Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 30(6): 1937-46, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20651337

ABSTRACT

BACKGROUND: Protein phosphorylation is one of the essential steps in cell signaling, and aberrant phosphorylation is a common event in human cancer. The expression of receptor type protein tyrosine phosphatase gamma (PTPRG) in normal breast is found to be approximately 50-60% higher than that of breast tumor tissue. Overexpression of PTPRG inhibits anchorage-independent growth and proliferation of breast cancer cells. To understand the tumor suppression characteristics of PTPRG, we studied its tumor suppressive function in an athymic mouse model and evaluated factors that can potentially regulate its expression in breast cancer cells. MATERIALS AND METHODS: To investigate the function of PTPRG in vivo, athymic nude mice were implanted with MCF-7 cells overexpressing PTPRG. For in vitro study, protein levels of cell cycle regulators, cell cycle re-entry, and the phosphorylation levels of extracellular signal-regulated protein kinases 1/2 (ERK1/2) were examined. In addition, methylation assays were conducted to investigate the epigenetic modification on the promoter of PTPRG. RESULTS: Athymic nude mice bearing MCF-7 cells overexpressing PTPRG showed a reduction in tumor burden in comparison to animals implanted with MCF-7 cells transfected with vector alone. When these two cell lines were studied in an in vitro system, elevated mRNA and protein levels of cell cycle regulators, p21(cip) and p27(kip) were detected in MCF-7 cells overexpressing PTPRG compared to cells transfected with vector alone. Similarly, overexpression of PTPRG also delayed the re-entry of breast cancer cells into the cell cycle after serum starvation, and reduced the phosphorylation levels ERK1/2 in MCF-7 cells. In addition, methylation assays in PTPRG promoter in breast cancer cell lines (including SK-Br-3) revealed an aberrant methylation pattern. When SK-Br-3 and MCF-7 cells were treated with deoxy-5-azacytidine (DAC) and trichostatin A (TSA), these compounds reactivated the expression of PTPRG, suggesting an epigenetic control on its expression. CONCLUSION: Our results indicated that PTPRG inhibited breast tumor formation in vivo; PTPRG may up-regulate p21(cip) and p27(kip) proteins through the ERK1/2 pathway. This study also showed methylation-mediated silencing of PTPRG in breast cancer cell lines. These data indicate that PTPRG exhibits the characteristics of a breast tumor suppressor.


Subject(s)
Breast Neoplasms/prevention & control , Receptor-Like Protein Tyrosine Phosphatases, Class 5/physiology , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Breast Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , CpG Islands , Cyclin-Dependent Kinase Inhibitor p21/analysis , Cyclin-Dependent Kinase Inhibitor p27 , DNA Methylation , Decitabine , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Hydroxamic Acids/pharmacology , Intracellular Signaling Peptides and Proteins/analysis , Mice , Phosphorylation
2.
Int J Mol Med ; 26(1): 113-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20514430

ABSTRACT

Induction of apoptosis is one of the mechanisms of chemotherapeutic agents against breast cancer. In addition, recent studies have shown that diets containing polyphenolic components possess anticancer activities either in vitro or in vivo by inhibiting cell proliferation and inducing apoptosis. The aim of our study was to explore the effects of (-)-gossypol-enriched cottonseed oil [(-)-GPCSO], a polyphenolic compound, on the proliferation of the breast cancer cell line MCF-7 as well as primary cultured human breast cancer epithelial cells (PCHBCEC). We investigated whether the mechanism of the effects of (-)-GPCSO was mediated via the induction of cell apoptosis and the regulation of Bcl-2 gene expression at both the mRNA and protein levels. Our results showed that (-)-GPCSO inhibited the proliferation of MCF-7 and PCHBCEC in a dose-dependent manner. (-)-GPCSO (0.1 and 0.2%) induced DNA fragmentation in both MCF-7 cells and PCHBCEC. (-)-GPCSO suppressed the expression of Bcl-2 at both the mRNA and protein levels in MCF-7 cells and PCHBCEC in a dose-dependent fashion. Our results suggest that the growth inhibitory effect of (-)-GPCSO on MCF-7 and PCHBCEC is due, at least partially, to the induction of cell apoptosis, which is mediated by down-regulation of Bcl-2 expression at both the mRNA and protein levels. It might be possible for (-)-GPCSO to be developed as a novel chemotherapeutic agent for breast cancer patients.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Cottonseed Oil/pharmacology , Gossypol/pharmacology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Cottonseed Oil/chemistry , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Electrophoresis, Agar Gel , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gossypol/chemistry , Humans , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction
3.
Anticancer Res ; 29(8): 3195-205, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19661335

