Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Spinal Cord ; 52 Suppl 1: S9-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24902647

ABSTRACT

STUDY DESIGN/METHODS: Case report. OBJECTIVES: In this case report, a consequence of not following proper care of the bowel affecting the genitourinary system is reported and discussed. SETTING: United States. RESULTS/CONCLUSION: Neurogenic bowel and bladder can result from a spinal cord injury. It is necessary for spinal cord injury patients to continually follow recommended bladder and bowel care programs to decrease complications.


Subject(s)
Hydronephrosis/etiology , Spinal Cord Injuries/complications , Aged , Humans , Hydronephrosis/pathology , Magnetic Resonance Imaging , Male , Ultrasonography
3.
Breast Cancer Res Treat ; 132(1): 75-85, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21553120

ABSTRACT

Mammosphere culture has been used widely for the enrichment of mammary epithelial stem cells and breast cancer stem cells (CSCs). Epithelial-to-mesenchymal transition (EMT) also induces stem cell features in normal and transformed mammary cells. We examined whether mammosphere culture conditions per se induced EMT in the epithelial MCF-7 breast cancer cell line. MCF-7 cells were cultured as mammospheres for 5 weeks, with dispersal and reseeding at the end of each week. This mammosphere culture induced a complete EMT by 3 weeks. Return of the cells to standard adherent culture conditions in serum-supplemented media generated a cell population (called MCF-7(M) cells), which displays a stable mesenchymal and CSC-like CD(44+)/CD(24-/low) phenotype. EMT was accompanied by a stable, marked increase in EMT-associated transcription factors and mesenchymal markers, and a decrease in epithelial markers and estrogen receptor α (ERα). MCF-7(M) cells showed increased motility, proliferation and chemoresistance in vitro, and produced larger tumors in immunodeficient mice with or without estrogen supplementation. MicroRNA analysis showed suppression of miR-200c, miR-203, and miR-205; and increases in miR-222 and miR-221. Antisense hairpin RNA inhibitor targeting miR-221 resulted in re-expression of ERα in MCF-7(M) cells. This study provides the first example of mammosphere culture conditions inducing EMT and of EMT regulating microRNAs that target ERα.


Subject(s)
Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Spheroids, Cellular/physiology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Movement , Cell Proliferation , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Estrogen Receptor alpha/metabolism , Female , Humans , Mice , Mice, SCID , MicroRNAs/genetics , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Phenotype , Tumor Burden/genetics
4.
Oncogene ; 29(48): 6378-89, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-20729917

ABSTRACT

The v-Crk oncogene product consists of two protein interaction modules, a Src homology 2 (SH2) domain and a Src homology 3 (SH3) domain. Overexpression of CrkI, the cellular homolog of v-Crk, transforms mouse fibroblasts, and elevated CrkI expression is observed in several human cancers. The SH2 and SH3 domains of Crk are required for transformation, but the identity of the critical cellular binding partners is not known. A number of candidate Crk SH3-binding proteins have been identified, including the nonreceptor tyrosine kinases c-Abl and Arg, and the guanine nucleotide exchange proteins C3G, SOS1 and DOCK180. The aim of this study is to determine which of these are required for transformation by CrkI. We found that short hairpin RNA-mediated knockdown of C3G or SOS1 suppressed anchorage-independent growth of NIH-3T3 cells overexpressing CrkI, whereas knockdown of SOS1 alone was sufficient to suppress tumor formation by these cells in nude mice. Knockdown of C3G was sufficient to revert morphological changes induced by CrkI expression. By contrast, knockdown of Abl family kinases or their inhibition with imatinib enhanced anchorage-independent growth and tumorigenesis induced by Crk. These results show that SOS1 is essential for CrkI-induced fibroblast transformation, and also reveal a surprising negative role for Abl kinases in Crk transformation.


Subject(s)
Cell Transformation, Neoplastic , Proto-Oncogene Proteins c-crk/physiology , src Homology Domains , Animals , Apoptosis , Guanine Nucleotide-Releasing Factor 2/physiology , HEK293 Cells , Humans , Male , Mice , Mice, Nude , NIH 3T3 Cells , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/physiology , Proto-Oncogene Proteins c-crk/chemistry , SOS1 Protein/physiology , Signal Transduction
5.
Oncogene ; 25(16): 2379-92, 2006 Apr 13.
Article in English | MEDLINE | ID: mdl-16369494

