosis. We found that Hdm2 (human homolog of murine double minute) has an ability to ubiquitinate Ku70 and that Hdm2 overexpression in cultured cells causes a decrease in Ku70 expression levels. An interaction between Ku70 and Hdm2 was shown by means of immunoprecipitation, whereas none could be shown between 80 kDa subunit of Ku protein heterodimer and Hdm2. Vascular endothelial growth factor (VEGF) is known to inhibit endothelial cell (EC) apoptosis through an Akt-mediated survival kinase signal; however, the mechanism underlying this inhibition of apoptosis has not been fully elucidated. We found that VEGF inhibited cytosolic Ku70 degradation induced by apoptotic stress. It is known that Akt-dependent phosphorylation of Hdm2 causes nuclear translocation of Hdm2 followed by Hdm2-mediated inactivation of p53. We found that VEGF stimulated nuclear translocation of Hdm2 in EC and efficiently inhibited Ku70 degradation. We also found that constitutively active Akt, but not kinase-dead Akt, inhibited Ku70 degradation in the cytosol. Furthermore, Ku70 knockdown diminished antiapoptotic activity of Akt. Taken together, we propose that Hdm2 is a Ku70 Ub ligase and that Akt inhibits Bax-mediated apoptosis, at least in part, by maintaining Ku70 levels through the promotion of Hdm2 nuclear translocation.


Subject(s)
Antigens, Nuclear/metabolism , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Apoptosis , Cell Line , Cell Survival , HeLa Cells , Humans , Ku Autoantigen , Phosphorylation , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Tumor Suppressor Protein p53/metabolism , Ubiquitination , Vascular Endothelial Growth Factor A/metabolism , bcl-2-Associated X Protein/metabolism
5.
Oncogene ; 27(54): 6791-805, 2008 Nov 20.
Article in English | MEDLINE | ID: mdl-18762808

ABSTRACT

We recently reported that hydrogen peroxide-inducible clone-5 (Hic-5, also named androgen receptor-associated protein 55) can bind to the transforming growth factor-beta (TGF-beta)-signaling regulator Smad3, thereby inhibiting certain Smad3-dependent TGF-beta responses. We now show that Hic-5 can also control TGF-beta responses through an alternative mechanism involving Smad7, a key negative regulator of TGF-beta signaling. Hic-5 binds directly to Smad7. This interaction requires the LIM3 domain of Hic-5, and enhances TGF-beta signaling through causing loss of Smad7 protein but not mRNA. Enforced expression of Hic-5 reverses the ability of Smad7 to suppress TGF-beta-induced phosphorylation of Smads 2 and 3 and activation of the plasminogen activator inhibitor-1 promoter (in NRP-154 and PC3 prostate carcinoma and WPMY-1 prostate myofibroblast cell lines). Lentiviral-mediated small-hairpin RNA silencing of endogenous Hic-5 reduced TGF-beta responses in PC3 and WPMY-1 cells. Further work suggests that the level of Smad7 is modulated by its physical interaction with Hic-5 and targeted to a degradation pathway not likely to be proteasomal. Our findings support that Hic-5 functions as a cell-type-specific activator of TGF-beta signaling through its ability to physically interact with and neutralize Smad7.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Smad7 Protein/antagonists & inhibitors , Transforming Growth Factor beta/physiology , 3T3 Cells , Animals , Cell Division/physiology , Cell Line , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney/embryology , LIM Domain Proteins , Lentivirus/genetics , Male , Mice , Phosphorylation , Plasminogen Activator Inhibitor 1/metabolism , Prostatic Neoplasms/physiopathology , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA Interference , Signal Transduction , Smad7 Protein/metabolism , Transforming Growth Factor beta/antagonists & inhibitors
6.
Oncogene ; 27(40): 5326-38, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18504435

