transforming-growth-factor-alpha and Prostatic-Neoplasms

transforming-growth-factor-alpha has been researched along with Prostatic-Neoplasms* in 88 studies

Reviews

7 review(s) available for transforming-growth-factor-alpha and Prostatic-Neoplasms

ArticleYear
ErbB receptors: possible therapeutic targets in prostate cancer?
    BJU international, 2003, Volume: 92, Issue:9

    Topics: Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Cetuximab; ErbB Receptors; Humans; Male; Prostatic Neoplasms; Protein-Tyrosine Kinases; Receptor, ErbB-2; Transforming Growth Factor alpha

2003
Role of growth factors, steroid and peptide hormones in the regulation of human prostatic tumor growth.
    The Journal of steroid biochemistry and molecular biology, 1996, Volume: 56, Issue:1-6 Spec N

    Previous work carried out in the authors' laboratory has shown that LHRH agonists directly inhibit the proliferation of hormone-responsive and hormone-independent human prostatic cancer cell lines (respectively LNCaP and DU145). In addition, the hormone-dependent LNCaP cells respond to a challenge with testosterone with an increase in growth rate. The following experiments have been performed to investigate whether the LHRH agonists might act by interfering with the stimulatory actions of either the EGF/TGF alpha system or androgens. The results obtained in LNCaP and DU145 cells show that LHRH agonists counteract the mitogenic action of the EGF/TGF alpha system. This effect is mediated by a decrease in the concentration of EGF receptors. In addition, in the hormone-dependent LNCaP cells, the treatment with LHRH agonists antagonizes the proliferation promoting effect of testosterone, which in turn appears to be mediated by the activation of the locally expressed EGF/TGF alpha system. Finally, the results suggest the presence in LNCaP cells of a soluble peptidase able to degrade LHRH. In conclusion, the present data suggest an intimate interplay among the actions of LHRH agonists, of androgens and of growth factors, thus, supporting the hypothesis that LHRH agonists may interfere with the EGF/TGF alpha stimulatory loop and with androgens in the control of the proliferation of human prostatic tumors.

    Topics: Androgen Antagonists; Androgens; Antineoplastic Agents, Hormonal; Brain Neoplasms; Carcinoma; Cell Division; Endopeptidases; Epidermal Growth Factor; Gonadotropin-Releasing Hormone; Humans; Lymphatic Metastasis; Male; Neoplasm Proteins; Neoplasms, Hormone-Dependent; Prostatic Neoplasms; Somatostatin; Testosterone; Transforming Growth Factor alpha; Tumor Cells, Cultured

1996
Dual regulation of the epidermal growth factor family of growth factors in breast cancer by sex steroids and protein kinase C.
    The Journal of steroid biochemistry and molecular biology, 1996, Volume: 57, Issue:1-2

    There has been increased interest in the last few years in seeking a better understanding of the local regulation of polypeptide growth factors by systemic hormones, such as sex steroids and by polypeptide hormones. Growth factors and systemic hormones play pivotal roles in hormone-regulated cancers such as breast cancer. In this review, we discuss the regulation of members of the epidermal growth factor (EGF) family by sex steroids and by regulators of the polypeptide hormone signal transduction enzyme termed protein kinase C (PKC). Regulation of the EGF family of genes will be discussed as a model system to evaluate interactions between these two important types of regulatory pathways in breast cancer.

    Topics: Breast Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Gonadal Steroid Hormones; Humans; Male; Phorbol Esters; Prostatic Neoplasms; Protein Kinases; Receptors, Estrogen; Signal Transduction; Steroids; Transforming Growth Factor alpha

1996
Epidermal growth factor-related peptides and the epidermal growth factor receptor in normal and malignant prostate.
    World journal of urology, 1995, Volume: 13, Issue:5

    Epidermal growth factor (EGF) and transforming growth factor-alpha (TGF alpha) are two closely related peptides that interact with cell-surface epidermal growth factor receptors (EGFR) to induce receptor tyrosine phosphorylation and activation of intracellular signal-transduction pathways. EGF appears to be the predominant EGF-related growth factor in the normal prostate and in benign prostatic hyperplasia (BPH). Evidence indicates that EGF and TGF alpha are important for maintainence of the structural and functional integrity of the benign prostatic epithelium. The EGF-related peptides are primarily localized to the secretory epithelium of the benign prostate, and their production and secretion is augmented by the presence of circulating androgens. EGFR are located in the basal/neuroendocrine (NE) compartment of the benign prostate and exhibit relatively androgen-independent expression. The EGF-related peptides and EGFR are also present in neoplastic prostatic tissues. There is currently no direct evidence to implicate EGFR activation in the pathogenesis of BPH. However, the EGF-related peptides appear to play a functional role in the growth of prostatic carcinoma cells, with TGF alpha being the predominant growth factor. Numerous investigators have demonstrated the functional significance of a TGF alpha/EGFR-mediated autocrine growth pathway in cultured prostatic carcinoma cells. Studies of cultured prostate cancer cells, but not normal epithelial cells, demonstrate constitutive activation of EGFR. Androgen-independent cancer cells exhibit more EGFR expression and phosphorylation than do androgen-responsive prostate cancer cells. Most studies indicate that EGFR do not play a functional role in androgen-stimulated growth of prostate cancer cells. Several studies have correlated EGFR expression with increased nuclear size and tumor dedifferentiation. Future studies should focus on determining both the prognostic significance of EGFR expression and whether manipulation of EGFR-mediated growth can be exploited for therapeutic benefit in human prostate cancer.

    Topics: Animals; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation; Gene Expression Regulation, Neoplastic; Humans; Male; Prostate; Prostatic Neoplasms; Transforming Growth Factor alpha

1995
Androgen-dependent prostatic tumors: biosynthesis and possible actions of LHRH.
    The Journal of steroid biochemistry and molecular biology, 1994, Volume: 49, Issue:4-6

    Testosterone (T) is the major exogenous stimulus for the growth of prostatic carcinoma. It is believed that the proliferative action of T may be mediated by locally expressed growth modulatory factors. Recent evidence from our laboratory suggests that a LHRH (or a LHRH-like) loop might be expressed in human prostatic tumor cells. To verify this hypothesis, we have studied whether a mRNA for LHRH is expressed in the human androgen-responsive prostatic cancer cell line LNCaP, using the reverse transcription-polymerase chain reaction technique in the presence of a pair of specific oligonucleotide primers. A cDNA band of the expected size was obtained from LNCaP cells; this band hybridized with a 32P-labeled LHRH oligonucleotide probe and its sequence showed a complete match with the reported sequence of the human placental LHRH cDNA. These observations indicate that the mRNA coding for LHRH is expressed in LNCaP cells and suggest that a LHRH (or a LHRH-like) peptide might be produced by these cells. To clarify the possible action of this peptide, LNCaP cells were grown in a steroid-free medium and treated with a LHRH antagonist. The treatment resulted in a significant increase of tumor cell growth. These data clearly indicate that the LHRH system expressed in LNCaP cells plays an inhibitory role on cell proliferation, and that this system seems to be regulated in a negative way by steroids. An EGF/TGF alpha autocrine stimulatory loop (peptides, receptors, intracellular signals) is also functional in these cells. Treatment of LNCaP cells grown in serum-free conditions (i.e. in the absence of exogenous growth factors) with a monoclonal antibody against the EGF receptor, or with immunoneutralizing antibodies against EGF or TGF alpha, resulted in a significant decrease of cell proliferation. T positively regulates this EGF/TGF alpha system by increasing the concentration of EGF binding sites. The present data indicate that an inhibitory LHRH (or LHRH-like) system is expressed in LNCaP cells and participates in the local mechanisms regulating tumor cell proliferation together with an EGF/TGF alpha stimulatory loop. Both systems appear to be modulated by T.

    Topics: Androgens; Animals; Epidermal Growth Factor; Gonadotropin-Releasing Hormone; Humans; Male; Neoplasms, Hormone-Dependent; Prostatic Neoplasms; Transforming Growth Factor alpha

1994
[Peptide growth factors in the prostate].
    Pathologie-biologie, 1993, Volume: 41, Issue:8

    Growth and development of the prostate depend on androgen action and other factors. Androgens act directly by specific nuclear receptors and induce or control many effects mediated by peptide growth factors on both epithelial and stromal cells. The major important peptide growth factors detected in the prostate are Fibroblast Growth Factors, Transforming Growth Factors, and Epidermal Growth Factor. These factors are not prostate specific. They are produced by epithelial or stromal cells and regulate the growth of these two cell compartments through autocrine or paracrine pathways. Overproduction of these factors or modification in affinity or number of cell receptors may participate in the two main prostatic pathologies: benign hyperplasia or adenocarcinoma. Recently, other factors have been evidenced in the prostate: Interleukine 6, Nerve Growth Factor or Insulin-like Growth factors. The study of the localization and the effects of these factors may be crucial to understand the prostatic development and diseases and may suggest new targets for therapy.

    Topics: Adenocarcinoma; Epidermal Growth Factor; Fibroblast Growth Factors; Humans; Interleukin-6; Male; Nerve Growth Factors; Prostate; Prostatic Hyperplasia; Prostatic Neoplasms; Suramin; Transforming Growth Factor alpha; Transforming Growth Factor beta

1993
Oncogenes in prostate cancer. An update.
    Cancer, 1993, Feb-01, Volume: 71, Issue:3 Suppl

    Oncogenes have been implicated in the carcinogenic development of many diverse types of human malignancies. For some cancers, the expression of specific oncogenes has been shown to have diagnostic or prognostic value. By contrast currently, no oncogene has been correlated conclusively with the initiation or progression of prostate cancer. The ras oncogene has been investigated the most thoroughly for its involvement in prostate cancer, but ras does not appear to play a significant role in the development of this malignancy. Several years ago, limited studies hinted at the possibility of overexpression of the myc oncogene and aberrant expression of the sis oncogene in prostate cancer, but additional studies to clarify the involvement of these oncogenes have not been done. Oncogenic activity of growth factors or growth factor receptors in prostate cancer has been suggested but not amply demonstrated. Current dogma indicates that oncogenes exist in prostate cancer, but these will be identified only by more intensive investigation.

    Topics: Epidermal Growth Factor; Genes, myc; Genes, ras; Humans; Male; Oncogenes; Prostatic Neoplasms; Transforming Growth Factor alpha

1993

Other Studies

81 other study(ies) available for transforming-growth-factor-alpha and Prostatic-Neoplasms

ArticleYear
Prostate cancer expressing membrane-bound TGF-α induces bone formation mediated by the autocrine effect of prostaglandin E
    Biochemical and biophysical research communications, 2023, 02-12, Volume: 644

    Prostate cancer highly metastasizes to bone, and such cancer is associated with severe bone resorption and bone formation at the site of metastasis. Prostaglandin E

    Topics: Bone Resorption; Core Binding Factor Alpha 1 Subunit; Cyclooxygenase 2; Dinoprostone; ErbB Receptors; Humans; Male; Osteoblasts; Osteogenesis; Prostaglandins; Prostatic Neoplasms; Transforming Growth Factor alpha

2023
Circular RNA circANKS1B acts as a sponge for miR-152-3p and promotes prostate cancer progression by upregulating TGF-α expression.
    The Prostate, 2021, Volume: 81, Issue:5

    A growing number of studies indicate that circular RNAs (circRNAs) play critical roles in human diseases, and show great potential as biomarkers and therapeutic targets. This study aimed to investigate the expression and function of circANKS1B in prostate cancer (PC).. The expression of circANKS1B and miR-152-3p was analyzed by real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR). Cell migration and invasion were measured using a transwell assay. The interaction between circANKS1B and miR-152-3p was confirmed by a dual-luciferase reporter gene assay. Rescue experiments were conducted to determine whether circANKS1B regulated the invasion of PC cells via the circANKS1B-miR-152-3p-TGF-α pathway.. The expression of circANKS1B was markedly upregulated in both PC cells and tissues. Moreover, high circANKS1B expression was associated with poor prognosis in PC patients. Dual-luciferase reporter assay indicated that circANKS1B directly bound to miR-152-3p. Furthermore, circANKS1B negatively regulated miR-152-3p expression. Knockdown of circANKS1B markedly suppressed cell migration and invasion and TGF-α expression in PC cells, whereas the effects of circANKS1B silencing were reversed by miR-152-3p deficiency. In addition, the impact of miR-152-3p silencing on invasion of circANKS1B-deficient PC cells was also abrogated by TGF-α deficiency. Overall, circANKS1B acts as a sponge for miR-152-3p to promote PC progression by upregulating TGF-α expression.. Our findings reveal that circANKS1B may be a potential prognostic biomarker and therapeutic target for PC.

    Topics: Aged; Cell Line, Tumor; Cell Movement; Disease Progression; Gene Expression; Humans; Intracellular Signaling Peptides and Proteins; Male; MicroRNAs; Middle Aged; Neoplasm Grading; Neoplasm Invasiveness; PC-3 Cells; Prognosis; Prostatic Neoplasms; RNA, Circular; Transforming Growth Factor alpha; Up-Regulation

2021
CX3CL1 increases invasiveness and metastasis by promoting epithelial-to-mesenchymal transition through the TACE/TGF-α/EGFR pathway in hypoxic androgen-independent prostate cancer cells.
    Oncology reports, 2016, Volume: 35, Issue:2

    Epithelial-to-mesenchymal transition (EMT) endows cancer cells with enhanced invasive and metastatic potential during cancer progression. Fractalkine, also known as chemokine (C-X3-C motif) ligand 1 (CX3CL1), the only member recognized so far that belongs to the CX3C chemokine subfamily, was reported to participate in the molecular events that regulate cell adhesion, migration and survival of human prostate cancer cells. However, the relationship between CX3CL1 and EMT remains unknown. We treated DU145 and PC-3 cells with CX3CL1 under hypoxic conditions. The migration and invasion abilities of DU145 and PC-3 cells were detected by Transwell assays. Induction of EMT was verified by morphological changes in the DU145 and PC-3 cells and analysis of protein expression of EMT markers such as E-cadherin and vimentin. To identify the involved signaling pathway in CX3CL1-induced EMT, activation of epidermal growth factor receptor (EGFR) was measured using western blot analysis, and Slug expression was detected with or without an EGFR inhibitor prior to CX3CL1 treatment. Concentrations of soluble and total TGF-α in the CX3CL‑treated DU145 cells were detected by ELISA. Additionally, we determined the involvement of the TACE/TGF-α/EGFR pathway in CX3CL1‑induced EMT using RNA interference and specific inhibitors. CX3CL1 increased the migration and invasiveness of the DU145 and PC-3 cells, and resulted in characteristic alterations of EMT. Our results demonstrated that TACE/TGF-α/EGFR pathway activation and subsequent upregulation of Slug expression were responsible for CX3CL1‑induced EMT, and contributed to the migration and inva-sion of prostate cancer cells. Inhibition of TACE/TGF-α/EGFR signaling reversed EMT and led to reduced migration and invasion abilities of the prostate cancer cells. We provide initial evidence that CX3CL1 exposure resulted in EMT occurrence and enhancement of cell migration and invasion through a mechanism involving activation of TACE/TGF-α/EGFR signaling. These findings revealed that CX3CL1 may serve as a new target for the treatment of prostate cancer.

    Topics: ADAM Proteins; ADAM17 Protein; Adenocarcinoma; Cell Hypoxia; Cell Line, Tumor; Cell Movement; Chemokine CX3CL1; Epithelial-Mesenchymal Transition; ErbB Receptors; Humans; Male; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasm Proteins; Prostatic Neoplasms; RNA Interference; RNA, Small Interfering; Signal Transduction; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor alpha; Up-Regulation

2016
MicroRNA-124 regulates TGF-α-induced epithelial-mesenchymal transition in human prostate cancer cells.
    International journal of oncology, 2014, Volume: 45, Issue:3

    Transforming growth factor-α (TGF-α) is upregulated in advanced stages of prostate cancer and strongly correlated with metastasis. However, the effect of TGF-α on epithelial-mesenchymal transition (EMT) in prostate cancer and the underlying mechanisms remain unclear. Recently, microRNAs have emerged as new regulators of EMT. This study found that treatment of DU145 cells with TGF-α suppressed the expression of epithelial marker E-cadherin and increased the expression of mesenchymal marker Vimentin as well as changed the cell morphology from cobblestone shape to spindle shape. The level of miR-124 was downregulated by TGF-α in several different cancer cell lines. Enforced expression of miR-124 abolished TGF-α-induced EMT. Slug was proven to be a target of miR-124 and mediated the inhibitory effect of miR-124 on TGF-α-induced EMT. Furthermore, overexpression of miR-124 reduced the migratory and invasive capacity of TGF-α-treated DU145 cells. In conclusion, our findings suggest that miR-124 inhibits TGF-α-induced EMT in DU145 cells by targeting Slug. Thus, miR-124 may be a potential target for prostate cancer therapeutic intervention.

    Topics: Cell Line, Tumor; Cell Movement; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Hep G2 Cells; Humans; Male; MicroRNAs; Neoplasm Invasiveness; Prostatic Neoplasms; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor alpha

2014
Loss of EGFR signaling regulated miR-203 promotes prostate cancer bone metastasis and tyrosine kinase inhibitors resistance.
    Oncotarget, 2014, Jun-15, Volume: 5, Issue:11

    Activation of EGFR signaling pathway leads to prostate cancer bone metastasis; however, therapies targeting EGFR have demonstrated limited effectiveness and led to drug resistance. miR-203 levels are down-regulated in clinical samples of primary prostate cancer and further reduced in metastatic prostate cancer. Here we show that ectopic miR-203 expression displayed reduced bone metastasis and induced sensitivity to tyrosine kinase inhibitors (TKIs) treatment in a xenograft model. Our results demonstrate that the induction of bone metastasis and TKI resistance require miR-203 down regulation, activation of the EGFR pathway via altered expression of EGFR ligands (EREG and TGFA) and anti-apoptotic proteins (API5, BIRC2, and TRIAP1). Importantly, a sufficient reconstitution of invasiveness and resistance to TKIs treatment was observed in cells transfected with anti-miR-203. In prostate cancer patients, our data showed that miR-203 levels were inversely correlated with the expression of two EGFR ligands, EREG and TGFA, and an EGFR dependent gene signature. Our results support the existence of a miR-203, EGFR, TKIs resistance regulatory network in prostate cancer progression. We propose that the loss of miR-203 is a molecular link in the progression of prostate cancer metastasis and TKIs resistance characterized by high EGFR ligands output and anti-apoptotic proteins activation.

    Topics: 3' Untranslated Regions; Amphiregulin; Animals; Base Sequence; Bone Neoplasms; Cell Line, Tumor; Down-Regulation; EGF Family of Proteins; Epiregulin; ErbB Receptors; Heterografts; Humans; Male; Mice; Mice, Nude; MicroRNAs; Molecular Sequence Data; Neoplasm Metastasis; Prostatic Neoplasms; Protein Kinase Inhibitors; Proto-Oncogene Proteins; Proto-Oncogene Proteins p21(ras); ras Proteins; Signal Transduction; Transforming Growth Factor alpha

2014
[TGF-α increases neuroendocrine differentiation and strengthens chemoresistance in prostate cancer DU145 cells].
    Zhong nan da xue xue bao. Yi xue ban = Journal of Central South University. Medical sciences, 2013, Volume: 38, Issue:2

    To study whether TGF-α possesses similar EGF effect of enforcing neuroendocrine differentiation (NED) in prostate cancer cell line DU145 and determine the influence of NED induced by TGF-α on chemoresistance.. DU145 cells were divided into 3 groups: a group with 2% FBS, a group with 2%FBS+TGF-α 5 ng/mL and a group with 2%FBS+TGF-α 10 ng/mL. Morphological change in DU145 cells was observed after TGF-α treatment. Expression levels of NSE mRNA were detected with real time RT-PCR. Western blot was used to detect the expression levels of protein NSE, P-gp, MRP1 and Bcl-2. Cell cycles of DU145 cells in the 3 groups were examined with flow cytometry. MTT assay was used to evaluate the influence of TGF-α in chemoresistance.. Compared with DU145 cells cultured with 2% FBS, cells treated with 2% FBS+TGF-α were pleomorphic and pseudopodia extended. The expression level of NSE mRNA upregulated to (3.6±0.5) folds (P<0.05) and (10.1±0.1) folds (P<0.01). Western blot showed that the expression levels of protein NSE, Bcl-2, and MRP1 increased after treatment with different concentrations of TGF-α; P-gp was not detected. The proportion of DU145 cells in phase G1 decreased; proportions of cells in phase S and phase G2/M were increased after TGF-α treatment (5 μg/mL). At the same time, chemoresistance of DU145 cells to cisplatin increased.. TGF-α can increase NED in DU145 cells and enforce the chemoresistance to cisplatin.

    Topics: Antineoplastic Agents; Cell Line, Tumor; Cisplatin; Drug Resistance, Neoplasm; Endosomal Sorting Complexes Required for Transport; Humans; Male; Prostatic Neoplasms; Transforming Growth Factor alpha

2013
miR-152 controls migration and invasive potential by targeting TGFα in prostate cancer cell lines.
    The Prostate, 2013, Volume: 73, Issue:10

    MicroRNAs (miRNAs) are a class of short non-coding RNAs that function in diverse biological processes. Aberrant miR-152 expression has been frequently reported in various malignant tumors. However, the mechanism of miR-152 in prostate cancer (PCa) remains unclear. This study aims to determine the function of miR-152 in PCa cells and identify the novel molecular targets regulated by miR-152.. The expression levels of transforming growth factor-alpha (TGFα) were determined in three samples of PCa and adjacent non-tumorous tissues by Western blot analysis. miR-152 levels in 48 primary PCa and 15 non-malignant tissue samples were measured by qRT-PCR. The effects of forced miR-152 expression or TGFα knockdown on PCa cells were evaluated by cell migration and invasion assays, as well as Western blot analysis. Dual-luciferase reporter assay was used to identify binding sites between miR-152 and TGFα 3'-UTR.. TGFα was upregulated in PCa tissue samples compared with that in adjacent normal ones. miR-152 expression was significantly decreased in primary PCa samples compared with that in non-malignant samples. Patients with Gleason scores >7 exhibited lower miR-152 levels than those with lower scores. Moreover, low miR-152 expression is correlated with advanced pathological T-stages. Forced miR-152 expression or TGFα knockdown significantly reduced the migratory and invasive capabilities of PCa cells in vitro. TGFα is a direct target gene of miR-152.. Our findings suggest that miR-152 can act as a tumor suppressor that targets TGFα. miR-152 is a promising molecular target that inhibits PCa cell migration and invasion.