ABSTRACT

The process of cancer cell invasion involves degradation of the extracellular matrix (ECM) by proteases, integrin adhesion and cell motility. The role of ECM degrading proteases on the hypoxia-induced invasion of breast carcinoma cells was investigated. Hypoxia markedly increased the invasion capacity of MDA-MB-231 and MDA-MB-435 breast carcinoma cell lines. Matrix metalloproteinase (MMP) inhibitors blocked the hypoxia-induced invasion, whereas other protease inhibitors had no effect. Antibodies or siRNAs blocking either membrane type-1 MMP (MT1-MMP) or MMP-2 were effective in reducing the hypoxia-induced invasion. Serum-free reconstitution experiments confirmed the involvement of the MT1-MMP/MMP-2/tissue inhibitor of metalloproteinase-2 complex in this hypoxia-induced response. Overexpression of MT1-MMP in a poorly invasive breast cancer cell line, T47-D, promoted hypoxia-induced invasion and MMP-2 activation. Cell surface accumulation and activation of MT1-MMP without apparent regulation at the mRNA or protein levels indicated a post-translational adaptive response to hypoxia. Inhibition of the small GTPase RhoA eliminated the hypoxia-induced invasion and blocked the localization of MT1-MMP to the plasma membrane. Zymographic and molecular analysis of human breast tumors showed a strong correlation between hypoxic microenvironments and MMP-2 activation without changes in MT1-MMP expression. Our studies suggest that hypoxic tumor microenvironments promote breast cancer invasion through an MT1-MMP-dependent mechanism.


Subject(s)
Breast Neoplasms/pathology , Cell Hypoxia , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinases/physiology , Cell Line, Tumor , Enzyme Activation , Female , Humans , Matrix Metalloproteinases, Membrane-Associated , Neoplasm Invasiveness , RNA Interference , RNA, Messenger/analysis , Tissue Inhibitor of Metalloproteinase-2/physiology , rhoA GTP-Binding Protein/physiology
6.
Growth Factors ; 19(1): 19-34, 2001.
Article in English | MEDLINE | ID: mdl-11678207

ABSTRACT

Aberrant expression of vascular endothelial growth factor (VEGF) has been demonstrated to be associated with most human solid tumors. Here we report that TGF-beta potently induces VEGF expression in human HT-1080 fibrosarcomas primarily through transcriptional activation with no significant changes in mRNA turnover. The tyrosine kinase inhibitor genistein and AP-1 inhibitor curcumin significantly blocked TGF-beta induction of VEGF expression while SP-1 and MKK1 inhibitors did not. TGF-beta enhanced both AP-1 and HIF-1 DNA binding activities whereas SP-1, AP-2 and NF-1 did not show major changes. Transcriptional reporter assays provided further evidence that TGF-beta augmented both AP-1 and HIF-1 activities. Moreover, TGF-beta-treated HT-1080 cells contained higher levels of HIF-1alpha and c-jun proteins in nuclear extracts. TGF-beta and hypoxia synergistically induced VEGF mRNA expression. Given the fact that most tumors respond to hypoxic stress with increased VEGF expression via HIF-1-dependent transcription, this study identifies for the first time that TGF-beta also increases VEGF mRNA in an AP-l/HIF-1-dependent mechanism and may potentiate the hypoxic response.


Subject(s)
DNA-Binding Proteins/metabolism , Endothelial Growth Factors/biosynthesis , Lymphokines/biosynthesis , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta/pharmacology , Blotting, Western , Cell Hypoxia , Endothelial Growth Factors/genetics , Enzyme Inhibitors/pharmacology , Genes, Reporter , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Lymphokines/genetics , Proto-Oncogene Proteins c-jun/analysis , RNA, Messenger/metabolism , Transcription Factors/analysis , Transcription Factors/metabolism , Transcriptional Activation , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
Nat Med ; 7(2): 192-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11175850

ABSTRACT

Metastasis of breast cancer occurs primarily through the lymphatic system, and the extent of lymph node involvement is a key prognostic factor for the disease. Whereas the significance of angiogenesis for tumor progression has been well documented, the ability of tumor cells to induce the growth of lymphatic vessels (lymphangiogenesis) and the presence of intratumoral lymphatic vessels have been controversial. Using a novel marker for lymphatic endothelium, LYVE-1, we demonstrate here the occurrence of intratumoral lymphangiogenesis within human breast cancers after orthotopic transplantation onto nude mice. Vascular endothelial growth factor (VEGF)-C overexpression in breast cancer cells potently increased intratumoral lymphangiogenesis, resulting in significantly enhanced metastasis to regional lymph nodes and to lungs. The degree of tumor lymphangiogenesis was highly correlated with the extent of lymph node and lung metastases. These results establish the occurrence and biological significance of intratumoral lymphangiogenesis in breast cancer and identify VEGF-C as a molecular link between tumor lymphangiogenesis and metastasis.