ABSTRACT

BACKGROUND: It is suggested that the phosphatidylinositol 3-kinase (PI3K/Akt) pathway may lead to tamoxifen (Tam) resistance in the estrogen receptor-alpha (ER-alpha)-positive breast cancer cell line, MCF-7. Our previous results demonstrated that keratinocyte growth factor (KGF) down-regulates ER-alpha expression and increases Tam resistance in MCF-7 cells. Therefore, we hypothesized that a possible mechanism for developing Tam resistance could be the regulation of ER-alpha and Bcl-2 family proteins through modulation of Akt activity. MATERIALS AND METHODS: MCF-7 cells were treated with KGF, LY294002 (LY), a PI3 kinase inhibitor, 4-OH-Tam, KGF with LY, KGF with LY and 4-OH-Tam, or vehicles as control for 24 hours. Total RNA was extracted from MCF-7 cells and real-time PCR was employed to identify the ER-alpha expression in response to KGF. To determine that the resistance to 4-OH-Tam-inducing cell killing after the KGF treatment was due to the inactivation of the apoptotic pathway, low molecular weight DNA was isolated from cells of different treatments and inter-nucleosomal DNA fragmentation was investigated. The phosphorylation of signaling intermediates Akt, Bad, the activation of caspase-9, and the expression of ER-alpha, Bcl-2, Bcl-xL, and Bax were evaluated by immunoblot analysis for the study of KGF signaling effects. To determine the involvement of PI3K/Akt pathway in the survival effect of KGF, the growth rate of MCF-7 cell was measured by non-radioactive cell proliferation assay after treatments of KGF, LY, 4-OH-Tam, KGF with LY, KGF with LY and 4-OH-Tam, or vehicles as control for 3 days. The results of real-time PCR and cell proliferation assay were analyzed by Student's t-test and p-values of less than 0.05 were considered statistically significant. RESULTS: Our results showed that in MCF-7 cells KGF increased Akt phosphorylation and induced ER-alpha mRNA expression which could be blocked by a PI3K/Akt pathway inhibitor, LY. KGF treatment also induced apoptosis based on the observation of the suppression of DNA fragmentation, variable increase in the expression of the Bcl-2 and Bcl-xL proteins and the decrease of the active form of caspase-9 protein, whereas LY blocked the anti-apoptotic effects of KGF. In the cell proliferation assay, KGF maintained MCF-7 cell survival in the presence of 4-OH-Tam which could be blocked by LY. CONCLUSION: We confirmed the regulation of ER-alpha by KGF in human breast cancer cells at both mRNA and protein levels. We further demonstrated that KGF may play an inhibitory role in the induction of breast cancer cell apoptosis, conferring resistance against anticancer drugs on breast cancer cells.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Fibroblast Growth Factor 7/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Blotting, Western , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Estrogen Receptor alpha/genetics , Female , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tumor Cells, Cultured
4.
Anticancer Res ; 27(1A): 107-16, 2007.
Article in English | MEDLINE | ID: mdl-17352222

ABSTRACT

BACKGROUND: Multidrug resistance (MDR) is a major impediment to successful cancer chemotherapy. P-glycoprotein (P-gp), the product of the multidrug resistance 1 (MDR1) gene, acts as an efflux pump and prevents sufficient intracellular accumulation of several anticancer agents, thus, playing a major role in MDR. Tamoxifen (Tam), ICI 182 780 (ICI) and Adriamycin (Adr) alone or with (-)-gossypol-enriched cottonseed oil [(-)-GPCSO] possible effects on cell growth inhibition and regulation of MDR1, mRNA and P-gp expression were examined in both an MDR human breast cancer cell line, MCF-7/Adr cells, and primary cultured human breast cancer epithelial cells (PCHBCEC). MATERIALS AND METHODS: Cells were treated with 0.05% of (-)-GPCSO either in the absence or presence of either 0.1 microM Tam, ICI or Adr for 24 h. RESULTS: Using the non-radioactive cell proliferation MTS assay, none of these chemotherapeutic agents alone inhibited MCF-7/Adr cell and PCHBCEC proliferation; meanwhile, the combination of 0.1 microM Tam, ICI or Adr with 0.05% (-)-GPCSO significantly reduced MCF-7/Adr cell growth by approximately 34%, 32% and 23%, respectively, of that of the vehicle-treated cells. For PCHBCEC, the combination of 0.05% (-)-GPCSO with 0.1 microM of Tam, ICI and Adr reduced cell growth to about 94%, 90%, and 71% respectively, of the vehicle treated PCHBCEC. Furthermore, (-)-GPCSO inhibited MDR1/P-gp expression in both MCF- 7/Adr and PCHBCEC in a dose-dependent manner. Our results provide insight into the MDR-reversing potential of (-)-GPCSO in human breast cancer cells resistant to current chemotherapeutic agents.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cottonseed Oil/pharmacology , Genes, MDR/drug effects , Gossypol/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cottonseed Oil/administration & dosage , Cottonseed Oil/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Synergism , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estradiol/pharmacology , Fulvestrant , Gene Expression/drug effects , Gossypol/administration & dosage , Gossypol/chemistry , Humans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tamoxifen/administration & dosage , Tamoxifen/pharmacology , Tumor Cells, Cultured
5.
Anticancer Res ; 26(5A): 3613-20, 2006.
Article in English | MEDLINE | ID: mdl-17094491