    Topics: Cell Line, Tumor; Cell Movement; Cell Proliferation; Gene Expression Regulation, Neoplastic; Humans; Male; MicroRNAs; Neoplasm Invasiveness; Prostatic Neoplasms; Transforming Growth Factor alpha; Up-Regulation

2013
Recruitment of normal stem cells to an oncogenic phenotype by noncontiguous carcinogen-transformed epithelia depends on the transforming carcinogen.
    Environmental health perspectives, 2013, Volume: 121, Issue:8

    Cancer stem cells (CSCs) drive tumor initiation, progression, and metastasis. The microenvironment is critical to the fate of CSCs. We have found that a normal stem cell (NSC) line from human prostate (WPE-stem) is recruited into CSC-like cells by nearby, but noncontiguous, arsenic-transformed isogenic malignant epithelial cells (MECs).. It is unknown whether this recruitment of NSCs into CSCs by noncontact co-culture is specific to arsenic-transformed MECs. Thus, we used co-culture to examine the effects of neighboring noncontiguous cadmium-transformed MECs (Cd-MECs) and N-methyl-N-nitrosourea-transformed MECs (MNU-MECs) on NSCs.. After 2 weeks of noncontact Cd-MEC co-culture, NSCs showed elevated metalloproteinase-9 (MMP-9) and MMP-2 secretion, increased invasiveness, increased colony formation, decreased PTEN expression, and formation of aggressive, highly branched duct-like structures from single cells in Matrigel, all characteristics typical of cancer cells. These oncogenic characteristics did not occur in NSCs co-cultured with MNU-MECs. The NSCs co-cultured with Cd-MECs retained self-renewal capacity, as evidenced by multiple passages (> 3) of structures formed in Matrigel. Cd-MEC-co-cultured NSCs also showed molecular (increased VIM, SNAIL1, and TWIST1 expression; decreased E-CAD expression) and morphologic evidence of epithelial-to-mesenchymal transition typical for conversion to CSCs. Dysregulated expression of SC-renewal genes, including ABCG2, OCT-4, and WNT-3, also occurred in NSCs during oncogenic transformation induced by noncontact co-culture with Cd-MECs.. These data indicate that Cd-MECs can recruit nearby NSCs into a CSC-like phenotype, but MNU-MECs do not. Thus, the recruitment of NSCs into CSCs by nearby MECs is dependent on the carcinogen originally used to malignantly transform the MECs.

    Topics: Animals; Blotting, Western; Cadmium; Carcinogens; Carcinoma; Cell Line, Tumor; Cell Transformation, Neoplastic; Environmental Pollutants; Epithelial Cells; Fluorescent Antibody Technique; Gene Expression Regulation, Neoplastic; Humans; Male; Matrix Metalloproteinases; Methylnitrosourea; Mice; Mice, Nude; Neoplastic Stem Cells; Polymerase Chain Reaction; Prostatic Neoplasms; Transforming Growth Factor alpha

2013
ADAM17 targets MMP-2 and MMP-9 via EGFR-MEK-ERK pathway activation to promote prostate cancer cell invasion.
    International journal of oncology, 2012, Volume: 40, Issue:5

    ADAM17, also known as tumor necrosis factor-α converting enzyme (TACE), is involved in proteolytic ectodomain shedding of cell surface molecules and cytokines. Although aberrant expression of ADAM17 has been shown in various malignancies, the function of ADAM17 in prostate cancer has not been clarified. In the present study, we sought to elucidate whether ADAM17 contributes to prostate cancer cell invasion, as well as the mechanism involved in the process. The expression pattern of ADAM17 was investigated in human prostate cancer cells. The results showed that ADAM17 expression levels are correlated with the invasive ability of androgen-independent prostate cancer cell lines. Further, ADAM17 was overexpressed in cells showing high invasion characteristics, activation of the EGFR-MEK-ERK pathway, up-regulation of MMP-2, MMP-9, and an increased TGF-α release into the supernatant. However, AG1478, PD98059 and antibody against TGF-α deactivating the EGFR-MEK-ERK signaling pathway, abolished up-regulation of MMP-2, MMP-9 and prevented cell invasion. In addition, cells with knockdown of ADAM17 by siRNA exhibited low invasive ability, deactivated EGFR-MEK-ERK signaling pathway, reduced TGF-α released and down-regulation of MMP-2, MMP-9. However, these effects could be reversed by simultaneous addition of TGF-α. These data demonstrated that ADAM17 contributes to androgen-independent prostate cancer cell invasion by shedding of EGFR ligand TGF-α, which subsequently activates the EGFR-MEK-ERK signaling pathway, leading finally to overexpression of MMP-2 and MMP-9. This study suggests that the ADAM17 expression level may be a new predictive biomarker of invasion and metastasis of prostate cancer, and ADAM17 could provide a target for treating metastatic PCa.

    Topics: ADAM Proteins; ADAM17 Protein; Cell Line, Tumor; Cell Movement; ErbB Receptors; Extracellular Signal-Regulated MAP Kinases; Humans; Male; MAP Kinase Kinase Kinases; Matrix Metalloproteinase 2; Matrix Metalloproteinase 9; Neoplasm Invasiveness; Prostatic Neoplasms; Protein Kinase Inhibitors; RNA Interference; Signal Transduction; Transfection; Transforming Growth Factor alpha; Up-Regulation

2012
Sox2 targets cyclinE, p27 and survivin to regulate androgen-independent human prostate cancer cell proliferation and apoptosis.
    Cell proliferation, 2012, Volume: 45, Issue:3

    Sox2 is a major transcription factor and the transforming growth factor-α (TGF-α)/EGFR autocrine loop is a hallmark of prostate cancer progression. In this study, we have evaluated the effects and potential mechanisms of Sox2 on cell proliferation and apoptosis, and investigated effects of TGF-α on expression of Sox2 on androgen-independent human prostate cancer cells.. Expression of Sox2 has been determined by RT-PCR, western blot analysis and immunocytochemistry, using RNAi and over-expression strategy to study functions of Sox2 in DU145 and PC-3 cells. Changes in level of proliferation, cell cycle and apoptosis profiles were measured by MTT, colony-forming, bromodeoxyuridine incorporation assays, cell cycle and annexin V analysis.. Sox2 was expressed in six human prostate cancer cell lines, and its inhibition reduced cell proliferation and induced apoptosis in DU145 cells. We have shown that knock-down of Sox2 inhibited G(1) to S phase transition concomitantly with down-regulation of cyclin E and up-regulation of p27 proteins. Conversely, over-expression of Sox2 led to the opposite effect in PC-3 cells but its inhibition induced apoptosis by down-regulation of survivin in DU145 cells. We also found that TGF-α up-regulated Sox2 and survivin protein expression via the EGFR/PI3K/AKT pathway.. Sox2 expression is necessary for cell proliferation and evasion of apoptosis in prostate cancer cells and TGF-α could regulate Sox2 and survivin expression by activating the EGFR/PI3K/AKT pathway.

    Topics: Androgens; Apoptosis; Cell Line, Tumor; Cell Proliferation; Cyclin E; Cyclin-Dependent Kinase Inhibitor p27; Down-Regulation; ErbB Receptors; G1 Phase Cell Cycle Checkpoints; Humans; Inhibitor of Apoptosis Proteins; Male; Phosphatidylinositol 3-Kinases; Prostatic Neoplasms; Proto-Oncogene Proteins c-akt; RNA Interference; RNA, Small Interfering; Signal Transduction; SOXB1 Transcription Factors; Survivin; Transforming Growth Factor alpha; Up-Regulation

2012
Growth factor/growth factor receptor loops in autocrine growth regulation of human prostate cancer DU145 cells.
    Acta biochimica Polonica, 2011, Volume: 58, Issue:3

    Autocrine growth factors produced by epithelial cells mediate the development and proliferation of neoplastic human prostate tissue. Various approaches have been used to down-regulate neoplastic growth of prostate cancer using natural flavonoids, soluble receptors, pseudo-ligands, monoclonal antibodies and tyrosine kinase inhibitors (tyrphostins). Selected growth factor/growth factor receptor loops (mainly TGFα/EGFR and IGFs/IGFIR) have been proposed as regulators of prostate cancer cell growth. We have previously determined that blockade of IGFIR or VEGF2R signaling pathways by tyrphostin AG1024 and SU1498 inhibits autocrine growth and viability of DU145 cells in vitro. Recently, we compared the activity of AG1024 and SU1498 with the inhibiting effect of tyrphostin A23 (a selective inhibitor of EGFR). The results described in this paper confirm that DU145 cells do not produce IGFI or EGF. In contrast, DU145 cells produce a great amount of VEGF, much more than TGFα (about 60-fold), and VEGF may be the real autocrine growth factor of the investigated cells. The results indicate that the growth of DU145 may be regulated by at least three autocrine loops: TGFα/EGFR, IGFII/IGFIR and VEGF/VEGFR2. Neither AG1024 nor SU1498 affected the production of TGFα substantially, which excludes the possibility that IGFRs or VEGFR2 inhibitors arrest the growth of these cells by inhibition of synthesis and/or secretion of TGFα. The obtained data indicate that all tree investigated tyrphostins (AG1024, SU1498 and A23) inhibit signal transmission by Akt (PKB), ERK(1/2), Src and STAT in a similar manner. A comparison of the effects of the investigated tyrphostins indicates that TGFα, IGFII and VEGF stimulate cell growth by affecting the same signaling pathway. The hypothesis was confirmed by the effect of the investigated tyrphostins on activation of EGFR. All these inhibitors decreased phosphorylation of EGFR to the same extent, and after the same time of incubation with cell culture. These results strongly suggest that stimulation of EGFR kinase is the main step in the initiation of mitogen signaling in DU145 cells, regardless of the type of ligand (TGFα, IGFs or VEGF) and their specific receptors.

    Topics: Cell Cycle; Cell Line, Tumor; Cell Proliferation; Cinnamates; ErbB Receptors; Gene Expression Regulation, Neoplastic; Humans; Insulin-Like Growth Factor II; Male; Phosphorylation; Prostatic Neoplasms; Proto-Oncogene Proteins c-akt; Receptor, IGF Type 1; Signal Transduction; Transforming Growth Factor alpha; Tyrphostins; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factor Receptor-2

2011
ETS-1 oncogenic activity mediated by transforming growth factor alpha.
    Cancer research, 2010, Jan-15, Volume: 70, Issue:2

    Inappropriate expression of Ets-1 is observed in a variety of human cancers, and its forced expression in cultured cells results in transformation, autonomous proliferation, and tumor formation. The basis by which Ets-1 confers autonomous growth, one of the primary hallmarks of cancer cells and a critical component of persistent proliferation, has yet to be fully explained. Using a variety of cancer cell lines, we show that inhibition of Ets-1 blocks tumor formation and cell proliferation in vivo and autonomous growth in culture. A screen of multiple diffusible growth factors revealed that inhibition of Ets-1 results in the specific downregulation of transforming growth factor alpha (TGFalpha), the proximal promoter region of which contains multiple ETS family DNA binding sites that can be directly bound and regulated by Ets-1. Notably, rescuing TGFalpha expression in Ets-1-silenced cells was sufficient to restore tumor cell proliferation in vivo and autonomous growth in culture. These results reveal a previously unrecognized mechanism by which Ets-1 oncogenic activity can be explained in human cancer through its ability to regulate the important cellular mitogen TGFalpha.

    Topics: Cell Growth Processes; Cell Line, Tumor; Cell Transformation, Neoplastic; Glioma; Humans; Kidney Neoplasms; Male; Neoplasms; Promoter Regions, Genetic; Prostatic Neoplasms; Protein Binding; Proto-Oncogene Protein c-ets-1; Transfection; Transforming Growth Factor alpha

2010
Multigene targeting of signal transduction pathways for the treatment of breast and prostate tumors: comparison between combination therapies employing bispecific oligonucleotides with either Rapamycin or Paclitaxel.
    Medical oncology (Northwood, London, England), 2009, Volume: 26, Issue:2

    Previous studies have demonstrated that monospecific antisense oligonucleotides (oligos) directed against mRNA encoding proteins associated with tumor growth, death, and survival are efficacious against breast and prostate tumors. Targeted proteins, associated with different signal transduction pathways, have included transforming growth factor-alpha [TGF-alpha (MR(1))], its binding site the epidermal growth factor receptor [EGFR (MR(2))] sharing sequence homology to the breast cancer prognostic marker Her-2/neu, an apoptosis inhibiting protein [bcl-2 (MR(4))], and the androgen receptor [AR (MR(5))]. In attempts to enhance antisense therapy, recent reports describe how two of the binding sites mentioned above can be sequentially placed within a single complementary (bispecific) strand and administered either in the presence or absence of additional therapeutic agents. When tested against breast and prostate tumor cell lines specific differences were noted: MCF-7 breast cancer cells were more receptive to the inhibitory effects of monospecific oligos, whereas PC-3 and LNCaP prostate cells were particularly responsive to bispecifics. In an effort to identify agents which enhance the activity of oligos and which possess less toxicity than traditionally employed chemotherapeutics, Rapamycin, an immunosuppressive agent known to regulate tumor growth and signal transduction mediated by the mTOR receptor, is compared to paclitaxel in combination therapy employing monospecific or bispecific oligos. Bispecifics were constructed recognizing the binding sites for TGF-alpha and EGFR mRNA [TGF-alpha/EGFR (MR(12)) and EGFR/TGF-alpha (MR(21))]; another pair recognized binding sites for EGFR and bcl-2 [EGFR/bcl-2 (MR(24)) and bcl-2/EGFR (MR(42))]; while a third pair employed only against the LNCaP prostate cell line recognized bcl-2 and the androgen receptor [bcl-2/AR (MR4(45)) and AR/bcl-2 (MR(54))]. Oligo pairs differ in their 5'-3' linear binding site orientations, and were tested in vitro against MCF-7 breast and PC-3 and LNCaP prostate tumor cell lines. Following cell attachment, incubations were done for 2 days with the agents followed by 2 days in their absence. Five experiments evaluated the effect of monospecific or bispecific antisense oligos in combination with an LD(50) dosage of either Rapamycin or paclitaxel and led to the conclusion that although these agents act via different mechanisms, they are comparable in effectiveness.

    Topics: Androgen Receptor Antagonists; Antibiotics, Antineoplastic; Antineoplastic Agents, Phytogenic; Apoptosis Regulatory Proteins; Breast Neoplasms; Cell Line, Tumor; Combined Modality Therapy; ErbB Receptors; Female; Gene Targeting; Humans; Male; Oligonucleotides, Antisense; Paclitaxel; Prostatic Neoplasms; Receptors, Androgen; Signal Transduction; Sirolimus; Transforming Growth Factor alpha

2009
EGFR ligand switch in late stage prostate cancer contributes to changes in cell signaling and bone remodeling.
    The Prostate, 2009, Apr-01, Volume: 69, Issue:5

    Bone metastasis occurs frequently in advanced prostate cancer (PCa) patients; however, it is not known why this happens. The epidermal growth factor receptor (EGFR) ligand EGF is available to early stage PCa; whereas, TGF-alpha is available when PCa metastasizes. Since the microenvironment of metastases has been shown to play a role in the survival of the tumor, we examined whether the ligands had effects on cell survival and proliferation in early and late PCa.. We used LNCaP cells as a model of early stage, non-metastatic PCa and the isogenic C4-2B cells as a model of late stage, metastatic PCa.. We found that the proliferation factor MAPK and the survival factor AKT were differentially activated in the presence of different ligands. TGF-alpha induced growth of C4-2B cells and not of the parental LNCaP cells; however, LNCaP cells expressing a constitutively active AKT did proliferate with TGF-alpha. Therefore, AKT appeared to be the TGF-alpha-responsive factor for survival of the late stage PCa cells. LNCaP cells exposed to EGF produced more osteoprotegerin (OPG), an inhibitor of bone remodeling, than C4-2B cells with TGF-alpha, which had increased expression of RANKL, an activator of bone remodeling. In concordance, TGF-alpha-treated C4-2B conditioned medium was able to differentiate an osteoclast precursor line to a greater extent than EGF-treated C4-2B or TGF-alpha-treated LNCaP conditioned media.. The switch in EGFR ligand availability as PCa progresses affects cell survival and contributes to bone remodeling.

    Topics: Adenocarcinoma; Bone Neoplasms; Bone Remodeling; Cell Line, Tumor; Disease Progression; Epidermal Growth Factor; ErbB Receptors; Humans; Ligands; Male; Mitogen-Activated Protein Kinase Kinases; Models, Biological; Neoplasm Staging; Prostatic Neoplasms; Proto-Oncogene Proteins c-akt; RANK Ligand; Signal Transduction; Transforming Growth Factor alpha

2009
Silibinin impairs constitutively active TGFalpha-EGFR autocrine loop in advanced human prostate carcinoma cells.
    Pharmaceutical research, 2008, Volume: 25, Issue:9

    Epidermal growth factor (EGF) and transformation growth factor-alpha (TGFalpha) are potent mitogens that regulate proliferation of prostate cancer cells via autocrine and paracrine loops, and promote tumor metastasis. They exert their action through binding to the cell surface receptor, epidermal growth factor receptor (EGFR), and cause activation of Erk1/2 mediated mitogenic signaling in human prostate cancer (PCA) at both advanced and androgen-independent stages. Thus, we rationalized that inhibiting this mitogenic pathway could be useful in controlling advanced PCA growth.. LNCaP and DU145 human PCA cells were treated with silibinin (100-200 microM) for different time points, and the levels of TGFalpha, activated signaling molecules (EGFR, Erk1/2 and Jnk1/2) and Erk1/2 kinase activity were analyzed employing ELISA, immunoprecipitation and/or immunoblotting techniques. The mRNA levels of TGFalpha were analyzed by RT-PCR.. Treatment of cells (LNCaP and DU145) with silibinin resulted in a decrease in TGFalpha protein at both secreted and cellular levels together with a decrease in its mRNA level. Silibinin also caused an inhibition of EGFR activation followed by that of Erk1/2 without any change in their protein levels. The kinase activity of Erk1/2 to Elk1 was also inhibited by silibinin at least in DU145 cells. In other study, silibinin caused strong inhibition of Jnk1/2 activation in LNCaP cells while in DU145 cells, a strong induction in Jnk1/2 activation was observed. These results suggest that silibinin impairs TGFalpha-EGFR-Erk1/2 signaling in both androgen-dependent (LNCaP) and -independent (DU145) advanced human prostate carcinoma cells.. This study, for the first time, identifies the inhibitory effect of silibinin on constitutively active TGFalpha-EGFR autocrine loop in advanced human PCA cells, which plausible contributes to the strong efficacy of silibinin in PCA prevention and intervention, as reported in recent studies.

    Topics: Androgens; Anticarcinogenic Agents; Antineoplastic Agents, Phytogenic; Autocrine Communication; Cell Line, Tumor; Dose-Response Relationship, Drug; Down-Regulation; ErbB Receptors; Humans; Male; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinase 8; Mitogen-Activated Protein Kinase 9; Neoplasms, Hormone-Dependent; Prostatic Neoplasms; RNA, Messenger; Silybin; Silymarin; Time Factors; Transforming Growth Factor alpha

2008
Arachidonic acid modulates the crosstalk between prostate carcinoma and bone stromal cells.
    Endocrine-related cancer, 2008, Volume: 15, Issue:1

    Diets high in n-6 fatty acids are associated with an increased risk of bone metastasis from prostate carces (PCa). The molecular mechanism underlying this phenomenon is largely unknown. Arachidonic acid (AA) and its precursor linoleic acid can be metabolized to produce pro-inflammatory cytokines that act as autocrine and paracrine regulators of cancer behavior. We and other authors have previously reported that factors released by PCa cells excite an aberrant response in bone marrow stromal cells (BMSCs). We planned to study how AA may modulate in vitro the interaction between PCa cells and human BMSCs. First, we observed that AA is a potent mitogenic factor for PCa cells through the production of both 5-lipoxygenase (5-LOX) and cyclooxygenase-2 (COX-2) metabolites. While 5-LOX controls cell survival through the regulation of the Bcl-2/Bax ratio, COX-2 activity stimulates the release of transforming growth factor-alpha (TGF-alpha) and pro-inflammatory cytokines. The blockade of COX-2 activity through a specific inhibitor is sufficient to repress AA-induced gene transcription. The over-expression of transforming growth factor -alpha (TGF-alpha), tumour necrosis factor-alpha (TNF-alpha) and interleukin-1 beta (IL-1beta) by AA-primed PCa cells resulted particularly effective in modifying cell behavior of cultured human BMSCs. In fact, we observed an increment in the cell number of BMSCs, due prevalently to the action of TGF-alpha, the number of osteoblasts, and the production of receptor activator for nuclear factor kappa B ligand (RANKL), events mainly controlled by inflammatory cytokines. These findings provide a possible molecular mechanism by which dietary n-6 fatty acids accumulating in bone marrow may influence the formation of PCa-derived metastatic lesions and indicate new molecular targets for the therapy of metastatic PCa.

