Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 20(11): 1532-45, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23933817

ABSTRACT

Monopolar spindle 1 (MPS1), a mitotic kinase that is overexpressed in several human cancers, contributes to the alignment of chromosomes to the metaphase plate as well as to the execution of the spindle assembly checkpoint (SAC). Here, we report the identification and functional characterization of three novel inhibitors of MPS1 of two independent structural classes, N-(4-{2-[(2-cyanophenyl)amino][1,2,4]triazolo[1,5-a]pyridin-6-yl}phenyl)-2-phenylacetamide (Mps-BAY1) (a triazolopyridine), N-cyclopropyl-4-{8-[(2-methylpropyl)amino]-6-(quinolin-5-yl)imidazo[1,2-a]pyrazin-3-yl}benzamide (Mps-BAY2a) and N-cyclopropyl-4-{8-(isobutylamino)imidazo[1,2-a]pyrazin-3-yl}benzamide (Mps-BAY2b) (two imidazopyrazines). By selectively inactivating MPS1, these small inhibitors can arrest the proliferation of cancer cells, causing their polyploidization and/or their demise. Cancer cells treated with Mps-BAY1 or Mps-BAY2a manifested multiple signs of mitotic perturbation including inefficient chromosomal congression during metaphase, unscheduled SAC inactivation and severe anaphase defects. Videomicroscopic cell fate profiling of histone 2B-green fluorescent protein-expressing cells revealed the capacity of MPS1 inhibitors to subvert the correct timing of mitosis as they induce a premature anaphase entry in the context of misaligned metaphase plates. Hence, in the presence of MPS1 inhibitors, cells either divided in a bipolar (but often asymmetric) manner or entered one or more rounds of abortive mitoses, generating gross aneuploidy and polyploidy, respectively. In both cases, cells ultimately succumbed to the mitotic catastrophe-induced activation of the mitochondrial pathway of apoptosis. Of note, low doses of MPS1 inhibitors and paclitaxel (a microtubular poison) synergized at increasing the frequency of chromosome misalignments and missegregations in the context of SAC inactivation. This resulted in massive polyploidization followed by the activation of mitotic catastrophe. A synergistic interaction between paclitaxel and MPS1 inhibitors could also be demonstrated in vivo, as the combination of these agents efficiently reduced the growth of tumor xenografts and exerted superior antineoplastic effects compared with either compound employed alone. Altogether, these results suggest that MPS1 inhibitors may exert robust anticancer activity, either as standalone therapeutic interventions or combined with microtubule-targeting chemicals.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Drug Synergism , Female , HeLa Cells , Humans , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/pathology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Random Allocation , Transfection , Xenograft Model Antitumor Assays
2.
Gut ; 58(2): 261-70, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18829975

ABSTRACT

BACKGROUND AND AIMS: Current systemic therapies for neuroendocrine tumours (NETs) do not provide sufficient control of tumour growth. However, efficient evaluation of novel drugs is hindered by the lack of a suitable preclinical animal model. Here an orthotopic mouse model of pancreatic NET is established and used to study the action of ZK 304709, a first in class, oral multitarget tumour growth inhibitor. ZK 304709 is an inhibitor of cyclin-dependent kinases (Cdks) 1, 2, 4, 7 and 9, vascular endothelial growth factor receptor-type kinases (VEGF-RTKs) 1-3 and platelet-derived growth factor receptor-type kinase beta (PDGF-RTKss). METHODS: BON and QGP-1 human NET cells were used to study proliferation, survival and cell cycle distribution in vitro. For induction of orthotopic NETs, BON cells were injected into the pancreas of NMRI(nu/nu) mice. Primary tumour growth and metastatic spread were recorded after 9 weeks, and apoptosis, microvessel density and lymphatic vessel density were determined. RESULTS: ZK 304709 dose-dependently suppressed proliferation and colony formation of NET cells. Direct effects on NET cells were consistent with Cdk inhibition and involved G(2) cell cycle arrest and apoptosis induction, which was associated with reduced expression of MCL1 (myeloid cell leukaemia sequence 1), survivin and hypoxia-inducible factor 1alpha (HIF1alpha). Apoptosis similarly occurred in vivo in ZK 304709-treated orthotopic BON tumours, resulting in a 80% reduction of primary tumour growth. In contrast, treatment with lanreotide or 5-fluorouracil and streptozotocin failed to inhibit tumour gowth. ZK 304709 also reduced tumour microvessel density, implicating antiangiogenic mechanisms. CONCLUSION: BON orthotopic tumours provide an informative model for preclinical drug evaluation in NETs. In this model, ZK 304709 achieved efficacious tumour growth control via induction of apoptosis and inhibition of tumour-induced angiogenesis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Biomarkers/analysis , Cell Nucleus/chemistry , Dose-Response Relationship, Drug , Female , Flow Cytometry , Fluorescent Antibody Technique , Histocytochemistry , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Inhibitor of Apoptosis Proteins , Lymphatic Metastasis , Mice , Mice, Nude , Microtubule-Associated Proteins/analysis , Neovascularization, Pathologic/drug therapy , Neuroendocrine Tumors/blood supply , Pancreatic Neoplasms/blood supply , Survivin , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
3.
Biomed Pharmacother ; 60(6): 269-72, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16887322