ABSTRACT

BACKGROUND: Gossypol, a natural polyphenolic compound present in cottonseeds, possesses antiproliferative and pro-apoptotic effects in in vivo and in vitro models. There are two enantiomers, (+)-gossypol and (-)-gossypol, the latter being a more potent inhibitor of cancer cell growth. Here, the effect of bovine serum albumin (BSA) and dextran-coated charcoal-treated fetal bovine serum (DCC-FBS)-containing medium on the ability of (-)-gossypol to inhibit the growth of human prostate cancer cells was studied. MATERIALS AND METHODS: BSA- and DCC-FBS-supplemented medium were used to examine the influence of serum proteins on the antiproliferative effects of (-)-gossypol in DU-145 cells, a human prostate cancer cell line. The viability of the DU-145 cells was determined by CellTiter 96 Aqueous assay. The expressions of mRNA and protein for the cell cycle regulators, cyclin-D1, Rb, CDK, p21 and TGF-beta, were determined by RT-PCR and Western blot analyses, respectively. RESULTS: (-)-Gossypol caused growth suppression of the DU-145 cells. In comparison with BSA-supplemented medium, DCC-FBS blocked the antiproliferative effects of (-)-gossypol at 1 and 2.5 microM, but not at 5 microM. Furthermore, (-)-gossypol treatment down-regulated cyclin-D1, Rb, CDK4 and CDK6, and up-regulated p21 and TGF-beta1 at the mRNA and/or protein levels. CONCLUSION: The data suggested that (-)-gossypol-suppressed prostate cancer cell growth may be influenced through cell cycle regulators, which may lead to better prognosis. We further speculate that (-)-gossypol might serve as a chemotherapeutic agent for human prostate cancer patients.


Subject(s)
Cell Survival/drug effects , Gossypol/pharmacology , Prostatic Neoplasms/pathology , Serum Albumin, Bovine/pharmacology , Serum/metabolism , Animals , Blotting, Western , Cattle , Charcoal/chemistry , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dextrans/chemistry , Humans , Male , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serum/chemistry , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Cells, Cultured
6.
J Steroid Biochem Mol Biol ; 100(1-3): 24-33, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16854582

ABSTRACT

In normal prostate, keratinocyte growth factor (KGF), also known as fibroblast growth factor-7 (FGF-7) serves as a paracrine growth factor synthesized in stromal cells that acts on epithelial cells through its receptor, KGFR. KGF and KGFR were found in human cancer epithelial cells as well as stromal cells. Since KGF expressed in epithelial cells of benign prostatic hyperplasia (BPH) and in prostate cancer, it has been suggested that KGF might act as an autocrine factor in BPH and prostate cancer. To investigate the roles of KGF in cancerous stroma, primary cultured human prostate cancer stromal cells (PCSCs) were isolated and evaluated. These PCSCs possessed estrogen receptors and KGFR, but not androgen receptor as determined by RT-PCR and Western blot, respectively. KGF exhibited mitogenic and anti-apoptotic effects that correlated with induction of cyclin-D1, Bcl-2, Bcl-xL and phospho-Akt expression in PCSCs, where treatment with KGF antiserum abolished cell proliferation and anti-apoptotic protein expression. PCSCs exposed to KGF for various time periods resulted in phosphorylation of Akt and subsequent up-regulation of Bcl-2. KGF modulated dynamic protein expression indicated that KGF triggered cell cycle machinery and then activated anti-apoptotic actions in PCSCs. Cell proliferation analysis indicated that tamoxifen or ICI 182,780 reduced cell viability in a dose-dependent manner; however, KGF prevented this inhibition, which further demonstrated KGF triggered anti-apoptotic machinery through activating Bcl-2 and phospho-Akt expression. In summary, KGF has an autocrine effect and serves as a survival factor in primary cultured human prostate cancer stromal cells.


Subject(s)
Fibroblast Growth Factor 7/pharmacology , Prostatic Neoplasms/pathology , Stromal Cells/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , Male , Receptors, Estrogen/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Stromal Cells/cytology
7.
Anticancer Res ; 26(3A): 1773-84, 2006.
Article in English | MEDLINE | ID: mdl-16827106