    Topics: Arachidonate 5-Lipoxygenase; Arachidonic Acid; Blotting, Western; Bone Marrow; Breast Neoplasms; Cell Proliferation; Colony-Forming Units Assay; Cyclooxygenase 2; DNA Primers; Epidermal Growth Factor; Female; Humans; Interleukin-1beta; Male; Prostatic Neoplasms; RANK Ligand; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Stromal Cells; Transcription, Genetic; Transforming Growth Factor alpha; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

2008
Bispecific antisense oligonucleotides having binding sites directed against an autocrine regulated growth pathway and bcl-2 for the treatment of prostate tumors.
    Medical oncology (Northwood, London, England), 2007, Volume: 24, Issue:2

    Antisense oligonucleotides (oligos) against transforming growth factor-alpha (TGF-alpha) (MR1) and its binding site, the epidermal growth factor receptor (EGFR) (MR2), are efficacious against PC-3 and LNCaP prostate tumors. To enhance activity and aid in simultaneous delivery, "bispecific" 39-mer oligos were constructed containing portions of both MR1 and MR2 sequences. The first pair contained truncated sequences recognizing TGF-alpha and EGFR mRNA binding sites, about their respective AUG initiation codons. These bispecifics differ in their 5' to 3' tandem orientation (TGF-alpha/EGFR [MR12] and EGFR/TGF-alpha [MR21] sequences). A second pair was constructed having complementary sequences for EGFR and bcl-2 (EGFR/bcl-2 [MR24] and bcl-2/EGFR [MR42]). All bispecifics were tested in vitro against PC-3 and LNCaP prostate tumor cells, and compared to mono-specific oligos from which they were derived. The purpose of this study was: (1) to evaluate bispecific antitumor activity; (2) to identify dominant sequences; (3) to identify effects of binding site orientation; and (4) to determine whether bispecifics are more effective when targeting one versus different growth-dependent pathways. Comparisons were made between oligos tested against either PC-3 or LNCaP cells incubated for 2 d with the agents followed by 2 d in their absence. The first PC-3 cell experiment demonstrated that bispecific MR12 and MR21 oligos are at least as effective as their mono-specific counterparts and that the MR21 bispecific orientation is more effective than the MR1 mono-specific by 64% (p = 0.014). It also suggested that the sequence directed against EGFR contributed most to bispecific activity, particularly in the MR21 orientation. In a second PC-3 study a second bispecific pair of 37-mer oligos was constructed containing bases complementary to mRNA encoding EGFR and the apoptosis-associated protein bcl-2 (MR4). MR24 was constructed with the EGFR complementary site at the 5' end (EGFR/bcl-2), and MR42, containing the opposite orientation (bcl-2/EGFR). Each contained the dominant EGFR activity identified previously. MR1, MR2, MR4, MR12, MR21, MR24, and MR42 (1X and 2X in concentration) were cultured with cells and compared to controls. Each oligo significantly inhibited growth of PC-3 cells. MR42 was most effective and significantly better than MR1 (p = 0.0128), MR2 (p = 0.021), MR4 (p = 0.0002), and MR12 (p = 0.0032). 2X MR24 and 2X MR42 were better than their 1X concentration counte

    Topics: Antineoplastic Agents; Autocrine Communication; Base Sequence; Binding Sites; ErbB Receptors; Humans; Male; Molecular Sequence Data; Nucleic Acid Conformation; Oligonucleotides, Antisense; Prostatic Neoplasms; Proto-Oncogene Proteins c-bcl-2; Transforming Growth Factor alpha; Tumor Cells, Cultured

2007
Combination chemotherapy employing bispecific antisense oligonucleotides having binding sites directed against an autocrine regulated growth pathway and bcl-2 for the treatment of prostate tumors.
    Medical oncology (Northwood, London, England), 2007, Volume: 24, Issue:4

    In previous studies we demonstrated that antisense oligonucleotides (oligos) against transforming growth factor-alpha (TGF-alpha [MR1]), its binding site the epidermal growth factor receptor (EGFR [MR2]), and the anti-apoptosis protein bcl-2 (MR4) are efficacious against prostate tumors. In recent reports we also describe how two of these mRNA directed binding sites can be synthesized sequentially within a single linear complementary strand and administered either in the presence or absence of additional therapeutic agents. In these continuing experiments "bispecific" oligo pairs were further evaluated in the presence or absence of Cytoxan, Taxol, or DES. One oligo pair recognized the binding sites for TGF-alpha and EGFR mRNA (TGF-alpha/EGFR [MR12] and EGFR/TGF-alpha [MR21]); another pair recognized binding sites for EGFR and bcl-2 (EGFR/bcl-2 [MR24] and bcl-2/EGFR [MR42]). Oligo pairs differ in their linear 5' to 3' binding site orientations, and were tested in vitro against PC-3 and LNCaP prostate tumor cell lines. Following cell attachment, incubations were for 2 days with the agents followed by 2 days in their absence. When tested against PC-3 cells and combined with LD50 Cytoxan, MR2, MR4, MR24, MR42 significantly inhibited 47.3, 45.7, 68.3, and 64.9%; with LD50 Taxol MR2, MR4, MR24, MR42 significantly inhibited 49.8, 45.8, 64.1, and 59.2%; and with LD50 DES MR2, MR4, MR24, MR42 significantly inhibited 66.6, 67.6, 64.3, and 67.2% respectively. Each agent significantly increased the inhibition produced by either oligo alone.LNCaP cells were also incubated with mono- and bispecific oligos in either the presence or absence of chemotherapeutics. MR2, MR4, MR24, MR42 produced significant inhibitions of 57.4, 58.4, 69.4, and 68.6% with LD50 Cytoxan; 70.4, 70.1, 73.6, and 74.0% with LD50 Taxol; and 49.8, 50.1, 59.6, and 53.9%, respectively with LD50 DES.A complete PC-3 experiment compared MR1, MR2, MR4, MR12, MR21, MR24 and MR42, in the presence of LD50 Cytoxan. Each oligo combined with Cytoxan significantly inhibited: MR1 by 51.0, MR2 by 55.0, MR4 by 58.0; MR12 by 56.0; MR21 by 61.1, MR24 by 65.5 and MR42 by 66.0%. Bispecifics directed against two different pathways, MR24, and MR42 were the most effective.A complete LNCaP experiment compared the same series of oligos also in the presence of LD50 Cytoxan. Each oligo combined with Cytoxan significantly inhibited: MR1 by 49.0, MR2 by 50.0, MR4 by 53.0; MR12 by 52.0; MR21 by 58.6, MR24 by 53.9 and MR42 by 58.0

    Topics: Antineoplastic Agents, Alkylating; Antineoplastic Combined Chemotherapy Protocols; Binding Sites; Cell Line, Tumor; Cell Proliferation; Cyclophosphamide; ErbB Receptors; Humans; Male; Oligonucleotides, Antisense; Prostatic Neoplasms; Proto-Oncogene Proteins c-bcl-2; RNA, Messenger; Transforming Growth Factor alpha

2007
Bispecific antisense oligonucleotides with multiple binding sites for the treatment of prostate tumors and their applicability to combination therapy.
    Methods and findings in experimental and clinical pharmacology, 2006, Volume: 28, Issue:8

    Antisense oligonucleotides (oligos) against transforming growth factor-alpha (TGF-alpha; MR(1)) and its binding site, the epidermal growth factor receptor (EGFR; MR(2)), have proven efficacious against PC-3 and LNCaP prostate tumors when evaluated in both in vitro and in vivo models. To enhance their activity, and also to introduce a significantly different type of multifunctional agent into this field, "bispecific" oligos were constructed containing truncated sequences (derived from MR(1) and MR(2)) recognizing both TGF-alpha and EGFR mRNA internal binding sites, located about their respective AUG initiation codons. Two bispecifics were constructed, each having complementary sequences for TGF-alpha and EGFR mRNA, but differing in their 5' to 3' tandem orientation (TGF-alpha/EGFR [MR(12)] and EGFR/TGF-alpha [MR(21)] sequences). These bispecifics were tested in an in vitro system against PC-3 and LNCaP prostate tumor cells, with comparisons made to the original monospecific oligos from which they were derived. Efficacy was also compared when administered either alone or in combination with conventional chemotherapeutic agents. The purpose of this study was: 1) to validate the concept that these newly developed bispecific oligos have antitumor activity; 2) to enhance their efficacy through combination therapy; 3) to identify differences in effectiveness dependent upon binding site orientation; 4) identification of a dominant binding site that can be used to design other bispecifics that target additional tumor regulatory pathways. When fully evaluated against PC-3 cells in a series of experiments, newly developed bispecific oligos are at least as effective as their monospecific counterparts from which they were derived, and the bispecific with the MR(21) orientation is notably more effective than the MR(1) monospecific by 64% (p = 0.014 by Student t-test and p = 0.068 by the more stringent Mann-Whitney U test). Bispecifics were more effective when administered with chemotherapeutics (producing inhibition of 52.1% and 61.2% for MR(12) and MR(21), respectively, with Cytoxan (cyclophosphamide) inhibition of 59.0% and 65.1% for MR(12) and MR(21), respectively, with Taxol (paclitaxel) and 63.0% and 69.4% for MR(12) and MR(21), respectively, with DES [diethylstilbestrol]). Increasing the oligo concentration above 6.25 microM with cyclophosphamide had no additional effect. The sequence directed against EGFR was dominant and contributed most to bispecific activity,

    Topics: Antineoplastic Combined Chemotherapy Protocols; Binding Sites; Cell Line, Tumor; Cell Proliferation; Cyclophosphamide; Diethylstilbestrol; ErbB Receptors; Humans; Male; Oligonucleotides, Antisense; Paclitaxel; Prostatic Neoplasms; Transforming Growth Factor alpha

2006
Construction of a bispecific antisense oligonucleotide containing multiple binding sites for the treatment of hormone insensitive prostate tumors.
    Medical hypotheses, 2005, Volume: 65, Issue:5

    Antisense oligonucleotides (oligos) have demonstrated efficacy for the treatment of various cancers, infectious diseases and metabolic disorders. While most studies have utilized single oligos either administered alone, or more recently in combination therapy with other drugs, some investigators have administered more than one oligo in a combined administration or have designed oligos which target multiple proteins (those which share mRNA sequence homology). Antisense oligos inhibit mRNA translation through complementary base pair binding, often about the AUG initiation codon. This inhibition is further enhanced through destruction of the mRNA:oligo hybrid by RNAse H. Construction of an oligo with multiple binding sites located about the respective mRNA initiation codons could simultaneously block translation of more than one protein, even those unrelated in sequence. Such binding could produce a complex mRNA:oligo hybrid more prone to degradation and clearance. Furthermore, such a formulation would increase oligo specific activity and cellular uptake, reduce toxicity, and stabilize at 1:1 the ratio between multiple oligo active sites which otherwise must be comparably delivered (in amount) and individually targeted. The activity of these newly constructed oligos could be tested in both in vitro and in vivo prostate tumor models, utilizing the hormone sensitive LNCaP and the hormone insensitive PC-3 lines. In vitro testing would evaluate oligos administered either alone or in combination with other chemotherapeutics. In vivo testing would administer the oligos to tumors carried in athymic nude mice by either intratumoral inoculation or by using a diffusion pump. Antisense oligos which target proteins associated with growth factors or their receptors, could have a role in the treatment of human prostate cancers when administered with hormone deprivation therapy, or against tumors which have already become hormone insensitive. In the later case, such treatment could form the basis of a second tier of therapy based upon growth factor deprivation.

    Topics: Androgens; Binding Sites; Drug Resistance; ErbB Receptors; Gene Silencing; Gene Targeting; Genetic Therapy; Humans; Male; Neoplasm Proteins; Oligonucleotides, Antisense; Prostatic Neoplasms; Transcription Factors; Transforming Growth Factor alpha

2005
Characterization of a transplantable hormone-responsive human prostatic cancer xenograft TEN12 and its androgen-resistant sublines.
    The Prostate, 2004, Jan-01, Volume: 58, Issue:1

    Models for human prostate cancer can facilitate the study of resistance to endocrine therapy, aid drug discovery, and pre-clinical assessment.. Characteristics thought relevant to the growth in athymic nude mice of TEN12, an androgen-dependent transplantable prostatic cell line derived from a primary prostate carcinoma, and its two androgen-independent sublines, TEN12F and TEN12C, have been assessed immunocytochemically.. The xenografts of the parental TEN12 line are moderately differentiated with both papillary and glandular regions, pleomorphic nuclei and abundant mitotic figures and are extremely vascular. The cells express androgen receptor (AR), PSA, VEGF, EGFR, c-erbB2, and TGFalpha. Both TEN12F and TEN12C xenografts possessed a more anaplastic morphology and displayed significantly lower growth rates, reduced blood vessel density (BVD), decreased MIB-1 antigen and E-cadherin expression and increased cytoplasmic AR and HSP90 staining. Elevated EGFR (membrane) but not c-erbB2 expression was demonstrated in the TEN12F line only. Castration of mice bearing TEN12 xenografts rapidly induced the appearance of cytoplasmic AR in the cells, PSA levels decreased initially but recovered to below pre-castration levels whilst reduced TGFalpha and loss of VEGF expression was seen in the long-term castrates.. TEN12 and its sublines offer additional in vivo models to study the factors involved in the progression of prostatic cancer to androgen-independence.

    Topics: Animals; Cadherins; Cell Division; Cell Line, Tumor; ErbB Receptors; Female; HSP90 Heat-Shock Proteins; Humans; Immunohistochemistry; Ki-67 Antigen; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Transplantation; Neoplasms, Hormone-Dependent; Neovascularization, Pathologic; Prostate-Specific Antigen; Prostatic Neoplasms; Receptor, ErbB-2; Receptors, Androgen; Transforming Growth Factor alpha; Transplantation, Heterologous

2004
Ionizing radiation causes a dose-dependent release of transforming growth factor alpha in vitro from irradiated xenografts and during palliative treatment of hormone-refractory prostate carcinoma.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2004, Sep-01, Volume: 10, Issue:17

    Characterize the radiation response for transforming growth factor (TGF) alpha shedding in vitro and in vivo. We also report the shedding of TGF alpha by patients undergoing irradiation for hormone-refractory prostate cancer.. TGF alpha levels were determined by ELISA. DU145 xenografts were established on the flanks of athymic nu/nu mice. Expression of phospho-extracellular signal-regulated kinase (ERK)1/2 and phospho-epidermal growth factor receptor (EGFR) and the DNA repair proteins XRCC1 and ERCC1 were determined by Western analyses.. Exposure to ionizing radiation results in a dose-dependent release of TGF alpha. Once released, TGF alpha stimulates EGFR-ERK1/2 signaling in unirradiated cells. Blockade of the EGFR with the tyrphostin AG1478 eliminates the up-regulation XRCC1 and ERCC1 by TGF alpha or irradiation. After irradiation, cells are refractory to additional transactivation of EGFR by additional irradiation for 8 to 12 hours. Irradiation during this refractory period does not increase the expression of XRCC1 or ERCC1. Ligand activation of EGFR is maintained during the refractory period. Irradiation of DU145 xenografts also results in the activation of ERK1/2, release of TGF alpha, and a similar refractory period. Ionizing irradiation also results in the release of TGF alpha for patients undergoing radiation therapy for hormone-refractory prostate cancer.. Irradiation results in a dose-dependent increase in TGF alpha capable of enhancing the growth of DU145 xenografts. TGF alpha is also shed following radiation therapy of patients treated for hormone-refractory prostate cancer. Radiation transactivation of the EGFR produces a radio-refractory period, which lasts for several hours. During this period, additional irradiation fails to induce XRCC1, ERCC1, or additional TGF alpha release.

    Topics: Adenocarcinoma; Animals; Blotting, Western; Case-Control Studies; DNA Repair; DNA-Binding Proteins; Dose-Response Relationship, Radiation; Endonucleases; Enzyme-Linked Immunosorbent Assay; ErbB Receptors; Female; Gene Expression Regulation, Neoplastic; Humans; In Vitro Techniques; Male; Mice; Mice, Nude; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Neoplasms, Hormone-Dependent; Prostatic Neoplasms; Radiation, Ionizing; Transforming Growth Factor alpha; Transplantation, Heterologous; Tumor Cells, Cultured; Up-Regulation; Whole-Body Irradiation; X-ray Repair Cross Complementing Protein 1

2004
[Regulation of EGF-receptor expression by TGFalpha in human prostate androgen-unresponsive cancer cells].
    Zhonghua nan ke xue = National journal of andrology, 2004, Volume: 10, Issue:8

    To elucidate the regulation of epidermal growth factor receptor (EGFR) expression by transforming growth factor (TGFalpha) and epidermal growth factor (EGF) in human prostate androgen-unresponsive cancer cells.. EGFR mRNA expression and its protein level were measured by means of RT-PCR and Western blot respectively in human prostate cancer androgen-unresponsive cell lines, ARCaP and PC3, all treated with exogenous TGFalpha.. In the TGFalpha group, the levels of EGFR mRNA were 5.01 0.45 and 9.05 0.63 in PC3 and ARCaP respectively, significantly higher than in the control group (P < 0.05). The level of EGFR protein in PC3 treated with TGFalpha was 2.28 0.53, higher than in the control group (P < 0.05); however, the level of EGFR protein in ARCaP treated with TGFalpha was only 1.24 0.22, not different from the control (P > 0.05).. TGFalpha/EGF-EGFR pathway serves as a key growth regulator in prostate cancer. TGFalpha, but not EGF, preferentially maintains an autocrine loop in human androgen-unresponsive prostate cancer.

    Topics: Blotting, Western; Cell Line, Tumor; ErbB Receptors; Humans; Male; Prostatic Neoplasms; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Transforming Growth Factor alpha

2004
Adipocyte-fatty acid binding protein induces apoptosis in DU145 prostate cancer cells.
    Journal of experimental therapeutics & oncology, 2004, Volume: 4, Issue:2

    Adipocyte-fatty acid binding protein (A-FABP) is a 14-15 kDa cytoplasmic protein that binds unesterified fatty acids (FA). It is believed that A-FABP is present in normal cells and disappears in cancer cells. Prostate cancer DU145 cells lack expression of A-FABP. Here, we report that transfection of A-FABP blocked growth of DU145 cells suggesting its role as a tumor suppressor. A-FABP transfected- prostate cancer DU145 cells underwent apoptosis when induced to overexpress A-FABP using an ecdysone-controlled expression system. DU145 cell cultures in complete medium exhibited a maximum of approximately 28% of apoptotic cells after 96 h of exposure to an ecdysone analog, Ponasterone A. We found that the possible mechanisms leading to the observed apoptotic effect may be due, in part, to an overexpression of tumor necrosis factor-alpha (TNF-alpha) and a moderate downregulation of transforming growth factor-alpha (TGF-alpha) in DU145 cells overexpressing A-FABP. The epidermal growth factor receptor (EGFR)/phosphatidyl inositol 3-kinase (PI 3-kinase) signaling pathway was not altered in these cells, suggesting that A-FABP may cause apoptosis by inducing downregulation of essential autocrine growth factors and/or upregulation of pro-apoptotic ones.

    Topics: Apoptosis; Carrier Proteins; Cell Line, Tumor; Ecdysterone; Fatty Acid-Binding Proteins; Gene Expression Regulation; Humans; Male; Prostatic Neoplasms; Signal Transduction; Steroids; Transfection; Transforming Growth Factor alpha; Tumor Necrosis Factor-alpha

2004
Synergistic effects of combination therapy employing antisense oligonucleotides with traditional chemotherapeutics in the PC-3 prostate cancer model.
    Medical oncology (Northwood, London, England), 2004, Volume: 21, Issue:4

    Combination therapy including antisense oligonucleotides (ODNs) with traditional chemotherapeutic agents offers potential benefits by increasing the effectiveness of the chemotherapeutics, reducing their effective dosage, and simultaneously reducing toxicity. Previously we have reported that antisense ODNs specific for transforming growth factor-alpha (TGF-alpha) and its binding site, the epidermal growth factor receptor (EGFR) (MR1 and MR2, respectively), are effective against the PC-3 in vitro and in vivo prostate cancer models. In this series we evaluated these antisense ODNs in various combinations and treatment cycles with paclitaxel (Taxol), cyclophosphamide (Cytoxan), mitoxantrone, carboplatin, cisplatin, and oxaliplatin in order to identify synergistic effects.We found that when either of the ODNs were simultaneously administered with Taxol, no synergistic activity was noted. However, when sequentially administered in a series 1 d apart, a pretreatment with the ODN directed against TGF-alpha (6.64 microm) followed by Taxol (5 nm) had significantly (p <0.001) greater activity than these agents similarly administered in the reverse order or simultaneously. When Cytoxan was administered in sequence with both ODNs significantly increased growth inhibition was obtained compared to when Cytoxan was administered alone. A 1 d treatment of PC-3 cells with Cytoxan followed the next day with MR1 was significantly more effective (p <0.0001). The reverse order, a pretreatment with MR1 followed by Cytoxan, also resulted in significant additional inhibition (p=0.0004). Similarly sequenced, MR2 followed by Cytoxan, was also significantly more effective (p=0.0014) than Cytoxan treatment alone. For mitoxantrone, which was administered in combination therapy with ODNs: mitoxantrone with MR1 was significantly more inhibitory than the combination of both MR1 and MR2 ODNs (p=0.006) and also mitoxantrone administered alone (p=0.0012). Mitoxantrone administered with MR1 was not significantly different from mitoxantrone given in combination with MR2. Although mitoxantrone and MR2 was statistically (p=00015) more inhibitory than mitoxantrone alone, this combination was barely more effective (p=0.04) than the MR1 ODN administered alone.