Subject(s)
Breast Neoplasms/pathology , Endothelial Growth Factors/physiology , Neovascularization, Pathologic , Animals , Endothelial Growth Factors/genetics , Female , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymph Nodes , Lymphatic Metastasis , Mice , Mice, Nude , Neoplasm Metastasis , Tumor Cells, Cultured , Vascular Endothelial Growth Factor C
8.
Lab Invest ; 81(1): 61-75, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11204275

ABSTRACT

Angiogenesis is a key component of human cancer progression and metastasis. In an effort to recapitulate early events in tumor-induced angiogenesis, we have employed a subcutaneous Matrigel implant model using immunodeficient mice as hosts. Matrigel-containing fibroblast growth factor 2 (FGF-2; 1.2 microg/ml) induced stromal cell infiltration into the Matrigel/skin interface within 4 days and maximal neovascularization at 7 days. Cells staining positive for the endothelial cell marker, platelet-endothelial cell adhesion molecule 1 (PECAM-1), were present in neovessels and in isolated cells within the Matrigel matrix. Immunohistochemical analysis revealed high levels of vascular endothelial growth factor (VEGF) deposited in the stromal interface present only in the FGF-2-containing but not in control Matrigel implants. VEGF expression was confirmed with in situ hybridization. High VEGF mRNA levels were observed in the infiltrating stromal cells but not in endothelial or endothelial precursors as defined by PECAM-1 staining. In vitro analysis of FGF-2-treated embryonic fibroblasts, Balb/c 3T3 cells, showed an induction of VEGF transcription, mRNA synthesis, and protein secretion as defined by transcriptional reporter, Northern blot, and ELISA assays. The FGF-2-induced VEGF expression was not dependent on select matrix adherence or signaling components because VEGF mRNA expression induced by FGF-2 was equally activated on serum, basement membrane, and fibronectin matrix substrates. Systemic application of anti-VEGF antibodies significantly repressed FGF-2-induced angiogenesis over control antibody by 88% (p < 0.001). These data support an FGF-2 angiogenic model that is dependent on endothelial cell activation, stromal cell infiltration, and VEGF expression by the infiltrating stromal cell population.


Subject(s)
Endothelial Growth Factors/metabolism , Fibroblast Growth Factor 2/pharmacology , Lymphokines/metabolism , Neovascularization, Pathologic , Stromal Cells/drug effects , Stromal Cells/metabolism , 3T3 Cells , Animals , Endothelial Growth Factors/genetics , Female , Gene Expression/drug effects , Humans , Lymphokines/genetics , Mice , Mice, Nude , Neoplasms/blood supply , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
FASEB J ; 14(15): 2589-600, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11099478

ABSTRACT

We previously reported that the binding of two-chain high molecular weight kininogen (HKa) to endothelial cells may occur through interactions with endothelial urokinase receptors. Since the binding of urokinase to urokinase receptors activates signaling responses and may stimulate mitogenesis, we assessed the effect of HKa binding on endothelial cell proliferation. Unexpectedly, HKa inhibited proliferation in response to several growth factors, with 50% inhibition caused by approximately 10 nM HKa. This activity was Zn(2+) dependent and not shared by either single-chain high molecular weight kininogen (HK) or low molecular weight kininogen. HKa selectively inhibited the proliferation of human umbilical vein and dermal microvascular endothelial cells, but did not affect that of umbilical vein or human aortic smooth muscle cells, trophoblasts, fibroblasts, or carcinoma cells. Inhibition of endothelial proliferation by HKa was associated with endothelial cell apoptosis and unaffected by antibodies that block the binding of HK or HKa to any of their known endothelial receptors. Recombinant HK domain 5 displayed activity similar to that of HKa. In vivo, HKa inhibited neovascularization of subcutaneously implanted Matrigel plugs, as well as rat corneal angiogenesis. These results demonstrate that HKa is a novel inhibitor of angiogenesis, whose activity is dependent on the unique conformation of the two-chain molecule.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis , Endothelium, Vascular/drug effects , Kininogen, High-Molecular-Weight/pharmacology , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Carcinoma , Cornea/blood supply , Fibroblasts/drug effects , Humans , Molecular Sequence Data , Muscle, Smooth, Vascular/drug effects , Rats , Trophoblasts/drug effects , Tumor Cells, Cultured/drug effects
10.
Am J Pathol ; 156(1): 159-67, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623663

