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.
Br J Cancer ; 110(12): 2855-64, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24853186

ABSTRACT

BACKGROUND: Insulin-like growth factors (IGF-I and IGF-II) signal via the type 1 IGF receptor (IGF-1R) and IGF-II also activates the insulin receptor isoform A (IR-A). Signalling via both receptors promotes tumour growth, survival and metastasis. In some instances IGF-II action via the IR-A also promotes resistance to anti-IGF-1R inhibitors. This study assessed the efficacy of two novel modified IGF-binding protein-2 (IGFBP-2) proteins that were designed to sequester both IGFs. The two modified IGFBP-2 proteins were either protease resistant alone or also lacked the ability to bind extracellular matrix (ECM). METHODS: The modified IGFBP-2 proteins were tested in vitro for their abilities to inhibit cancer cell proliferation and in vivo to inhibit MCF-7 breast tumour xenograft growth. RESULTS: Both mutants retained low nanomolar affinity for IGF-I and IGF-II (0.8-2.1-fold lower than IGFBP-2) and inhibited cancer cell proliferation in vitro. However, the combined protease resistant, non-matrix-binding mutant was more effective in inhibiting MCF-7 tumour xenograft growth and led to inhibition of angiogenesis. CONCLUSIONS: By removing protease cleavage and matrix-binding sites, modified IGFBP-2 was effective in inhibiting tumour growth and reducing tumour angiogenesis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Insulin-Like Growth Factor Binding Protein 2/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Proliferation/drug effects , Extracellular Matrix/genetics , Female , Humans , Insulin-Like Growth Factor Binding Protein 2/administration & dosage , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Protein Binding , Recombinant Proteins/therapeutic use , Xenograft Model Antitumor Assays
3.
Clin Exp Pharmacol Physiol ; 29(11): 1024-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12366396

ABSTRACT

1. The vascular endothelial growth factor (VEGF) family is a focus of interest with respect to novel therapies for cardiovascular disease. Members of this family bind differentially to three receptor tyrosine kinases, namely VEGF-R1, -R2 and -R3, and to the semaphorin receptors neuropilin 1 and 2. The role of VEGF-R1 and the factors that interact exclusively with this receptor (VEGF-B and placenta growth factor) has remained controversial. 2. To further elucidate the role of VEGF-B in blood vessel formation and function, we have expressed, purified and refolded both naturally occurring VEGF-B isoforms and a truncated amino acid 10-108 form. All refolded proteins have been demonstrated to bind to VEGF-R1 with appropriate kinetics in biosensor-based analysis. 3. Robust cell assays for VEGF-R1 ligands, such as VEGF-B, have been problematic. We have developed an assay based on a chimeric receptor consisting of extracellular domains 1-4 of VEGF-R1 and the transmembrane and intracellular domains of gp130. The cell line expresses luciferase to high levels 24 h after exposure to VEGF-A and both refolded VEGF-B167 and the short 10-108 isoform have been demonstrated to be active in this assay. 4. The novel cell-based assay, in combination with a variety of immunochemical approaches, has been used to identify and characterize monoclonal antibodies that neutralize VEGF-B activity.


Subject(s)
Angiogenesis Inducing Agents/chemistry , Antibodies, Monoclonal/biosynthesis , Recombinant Proteins/chemistry , Vascular Endothelial Growth Factor B , Angiogenesis Inducing Agents/biosynthesis , Angiogenesis Inducing Agents/genetics , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Humans , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
4.
Protein Sci ; 9(10): 2018-25, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11106176

ABSTRACT

Vascular endothelial growth factor (VEGF)-A interacts with the receptor tyrosine kinases VEGF-R1 and R2, and the importance of this interaction in endothelial cell (EC) function and blood vessel development has been well documented. Other ligands that interact differentially with these receptors and that are structurally related to VEGF-A include VEGF-B, VEGF-C, VEGF-D, and placenta growth factor (PLGF). Compared with VEGF-A, relatively little is known about the biological role of the VEGF-R1 specific ligand, VEGF-B. Two splice variant isoforms that differ at the COOH-terminus and which retain unique solubility characteristics are widely expressed throughout embryonic and postnatal development. Recent analysis of mice with a targeted deletion of the VEGF-B gene has revealed a defect in heart development and function consistent with an important role in vascularization of the myocardium (Bellomo D et al., 2000, Circ Res 86:E29-E35). To facilitate further characterization of VEGF-B, we have developed a protocol for expression and purification of refolded recombinant protein from Escherichia coli inclusion bodies (IBs). The approach developed resolves a number of significant issues associated with VEGF-B, including the ability to heterodimerize with endogenous VEGF-A when co-expressed in mammalian cells, a complex secondary structure incorporating inter- and intrachain disulfide bonds and hydrophobic characteristics that preclude the use of standard chromatographic resins. The resulting purified disulfide-linked homodimer was demonstrated to bind to VEGF-R1 and to compete with VEGF-A for binding to this receptor.