    Topics: Antineoplastic Agents; Combined Modality Therapy; Drug Interactions; ErbB Receptors; Humans; Male; Oligonucleotides, Antisense; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured

2004
Epidermal growth factor receptor-related peptide inhibits growth of PC-3 prostate cancer cells.
    Molecular cancer therapeutics, 2004, Volume: 3, Issue:12

    Interference with the activation of growth factor receptors, specifically epidermal growth factor receptor (EGFR), represents a promising strategy for the development of novel and selective anticancer therapies. We reported that EGFR-related peptide (ERRP), a recently isolated negative regulator of EGFR, could be a potential therapeutic agent for colorectal cancer. To determine whether ERRP could potentially be a therapeutic agent for prostate carcinoma, we examined the effect of recombinant ERRP on the growth of the prostate cancer cell line PC-3 in vitro. Events of the EGFR signal transduction pathways were also examined. ERRP caused a marked inhibition of cell growth in a dose- and time-dependent manner and also induced apoptosis. The latter was evidenced by increased number of apoptotic cells, activation of caspase-3, and cleavage of poly(ADP-ribose)polymerase. The transforming growth factor-alpha-induced stimulation of cell growth and activation of EGFR was also inhibited by ERRP. These changes were accompanied by a concomitant attenuation of activation of Akt and mitogen-activated protein kinases as well as basal and transforming growth factor-alpha-induced activation of nuclear factor-kappaB. Inhibition of EGFR activation by ERRP could be partly attributed to increased sequestration of EGFR ligands. In summary, our data show that ERRP inhibits the growth of prostate cancer cells by attenuating EGFR signaling processes. ERRP could potentially be an effective therapeutic agent for prostate cancer.

    Topics: Apoptosis; ErbB Receptors; Glycoproteins; Humans; Male; Mitogen-Activated Protein Kinases; NF-kappa B; Prostatic Neoplasms; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Signal Transduction; Transforming Growth Factor alpha; Tumor Cells, Cultured

2004
Inhibition of PC-3 prostate cancer cell growth in vitro using both antisense oligonucleotides and taxol.
    Medical oncology (Northwood, London, England), 2003, Volume: 20, Issue:1

    Antisense oligonucleotides (oligos) directed against mRNA-encoding transforming growth factor-alpha (TGF-alpha) and the epidermal growth factor receptor (EGFR) have demonstrated in vitro and in vivo efficacy against prostate cancer tumor models. However, many therapeutic agents have increased effectiveness when given in combination with other more established agents. We evaluated the effectiveness of two oligos (3.32 and 6.64 microM/L) known to have significant activity against the PC-3 prostate cell line in combination therapy with the chemotherapeutic agent paclitaxel (Taxol) (2.5 and 5.0 nm). Therapy was evaluated when oligos and Taxol were administered either as (1) single agents, (2) simultaneously in a combined therapy, or (3) sequentially, a form of combination therapy with both agents being administered in a series. We found that when either of the two oligos were given simultaneously with Taxol, no synergistic activity was noted. However, when sequentially administered in a series 1 d apart, a pretreatment with the antisense directed against TGF-alpha (6.64 microM/L) followed by Taxol (5 nm) had significantly greater activity than these agents similarly administered in the reverse order or simultaneously.>

    Topics: Antineoplastic Agents, Phytogenic; Antineoplastic Combined Chemotherapy Protocols; Combined Modality Therapy; Dose-Response Relationship, Drug; Drug Administration Schedule; Drug Synergism; ErbB Receptors; Gene Expression Regulation, Neoplastic; Humans; Male; Oligonucleotides, Antisense; Paclitaxel; Phosphatidylethanolamines; Prostatic Neoplasms; RNA, Neoplasm; Transforming Growth Factor alpha; Tumor Cells, Cultured

2003
Inhibition of ErbB-2 and ErbB-3 expression by quercetin prevents transforming growth factor alpha (TGF-alpha)- and epidermal growth factor (EGF)-induced human PC-3 prostate cancer cell proliferation.
    International journal of oncology, 2003, Volume: 23, Issue:3

    Because ErbB-2 receptor is involved in hormone-independency for growth and metastasis of prostate cancer cells, the aim was to investigate the effects of quercetin on ErbB-2 and ErbB-3 expression and its critical components such as MAP kinase and PI-3 kinase. Hemocytometric counts and [3H]-thymidine incorporation were used to determine the effects of quercetin, EGF and TGF-alpha on cell proliferation and DNA synthesis in PC-3 and LnCap cells. Changes in ErbB-2, ErbB-3 and components of MAPK and PI-3K pathways were analyzed by Western blot analysis. Treatment of PC-3 and LnCap cells with quercetin resulted in a dose-dependent growth inhibition. The rate of DNA synthesis was decreased by 40, 55 and 65% on treatment with 14.5, 29.0 and 58.0 microM of quercetin, respectively. Concomitantly, these treatments led to a dose-dependent decrease in ErbB-2, ErbB-3 and their basal autophosphorylation levels as compared to controls. Cyclin D1 expression and basal phosphorylation of c-Raf, MAPK, Elk-1 and Akt-1 in PC-3 cells was also inhibited by quercetin treatment. Co-treating PC-3 cells with quercetin significantly attenuated EGF- and TGF-alpha-induced growth and phosphorylation of ErbB-2, ErbB-3, c-Raf, MAPK kinase 1/2 (MEK1/2), MAPK, Elk-1 and Akt-1. Since ErbB receptor is important for growth, metastasis and drug resistance, inhibition of ErbB-2 and ErbB-3 by pharmacological doses of quercetin may provide a new approach for treatment of prostate cancers.

    Topics: Blotting, Western; Cell Division; Cell Line, Tumor; Cyclin D1; DNA; Dose-Response Relationship, Drug; Epidermal Growth Factor; Humans; Male; MAP Kinase Signaling System; Phosphatidylinositol 3-Kinases; Phosphorylation; Prostatic Neoplasms; Proto-Oncogene Proteins c-raf; Quercetin; Receptor, ErbB-2; Receptor, ErbB-3; Transforming Growth Factor alpha

2003
Inhibition of ligand-mediated HER2 activation in androgen-independent prostate cancer.
    Cancer research, 2002, Oct-01, Volume: 62, Issue:19

    Hormone-independent tumor growth and metastasis are associated with increased mortality in human prostate cancer. In this study, we evaluate a potential role for ligand-mediated activation of HER2 receptor tyrosine kinase in androgen-independent prostate cancers. HER2, HER3, and epidermal growth factor receptor were detected in the androgen-independent cell line 22Rv1. Heregulin stimulation results in receptor phosphorylation and cell proliferation that is inhibited by increasing concentrations of anti-HER2 recombinant humanized monoclonal antibody (rhuMAb) 2C4. Furthermore, inhibition of tumor growth was observed in xenografts derived from 22Rv1 cells when treated with rhuMAb 2C4 in a dose-dependent manner. These studies provide a framework, both in vitro and in vivo, to examine the molecular mechanisms of ligand-driven HER2 activation in androgen-independent tumorigenesis.

    Topics: Androgens; Animals; Antibodies, Monoclonal; Cell Division; Dose-Response Relationship, Immunologic; Female; Humans; Ligands; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasms, Hormone-Dependent; Neuregulin-1; Prostatic Neoplasms; Receptor, ErbB-2; Receptor, ErbB-3; Recombinant Proteins; Transforming Growth Factor alpha; Tumor Cells, Cultured; Xenograft Model Antitumor Assays

2002
Backbone modification alters the efficacy of antisense oligonucleotides directed against mRNA encoding either TGF-alpha or EGFR in the treatment of prostate cancer cell lines.
    Methods and findings in experimental and clinical pharmacology, 2002, Volume: 24, Issue:10

    Antisense oligonucleotides (oligos) directed against mRNA-encoding, transforming growth factor-alpha (TGF-alpha) and the epidermal growth factor receptor (EGFR), have been shown to significantly inhibit in vitro and in vivo growth of prostate tumor models. Recently, second generation oligos have been employed with identical base sequences, but containing backbome modifications that enhance stability, solubility and circulatory patterns. Using relatively low concentrations of oligos, we compared the efficacy of the first generation phosphorothioated oligos against TGF-alpha (MR1) and EGFR (MR2) with second generation oligos containing completely phosphorothioated backbones and different patterns of 2'-methoxyethyl (2'-MOE) backbone modifications, while retaining the original designated base sequence using, the LNCaP and PC-3 prostate cancer cell lines, respectively. All experiments were conducted in vitro with lipofectin to enhance oligo entry. Under these conditions, using oligo concentrations between 0.83 and 3.32 microM for LNCaP cells treated with oligos directed against TGF-alpha only the first generation MR1 had inhibitory activity. When treated with oligos directed against EGFR, none of the oligos had inhibitory activity and they behaved similarly. Using the PC-3 cell line and treatment directed against TGF-alpha with oligo concentrations between 0.42 and 3.32 microM, first generation MR1 and second generation 5005 behaved similarly with no notable effect, while second generation 5007 produced dramatic growth stimulation. When PC-3 cells were treated with oligos directed against EGFR, second generation 5006 and 5008 had similar and apparently dose-dependent inhibition. We conclude that backbone modifications influence oligo efficacy and may result in either enhanced or diminished activity. Because of their activity against the hormone insensitive PC-3 cells, the 5006 and 5008 compounds warrant additional study at greater concentrations and also merit in vivo testing.

    Topics: ErbB Receptors; Gene Expression Regulation, Neoplastic; Humans; Male; Nucleic Acid Conformation; Oligonucleotides, Antisense; Prostatic Neoplasms; RNA, Messenger; RNA, Neoplasm; Thionucleotides; Transforming Growth Factor alpha; Treatment Outcome; Tumor Cells, Cultured

2002
Multiple responses to EGF receptor activation and their abrogation by a specific EGF receptor tyrosine kinase inhibitor.
    The Prostate, 2002, Jun-01, Volume: 52, Issue:1

    Epidermal growth factor receptor (EGF-R) autophosphorylation is essential for its intracellular mitogenic signaling via the MAPK pathway and for interaction in other cellular processes. Inhibition of this activity in tumor cells that predominantly utilise EGF-R therefore offers an alternative approach to therapy.. The ability of a specific inhibitor of EGF-R tyrosine kinase, ZM 252868, (TKI) to alter various parameters related to growth in DU145 and PC3 cell lines was investigated, by immunocytochemistry, Northern blotting, Western blotting and invasion assays.. In DU145 cultures, the total cell population and number of cells in cell cycle decreased in the presence of TKI whilst the apoptotic rate was significantly increased. Reduction in autophosphorylation of the EGF-R, membrane expression of EGF-R, activation of the MAPK, p38, and JNK enzymes and the invasive capacity of DU145 cells was observed in the TKI treated cells. Under the same conditions, PC3 cell growth and EGF-R expression and MAPK activation were not affected. The use of inhibitors of intracellular signaling indicated that the DU145 cells, in contrast to PC3 cells, predominantly utilize EGF-R activation of the MAPK signaling pathway for growth.. In prostatic cancer patients, in whom androgen resistance has developed and whose tumors have upregulated EGF-R for growth, specific TKI's may offer an important therapy option.

    Topics: Adenocarcinoma; Apoptosis; Blotting, Northern; Blotting, Western; Cell Count; Cell Cycle; Cell Division; Enzyme Activation; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Humans; Immunohistochemistry; Male; Mitogen-Activated Protein Kinases; Neoplasm Invasiveness; Phosphorylation; Prostatic Neoplasms; Quinazolines; RNA, Messenger; Signal Transduction; Transforming Growth Factor alpha; Tumor Cells, Cultured

2002
Detrimental effect of cancer preventive phytochemicals silymarin, genistein and epigallocatechin 3-gallate on epigenetic events in human prostate carcinoma DU145 cells.
    The Prostate, 2001, Feb-01, Volume: 46, Issue:2

    Targeting epigenetic events associated with autonomous growth of advanced prostate cancer (PCA) is a practical approach for its control, prevention, and treatment. Recently we showed that treatment of prostate carcinoma DU145 cells with cancer preventive flavonoid silymarin at 100-200 microM doses inhibits erbB1-Shc mitogenic signaling and modulates cell cycle regulators leading to a G1 arrest and inhibition of cell growth and anchorage-independent colony formation. Here, we asked the question whether these important findings could be extended to other cancer preventive flavonoids and isoflavones such as epigallocatechin 3-gallate (EGCG) and genistein.. DU145 cells were treated with similar doses (100-200 microM) of silymarin, genistein or EGCG, cell lysates prepared, and levels of activated signaling molecules (erbB1-Shc-ERK1/2) and cell cycle regulators (CDKIs, CDKs, and cyclins) analyzed employing immunoprecipitation and/or immunoblotting techniques. Cell growth studies were done by cell counting during 5 days of treatment with these agents, and cell death was determined by Trypan blue staining.. Treatment of cells with silymarin, genistein or EGCG at 100-200 microM resulted in a complete inhibition of TGFalpha-caused activation of erbB1 followed by a moderate to strong inhibition (10-90%) of Shc activation without an alteration in their protein levels. Silymarin and genistein, but not EGCG, also inhibited (10% to complete) ERK1/2 activation suggesting that these agents impair erbB1-Shc-ERK1/2 signaling in DU145 cells. In other studies, silymarin, genistein or EGCG caused a strong induction of Cip1/p21 (up to 2.4-fold) and Kip1/p27 (up to 150-fold), and a strong decrease in CDK4 (40-90%) but had moderate effect on CDK2, and cyclins D1 and E. An enhanced level of CDKIs also led to an increase in their binding to CDK4 and CDK2. Treatment of cells with silymarin, genistein or EGCG also resulted in 50-80% cell growth inhibition at lower doses, and complete inhibition at higher doses. In contrast to silymarin, higher doses of genistein showed cytotoxic effect causing 30-40% cell death. A more profound cytotoxic effect was observed with EGCG accounting for 50% cell death at lower doses and complete loss of viability at higher doses.. These results suggest that similar to silymarin, genistein and EGCG also inhibit mitogenic signaling pathway(s) and alter cell cycle regulators, albeit at different levels, leading to growth inhibition and death of advanced and androgen-independent prostate carcinoma cells. More studies are, therefore, needed with these agents to explore their anti-carcinogenic potential against human prostate cancer.

    Topics: Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Anticarcinogenic Agents; Catechin; Cell Cycle; Cell Death; Cell Division; ErbB Receptors; Genistein; Humans; Male; MAP Kinase Signaling System; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Plant Extracts; Prostatic Neoplasms; Protein Binding; Proteins; Shc Signaling Adaptor Proteins; Silymarin; Src Homology 2 Domain-Containing, Transforming Protein 1; Transforming Growth Factor alpha

2001
Ionizing radiation-induced mitogen-activated protein (MAP) kinase activation in DU145 prostate carcinoma cells: MAP kinase inhibition enhances radiation-induced cell killing and G2/M-phase arrest.
    Radiation research, 2000, Volume: 153, Issue:4

    These studies examine the role(s) played by the mitogen-activated protein kinase (MAPK) pathway after exposure of DU145 prostate carcinoma cells to radiation. Radiation (2 Gy) was found to cause both immediate primary (0-30 min) and prolonged secondary activations (90-1440 min) of the MAPK pathway. These activations of the MAPK pathway were abolished by inhibition of epidermal growth factor receptor (EGFR) function. The secondary activation was also abolished by addition of a neutralizing monoclonal antibody against transforming growth factor alpha (TGFA). Activation of the MAPK pathway could be induced in nonirradiated cells by the transfer of medium from irradiated cultures. Neutralizing antibody to TGFA blocked this effect, indicating that radiation causes secondary activation of the MAPK pathway by release of TGFA in DU145 cells. Radiation induced a transient G(2)/M-phase growth arrest that was prolonged for up to 24 h by inhibition of the MAPK pathway. Inhibition of the MAPK pathway significantly increased the ability of radiation to cause apoptosis 24 h after exposure. The ability of DU145 cells to proliferate after irradiation became dependent on MAPK signaling. When cells were subjected to single doses or fractionated radiation exposure, continuous inhibition of the MAPK pathway significantly decreased clonogenic survival. Only a small fraction of this cell killing could be accounted for by apoptosis within the first 96 h. Thus inhibition of the MAPK pathway increased radiation-induced cell killing likely by both apoptotic and nonapoptotic mechanisms. Collectively, our findings indicate that disruption of the TGFA/EGFR/MAPK pathway may represent a strategy that could be exploited to manipulate prostate carcinoma growth and cell survival after irradiation.

    Topics: Apoptosis; CDC2 Protein Kinase; Dose Fractionation, Radiation; Enzyme Activation; Enzyme Inhibitors; ErbB Receptors; G2 Phase; Humans; Male; Mitogen-Activated Protein Kinases; Mitosis; Prostatic Neoplasms; Radiation, Ionizing; Signal Transduction; Transforming Growth Factor alpha; Tumor Cells, Cultured

2000
Impairment of erbB1 receptor and fluid-phase endocytosis and associated mitogenic signaling by inositol hexaphosphate in human prostate carcinoma DU145 cells.
    Carcinogenesis, 2000, Volume: 21, Issue:12

    Recently, we observed that epidermal growth factor receptor (EGFR or erbB1) endocytosis and associated mitogenic signaling occur in human prostate cancer (PCA) cells, suggesting that erbB1 endocytosis might be involved in advanced and androgen-independent PCA growth. Based on these findings, and the fact that aberrant expression of erbB family members is common in human prostatic intraepithelial neoplasia and invasive PCA, we reasoned that impairment of erbB1 endocytosis and associated mitogenic signaling might inhibit PCA growth. Inositol hexaphosphate (IP6) interacts with plasma membrane clathrin-associated protein complex 2 (AP2) and inhibits phosphatidylinositol 3-kinase (PI3K). As these are essential components of receptor-mediated and fluid-phase endocytosis, respectively, we reasoned that IP6 might impair erbB1 endocytosis and associated signaling in human PCA cells, leading to their growth inhibition. IP6 strongly to completely inhibited (26-100%; P < 0.05) transforming growth factor alpha-induced binding of activated erbB1 to AP2 in human PCA DU145 cells, demonstrating the impairment of the initial step in ligand-induced erbB1 endocytosis. IP6 treatment of cells resulted in a dose-dependent increase (1.8- to 7. 7-fold compared with cells treated with ligand alone; P < 0.05) in levels of activated erbB1. These two findings suggest that the inhibitory effect of IP6 on receptor endocytosis is independent of its lack of effect on ligand-induced erbB1 activation. These effects of IP6, however, were associated with strong inhibition of ligand-induced Shc phosphorylation (77-84% decrease; P < 0.05) and its binding to erbB1 (58-100% decrease; P < 0.05). IP6 also significantly and dose-dependently inhibited fluid-phase endocytosis (19-52%; P < 0.05). It inhibited PI3K-AKT signaling pathway as an upstream response in its effect on the inhibition of fluid-phase endocytosis. The inhibition of erbB1 receptor and fluid-phase endocytosis, and associated signaling by IP6, was corroborated by very strong to complete inhibition (70-100%; P < 0.05) of extracellular signal-regulated protein kinase 1/2 activation by IP6. IP6 significantly (P < 0.05) inhibited anchorage-dependent and -independent inhibition (50-100% and 30-75%, respectively) in DU145 cells. Targeting the impairment of erbB1 endocytosis and associated mitogenic signaling by IP6 in advanced and androgen-independent human PCA DU145 cells could be a useful approach for treating PCA.

    Topics: Adaptor Protein Complex 2; Adaptor Protein Complex alpha Subunits; Adaptor Proteins, Vesicular Transport; Androstadienes; Biological Transport; Cell Division; Endocytosis; ErbB Receptors; Horseradish Peroxidase; Humans; Kinetics; Male; Membrane Proteins; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Models, Biological; Phytic Acid; Prostatic Neoplasms; Signal Transduction; Transforming Growth Factor alpha; Tumor Cells, Cultured; Wortmannin

2000
Nuclear matrix targeting of the protein kinase CK2 signal as a common downstream response to androgen or growth factor stimulation of prostate cancer cells.
    Cancer research, 1999, Mar-01, Volume: 59, Issue:5

    Protein kinase CK2, a messenger-independent serine/threonine kinase, has been implicated in cell growth. Androgenic stimulus in rat prostate modulates its association with nuclear matrix (NM) and chromatin. Because the growth of human prostate carcinoma cells is influenced by androgens and/or growth factors, we determined the nature of CK2 signaling in the NM in response to androgen and growth factor stimuli. Androgen-sensitive LNCaP and androgen-insensitive PC-3 cells were cultured in media to regulate their growth in the presence of 5alpha-dihydrotestosterone (5alpha-DHT) or growth factors (epidermal growth factor, keratinocyte growth factor, and transforming growth factor alpha). The activity of CK2 was measured in the cytosolic and NM fractions isolated from these cells after treatment with growth stimuli. The changes in CK2 in various fractions were also confirmed by immunoblotting with a specific antibody. LNCaP cells responded to both 5alpha-DHT and growth factors for growth. The presence of these agents in the culture medium evoked a translocation of CK2 to the NM from the cytosol. The PC-3 cells did not respond to 5alpha-DHT for growth but did respond to growth factors. Under these conditions, there was also a translocation of CK2 to the NM concomitant with a decrease in the cytosolic fraction. These results suggest that CK2 translocation to the NM occurs in response to various growth stimuli in cells in culture. Thus, CK2 is a common downstream signal transducer in response to diverse growth stimuli that may relate to the pathobiology of prostate cancer cells.

