Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
PLoS One ; 11(8): e0160700, 2016.
Article in English | MEDLINE | ID: mdl-27490118

ABSTRACT

BACKGROUND: During short-term hypoxia, Hypoxia Inducible Factors (particular their subunits HIF-1α and HIF-2α) regulate the expression of many genes including the potent angiogenesis stimulator VEGF. However, in some pathological conditions chronic hypoxia occurs and is accompanied by reduced angiogenesis. OBJECTIVES: We investigated the effect of prolonged hypoxia on the proliferation and sprouting ability of human microvascular endothelial cells and the involvement of the HIFs and Dll4/Notch signaling. METHODS AND RESULTS: Human microvascular endothelial cells (hMVECs), cultured at 20% oxygen for 14 days and seeded on top of 3D fibrin matrices, formed sprouts when stimulated with VEGF-A/TNFα. In contrast, hMVECs precultured at 1% oxygen for 14 days were viable and proliferative, but did not form sprouts into fibrin upon VEGF-A/TNFα stimulation at 1% oxygen. Silencing of HIF-2α with si-RNA partially restored the inhibition of endothelial sprouting, whereas HIF-1α or HIF-3α by si-RNA had no effect. No involvement of Dll4/Notch pathway in the inhibitory effect on endothelial sprouting by prolonged hypoxia was found. In addition, hypoxia decreased the production of urokinase-type plasminogen activator (uPA), needed for migration and invasion, without a significant effect on its inhibitor PAI-1. This was independent of HIF-2α, as si-HIF-2α did not counteract uPA reduction. CONCLUSION: Prolonged culturing of hMVECs at 1% oxygen inhibited endothelial sprouting into fibrin. Two independent mechanisms contribute. Silencing of HIF-2α with si-RNA partially restored the inhibition of endothelial sprouting pointing to a HIF-2α-dependent mechanism. In addition, reduction of uPA contributed to reduced endothelial tube formation in a fibrin matrix during prolonged hypoxia.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Fibrin/chemistry , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Culture Techniques , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Microvessels/cytology , Plasminogen Activator Inhibitor 1/analysis , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Urokinase-Type Plasminogen Activator/analysis , Vascular Endothelial Growth Factor A/pharmacology
3.
Cell Oncol ; 31(3): 179-90, 2009.
Article in English | MEDLINE | ID: mdl-19478386

ABSTRACT

BACKGROUND: 1F6 human melanoma xenografts overexpressing either the 18 kD (18kD) form or all (ALL) forms of human basic fibroblast growth factor (bFGF) demonstrate an abundant number of microvessels and accelerated growth. We now examined whether bFGF mediates vascular endothelial growth factor (VEGF) expression. METHODS: Quantitative RT-PCR was used to determine bFGF and VEGF mRNA, VEGF protein secretion was measured by ELISA and VEGF promoter activation was assessed by a dual luciferase activity assay. Western blot was carried out to detect phosphorylation of bFGF-regulated target proteins. RESULTS: In 1F6-18kD and 1F6-ALL clones VEGF mRNA was increased 4- to 5-fold and VEGF protein secretion was highly stimulated due to activation of the VEGF promotor. PI-3K, p38 MAPK and ERK1/2 MAPK pathways were activated, while inhibition of PI-3K or p38 resulted in, respectively, 55% and up to 70% reduction of VEGF mRNA overexpression. A concurrent 60% decrease in VEGF protein secretion was mostly apparent upon inhibition of PI-3K. Inhibition of ERK1/2 hardly affected VEGF mRNA or protein secretion. Two unselected human melanoma cell lines with high metastatic potential contained high bFGF and VEGF, while three non- or sporadically metastatic cell lines displayed low bFGF and VEGF. CONCLUSION: These data indicate that stimulation of VEGF protein secretion in response to bFGF overexpression may contribute to increased vascularization and enhanced aggressiveness in melanoma.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Vascular Endothelial Growth Factor A/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line, Tumor , Enzyme Activation , Fibroblast Growth Factor 2/genetics , Humans , Melanoma/genetics , Melanoma/metabolism , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
4.
Biochem Pharmacol ; 74(2): 191-201, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17498666

