Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biol Reprod ; 89(3): 73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23843242

ABSTRACT

Postmenopausal women are at a higher risk of ovarian cancer due, in part, to increased levels of gonadotropins such as luteinizing hormone (LH). Gonadotropins and other stimuli are capable of activating two pathways, PKA and PKC, that are altered in ovarian cancer. To determine the role of LH on ovarian cancer, we explored the effects of human chorionic gonadotropin (hCG), an LH mimic, and an activator of the PKC pathway, phorbol-12-myristate 13-acetate (PMA), on ovarian cancer cell-cycle kinetics and apoptosis in Ovcar3 cells. PMA treatment increased cells in the S phase of the cell cycle and initially increased apoptosis after 4 h before diminishing apoptosis after 8 h. Treatment of ovarian cancer cells with hCG had no effect on these parameters. The PKC pathway is known to differentially regulate matrix metalloproteinase (MMP) expression. Results showed that ovarian cancer cells treated with PMA increased MMP7 and MMP10 mRNA levels after 8 h of treatment, and expression remained high after 12 h before decreasing at 24 h. The mRNA expression of extracellular matrix metalloproteinase inducer (BSG), an activator of MMPs, was unaffected by PMA. Due to the role that MMPs play in migration, we investigated the effect of PMA activation of MMPs on ovarian cancer cell migration. The use of the MMP inhibitor GM6001 blocked the increased migratory effects of PMA on ovarian cancer cells. Together, these studies show that activating the PKC pathway causes significant changes in cell cycle kinetics and selective expression of MMPs that are involved in enhancing ovarian cancer cell proliferation and migration.


Subject(s)
Cell Movement , Cell Proliferation , Chorionic Gonadotropin/pharmacology , Matrix Metalloproteinase 10/genetics , Matrix Metalloproteinase 7/genetics , Ovarian Neoplasms/pathology , Protein Kinase C/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Dipeptides/pharmacology , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase 7/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Signal Transduction/physiology , Tumor Cells, Cultured
2.
Anticancer Res ; 31(3): 813-23, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21498701

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor gamma (PPARγ) is overexpressed in many types of cancer, including breast cancer, and it is regulated by ligand binding and post-translational modifications. It was previously demonstrated that endogenous transactivation promotes an aggressive phenotype of malignant breast cells. This study examines whether selective antagonism of PPARγ with T0070907 is a potential strategy for breast cancer therapy. MATERIALS AND METHODS: PPARγ activation was inhibited using both pharmacological and molecular approaches and proliferation, apoptosis, migration and invasion were measured in MDA-MB-231 and MCF-7 breast cancer cells. RESULTS: T0070907 treatment inhibited proliferation, invasion and migration but did not significantly affect apoptosis. Molecular inhibition using a dominant negative (Δ462) receptor yielded similar results. T007 also mediated a dose-dependent decrease in phosphorylation of PPARγ, and its ability to bind to DNA, and may directly affect mitogen-activated protein kinase signaling. CONCLUSION: These data indicate that inhibiting endogenous PPARγ signaling may be a promising new approach to breast cancer therapy.


Subject(s)
Benzamides/pharmacology , Benzamides/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Movement/drug effects , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA, Neoplasm/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Genes, Dominant/genetics , Humans , Mitogen-Activated Protein Kinases/metabolism , Mutation/genetics , Neoplasm Invasiveness , PPAR gamma/genetics , Phenotype , Phosphorylation/drug effects , Protein Binding/drug effects , Response Elements/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects
3.
PLoS One ; 6(1): e16179, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21283708

ABSTRACT

BACKGROUND: Peroxisome Proliferator Activated Receptor gamma (PPARγ) agonists, such as the thiazolinediones (TZDs), have been studied for their potential use as cancer therapeutic agents. We investigated the effect of four TZDs--Rosiglitazone (Rosi), Ciglitazone (CGZ), Troglitazone (TGZ), and Pioglitazone (Pio)--on ovarian cancer cell proliferation, PPARγ expression and PPAR luciferase reporter activity. We explored whether TZDs act in a PPARγ dependent or independent manner by utilizing molecular approaches to inhibit or overexpress PPARγ activity. PRINCIPAL FINDINGS: Treatment with CGZ or TGZ for 24 hours decreased proliferation in three ovarian cancer cell lines, Ovcar3, CaOv3, and Skov3, whereas Rosi and Pio had no effect. This decrease in Ovcar3 cell proliferation was due to a higher fraction of cells in the G(0)/G(1) stage of the cell cycle. CGZ and TGZ treatment increased apoptosis after 4 hours of treatment but not after 8 or 12 hours. Treatment with TGZ or CGZ increased PPARγ mRNA expression in Ovcar3 cells; however, protein levels were unchanged. Surprisingly, luciferase promoter assays revealed that none of the TZDs increased PPARγ activity. Overexpression of wild type PPARγ increased reporter activity. This was further augmented by TGZ, Rosi, and Pio indicating that these cells have the endogenous capacity to mediate PPARγ transactivation. To determine whether PPARγ mediates the TZD-induced decrease in proliferation, cells were treated with CGZ or TGZ in the absence or presence of a dominant negative (DN) or wild type overexpression PPARγ construct. Neither vector changed the TZD-mediated cell proliferation suggesting this effect of TZDs on ovarian cancer cells may be PPARγ independent. CONCLUSIONS: CGZ and TGZ cause a decrease in ovarian cancer cell proliferation that is PPARγ independent. This concept is supported by the finding that a DN or overexpression of the wild type PPARγ did not affect the changes in cell proliferation and cell cycle.


Subject(s)
Cell Cycle/drug effects , Cell Proliferation/drug effects , Ovarian Neoplasms/pathology , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Cell Line, Tumor , Chromans/pharmacology , Female , Gene Expression , Humans , Ovarian Neoplasms/drug therapy , PPAR gamma/genetics , Pioglitazone , Rosiglitazone , Troglitazone
4.
Mol Cancer ; 7: 90, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-19061500

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor gamma (PPARgamma) is a member of the nuclear hormone receptor superfamily and is highly expressed in many human tumors including breast cancer. PPARgamma has been identified as a potential target for breast cancer therapy based on the fact that its activation by synthetic ligands affects the differentiation, proliferation, and apoptosis of cancer cells. However, the controversial nature of current studies and disappointing results from clinical trials raise questions about the contribution of PPARgamma signaling in breast cancer development in the absence of stimulation by exogenous ligands. Recent reports from both in vitro and in vivo studies are inconsistent and suggest that endogenous activation of PPARgamma plays a much more complex role in initiation and progression of cancer than previously thought. RESULTS: We have previously demonstrated that an increase in expression of PPARgamma1 in MCF-7 breast cancer cells is driven by a tumor-specific promoter. Myc-associated zinc finger protein (MAZ) was identified as a transcriptional mediator of PPARgamma1 expression in these cells. In this study, using RNA interference (RNAi) to inhibit PPARgamma1 expression directly or via down-regulation of MAZ, we report for the first time that a decrease in PPARgamma1 expression results in reduced cellular proliferation in MCF-7 breast cancer cells. Furthermore, we demonstrate that these changes in proliferation are associated with a significant decrease in cell transition from G1 to the S phase. Using a dominant-negative mutant of PPARgamma1, Delta462, we confirmed that PPARgamma1 acts as a pro-survival factor and showed that this phenomenon is not limited to MCF-7 cells. Finally, we demonstrate that down-regulation of PPARgamma1 expression leads to an induction of apoptosis in MCF-7 cells, confirmed by analyzing Bcl-2 expression and PARP-1 cleavage. CONCLUSION: Thus, these findings suggest that an increase in PPARgamma1 signaling observed in breast cancer contributes to an imbalance between proliferation and apoptosis, and may be an important hallmark of breast tumorigenesis. The results presented here also warrant further investigation regarding the use of PPARgamma ligands in patients who are predisposed or already diagnosed with breast cancer.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Down-Regulation , PPAR gamma/deficiency , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , PPAR gamma/genetics , PPAR gamma/metabolism , Phenotype , RNA Interference
5.
Exp Cell Res ; 314(20): 3724-36, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-18929560

ABSTRACT

The inhibition of apoptosis by Toxoplasma gondii is governed by its modulation of several signaling cascades including the NFkappaappaB and JNK pathways. This is evident in the dysregulation of JNK activation following treatment with UV and TNFalpha, both apoptogenic stimuli. Infection-mediated interference with the JNK cascade was found to be highly reproducible in HeLa cells. In light of emerging evidence regarding cross talk between the JNK and NFkappaB cascades, we examined the impact of infection in wild type and RelA/p65-/- mouse embryonic fibroblasts (MEF). Remarkably, parasite infection failed to significantly impact both UV and TNFalpha-mediated JNK phosphorylation in both cell lines suggesting a cell type specific effect. Furthermore siRNA-mediated knockdown of RelA/p65 failed to impact the parasite mediated effects on stimulus dependent activation of JNK in HeLa cells. Finally, the infection mediated suppression of JNK phosphorylation in HeLa cells did not result in decreased JNK kinase activity. Rather, the reduced levels of phospho-JNK in infected cells correlated with increased phosphatase activity noted by the partial rescue of the phenotype following treatment with okadaic acid. Taken together the results indicate that manipulation of the JNK pathway does not involve NFkappaB and is furthermore not a central component of the parasite enforced block of apoptosis. It further highlights the complexity of these systems and the danger of extrapolating results both within and across pathogen-host cell systems based on limited studies.


Subject(s)
Host-Pathogen Interactions/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/physiology , Toxoplasma/physiology , Toxoplasmosis/metabolism , Animals , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , HeLa Cells , Humans , Okadaic Acid/pharmacology , Phosphorylation/drug effects , Phosphorylation/radiation effects , Toxoplasmosis/enzymology , Toxoplasmosis/pathology , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/pharmacology , Ultraviolet Rays
6.
J Nutr ; 138(2): 250-6, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18203887

ABSTRACT

Diets high in (n-3) PUFA decrease colon cancer development and suppress colon tumor growth, but the molecular mechanism through which these compounds act is largely unknown. We sought to determine whether PPARgamma1 serves as a molecular link between the physiological actions of eicosapentaenoic acid (EPA) in human colon cancer cells (HT-29). At nutritionally relevant concentrations, EPA stimulated a PPAR response element (PPRE) reporter assay in a dose-responsive manner in HT-29 cells. Cotreatment with GW9662 (GW), a PPARgamma antagonist, significantly inhibited this effect, whereas overexpressing the receptor enhanced it. EPA also stimulated the PPRE reporter in a PPARgamma negative cancer cell line (22Rv1) when the cells were cotransfected with a PPARgamma1 expression plasmid and this effect was again inhibited by GW. Furthermore, in vitro incubation of EPA with PPARgamma1 enhanced binding of the protein to DNA containing a PPRE. Next, we sought to determine whether EPA or a prostaglandin formed from EPA is the functional ligand of PPARgamma. Cotreatment in HT-29 and 22Rv1 cells with EPA and acetyl salicylic acid, an inhibitor of cyclooxygenase activity, activated the PPRE reporter at levels similar to EPA alone, suggesting that EPA itself is a ligand of PPARgamma. Finally, EPA suppressed HT-29 cell growth and this effect was significantly reversed by the addition of GW, suggesting that in part the physiological actions of EPA are the result of PPARgamma activation. These studies identify PPARgamma as a molecular mediator of (n-3) PUFA actions in colon cancer cells.


Subject(s)
Colonic Neoplasms/metabolism , Eicosapentaenoic Acid/pharmacology , PPAR gamma/metabolism , Anilides/pharmacology , Cell Proliferation , Gene Expression Regulation , HT29 Cells , Humans , Male , PPAR gamma/genetics , Prostatic Neoplasms/metabolism , Protein Binding , Response Elements/physiology , Rosiglitazone , Thiazolidinediones/pharmacology
7.
Breast Cancer Res Treat ; 111(1): 103-11, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17902047

ABSTRACT

The peroxisome proliferator-activated receptor gamma 1 (PPARgamma1) is a nuclear receptor that plays a pivotal role in breast cancer and is highly over-expressed relative to normal epithelia. We have previously reported that the expression of PPARgamma1 is mediated by at least six distinct promoters and expression in breast cancer is driven by a tumor-specific promoter (pA1). Deletional analysis of this promoter fragment revealed that the GC-rich, 263 bp sequence proximal to the start of exon A1, is sufficient to drive expression in breast cancer cells but not in normal, human mammary epithelial cells (HMEC). By combining the disparate technologies of microarray and computer-based transcription factor binding site analyses on this promoter sequence the myc-associated zinc finger protein (MAZ) was identified as a candidate transcription factor mediating tumor-specific expression. Western blot analysis and chromatin immunoprecipitation assays verify that MAZ is overexpressed in MCF-7 cells and is capable of binding to the 263 bp promoter fragment, respectively. Furthermore, the over-expression of MAZ in HMEC is sufficient to drive the expression of PPARgamma1 and does so by recruiting the tumor-specific promoter. This results in an increase in the amount of PPARgamma1 capable of binding to its DNA response element. These findings help to define the molecular mechanism driving the high expression of PPARgamma1 in breast cancer and raise new questions regarding the role of MAZ in cancer progression.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , PPAR gamma/biosynthesis , Transcription Factors/metabolism , Blotting, Western , Cell Line, Tumor , Disease Progression , Female , Gene Expression , Humans , Immunoprecipitation , Oligonucleotide Array Sequence Analysis , PPAR gamma/genetics , Plasmids , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Transfection
8.
Cancer Res ; 64(16): 5592-6, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15313896

ABSTRACT

Peroxisome proliferator-activated receptor-gamma1 (PPARgamma1) is transactivated by a wide range of ligands in normal human mammary epithelial and breast cancer cells. Although transactivation of PPARgamma mediates the expression of genes that are markers of differentiation, its overexpression in cancers of the breast, thyroid, colon, and lung suggests its dysregulation may play a role in oncogenesis, cancer progression, or both. We report the overexpression of PPARgamma is caused by the use of a tumor-specific promoter in breast cancer cells that is distinct from the promoter used in normal epithelia. Thus, the increase in PPARgamma expression seen in breast cancer cells results from promoter recruitment, providing new insights into the expression and actions of PPARgamma in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Cell Line, Tumor , Epithelial Cells/metabolism , Exons , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...