ABSTRACT

Loss of cell cycle control and tumor-induced neovascularization are major drivers of human tumor growth. The multi-target tumor growth inhibitor ZK 304709 is a nanomolar inhibitor of cyclin-dependent kinases 1, 2, 4, 7 and 9, as well as vascular endothelial growth factor receptor tyrosine kinase 1-3 and of platelet-derived growth factor receptor beta tyrosine kinase. The multi-targeted mode of action of ZK 304709 acting on cell cycle and angiogenesis resulted in superior efficacy compared to standard chemotherapeutic compounds both in s.c. human tumor xenografts as well as orthotopic human pancreatic carcinoma models.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Transplantation, Heterologous
4.
Angiogenesis ; 4(2): 123-31, 2001.
Article in English | MEDLINE | ID: mdl-11806244

ABSTRACT

The transmembrane tyrosine kinase TIE-2, the receptor for the angiopoietins-1 and -2, has been shown to be involved in angiogenic processes. Investigating the regulation of TIE-2 expression on endothelial cells, we found that stimulators such as PMA induce a decrease of TIE-2 protein from the cell surface without affecting TIE-2 mRNA. In conditioned media of PMA stimulated endothelial cells, a soluble form of this receptor comprising parts of the extracellular domain can be detected. Using a sandwich ELISA, we were able to detect and quantify TIE-2 receptors in cell lysates (representing the whole transmembrane receptor) and in cell culture supernatants (representing a soluble form of this receptor, sTIE-2). Several factors influencing the shedding process e.g. basic FGF could be identified. Finally, the soluble form of TIE-2 could also be detected in human biological fluids such as sera and plasma from healthy controls.


Subject(s)
Endothelium, Vascular/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins , Animals , Base Sequence , Culture Media, Conditioned , DNA Primers , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Proteins/blood , Precipitin Tests , Receptor, TIE-2 , Tetradecanoylphorbol Acetate/pharmacology
5.
Biochim Biophys Acta ; 1466(1-2): 71-8, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10825432

ABSTRACT

Immunoliposomes (IL) containing anti-angiogenic drugs directed selectively to the easily accessible kinase insert domain containing receptor (KDR) vascular endothelial growth factor (VEGF), which is predominantly expressed on tumour vessels are a promising tool to inhibit tumour angiogenesis. To explore this strategy, we have prepared fluorescent-labelled IL presenting antibodies against the KDR receptor (3G2) on their surface. 3G2-IL were composed of egg phosphatidylcholine and cholesterol (6:4), containing 2 mol% of the new thiol reactive linker lipid O-(3-cholesteryloxycarbonyl)propionyl-O'-m-maleimido-benzoyl tetraethylene glycol. Specific binding of 3G2-IL to immobilised recombinant KDR was used to show the maintenance of sufficient immunoreactivity of 3G2 antibodies upon the coupling procedure. 3G2-IL bound to Chinese hamster ovarian (CHO) cells stably transfected to overexpress KDR to a five times higher amount as compared to mock-transfected CHO cells. Subsequently, specific binding of 3G2-IL to KDR could also be demonstrated on KDR expressing cells, human umbilical vein endothelial cells and human microvascular endothelial cells, whereas only low binding of 3G2-IL to NIH-3T3 mouse fibroblast cells, which do not express KDR, was found. The binding of 3G2-IL to KDR receptors could not be blocked by VEGF, suggesting that the binding site for VEGF is not identical with the epitope recognised by 3G2. We could demonstrate that 3G2-IL is able to bind in vitro even in the presence of high levels of VEGF.