ABSTRACT

Vascular Endothelial Growth Factor (VEGF) and its transcriptional regulator Hypoxia-inducible Factor 1 (HIF-1) play an important role in the process of angiogenesis in many types of cancer, including ovarian cancer. We have examined whether the DNA-damaging drugs cisplatin and doxorubicin and the microtubule inhibitors docetaxel and paclitaxel can affect VEGF expression and HIF-1 activity in three human ovarian cancer cell lines. We demonstrate that cisplatin and doxorubicin abolish hypoxia-induced VEGF mRNA expression in all cell lines, while basal VEGF mRNA expression was also downregulated. Transient transfection with a HIF-1-responsive luciferase construct indicated that cisplatin and doxorubicin inhibited hypoxic activation of HIF-1. Cisplatin repressed HIF-1alpha protein expression in all cell lines. Stimulation of HIF-1alpha protein degradation by cisplatin was observed in the only cell line expressing wild-type p53. Cisplatin also inhibited the synthesis of HIF-1alpha protein for which p53 was dispensable. Interestingly, cisplatin strongly reduced the protein levels of the HIF-1 coactivators p300 and CREB-binding protein (CBP) under hypoxia in all cell lines. Although doxorubicin inhibited hypoxic activation of HIF-1, this drug had no significant effect on the expression levels of HIF-1alpha and hypoxic expression of p300 and CBP was only weakly reduced. Docetaxel and paclitaxel did neither influence VEGF expression nor hypoxia-induced HIF-1 activity. In total, our findings indicate that cisplatin and doxorubicin can repress hypoxic induction of VEGF expression by inhibiting HIF-1 through different mechanisms. This knowledge may be useful for future treatment schedules including agents that target the HIF-1 signalling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ovarian Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/genetics , Antigens, Neoplasm/genetics , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carrier Proteins/genetics , Cell Line, Tumor , Corticosterone , Down-Regulation , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , p300-CBP Transcription Factors/genetics
5.
Melanoma Res ; 17(3): 155-68, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17505261

ABSTRACT

Basic fibroblast growth factor is the best-characterized autocrine growth factor in melanoma development and progression. We hypothesized that basic fibroblast growth factor might induce a more aggressive phenotype dependent on the amount of protein expressed in melanoma. Two human melanoma cell lines, M14 and 1F6, known to have low endogenous basic fibroblast growth factor expression and slow growth as subcutaneous xenografts, were stably transfected with vectors encoding either the 18 kDa or all (ALL) isoform proteins of human basic fibroblast growth factor. Different clones overexpressing the 18 kDa or ALL basic fibroblast growth factor proteins were easily obtained. Increased levels of basic fibroblast growth factor were secreted in conditioned medium and stored on the extracellular membrane. Biological activity of the overexpressed basic fibroblast growth factor was confirmed in a human umbilical vein endothelial cell proliferation assay. In 1F6 cells, overexpression of either 18 kDa or ALL basic fibroblast growth factor proteins resulted in up to two-fold shorter in-vitro doubling times (P<0.05). In addition, in vivo, both 18 kDa and ALL basic fibroblast growth factor-overexpressing 1F6 subcutaneous xenografts displayed significantly higher growth rates (P<0.05). In contrast, no major differences in in-vitro and in-vivo doubling times were observed when 18 kDa or ALL isoforms of basic fibroblast growth factor were overexpressed in M14 cells. Interestingly, basic fibroblast growth factor overexpression only affected the microvasculature in 1F6 xenografts. Although blood vessels in 1F6 parent tumors were large, 1F6 tumors overexpressing basic fibroblast growth factor contained numerous small, compressed vessels. Taken together, overexpression of the 18 kDa basic fibroblast growth factor protein only can promote autocrine melanoma cell growth and paracrine-driven angiogenesis.