ABSTRACT

Inhibition of the vascular endothelial growth factor (VEGF) receptor Flk-1 has been shown to prevent invasion of experimental squamous cell carcinomas (SCC). To directly investigate the role of VEGF in tumor invasion, we stably transfected human SCC-13 cells, which are characterized by a noninvasive phenotype in vivo, with expression vectors containing murine VEGF(164) in sense (SCC/VEGF+) or antisense (SCC/VEGF-) orientation or with vector alone (SCC/vec). SCC/vec cells formed slowly growing, well-differentiated tumors with well-defined borders between tumor and stroma, after intradermal or subcutaneous injection. In contrast, SCC/VEGF+ tumors were characterized by rapid tumor growth, with small cell groups and single cells invading into the surrounding tissue, and by admixture of blood vessels and tumor cells in areas of tumor invasion. We detected an increase in tumor vessel density and size in VEGF-overexpressing tumors, resulting in a more than fourfold increase in total vascular areas. In contrast, SCC/VEGF- clones formed noninvasive, sharply circumscribed tumors with reduced vascular density. These findings demonstrate that selective VEGF overexpression was sufficient to induce tumor invasiveness, and they provide further evidence for an active role of the tumor stroma in cancer progression.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Animals , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Cell Division , DNA, Complementary/genetics , Endothelial Growth Factors/genetics , Humans , Immune System Diseases/genetics , Lymphokines/genetics , Mice , Mice, Inbred BALB C/genetics , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Oligonucleotides/pharmacology , Oligonucleotides, Antisense/pharmacology , Phenotype , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Cell ; 99(3): 301-12, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10555146

ABSTRACT

Vascular endothelial cells undergo morphogenesis into capillary networks in response to angiogenic factors. We show here that sphingosine-1-phosphate (SPP), a platelet-derived bioactive lipid, activates the EDG-1 and -3 subtypes of G protein-coupled receptors on endothelial cells to regulate angiogenesis. SPP induces the Gi/mitogen-activated protein kinase/cell survival pathway and the small GTPase Rho- and Raccoupled adherens junction assembly. Both EDG-1-and EDG-3-regulated signaling pathways are required for endothelial cell morphogenesis into capillary-like networks. Indeed, SPP synergized with polypeptide angiogenic growth factors in the formation of mature neovessels in vivo. These data define SPP as a novel regulator of angiogenesis.


Subject(s)
Cadherins/physiology , Cell Adhesion/physiology , Endothelium, Vascular/physiology , I-kappa B Proteins , Intercellular Junctions/physiology , Lysophospholipids , Neovascularization, Physiologic/physiology , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Animals , Antigens, CD , Cadherins/analysis , Calcium/metabolism , Cell Adhesion/drug effects , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Endothelium, Vascular/drug effects , Female , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Intercellular Junctions/drug effects , Mice , Mice, Nude , Models, Biological , Morphogenesis/drug effects , NF-KappaB Inhibitor alpha , Neovascularization, Physiologic/drug effects , Oocytes/physiology , Receptors, Cell Surface/physiology , Receptors, Lysophospholipid , Recombinant Proteins/metabolism , Sphingosine/pharmacology , Umbilical Veins , Xenopus laevis
12.
Cancer Res ; 59(20): 5392-7, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10537325

ABSTRACT

Gangliosides are sialated glycosphingolipids present on the plasma membranes of all vertebrate cells. Tumors shed gangliosides into the extracellular microenvironment, which may influence tumor-host cell interactions. We have investigated the role of gangliosides on the growth and angiogenesis of the EPEN experimental mouse brain tumor. EPEN cells express only ganglioside G(M3), and the solid tumors formed in vivo are sparsely vascularized with extensive necrosis. We stably transfected the EPEN cells with the cDNA for N-acetylgalactosaminyl transferase, a key enzyme for the synthesis of complex gangliosides. In addition to G(M3), the transfected cell line (EPEN-GNT) expressed complex gangliosides G(M2), G(M1), and G(D1a). The EPEN-GNT tumor was more densely vascularized with less necrosis and grew more rapidly than the nontransfected EPEN or mock-transfected (EPEN-V) control tumors. Also, VEGF gene expression was higher in the EPEN-GNT tumor than in the control tumors. The synthesis of complex gangliosides in the EPEN-GNT tumor cells also stimulated vascularization in an in vivo Matrigel assay for angiogenesis. These results indicate that the ratio of G(M3) to complex gangliosides can influence the growth and angiogenic properties of the EPEN experimental brain tumor and are consistent with previous findings in other systems. We conclude that gangliosides may be important modulators of brain tumor angiogenesis.