ABSTRACT

BACKGROUND: Both estrogen receptor-alpha (ER-alpha) and progesterone receptor (PR) are good prognostic factors and indicators of benefit from endocrine therapy in breast cancer patients. The relationship of the ER-alpha and PR status and the difference in clinical benefit from endocrine therapy in breast cancer is currently unclear. It has been suggested that keratinocyte growth factors (KGFs) are important regulatory factors in breast cancer. Our laboratory has demonstrated that KGF can act as an estromedin for the stimulation of breast cancer cell growth. Also, KGF stimulates aromatase activity in primary cultured human breast cells. This enzyme is a key to the conversion of androgens to estrogens. In the present study, ER-alpha, two estrogen-regulated genes, PR and PTPgamma, KGF and their relationship with endocrine resistance in human breast cancer cells were investigated. MATERIALS AND METHODS: MCF-7 cells were treated with KGF (1, 5, 10, 20 ng/ml), KGF-13 (0.1, 1, 10 microM) or vehicles as control for 24 hours. KGF-13 is a potential receptor-binding pentapeptide constructed using the KGF peptide residues 101-105 (RTVAV) as a template, located within the beta 4--beta 5 loop. Total RNA were isolated and real-time PCR was employed to identify ER-alpha, PR and PTPgamma gene expressions in response to KGF and KGF-13. Western blot analysis was used to verify the levels of ER-alpha and PR protein, whereas immunohistochemical staining was used to detect PTPgamma expression in MCF-7 cells. To determine the response of MCF-7 cells to endocrine therapy, MCF-7 was treated with either 20 ng/ml KGF or 10 microM KGF-13 combined with 1, 3 and 5 microM of 4-hydroxytamoxifen (4OH-Tam). A non-radioactive cell proliferation assay was applied to determine the growth rate of MCF-7 cells. The results of real-time PCR and the cell proliferation assay were analyzed by Student's t-test and p-values of less than 0.05 were considered statistically significant. RESULTS: Our data showed that KGF significantly suppressed ER-alpha, PR and PTPgamma expression in MCF-7 cells. KGF suppressed ER-alpha, PR and PTPgamma mRNA to a maximal inhibition at 20 ng/ml by 88%, 57% and 61%, respectively. Western blot analysis and immunohistochemical staining confirmed the down-regulation of ER-alpha, PR and PTPgamma by KGF in protein levels. Ten microM KGF-13 also decreased ER-alpha expression. Ten microM KGF-13 significantly decreased ER-alpha, PR and PTPgamma mRNA expressions by 51%, 57% and 67%, respectively. These KGF-13-mediated mRNA down-regulations were also observed in protein levels. In a cell proliferation assay, 4OH-Tam (3, 5 microM) induced MCF-7 cell death. KGF and KGF-13 alone did not stimulate MCF-7 cell growth. KGF significantly disrupted 4OH-Tam cell killing effects by 1.2- and 1.3-fold at 4OH-Tam concentrations of 3 microM and 5 microM, respectively. KGF-13 significantly disrupted 4OH-Tam cell killing effects by 1.2- and 1.7-fold at 4OH-Tam concentrations of 3 microM and 5 microM, respectively. CONCLUSION: Our results suggested that not only ER-alpha and PR but also PTPgamma could be potential bio-makers for growth factor-induced endocrine resistant in breast cancer. KGF might increase the endocrine resistance via decreasing ER-alpha, PR and PTPgamma as well. Moreover, the functional analysis of KGF-13 implied possible applications of using short receptor-binding peptides derived from intact KGF as breast cancer therapeutic agents. Thus, our experimental data provided evidence of KGF-induced anti-hormone resistance in human breast cancer and suggested novel strategies for breast cancer via interference with KGF signaling.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Fibroblast Growth Factor 7/metabolism , Tamoxifen/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation , Drug Resistance, Neoplasm , Estrogen Receptor alpha/biosynthesis , Humans , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Protein Tyrosine Phosphatases/biosynthesis , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Receptors, Progesterone/biosynthesis , Signal Transduction , Tamoxifen/analogs & derivatives
8.
Anticancer Res ; 26(3A): 1925-33, 2006.
Article in English | MEDLINE | ID: mdl-16827126

ABSTRACT

BACKGROUND: Gossypol, a natural compound present in cottonseeds, displays antiproliferative and pro-apoptotic effects against various cancer cells. The (-)-gossypol enantiomer is a more potent inhibitor of cancer cell growth. Here, the molecular mechanisms of apoptosis induced by (-)-gossypol were studied in human prostate cancer cells. MATERIALS AND METHODS: After the prostate cancer cell DU-145 had been treated with (-)-gossypol, the trypan blue exclusion assay and DNA fragment end-labeling assay were used to stain the dead cells and to detect DNA laddering, respectively. The effects of (-)-gossypol on the expression of apoptotic-regulated gene markers in both death receptor- and mitochondria-mediated apoptotic pathways, such as the Bcl-2 family and caspase, etc., were detected by RT-PCR and Western blot analysis. To further investigate the apoptotic pathways induced by (-)-gossypol, different caspase inhibitors were used to block caspase activities and cell viability was detected by the CellTiter 96 AQueous assay in DU-145 cells. RESULTS: At a 5-10 microM dose-level, (-)-gossypol significantly enhanced apoptosis measured by DNA fragmentation. (-)-Gossypol caused apoptosis in DU-145 cells through the down-regulation of Bcl-2 and Bcl-xL and the up-regulation of Bax at the mRNA and protein levels. (-)-Gossypol also activated caspases-3, -8 and -9 and increased PARP [poly (ADP-ribose) polymerase] cleavage. Furthermore, (-)-gossypol-induced apoptosis might be due to an increase in CAD (caspase-activated deoxyribonuclease) proteins and a decrease in ICAD (inhibitor of CAD) proteins. By using caspase inhibitors, (-)-gossypol caused apoptosis via the caspase-dependent pathways. CONCLUSION: Our results indicated that the apoptotic processes caused by (-)-gossypol are mediated by the regulation of the Bcl-2 and caspase families in human prostate cancer cells. Our data also suggested that (-)-gossypol may have chemotheraputic benefits for prostate cancer patients.