ABSTRACT

Survivin is a prosurvival protein overexpressed in many cancers through mechanisms that remain poorly explored, and is implicated in control of tumor progression and resistance to cancer chemotherapeutics. Here, we report a critical role for survivin in the induction of apoptosis by transforming growth factor-beta (TGF-beta). We show that TGF-beta rapidly downregulates survivin expression in prostate epithelial cells, through a unique mechanism of transcriptional suppression involving Smads 2 and 3, Rb/E2F4, and the cell-cycle repressor elements CDE and CHR. This TGF-beta response is triggered through a Smad2/3-dependent hypophosphorylation of Rb and the subsequent association of the Rb/E2F4 repressive complex to CDE/CHR elements in the proximal region of the survivin promoter. Viral-mediated gene delivery experiments, involving overexpressing or silencing survivin, reveal critical roles of survivin in apoptosis induced by TGF-beta alone or in cooperation with cancer therapeutic agents. We propose a novel TGF-beta/Rb/survivin axis with a putative role in the functional switch of TGF-beta from tumor suppressor to tumor promoter.


Subject(s)
Apoptosis/physiology , E2F4 Transcription Factor/metabolism , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Retinoblastoma Protein/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Blotting, Western , E2F4 Transcription Factor/genetics , Electrophoretic Mobility Shift Assay , Humans , Inhibitor of Apoptosis Proteins , Male , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Response Elements , Retinoblastoma Protein/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/genetics , Smad3 Protein/genetics , Survivin , Transcription, Genetic , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured
7.
Oncogene ; 25(40): 5547-60, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16619037

ABSTRACT

Cited (CBP/p300-interacting transactivators with glutamic acid (E)/aspartic acid (D)-rich C-terminal domain) 2, which is a CBP/p300-binding transcription co-activator without typical DNA-binding domains, has been implicated in control of cell growth and malignant transformation in Rat1 cells. In this report, we provide evidence that Cited2 is an important regulator of transforming growth factor (TGF)-beta signaling. Overexpression of Cited2 enhanced TGF-beta-mediated transcription of a Smad-Binding Element-containing luciferase reporter construct, SBE4-Luc. This may occur through a direct physical association of Cited2 with Smads 2 and 3, as supported by co-immunoprecipitation, mammalian two-hybrid and glutathione S-transferase-pull down assays. The transcription factor p300, which binds to Smad3, was shown to further enhance the interaction between Cited2 and Smad3, and the transcriptional responses of Smad3 by Cited2 in reporter assays. Cited2 enhances TGF-beta-mediated upregulation of matrix metalloproteinase 9 (MMP9) in Cited2 inducible mouse embryo fibroblasts. Overexpression of Cited2 enhanced TGF-beta-mediated MMP9 promoter reporter activity. Moreover, knockdown of Cited2 in MDA-MB-231 cells attenuated TGF-beta-mediated upregulation of MMP9 and TGF-beta-mediated cell invasion. Chromatin immunoprecipitation showed that Cited2 and Smad3 were recruited to MMP9 promoter upon TGF-beta stimulation. This is the first demonstration that Cited2 functions as a Smad3/p300-interacting transcriptional co-activator in modulating the expression of MMP9, which could affect tumor cell invasion mediated by TGF-beta.


Subject(s)
DNA-Binding Proteins/metabolism , Matrix Metalloproteinase 9/genetics , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Deletion , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Polymerase Chain Reaction , Promoter Regions, Genetic , Rats , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Trans-Activators/genetics , Transfection , Up-Regulation , p300-CBP Transcription Factors
8.
Oncogene ; 25(41): 5591-600, 2006 Sep 14.
Article in English | MEDLINE | ID: mdl-16636667

ABSTRACT

Transforming growth factor-beta (TGF-beta) elicits a potent growth inhibitory effect on many normal cells by binding to specific serine/threonine kinase receptors and activating specific Smad proteins, which regulate the expression of cell cycle genes, including the p21 cyclin-dependent kinase (CDK) inhibitor gene. Interestingly, cancer cells are often insensitive to the anti-mitogenic effects of TGF-beta for which the molecular mechanisms are not well understood. In this study, we found that the candidate prostate cancer susceptibility gene ELAC2 potentiates TGF-beta/Smad-induced transcriptional responses. ELAC2 associates with activated Smad2; the C-terminal MH2 domain of Smad2 interacts with the N-terminal region of ELAC2. Small interfering siRNA-mediated knock-down of ELAC2 in prostate cells suppressed TGF-beta-induced growth arrest. Moreover, ELAC2 was shown to specifically associate with the nuclear Smad2 partner, FAST-1 and to potentiate the interaction of activated Smad2 with transcription factor Sp1. Furthermore, activation of the p21 CDK inhibitor promoter by TGF-beta is potentiated by ELAC2. Taken together our data indicate an important transcriptional scaffold function for ELAC2 in TGF-beta/Smad signaling mediated growth arrest.