Subject(s)
Autocrine Communication , Cell Proliferation , Fibroblast Growth Factor 2/metabolism , Melanoma, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Paracrine Communication , Animals , Autocrine Communication/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/pharmacology , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/pharmacology , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Weight , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Paracrine Communication/drug effects , Phosphorylation , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Transfection
6.
Clin Cancer Res ; 10(3): 1180-91, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871998

ABSTRACT

PURPOSE: Expression of aminopeptidase N/CD13 can be detected in several solid tumor types. Thus far, the role of CD13 in ovarian cancer has not been studied. We have investigated the expression pattern and biological function of CD13 in ovarian cancer. EXPERIMENTAL DESIGN: First, we studied the expression of CD13 in ovarian cancer tissue of 15 patients representing three different histological types (5 patients each) by immunohistochemistry. We then stably transfected the IGROV-1 human ovarian cancer cell line with a CD13 expression vector and examined the biological effect of CD13 in vitro and in vivo. RESULTS: The expression of CD13 in ovarian cancer was associated with the histological subtype: CD13 expression in tumor cells was observed in 80-100% of the patients with a serous or mucinous carcinoma and in only 20% of the clear cell carcinoma patients. In all patients' tumor samples, CD13-positive blood vessels were present. CD13 overexpression in IGROV-1 cells did not affect in vitro cell growth and sensitivity to doxorubicin, cisplatin, or gemcitabine. CD13 overexpression reduced invasion in Matrigel, which appeared to be independent of the aminopeptidase activity of CD13. Furthermore, the growth rate of IGROV-1/CD13 xenografts was reduced. The area of the vessel lumens was enlarged in a small percentage of vessels in the CD13-overexpressing xenografts. In addition, the CD13-overexpressing tumors were less sensitive to cisplatin. CONCLUSIONS: CD13 is expressed in tumor as well as endothelial cells in human ovarian cancer. Our results suggest that CD13 overexpression affects ovarian cancer growth, vascular architecture, and response to chemotherapy. Further elucidation of the mechanism of the observed effects of CD13 is warranted to better understand its role in the pathophysiology of ovarian cancer.


Subject(s)
CD13 Antigens/biosynthesis , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Mucinous/metabolism , Aminopeptidases/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Division , Cell Line, Tumor , Cell Membrane/metabolism , Collagen/pharmacology , Cystadenocarcinoma, Serous/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Female , Humans , Immunohistochemistry , Laminin/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Plasmids/metabolism , Proteoglycans/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S Phase , Time Factors , Transfection
7.
J Biol Chem ; 278(9): 6885-95, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12482858

ABSTRACT

Recently we have demonstrated that sodium arsenite induces the expression of hypoxia-inducible factor 1alpha (HIF-1alpha) protein and vascular endothelial growth factor (VEGF) in OVCAR-3 human ovarian cancer cells. We now show that arsenic trioxide, an experimental anticancer drug, exerts the same effects. The involvement of phosphatidylinositol 3-kinase and mitogen-activated protein kinase (MAPK) pathways in the effects of sodium arsenite was investigated. By using kinase inhibitors in OVCAR-3 cells, both effects of sodium arsenite were found to be independent of phosphatidylinositol 3-kinase and p44/p42 MAPKS but were attenuated by inhibition of p38 MAPK. A role for p38 in the regulation of HIF-1alpha and VEGF expression was supported further by analysis of activation kinetics. Experiments in mouse fibroblast cell lines, lacking expression of c-Jun N-terminal kinases 1 and 2, suggested that these kinases are not required for induction of HIF-1alpha protein and VEGF mRNA. Unexpectedly, sodium arsenite did not activate a HIF-1-dependent reporter gene in OVCAR-3 cells, indicating that functional HIF-1 was not induced. In agreement with this hypothesis, up-regulation of VEGF mRNA was not reduced in HIF-1alpha(-/-) mouse fibroblast cell lines. Altogether, these data suggest that not HIF-1, but rather p38, mediates induction of VEGF mRNA expression by sodium arsenite.