Subject(s)
Apoptosis/drug effects , Gossypol/pharmacology , Prostatic Neoplasms/drug therapy , Apoptosis/physiology , Caspases/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Male , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/biosynthesis
9.
Anticancer Res ; 26(2A): 889-98, 2006.
Article in English | MEDLINE | ID: mdl-16619484

ABSTRACT

BACKGROUND: Conjugated linoleic acid (CLA), a naturally occurring linoleic acid isomer found in ruminant-produced foods, has the potential to serve as an effective chemopreventive nutriceutical factor for breast cancer prevention based upon previous published studies. There are several CLA isomers in ruminant-produced food products, among which t10,c12-CLA and c9,t11-CLA are more potent. Expression of cyclooxygenase 2 (COX-2) in mammary tumors has been correlated with poor prognosis. Prostaglandin E2 (PGE2) is a major COX-2 product in various cancers and, as in humans, PGE2 concentrations in canine tumor tissues were frequently elevated. Moreover, a PGE2 receptor subtype, EP2, is highly expressed in mammary tumors. Thus, various studies have implicated the important role of PGE2 and EP2 in COX-2-regulated tumor development. MATERIALS AND METHODS: Mammary tumor and normal mammary tissues were both collected from a female dog with mammary tumor. Both malignant and normal mammary tissues were subjected to isolation of epithelial and stromal cells. The effects of t10,c12-CLA and c9,t11-CLA on proliferation, as well as COX-2 and EP2 protein expression in canine mammary normal and cancerous cells, were detected by CellTiter 96 AQueous assay and Western blot assay, respectively. RESULTS: Both t10,c12-CLA and c9,t11-CLA not only suppressed malignant mammary cell growth, but also exerted inhibitory effects on tumor-associated non-malignant mammary cells. Similarly, both t10,c12-CLA and c9,t11-CLA suppressed EP2 protein expression in both normal and malignant mammary cells. t10,c12-CLA was more effective in decreasing COX-2 protein expression in malignant mammary cells, while, in contrast, c9,t11-CLA down-regulated COX-2 protein expression in both normal and malignant mammary cells. CONCLUSION: The results indicate that the dietary component CLA regulates COX-2 and EP2 protein expression in both malignant mammary cells and cells from the tumor-associated stromal compartment. In turn, this may suppress PGE2 signaling, leading to better prognosis. We further speculate that the knowledge obtained from canine studies may also be beneficial to study human breast cancer.


Subject(s)
Dinoprostone/metabolism , Dog Diseases/drug therapy , Linoleic Acids, Conjugated/pharmacology , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/metabolism , Animals , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2 Inhibitors/pharmacology , Dogs , Female , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/biosynthesis , Receptors, Prostaglandin E, EP2 Subtype , Signal Transduction/drug effects , Tumor Cells, Cultured
10.
Anticancer Res ; 26(1A): 27-34, 2006.
Article in English | MEDLINE | ID: mdl-16475675

ABSTRACT

BACKGROUND: Conjugated linoleic acid (CLA), a naturally occurring compound found in ruminants products, has been shown to possess anticancer properties in vivo and in vitro. There are several CLA isomers in ruminant-produced foods, among which t10, c12-CLA and c9, t11-CLA are the most potent. Protein tyrosine phosphatase gamma (PTPgamma) has been implicated as a tumor suppressor gene in kidney and lung cancers. Our previous results indicated that estradiol-17beta (E2)-induced suppression of PTPgamma may play a role in mammary tumorigenesis. MATERIALS AND METHODS: The effects of t10, c12-CLA and c9, t11-CLA on PTPgamma mRNA expression in human breast epithelial cells and stromal cells, isolated from surgical specimens of mammoplasty and breast cancer patients, were detected and quantified by RT-PCR RESULTS: The PTPgamma mRNA expression was lower in cancer than in normal breast cells. Both t10, c12-CLA and c9, t11-CLA significantly (p < 0.05) increased the PTPgamma mRNA levels in primary cultured normal breast epithelial cells, normal breast stromal cells and breast cancer epithelial cells, but not in breast cancer stromal cells. t10, c12-CLA appeared to be the most active isomer in estrogen receptor a (ERalpha)-positive human breast cancer epithelial cells. CONCLUSION: The results indicate that dietary CLA might serve as a chemo-preventive and chemo-therapeutic agent in human breast cancers by up-regulating the estrogen-regulated tumor suppressor gene, PTPgamma expression.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Linoleic Acids, Conjugated/pharmacology , Nerve Tissue Proteins/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Estrogen Receptor alpha/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , Humans , Nerve Tissue Proteins/genetics , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Stromal Cells/drug effects , Stromal Cells/enzymology , Up-Regulation/drug effects
11.
Anticancer Res ; 25(6B): 4061-8, 2005.
Article in English | MEDLINE | ID: mdl-16312046