Subject(s)
Cell Division/genetics , Genetic Predisposition to Disease , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Smad Proteins/physiology , Transforming Growth Factor beta/physiology , Animals , Base Sequence , COS Cells , Cell Division/physiology , Cell Line, Tumor , Chlorocebus aethiops , DNA Primers , Humans , Male , Neoplasm Proteins/metabolism , Prostatic Neoplasms/pathology , Protein Binding , RNA, Small Interfering , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
9.
J Biol Chem ; 276(28): 26614-21, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11320089

ABSTRACT

The mechanism by which transforming growth factor-beta1 (TGF-beta1) induces apoptosis of prostate epithelial cells was studied in the NRP-154 rat prostate epithelial cell line. TGF-beta 1 down-regulates expression of Bcl-xL and poly(ADP-ribosyl)polymerase (PARP), promotes cytochrome c release, up-regulates expression of latent caspase-3, and activates caspases 3 and 9. We tested the role of Bcl-xL in this cascade by stably overexpressing Bcl-xL to prevent loss by TGF-beta 1. Clones overexpressing Bcl-xL are resistant to TGF-beta 1 with respect to induction of apoptosis, cytochrome c release, activation of caspases 9 and 3, and cleavage of PARP; yet they remain sensitive to TGF-beta 1 by cell cycle arrest, induction of both fibronectin and latent caspase-3 expression, and loss of PARP expression. We show that Bcl-xL associates with Apaf-1 in NRP-154 cells; but this association does not inhibit the activation of caspases 9 and 3 by cytochrome c. Together, our data suggest that TGF-beta1 induces apoptosis through loss of Bcl-xL, leading to cytochrome c release and the subsequent activation of caspases 9 and 3. Moreover, our data demonstrate that the antiapoptotic effect of Bcl-xL occurs by inhibition of mitochondrial cytochrome c release and not through antagonizing Apaf-1-dependent processing of caspases 9 and 3.


Subject(s)
Apoptosis , Epithelial Cells/metabolism , Epithelial Cells/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Transforming Growth Factor beta/metabolism , Apoptosis/drug effects , Apoptotic Protease-Activating Factor 1 , Caspases/metabolism , Cell Line , Cytochrome c Group/metabolism , Enzyme Activation , Humans , Male , Prostate/metabolism , Prostate/pathology , Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/pharmacology , bcl-X Protein
10.
Proc Natl Acad Sci U S A ; 97(11): 5924-9, 2000 May 23.
Article in English | MEDLINE | ID: mdl-10811875

ABSTRACT

The c-ski protooncogene encodes a transcription factor that binds DNA only in association with other proteins. To identify co-binding proteins, we performed a yeast two-hybrid screen. The results of the screen and subsequent co-immunoprecipitation studies identified Smad2 and Smad3, two transcriptional activators that mediate the type beta transforming growth factor (TGF-beta) response, as Ski-interacting proteins. In Ski-transformed cells, all of the Ski protein was found in Smad3-containing complexes that accumulated in the nucleus in the absence of added TGF-beta. DNA binding assays showed that Ski, Smad2, Smad3, and Smad4 form a complex with the Smad/Ski binding element GTCTAGAC (SBE). Ski repressed TGF-beta-induced expression of 3TP-Lux, the natural plasminogen activator inhibitor 1 promoter and of reporter genes driven by the SBE and the related CAGA element. In addition, Ski repressed a TGF-beta-inducible promoter containing AP-1 (TRE) elements activated by a combination of Smads, Fos, and/or Jun proteins. Ski also repressed synergistic activation of promoters by combinations of Smad proteins but failed to repress in the absence of Smad4. Thus, Ski acts in opposition to TGF-beta-induced transcriptional activation by functioning as a Smad-dependent co-repressor. The biological relevance of this transcriptional repression was established by showing that overexpression of Ski abolished TGF-beta-mediated growth inhibition in a prostate-derived epithelial cell line.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation/drug effects , Neoplasm Proteins/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/pharmacology , Cell Nucleus/chemistry , DNA-Binding Proteins/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Macromolecular Substances , Melanoma/pathology , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Smad2 Protein , Smad3 Protein , Trans-Activators/antagonists & inhibitors , Transcription Factors/physiology , Transcription, Genetic/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Two-Hybrid System Techniques
12.
Cell Growth Differ ; 11(1): 1-10, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10672898