Subject(s)
Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , 3T3 Cells , Animals , CHO Cells , Cells, Cultured , Cricetinae , Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Fluoresceins , Humans , Liposomes , Lymphokines/metabolism , Mice , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/genetics , Receptors, Growth Factor/immunology , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
Cancer Res ; 60(8): 2178-89, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10786682

ABSTRACT

PTK787/ZK 222584 (1-[4-chloroanilino]-4-[4-pyridylmethyl] phthalazine succinate) is a potent inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinases, active in the submicromolar range. It also inhibits other class III kinases, such as the platelet-derived growth factor (PDGF) receptor beta tyrosine kinase, c-Kit, and c-Fms, but at higher concentrations. It is not active against kinases from other receptor families, such as epidermal growth factor receptor, fibroblast growth factor receptor-1, c-Met, and Tie-2, or intracellular kinases such as c-Src, c-Abl, and protein kinase C-alpha. PTK787/ZK 222584 inhibits VEGF-induced autophosphorylation of kinase insert domain-containing receptor (KDR), endothelial cell proliferation, migration, and survival in the nanomolar range in cell-based assays. In concentrations up to 1 microM, PTK787/ZK 222584 does not have any cytotoxic or antiproliferative effect on cells that do not express VEGF receptors. After oral dosing (50 mg/kg) to mice, plasma concentrations of PTK787/ZK 222584 remain above 1 microM for more than 8 h. PTK787/ZK 222584 induces dose-dependent inhibition of VEGF and PDGF-induced angiogenesis in a growth factor implant model, as well as a tumor cell-driven angiogenesis model after once-daily oral dosing (25-100 mg/kg). In the same dose range, it also inhibits the growth of several human carcinomas, grown s.c. in nude mice, as well as a murine renal carcinoma and its metastases in a syngeneic, orthotopic model. Histological examination of tumors revealed inhibition of microvessel formation in the interior of the tumor. PTK787/ZK 222584 is very well tolerated and does not impair wound healing. It also does not have any significant effects on circulating blood cells or bone marrow leukocytes as a single agent or impair hematopoetic recovery after concomitant cytotoxic anti-cancer agent challenge. This novel compound has therapeutic potential for the treatment of solid tumors and other diseases where angiogenesis plays an important role.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma/drug therapy , Carcinoma/pathology , Endothelial Growth Factors/antagonists & inhibitors , Lymphokines/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Phthalazines , Pyridines , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/blood , Angiogenesis Inhibitors/therapeutic use , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Carcinoma/blood supply , Cell Division/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Hematopoiesis/drug effects , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Leukocytes/cytology , Leukocytes/drug effects , Lymphokines/pharmacology , Mice , Mice, Nude , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Phosphorylation/drug effects , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Wound Healing/drug effects
7.
Histochem Cell Biol ; 114(5): 373-85, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11151407

ABSTRACT

We have used the avian chorioallantoic membrane (CAM) to study the interaction of tumor cells with the lymphatics in vivo. The vascular endothelial growth factor-C (VEGF-C) has been shown to be lymphangiogenic. We have therefore grown VEGF-C-expressing human A375 melanoma cells on the CAM. These tumors induced numerous lymphatics at the invasive front, and compressed or destroyed VEGF receptor (R)-3-positive lymphatics were observed within the solid tumors. The lymphatics in the CAM and in the A375 melanomas could also be demonstrated with an antibody against Prox 1, a highly specific marker of lymphatic endothelial cells. Proliferation studies revealed a BrdU labeling index of 11.6% of the lymphatic endothelial cells in the tumors and at their margins. A great number of melanoma cells invaded the lymphatics. Such interactions were not observed with VEGF-C-negative Malme 3 M melanoma cells. Lymphangiogenesis was inhibited to some extent when A375 melanoma cells were transfected with cDNA encoding soluble VEGFR-3 (sflt4), and the BrdU labeling index of the lymphatics in these tumors was 3.9%. Invasion of lymphatics and growth of blood vascular capillaries were not inhibited by the transfection. Therefore, tumor-induced lymphangiogenesis seems to be dependent to some extent on VEGF-C/flt4 interactions, but invasion of lymphatics seems to be a distinct mechanism.