    Topics: Adenocarcinoma; Animals; Casein Kinase II; Cell Division; Chromatin; Cytosol; Dihydrotestosterone; DNA-Binding Proteins; Epidermal Growth Factor; Fibroblast Growth Factor 10; Fibroblast Growth Factor 7; Fibroblast Growth Factors; Growth Substances; Humans; Kinetics; Male; Nuclear Matrix; Prostatic Neoplasms; Protein Serine-Threonine Kinases; Rats; Signal Transduction; Transforming Growth Factor alpha; Tumor Cells, Cultured

1999
Complex post-transcriptional regulation of EGF-receptor expression by EGF and TGF-alpha in human prostate cancer cells.
    British journal of cancer, 1999, Volume: 80, Issue:5-6

    The epidermal growth factor receptor (EGFR) plays an important role in the development and progression of prostate cancer and its overexpression is associated with decreased survival. With progression, prostate cancer cells switch from epidermal growth factor (EGF) to transforming growth factor alpha (TGF-alpha) synthesis, which contributes to autocrine growth and unrestrained proliferation. To define the molecular mechanisms involved in the regulation of EGFR expression by EGF and TGF-alpha we studied three human prostate cancer cell lines, androgen-responsive (LNCaP) and -unresponsive (DU145 and PC3). Here we show that TGF-alpha stabilized EGFR mRNA two- to threefold in all three cell lines, whilst EGF stabilized EGFR mRNA approximately twofold in LNCaP and DU145 cells, but not in PC3 cells. Both ligands increased EGFR transcription in LNCaP and DU145 cells, with less effect in PC3 cells. In all three cell lines EGF reduced total EGFR protein levels more than TGF-alpha, but this was associated with a greater increase in de novo protein synthesis with EGF compared to TGF-alpha. Only EGF, however, shortened EGFR protein stability (half-life decreased from 5 h to 120 min), resulting in rapid disappearance of newly synthesized EGFR protein. Both ligands increased total LNCaP and DU145 cell numbers. These studies demonstrate that the EGF- and TGF-alpha-induced upregulation of EGFR mRNA and protein in human prostate cancer cell lines is complex and occurs at multiple, transcriptional and post-transcriptional levels. Taken together, these data provide novel insight into the molecular mechanisms by which TGF-alpha would preferentially maintain an autocrine loop in human prostate cancer cells. Furthermore, this work suggests that in human prostate cancer cells ligand-specific differential intracellular trafficking of the EGFR plays a major role in regulating its expression.

    Topics: Androgens; Cell Division; Down-Regulation; Epidermal Growth Factor; ErbB Receptors; Humans; Male; Neoplasms, Hormone-Dependent; Prostatic Neoplasms; Protein Processing, Post-Translational; RNA, Messenger; Transcription, Genetic; Transforming Growth Factor alpha; Tumor Cells, Cultured; Up-Regulation

1999
Androgenic regulation of growth factor and growth factor receptor expression in the CWR22 model of prostatic adenocarcinoma.
    International journal of cancer, 1999, Jul-30, Volume: 82, Issue:3

    The effects of androgen manipulation on epidermal growth factor (EGF) receptor, p185erbB-2 and transforming growth factor-alpha (TGF-alpha) levels were examined in prostatic adenocarcinoma. Male nude mice were inoculated with the CWR22 androgen-dependent human prostatic tumor or an androgen-independent (CWR22R) derivative. Mice with CWR22 tumors were castrated and subsequently killed at 3, 7, 21, 28 or 42 days post-castration. Other CWR22-bearing mice received s.c. testosterone pellets at 21 days post-castration and were killed 7 days later. EGF receptor, p185erbB-2 and TGF-alpha levels were examined by immuno-histochemistry. Strong EGF receptor and p185erbB-2 immunostaining was detected in CWR22 tumors from intact controls. EGF receptor immunostaining decreased by 65% to 70% at 21 to 42 days post-castration. Testosterone treatment at 21 to 28 days post-castration resulted in a 2-fold increase in EGF receptor immunostaining. p185erbB-2 immunostaining within CWR22 tumors did not decrease following castration and, in fact, was slightly increased at 7 days post-castration. The effects of castration on EGF receptor and p185erbB-2 levels were confirmed by Western blot analysis. Fewer than 10% of CWR22 tumor cells demonstrated strong TGF-alpha immunostaining, and androgen manipulation did not effect TGF-alpha immunostaining. In contrast, 30% of androgen-independent CWR22R tumor cells were strongly immunostained for TGF-alpha. Our findings indicate that EGF receptor levels, but not p185erbB-2 levels, are strongly dependent on testosterone in CWR22 tumors. The co-localization of TGF-alpha and the EGF receptor in CWR22R tumors suggests that these factors may constitute an autocrine pathway that regulates androgen-independent growth.

    Topics: Adenocarcinoma; Androgens; Animals; Cell Division; Disease Models, Animal; ErbB Receptors; Female; Humans; Immunohistochemistry; Male; Mice; Mice, Nude; Orchiectomy; Prostatic Neoplasms; Receptor, ErbB-2; Transforming Growth Factor alpha; Transplantation, Heterologous; Tumor Cells, Cultured

1999
Growth factors in expressed prostatic fluid from men with prostate cancer, BPH, and clinically normal prostates.
    The Prostate, 1999, Sep-01, Volume: 40, Issue:4

    Although growth factors such as epidermal growth factor (EGF), transforming growth factor (TGF)-alpha, and TGF-beta are important regulators of prostate cell growth in vitro and in animal models, evidence to support their role in human prostate cancer development remains sparse. We previously showed that men without prostate cancer have concentrations of EGF and TGF-alpha in expressed prostatic fluid (EPF) that are individually distinct and stable over time. This study addressed whether growth factor levels in EPF are associated with the presence or progression of prostate cancer.. We measured levels of immunoreactive EGF, TGF-alpha, and TGF-beta1 in stored EPF samples from three age-matched groups: 19 men with untreated, histologically diagnosed prostate cancer (CaP), 38 with benign prostate hyperplasia (BPH), and 19 with normal prostate glands (NPD).. Median TGF-alpha was lower in the BPH group (0.45 ng/ml) than in either CaP (0.63 ng/ml) or NPD (0.58 ng/ml) groups (P = 0.03 and 0.12, respectively). For EGF, the median was lowest in the CaP group and highest in the NPD group (92.5 ng/ml vs. 175.5 ng/ml, P = 0.006). For TGF-beta1, the median level in CaP was 2.7 times higher than the median level among all controls (6.65 ng/ml vs. 2.46 ng/ml, P = 0.002). Growth factor levels were not associated with tumor stage or Gleason score. However, the single case with distant metastases had TGF-beta1 levels 23-fold higher than the CaP median.. The results suggest that at the time of CaP diagnosis, EGF levels in EPF are significantly lower, and TGF-beta1 levels significantly higher, than normal. Marked overexpression of TGF-beta1 in advanced CaP might be reflected in extremely high EPF levels.

    Topics: Aged; Disease Progression; Epidermal Growth Factor; Growth Substances; Humans; Male; Prostate; Prostatic Hyperplasia; Prostatic Neoplasms; Radioimmunoassay; Transforming Growth Factor alpha; Transforming Growth Factor beta

1999
Immunohistochemical comparative analysis of transforming growth factor alpha, epidermal growth factor, and epidermal growth factor receptor in normal, hyperplastic and neoplastic human prostates.
    Cytokine, 1999, Volume: 11, Issue:9

    Immunoreaction to TGF-alpha was limited to the basal epithelial cells of focal areas in the normal prostates. In benign prostatic hyperplasia (BPH) the immunostained areas were more widespread and immunolabelling was observed in both basal and columnar (secretory) cells of the epithelium. Some cells in the connective tissue stroma were also stained. In prostatic adenocarcinoma, epithelial immunostaining was even more extensive and intense than in BPH, and some stromal cells were also stained. Epidermal growth factor (EGF) immunostaining was only present in some basal cells in normal prostates. In BPH, this immunoreaction was strong in the basal cells and even stronger in the secretory cells. In prostatic cancer, the intensity of epithelial cell immunoreactivity was intermediate between that of normal prostates and that of BPH specimens. EGF-receptor immunostaining was focal and located in the basal cells in normal prostates. In BPH, labelling was also localized in basal cells but extended to wider areas. Some stromal cells appeared weakly labelled. In the prostatic carcinoma, both basal and columnar cells appeared stained and the number of immunolabelled stromal cells was higher than in BPH. The results presented suggest that, in normal conditions, EGF and TGF-alpha act as autocrine growth factors for the basal cells of the prostatic epithelium. In BPH this action is maintained and, in addition, the columnar cells start to secrete both factors which are bound by the basal cell receptors, giving rise to a paracrine regulation which probably overstimulates basal cell proliferation. In prostatic carcinoma, besides these regulatory mechanisms, the acquisition of EGF-receptors by the secretory cells develops an autocrine regulation which might induce their proliferation.

    Topics: Adenocarcinoma; Aged; Aged, 80 and over; Animals; Antibody Specificity; Connective Tissue; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Humans; Immunoenzyme Techniques; Male; Mice; Middle Aged; Neoplasm Proteins; Organ Specificity; Prostate; Prostatic Hyperplasia; Prostatic Neoplasms; Rabbits; Stromal Cells; Transforming Growth Factor alpha

1999
Modulation of cellular proliferation and production of prostate-specific antigen and matrix adhesion molecules in human prostate carcinoma cells by polypeptide growth factors: comparative analyses of MDA PCa2a with established cell lines.
    International journal of oncology, 1998, Volume: 12, Issue:3

    The cellular responses of a newly established and early-passage human prostate adenocarcinoma cell line, MDA PCa2a, to transforming growth factor (TGF) beta1, epidermal growth factor (EGF), and TGFalpha were characterized in terms of proliferation, production of prostate-specific antigen (PSA), fibronectin (FN) and laminin (LM). The responses of the MDA PCa2a cells were compared with those of the well-established human prostate carcinoma cell lines LNCap, PC3, and DU145. The MDA PCa2a cells were more responsive to the growth-inhibitory effect of TGFbeta1 than the established cell lines. The androgen-responsive cell lines (MDA PCa2a and LNCap) were relatively responsive to the growth-stimulatory effect of EGF and TGFalpha whereas the androgen-independent lines (PC3 and DU145) were not. Only the androgen-responsive cells produced PSA, which was further upregulated by treatment with growth factors. The androgen-independent cells did not produce PSA, and growth factors had no effect on PSA production. However, all cell lines produced abundant amounts of FN and LM, and the levels of production of these molecules were subject to modulation by growth factors. It is concluded that each growth factor elicits diverse and distinct responses in prostate carcinoma cells, which may reflect the involvement of diverse post-receptor signal pathways.

    Topics: Adenocarcinoma; Cell Division; Epidermal Growth Factor; Fibronectins; Growth Substances; Humans; Laminin; Male; Prostate-Specific Antigen; Prostatic Neoplasms; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Cells, Cultured

1998
Basic FGF, EGF, and PDGF modify TGFbeta-induction of smooth muscle cell phenotype in human prostatic stromal cells.
    The Prostate, 1998, Volume: 35, Issue:2

    We investigated the ability of a variety of growth factors to regulate the differentiation of prostatic fibroblasts into smooth muscle cells.. Smooth muscle actin levels were monitored by immunoblot analysis and immunocytochemistry. Proliferation was measured in clonal growth assays and by cell counts.. We determined that TGFbeta inhibited proliferation and induced smooth muscle differentiation of stromal cells derived from prostatic adenocarcinomas, as we previously reported for cells derived from the normal peripheral zone. Basic FGF, EGF, TGFalpha, and PDGF, but not IGF, retinoic acid, 1,25-dihydroxyvitamin D3, or androgen, attenuated induction of differentiation by TGFbeta, by a mechanism apparently unrelated to proliferation.. Regulation of growth and differentiation occurs equivalently in prostatic stromal cells derived from adenocarcinomas and normal peripheral zone. TGFbeta is a potent inducer of the smooth muscle phenotype. Basic FGF, EGF and/or TGFalpha, and PDGF attenuate TGFbeta's activity, and promote a fibroblastic phenotype. Our studies provide an in vitro model system in which fibroblastic or smooth muscle cells can be promoted, maintained, and investigated in a defined manner. The results suggest that the ratio of fibroblasts to smooth muscle cells in the stroma reflects the relative levels of growth factors, which may be altered in diseased states.

    Topics: Actins; Adenocarcinoma; Epidermal Growth Factor; Fibroblast Growth Factor 2; Humans; Immunohistochemistry; Male; Muscle, Smooth; Phenotype; Platelet-Derived Growth Factor; Prostatic Neoplasms; Transforming Growth Factor alpha

1998
The localization of transforming growth factor alpha and epidermal growth factor receptor in stromal and epithelial compartments of developing human prostate and hyperplastic, dysplastic, and carcinomatous lesions.
    Human pathology, 1998, Volume: 29, Issue:7

    To gain insight into autocrine/paracrine mechanisms that may influence normal and abnormal growth of the human prostate, we studied the immunohistochemical localization of transforming growth factor alpha (TGF-alpha) and epidermal growth factor receptor (EGFr) in fetal, neonatal, prepubertal, and young adult glands. Results were compared with findings in specimens of benign prostatic hyperplasia (BPH), dysplasia (prostatic intraepithelial neoplasia--PIN), and carcinoma. EGFr was strongly and exclusively expressed in fetal basal cells, whereas TGF-alpha was localized in these and secretory cells as well as in differentiating smooth muscle cells. In neonatal and prepubertal glands, EGFr continued to be found only in basal cells, whereas TGF-alpha was now present in smooth muscle and infrequently in secretory cells. In the normal adult prostate, the receptor was strictly localized in basal cells and in the lateral plasma membranes of secretory cells, whereas its ligand was exclusively expressed in smooth muscle. This pattern persisted in PBH, but both EGFr and TGF-alpha staining appeared to be enhanced in their respective cellular compartments. Irrespective of grade, in dysplasia diffuse-moderate EGFr and strong TGF-alpha staining were both present in a majority of secretory cells. Similarly, most cells in Gleason grade 3 and 4 carcinomas expressed both EGFr and TGF-alpha. Our findings suggest that an unregulated paracrine mode of growth attends the development of BPH, whereas malignant transformation and progression involves autocrine/paracrine mechanisms reminiscent of those found in the developing prostate.

    Topics: Adult; Aged; Carcinoma; Child; Epithelial Cells; ErbB Receptors; Fetus; Humans; Immunoenzyme Techniques; Infant, Newborn; Male; Middle Aged; Prostate; Prostatic Hyperplasia; Prostatic Intraepithelial Neoplasia; Prostatic Neoplasms; Stromal Cells; Transforming Growth Factor alpha; Up-Regulation

1998
Contrasting patterns of regulation of the antioxidant selenoproteins, thioredoxin reductase, and glutathione peroxidase, in cancer cells.
    Biochemical and biophysical research communications, 1998, Oct-20, Volume: 251, Issue:2

    There is strong evidence that selenium protects against certain human cancers, but the underlying mechanism is unknown. Glutathione peroxidase (GPX1) and thioredoxin reductase (TR), the most abundant antioxidant selenium-containing proteins in mammals, have been implicated in this protection. We analyzed the expression of TR and GPX1 in the following model cancer systems: (1) liver tumors in TGFalpha/c-myc transgenic mice; (2) human prostate cell lines from normal and cancer tissues; and (3) p53-induced apoptosis in a human colon cancer cell line. TR was induced while GPX1 was repressed in malignancies relative to controls in transgenic mice and prostate cell lines. In the colon cell line, p53 expression resulted in elevated GPX1, but repressed TR. The data indicate that TR and GPX1 are regulated in a contrasting manner in the cancer systems tested and reveal the p53-dependent regulation of selenoprotein expression. The data suggest that additional studies on selenoprotein regulation in different cancers are required to evaluate future implementation of selenium as a dietary supplement in individuals at risk for developing certain cancers.

    Topics: Animals; Apoptosis; Carcinoma, Hepatocellular; Cell Line; Colonic Neoplasms; Enzyme Induction; Epithelial Cells; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Neoplastic; Genes, myc; Glutathione Peroxidase; Humans; Liver Neoplasms; Male; Mice; Mice, Inbred CBA; Mice, Transgenic; Prostate; Prostatic Neoplasms; Proto-Oncogene Proteins c-myc; Thioredoxin-Disulfide Reductase; Transcription, Genetic; Transforming Growth Factor alpha; Tumor Cells, Cultured; Tumor Suppressor Protein p53

1998
Expression of androgen receptor and growth factors in premalignant lesions of the prostate.
    The Journal of pathology, 1998, Volume: 186, Issue:2

    Analysis of growth factors and receptors in putative premalignant lesions of prostatic adenocarcinoma should aid our understanding of their growth pathways. Sixty prostatic TURP (transurethral resection of the prostate) specimens exhibiting atypical adenomatous hyperplasia (AAH) and/or prostatic intraepithelial neoplasia (PIN) lesions were assayed by immunohistochemistry for androgen receptor (AR), epidermal growth factor receptor (EGFR), c-erbB-2, transforming growth factor-alpha (TGF-alpha), vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2), MIB-1, E-cadherin, and high molecular weight keratin. Expression of these factors in the lesions was compared with that in the co-existing benign prostatic hyperplasia (BPH) or prostatic adenocarcinoma. Strong AR nuclear staining was observed in the luminal cells, but not the basal cells, of BPH and PIN lesions and in all the carcinomas examined. A similar growth factor and receptor profile was demonstrated in the secretory epithelium of high-grade PIN and carcinoma with a tendency to higher expression of membranous EGFR and c-erbB-2 and cytoplasmic TGF-alpha, and lower levels of FGF-2 than in low-grade PIN or BPH glands. Also, increased rates of proliferation, as estimated by MIB-1 stained cells, were observed in high-grade PIN in comparison with low-grade PIN and BPH and were not confined to the basal layer. AAH lesions resembled neither BPH nor carcinoma. Proliferation was virtually absent (MIB-1 expression); both AR and E-cadherin expression was significantly reduced; and, with the exception of FGF-2, all the other growth factors and receptors studied were absent. The results presented would support a premalignant role for high-grade PIN, whilst AAH would appear to represent a quiescent phenotype unlikely to progress to neoplasia.

    Topics: Antigens, Nuclear; Cadherins; Endothelial Growth Factors; ErbB Receptors; Fibroblast Growth Factor 2; Growth Substances; Humans; Ki-67 Antigen; Lymphokines; Male; Nuclear Proteins; Precancerous Conditions; Prostatic Hyperplasia; Prostatic Intraepithelial Neoplasia; Prostatic Neoplasms; Receptor, ErbB-2; Receptors, Androgen; Transforming Growth Factor alpha; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

1998
Growth factor deprivation therapy of hormone insensitive prostate and breast cancers utilizing antisense oligonucleotides.
    Methods and findings in experimental and clinical pharmacology, 1998, Volume: 20, Issue:10

    Antisense oligonucleotides (oligos) are artificial sequences of nucleotide bases which may be synthesized complementary to known regions within specific mRNAs. When these constructed oligos interact with protein encoding mRNA they may regulate expression of various growth factors and/or their receptors. Oligos directed against transforming growth factor-alpha (TGF-alpha) and its binding site, the epidermal growth factor receptor (EGFR), were employed: A) in vitro to affect the growth of hormone insensitive human derived PC-3 prostate cancer cells as well as the human derived UACC-893 breast cancer cell line; and B) in vivo to treat tumors established by these cell lines in athymic nude mice. The in vitro results for each oligo, and their combination, produced significant inhibition of both prostate and breast cell lines. In addition, the combination of oligos most efficiently diminished the immunohistochemical expression of both TGF-alpha and EGFR in PC-3 cells. Direct in vivo inoculation of oligos into established PC-3 or UACC-893 tumors in nude mice produced hemorrhagic necrosis within 2-3 days. Such therapy could represent a new tier of therapy for recurrent, hormone insensitive, tumors based upon the concept of growth factor deprivation.

    Topics: Animals; Binding Sites; Breast Neoplasms; ErbB Receptors; Growth Substances; Humans; Male; Mammary Neoplasms, Animal; Mice; Mice, Nude; Oligonucleotides, Antisense; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured

1998
The prostate gland and prostate carcinogenesis.
    Italian journal of anatomy and embryology = Archivio italiano di anatomia ed embriologia, 1998, Volume: 103, Issue:4 Suppl 1

    Despite extensive research, the mechanisms of prostate carcinogenesis are not well understood. The slow progress in this area is due, at least in part, to lack of a suitable animal model for prostate carcinogenesis. We have developed an animal model, based on the existing sex hormone-induced prostate carcinogenesis in the Noble rat, by substantially increasing the dosage of testosterone while keeping the level of estrogen unchanged. Using the modified method of combination of testosterone and estradiol-17beta (T+E2), it has been shown in Noble rats that prostate carcinogenesis followed a multi-step process involving hyperplasia, dysplasia, and carcinoma. We have demonstrated the importance of TGF-alpha, TGF-beta1 and bFGF in the development of prostate carcinogenesis. This study also established the roles of VEGF and IGF-1, initially as paracrine factors in epithelial-stromal interactions during the process of carcinogenesis and subsequently switching over to an autocrine mode during the establishment of carcinoma.