Subject(s)
Brain Neoplasms/blood supply , Gangliosides/physiology , Neovascularization, Pathologic/etiology , Animals , Endothelial Growth Factors/genetics , Gangliosides/genetics , Gene Expression Regulation, Neoplastic , Lymphokines/genetics , Male , Mice , Mice, Inbred C57BL , Mitotic Index , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
J Biol Chem ; 274(22): 15407-14, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10336429

ABSTRACT

Aberrant expression of the potent angiogenic cytokine, vascular endothelial growth factor (VEGF), has been demonstrated to be associated with most human solid tumors. Both transcriptional and post-transcriptional mechanisms have been shown to modulate VEGF expression in a multitude of cell types. Here we report that when protein kinase C (PKC) pathways were activated in human glioblastoma U373 cells by phorbol 12-myristate 13-acetate (PMA), VEGF mRNA expression was up-regulated via a post-transcriptional mRNA stabilization mechanism. PMA treatment exhibited no increase in VEGF-specific transcriptional activation as determined by run-off transcription assays and VEGF promoter-luciferase reporter assays. However, PMA increased VEGF mRNA half-life from 0.8 to 3.6 h which was blocked by PKC inhibitors but not by protein kinase A or cyclic nucleotide-dependent protein kinase inhibitors. When U373 cells were transfected with antisense oligonucleotide sequences to the translation start sites of PKC-alpha, -beta, -gamma, -delta, -epsilon, or -zeta isoforms, both PKC-alpha and -zeta antisense oligonucleotides showed substantial inhibition of PMA-induced VEGF mRNA. In addition, overexpression of PKC-zeta resulted in a strong constitutive up-regulation of VEGF mRNA expression. This study demonstrates for the first time that specific PKC isoforms regulate VEGF mRNA expression through post-transcriptional mechanisms.


Subject(s)
Endothelial Growth Factors/genetics , Isoenzymes/metabolism , Lymphokines/genetics , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Glioblastoma/genetics , Humans , Oligonucleotides, Antisense/pharmacology , Protein Kinase C/antagonists & inhibitors , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Staurosporine/pharmacology , Transcriptional Activation , Transfection , Tumor Cells, Cultured , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Am J Physiol ; 276(5): E849-55, 1999 05.
Article in English | MEDLINE | ID: mdl-10329978

ABSTRACT

Physiological stressors such as sepsis and tissue damage initiate an acute immune response and cause transient systemic insulin resistance. This study was conducted to determine whether tumor necrosis factor-alpha (TNF-alpha), a cytokine produced by immune cells during skeletal muscle damage, decreases insulin responsiveness at the cellular level. To examine the molecular mechanisms associated with TNF-alpha and insulin action, we measured insulin receptor substrate (IRS)-1- and IRS-2-mediated phosphatidylinositol 3-kinase (PI 3-kinase) activation, IRS-1-PI 3-kinase binding, IRS-1 tyrosine phosphorylation, and the phosphorylation of two mitogen-activated protein kinases (MAPK, known as p42(MAPK) and p44(MAPK)) in cultured C2C12 myotubes. Furthermore, we determined the effects of TNF-alpha on insulin-stimulated 2-deoxyglucose (2-DG) uptake. We observed that TNF-alpha impaired insulin stimulation of IRS-1- and IRS-2-mediated PI 3-kinase activation by 54 and 55% (P < 0.05), respectively. In addition, TNF-alpha decreased insulin-stimulated IRS-1 tyrosine phosphorylation by 40% (P < 0.05). Furthermore, TNF-alpha repressed insulin-induced p42(MAPK) and p44(MAPK) tyrosine phosphorylation by 81% (P < 0.01). TNF-alpha impairment of insulin signaling activation was accompanied by a decrease (P < 0.05) in 2-DG uptake in the muscle cells (60 +/- 4 vs. 44 +/- 6 pmol. min-1. mg-1). These data suggest that increases in TNF-alpha may cause insulin resistance in skeletal muscle by inhibiting IRS-1- and IRS-2-mediated PI 3-kinase activation as well as p42(MAPK) and p44(MAPK) tyrosine phosphorylation, leading to impaired insulin-stimulated glucose uptake.


