Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 160(4): 1016-28, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20590596

ABSTRACT

BACKGROUND AND PURPOSE: The overexpression of epidermal growth factor receptor (EGFR) and its mutated variant EGFRvIII occurs in 50% of glioblastoma multiforme. We developed antibody fragments against EGFR/EGFRvIII for molecular imaging and/or therapeutic targeting applications. EXPERIMENTAL APPROACH: An anti-EGFR/EGFRvIII llama single-domain antibody (EG(2)) and two higher valency format constructs, bivalent EG(2)-hFc and pentavalent V2C-EG(2) sdAbs, were analysed in vitro for their binding affinities using surface plasmon resonance and cell binding studies, and in vivo using pharmacokinetic, biodistribution, optical imaging and fluorescent microscopy studies. KEY RESULTS: Kinetic binding analyses by surface plasmon resonance revealed intrinsic affinities of 55 nM and 97 nM for the monovalent EG(2) to immobilized extracellular domains of EGFR and EGFRvIII, respectively, and a 10- to 600-fold increases in apparent affinities for the multivalent binders, V2C-EG(2) and EG(2)-hFc, respectively. In vivo pharmacokinetic and biodistribution studies in mice revealed plasma half-lives for EG(2), V2C-EG(2) and EG(2)-hFc of 41 min, 80 min and 12.5 h, respectively, as well as a significantly higher retention of EG(2)-hFc compared to the other two constructs in EGFR/EGFRvIII-expressing orthotopic brain tumours, resulting in the highest signal in the tumour region in optical imaging studies. Time domain volumetric optical imaging fusion with high-resolution micro-computed tomography of microvascular brain network confirmed EG(2)-hFc selective accumulation/retention in anatomically defined tumour regions. CONCLUSIONS: Single domain antibodies can be optimized for molecular imaging applications by methods that improve their apparent affinity and prolong plasma half-life and, at the same time, preserve their ability to penetrate tumour parenchyma.


Subject(s)
Brain Neoplasms/diagnosis , ErbB Receptors/metabolism , Glioblastoma/diagnosis , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Variable Region/metabolism , Animals , Antibodies/metabolism , Antibody Affinity , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Brain Neoplasms/ultrastructure , Cell Line, Tumor , ErbB Receptors/immunology , Glioblastoma/blood supply , Glioblastoma/metabolism , Glioblastoma/ultrastructure , Half-Life , Humans , Immobilized Proteins/metabolism , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Kinetics , Male , Mice , Mice, Nude , Molecular Imaging/methods , Molecular Weight , Tomography, Optical/methods , Whole Body Imaging/methods , X-Ray Microtomography/methods
2.
Oncogene ; 29(6): 831-44, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-19935703

ABSTRACT

Transforming growth factor (TGF)-beta plays a dual role in tumorigenesis, switching from acting as a growth inhibitory tumor suppressor early in the process, to a tumor promoter in late-stage disease. Since TGF-beta's prometastatic role may be linked to its ability to induce tumor cell epithelial-to-mesenchymal transition (EMT), we explored TGF-beta's EMT-promoting pathways by analysing the transcriptome changes occurring in BRI-JM01 mammary tumor epithelial cells undergoing a TGF-beta-induced EMT. We found the clusterin gene to be the most highly upregulated throughout most of the TGF-beta time course, and showed that this results in an increase of the secreted form of clusterin. By monitoring several hallmark features of EMT, we demonstrated that antibodies targeting secreted clusterin inhibit the TGF-beta-induced EMT of BRI-JM01 cells, as well as the invasive phenotype of several other breast and prostate tumor cell lines (4T1, NMuMG, MDA-MB231LM2 and PC3), without affecting the proliferation of these cells. These results indicate that secreted clusterin is a functionally important EMT mediator that lies downstream within TGF-beta's EMT-promoting transcriptional cascade, but not within its growth-inhibitory pathways. To further investigate the role played by secreted clusterin in tumor metastasis, we assessed the effect of several anti-clusterin monoclonal antibodies in vivo using a 4T1 syngeneic mouse breast cancer model and found that these antibodies significantly reduce lung metastasis. Taken together, our results reveal a role for secreted clusterin as an important extracellular promoter of EMT, and suggest that antibodies targeting clusterin may inhibit tumor metastasis without reducing the beneficial growth inhibitory effects of TGF-beta.