ABSTRACT

Retroviral insertional mutagenesis was used to select mutant NRP-154 rat prostate carcinoma cells resistant to transforming growth factor (TGF)-beta-induced cell death. Similar to the parental cells, a mutant clone, M-NRP1, expressed TGF-beta receptors and was still responsive to induction both of direct target genes by TGF-beta and of apoptosis by staurosporine or okadaic acid. In contrast, indicators of cell growth, strongly suppressed by TGF-beta in the parental cells, were unaffected in M-NRP1 cells. M-NRP1 cells overexpress the antiapoptotic protein, Bcl-xL, and show dysregulated expression and localization of a protein related to a novel human septin, ARTS (designation of apoptotic response to TGF-beta signals), cloned by homology to an exonic sequence flanked by the viral long terminal repeats in M-NRP1 cells and shown to make cells competent to undergo apoptosis in response to TGF-beta. We propose that ARTS might operate within the same apoptotic pathway as Bcl-xL and that M-NRP1 cells could serve as a useful model for characterization of this pathway.


Subject(s)
Apoptosis/drug effects , Signal Transduction/genetics , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis/genetics , Cell Division/drug effects , In Situ Nick-End Labeling , Male , Mutagenesis, Insertional , Okadaic Acid/pharmacology , Prostate , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Rats , Receptors, Transforming Growth Factor beta/metabolism , Retroviridae/genetics , Staurosporine/pharmacology , Transfection , Tumor Cells, Cultured , bcl-X Protein
13.
Nat Cell Biol ; 2(12): 915-21, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11146656

ABSTRACT

Here we describe a protein product of the human septin H5/PNUTL2/CDCrel2b gene, which we call ARTS (for apoptosis-related protein in the TGF-beta signalling pathway). ARTS is expressed in many cells and acts to enhance cell death induced by TGF-beta or, to a lesser extent, by other apoptotic agents. Unlike related septin gene products, ARTS is localized to mitochondria and translocates to the nucleus when apoptosis occurs. Mutation of the P-loop of ARTS abrogates its competence to activate caspase 3 and to induce apoptosis. Taken together, these observations expand the functional attributes of septins previously described as having roles in cytokinesis and cellular morphogenesis.


Subject(s)
Apoptosis/physiology , Cytoskeletal Proteins/physiology , GTP Phosphohydrolases/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Caspases/metabolism , Cloning, Molecular , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , DNA Primers/genetics , Enzyme Activation , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , Gene Expression , Humans , Mitochondria/metabolism , Molecular Sequence Data , Septins , Sequence Homology, Amino Acid , Signal Transduction , Transfection , Transforming Growth Factor beta/physiology
14.
Cancer Res ; 59(19): 4834-42, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10519393