Subject(s)
Glucose/metabolism , Insulin/pharmacology , Mitogen-Activated Protein Kinases , Muscles/drug effects , Muscles/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Deoxyglucose/metabolism , Enzyme Activation , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphoproteins/pharmacology , Phosphorylation , Phosphotyrosine/metabolism
15.
J Biol Chem ; 274(3): 1359-65, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9880507

ABSTRACT

A 126-base region of human vascular endothelial growth factor (VEGF) 3'-untranslated region, which we identified as the hypoxia stability region, forms seven hypoxia-inducible RNA-protein complexes with apparent molecular masses ranging from 40 to 90 kDa in RNA-UV-cross-linking assays. In this study, we show that proteins that form the 60-kDa RNA-protein complex with the hypoxia stability region were present in both cytoplasmic and nuclear compartments. We purified the protein associated in the 60-kDa complex and identified it as heterogeneous nuclear ribonucleoprotein L (hnRNP L) by protein sequencing. Removal of hnRNP L by immunoprecipitation specifically abolished formation of the 60-kDa complex. Synthetic deoxyribonucleotide competition studies defined the RNA-binding site of hnRNP L as a 21-base-long sequence, 5'-CACCCACCCACAUACAUACAU-3'. Immunoprecipitation of hnRNP L followed by reverse transcription-polymerase chain reaction showed that hnRNP L specifically interacts with VEGF mRNA in hypoxic cells in vivo. Furthermore, when M21 cells transfected with antisense oligodeoxyribonucleotide to the hnRNP L RNA-binding site, the VEGF mRNA half-life was significantly reduced under hypoxic conditions. Thus, we propose that specific association of hnRNP L with VEGF mRNA under hypoxia may play an important role in hypoxia-induced post-transcriptional regulation of VEGF mRNA expression.


Subject(s)
Endothelial Growth Factors/genetics , Hypoxia/metabolism , Lymphokines/genetics , RNA, Heterogeneous Nuclear/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Base Sequence , Binding Sites , Electrophoresis, Polyacrylamide Gel , Heterogeneous-Nuclear Ribonucleoprotein L , Heterogeneous-Nuclear Ribonucleoproteins , Humans , Molecular Sequence Data , Molecular Weight , Oligodeoxyribonucleotides, Antisense/metabolism , RNA Processing, Post-Transcriptional , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Clin Exp Metastasis ; 17(8): 687-94, 1999.
Article in English | MEDLINE | ID: mdl-10919714

ABSTRACT

Upregulation of vascular endothelial growth factor (VEGF) expression induced by hypoxia is crucial event leading to neovascularization. Cyclooxygenase-2, an inducible enzyme that catalyzes the formation of prostaglandins (PGs) from arachidonic acid, has been demonstrated to be induced by hypoxia and play role in angiogenesis and metastasis. To investigate the potential effect of COX-2 on hypoxia-induced VEGF expression in prostate cancer. We examined the relationship between COX-2 expression and VEGF induction in response to cobalt chloride (CoCl2)-simulated hypoxia in three human prostate cancer cell lines with differing biological phenotypes. Northern blotting and ELISA revealed that all three tested cell lines constitutively expressed VEGF mRNA, and secreted VEGF protein to different degrees (LNCaP > PC-3 > PC3ML). However, these cell lines differed in the ability to produce VEGF in the presence of CoCl2-simulated hypoxia. CoCl2 treatment resulted in 40% and 75% increases in VEGF mRNA, and 50% and 95% in protein secretion by LNCaP and PC-3 cell lines, respectively. In contrast, PC-3ML cell line, a PC-3 subline with highly invasive, metastatic phenotype, exhibits a dramatic upregulation of VEGF, 5.6-fold in mRNA and 6.3-fold in protein secretion after treatment with CoCl2. The upregulation of VEGF in PC-3ML cells is accompanied by a persistent induction of COX-2 mRNA (6.5-fold) and protein (5-fold). Whereas COX-2 expression is only transiently induced in PC-3 cells and not affected by CoCl2 in LNCaP cells. Moreover, the increases in VEGF mRNA and protein secretion induced by CoCl2 in PC-3ML cells were significantly suppressed following exposure to NS398, a selective COX-2 inhibitor. Finally, the effect of COX-2 inhibition on CoCl2-induced VEGF production was reversed by the treatment with exogenous PGE2. Our data demonstrate that VEGF induction by cobalt chloride-simulated hypoxia is maintained by a concomitant, persistent induction of COX-2 expression and sustained elevation of PGE2 synthesis in a human metastatic prostate cancer cell line, and suggest that COX-2 activity, reflected by PGE2 production, is involved in hypoxia-induced VEGF expression, and thus, modulates prostatic tumor angiogenesis.