Subject(s)
Arsenites/pharmacology , DNA-Binding Proteins/metabolism , Endothelial Growth Factors/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Lymphokines/biosynthesis , Mitogen-Activated Protein Kinases/physiology , Nuclear Proteins/metabolism , Sodium Compounds/pharmacology , Transcription Factors , Animals , Blotting, Western , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , JNK Mitogen-Activated Protein Kinases , Kinetics , Luciferases/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , RNA, Messenger/metabolism , Ribonucleases/metabolism , Subcellular Fractions , Time Factors , Transfection , Tumor Cells, Cultured , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , p38 Mitogen-Activated Protein Kinases
8.
Biochem Pharmacol ; 63(4): 733-43, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11992642

ABSTRACT

Paclitaxel is able to cause cell death through the induction of apoptosis. Cell death characteristics for docetaxel have not yet been described in detail. We investigated four unselected human ovarian cancer cell lines for the sensitivity to a 1hr exposure to docetaxel and calculated the concentrations inhibiting 50% (IC(50)) and 90% (IC(90)) of cell growth. Of the cell lines A2780, H134, IGROV-1 (all wild-type p53) and OVCAR-3 (mutant, mt p53) A2780 was most sensitive and OVCAR-3 least sensitive. Equitoxic drug concentrations representing IC(90) values (25-510nM) were applied for 1hr to measure cell cycle distribution, DNA degradation, and to count apoptotic cell bodies and cells with multifragmented nuclei at various time-points after drug exposure. H134, IGROV-1 and OVCAR-3 showed a continued mitotic block up to at least 72hr and prolonged presence of cells with multifragmented nuclei. High percentages of apoptosis were calculated at 48hr and at later time-points. In contrast, A2780 cells accumulated in the S-phase of the cell cycle and apoptosis was hardly present. The changes in the expression levels of p53, p21/WAF1, Bax and Bcl-2, were not predictive for docetaxel-induced apoptosis. Caspase-3 activation occurred only in cells with accumulation in the G2/M phase starting as early as 8hr in OVCAR-3. Prolonged Bcl-2 phosphorylation was evident in OVCAR-3, visible at 24hr in H134 and IGROV-1, while this phenomenon did not occur in A2780. The mitogen-activated protein kinase pathway (JNKs/SAPKs or c-Jun N-terminal kinases/stress-activated protein kinases, JNK1/2; extracellular response kinase, ERK1/2; p38) did not seem to be directly involved in Bcl-2 phosphorylation or apoptosis. We conclude that docetaxel is able to activate caspase-3, induce Bcl-2 phosphorylation and apoptosis in cells that show a prolonged G2/M arrest, but cells may also die by a caspase-3-independent cell death mechanism.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Caspases/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Taxoids , Apoptosis , Caspase 3 , Cell Cycle/drug effects , Docetaxel , Enzyme Activation/drug effects , Female , Humans , Kinetics , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Tumor Cells, Cultured
9.
Am J Pathol ; 160(2): 537-48, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11839574

ABSTRACT

Vascular endothelial growth factor (VEGF) is suggested to be an important regulator of angiogenesis in ovarian cancer. We have evaluated the effects of VEGF overexpression on the histology and growth rate of human ovarian cancer xenografts. OVCAR-3 human ovarian cancer cells were stably transfected with an expression vector encoding the 165-amino acid isoform of VEGF. As subcutaneous xenografts, moderately and highly VEGF(165)-overexpressing OVCAR-3 cells formed tumors with large cysts. Immunohistochemistry demonstrated an increase in the number of CD31-positive microvessels, some of which were larger in diameter than those in the parental tumors, as well as extensive vascular rimming around the cysts. Weakly VEGF(165)-overexpressing tumors also contained an increased number of CD31-positive microvessels and occasional vascular rimming, but cysts were not present. Immunohistochemistry further revealed the presence of monocytes and macrophages in both parental and VEGF(165)-overexpressing xenografts. Interestingly, the number of monocytes/macrophages was greatly increased in moderately and highly VEGF(165)-overexpressing xenografts and large areas populated with monocytes/macrophages were detected within the tumor stroma. Although the higher number of CD31-positive cells would suggest a better vascularization pattern in VEGF(165)-overexpressing xenografts, tumor growth rates were not increased when compared with that of parental xenografts. These data provide functional evidence for a role of VEGF(165) in cyst formation and monocyte/macrophage infiltration.


Subject(s)
Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Macrophages/physiology , Neovascularization, Pathologic , Ovarian Neoplasms/physiopathology , Transplantation, Heterologous , Animals , Endothelial Growth Factors/genetics , Female , Humans , Immunohistochemistry , Lymphokines/genetics , Mice , Mice, Nude , Monocytes/physiology , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...