Subject(s)
Endothelial Growth Factors/chemistry , Animals , Biosensing Techniques , Chromatography, Affinity , Cloning, Molecular , Dimerization , Endothelial Growth Factors/isolation & purification , Endothelial Growth Factors/metabolism , Enzyme-Linked Immunosorbent Assay , Escherichia coli , Protein Denaturation , Protein Folding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/chemistry , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Surface Plasmon Resonance , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor B , Vascular Endothelial Growth Factor Receptor-1
5.
Proc Natl Acad Sci U S A ; 97(12): 6493-8, 2000 Jun 06.
Article in English | MEDLINE | ID: mdl-10829066

ABSTRACT

Suppressor of cytokine signaling-3 (SOCS-3) is one member of a family of intracellular inhibitors of signaling pathways initiated by cytokines that use, among others, the common receptor subunit gp130. The SH2 domain of SOCS-3 has been shown to be essential for this inhibitory activity, and we have used a quantitative binding analysis of SOCS-3 to synthetic phosphopeptides to map the potential sites of interaction of SOCS-3 with different components of the gp130 signaling pathway. The only high-affinity ligand found corresponded to the region of gp130 centered around phosphotyrosine-757 (pY757), previously shown to be a docking site for the tyrosine phosphatase SHP-2. By contrast, phosphopeptides corresponding to other regions within gp130, Janus kinase, or signal transducer and activator of transcription proteins bound to SOCS-3 with weak or undetectable affinity. The significance of pY757 in gp130 as a biologically relevant SOCS-3 docking site was investigated by using transfected 293T fibroblasts. Although SOCS-3 inhibited signaling in cells transfected with a chimeric receptor containing the wild-type gp130 intracellular domain, inhibition was considerably impaired for a receptor carrying a Y-->F point mutation at residue 757. Taken together, these data suggest that the mechanism by which SOCS-3 inhibits the gp130 signaling pathway depends on recruitment to the phosphorylated gp130 receptor, and that some of the negative regulatory roles previously attributed to the phosphatase SHP-2 might in fact be caused by the action of SOCS-3.


Subject(s)
Antigens, CD/metabolism , Membrane Glycoproteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Proteins/metabolism , Repressor Proteins , Signal Transduction , Transcription Factors , Amino Acid Sequence , Binding Sites , Cytokine Receptor gp130 , Intracellular Signaling Peptides and Proteins , Janus Kinase 1 , Molecular Sequence Data , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein-Tyrosine Kinases/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
6.
Growth Factors ; 16(4): 265-78, 1999.
Article in English | MEDLINE | ID: mdl-10427501

ABSTRACT

Interleukin-6 (IL-6) mediates its activity through binding to two cell-surface receptors. The high-affinity human IL-6 receptor complex consists of two transmembrane anchored subunits: a ligand-specific, low-affinity IL-6 receptor and the high-affinity converter and signal transducing, gp130. Previously, using recombinant forms of human IL-6 and the extracellular ('soluble') domains of the IL-6 receptor (sIL-6R) and gp130 (sgp130), we have shown that the high-affinity IL-6R complex is hexameric, consisting of two molecules each of IL-6, sIL-6R and sgp130 (Ward et al., 1994, J. Biol. Chem. 269: 23286-23289). This paper investigates the role of the N-terminal region of gp130 in the formation of the high-affinity IL-6R complex. Using recombinant sgp130 produced with a FLAG octapeptide epitope (DYKDDDDK) at the N-terminus (sgp130-FLAG), we demonstrate, using biosensor analysis and size-exclusion chromatography, that modification of the N-terminus of sgp130 interferes with the in vitro in solution formation of the stable hexameric IL-6 receptor complex. Rather, sgp130-FLAG interacts with IL-6 and sIL-6R with a much lower affinity and forms a stable lower-order ternary complex. However, this lower-order complex is inconsistent with the solution molecular weight of a trimeric complex, as measured by size-exclusion chromatography. In contrast, N-terminal modification of the sgp130 with the FLAG epitope did not interfere with the binding of leukemia inhibitory factor or oncostatin-M (other cytokines that signal through gp130) to sgp130. These data support our model of the hexameric IL-6 receptor complex, which is biased towards the association of two IL-6.IL-6R.gp130 trimers, and postulates the critical involvement of the N-terminal Ig-like domain of gp130 in tethering the two trimers to form the stable hexamer (Simpson et al., 1997, Prot. Sci. 6: 929-955).


