Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Prostate ; 73(8): 801-12, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23192379

ABSTRACT

BACKGROUND: Neuroendocrine (NE) cells promote the progression of prostate cancer to a castration-resistant state through the production of paracrine growth factors. We have demonstrated this principle using in vitro and in vivo proliferative endpoints; however, the contributions of NE-derived pro-survival factors and anti-apoptosis to this phenomenon have not been thoroughly investigated. METHODS: Here, we utilized conditioned-medium (CM) from LNCaP cells, engineered to undergo NE differentiation, and examined its effects on PC3 and LNCaP cell survival. RESULTS: Statistically significant changes in clonogenic survival, Annexin V staining, PARP cleavage and trypan blue positivity of approximately twofold were observed in the presence of NE-derived CM relative to control-CM for both LNCaP and PC3 cells. These changes were partially abrogated by antagonists of the neuropeptides neurotensin, bombesin, and PTHrP. Selective inhibitors of IGF-1R, EGFR or Src caused significant and nearly complete blockade of prostate cancer cell survival due to NE secretions. Similar increases in cell survival were observed for LNCaP or PC3 cells treated with NE-derived medium in the presence of docetaxel. Increased phosphorylation of IGF-1R, following treatment with NE-derived medium, was accompanied by decreased protein tyrosine phosphatase, receptor type F (PTPRF) mRNA, and protein levels. Overexpression of PTPRF decreased cell survival, the amplitude and duration of IGF-1R phosphorylation, and enhanced PARP cleavage in the presence of NE-derived medium. CONCLUSIONS: These data support the hypothesis that NE-derived factors act upon prostate cancer cells to stimulate pro-survival signaling and describe a novel mechanism of cross-talk between NE-derived factors and IGF-1R, mediated in part by PTPRF.


Subject(s)
Neoplasms, Hormone-Dependent/metabolism , Neurosecretory Systems/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, IGF Type 1/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Blotting, Western , Cell Line, Tumor , Cell Survival/physiology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/genetics , Parathyroid Hormone-Related Protein/antagonists & inhibitors , Parathyroid Hormone-Related Protein/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 1/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Signal Transduction
2.
Cancer Res ; 67(17): 8316-24, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804747

ABSTRACT

2-Methoxyestradiol (2ME2) is an endogenous estradiol metabolite that inhibits microtubule polymerization, tumor growth, and angiogenesis. Because prostate cancer is often treated with radiotherapy, and 2ME2 has shown efficacy as a single agent against human prostate carcinoma, we evaluated 2ME2 as a potential radiosensitizer in prostate cancer models. A dose-dependent decrease in mitogen-activated protein kinase phosphorylation was observed in human PC3 prostate cancer cells treated with 2ME2 for 18 h. This decrease correlated with in vitro radiosensitization measured by clonogenic assays, and these effects were blocked by the expression of constitutively active MEK. Male nude mice with subcutaneous PC3 xenografts in the hind leg were treated with 2ME2 (75 mg/kg) p.o. for 5 days, and 2 Gy radiation fractions were delivered each day at 4 h after drug treatment. A statistically significant super-additive effect between radiation and 2ME2 was observed in this subcutaneous model, using analysis of within-animal slopes. A PC-3M orthotopic model was also used, with bioluminescence imaging as an end point. PC-3M cells stably expressing the luciferase gene were surgically implanted into the prostates of male nude mice. Mice were given oral doses of 2ME2 (75 mg/kg), with radiation fractions (3 Gy) delivered 4 h later. Mice were then imaged weekly for 4 to 5 weeks with a Xenogen system. A significant super-additive effect was also observed in the orthotopic model. These data show that 2ME2 is an effective radiosensitizing agent against human prostate cancer xenografts, and that the mechanism may involve a decrease in mitogen-activated protein kinase phosphorylation by 2ME2.


Subject(s)
Carcinoma/metabolism , Carcinoma/radiotherapy , Estradiol/analogs & derivatives , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , 2-Methoxyestradiol , Animals , Carcinoma/enzymology , Estradiol/pharmacology , Estradiol/therapeutic use , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Prostatic Neoplasms/enzymology , Subcutaneous Tissue , Transplantation, Heterotopic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Cancer Res ; 67(8): 3663-72, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440078