ABSTRACT

Transforming growth factor (TGF)-betas are multifunctional growth factors, the properties of which include the potent inhibition of epithelial cell growth. Expression patterns of TGF-betas and TGF-beta receptors in the normal prostate indicate that these growth regulators play key roles in prostatic development and proliferative homeostasis. Importantly, TGF-beta receptor levels are frequently diminished in malignant human prostate tissue. To test the hypothesis that loss of TGF-beta responsiveness is causally involved in the tumorigenic process, we have used retroviral transduction to introduce a dominant-negative mutant type II TGF-beta receptor (DNR) into the premalignant rat prostatic epithelial cell line, NRP-152. High-level expression of the DNR abolished the ability of TGF-beta to inhibit cell growth, to promote cell differentiation, and to induce apoptosis, and it partially blocked the induction of extracellular matrix gene expression. When injected into nude mice, NRP-152-DNR cells formed carcinomas at 13 of 34 sites, compared with 0 of 30 sites for parental and control cells (P = 0.0001). We conclude that the type II TGF-beta receptor is an important tumor suppressor in the prostate, and furthermore, that loss of TGF-beta responsiveness can contribute early in the tumorigenic process by causing the malignant transformation of preneoplastic cells.


Subject(s)
Cell Transformation, Neoplastic , Extracellular Matrix Proteins/genetics , Gene Expression Regulation/drug effects , Prostatic Neoplasms/pathology , Receptors, Transforming Growth Factor beta/physiology , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Epithelial Cells , Humans , Male , Mice , Mice, Nude , Prostate , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases , Rats , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Recombinant Proteins/metabolism , Transfection , Transplantation, Heterologous
15.
Mol Cell Endocrinol ; 153(1-2): 29-38, 1999 Jul 20.
Article in English | MEDLINE | ID: mdl-10459851

ABSTRACT

Recent in vivo and in vitro studies suggest that retinoic acid receptor (RAR)-mediated processes may be involved in androgen regulation of prostate cells in a manner that may be altered during prostatic carcinogenesis. We tested this hypothesis in the newly established carcinoma and non-carcinoma rat prostate epithelial cell lines, NRP-154 and NRP-152, respectively. In DMEM/F-12 medium supplemented with 10% charcoal stripped fetal calf serum (cFCS), the number of both NRP-152 and NRP-154 cells were stimulated by testosterone (T), with a 4-fold greater effect in NRP-152 than in NRP-154 cells. Retinoic acid (RA) alone also stimulated the growth of NRP-152 cells, but failed to induce cell growth of NRP-154 cells. Importantly, the level of RAR alpha mRNA was elevated whereas the levels of RAR gamma and androgen receptor (AR) mRNA were lower in NRP-154 cells compared to those in NRP-152 cells. Treatment of NRP-152 cells with increasing doses of T resulted in a dose-dependent decrease and rebound of the level of RAR alpha and gamma mRNA in NRP-152 cells; these effects were not apparent, if not reversed, in NRP-154 cells. Both ligand binding and Western blot analyses revealed that epidermal growth factor receptor (EGF-R) was stimulated by 20 nM T but was suppressed by 0.1 microM RA, which also attenuated the stimulating effects of T on EGF-R in NRP-152 and to a lesser extent in NRP-154 cells. The differences in the level and androgen regulation of RAR mRNAs and reciprocal regulation of EGF-R expression by T and RA between NRP-154 and NRP-152 cells suggest that variations in the EGF-R and RAR signal events may contribute to differences in growth rate between these two cell lines.


Subject(s)
Cell Division/drug effects , ErbB Receptors/genetics , Prostatic Neoplasms/metabolism , Receptors, Retinoic Acid/genetics , Testosterone/pharmacology , Tretinoin/pharmacology , Animals , Blotting, Western , Cell Line , Culture Media , DNA/biosynthesis , ErbB Receptors/metabolism , Gene Expression/drug effects , Male , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology , RNA, Messenger/metabolism , Rats , Receptors, Retinoic Acid/metabolism , Tumor Cells, Cultured
16.
Prostate ; 39(3): 205-12, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10334110