Subject(s)
Antigens, CD/pharmacology , Interleukin-6/metabolism , Membrane Glycoproteins/pharmacology , Protein Binding , Receptors, Interleukin-6/metabolism , Amino Acid Sequence , Antigens, CD/chemistry , Antigens, CD/immunology , Antineoplastic Agents , Binding Sites , Biosensing Techniques/methods , Chromatography, Gel , Cytokine Receptor gp130 , Cytokines , Epitopes , Fungal Proteins/metabolism , Growth Inhibitors , Humans , Inflammation Mediators , Leukemia Inhibitory Factor , Lymphokines , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/immunology , Oncostatin M , Peptides , Pichia , Protein Conformation , Recombinant Proteins/analysis , Surface Plasmon Resonance
7.
Biochem J ; 325 ( Pt 3): 693-700, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9271090

ABSTRACT

Leukaemia inhibitory factor (LIF) is a polyfunctional cytokine that is known to require at least two distinct receptor components (LIF receptor alpha-chain and gp130) in order to form a high-affinity, functional, receptor complex. Human LIF binds with unusually high affinity to a naturally occurring mouse soluble LIF receptor alpha-chain, and this property was used to purify a stable complex of human LIF and mouse LIF receptor alpha-chain from pregnant-mouse serum. Recombinant soluble human gp130 was expressed, with a FLAG(R) epitope (DYKDDDDK) at the N-terminus, in the methylotropic yeast Pichia pastoris and purified using affinity chromatography. The formation of a trimeric complex in solution was established by native gel electrophoresis, gel-filtration chromatography, sedimentation equilibrium analysis, surface plasmon resonance spectroscopy and chemical cross-linking. The stoichiometry of this solution complex was 1:1:1, in contrast with that of the complex of interleukin-6, the interleukin-6-specific low-affinity receptor subunit and gp130, which is 2:2:2.


Subject(s)
Growth Inhibitors/chemistry , Lymphokines/chemistry , Antigens, CD/genetics , Antigens, CD/metabolism , Cytokine Receptor gp130 , Growth Inhibitors/genetics , Growth Inhibitors/metabolism , Humans , Interleukin-6/metabolism , Leukemia Inhibitory Factor , Lymphokines/genetics , Lymphokines/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Pichia/genetics , Protein Binding , Recombinant Fusion Proteins/genetics , Signal Transduction , Solutions
8.
J Biol Chem ; 266(35): 23815-23, 1991 Dec 15.
Article in English | MEDLINE | ID: mdl-1721056

ABSTRACT

Human granulocyte colony-stimulating factor (G-CSF) is a hemopoietic growth factor that is being used successfully to treat various forms of neutropenia. To define functionally important regions of G-CSF, we have prepared 37 monoclonal anti-G-CSF antibodies and mapped the regions of G-CSF recognized by different antibody groups. Antibodies recognizing similar epitopes were identified by competition assays, neutralization assays, conformation dependence and cross-reactivity with canine G-CSF. Seven of eight neutralizing antibodies fell into two related epitope groups and were conformation-dependent. The eighth was unrelated and conformation-independent. Peptides of G-CSF were generated by chemical or enzymatic digestion and tested for antibody reactivity. One of the neutralizing antibodies (LMM351) recognized a small, disulfide-bonded peptide from the V8 protease digest (residues 34-46). A synthetic peptide (residues 20-58) was recognized by all the neutralizing antibodies, implicating this disulfide-bonded loop in receptor binding. The epitopes recognized by nonneutralizing antibodies were found throughout G-CSF. Thus, regions of G-CSF that are not involved in receptor binding have also been defined. A CNBr peptide (residues 1-121) had greatly reduced biological activity, indicating that the COOH terminus is required for receptor binding. We predict that residues 20-46 and the COOH terminus bind to the G-CSF receptor.


Subject(s)
Antibodies, Monoclonal , Epitopes/analysis , Granulocyte Colony-Stimulating Factor/physiology , Peptide Fragments/pharmacology , Amino Acid Sequence , Binding Sites, Antibody , Cell Division/drug effects , Cell Line , Chromatography, High Pressure Liquid , Endopeptidases , Enzyme-Linked Immunosorbent Assay , Granulocyte Colony-Stimulating Factor/immunology , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Models, Molecular , Molecular Sequence Data , Neutralization Tests , Peptide Fragments/immunology , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Software
9.
FEBS Lett ; 224(1): 128-32, 1987 Nov 16.
Article in English | MEDLINE | ID: mdl-3678486

ABSTRACT

The complete amino acid sequence of basic FGF (146 residues) from ovine pituitary glands has been established. This has been achieved by the sequence analysis of subnanomole amounts of the intact molecule and of peptides derived by enzymatic digestions with clostripain, chymotrypsin, pepsin and Staphylococcus aureus V8 protease. Microbore HPLC, employing 1-2 mm i.d. columns, was used to purify, concentrate and buffer-exchange the FGF peptides. A novel application of ion-pairing chromatography was employed to isolate peptides which were not retained on conventional reversed-phase systems. There is only one positional difference between the ovine and bovine basic FGFs, but there are 3 positional differences between ovine and human basic FGFS.


Subject(s)
Fibroblast Growth Factors/genetics , Pituitary Gland/analysis , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Fibroblast Growth Factors/isolation & purification , Humans , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Sheep/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...