Subject(s)
Cobalt/pharmacology , Endothelial Growth Factors/biosynthesis , Isoenzymes/biosynthesis , Isoenzymes/pharmacology , Lymphokines/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/pharmacology , Prostatic Neoplasms/metabolism , Androgens/physiology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/biosynthesis , Dinoprostone/pharmacology , Drug Interactions , Enzyme Induction/drug effects , Humans , Male , Membrane Proteins , Neoplasm Metastasis , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Nitrobenzenes/pharmacology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Stimulation, Chemical , Sulfonamides/pharmacology , Tumor Cells, Cultured/drug effects , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
J Biol Chem ; 273(41): 26277-80, 1998 Oct 09.
Article in English | MEDLINE | ID: mdl-9756852

ABSTRACT

The transcription factor Sp1 is ubiquitously expressed and plays a significant role in the constitutive and induced expression of a variety of mammalian genes and may even contribute to tumorigenesis. Here, we describe a novel pathway whereby Sp1 promotes the transcription of vascular permeability factor/vascular endothelial growth factor (VPF/VEGF), a potent angiogenic factor, by interacting directly and specifically with protein kinase C zeta (PKC zeta) isoform in renal cell carcinoma. PKC zeta binds and phosphorylates the zinc finger region of Sp1. Moreover, in the presence of the wild type von Hippel-Lindau gene product, the interaction of Sp1 with PKC zeta is inhibited, and in this manner steady state levels of Sp1 phosphorylation are decreased significantly. Co-transfection of renal cell carcinoma cells and human fibrosarcoma cells with a plasmid overexpressing PKC zeta and VPF/VEGF promoter luciferase constructs results in activation of Sp1-mediated transcription, whereas expression of a dominant-negative mutant of PKC zeta repressed this activation. Taken together, our results suggest a new pathway of cell signaling through PKC zeta and provide an insight into PKC zeta and Sp1-dependent transcriptional regulation of VPF/VEGF expression and thus tumor angiogenesis.


Subject(s)
Endothelial Growth Factors/genetics , Ligases , Lymphokines/genetics , Protein Kinase C/metabolism , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Fibrosarcoma/enzymology , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Von Hippel-Lindau Tumor Suppressor Protein
18.
J Exp Med ; 188(6): 1135-45, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9743532

ABSTRACT

Vascular permeability factor/vascular endothelial cell growth factor (VPF/VEGF) can both potently enhance vascular permeability and induce proliferation of vascular endothelial cells. We report here that mouse or human mast cells can produce and secrete VPF/VEGF. Mouse mast cells release VPF/VEGF upon stimulation through Fcepsilon receptor I (FcepsilonRI) or c-kit, or after challenge with the protein kinase C activator, phorbol myristate acetate, or the calcium ionophore, A23187; such mast cells can rapidly release VPF/VEGF, apparently from a preformed pool, and can then sustain release by secreting newly synthesized protein. Notably, the Fc epsilonRI-dependent secretion of VPF/VEGF by either mouse or human mast cells can be significantly increased in cells which have undergone upregulation of Fc epsilonRI surface expression by a 4-d preincubation with immunoglobulin E. These findings establish that at least one cell type, the mast cell, can be stimulated to secrete VPF/VEGF upon immunologically specific activation via a member of the multichain immune recognition receptor family. Our observations also identify a new mechanism by which mast cells can contribute to enhanced vascular permeability and/or angiogenesis, in both allergic diseases and other settings.