ABSTRACT

BACKGROUND: Conjugated linoleic acid (CLA), a naturally occurring compound found in ruminant dairy and beef products, has been shown to possess anti-cancer ability in vivo and in vitro. There are several CLA isomers in ruminant-produced food products, among which t10,c12-CLA and c9,t11-CLA are most potent. Vascular endothelial growth factor-A (VEGF-A) has been implicated as an angiogenesis-activating cytokine. Our previous results indicated that CLA induced suppression of VEGF-A in MCF-7 cells, which may be one of CLA's anticancer mechanisms. MATERIALS AND METHODS: The effects of 10,c12-CLA and c9,t11-CLA on VEGF-A mRNA and protein expression in MCF-7 cells, which were co-cultured with human breast stromal cells isolated from breast tissues of surgical specimens of mammoplasty and breast cancer patients, were detected by RT-PCR and Western blot analysis. RESULTS: VEGF-A mRNA and protein expressions were significantly (p < 0.05) elevated in co-cultured MCF-7 cells in comparison with cultured MCF-7 cells alone. Normal human breast stromal cells contribute greater effects in increasing VEGF-A protein expression in MCF-7 cells. Both t10,c12-CLA and c9,t11-CLA significantly (p < 0.05) decreased VEGF-A mRNA and protein levels in co-cultured MCF-7 cells. t10,c12-CLA appeared to be the more active isomer than c9, t11-CLA. CONCLUSION: The results indicate that dietary CLA might serve as a chemo-therapeutic agent in human breast cancers by down-regulating VEGF-A expression.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Communication/physiology , Linoleic Acids, Conjugated/pharmacology , Stromal Cells/cytology , Vascular Endothelial Growth Factor A/biosynthesis , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Cell Line, Tumor , Coculture Techniques , Down-Regulation , Humans , Isomerism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Vascular Endothelial Growth Factor A/genetics
12.
Anticancer Res ; 25(6B): 4081-90, 2005.
Article in English | MEDLINE | ID: mdl-16312047

ABSTRACT

BACKGROUND: Benign prostatic hyperplasia (BPH) is an age-dependent prostatic disease in human males and dogs. The prostatic stromal estrogen level of health control and BPH patients increases significantly with age, while the dihydrotestosterone (DHT) level is not connected with age. Moreover, experimentally estrogens have induced BPH in the presence of androgens. Our aim was to investigate the effects of age on the proliferation and estrogen receptor beta (ER/3) mRNA of canine prostatic epithelial and stromal cells. MATERIALS AND METHODS: Epithelial and stromal cells were isolated from canine prostatic tissues. The proliferation of these cell types from dogs of different ages was assessed by thymidine incorporation assay, while the expression and identification of ERbeta mRNA were performed by RT-PCR and DNA sequence. RESULTS: Prostatic epithelial cells isolated from 1-year-old dogs exhibited a greater proliferative activity than those of 4-year-old dogs. In contrast, the prostatic stromal cells from 4-year-old dogs proliferated more rapidly than the cells from 1-year-old dogs. ERbeta mRNA expression was detected in the canine prostatic epithelial and stromal cells, decreasing with age. The partial DNA sequence showed that the canine ERbeta sequence shares 90.0%, 87.0% and 83.0% of its nucleotide homology with human, rat and mouse ERbeta, respectively. CONCLUSION: The decrease in the expression of ERbeta in prostatic cells with age reduces its negative control over the androgen receptor, associated with the overgrowth of canine prostatic stromal cells, which further induces the development of canine BPH.


Subject(s)
Estrogen Receptor beta/biosynthesis , Prostate/cytology , Prostate/metabolism , RNA, Messenger/biosynthesis , Age Factors , Animals , Base Sequence , Cell Growth Processes/physiology , Cells, Cultured , Dogs , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Estrogen Receptor beta/genetics , Male , Molecular Sequence Data , Prostate/physiology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/physiology
13.
Anticancer Res ; 24(1): 91-100, 2004.
Article in English | MEDLINE | ID: mdl-15015581

ABSTRACT

BACKGROUND: Racemic gossypol [(+/-)-GP], a naturally occurring polyphenolic yellow pigment present in cottonseed products, inhibits in vitro proliferation of Dunning prostate cancer cells (MAT-LyLu), human prostate cancer cells derived from a bone marrow metastasis (PC3), MCF-7 and primary cultured human prostate cells. (+/-)-GP also has the ability to inhibit the metastasis of lung and lymph nodes of the androgen-independent rodent prostate cancer cell line, MAT-LyLu, after implantation into Copenhagen rats. MATERIALS AND METHODS: The effects of (+/-)-GP on the proliferation of human prostate cancer PC3 cells were determined by thymidine incorporation assay and doubling-time (DT) determination. The mechanisms of action of (+/-)-GP on the proliferation of PC3 cells were determined by RT-PCR analysis, ELISA assay and Western blot analysis. RESULTS: The results show that (+/-)-GP caused reductions in DNA synthesis and prolonged the DTs in PC3 cells. RT-PCR and ELISA results show that (+/-)-GP elevate the mRNA expression and protein secretion of transforming growth factor beta1 (TGFbeta1) in PC3 cells. Consistent with these findings, (+/-)-GP has been shown to decrease the cyclin D1 mRNA expression and protein expression in PC3 cells. Furthermore, the growth inhibition of PC3 cells by conditioned media collected from the (+/-)-GP-treated-PC3 cells was completely reversed by addition of 25 microg/ml of mouse monoclonal anti-TGFbeta1, -beta2, -beta3 antibody, suggesting the involvement of TGFbeta1 in (+/-)-GP-induced growth inhibition of PC3 cells. CONCLUSION: These results indicate that the inhibitory effects of (+/-)-GP on the proliferation of human prostate cancer PC3 cells are associated with induction of TGFbeta1, which in turn influences the expression of the cell cycle-regulatory protein, cyclin D1, in prostate cancer cells.