Subject(s)
Antibodies/therapeutic use , Clusterin/antagonists & inhibitors , Clusterin/genetics , Epithelial Cells/pathology , Extracellular Space/metabolism , Gene Expression Profiling , Mesoderm/pathology , Transforming Growth Factor beta/pharmacology , Animals , Antibodies/immunology , Antibodies/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Clusterin/immunology , Clusterin/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Space/drug effects , Female , Humans , Immunoglobulin G/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Transcription, Genetic
3.
Oncogene ; 28(7): 1005-15, 2009 Feb 19.
Article in English | MEDLINE | ID: mdl-19079339

ABSTRACT

Transforming growth factor (TGF)-beta signaling is a potent modulator of the invasive and metastatic behavior of breast cancer cells. Indeed, breast tumor responsiveness to TGF-beta is important for the development of osteolytic bone metastases. However, the specific TGF-beta isoforms that promote breast cancer outgrowth in bone is unknown. We demonstrate that expression of a TGF-beta ligand trap, which neutralizes TGF-beta1 and TGF-beta3, in MDA-MB-231 breast cancer cells diminished their outgrowth in bone and reduced the severity of osteolytic lesion formation when compared with controls. We further show that a reduction or loss of TGF-beta1 expression within the bone microenvironment of TGF-beta1+/- and TGF-beta1-/- mice significantly reduced the incidence of breast tumor outgrowth compared with wild-type animals. Interestingly, those tumors capable of growing within the tibiae of TGF-beta1-deficient mice had upregulated expression of all three TGF-beta isoforms. Finally, breast cancer cells expressing the TGF-beta ligand trap showed a pronounced reduction in their ability to form osteolytic lesions when injected into the tibiae of TGF-beta1+/- mice. Thus, our studies show that both host- and tumor-derived TGF-beta expression plays a critical role during the establishment and outgrowth of breast cancer cells in bone.


Subject(s)
Bone Neoplasms/pathology , Breast Neoplasms/pathology , Osteolysis/prevention & control , Transforming Growth Factor beta1/physiology , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , DNA-Binding Proteins/physiology , Female , Humans , Immunoenzyme Techniques , Mice , Mice, Knockout , Mice, Nude , Osteolysis/pathology , Phosphorylation , Protein Isoforms , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta3/antagonists & inhibitors , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism
4.
Cell Mol Life Sci ; 64(14): 1752-62, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17415519

ABSTRACT

Genomic alterations lead to cancer complexity and form a major hurdle for comprehensive understanding of the molecular mechanisms underlying oncogenesis. In this review, we describe recent advances in studying cancer-associated genes from a systems biology point of view. The integration of known cancer genes onto protein and signaling networks reveals the characteristics of cancer genes within networks. This approach shows that cancer genes often function as network hub proteins which are involved in many cellular processes and form focal nodes in information exchange between many signaling pathways. Literature mining allows constructing gene-gene networks, in which new cancer genes can be identified. The gene expression profiles of cancer cells are used for reconstructing gene regulatory networks. By doing so, genes which are involved in the regulation of cancer progression can be picked up from these networks, after which their functions can be further confirmed in the laboratory.


Subject(s)
Gene Regulatory Networks , Genes, Neoplasm , Neoplasms/genetics , Systems Biology , Data Interpretation, Statistical , Databases, Genetic , Gene Expression Regulation, Neoplastic , Genomics , Humans , Models, Biological , Oligonucleotide Array Sequence Analysis , Signal Transduction/genetics
5.
Cell Death Differ ; 10(11): 1234-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14576775

ABSTRACT

Smac/Diablo and HtrA2/Omi promote apoptosis by binding to and antagonizing IAP proteins, including the 'X chromosome-linked inhibitor of apoptosis' (XIAP). Here we show that caspase-mediated proteolysis of a limited subset of cell death substrates exposes functional Smac/Diablo-like N-termini after cleavage, which are able to bind to and antagonize XIAP. We propose that this mechanism may establish a feedforward sensitization of the apoptotic pathway and contribute to the functional redundancy of IAP antagonism. In addition, this may be particularly relevant in Alzheimer's disease since the caspase-generated C31 peptide, an established cytotoxin, acquires Smac/Diablo-like properties after apoptotic processing.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Peptide Fragments/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , Apoptosis Regulatory Proteins , Carrier Proteins/physiology , Caspase 3 , Cell Line, Tumor , Cytochromes c/metabolism , Enzyme Inhibitors/pharmacology , Feedback, Physiological/physiology , Histocompatibility Antigens Class I/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mitochondria/metabolism , Mitochondrial Proteins/physiology , Peptide Fragments/pharmacology , Protein Structure, Tertiary/physiology , Proteins/antagonists & inhibitors , Proteins/metabolism , Signal Transduction/physiology , X-Linked Inhibitor of Apoptosis Protein
6.
Gene Ther ; 9(18): 1238-46, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12215891