ABSTRACT

The neuroendocrine status of prostatic adenocarcinomas is considered a prognostic indicator for development of aggressive, androgen-independent disease. Neuroendocrine-like cells are thought to function by providing growth and survival signals to surrounding tumor cells, particularly following androgen ablation therapy. To test this hypothesis directly, LNCaP cells were engineered to inducibly express a constitutively activated form of the cyclic AMP-dependent protein kinase A catalytic subunit (caPKA), which was previously found upon transient transfection to be sufficient for acquisition of neuroendocrine-like characteristics and loss of mitotic activity. Clonal cells that inducibly expressed caPKA enhanced the growth of prostate tumor cells in anchorage-dependent and anchorage-independent in vitro assays as well as the growth of prostate tumor xenografts in vivo, with the greatest effects seen under conditions of androgen deprivation. These results suggest that neuroendocrine-like cells of prostatic tumors have the potential to enhance androgen-independent tumor growth in a paracrine manner, thereby contributing to progression of the disease.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Neoplasms, Hormone-Dependent/pathology , Neuroendocrine Tumors/pathology , Prostatic Neoplasms/pathology , Animals , Catalytic Domain , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mitosis/physiology , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/genetics , Neuroendocrine Tumors/enzymology , Oligopeptides , Peptides/genetics , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics
4.
Mol Biol Cell ; 13(7): 2233-44, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12134064

ABSTRACT

Ionizing radiation (1-5 Gy) activates the epidermal growth factor receptor (EGFR), a major effector of the p42/44 mitogen-activated protein kinase (MAPK) pathway. MAPK and its downstream effector, p90 ribosomal S6 kinase (p90RSK), phosphorylate transcription factors involved in cell proliferation. To establish the role of the EGFR/MAPK pathway in radiation-induced transcription factor activation, MDA-MB-231 human breast carcinoma cells were examined using specific inhibitors of signaling pathways. Gel-shift analysis revealed three different profile groups: 1) transcription factors that responded to both radiation (2 Gy) and epidermal growth factor (EGF) (CREB, Egr, Ets, and Stat3); 2) factors that responded to radiation, but not EGF (C/EBP and Stat1); and 3) those that did not respond significantly to either radiation or EGF (AP-1 and Myc). Within groups 1 and 2, a two- to fivefold maximum stimulation of binding activity was observed at 30-60 min after irradiation. Interestingly, only transcription factors that responded to EGF had radiation responses significantly inhibited by the EGFR tyrosine kinase inhibitor, AG1478; these responses were also abrogated by farnesyltransferase inhibitor (FTI) or PD98059, inhibitors of Ras and MEK1/2, respectively. Moreover, radiation-induced increases in CREB and p90RSK phosphorylation and activation of Stat3 and Egr-1 reporter constructs by radiation were all abolished by AG1478. These data demonstrate a distinct radiation response profile at the transcriptional level that is dependent on enhanced EGFR/Ras/MAPK signaling.


Subject(s)
Breast Neoplasms/metabolism , ErbB Receptors/metabolism , MAP Kinase Signaling System/radiation effects , Mitogen-Activated Protein Kinases/metabolism , Radiation, Ionizing , Transcription Factors/metabolism , Binding Sites , Cell Fractionation , Enzyme Inhibitors/metabolism , Epidermal Growth Factor/metabolism , Female , Genes, Reporter , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nuclear Proteins/metabolism , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Time Factors , Transcription Factors/antagonists & inhibitors , Transcription, Genetic/radiation effects , Tumor Cells, Cultured , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism
5.
Oncogene ; 22(36): 5545-53, 2003 Aug 28.
Article in English | MEDLINE | ID: mdl-12944901

ABSTRACT

The constitutively active, truncated epidermal growth factor receptor EGFRvIII lacks the ability of EGF binding due to a deletion of the NH(2)-terminal domain. EGFRvIII confers increased tumorigenicity, is coexpressed with EGFR wild type (wt) in human carcinoma and malignant glioma cells when grown as xenografts, but is not expressed in vitro. The effects of EGFRvIII expression on cellular radiation responses were studied in Chinese hamster ovary (CHO) cells transfected with plasmids expressing EGFRvIII (CHO.EGFRvIII) or EGFRwt (CHO.EGFRwt). CHO cells expressing similar levels of either receptor were employed to define their roles in response to EGF and ionizing radiation. EGF activated EGFRwt with no effect on EGFRvIII. In contrast, a single radiation exposure of 2 Gy resulted in a 2.8- and 4.3-fold increase in Tyr phosphorylation of EGFRwt and EGFRvIII, respectively. Downstream consequences of this radiation-induced activation were examined by inhibiting EGFRwt and EGFRvIII with AG1478 (kinase inhibitor). The radiation-induced 8.5-fold activation of the pro-proliferative mitogen-activated protein kinase and the 3.2-fold stimulation of the antiapoptotic AKT/phosphatidylinositol-3-kinase pathways by EGFRvIII far exceeded that in CHO.EGFR wt cells. Thus, based on colony formation and apoptosis assays, EGFRvIII expression conferred a stronger cytoprotective response to radiation than EGFRwt, resulting in relative radioresistance. Therefore, disabling EGFRvIII in addition to EGFRwt needs to be considered in any therapeutic approach aimed at targeting EGFR for tumor cell radiosensitization.