Subject(s)
Gossypol/pharmacology , Prostatic Neoplasms/drug therapy , Transforming Growth Factor beta/biosynthesis , Animals , Cell Division/drug effects , Cyclin D1/biosynthesis , Cyclin D1/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Xenograft Model Antitumor Assays
14.
Anticancer Res ; 22(3): 1491-6, 2002.
Article in English | MEDLINE | ID: mdl-12168828

ABSTRACT

BACKGROUND: Gossypol (GP) is a potent antifertility agent contained in cottonseed and other parts of cotton plants. We have shown that GP inhibits in vitro growth of Dunning rodent prostate cancer cells (MAT-LyLu), PC3, MCF-7 and primary cultured human prostate cells, as well as the in vivo tumor growth of the MAT-LyLu cell line after implantation into Copenhagen rats. MATERIALS AND METHODS: GP's effects on O2 consumption/CO2 production of PC3 cells were studied using the Micro-Oxymax respirometer. The effects of GP on oxidative phosphorylation in PC3 cells were determined by the succinic dehydrogenase assay. RESULTS: GP at the concentration of 1.0 microM reduced DNA synthesis by 25.6%, 54.6% and 81.25% after treatment times of 24, 28, and 32 hours, respectively, while GP at 2 microM reduced DNA synthesis by 78.57%, 81.44% and 83.72% after treatment durations of 24, 28 and 32 hours, respectively. GP at the concentration of 2.0 microM decreased significantly CO2 production by 37.5%, 42.4% and 44.7% and O2 consumption by 6.4%, 6.9% and 7.9% in PC3 cells after the different periods of treatment (24, 28 and 32 hours, respectively). GP at the concentration of 1.0 microM did not significantly inhibit O2 consumption/CO2 production of PC3 cells. GP also reduced the activity of mitochondrial succinic dehydrogenase in PC3 cells to 31% of control at the concentration of 1.0 microM and 15% of control at the concentration of 2.0 microM. CONCLUSION: The inhibitory action of GP on O2 consumption/CO2 production of PC3 cells may be due to disruption of oxidative phosphorylation by inhibition of mitochondrial succinic dehydrogenase.


Subject(s)
Carbon Dioxide/metabolism , Gossypol/pharmacology , Growth Inhibitors/pharmacology , Oxygen Consumption/drug effects , Prostatic Neoplasms/metabolism , DNA, Neoplasm/biosynthesis , Humans , Male , Mitochondria/drug effects , Mitochondria/enzymology , Oxygen/antagonists & inhibitors , Oxygen/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Succinate Dehydrogenase/metabolism , Tumor Cells, Cultured
15.
Anticancer Res ; 22(1A): 33-8, 2002.
Article in English | MEDLINE | ID: mdl-12017312

ABSTRACT

Natural gossypol (GP), a polyphenolic pigment in cottonseed, is a racemic mixture of two enantiomers, (+)GP and (-)GP. Our aim was to compare the abilities of (+/-)GP, (+)GP and (-)GP to reduce proliferation of breast cancerous epithelial cells (cEC) and cancerous stromal cells (cSC). Proliferation was measured by 3H-thymidine uptake. Results showed that (+)GP had no effect on both cEC and cSC. In contrast, in both cell types, (+/-)GP and (-)GP significantly inhibited proliferation. (+/-)GP caused reductions of 15, 46 and 82% at 25, 5.0 and 7.5 microM, respectively, in cEC, and reductions of 17, 28, 39 and 56% at 2.0, 3.0, 4.0 and 5.0 microM, respectively, in cSC. (-)GP induced reductions of 33, 89 and 98% at 2.5, 5.0 and 7.5 microM, respectively, in cEC, and reductions of 29, 51, 64 and 72% at 2.0, 3.0, 4.0 and 5.0 microM, respectively, in cSC. By RT-PCR, we found that 3 microM of (+/-)GP and (-)GP decreased cyclin D1 mRNA expression in both cell types (52% and 71%, respectively, in cEC; and 47% and 71%, respectively, in cSC), and increased transforming growth factor beta (TGFbeta) mRNA expression in both cell types (93% and 130%, respectively, in cEC; and 45% and 89%, respectively, in cSC). Interestingly, (-)GP was significantly more potent than (+/-)GP. These results show that (-)GP is the major inhibitory component of (+/-)GP, (-)GP is the more potent inhibitor of cancerous breast cell growth, and the inhibitory activity of (-)GP and (+/-)GP is related to the reduction of the cell cycle regulator, cyclin D1, and the induction of the cell proliferation inhibitor, TGFbeta.