Subject(s)
Endothelial Growth Factors/metabolism , Immunoglobulin E/physiology , Lymphokines/metabolism , Mast Cells/metabolism , Receptors, IgE/biosynthesis , Up-Regulation/immunology , Animals , Calcimycin/pharmacology , Cell Line , Cells, Cultured , Dinitrophenols/immunology , Dinitrophenols/pharmacology , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Female , Haptens/pharmacology , Humans , Lymphokines/biosynthesis , Lymphokines/genetics , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, IgE/physiology , Serum Albumin/immunology , Serum Albumin/pharmacology , Stem Cell Factor/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/immunology , Umbilical Cord/cytology , Umbilical Cord/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Cancer Res ; 58(18): 4185-92, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9751633

ABSTRACT

Vascular endothelial growth factor (VEGF), also known as vascular permeability factor, has been investigated as a potent mediator of brain tumor angiogenesis and tumor growth. We evaluated the effect of VEGF expression on the pathophysiology of tumor growth in the brain. Human SK-MEL-2 melanoma cells, with minimal VEGF expression, were stably transfected with either sense or antisense mouse VEGF cDNA and used to produce intracerebral xenografts. Vascular permeability, blood volume, blood flow, and tumor fluorodeoxyglucose metabolism were assessed using tissue sampling and quantitative autoradiography. Tumor proliferation was assessed by measuring bromodeoxyuridine labeling indices. Tumor vascular density and morphological status of the blood-brain barrier were evaluated by immunohistochemistry. SK-MEL-2 cells transfected with sense VEGF (V+) expressed large amounts of mouse and human VEGF protein; V+ cells formed well-vascularized, rapidly growing tumors with minimal tumor necrosis. V+ tumors had substantial and significant increases in blood volume, blood flow, vascular permeability, and fluorodeoxyglucose metabolism compared to wild-type and/or V- (antisense VEGF) tumors. VEGF antisense transfected V- expressed no detectable VEGF protein and formed minimally vascularized tumors. V- tumors had a very low initial growth rate with central necrosis; blood volume, blood flow, vascular permeability, and glucose metabolism levels were low compared to wild-type and V+ tumors. A substantial inhibition of intracerebral tumor growth, as well as a decrease in tumor vascularity, blood flow, and vascular permeability may be achieved by down-regulation of endogenous VEGF expression in tumor tissue. VEGF-targeted antiangiogenic gene therapy could be an effective component of a combined strategy to treat VEGF-producing brain tumors.


Subject(s)
Brain Neoplasms/blood supply , Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Melanoma/blood supply , Neoplasm Proteins/metabolism , Neovascularization, Pathologic , Animals , Blood Volume , Brain Neoplasms/metabolism , Capillary Permeability , Cerebrovascular Circulation , Endothelial Growth Factors/genetics , Enzyme-Linked Immunosorbent Assay , Fluorodeoxyglucose F18/metabolism , Humans , Lymphokines/genetics , Melanoma/metabolism , Mice , Neoplasm Proteins/genetics , RNA, Antisense/metabolism , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
J Invest Dermatol ; 111(1): 1-6, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9665379

ABSTRACT

Vascular endothelial growth factor (VEGF) has been implicated in the pathologic angiogenesis observed in psoriasis and other chronic inflammatory skin diseases that are characterized by enhanced expression of VEGF by epidermal keratinocytes and of VEGF receptors by tortuous microvessels in the upper dermis. To investigate the functional importance of chronic VEGF overexpression in vivo, we used a keratin 14 promoter expression cassette containing the gene for murine VEGF164 to selectively target VEGF expression to basal epidermal keratinocytes in transgenic mice. These mice demonstrated an increased density of tortuous cutaneous blood capillaries with elevated expression levels of the high affinity VEGF receptors, VEGFR-1 and VEGFR-2, most prominently during the neonatal period. In contrast, no abnormalities of lymphatic vessels were detected. In addition, the number of mast cells in the upper dermis was significantly increased in transgenic skin. Intravital fluorescence microscopy revealed highly increased leukocyte rolling and adhesion in postcapillary skin venules that were both inhibited after injection of blocking antibodies against E- and P-selectin. Combined blocking antibodies against intercellular adhesion molecule-1 and lymphocyte function-associated antigen-1 were without effect, whereas an anti-vascular cell adhesion molecule-1/VLA-4 antibody combination almost completely normalized the enhanced leukocyte adhesion in transgenic mice. This study reveals VEGF as a growth factor specific for blood vessels, but not lymphatic vessels, and demonstrates that chronic orthotopic overexpression of VEGF in the epidermis is sufficient to induce cardinal features of chronic skin inflammation, providing a molecular link between angiogenesis, mast cell accumulation, and leukocyte recruitment to sites of inflammation.


Subject(s)
Endothelial Growth Factors/physiology , Leukocytes/physiology , Lymphokines/physiology , Skin/blood supply , Animals , Cell Adhesion , Cell Movement , Endothelial Growth Factors/genetics , Lymphokines/genetics , Mast Cells/physiology , Mice , Mice, Transgenic , Microcirculation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...