ABSTRACT

BACKGROUND: The clonally derived rat prostatic epithelial cell line NRP-152 was examined to determine its ability to differentiate in a tissue recombination model. METHODS: NRP-152 cells alone, or combined with urogenital mesenchyme (UGM) or 10T1/2 fibroblasts, were grafted beneath the renal capsule of athymic rodent hosts. After 1 and 3 months, grafts were examined grossly and immunohistochemically. RESULTS: NRP-152 cells grafted alone formed small (10-25 mg) grafts without recognizable architecture. NRP-152 cells recombined with UGM formed larger grafts (50-100 mg after 28 days) containing glandular epithelium. Columnar luminal cells expressed cytokeratins 8 and 18 and rat prostatic secretory markers (DP-1 and DP-2). The epithelial ducts were surrounded by well-differentiated smooth muscle. The glandular epithelial cells were shown to be of rat origin. NRP-152 + 10T1/2 tissue recombinants formed small grafts (10-40 mg wet weight) after 1 month. The epithelial component of these grafts formed solid unbranched cords expressing cytokeratins 5 and 14; no glandular epithelial structures were observed. The stromal matrix was densely packed with a few cells expressing alpha-actin. CONCLUSIONS: A clonally derived prostatic epithelial cell line can form structurally and functionally normal prostatic tissue. This suggests that prostatic basal and luminal epithelial cells can be derived from a common progenitor.


Subject(s)
Cell Communication , Prostate/transplantation , Animals , Cell Differentiation , Cell Line , Epithelial Cells/cytology , Epithelial Cells/transplantation , Fibroblasts/cytology , Fibroblasts/transplantation , Male , Mesoderm/cytology , Mesoderm/transplantation , Mice , Mice, Nude , Prostate/cytology , Rats , Stromal Cells/cytology , Stromal Cells/transplantation , Transplantation, Heterologous
17.
J Cell Sci ; 112 ( Pt 2): 169-79, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9858470

ABSTRACT

The role of basal epithelial cells in prostatic function, development and carcinogenesis is unknown. The ability of basal prostatic epithelial cells to acquire a luminal phenotype was explored in vitro using the NRP-152 rat dorsal-lateral prostate epithelial cell line as a model system. NRP-152, which was spontaneously immortalized and clonally derived, is an androgen-responsive and nontumorigenic cell line that has a basal cell phenotype under normal growth conditions. However, when placed in mitogen-deficient media, these cells undergo a dramatic morphological change to a luminal phenotype. Under these growth-restrictive conditions, immunocytochemical analysis shows that NRP-152 cells acquire the luminal markers Z0-1 (a tight-junction associated protein), occludin (integral tight-junction protein), and cytokeratin 18, and lose the basal markers cytokeratins 5 and 14. Total protein and mRNA levels of cytokeratins 8, 18, c-CAM 105 (the calcium-independent cell adhesion molecule) and Z0-1, as detected by western and/or northern blot analyses, respectively, are induced, while cytokeratin 5 and 15 are lost, and occludin is unchanged. Concomitant with this differentiation, expression of transforming growth factor-beta2 (TGF-beta2), TGF-beta3, and TGF-beta receptor type II (TbetaRII) is induced, while those of TGF-beta1 and TbetaRI remain essentially unchanged. Mitogens, such as insulin-like growth factor-I and dexamethasone inhibit luminal differentiation, while exogenous TGF-beta induces such differentiation. These data together with TGF-beta neutralization experiments using pan-specific antibody implicate an important role for autocrine TGF-beta in the induction of the luminal differentiation.


Subject(s)
Prostate/cytology , Prostate/metabolism , Animals , Apoptosis/drug effects , Biomarkers , Cell Differentiation/drug effects , Cell Line , Dexamethasone/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Glucocorticoids/metabolism , Immunohistochemistry , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Male , Phenotype , Prostate/drug effects , Rats , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
18.
J Cell Physiol ; 175(2): 184-92, 1998 May.
Article in English | MEDLINE | ID: mdl-9525477