Subject(s)
Breast Neoplasms/drug therapy , Gossypol/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/drug effects , Cyclin D1/biosynthesis , Cyclin D1/genetics , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic/drug effects , Growth Inhibitors/pharmacology , Humans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Stereoisomerism , Stromal Cells/drug effects , Stromal Cells/pathology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured
16.
Breast Cancer Res Treat ; 71(1): 21-35, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11859871

ABSTRACT

BACKGROUND: Protein tyrosine phosphatase gamma (PTPgamma) has been implicated as a tumor suppressor gene in kidney and lung cancers. Our previous results indicate that estradiol-17beta (E2)-induced suppression of PTPgamma may play a role in mammary tumorigenesis. Zeranol (Z), a nonsteroidal growth promoter with estrogenic activity that is used by the US meat industry, induces estrogenic responses in primary cultured breast cells and breast cancer cell lines. METHODS: PTPgamma mRNA expression in human breast tissues and cells isolated from surgical specimens of mammoplasty and breast cancer patients were detected and quantified by RT-PCR. Immunohistochemical staining was used to localize PTPgamma in human breast tissues. Breast epithelial and stromal cells were isolated and co-cultured to determine the involvement of cell-cell interaction in the regulation of PTPgamma mRNA expression by E2 and Z. RESULTS: PTPgamma mRNA expression was lower in cancerous than in normal breast tissues. Both E2 and Z suppressed PTPgamma mRNA levels in cultured normal breast tissues by approximately 80%, but had a lesser effect in cultured epithelial cells isolated from normal breast tissues. In the co-culture system, both E2 and Z suppressed PTPgamma mRNA to a greater degree in epithelial cells than in stromal cells. In whole breast tissues, PTPgamma was immunolocalized to the epithelium. Treatment with E2 or Z diminished PTPgamma staining indicating reductions in PTPgamma at the protein level. CONCLUSIONS: The results indicate that both E2 and Z regulate PTPgamma expression in human breast and that epithelial-stromal cells interaction is important in the regulation of PTPgamma expression by estrogenically active agents.


Subject(s)
Breast Neoplasms/pathology , Cell Communication , Cell Transformation, Neoplastic , Estradiol/pharmacology , Estrogens, Non-Steroidal/pharmacology , Gene Expression Regulation, Neoplastic , Nerve Tissue Proteins/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , Zeranol/pharmacology , Breast/cytology , Breast/physiology , Epithelium/physiology , Female , Humans , Immunohistochemistry , Nerve Tissue Proteins/genetics , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/physiology , Tumor Cells, Cultured
17.
Anticancer Res ; 22(6C): 3917-23, 2002.
Article in English | MEDLINE | ID: mdl-12553013

ABSTRACT

We have reported PTP gamma expression was downregulated by 17 beta-estradiol (E2) and Zeranol (Z) and that PTP gamma may function as an estrogen-regulated cancer suppressor in human breast. We utilized RT-PCR to examine expression of estrogen receptor alpha (ER alpha) and beta (ER beta) mRNA in MCF-7 and MDA-MB-231 cells and to investigate the regulation of PTP gamma expression by E2 and Z in the absence or presence of ICI 182,780 (ICI) in both cells, and immunohistochemistry to examine ER alpha and ER beta protein in normal and cancerous human breast. Results show that MCF-7 express both ER alpha and ER beta, and MDA-MB-231 express only ER beta. Both E2 and Z (30 nM; 24 h) suppressed PTP gamma by approximately 56% in MCF-7 cells and these effects were completely blocked by 1 mM of ICI. In contrast, E2, Z and ICI had no effects on PTP gamma expression in MDA-MB-231 cells. Interestingly, both E2 and Z suppressed PTP gamma by approximately 45% in MDA-MB-231 cells transfected with ER alpha, and these effects were completely blocked by 100 nM of ICI. Both RT-PCR and immunohistochemical staining showed that ER alpha expression was significantly higher in cancerous human breast than in normal breast, while ER beta was higher in normal human breast than in cancerous breast. In combination with our previous findings of greater PTP gamma expression levels in normal human breast than cancerous breast, current results show that lower PTP gamma was associated with higher ER alpha in cancerous human breast tissues. In conclusion, results indicate that Z induces estrogenic effects in human breast relative of PTP gamma expression and the estrogenic down-regulation of PTP gamma expression in human breast is associated with ER alpha.


Subject(s)
Breast/metabolism , Estradiol/analogs & derivatives , Estrogens/pharmacology , Nerve Tissue Proteins/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , Receptors, Estrogen/physiology , Breast/drug effects , Breast/enzymology , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Fulvestrant , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Nerve Tissue Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Zeranol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...