    Topics: Adenocarcinoma; Animals; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Therapy, Combination; Endothelial Growth Factors; Estradiol; Fibroblast Growth Factor 2; Hyperplasia; Immunohistochemistry; Lymphokines; Male; Precancerous Conditions; Prostate; Prostatic Neoplasms; Rats; Rats, Inbred Strains; Receptor Protein-Tyrosine Kinases; Receptors, Growth Factor; Receptors, Vascular Endothelial Growth Factor; Testosterone; Transforming Growth Factor alpha; Transforming Growth Factor beta; Transforming Growth Factor beta1; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

1998
Comparative studies of the mitogenic effects of epidermal growth factor and transforming growth factor-alpha and the expression of various growth factors in neoplastic and non-neoplastic prostatic cell lines.
    The Prostate, 1997, Mar-01, Volume: 30, Issue:4

    The role of growth factors in prostate cell growth has been investigated as these peptides may be involved in the autonomous growth of hormone-independent prostate cancer.. Responses of neoplastic (PC-3 and CPA) and non-neoplastic (CAPE) prostatic cell lines to epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-alpha) were determined using clonogenic and growth curve analysis. The constitutive expression of EGF, TGF-alpha, and TGF-beta 1-3 mRNA was examined using Northern blotting and EGF and TGF-alpha protein levels were determined immunohistochemically.. Growth curve and clonogenic analysis indicated that EGF and TGF-alpha were mitogenic in each cell line. The magnitude of the clonogenic response varied between the cell lines, with CPA cells showing the greatest growth increases. CPA cells also displayed the highest levels of EGF and TGF-alpha mRNA and protein. TGF-beta 1 mRNA was detected in the order of magnitude, PC-3 > CPA > CAPE. Furthermore, PC-3 and CPA cells expressed TGF-beta 3 and TGF-beta 2 transcripts respectively. In each cell line, the expression of any growth factor mRNA was not affected by exogenous EGF.. The growth responses of the cell lines to EGF and TGF-alpha did not correlate with their constitutive levels of EGF and TGF-alpha mRNA and protein, thus whilst growth factors may be important in malignant cell growth, other pathways may also be involved in the autocrine regulation of cell proliferation.

    Topics: Animals; Cell Division; Cell Line; Dogs; Epidermal Growth Factor; Growth Substances; Humans; Male; Mitogens; Prostate; Prostatic Neoplasms; RNA, Messenger; Transforming Growth Factor alpha; Transforming Growth Factor beta

1997
Enhanced expression of bcl-2 following antisense oligonucleotide mediated growth factor deprivation.
    Medical oncology (Northwood, London, England), 1997, Volume: 14, Issue:1

    Although the role of bcl-2 in apoptosis has been described, its involvement in prostate cancer (CAP) progression is less well understood, but thought to be involved with the transition of CAP from androgen-sensitivity to androgen-independence, where its expression is augmented following androgen ablation. For treating these recurrent androgen-independent tumors, following hormone treatment failure, a new tier of therapy based upon growth factor deprivation has been suggested, implemented by antisense oligonucleotides (oligos) directed against mRNA encoding a critical growth regulatory autocrine loop (comprised of transforming growth factor-alpha (TGF-alpha) and its binding site, the epidermal growth factor receptor (EGFR). To determine whether oligo-induced growth factor deprivation therapy similarly enhanced expression of bcl-2 (as follows androgen deprivation) human prostate cancer derived PC-3 cells were treated in vitro with oligos directed against TGF-alpha (MR-1) and/or EGFR (MR-2). After 5 days of treatment cells were immunochemically stained for human bcl-2. In similar experiments, cells were treated for 3 days prior to extraction of proteins, Western blot analysis, photography and computer evaluation of protein density by SigmaScan software. Immunostained cells treated with oligos directed against mRNA encoding TGF-alpha (MR-1) either alone or in combination with that directed against EGFR (MR-2) had increased bcl-2 expression (+3 to +5). In addition, the intensity of Western blots scanned for bcl-2 expression were 19%, 32% and 30% greater in cells treated with oligos directed against TGF-alpha, EGFR or their combination, respectively. We conclude that enhanced bcl-2 expression followed antisense oligo induced growth factor deprivation. This result is similar to that found upon androgen deprivation therapy, and also demonstrates additional biologic activity of this new class of molecular therapeutic agents.

    Topics: Apoptosis; Blotting, Western; ErbB Receptors; Humans; Immunohistochemistry; Male; Oligonucleotides, Antisense; Prostatic Neoplasms; Proto-Oncogene Proteins c-bcl-2; Transforming Growth Factor alpha; Tumor Cells, Cultured

1997
ErbB kinases and NDF signaling in human prostate cancer cells.
    Oncogene, 1997, Nov-27, Volume: 15, Issue:22

    Prostate carcinoma (PCA) is the most commonly diagnosed malignancy in American men. Our knowledge of PCA growth regulation lags behind that of other cancers, such as breast and colon carcinomas. Among receptor tyrosine kinases, the ErbB family is most frequently implicated in neoplasia. We report here the expression of ErbB family kinases and their ligands in PCA cell lines and a xenograft. While ErbB1/EGFR, ErbB2/NEU, and ErbB3 were always observed in a distinct pattern, ErbB4 was not observed. Interestingly, while TGF-alpha was expressed in the majority of PCA lines, the ligand Neu Differentiation Factor/Heregulin (NDF) was expressed only in an immortalized, non-transformed prostate epithelial line. Concomitantly, there was a significant difference in biological response to these ligands. NDF inhibited LNCaP growth and induced an epithelial-like morphological change, in contrast to TGF-alpha, which accelerated cell growth. We also performed the first comprehensive analysis of NDF signaling in a prostate line. LNCaP stimulated with NDF demonstrated crosstalk between ErbB3 and ErbB2 which did not involve ErbB1. NDF also turned on several cascades, including those of PI3-K, ERK/MAPK, mHOG/p38 and JNK/SAPK, but not those of PLCgamma or the STAT family. This signaling pattern is distinct from that of TGF-alpha. The activation of mHOG by ErbB2 or ErbB3 has not been reported, and may contribute to the unusual phenotype. PI3-K activation is characterized by the formation of a striking 'activation complex' with multiple tyrosine-phosphorylated species, including ErbB3. Our studies provide a framework in which to dissect the growth and differentiation signals of prostate cancer cells.

    Topics: Calcium-Calmodulin-Dependent Protein Kinases; Carcinoma; Cell Division; DNA-Binding Proteins; Enzyme Activation; ErbB Receptors; Glycoproteins; Humans; Isoenzymes; Male; Milk Proteins; Mitogen-Activated Protein Kinases; Neuregulins; p38 Mitogen-Activated Protein Kinases; Phosphatidylinositol 3-Kinases; Phospholipase C gamma; Phosphorylation; Prostatic Neoplasms; Proto-Oncogene Proteins; Receptor, ErbB-2; Receptor, ErbB-3; Recombinant Proteins; Signal Transduction; STAT1 Transcription Factor; STAT3 Transcription Factor; STAT5 Transcription Factor; Trans-Activators; Transforming Growth Factor alpha; Transplantation, Heterologous; Tumor Cells, Cultured; Type C Phospholipases; Tyrosine

1997
Modulation of aromatase activity by growth factors in an androgen sensitive human prostate cancer cell line, LNCaP.
    Cancer letters, 1996, Apr-19, Volume: 102, Issue:1-2

    The effects of steroids and peptide growth factors on aromatase activity in an androgen sensitive human prostate cancer cell line (LNCaP) were investigated. Factors were selected based on their observed modulation of the enzyme in other tissues. Incubation with epidermal growth factor and transforming growth factor-I decreased aromatase activity in LNCaP cells by 25-40%. Insulin like growth factor-1, dexamethasone, dibutyryl cAMP and phorbol 12-myristate 13-acetate, all of which are modulators of aromatase in other tissues, had no significant effect on aromatase activity in LNCaP cells. In addition, the cAMP-dependent protein kinase and protein kinase C inhibitor 1-(5-isoquinolinylsulfonyl)-2 methylpiperazine (H-7) had no effect on the enzyme activity. Factors affecting prostatic aromatase may be distinct from those for other known species.

    Topics: Androgens; Aromatase; Cell Division; Epidermal Growth Factor; Growth Substances; Humans; Kinetics; Male; Neoplasms, Hormone-Dependent; Prostatic Neoplasms; Signal Transduction; Transforming Growth Factor alpha; Tumor Cells, Cultured

1996
Comparative analysis of mRNA and protein expression for epidermal growth factor receptor and ligands relative to the proliferative index in human prostate tissue.
    Human pathology, 1996, Volume: 27, Issue:7

    The expression of epidermal growth factor receptor (EGF-R), transforming growth factor alpha (TGFalpha), and epidermal growth factor (EGF) was evaluated in a series of prostate cancer (CaP; n = 55) and benign prostate hyperplasia (BPH; n = 44) specimens using immunocytochemistry (ICC) and Northern blotting. In situ hybridization (ISH), performed on a subgroup of these specimens, proved to be a more informative technique for the assessment of messenger RNA (mRNA) in this heterogeneous tissue. A comparative analysis was made in relation to the proliferative index, assessed using the MIB-1 antibody. Elevated levels of EGF-R and TGFalpha, mRNA, and protein were observed in carcinoma cells compared with benign, secretory epithelium using in situ hybridization and immunocytochemistry. In carcinoma specimens evidence of an autocrine growth loop is provided by a correlation between EGF-R and TGFalpha, mRNA (P < .0001), and protein expression (P < .01). A trend toward increased expression of EGF-R and TGFalpha protein with dedifferentiation and a similar trend in the growth fraction suggest a role in tumor progression. Although there was a correlation between EGF-R and the proliferative index (P < .01), no relationship was found between this latter parameter and TGFalpha immunoreactivity (P > .05), indicating that this growth factor may be linked with other aspects of malignant activity rather than directly stimulating proliferation.

    Topics: Cell Division; Epidermal Growth Factor; ErbB Receptors; Humans; Immunohistochemistry; In Situ Hybridization; Ligands; Male; Prostate; Prostatic Hyperplasia; Prostatic Neoplasms; RNA, Messenger; RNA, Neoplasm; Transforming Growth Factor alpha

1996
Tyrosine kinase inhibitor as a novel signal transduction and antiproliferative agent: prostate cancer.
    Molecular and cellular endocrinology, 1996, Mar-01, Volume: 117, Issue:1

    In prostate cancer cells, the binding of peptide growth factors to specific receptors increases tyrosine kinases (TK) activity to regulate cell proliferation, cell differentiation, and signaling processes. To determine whether inhibition of receptor TK activity inhibits tumor growth, we studied the effects of a tyrosine kinase inhibitor, RG-13022 (tyrphostin), on cultured human prostate cancer cells. RG-13022 significantly inhibited TGF alpha-induced phosphorylation of EGF receptor (EGFR). This compound inhibited TGF alpha-stimulated [3H]thymidine incorporation in a dose-dependent manner with IC50 being 30 microM. Clonogenicity in soft agar was reduced in the presence of RG-13022. Inhibitory effects were also observed in androgen-positive LNCaP cells and androgen-negative PC3 cells. RG-13022 not only inhibited TGF alpha-induced growth but also growth stimulated by epidermal growth factor (EGF), acidic fibroblast growth factor (aFGF) and serum. In addition, RG-13022 also blocked androgen-stimulated cell proliferation, suggesting that functioning TK pathways are required for androgen-induced growth. This novel synthetic inhibitor may be useful in providing a new strategy for future therapeutic intervention for prostate cancer.

    Topics: Androgens; Antineoplastic Agents; Cell Division; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Humans; Male; Nitriles; Phosphatidylinositol 3-Kinases; Phosphorylation; Phosphotransferases (Alcohol Group Acceptor); Prostate; Prostatic Neoplasms; Protein-Tyrosine Kinases; Pyridines; Signal Transduction; Transforming Growth Factor alpha; Tumor Cells, Cultured; Tyrphostins

1996
Steroid-growth factor interaction in human prostate cancer. 2. Effects of transforming growth factors on androgen metabolism of prostate cancer cells.
    Steroids, 1996, Volume: 61, Issue:1

    The ability of human prostate cancer cells to metabolize androgens was assessed through administration of physiological concentration (0.5-10 nM) of tritiated testosterone (T) as precursor and one-step analysis of both T degradation and products' formation by reverse-phase HPLC and on-line radioactive detection after either 24 h or 72 h incubation. Overall, different prostate cancer cells degraded T quite differently, favoring alternatively reductive or oxidative metabolic pathways. In particular, both LNCaP and DU145 cells retained high levels of unconverted T, with a limited production of androstenedione and its 17-keto derivatives and relatively high amounts of dihydrotestosterone (DHT) and 3 alpha-androstanediol (3 alpha-diol). In contrast, PC3 cells quickly degraded T and exhibited high formation rates of androstenedione and 17-keto metabolites, while neither dihydrotestosterone nor 3 alpha-diol were detected after short or longer incubation times. The effects of both TGF alpha (50 ng/mL) and TGF beta 1 (5 ng/mL) on rates and direction of T metabolism were also explored. In LNCaP cells TGF alpha induced a significant (P < 0.04) decrease of the reductive metabolism of T with a corresponding enhancement of the oxidative pathway (P < 0.002), while TGF beta 1 did not significantly affect T metabolism. On the other hand, both reductive and oxidative pathways were only partially influenced by either growth factor in DU145 and PC3 cells, although TGF alpha significantly raised 5 alpha-androstanedione formation and reduced androsterone production in DU145 cells. All the above evidence was confirmed at both 24 h and 72 h or using increasing doses of TGF alpha and TGF beta 1, a peak activity of 50 ng/mL and 5 ng/mL, respectively, being generally encountered. Overall, our data suggest that TGFs may have a role in the growth regulation of hormone-responsive prostate tumor cells through changes of the intracellular contents of biologically active androgen metabolites.

    Topics: Androgens; Humans; Male; Prostatic Neoplasms; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Cells, Cultured

1996
Transforming growth factor-alpha, epidermal growth factor, and epidermal growth factor receptor in human testis obtained from biopsy and castration: immunohistochemical study.
    The Tohoku journal of experimental medicine, 1996, Volume: 178, Issue:4

    We analyzed expression of transforming growth factor (TGF)-alpha, epidermal growth factor (EGF) and their receptor, EGF receptor (EGFR), by immunohistochemistry in the human testis to determine the possible roles of these growth factors in human testicular function. Specimens were obtained from 17 patients including 9 patients with infertility, 4 patients with prostatic carcinoma and 4 patients with contralateral testicular tumor. EGF immunoreactivity was positive in the hyperplasic Leydig cells of one patient but negative in the other cases. On the other hand, strong TGF-alpha immunoreactivity was observed in Leydig cells, with weak staining in Sertoli cells and germ cells in cases with normal spermatogenesis. EGFR immunoreactivity was observed in the Leydig and peritubular cells, appearing as membrane staining. Marked immunoreactivity for TGF-alpha was observed in the Sertoli cells in testes with decreased spermatogenesis, especially in the Sertoli-cell-only syndrome. This finding may indicate a compensatory increase of TGF-alpha expression in the Sertoli cells accompanying a decrease in spermatogenesis. No significant correlation was found between the degrees of spermatogenesis and immunolocalization of the EGF receptor. These findings suggest that TGF-alpha is a locally produced growth factor that is involved in spermatogenesis in the human testis via an autocrine and/or paracrine mechanism.

    Topics: Adolescent; Adult; Aged; Aged, 80 and over; Epidermal Growth Factor; ErbB Receptors; Humans; Immunohistochemistry; Infertility, Male; Male; Middle Aged; Orchiectomy; Prostatic Neoplasms; Spermatogenesis; Testicular Neoplasms; Testis; Transforming Growth Factor alpha

1996
Vascular endothelial growth factor (VEGF) expression in prostatic tumours and its relationship to neuroendocrine cells.
    British journal of cancer, 1996, Volume: 74, Issue:6

    Vascular endothelial growth factor (VEGF) expression was examined by immunohistochemistry in 45 prostatic carcinoma specimens and ten benign prostatic tumours (BPH). The majority of carcinoma specimens exhibited cytoplasmic staining for VEGF and showed a trend of increasing expression with dedifferentiation (2p = 0.003). Immunoreactive VEGF was also seen in the prostatic carcinoma cell lines, the order of staining intensity was PC3 > DU145 > LNCaP. Intense granular cytoplasmic staining for VEGF was observed in neuroendocrine-like cells which were seen focally in many of the prostatic specimens. Consecutive sections were incubated with a chromogranin A antibody to confirm the neuroendocrine phenotype of these cells. A significant correlation (P < 0.0001) between the total number of intensely stained VEGF-positive cells and chromogranin A-positive cells was found. A subpopulation of neuroendocrine-like cells also showed intense immunoreactivity for transforming growth factor alpha (TGF-alpha). A correlation was observed (2p = 0.0092) between the intensity of VEGF and TGF-alpha immunostaining in carcinoma cells which were not of neuroendocrine differentiation. The presence of these two angiogenic factors may aid the neovascularisation of carcinomas and their increased expression in tumour-associated neuroendocrine cells may contribute to a more aggressive phenotype.

    Topics: Chromogranin A; Chromogranins; Endothelial Growth Factors; Humans; Immunohistochemistry; Lymphokines; Male; Neovascularization, Pathologic; Neurosecretory Systems; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

1996
Involvement of transforming growth factor alpha (TGFalpha) and epidermal growth factor receptor (EGFR) in sex hormone-induced prostatic dysplasia and the growth of an androgen-independent transplantable carcinoma of the prostate.
    Carcinogenesis, 1996, Volume: 17, Issue:12

    We previously reported the induction of dysplasia, a putative precursor of carcinoma, in the dorsolateral prostates (DLPs) of Noble rats by the combined administration of testosterone (T) and estradiol-17beta (E2) for 16 weeks. Additionally, we demonstrated growth of the AIT, a DLP-derived, androgen-independent, transplantable solid tumor, in castrated syngeneic hosts. In this investigation, using Northern blot hybridization, radioimmunoassays and radioligand assays, we showed that transforming growth factor-alpha (TGFalpha) and epidermal growth factor receptor (EGFR) were expressed at close to non-detectable levels in the ventral prostates but at low, but measurable, levels in the DLPs of untreated rats. Enhanced expression of this ligand and its receptor was detected in the DLPs harboring dysplasia and marked overexpression of these molecules was noted in the AIT. In contrast, epidermal growth factor (EGF) expression was found to be constitutively expressed, at high levels, in both normal and dysplastic DLPs, but virtually absent in the AIT. Immunohistochemical data suggested that EGF, TGFalpha and EGFR were aprocine secretory products of the normal DLP, with TGFalpha and EGF localized to the supranuclear complexes and EGFR to the apical membranes of epithelial cells. Alterations in immunostaining patterns for TGFalpha and EGFR were exclusively detected in the dysplastic lesions in the DLPs of T + E2-treated rats. Enhanced intracytoplasmic localization for both peptides were found to accompany the loss of cell polarity in dysplastic foci. Strong intracytoplasmic immunostaining for TGFalpha was observed in some AIT cells whilst staining for EGFR was present in the membranes of tumor cells that formed psuedoacini. Taken together, our findings suggest that autocrine mechanisms may play an important role early in the carcinogenic process and that progression to an androgen-independent neoplastic growth may be modulated by this signaling pathway.

    Topics: Animals; ErbB Receptors; Gonadal Steroid Hormones; Male; Precancerous Conditions; Prostatic Neoplasms; Rats; RNA, Messenger; Transforming Growth Factor alpha

1996
In vitro invasiveness of DU-145 human prostate carcinoma cells is modulated by EGF receptor-mediated signals.
    Clinical & experimental metastasis, 1995, Volume: 13, Issue:6

    Prostate carcinomas often present an autocrine stimulatory loop in which the transformed cells both express the EGF receptor (EGFR) and produce activating ligands (TGF alpha and EGF forms). Up-regulated EGFR signalling has been correlated with tumor progression in other human neoplasia; however, the cell behaviour which is promoted remains undefined. To determine whether an EGFR-induced response contributes to cell invasiveness, we transduced DU-145 human prostate carcinoma cells with either a full-length (WT) or a mitogenically-active but motility-deficient truncated (c'973) EGFR. The DU-145 Parental and two transgene sublines all produced EGFR and TGF alpha, but the transduced WT and c'973 EGFR underwent autocrine downregulation to a lesser degree, with more receptor remaining intact. DU-145 cells transduced with WT EGFR transmigrated a human amniotic basement membrane matrix (Amgel) to a greater extent than did Parental DU-145 cells (175 +/- 22%). Cells expressing the c'973 EGFR invaded through the Amgel only to about two thirds the extent of the Parental cells (62 +/- 23%). A monoclonal antibody which prevents ligand-induced activation of EGFR decreased the invasiveness of WT-expressing cells by half and Parental cells by a fifth, but had little effect on the invasiveness of c'973-expressing cells; with the result that in the presence of antibody, all three cell lines transmigrated the Amgel to the same extent. The different levels of invasiveness between the three sublines were independent of cell proliferation. These findings demonstrated that EGFR-mediated signals increase tumor cell invasiveness and suggested that domains in the carboxy-terminus are required to signal invasiveness. As an initial investigation into the mechanisms underlying the EGFR-mediated enhanced invasiveness, we determined whether these cells presented different collagenolytic activity, as the major constituents of Amgel are collagen types I and IV. All three sublines secreted easily detectable levels of gelatin-directed proteases and TIMP-1, with WT cells secreting equivalent or lower levels of proteases. The proteolytic balance in these cells did not correlate with invasiveness. These data suggest that the TGF alpha-EGFR autocrine loop promotes invasiveness and that this is accomplished by signalling cell properties other than differential secretion of collagenolytic activity.