Subject(s)
Cytoprotection , ErbB Receptors/physiology , Protein Serine-Threonine Kinases , Radiation Tolerance , Animals , Apoptosis/radiation effects , CHO Cells , Cricetinae , Enzyme Activation , Epidermal Growth Factor/pharmacology , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Quinazolines , Tyrphostins/pharmacology
6.
Crit Rev Eukaryot Gene Expr ; 14(4): 287-300, 2004.
Article in English | MEDLINE | ID: mdl-15663358

ABSTRACT

Neuroendocrine (NE) cells are found in prostate tumors, and their incidence is considered a promising prognostic indicator for the development of androgen-independent disease. NE cells are derived from non-NE prostate cancer cells and secrete factors that can act in a paracrine manner to stimulate the survival, growth, motility, and metastatic potential of prostatic carcinoma cells. Factors such as IL-6, epinephrine, and forskolin induce NE differentiation in prostate cancer cells; the mechanisms involve increases in intracellular cAMP, protein kinase A (PKA) activation and reduced intracellular calcium levels. Transcription factors implicated in the acquisition of NE characteristics by prostate cancer cells include STAT3, CREB, EGR1, c-fos, and NF-kappaB. Expression of Chromogranin A, neuron-specific enolase, bcl-2, and the androgen receptor are modulated during NE differentiation and serve as molecular markers for NE cells. Most importantly, NE cells secrete neuropeptides, such as bombesin, neurotensin, PTHrP, serotonin, and calcitonin, which trigger growth and survival responses in androgen-independent prostate cancer cells. Prostate cancer cell receptors that play a role in these processes include the gastrin-releasing peptide (GRP) receptor, neurotensin receptors, and the epidermal growth-factor receptor (EGFR). Signal-transduction molecules activated by these neuropeptides include Src, focal adhesion kinase (FAK), ERK, and PI3K/Akt, with subsequent activation of Elk-1, NF-kappaB, and c-myc transcription factors. A multitude of genes are then expressed by prostate cancer cells, which are involved in proliferation, anti-apoptosis, migration, metastasis, and angiogenesis. Targeting of these pathways at multiple levels can be exploited to inhibit the process by which NE cells contribute to the progression of androgen-independent, treatment-refractory prostate cancer.


Subject(s)
Neurosecretory Systems/pathology , Prostatic Neoplasms/pathology , Disease Progression , Humans , Male , Prostatic Neoplasms/genetics
7.
Radiother Oncol ; 62(1): 61-7, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11830313

ABSTRACT

BACKGROUND AND PURPOSE: We have tested the camptothecin analogs, RFS-2000 or CPT-11, in combination with both etoposide and ionizing radiation in vitro to examine the radiation enhancing potential of topoisomerase I plus topoisomerase II (Topo I+Topo II) inhibition in human cancer cells. MATERIALS AND METHODS: H460 human lung carcinoma cells were plated and treated with 10nM RFS-2000 or 4.5microM CPT-11 for 4h. Cells were then irradiated with various doses and treated with 1microM etoposide for 1.5h. Cell survival and sublethal damage recovery (SLDR) were determined by clonogenic assay. 7-aminoactinomycin D (7-AAD) staining and flow cytometry were used to analyze cell viability/apoptosis after combined treatment of drugs with radiation. RESULTS: Survival experiments showed radiation dose enhancement ratios (DER) of 1.26, 1.34, and 1.63 for RFS-2000, etoposide, and RFS-2000 plus etoposide, respectively; the corresponding DER values were 1.30, 1.39, and 1.65 for CPT-11, etoposide, and CPT-11 plus etoposide. The analysis of cell viability/apoptosis using 7-AAD staining and flow cytometry showed an additive effect. Greater inhibition of SLDR was observed with RFS-2000 plus etoposide than with either agent separately, but CPT-11 plus etoposide showed a more modest effect upon SLDR. CONCLUSIONS: These data show that the combination of Topo I inhibitors, RFS-2000 or CPT-11 plus Topo II inhibitor etoposide, is a more effective radiation enhancer than either agent alone in human lung cancer cells. The mechanism of radiation enhancement may involve inhibition of SLDR with RFS-2000 plus etoposide, but other mechanisms may be involved in the combined treatment including CPT-11.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Etoposide/pharmacology , Radiation-Sensitizing Agents/pharmacology , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Cell Survival/drug effects , Colony-Forming Units Assay , Combined Modality Therapy , Dose-Response Relationship, Drug , Drug Combinations , Enzyme Inhibitors/pharmacology , Humans , Irinotecan , Lung Neoplasms , Tumor Cells, Cultured
8.
Cancer Res ; 71(21): 6817-26, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21903767