Subject(s)
Lymphatic System/pathology , Melanoma, Experimental/pathology , Allantoin/metabolism , Animals , Blotting, Northern , Cell Division , Cell Nucleus/ultrastructure , Chick Embryo , Coturnix , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/biosynthesis , Humans , Immunohistochemistry , In Situ Hybridization , Lymphatic System/ultrastructure , Melanoma, Experimental/genetics , Melanoma, Experimental/ultrastructure , Mice , Mice, Nude , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Transfection , Tumor Cells, Cultured , Tumor Suppressor Proteins , Vascular Endothelial Growth Factor Receptor-3
8.
Cancer Res ; 59(13): 3185-91, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10397264

ABSTRACT

Protein ligands and receptor tyrosine kinases that specifically regulate endothelial cell function are mainly involved in physiological as well as in disease-related angiogenesis. These ligand/receptor systems include the vascular endothelial growth factor (VEGF) and the angiopoietin (Ang) families, and their receptors, the VEGF receptor family and the tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (Tie) family. In the present study, the contribution of these endothelium-specific ligand/receptor systems to tumor angiogenesis was evaluated. A375v human melanoma cells, which express at least the angiogenic growth factors VEGF, VEGF-C, and Ang-1, were stably transfected to overexpress the extracellular ligand-binding domains of the endothelium-specific receptor tyrosine kinases fms-like tyrosine kinase-1 (Flt-1), Flt-4, Tie-1, and Tie-2, respectively. In vitro proliferation and colony formation assays confirmed that expression of the extracellular receptor domains inhibited neither tumor cell mitogenesis nor the ability to produce anchorage-independent growth. Nude mouse xenografts revealed that interference with either the VEGF receptor pathway or the Tie-2 pathway resulted in a significant inhibition of tumor growth and tumor angiogenesis. In contrast, interference with the Flt-4 pathway or the Tie-1 pathway was without significant effect. Our results show that both the VEGF receptor pathway and the Tie-2 pathway are essential for A375v melanoma xenograft growth. The inhibition of the VEGF receptor pathway cannot be compensated by the Tie-2 pathway, nor vice versa. These findings suggest that the VEGF receptor pathway and the Tie-2 pathway have to be considered as two independent mediators essential for the process of in vivo angiogenesis.


Subject(s)
Melanoma/blood supply , Melanoma/pathology , Neovascularization, Pathologic/prevention & control , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/physiology , Animals , Endothelial Growth Factors/biosynthesis , Humans , Lymphokines/biosynthesis , Mice , Mice, Nude , Microcirculation/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-2 , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
9.
J Cancer Res Clin Oncol ; 125(6): 336-42, 1999.
Article in English | MEDLINE | ID: mdl-10363565

ABSTRACT

Vascular endothelial growth factor (VEGF) is an important mediator of tumor-induced angiogenesis and represents a potential target for anticancer therapy. Therefore, we prepared a panel of monoclonal antibodies (mAb) against both the VEGF121 and VEGF165 isoforms. Three of them completely neutralized the mitogenic stimulation by VEGF of human umbilical vein endothelial cells at mAb concentrations below 0.1 microg/ml. The most potent one, with a dissociation constant (Kd) of 8 pM, inhibited, in a dose-dependent manner, VEGF-induced angiogenesis in a growth factor implant model in mice. A complete inhibition of the angiogenic response was obtained by daily intraperitoneal injections of 10 microg mAb/mouse. Angiogenesis induced by basic fibroblast growth factor was not inhibited by the mAb. Epitope mapping of the mAb, performed by competitive enzyme-linked immunosorbent assay and Western blot analysis, showed that it did not bind to the reduced and denatured monomer of VEGF. Substitutions of three residues (Q87R, G88K, Q89K), located on the major surface loop beta5 to beta6 of VEGF, resulted in the complete loss of binding (more than 400-fold reduction). The results suggest that the mAb binds primarily to a conformation-dependent epitope on the VEGF dimeric form, encompassing one of the loop regions involved in KDR receptor binding. The mAb with its strong neutralizing properties represents a useful agent for effective blocking of VEGF-mediated tumor neovascularization.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigen-Antibody Reactions , Endothelial Growth Factors/immunology , Lymphokines/immunology , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Epitope Mapping , Humans , Mice , Neovascularization, Pathologic/therapy , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
J Biol Chem ; 273(18): 11115-20, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9556597

ABSTRACT

Vascular endothelial growth factor (VEGF) is an endothelial cell-specific mitogen and a key mediator of aberrant endothelial cell proliferation and vascular permeability in a variety of human pathological situations such as tumor angiogenesis, diabetic retinopathy, or psoriasis. By amino-terminal deletion analysis and by site-directed mutagenesis we have identified a new domain within the amino-terminal alpha-helix that is essential for dimerization of VEGF. VEGF121 variants containing amino acids 8 to 121 or 14 to 121, respectively, either expressed in Escherichia coli and refolded in vitro, or expressed in Chinese hamster ovary cells, were in a dimeric conformation and showed full binding activity to VEGF receptors and stimulation of endothelial cell proliferation as compared with wild-type VEGF. In contrast, a VEGF121 variant covering amino acids 18 to 121, as well as a variant in which the hydrophobic amino acids Val14, Val15, Phe17, and Met18 within the amphipathic alpha-helix near the amino terminus were replaced by serine, failed to form biological active VEGF dimers. From these data we conclude that a domain between amino acids His12 and Asp19 within the amino-terminal alpha-helix is essential for formation of VEGF dimers, and we propose hydrophobic interactions between VEGF monomers to stabilize or favor dimerization.


Subject(s)
Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , CHO Cells , Cricetinae , Dimerization , Endothelial Growth Factors/chemistry , Endothelial Growth Factors/genetics , Escherichia coli/genetics , Lymphokines/chemistry , Lymphokines/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Proc Natl Acad Sci U S A ; 95(8): 4625-9, 1998 Apr 14.
Article in English | MEDLINE | ID: mdl-9539788

ABSTRACT

Vascular endothelial growth factor (VEGF) is a potent mitogen with a unique specificity for endothelial cells and a key mediator of aberrant endothelial cell proliferation and vascular permeability in a variety of human pathological situations, such as tumor angiogenesis, diabetic retinopathy, rheumatoid arthritis, or psoriasis. VEGF is a symmetric homodimeric molecule with two receptor binding interfaces lying on each pole of the molecule. Herein we report on the construction and recombinant expression of an asymmetric heterodimeric VEGF variant with an intact receptor binding interface at one pole and a mutant receptor binding interface at the second pole of the dimer. This VEGF variant binds to VEGF receptors but fails to induce receptor activation. In competition experiments, the heterodimeric VEGF variant antagonizes VEGF-stimulated receptor autophosphorylation and proliferation of endothelial cells. A 15-fold excess of the heterodimer was sufficient to inhibit VEGF-stimulated endothelial cell proliferation by 50%, and a 100-fold excess resulted in an almost complete inhibition. By using a rational approach that is based on the structure of VEGF, we have shown the feasibility to construct a VEGF variant that acts as an VEGF antagonist.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Lymphokines/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Binding Sites , Binding, Competitive , Cell Division/drug effects , Cells, Cultured , Cloning, Molecular , Dimerization , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Genetic Variation , Humans , Kinetics , Lymphokines/biosynthesis , Lymphokines/chemistry , Models, Molecular , Mutagenesis, Site-Directed , Phosphorylation , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/drug effects , Receptors, Growth Factor/chemistry , Receptors, Growth Factor/drug effects , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/biosynthesis , Recombinant Proteins/drug effects , Recombinant Proteins/pharmacology , Umbilical Arteries , Umbilical Veins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
Cancer Res ; 57(23): 5421-5, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9393770

ABSTRACT

The growth of solid tumors and the formation of metastases are dependent on neoangiogenesis. One of the most important factors in inducing the formation of new blood vessels is the vascular endothelial growth factor (VEGF), which acts specifically on endothelial cells. VEGF is expressed and secreted by almost all solid tumors. The molecular mechanisms leading to enhanced production of this angiogenic mitogen are manyfold and have been elucidated to some degree. Two VEGF receptors, fms-like tyrosine kinase 1 (FLT-1) and KDR, have been identified almost specifically on human endothelial cells. They are expressed preferentially in the proliferating endothelium of vessels lining and/or penetrating solid tumors, whereas they are almost undetectable by convenient methods in vessels of healthy tissue. However, the underlying mechanisms are not understood. We could show that media conditioned by various cancer cell lines grown under hypoxic conditions were able to up-regulate expression of FLT-1 mRNA and protein but not of KDR mRNA. Furthermore, up-regulation of a shorter mRNA species was observed that most probably codes for the soluble variant of FLT-1. These effects were completely inhibited by VEGF-neutralizing extracellular VEGF receptor domains. The effect could be mimicked by adding recombinant VEGF instead of conditioned cancer cell medium to the endothelial cell cultures. Both mutant VEGF, which activates only KDR, and placenta growth factor, which activates only FLT-1, were able to enhance FLT-1 expression. VEGF-stimulated FLT-1 mRNA expression was inhibited by actinomycin D. These data suggest that VEGF itself is the main factor secreted by tumor cells that is able to enhance the expression of its receptor FLT-1 and of a soluble variant of FLT-1 in endothelial cells.


Subject(s)
Endothelial Growth Factors/pharmacology , Gene Expression Regulation/drug effects , Lymphokines/pharmacology , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Transcription, Genetic/drug effects , Cell Hypoxia , Cells, Cultured , Culture Media, Conditioned , Enzyme-Linked Immunosorbent Assay , Genetic Variation , Humans , Proto-Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/biosynthesis , Umbilical Veins , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
14.
Hybridoma ; 16(5): 465-71, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9388030

ABSTRACT

Vascular endothelial cell growth factor (VEGF) plays a pivotal role in the regulation of angiogenesis by binding to its cognate receptor molecule type II (VEGFr-II, KDR). VEGFr-II is an endothelial cell-specific transmembrane tyrosine kinase important for vascular endothelial cell development and differentiation during embryogenesis, angiogenic processes under physiological conditions, and various diseases. An increasing number of reports indicate that VEGF/VEGFr-II also play a fundamental role for tumor angiogenesis. We present the generation and in vitro characterization of the monoclonal antibodies 2-7-9 and 2-10-1. Both antibodies are highly specific for VEGFr-II as demonstrated by Western blotting and immunoprecipitation. MAbs 2-10-1 and 2-7-9 bind to a disulphide bridge-stabilized epitope within domains 6 and 7 of the human VEGFr-II with an affinity of 8 and 80 nM, respectively. Furthermore, the antibodies are suitable for immunohistochemistry and ELISA techniques. Because both antibodies recognize their epitope on living cells, they also have the potential for drug targeting and diagnostic purposes.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Receptors, Mitogen/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Cells, Cultured , Epitope Mapping , Female , Humans , Mice , Mice, Inbred BALB C , Neovascularization, Physiologic , Protein Conformation , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/chemistry , Recombinant Proteins/immunology
15.
Oncogene ; 15(6): 669-76, 1997 Aug 07.
Article in English | MEDLINE | ID: mdl-9264407

ABSTRACT

Stimulation of NIH3T3 cells with platelet-derived growth factor (PDGF)-BB enhances expression of vascular endothelial growth factor (VEGF), an endothelial cell-specific mitogen and a key mediator of tumor angiogenesis. Here, we identified cis-acting VEGF promoter elements and trans-acting factors which are involved in PDGF-stimulated VEGF expression. By 5'-deletion and transient transfection analysis, a G + C-rich region at -85 to -50 of the human VEGF promoter was shown to be necessary and sufficient for both PDGF inducible and basal expression. The region contains three potential recognition sites for Sp1 transcription factors, which overlap with two Egr-1 sites. Mutations that abolish the ability of Sp1 to interact with the VEGF promoter element also abrogate expression induced by PDGF. Mutations of the potential Egr-1 binding sites did not affect PDGF responsiveness. Gel shift and antibody supershift analyses showed that Sp1 and Sp3 interact constitutively with the VEGF promoter element. Our data strongly suggest that enhanced VEGF gene expression in PDGF-induced NIH3T3 cells is mediated by Sp1 and/or Sp3 transcription factors bound to the -85 to -50 promoter region of the VEGF gene.


Subject(s)
Endothelial Growth Factors/genetics , Immediate-Early Proteins , Lymphokines/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/genetics , Transcription Factors , 3T3 Cells , Animals , Antibodies, Blocking/immunology , Base Composition , Cells, Cultured , Cloning, Molecular , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Gene Expression Regulation, Neoplastic , Humans , Luciferases/genetics , Mice , Platelet-Derived Growth Factor/pharmacology , Retroviridae Proteins, Oncogenic/genetics , Sequence Deletion , Sp1 Transcription Factor/immunology , TATA Box , Thymidine Kinase/genetics , Trans-Activators/genetics , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Biochem Biophys Res Commun ; 234(3): 626-30, 1997 May 29.
Article in English | MEDLINE | ID: mdl-9175764

ABSTRACT

The action of insulin and IGF-1 in comparison to non-diabetic controls was studied in cultured fibroblasts of a patient with an inherited syndrome of insulin resistance (Type A syndrome). Insulin binding was reduced due to decreased receptor affinity, but sequence analyses revealed no alterations of splicing or primary insulin receptor (IR) structure. Most likely due to the IR affinity defect analyses of signal transduction pathways showed an impairment of insulin action on glucose uptake, total RNA synthesis and phosphorylation as well as activity of MAP-kinase. In addition inducibility of c-fos mRNA level was strongly impaired by insulin and IGF-1, but comparable to controls by PDGF indicating a postreceptor defect. In conclusion, we provide evidence that genetic syndromes of insulin resistance can be associated with both, receptor and postreceptor defects.


Subject(s)
Insulin Resistance/genetics , Insulin-Like Growth Factor I/physiology , Insulin/physiology , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Cells, Cultured , Female , Gene Expression Regulation/genetics , Genes, fos , Humans , Insulin/metabolism , Mutation , Protein Binding , Receptor, IGF Type 1/metabolism
17.
J Biol Chem ; 272(16): 10382-8, 1997 Apr 18.
Article in English | MEDLINE | ID: mdl-9099677

ABSTRACT

The vascular endothelial growth factor (VEGF) receptor FLT-1 has been shown to be involved in vasculogenesis and angiogenesis. The receptor is characterized by seven Ig-like loops within the extracellular domain. Upon VEGF binding FLT-1 becomes phosphorylated, which has been thought to be preceded by receptor dimerization. To further investigate high affinity binding of VEGF to FLT-1 and ligand-induced receptor dimerization, we expressed in Sf9 cells the entire extracellular domain comprising all seven Ig-like loops: sFLT-1(7) and several truncated mutants consisting of loop one, one and two, one to three, one to four, and one to five. The corresponding proteins, named sFLT-1(1), (2), (3), (4), and (5) were purified. Only mutants sFLT-1(3) to (7) were able to bind 125I-VEGF with high affinity. No binding of VEGF was observed with sFLT-1(1) and sFLT-1(2), indicating that the first three Ig-like loops are involved in high affinity binding of VEGF. The binding of VEGF to sFLT-1(3) could be competed with placenta growth factor (PlGF), a VEGF-related ligand, suggesting that high affinity binding of VEGF and PlGF is mediated by the same or closely related contact sites on sFLT-1. Deglycosylation of the sFLT-1(3), (4), (5), and (7) did not abolish VEGF binding. Furthermore, unglycosylated sFLT-1(3), expressed in Escherichia coli, was able to bind VEGF with similar affinity as sFLT-1(3) or sFLT-1(7), both expressed in Sf9 cells. This indicates that receptor glycosylation is not essential for high affinity binding. Dimerization of the extracellular domains of FLT-1 upon addition of VEGF was detected with all mutants containing the Ig-like loop four. Although sFLT-1(3) was able to bind VEGF, dimerization of this mutant was inefficient, indicating that sites on Ig-like loop four are essential to stabilize receptor dimers.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/metabolism , Lymphokines/metabolism , Lymphokines/pharmacology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Binding Sites , Cell Line , Cloning, Molecular , DNA/biosynthesis , DNA Replication/drug effects , Dimerization , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Escherichia coli , Humans , Kinetics , Ligands , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/isolation & purification , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/isolation & purification , Receptors, Growth Factor/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spodoptera , Transfection , Umbilical Veins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
18.
Z Gastroenterol ; 34 Suppl 3: 49-50, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8767460