ABSTRACT

We have investigated the role of autocrine/paracrine TGF-beta secretion in the regulation of cell growth by androgens as demonstrated by its inhibition by two androgen response modifiers; the nonsteroidal antiandrogen hydroxyflutamide (OHF), believed to act by inhibiting androgen binding to androgen receptors, or finasteride, an inhibitor of 5alpha-reductase, the enzyme necessary for the conversion of testosterone to 5alpha-dihydrotestosterone (DHT), using the nontumorigenic rat prostatic epithelial cell line NRP-152. Growth of these cells was stimulated three- to sixfold over control by either testosterone or DHT under serum-free culture conditions. This was accompanied by a two- to threefold decrease in the secretion rate of TGF-beta1, -beta2, and -beta3. Finasteride reversed the ability of testosterone but not DHT to stimulate growth and downregulate expression of TGF-beta1, -beta2, and -beta3 in a dose-dependent fashion, suggesting that this activity of testosterone required its conversion to DHT. OHF antagonized the stimulatory effects of DHT on NRP-152 cell growth but could reverse the inhibitory effects of DHT only on TGF-beta2 and TGF-beta3 and not TGF-beta1 secretion. This suggests that either TGF-beta1 regulation by DHT or the androgen antagonism of OHF occurs independent of androgen receptor binding. Neutralizing antibodies to TGF-beta (pantropic and isoform-specific) were able to block the ability of finasteride to antagonize the effects of testosterone nearly completely while only partially inhibiting the antiandrogenic effects of OHF. Thus, the ability of androgens to stimulate growth of NRP-152 cells involves the downregulation of the production of TGF-beta1, -beta2, and -beta3 in addition to other growth-stimulatory mechanisms.


Subject(s)
Androgens/pharmacology , Cell Division/drug effects , Prostate/drug effects , Transforming Growth Factor beta/metabolism , Androgen Antagonists/pharmacology , Animals , Antibodies/immunology , Antibodies/pharmacology , Cell Line , Dihydrotestosterone/antagonists & inhibitors , Dihydrotestosterone/pharmacology , Down-Regulation/physiology , Finasteride/pharmacology , Flutamide/analogs & derivatives , Flutamide/pharmacology , Male , Rats , Testosterone/antagonists & inhibitors , Testosterone/pharmacology
19.
Anal Biochem ; 250(2): 147-52, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9245431

ABSTRACT

Apoptosis is an important and common pathway of cellular death. Differentiation from cellular necrosis and quantitation of apoptosis within the milieu of necrosis are analytical challenges. We describe the use of the RIT120 digital imaging software package for quantitative and qualitative analysis of apoptotic DNA ladders induced by a variety of agents, such as serum, tumor necrosis factor-alpha, transforming growth factor-beta1, and nitric oxide. Autoradiographs of DNA ladders are densitometrically scanned to yield a set of curves with peaks corresponding to specific DNA fragments, thereby allowing quantitative subtraction of concurrent DNA degradation from necrotic death. Integration of the areas specifically under the peaks yields a quantitative measure of apoptosis. We provide a useful, rapid, and objective means to quantitate apoptosis, using relatively inexpensive hardware and software.


Subject(s)
DNA Fragmentation , Image Processing, Computer-Assisted/methods , Animals , Apoptosis/genetics , Autoradiography , Humans , Male , Rats , Tumor Cells, Cultured
20.
Cancer Res ; 56(22): 5146-9, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8912849

ABSTRACT

Transforming growth factor-beta1 (TGF-beta1), which is induced in the prostate following castration, has been speculated to mediate apoptosis of epithelial cells during prostatic involution. Here, we report the first evidence of a direct effect of TGF-beta on induction of apoptosis in prostatic epithelial cells in vitro, using NRP-152 nontumorigenic and NRP-154 tumorigenic rat prostatic epithelial cell lines. TGF-beta1 induces apoptosis of both cell lines within 24 h, as shown by a decrease in cell viability, in situ DNA nick-end labeling, and internucleosomal DNA fragmentation. Moreover, the ability of TGF-beta to induce apoptosis of NRP-152 is strictly dependent on culture conditions, because dexamethasone enhances while insulin and insulin-like growth factor-I specifically block apoptosis induced by TGF-beta. We suggest that TGF-betas are direct physiological regulators of apoptosis of prostatic epithelial cells.


Subject(s)
Apoptosis/physiology , DNA, Neoplasm/drug effects , Prostate/physiopathology , Prostatic Neoplasms/physiopathology , Transforming Growth Factor beta/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , DNA Fragmentation , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Male , Nucleosomes , Rats , Transforming Growth Factor beta/antagonists & inhibitors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...