ABSTRACT

Radiotherapy combined with androgen depletion is generally successful for treating locally advanced prostate cancer. However, radioresistance that contributes to recurrence remains a major therapeutic problem in many patients. In this study, we define the high-affinity neurotensin receptor 1 (NTR1) as a tractable new molecular target to radiosensitize prostate cancers. The selective NTR1 antagonist SR48692 sensitized prostate cancer cells in a dose- and time-dependent manner, increasing apoptotic cell death and decreasing clonogenic survival. The observed cancer selectivity for combinations of SR48692 and radiation reflected differential expression of NTR1, which is highly expressed in prostate cancer cells but not in normal prostate epithelial cells. Radiosensitization was not affected by androgen dependence or androgen receptor expression status. NTR1 inhibition in cancer cell-attenuated epidermal growth factor receptor activation and downstream signaling, whether induced by neurotensin or ionizing radiation, establish a molecular mechanism for sensitization. Most notably, SR48692 efficiently radiosensitized PC-3M orthotopic human tumor xenografts in mice, and significantly reduced tumor burden. Taken together, our findings offer preclinical proof of concept for targeting the NTR1 receptor as a strategy to improve efficacy and outcomes of prostate cancer treatments using radiotherapy.


Subject(s)
Adenocarcinoma/radiotherapy , Neoplasm Proteins/antagonists & inhibitors , Prostatic Neoplasms/radiotherapy , Pyrazoles/therapeutic use , Quinolines/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Receptors, Neurotensin/antagonists & inhibitors , Adenocarcinoma/pathology , Androgens , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Mice, Nude , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/radiotherapy , Phosphorylation/drug effects , Phosphorylation/radiation effects , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/radiation effects , Pyrazoles/pharmacology , Quinolines/pharmacology , Radiation Tolerance/drug effects , Radiation Tolerance/physiology , Radiation-Sensitizing Agents/pharmacology , Receptors, Androgen/analysis , Receptors, Neurotensin/physiology , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
9.
J Biol Chem ; 278(32): 29394-9, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12734192

ABSTRACT

Selective inhibition of the epidermal growth factor receptor or mitogen-activated protein kinase (MAPK) results in radiosensitization of cancer cells. One potential mechanism involves cAMP-responsive element-binding protein, which is activated by radiation via the epidermal growth factor receptor/MAPK pathway and which regulates synthesis of proliferating cell nuclear antigen (PCNA), a protein involved in repair of ionizing radiation-induced DNA damage. To test for a role of CREB in cellular radiosensitivity, CHO cells were transfected with plasmids expressing dominant-negative CREB mutants (CR133 or KCREB), and various end-points were measured 48 h later. Basal levels of PCNA-CAT reporter construct activity were reduced by 60 and 40% following expression of CR133 and KCREB, respectively; similar decreases were observed in PCNA protein levels. Pulsed-field gel electrophoresis measurements showed that CR133 inhibited the repair of radiation-induced DNA double-strand breaks, and this effect was reversed by over-expression of PCNA; dominant-negative CREB also significantly inhibited split-dose recovery. Clonogenic assays were used to determine surviving fraction; the dose enhancement ratios for dominant-negative CREB-expressing cells compared with control (vector alone) were 1.5 and 1.3 for CR133 and KCREB, respectively. Importantly, co-transfection of mutant CREB and a construct constitutively expressing PCNA protein restored radiosensitivity of CHO cells back to wild-type levels. Moreover, cells expressing either CREB mutant showed no significant cell cycle redistribution. These data demonstrate that genetic disruption of CREB results in radiosensitization, and that this effect can be explained by a mechanism involving decreased PCNA expression and inhibition of DNA repair.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/physiology , Genes, Dominant , Proliferating Cell Nuclear Antigen/metabolism , Animals , Blotting, Western , CHO Cells , Cell Cycle , Cell Division , Cricetinae , DNA Damage , DNA Repair , Dose-Response Relationship, Radiation , Flow Cytometry , Genes, Reporter , Genetic Vectors , Kinetics , MAP Kinase Signaling System , Mutation , Protein Binding , Time Factors , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL