Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 37(35): 4854-4870, 2018 08.
Article in English | MEDLINE | ID: mdl-29773902

ABSTRACT

Neural precursor cell expressed, developmentally downregulated 9 (NEDD9) supports oncogenic signaling in a number of solid and hematologic tumors. Little is known about the role of NEDD9 in ovarian carcinoma (OC), but available data suggest elevated mRNA and protein expression in advanced stage high-grade cancers. We used a transgenic MISIIR-TAg mouse OC model combined with genetic ablation of Nedd9 to investigate its action in the development and progression of OC. A Nedd9-/- genotype delayed tumor growth rate, reduced incidence of ascites, and reduced expression and activation of signaling proteins including SRC, STAT3, E-cadherin, and AURKA. Cell lines established from MISIIR-TAg;Nedd9-/- and MISIIR-TAg;Nedd9+/+ mice exhibited altered migration and invasion. Growth of these cells in a syngeneic allograft model indicated that systemic Nedd9 loss in the microenvironment had little impact on tumor allograft growth, but in a Nedd9 wild-type background Nedd9-/- allografts exhibited significantly reduced growth, dissemination, and oncogenic signaling compared to Nedd9+/+ allografts. Gene expression analysis revealed that Nedd9+/+ tumors exhibited more mesenchymal "stem-like" transcriptional program, including increased expression of Aldh1a1 and Aldh1a2. Conversely, loss of Nedd9 resulted in increased expression of differentiation genes, including fallopian tube markers Foxj1, Ovgp1, and Pax8. Collectively, these data suggest that tumor cell-intrinsic Nedd9 expression promotes OC development and progression by broad induction of oncogenic protein signaling and stem/mesenchymal gene expression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Mesenchymal Stem Cells/metabolism , Ovarian Neoplasms/genetics , Phosphoproteins/genetics , Signal Transduction/genetics , Animals , Cadherins/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/genetics , Female , Humans , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Oncogenes/genetics , Ovarian Neoplasms/pathology , Transcription, Genetic/genetics
2.
Oncotarget ; 8(31): 50376-50392, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28881569

ABSTRACT

Ovarian cancer is usually diagnosed at late stages when cancer has spread beyond the ovary and patients ultimately succumb to the development of drug-resistant disease. There is an urgent and unmet need to develop therapeutic strategies that effectively treat ovarian cancer and this requires a better understanding of signaling pathways important for ovarian cancer progression. Aurora A kinase (AURKA) plays an important role in ovarian cancer progression by mediating mitosis and chromosomal instability. In the current study, we investigated the role of AURKA in regulating the DNA damage response and DNA repair in ovarian carcinoma cells. We discovered that AURKA modulated the expression and activity of PARP, a crucial mediator of DNA repair that is a target of therapeutic interest for the treatment of ovarian and other cancers. Further, specific inhibition of AURKA activity with the small molecule inhibitor, alisertib, stimulated the non-homologous end-joining (NHEJ) repair pathway by elevating DNA-PKcs activity, a catalytic subunit required for double-strand break (DSB) repair, as well as decreased the expression of PARP and BRCA1/2, which are required for high-fidelity homologous recombination-based DNA repair. Further, AURKA inhibition stimulates error-prone NHEJ repair of DNA double-strand breaks with incompatible ends. Consistent with in vitro findings, alisertib treatment increased phosphorylated DNA-PKcs(pDNA-PKcsT2609) and decreased PARP levels in vivo. Collectively, these results reveal new non-mitotic functions for AURKA in the regulation of DNA repair, which may inform of new therapeutic targets and strategies for treating ovarian cancer.

3.
Mol Cancer Res ; 6(5): 695-705, 2008 May.
Article in English | MEDLINE | ID: mdl-18505915

ABSTRACT

Transforming growth factor-beta (TGF-beta) is thought to play a role in the pathobiological progression of ovarian cancer because this peptide hormone is overexpressed in cancer tissue, plasma, and peritoneal fluid. In the current study, we investigated the role of the TGF-beta/Smad3 pathway in ovarian cancer metastasis by regulation of an epithelial-to-mesenchymal transition. When cancer cells were cultured on plastic, TGF-beta1, TGF-beta2, and TGF-beta3 induced pro-matrix metalloproteinase (MMP) secretion, loss of cell-cell junctions, down-regulation of E-cadherin, up-regulation of N-cadherin, and acquisition of a fibroblastoid phenotype, consistent with an epithelial-to-mesenchymal transition. Furthermore, Smad3 small interfering RNA transfection inhibited TGF-beta-mediated changes to a fibroblastic morphology, but not MMP secretion. When cancer cells were cultured on a three-dimensional collagen matrix, TGF-beta1, TGF-beta2, and TGF-beta3 stimulated both pro-MMP and active MMP secretion and invasion. Smad3 small interfering RNA transfection of cells cultured on a collagen matrix abrogated TGF-beta-stimulated invasion and MMP secretion. Analysis of Smad3 nuclear expression in microarrays of serous benign tumors, borderline tumors, and cystadenocarcinoma revealed that Smad3 expression could be used to distinguish benign and borderline tumors from carcinoma (P = 0.006). Higher Smad3 expression also correlated with poor survival (P = 0.031). Furthermore, a direct relationship exists between Smad3 nuclear expression and expression of the mesenchymal marker N-cadherin in cancer patients (P = 0.0057). Collectively, these results implicate an important role for the TGF-beta/Smad3 pathway in mediating ovarian oncogenesis by enhancing metastatic potential.


Subject(s)
Epithelium/metabolism , Gene Expression Regulation, Neoplastic , Mesoderm/metabolism , Neoplasm Metastasis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta3/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Female , Humans , RNA, Small Interfering/metabolism
4.
Endocrinology ; 149(8): 3809-16, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18450971

ABSTRACT

Elevated activin A levels in serum, cyst fluid, and peritoneal fluid of ovarian cancer patients suggest a role for this peptide hormone in disease development. We hypothesize that activin A plays a role in ovarian tumor biology, and analyzed activin-mediated pro-oncogenic signaling in vitro and the expression of activin signaling pathway molecules in vivo. Activin A regulation of Akt and GSK, and the effects of repressing the activities of these molecules (with pharmacological inhibitors) on cellular proliferation were assessed in the cell line, OVCA429. Activin A activated Akt, which phosphorylated GSK, repressing GSK activity in vitro. Activin A stimulated cellular proliferation and repression of GSK augmented activin-regulated proliferation. To validate in vitro observations, immunostaining of the betaA-subunit of activin A and phospho-GSKalpha/beta (Ser9/21) was performed, and the correlation between immunoreactivity levels of these markers and survival was evaluated in benign serous cystadenoma, borderline tumor, and cystadenocarcinoma microarrays. Analysis of tissue microarrays revealed that betaA expression in epithelia did not correlate with survival or malignancy, but expression was elevated in stromal cells from carcinomas when compared with benign tumors. Phospho-GSKalpha/beta (Ser9/21) staining was more intense in mitotically active carcinoma cells and exhibited a polarized localization in benign neoplasms that was absent in carcinomas. Notably, lower phospho-GSKalpha/beta (Ser9/21) immunoreactivity correlated with better survival for carcinoma patients (P=0.046). Our data are consistent with a model in which activin A may mediate ovarian oncogenesis by activating Akt and repressing GSK to stimulate cellular proliferation.


Subject(s)
Activins/physiology , Carcinoma/pathology , Glycogen Synthase Kinase 3/physiology , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-akt/physiology , Activins/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma/metabolism , Carcinoma/mortality , Cell Proliferation , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Cystadenoma, Serous/metabolism , Cystadenoma, Serous/pathology , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , Middle Aged , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Phosphorylation , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Survival Analysis , Tumor Cells, Cultured
5.
Mol Cancer Res ; 5(2): 121-31, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17314270

ABSTRACT

Epithelial ovarian cancer (EOC) is asymptomatic at early stages and is often diagnosed late when tumor cells are highly metastatic. Lysophosphatidic acid (LPA) has been implicated in ovarian oncogenesis as levels of this lipid are elevated in patient ascites and plasma. Because the underlying mechanism governing LPA regulation of matrix metalloproteinase-2 (MMP-2) activation remains undefined, we investigated the relationship between LPA-induced changes in actin microfilament organization and MMP-2 enzymatic activity. We report that when cells were cultured at a high density, LPA mediated stress fiber and focal adhesion disassembly and significantly repressed RhoA activity in EOC cells. Inhibition of Rho-kinase/ROCK enhanced both LPA-stimulated loss of stress fibers and pro-MMP-2 activation. In contrast, expression of the constitutively active RhoA(G14V) mutant diminished LPA-induced pro-MMP-2 activation. LPA had no effects on membrane type 1-MMP or tissue inhibitor of metalloproteinase-2 expression, but up-regulated MMP-2 levels, contributing to the induction of MMP-2 activation. Interestingly, when cells were cultured at a low density, stress fibers were present after LPA stimulation, and ROCK activity was required for EOC cell migration. Collectively, these results were consistent with a model in which LPA stimulates the metastatic dissemination of EOC cells by initiating loss of adhesion and metalloproteinase activation.


Subject(s)
Enzyme Precursors/metabolism , Gene Expression Regulation, Neoplastic , Lysophospholipids/pharmacology , Matrix Metalloproteinase 2/metabolism , Ovarian Neoplasms/metabolism , Stress Fibers/metabolism , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ovarian Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , Stress Fibers/drug effects , Transfection , Up-Regulation , rho-Associated Kinases , rhoA GTP-Binding Protein/metabolism
6.
Gynecol Oncol ; 95(3): 488-95, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15581951

ABSTRACT

OBJECTIVE: Constitutive expression and upregulation of FasL by malignant epithelial cells counterattack infiltrating natural killer (NK) cells and cytotoxic T lymphocytes (CTLs) and induce apoptosis of normal cells within the tumor, which may induce metastasis. As little is known about the mechanisms that regulate expression of Fas ligand and the subsequent release of FasL in epithelial ovarian cancer cells (EOC), we investigated the effects of lysophosphatidic acid (LPA) on FasL expression and associated signaling pathways. METHODS: We used established EOC cell lines that were incubated with or without LPA and FasL expression was detected by flow cytometry. Cells were additionally lysed and detected for total protein expression. Activated CD4+ T cells, after coculture with or without EOC, were collected for apoptosis staining and analysis by flow cytometry. RESULTS: Flow cytometry showed that LPA strongly upregulated FasL expression on the OVCAR3 cell surface (P < 0.01), yet in Dov13 cells, LPA significantly upregulated FasL expression only in the presence of the general matrix metalloproteinase (MMP) inhibitors GM6001 and MMP inhibitor II (P < 0.01). The MEK/ERK1/2 kinase cascade is required for FasL upregulation, since the MEK inhibitor PD98059 significantly inhibited FasL upregulation induced by LPA (P < 0.01). Type II secretory phospholipase A2 (sPLA2-II), which promotes protein exocytosis from secretory vesicles and gelatinase granules, affects FasL translocation from intracellular to the cell surface. Pretreatment of Dov13 cells with LPA increased activated T cell apoptosis in cocultures. CONCLUSIONS: These data suggest that upregulation of FasL by LPA provides EOC immune-privilege and leads to apoptosis of activated T lymphocytes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lysophospholipids/pharmacology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Ovarian Neoplasms/immunology , Apoptosis/drug effects , Apoptosis/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Cell Membrane/immunology , Cell Membrane/metabolism , Coculture Techniques , Enzyme Activation , Fas Ligand Protein , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Membrane Glycoproteins/genetics , Mitogen-Activated Protein Kinases/metabolism , Ovarian Neoplasms/metabolism , Phospholipases A/metabolism , Phospholipases A2 , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...