    Topics: Carcinoma; Collagenases; Endopeptidases; ErbB Receptors; Extracellular Matrix; Glycoproteins; Humans; Male; Neoplasm Invasiveness; Prostatic Neoplasms; Protease Inhibitors; Tissue Inhibitor of Metalloproteinases; Transfection; Transforming Growth Factor alpha; Tumor Cells, Cultured

1995
Androgen up-regulates epidermal growth factor receptor expression and binding affinity in PC3 cell lines expressing the human androgen receptor.
    Cancer research, 1995, Jul-15, Volume: 55, Issue:14

    Androgens are required for the optimal growth and development of both the normal prostate and steroid-sensitive prostate cancer. PC3 prostate cancer cell lines stably expressing the human androgen receptor (AR) and possessing an androgen-sensitive phenotype (PC3-hAR) were used to examine the role of the epidermal growth factor receptor (EGFR) in androgen-stimulated prostate cancer cell growth. Epidermal growth factor (EGF) and dihydrotestosterone (DHT) independently induced the growth of PC3-hAR cells. Moreover, EGF and DHT in combination exerted a synergistic effect on PC3-hAR cell growth. DHT-exposed PC3-hAR cells expressed a greater than 2-fold increase in EGFR mRNA and 50% more EGFR protein than controls. Time course radioligand-binding assays confirmed these findings by showing an elevation in EGF binding in the DHT-exposed PC3-hAR cells. In addition, radioligand competition-binding studies revealed a 2-fold increase in EGFR-EGF binding affinity in the PC3-hAR cells after DHT treatment. However, no enhancement of transforming growth factor alpha or EGF expression was detected because DHT did not affect the levels of these cytokines in the PC3-hAR cell lysate or conditioned media. Our observations suggest that DHT increases both EGFR number and receptor-ligand affinity in androgen-sensitive prostate cancer cells and that these effects correlate with increased EGF binding and an enhanced mitogenic response to EGF.

    Topics: Adenocarcinoma; Androgens; Animals; Bone Neoplasms; Cell Division; Dihydrotestosterone; Drug Synergism; Epidermal Growth Factor; ErbB Receptors; Gene Expression; Gene Expression Regulation, Neoplastic; Humans; Lymphatic Metastasis; Male; Mice; Neoplasms, Hormone-Dependent; Prostatic Neoplasms; Receptors, Androgen; Stimulation, Chemical; Transforming Growth Factor alpha; Tumor Cells, Cultured; Up-Regulation

1995
Growth of the androgen-dependent tumor of the prostate: role of androgens and of locally expressed growth modulatory factors.
    The Journal of steroid biochemistry and molecular biology, 1995, Volume: 53, Issue:1-6

    The crucial role played by androgens in the growth of prostatic carcinoma is now well established. However, the mechanisms of this proliferative action are still poorly understood. Experiments have been performed to clarify: (1) the metabolism of androgens in prostatic tumor cells; and (2) the role played by locally produced growth factors in the autocrine regulation of prostatic tumor cell proliferation and the possible regulation exerted by testosterone (T) on the activity of these factors. These studies have been performed by utilizing the human androgen-responsive prostatic cancer LNCaP cell line. (1) By incubating LNCaP cells with different 14C-labeled androgenic precursors, it has been shown that all the major key enzymes involved in the metabolism of androgens (5 alpha-reductase, 17 beta-hydroxysteroid-oxidoreductase, 3 alpha- and 3 beta-hydroxysteroid-oxidoreductases) are present and active in these cells. In particular, the 5 alpha-reductase, which converts T and delta 4 to DHT and 5 alpha-A respectively, seems to be more active when delta 4 is the substrate, suggesting a preference for this precursor. (2) The hypothesis that LNCaP cells might produce LHRH (or a LHRH-like peptide) has been verified by RT-PCR, performed in the presence of a pair of specific oligonucleotide primers. A cDNA band of the expected size (228 bp), which specifically hybridized with a 32P-labeled LHRH oligonucleotide probe, has been obtained in LNCaP cells. To clarify the possible role played by this factor in the regulation of tumor growth, LNCaP cells, cultured in steroid-free conditions, have been treated with a LHRH antagonist; the treatment resulted in a significant increase of cell proliferation. Taken together, these data indicate that a LHRH (or LHRH-like) growth modulatory system is expressed in LNCaP cells and plays an inhibitory role in the regulation of tumor cell proliferation. This system seems to be regulated in a negative way by steroids. Growth factors endowed with stimulatory activity, such as EGF and TGF alpha, have also been shown to be produced by LNCaP cells. The present studies show that the immunoprecipitation of the EGF receptor with a specific monoclonal antibody (Ab225) reveals a protein band of the expected size (170 kDa) which is phosphorylated even in basal conditions. Moreover, the treatment of LNCaP cells, cultured in serum-free conditions, either with a monoclonal antibody against the EGF receptor, or with immunoneutralizing antibodies aga

    Topics: Androgens; Cell Division; Epidermal Growth Factor; ErbB Receptors; Gonadotropin-Releasing Hormone; Growth Substances; Humans; In Vitro Techniques; Male; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured

1995
Changing pattern of expression of the epidermal growth factor receptor and transforming growth factor alpha in the progression of prostatic neoplasms.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 1995, Volume: 1, Issue:5

    The autocrine/paracrine interaction of the epidermal growth factor receptor (EGFr) and transforming growth factor alpha (TGF-alpha) has been implicated in prostate cancer cell growth and proliferation. To evaluate the role of EGFr and TGF-alpha in prostate cancer progression, we studied the immunohistochemical staining pattern of EGFr and TGF-alpha in malignant primary and hormone-independent metastatic prostate lesions. The specimens evaluated included 37 primary carcinomas (34 hormone-naive and 3 hormone-refractory tumors) and 22 metastases. For each specimen, the pattern of expression was evaluated and staining reactivities graded from 0-3, with 0 representing no staining and 3 representing homogeneous and intense staining. Primary malignant prostate epithelial cells in areas with discrete gland formation showed strong EGFr immunostaining, while stromal cells were generally nonreactive. In untreated primary tumors, TGF-alpha expression was primarily in the stroma, while epithelial cells were weakly positive in several cases. Malignant epithelial cells adjacent to neural elements that stained positive for TGF-alpha was frequently observed. A homogeneous staining pattern for EGFr was noted in 17 (89%) of 19 evaluable androgen-independent-refractory metastases, while TGF-alpha expression was found in 14 (78%) of 18 evaluable cases. Overall, 14 of 18 androgen-independent metastases coexpressed the receptor and the ligand. These results suggest that, unlike primary prostate tumors where a paracrine relationship between EGFr and TGF-alpha appears to predominate, the potential for autocrine stimulation may exist in the majority of metastatic androgen-independent tumors. Furthermore, the changing pattern of expression as the disease evolves from the localized hormone-naive to metastatic androgen-independent condition suggests that strategies aimed at blocking this growth factor pathway may be of therapeutic importance for androgen-independent disease.

    Topics: Adrenal Gland Neoplasms; Bone Neoplasms; Disease Progression; ErbB Receptors; Humans; Liver Neoplasms; Lymphatic Metastasis; Male; Prognosis; Prostatic Neoplasms; Transforming Growth Factor alpha

1995
Strategy for boron neutron capture therapy against tumor cells with over-expression of the epidermal growth factor-receptor.
    International journal of radiation oncology, biology, physics, 1994, Aug-30, Volume: 30, Issue:1

    Gliomas, squamous carcinomas and different adenocarcinomas from breast, colon and prostate might have an increased number of epidermal growth factor (EGF) receptors. The receptors are, in these cases, candidates for binding of receptor specific toxic conjugates that might inactivate cellular proliferation. The purpose of this study was to evaluate whether it is reasonable to try ligand-dextran based conjugates for therapy.. EGF or TGF alpha were conjugated to dextran and binding, internalization, retention and degradation of eight types of such conjugates were analyzed in EGF-receptor amplified glioma cells. The conjugates were labelled with radioactive nuclides to allow detection and two of the conjugates were carrying boron in the form of carboranyl amino acids or aminoalkyl-carboranes. Comparative binding tests, applying 125I-EGF, were made with cultured breast, colon and prostate adenocarcinoma, glioma and squamous carcinoma cells. Some introductory tests to label with 76Br for positron emission tomography and with 131I for radionuclide therapy were also made.. The dextran part of the conjugates did not prevent receptor specific binding. The amount of receptor specific binding varied between the different types of conjugates and between the tested cell types. The dextran part improved intracellular retention and radioactive nuclides were retained for at least 20-24 h. The therapeutical effect improved when 131I was attached to EGF-dextran instead of native EGF.. The improved cellular retention of the ligand-dextran conjugates is an important property since it gives extended exposure time when radionuclides are applied and flexibility in the choice of time for application of neutrons in boron neutron capture therapy (BNCT). It is possible that ligand-dextran mediated BNCT might allow, if the applied neutron fields covers rather wide areas around the primary tumor, locally spread cells that otherwise would escape treatment to be inactivated.

    Topics: Adenocarcinoma; Animals; Boron Compounds; Boron Neutron Capture Therapy; Carcinoma; Colonic Neoplasms; Dextrans; Drug Carriers; Epidermal Growth Factor; ErbB Receptors; Glioma; Humans; Iodine Radioisotopes; Male; Mammary Neoplasms, Experimental; Models, Biological; Neoplasms; Neoplasms, Experimental; Prostatic Neoplasms; Rats; Rats, Sprague-Dawley; Transforming Growth Factor alpha; Tumor Cells, Cultured

1994
Comparative analysis of epidermal growth factor receptor gene expression and protein product in benign, premalignant, and malignant prostate tissue.
    The Prostate, 1994, Volume: 25, Issue:4

    In order to more clearly define the status of epidermal growth factor receptor (EGFR) in prostate cancer, expression of EGFR transcript and protein was analyzed in paired samples of benign and malignant tissues from 30 radical prostatectomy specimens. Prostate tumors and high grade prostatic intraepithelial neoplasias (PINs) expressed significantly less EGFR protein than benign tissues or low grade PINs (P < 0.001). Expression of EGFR mRNA was analyzed in a subset of the same samples, and was higher in more prostate tumors than benign specimens (P < 0.05). However, differences in mean mRNA expression between malignant and benign tissues were not significant. EGFR mRNA was expressed at moderate or low levels in equivalent numbers of PIN lesions. These results suggest that, although EGFR mRNA expression is somewhat elevated in prostate tumors, EGFR protein expression may be down-regulated in the same malignant tissues. Furthermore, our data demonstrate phenotypic similarity between prostate tumors and high grade PIN at the level of EGFR protein expression.

    Topics: Aged; Atrophy; Carcinoma in Situ; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation, Neoplastic; Humans; Immunohistochemistry; In Situ Hybridization; Male; Middle Aged; Precancerous Conditions; Prostate; Prostatic Hyperplasia; Prostatic Neoplasms; RNA, Messenger; Transforming Growth Factor alpha

1994
Expression of transforming growth factor-alpha and the epidermal growth factor receptor in human prostate tissues.
    The Journal of urology, 1994, Volume: 152, Issue:6 Pt 1

    Cells respond to certain soluble factors that bind to cell surface receptors possessing intrinsic tyrosine kinase activity. Overexpression of these molecules has been associated with tumor progression. Enhanced prostatic cancer cell growth in vitro has been reported in the presence of certain growth factors. To characterize the patterns of expression of the epidermal growth factor receptor (EGFr) and transforming growth factor-alpha (TGF alpha), we studied tissue from 107 prostate specimens using immunohistochemistry. We observed that epithelial cells of normal (n = 4) and benign prostatic (n = 56) tissues express EGFr but were unreactive for TGF alpha, while stroma cells in these tissues express TGF alpha but not EGFr. However, coexpression of EGFr and TGF alpha was identified in 22 of 46 prostatic adenocarcinomas studied. These results suggest that the major mode of action of EGFr/TGF alpha in normal and benign prostate is that of a paracrine or juxtacrine loop, the ligand being expressed in the stroma cells and the receptor in the epithelial cells. Since a subset of prostatic carcinomas coexpressed the ligand and the receptor in their tumor cells, it is suggested that an independent autocrine signaling mechanism may occur and grant a selective advantage for the growth of prostate cancers.

    Topics: Adenocarcinoma; ErbB Receptors; Humans; Male; Prostatic Hyperplasia; Prostatic Neoplasms; Transforming Growth Factor alpha

1994
Differential immunoreactivity of transforming growth factor alpha in benign, dysplastic and malignant prostatic tissues.
    Surgical oncology, 1994, Volume: 3, Issue:4

    Immunohistochemical examination of radical prostatectomy specimens from 57 patients was performed to determine the differential expression of transforming growth factor alpha in the human prostate. In addition, epidermal growth factor receptor (EGFr) immunoreactivity was assessed in each case. Stromal versus epithelial staining was determined for each histological subtype: benign prostatic hypertrophy (BPH), prostatic intra-epithelial neoplasia (PIN), and prostatic cancer (CaP) by a single pathologist reviewer. TGFa staining was predominant in stroma while EGFr was localized to the epithelial basal cell layer. Immunoreactivity of both TGFa (P = 0.002) and EGFr (P < 0.001) revealed a significant reduction in CaP compared to BPH or PIN. Autocrine stimulation of EGFr by TGFa or other unrecognized factors may be present in CaP. Conversely, altered stromal influence of CaP via TGFa may be present. These observations could form the basis for future cancer therapeutic strategies using antagonist factors.

    Topics: Aged; ErbB Receptors; Humans; Immunohistochemistry; Male; Middle Aged; Precancerous Conditions; Prostatic Hyperplasia; Prostatic Neoplasms; Transforming Growth Factor alpha

1994
Steroid-growth factor interaction in human prostate cancer. 1. Short-term effects of transforming growth factors on growth of human prostate cancer cells.
    Steroids, 1994, Volume: 59, Issue:7

    In order to better define potential mechanisms of growth regulation in human prostate cancer cells, we have compared biological responses (such as short-term response to both transforming growth factor alpha and beta; TFG alpha and TFG beta) in relation to hormone sensitivity of LNCaP, DU145, and PC3 cells. Androgen receptor (AR) and epidermal growth factor receptor (EGF-R) content of each cell line was also investigated. In addition, expression of EGF, TGF alpha, and TGF beta was evaluated through immunofluorescent staining. Growth of androgen non-responsive PC3 cells was stimulated by TGF alpha (about 35%) and inhibited by TGF beta (more than 50%), with respect to controls, after 48 h exposure. Conversely, AR-positive, hormone-responsive LNCaP cells proved to be poorly sensitive, at least short-term, to either growth factor. Furthermore, high levels of both EGF-R and TGF alpha, and a fairly high amount of EGF, were found in DU145 cells and, to a lesser extent, in LNCaP cells; in contrast, PC3 cells exhibited low expression levels of both receptors (EGF-R) and ligands (EGF, TGF alpha), but displayed remarkable TGF beta binding and relatively high levels of endogenous TGF beta. Overall, these results suggest a differential sensitivity to TGF alpha and TGF beta by prostate cancer cells; TGF alpha response seems not to be proportional to the EGF-R content of individual cell lines.

    Topics: Androgens; Cell Division; Fluorescent Antibody Technique; Humans; Male; Prostatic Neoplasms; Receptors, Androgen; Receptors, Growth Factor; Time Factors; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Cells, Cultured

1994
Tumorigenicity of SV40 T antigen immortalized human prostate epithelial cells: association with decreased epidermal growth factor receptor (EGFR) expression.
    International journal of cancer, 1994, Sep-01, Volume: 58, Issue:5

    Our primary objectives were to: 1) develop a system for the study of prostatic tumor evolution; and 2) examine the role of the epidermal growth factor/epidermal growth factor receptor (EGF/EGFR) pathway in prostate tumor progression. Adult human prostate epithelial cells previously immortalized by transfection with the SV40 T antigen gene (P69SV40T) produced tumors in only 2/18 mice with a 6 month latency period. Reinjection of cells recovered from these tumors after 1 or 2 cycles of growth in nude mice produced tumors in 2/4 and 2/3 mice with markedly decreased latent intervals of 12, 25, 25 and 25 days each. The chromosomal complement of each tumor was human, consistently pseudodiploid, and retained the Y chromosome. In both anchorage-independent and adherent cell growth assays, EGF stimulated proliferation by approximately 2-fold in both the parental P69SV40T line and the tumor sublines. The tumor sublines expressed less EGFR protein than the parental line, as assessed by Western immunoblotting and flow cytometric analysis. Immunoprecipitation revealed increased production of the 18 and 25 kDa TGF-alpha precursors parallel to decreases in detectable EGFR. The growth of both the parental P69SV40T line and the tumor sublines was inhibited by a neutralizing antibody to TGF-alpha under serum-free defined conditions. Inclusion of the TGF-alpha neutralizing antibody consistently inhibited the proliferation of the tumor sublines more than P69SV40T in both proliferation and [3H]thymidine incorporation assays. This finding suggests that the increased tumorigenicity and decreased latent interval observed among the human prostate tumor cells is partially due to activation of the TGF-alpha/EGFR autocrine network.

    Topics: Animals; Antigens, Polyomavirus Transforming; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Karyotyping; Male; Mice; Mice, Nude; Neoplasm Transplantation; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured

1994
Expression of mRNA for epidermal growth factor, transforming growth factor-alpha and their receptor in human prostate tissue and cell lines.
    Molecular and cellular biochemistry, 1993, Sep-22, Volume: 126, Issue:2

    Enhanced expression of the epidermal growth factor receptor (EGFR) or its ligands, epidermal growth factor (EGF) and transforming growth factor alpha (TGF-alpha) can increase signalling via receptor-mediated pathways which may lead to excessive proliferation and cellular transformation. Such autocrine regulation of growth has been demonstrated for prostate cancer cell lines in culture but its role in prostate cancer in vivo has not been established. To assess the potential of such a mechanism, we have examined the pathway components in prostate carcinomas (CaP) in comparison with non-malignant benign prostatic hyperplasias (BPH). In the present study, we investigate the dosage, structure and expression of EGF, TGF-alpha and EGFR genes in a series of 34 human prostate samples and 3 prostate cancer cell lines. All of the samples contained transcripts from each of the genes. The expression of pre-pro-TGF-alpha mRNA and pre-pro-EGF mRNA were significantly higher in CaP (n = 13) than BPH (n = 21) specimens (p < 0.05). The androgen-responsive prostatic carcinoma cell line, LNCaP, expressed high levels of EGF mRNA while the androgen-independent DU145 and PC-3 cell lines expressed high levels of TGF-alpha mRNA and EGFR mRNA. In general, overexpression of these mRNAs was not associated with amplification or detectable gene rearrangement; only DU145 cells demonstrated any alteration in these genes, with apparent amplification of the TGF-alpha gene. Relative to BPH, all prostate carcinomas and cell lines studied had elevated levels of mRNA for one or both mRNA coding for the ligands for EGFR.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation; Humans; Male; Prostate; Prostatic Hyperplasia; Prostatic Neoplasms; RNA, Messenger; Transforming Growth Factor alpha; Tumor Cells, Cultured

1993
Expression of transforming growth factor-alpha, epidermal growth factor and the epidermal growth factor receptor in adenocarcinoma of the prostate and benign prostatic hyperplasia.
    Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc, 1993, Volume: 6, Issue:6

    The present immunohistochemical study examines the expression of TGF alpha, EGF, and the EGF receptor in prostatic tissues obtained from patients with either benign prostatic hyperplasia (BPH) or adenocarcinoma of the prostate. Frozen sections were cut at 5 microns and were incubated with monoclonal antibodies directed against TGF alpha, EGF, or the EGF receptor. The remainder of the staining procedure was performed with an avidin-biotin-complex peroxidase procedure. Strong TGF alpha expression was not detected in any of the 15 BPH specimens examined or in the normal/hyperplastic tissue intermixed within the adenocarcinomas. In contrast, malignant cells in 9 of 11 adenocarcinomas strongly expressed TGF alpha. EGF reactivity was observed in both hyperplastic and malignant epithelial tissues. EGF receptor expression was strongest along the basal aspects of cells of normal or hyperplastic glands. Although most malignant cells also were positive for the EGF receptor the level of staining was less than that observed in cells forming normal or hyperplastic glands. These results suggest that EGF and the EGF receptor may be components of an autocrine/paracrine regulatory pathway involved in hyperplastic and/or malignant disease of the prostate. The finding that TGF alpha is expressed only in malignant cells suggests that TGF alpha may represent a locally active autocrine regulator of malignant prostate cells.