ABSTRACT

To localize transcriptional active cis elements and to study the effect of a single base transition at -86 derived from a patient (PS) with an inherited severe form of hypertriglyceridemia a reporter gene transfection assay with apo C-II promoter fragments was performed. Sequences from -170 to -140, -140 to -59 and -59 to -39 were transcriptional active. The A to G transition at -86 reduced promoter activity to 25% (pC2PS-13). A nuclear protein bound sequence specific to a DNA fragment from -102 to -70, but disappeared when the point mutation at -86 was introduced. We found that one of these cis elements enhances promoter activity as a response of cAMP elevation. Apo CII mRNA level increases after incubation of HepG2 cells with 10 microM forskolin within 2 hours as measured by northern blot analyses. Incubation of transfected cells (pC2W) with forskolin [10 microM] or a cAMP analogue resulted in an 6-fold increase of luciferase activity within 6 hours and a subsequent decrease. The results suggest that several positive acting elements are located between -170 and -36 and that one of them may be responsible for promoter activation by cAMP. The reduction of promoter activity by the point mutation may be a consequence of impaired protein-DNA interaction.


Subject(s)
Apolipoproteins C/genetics , Hyperlipoproteinemia Type IV/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics , Apolipoprotein C-II , Carcinoma, Hepatocellular , Cell Line, Transformed , Genetic Complementation Test , HeLa Cells , Humans , Liver Neoplasms , Point Mutation/genetics , Transfection/genetics
19.
Cancer Res ; 56(10): 2299-301, 1996 May 15.
Article in English | MEDLINE | ID: mdl-8625303

ABSTRACT

Mutations or loss of both alleles of the von Hippel-Lindau (VHL) tumor suppressor gene has been documented in sporadic renal cell carcinomas and neoplasms that arise in individuals having the VHL syndrome. The well-vascularized phenotype of tumors that form in VHL disease let us consider vascular endothelial growth factor (VEGF) as a mediator of tumor growth in VHL disease. Human renal carcinoma cells that either lacked endogenous wild-type VHL or were transfected with an inactive mutant VHL showed deregulated expression of VEGF on the mRNA and protein level that was reverted by introduction of wild-type VHL. Stimulation of proliferation of endothelial cells by conditioned medium of cells expressing mutant VHL was almost abolished by neutralizing the VEGF. In contrast, expression of basic fibroblast growth factor and of c-myc proto-oncogene was not affected by VHL. Our data suggest VEGF as the key tumor angiogenesis factor in VHL disease.


Subject(s)
Carcinoma, Renal Cell/pathology , Endothelial Growth Factors/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , Kidney Neoplasms/pathology , Ligases , Lymphokines/biosynthesis , Neoplasm Proteins/biosynthesis , Proteins/physiology , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , von Hippel-Lindau Disease/genetics , Carcinoma, Renal Cell/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/physiology , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Genes, myc , Humans , Kidney Neoplasms/metabolism , Lymphokines/genetics , Lymphokines/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neovascularization, Pathologic/genetics , Phenotype , Protein Biosynthesis , Proteins/genetics , Proto-Oncogene Mas , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Von Hippel-Lindau Tumor Suppressor Protein
20.
Biochem Biophys Res Commun ; 222(2): 249-55, 1996 May 15.
Article in English | MEDLINE | ID: mdl-8670191

ABSTRACT

Vascular endothelial growth factor (VEGF) was identified as an endothelial cell specific mitogen that induces angiogenesis and vascular permeability in vivo. VEGF is a homodimeric protein which contains three intramolecular and two intermolecular disulfide bridges. Here, we report on an efficient procedure for recombinant production of VEGF isoforms VEGF121 and VEGF165 in Escherichia coli. The proteins were solubilized from inclusion bodies, refolded, and purified by chromatographic methods. The final protein products were almost completely in the dimeric conformation, bound to VEGF receptor FLT1 with a Kd of 30 pM, stimulated proliferation of human umbilical vein endothelial cells half-maximally at a concentration of 30 pM, and induced in vivo neovascularization and vascular permeability on the chicken chorioallantoic membrane.


Subject(s)
Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Lymphokines/biosynthesis , Lymphokines/pharmacology , Neovascularization, Physiologic/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Recombinant Proteins/biosynthesis , Allantois/blood supply , Animals , Binding, Competitive , Capillary Permeability/drug effects , Cells, Cultured , Chick Embryo , Chorion/blood supply , Cloning, Molecular , Endothelial Growth Factors/chemistry , Endothelium, Vascular/drug effects , Escherichia coli , Humans , Kinetics , Lymphokines/chemistry , Macromolecular Substances , Protein Conformation , Protein Folding , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Umbilical Veins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...