    Topics: Adenocarcinoma; Epidermal Growth Factor; ErbB Receptors; Humans; Immunohistochemistry; Male; Prostatic Hyperplasia; Prostatic Neoplasms; Staining and Labeling; Transforming Growth Factor alpha

1993
[Androgen regulation of secreted growth factors in prostate carcinoma cell and tumor lines].
    Verhandlungen der Deutschen Gesellschaft fur Pathologie, 1993, Volume: 77

    Previous studies have shown that part of steroid hormone action on hormone dependent carcinoma cells is mediated through secreted autocrine and paracrine growth factors. Coculture experiments using the androgen receptor positive human prostate carcinoma cell line LNCaP as feeder cells and the androgen receptor negative prostate cell line DU 145 as indicator cells, such as experiments with conditioned medium suggest that androgens might regulate proliferation of prostate carcinoma through a similar mechanism. LNCaP and DU 145 cells express high affinity EGF-receptors and show an increased growth rate under treatment with EGF, TGF alpha and FGF. The growth stimulating potential of LNCaP-conditioned medium can be enhanced by androgens. The polyanionic compounds suramin and dextran sulfates which have been shown to inactivate a variety of growth factors e.g. EGF/TGF alpha inhibit growth of LNCaP cells and DU 145 cells in a dose dependent and reversible fashion. Growth stimulation of LNCaP cells by EGF/TGF alpha can be completely reversed by simultaneous addition of polyanions but they inhibit androgen stimulation only partially. These data suggest the existence of at least two different mechanisms of growth regulation by androgens which can be distinguished by their different sensitivity of prostatic carcinoma cells to growth factor inhibitory agents. In order to investigate the therapeutic potential of these substances in complex, heterogeneous cell systems of solid tumors we treated 8 representative human prostate cancer lines in the nude mouse model. Systemic applications of polyanions revealed significant growth inhibition in hormone dependent as well as hormone independent xenografts. In androgen responsive lines growth inhibition was intensified by additional androgen withdrawal.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Androgens; Cell Division; Cell Line; Epidermal Growth Factor; ErbB Receptors; Fibroblast Growth Factors; Growth Substances; Humans; Male; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured

1993
Epidermal growth factor and transforming growth factor alpha concentrations in BPH and cancer of the prostate: their relationships with tissue androgen levels.
    British journal of cancer, 1993, Volume: 67, Issue:1

    We measured immunoreactive EGF and TGF alpha in prostate tissue extracts obtained from 19 patients with benign prostatic hyperplasia (BPH) and 19 with cancer of the prostate (CaP). Whilst both BPH and CaP expressed EGF (BPH = 195.61 +/- 19.94 ng g-1 protein; CaP = 235.60 +/- 24.45 ng g-1 protein) and TGF alpha (BPH = 92.57 +/- 7.60 ng g-1 protein; CaP = 100.73 +/- 15.47 ng g-1 protein) in equal concentrations, the levels of EGF in any tissue extract were on average twice those of TGF alpha. Furthermore analysis of the individual growth factor data revealed a direct correlation between EGF and TGF alpha in both BPH (r = 0.72, P < 0.001) and CaP (r = 0.69, P < 0.001). When the tumours were classified according to their Gleason score, a slight but significant increase in growth factor concentrations was noted as the tumour became less differentiated. We also measured the concentrations of testosterone and dihydrotestosterone (DHT) in prostate extracts with a view of elucidating the relationship between androgen and growth factors in this gland. There was a small positive correlation only between testosterone and EGF (r = 0.62, P < 0.05) and testosterone and TGF alpha (r = 0.61, P < 0.05) in CaP. The absence of any similar correlation in BPH where DHT becomes the predominant hormone may suggest an indirect role for testosterone in the regulation of growth factor production.

    Topics: Androgens; Dihydrotestosterone; Epidermal Growth Factor; Humans; Male; Prostatic Hyperplasia; Prostatic Neoplasms; Testosterone; Transforming Growth Factor alpha

1993
Growth factor regulation of gene expression in the human prostatic carcinoma cell line LNCaP.
    Cancer research, 1993, Mar-01, Volume: 53, Issue:5

    In order to characterize the effects of growth factors on the regulation of expression of the genes coding for prostatic differentiation markers, prostatic acid phosphatase and prostate-specific antigen, we studied changes occurring in the biosynthesis of these enzymes in LNCaP prostatic cancer cells treated with growth factors. Epidermal growth factor was found to reduce the secretion of prostatic acid phosphatase and prostate-specific antigen by the cells, as the result of lowered steady-state levels of the corresponding messenger RNAs (mRNAs). In addition, epidermal growth factor (EGF) interfered with the androgen regulation of these genes. EGF evoked these changes in a concentration- and time-dependent fashion, in both the presence and absence of serum and most likely through interactions with the epidermal growth factor receptor, inasmuch as similar effects were achieved by treating the cells with transforming growth factor alpha. The regulation of the human glandular kallikrein 1 gene was quite similar to the regulation of the prostate-specific antigen gene. In addition to the expression of the genes coding for prostatic secretory proteins, the amount of the human androgen receptor mRNA was down-regulated by EGF. This reduction was more pronounced than the autologous down-regulation of human androgen receptor (hAR) mRNA by androgen and could be maintained for at least 5 days. In the presence of androgen, some of the effects of EGF and transforming growth factor alpha on the levels of androgen-regulated mRNAs may be due to down-regulation of the expression of the hAR gene. Transforming growth factor beta 1, which blocked the growth induction of LNCaP cells by EGF, increased the level of prostatic acid phosphatase and hAR mRNAs, but when given to the cells together with EGF its up-regulatory effect could not be discerned. In summary, regulation of the prostatic acid phosphatase and prostate-specific antigen genes is a complex matter, inasmuch as androgens and growth factors regulate the levels of the mRNAs originating from them. Furthermore, the interactions between the androgen-regulatory system and the growth factor-regulatory systems are likely to be at multiple levels in prostatic cells, as suggested by the modulation of the hAR gene expression by these growth factors.

    Topics: Acid Phosphatase; Base Sequence; Carcinoma; Epidermal Growth Factor; Gene Expression Regulation; Humans; Kallikreins; Male; Molecular Sequence Data; Prostate-Specific Antigen; Prostatic Neoplasms; Receptors, Androgen; RNA, Messenger; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Cells, Cultured

1993
An immunocytochemical analysis of TGF alpha expression in benign and malignant prostatic tumors.
    The Prostate, 1993, Volume: 23, Issue:1

    Transforming growth factor alpha (TGF alpha) expression was analyzed immunocytochemically on formalin-fixed wax-embedded sections obtained from 24 benign prostatic hyperplasia (BPH) specimens and 76 prostatic carcinoma tissues, 3 human prostatic tumor xenografts, normal kidney, and salivary gland. Low amounts of TGF alpha immunopositivity were encountered in the epithelium of BPH glandular tissues, whereas in the prostatic adenocarcinoma samples, a greater heterogeneity and intensity of TGF alpha immunostaining was observed. The most intense staining was exhibited by the least differentiated tumors, although a few of these were weakly stained. Statistical analysis of the relationship of histopathological grade of tumor with TGF alpha expression in the carcinomas showed a significant correlation of these parameters, 0.01 > P > 0.001. The expression of the proliferation markers Ki-67 and PCNA was also analyzed in the carcinoma specimens, and the relationship of these to TGF alpha expression indicated that there was no significant correlation in this series of tumors between increased growth activity and TGF alpha expression (p approximately 0.25 with both markers). The prostatic carcinoma xenografts TEN12 and TEN15 contained low levels of immunoreactive TGF alpha, which was uniformly distributed, whilst heterogeneous immunostaining was observed in the uroepithelial xenograft TEN16. In the normal human kidney, TGF alpha was concentrated in the epithelium of the distal convoluted tubules (DCT) and the collecting tubules (CT), and lower amounts were identified in the proximal convoluted tubules (PCT). As in the prostatic carcinomas, the immunostaining was eliminated by prior absorption of the antibody with pure TGF alpha and not with human or mouse EGF. No crossreactivity of the TGF alpha antibody with salivary EGF was demonstrated. This study concludes that, in prostate carcinoma, the least differentiated tumors more often expressed greater amounts immunoreactive TGF alpha; however, no relationship between TGF alpha expression and cellular proliferation markers was found.

    Topics: Adenocarcinoma; Adenocarcinoma, Papillary; Animals; Antigens, Neoplasm; Carcinoma, Squamous Cell; Humans; Immunohistochemistry; Ki-67 Antigen; Kidney; Male; Mice; Mice, Nude; Neoplasm Proteins; Neoplasm Transplantation; Nuclear Proteins; Proliferating Cell Nuclear Antigen; Prostatic Hyperplasia; Prostatic Neoplasms; Submandibular Gland; Transforming Growth Factor alpha

1993
Androgens regulate proliferation of human prostate cancer cells in culture by increasing transforming growth factor-alpha (TGF-alpha) and epidermal growth factor (EGF)/TGF-alpha receptor.
    The Journal of clinical endocrinology and metabolism, 1993, Volume: 77, Issue:6

    Androgens affect growth of the prostate gland and many prostate cancers. Androgens could mediate their mitogenic effects on prostate cells by an autocrine loop involving epidermal growth factor (EGF) and transforming growth factor (TGF)-alpha that bind to the EGF/TGF-alpha receptor. We examined the effects of 5 alpha-dihydrotestosterone (DHT) and testosterone (T), EGF, and EGF-alpha on cell proliferation and 3H-thymidine incorporation in an androgen-dependent human prostate cancer cell line, ALVA101, in serum-free medium. The regulation of TGF-alpha and EGF/TGF-alpha receptor messenger RNA (mRNA) levels were determined by Northern blot analysis and EGF/TGF-alpha receptor protein by immunoblot. After 24 h of treatment of ALVA101 cells with DHT (10(-8) M) or T (10(-8) M), TGF-alpha mRNA levels increased 3- and 2.5-fold, respectively, and EGF/TGF-alpha receptor mRNA levels 2- and 1.5-fold, respectively. Cell numbers increased at day 5 in response to 10(-8) M DHT (18%, P < 0.01), 10(-8) M T (15%, P < 0.01), 20 ng/ml EGF (16%, P < 0.01), and 50 ng/mL TGF-alpha (34%, P < 0.01). DHT combined with TGF-alpha or T combined with EGF increased cell number 43% and 40% above control, respectively (P < 0.01 vs. DHT, P < 0.05 vs. TGF-alpha, T, EGF alone). The anti-EGF/TGF-alpha receptor antibody (528) blocked the cell proliferation induced by either DHT or TGF-alpha. We conclude that DHT and T stimulate synthesis of TGF-alpha and EGF/TGF-alpha receptor mRNAs and EGF/TGF-alpha receptor content in ALVA101 cells. This mitogenic effect of androgen on ALVA101 cells may involve TGF-alpha and the EGF/TGF-alpha receptor autocrine loop.

    Topics: Androgens; Antibodies, Monoclonal; Cell Division; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation; Humans; Male; Prostatic Neoplasms; RNA, Messenger; Transforming Growth Factor alpha; Tumor Cells, Cultured

1993
Expression of nerve growth factor and nerve growth factor receptor genes in human tissues and in prostatic adenocarcinoma cell lines.
    Journal of neurochemistry, 1992, Volume: 59, Issue:4

    Nerve growth factor (NGF) mRNAs were detected and quantified in a variety of normal and neoplastic human tissues by northern blot hybridization. Human heart contained the highest NGF mRNA levels, whereas lower but comparable levels were found in the placenta, prostate, and kidney. All tissues examined coexpressed the low-affinity NGF receptor (LNGFR), whereas none of these tissues expressed the high-affinity NGF receptor encoded by the trk protooncogene. The widespread distribution of the LNGFR suggests that it plays a role in the regulation of normal cell growth. No overexpression of NGF or LNGFR mRNA was detected in neoplastic tissues, whereas LNGFR-like immunoreactivity was localized outside of tumor cells. Transforming growth factor-alpha and protooncogene c-fos expression in these tissues did not show a systematic correlation with NGF/LNGFR expression. Furthermore, regulation of the human NGF gene was studied in DU145 cells, a prostatic adenocarcinoma cell line that synthesizes significant NGF mRNA levels. Serum induced, whereas dexamethasone inhibited, NGF mRNA synthesis in these cells. Serum induction was preceded by a rapid and transient activation of the c-fos protooncogene.

    Topics: Adenocarcinoma; Binding, Competitive; Dexamethasone; Gene Expression; Humans; Kidney; Male; Nerve Growth Factors; Prostatic Hyperplasia; Prostatic Neoplasms; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-fos; Receptor, trkA; Receptors, Nerve Growth Factor; RNA, Messenger; Testis; Transforming Growth Factor alpha; Tumor Cells, Cultured

1992
[Analysis of anchorage independent growth of human prostate cancer cell line LNCap].
    Nihon Hinyokika Gakkai zasshi. The japanese journal of urology, 1992, Volume: 83, Issue:9

    LNCap, a human prostate cancer cell line, possess androgen dependent growth characteristics. We studied anchorage independent proliferation of LNCap cells using semi-solid agarose double layer culture. The cells formed colonies in the semi-solid medium supplemented with charcoal filtered steroid free fetal calf serum and maximal colony formation was obtained in the medium with 10% serum. The addition of several steroids (testosterone, dihydrotestosterone, ethinylestradiol) influenced the colony formation. Testosterone at the concentration of 10(-8) M to 10(-10) M stimulated colony formation with optiman of 10(-9) M. When LNCap cells were placed under the basal layer of the semi-solid culture as feeder cells, also stimulated was the colony formation of the LNCap cells cultured in upper layer of semi-solid medium. The addition of EGF, TGF alpha and TGF beta to the medium also stimulated the colony formation. The combined effect of EGF and TGF alpha was shown to be cooperative with testosterone. TGF beta, however, did not show such cooperative effect with testosterone on colony formation. The addition of the anti-body to EGF, TGF alpha or TGF beta to the medium decreased the colony formation of LNCap cells. These results suggest that LNCap cells excrete EGF, TGF alpha, TGF beta and/or similar substances and these factors autocrinely decorate the cell proliferation of LNCap human prostate cancer cells.

    Topics: Cell Division; Culture Media; Dihydrotestosterone; Epidermal Growth Factor; Ethinyl Estradiol; Humans; Male; Prostatic Neoplasms; Testosterone; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Cells, Cultured

1992
Density-dependent regulation of epidermal growth factor receptor expression in DU 145 human prostate cancer cells.
    The Prostate, 1991, Volume: 19, Issue:1

    Androgen-independent prostate cancer cells may rely on an autocrine loop for growth stimulation, and have been shown to express both the epidermal growth factor receptor (EGFR) and its stimulatory ligands. We have shown here that DU 145 prostate cancer cells have a decreased amount of EGFR in confluent cultures when compared to levels seen in subconfluent cultures. This down-regulation of EGFR numbers is not due to cell proliferation or nutrient depletion, but can be correlated only with whether cell-cell contact exists throughout the culture. This is reminiscent of the situation existing in some tumors whereby EGFR expression is higher in cells at the invading margins of the tumor.

    Topics: Antibodies, Monoclonal; Autoradiography; Cell Count; Densitometry; Down-Regulation; Electrophoresis, Polyacrylamide Gel; ErbB Receptors; Humans; Male; Precipitin Tests; Prostatic Neoplasms; Receptor, Insulin; Sulfur Radioisotopes; Time Factors; Transforming Growth Factor alpha; Tumor Cells, Cultured

1991
Autocrine regulation of DU145 human prostate cancer cell growth by epidermal growth factor-related polypeptides.
    The Prostate, 1991, Volume: 19, Issue:2

    The DU145 human prostate cancer cell line possesses epidermal growth factor (EGF) receptors and synthesizes both EGF and the related polypeptide transforming growth factor-alpha (TGF-alpha). A monoclonal antibody to the EGF receptor was used to determine whether these characteristics were indicative of a functional autocrine regulatory system. This antibody competed effectively with [125I]EGF for binding to DU145 cell binding sites over a 1 x 10(-11) to 1 x 10(-7) M concentration range, and did so with a capability similar to that of the two natural ligands. It inhibited growth of these cells in both 3% fetal bovine serum-supplemented and serum-free medium; in experiments with incubation times of 3-5 days there was a 45-50% reduction in cell number. Growth suppression by the EGF receptor blockade of cells plated at a density of 1.5 x 10(4) cells/ml/well was reversed competitively by the addition of EGF to the medium; 0.3 nM completely eliminated the inhibitory effect of a 1 x 10(-9) M antibody concentration. It is concluded that DU145 cell growth is regulated by an EGF-mediated autocrine loop.

    Topics: Antibodies, Monoclonal; Binding, Competitive; Cell Division; Cell Line; Epidermal Growth Factor; ErbB Receptors; Humans; Male; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured

1991
Autonomous growth of androgen-independent human prostatic carcinoma cells: role of transforming growth factor alpha.
    Cancer research, 1991, Jun-01, Volume: 51, Issue:11

    The androgen-independent prostatic carcinoma cell line PC3 is known to exhibit autonomous growth in vitro and in vivo. The purpose of the present study was to investigate the role of transforming growth factor alpha (TGF-alpha) and its receptor, the epidermal growth factor (EGF) receptor, in the regulation of PC3 cell proliferation. Results showed that PC3 cells secrete factors into conditioned medium that are mitogenic for the less aggressive prostatic carcinoma lines DU145 and LNCaP. Gel filtration chromatography of PC3-conditioned medium revealed a major peak of mitogenic activity at a molecular weight of 5,000 to 10,000 which was inhibited by the addition of antibody to TGF-alpha. The synthesis and secretion of TGF-alpha by PC3 cells were further demonstrated by immunoblotting and radioimmunoassay. Radioreceptor analysis showed a single class (Kd 5.3 nM) of EGF receptors on PC3 cells. The presence of Mr 170,000 EGF receptors on PC3 cells was further demonstrated by immunoprecipitation of metabolically labeled proteins. TGF-alpha was effective in stimulating the growth of low-density, but not high-density, PC3 cultures. In addition, the proliferation of PC3 cells under serum-free defined conditions was inhibited by antibodies to TGF-alpha and/or the EGF receptor. These data indicate that TGF-alpha/EGF receptor interactions are partially responsible for autonomous growth of the PC3 cell line and may explain one mechanism of escape from androgen-dependent growth in human prostatic carcinoma.

    Topics: Cell Division; Culture Media; ErbB Receptors; Humans; Male; Molecular Weight; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured

1991
Antitumor activity of a transforming growth factor alpha-Pseudomonas exotoxin fusion protein (TGF-alpha-PE40).
    Cancer research, 1991, Jun-01, Volume: 51, Issue:11

    TGF-alpha-PE40 is a chimeric protein composed of transforming growth factor alpha (TGF-alpha) linked to a modified Pseudomonas toxin from which the cell recognition domain has been deleted (PE40). TGF-alpha-PE40 has been shown to have cytotoxic effects on human cancer cell lines that express the epidermal growth factor (EGF) receptor on their surface, and when given i.p., it prolongs the survival of nude mice bearing i.p. tumors. Because several normal tissues, including liver, express EGF receptors on their surfaces, it has not been clear that this agent can be used systemically to treat EGF receptor-bearing tumors. In this study, we have delivered TGF-alpha-PE40 for 7 days by continuous infusion through a miniosmotic pump placed in the peritoneal cavity of nude immunodeficient mice. Two different human cancer cell lines that express EGF receptors on their surface were implanted s.c. One was A431, an epidermoid carcinoma; the other was DU-145, a prostate carcinoma. By using this mode of continuous i.p. delivery, we were able to achieve a constant serum level of TGF-alpha-PE40 that was nontoxic to the mice and yet delayed the growth of both tumors implanted s.c. and caused partial regression of one. We conclude that it is possible to deliver TGF-alpha-PE40 systemically and achieve a therapeutic serum level in mice without major toxicity. Although side effects may be expected, this study establishes that there is a therapeutic window for this agent in the therapy of cancers with high numbers of EGF receptors.

    Topics: Animals; Carcinoma, Squamous Cell; Drug Screening Assays, Antitumor; Drug Stability; Exotoxins; Female; Infusion Pumps; Male; Mice; Mice, Nude; Prostatic Neoplasms; Recombinant Fusion Proteins; Transforming Growth Factor alpha; Tumor Cells, Cultured

1991
Inhibition of prostatic tumor cell proliferation by suramin: alterations in TGF alpha-mediated autocrine growth regulation and cell cycle distribution.
    The Journal of urology, 1991, Volume: 146, Issue:1

    Suramin is a trypanocidal drug that has generated recent interest as an antineoplastic agent because of its ability to inhibit the binding of growth factors to their cell surface receptors. Our studies, and others, suggest that the androgen-independent human prostatic carcinoma cell lines PC3 and DU145 proliferate via autocrine growth mechanisms mediated by transforming growth factor alpha (TGFa) and its receptor, the epidermal growth factor (EGF) receptor. The present studies were designed to evaluate the ability of suramin to inhibit PC3 and DU145 proliferation by interfering with TGFa-mediated autocrine growth. Suramin induced a dose-dependent reduction of prostatic tumor cell proliferation which was reversed by removal of suramin from the culture medium. 3H-thymidine release studies showed that suramin had little direct cytotoxicity to either cell line. These findings suggest that the effects of suramin are mediated by cytostatic, rather than cytotoxic, mechanisms. Suramin also interfered with TGFa-mediated growth mechanisms. Specifically, suramin reduced the specific binding of TGFa to PC3 and DU145 cells. Additionally, the inhibitory effect of suramin on DU145 was reversed by cultivation of cells in the presence of excess TGFa. Further investigations revealed that suramin increased the percentage of cells in the S phase of the cell cycle for both cell lines. These studies show that the inhibitory effect of suramin on PC3 and DU145 cell growth is mediated, in part, by alteration of TGFa-mediated autocrine growth mechanisms and cell cycle kinetics.

    Topics: Antineoplastic Agents; Cell Cycle; Cell Division; Cell Line; Depression, Chemical; Dose-Response Relationship, Drug; Drug Screening Assays, Antitumor; ErbB Receptors; Humans; Male; Prostatic Neoplasms; Suramin; Transforming Growth Factor alpha; Tumor Cells, Cultured

1991
Production and response of a human prostatic cancer line to transforming growth factor-like molecules.
    British journal of cancer, 1990, Volume: 62, Issue:4

    Serum-free media conditioned by the androgen insensitive human prostate cancer cell line DU145 showed immunological transforming growth factor-alpha (TGF alpha) activity, as well as competing activity in epidermal growth factor (EGF) radioreceptor assays (RRA). Furthermore, there were factors in the conditioned media which inhibited and stimulated DNA synthesis by DU145 cells in a dose-dependent fashion. Fractionation of the concentrated conditioned media by reverse-phase high performance liquid chromatography revealed several peaks containing EGF-like competitive activity only one of which demonstrated TGF alpha activity. However, none of the peaks corresponded to immunoreactive EGF. Measurement of EGF receptors on DU145 cells by competition and saturation analysis revealed high levels of receptors (mean +/- s.d. = 2.5 +/- 1 x 10(5) surface receptors per cell) which were of high affinity (Kd +/- s.d. = 1.0 +/- 0.5 nmol l-1). Although DU145 cells express high levels of EGF receptors, DNA synthesis was only minimally affected by exogenous EGF and TGF alpha.

    Topics: Chromatography, High Pressure Liquid; DNA; Epidermal Growth Factor; ErbB Receptors; Humans; Male; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured

1990