ABSTRACT

Adenoviruses (Ad) deleted in the protease (PS) gene are capable of only one round of replication in non-complementing cells. This feature was exploited to develop a positive selection method for constructing adenoviral recombinants using ectopic expression of the PS gene in the E1 region. Very low levels of PS were sufficient to ensure the rescue of a PS-deleted Ad genome (Ad(Delta)PS), thereby eliminating deleterious effects PS over-expression might exert on cell or virus growth. In addition to the standard co-transfection method, an alternative protocol was developed in which the Ad5-(Delta)PS viral DNA was delivered by infection before subsequent transfection of 293 cells with the transfer vector. Under optimal conditions, at least one recombinant Ad per 10(3) cells was generated with 100% of the plaques being recombinant. Since the infection/transfection protocol is readily scalable, this represents the first method that allows for the easy construction of adenovirus vector (AdV) libraries with high diversities. This approach addresses in a novel way the bottleneck encountered when converting plasmid libraries, constructed in E. coli using a variety of well-established strategies, into corresponding AdV libraries. It maintains high diversity while generating recombinant viruses with 100% efficiency.


Subject(s)
Adenoviridae/genetics , Endopeptidases/genetics , Gene Library , Genetic Vectors/genetics , Animals , Cell Line , Genetic Engineering , Transfection/methods , Virus Replication/genetics
7.
Calcif Tissue Int ; 68(5): 304-15, 2001 May.
Article in English | MEDLINE | ID: mdl-11683538

ABSTRACT

Previous studies have shown endothelial cells to be a major target for endocrine TGF-beta in several soft tissues in the normal growing rat. The potent effect of TGF-beta1 on bone formation prompted us to analyze in detail the localization of specific binding sites for endocrine TGF-beta in hard tissues. At 2.5 minutes after injection of 125I-TGF-beta1, specific binding, as demonstrated by quantitative radioautography, was localized to fenestrated endothelium participating in angiogenesis in the vascular invasion region of the growth plate in bone as well as to anatomizing capillary networks in the maturation zone of the enamel organ. At 15 minutes after injection, the bound ligand was internalized into endocytic vesicles of endothelial cells. In bone, quantitation revealed significant differences in receptor density between endothelia undergoing proliferation vs those in a state of elongation and anastomosis with neighboring endothelial cells. In the rat incisor, specific binding of 125I-TGF-beta1 to endothelium correlated with increased formation of anastomotic capillary networks. These studies identify differential specific binding sites of 125I-TGF-beta1 in angiogenically active endothelium, providing an important link between TGF-beta1, the endothelium, and hard tissue development.


Subject(s)
Capillaries/metabolism , Enamel Organ/blood supply , Endothelium, Vascular/metabolism , Neovascularization, Physiologic/physiology , Tibia/metabolism , Transforming Growth Factor beta/metabolism , 3T3 Cells , Animals , Autoradiography , Binding Sites , Capillaries/ultrastructure , Enamel Organ/metabolism , Enamel Organ/ultrastructure , Endothelium, Vascular/ultrastructure , Incisor , Iodine Radioisotopes , Mice , Microscopy, Electron , Rats , Transforming Growth Factor beta1
8.
Mol Cell Biol ; 21(15): 5200-13, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11438674

ABSTRACT

The cap structure and the poly(A) tail of eukaryotic mRNAs act synergistically to enhance translation. This effect is mediated by a direct interaction of eukaryotic initiation factor 4G and poly(A) binding protein (PABP), which brings about circularization of the mRNA. Of the two recently identified PABP-interacting proteins, one, Paip1, stimulates translation, and the other, Paip2, which competes with Paip1 for binding to PABP, represses translation. Here we studied the Paip2-PABP interaction. Biacore data and far-Western analysis revealed that Paip2 contains two binding sites for PABP, one encompassing a 16-amino-acid stretch located in the C terminus and a second encompassing a larger central region. PABP also contains two binding regions for Paip2, one located in the RNA recognition motif (RRM) region and the other in the carboxy-terminal region. A two-to-one stoichiometry for binding of Paip2 to PABP with two independent K(d)s of 0.66 and 74 nM was determined. Thus, our data demonstrate that PABP and Paip2 could form a trimeric complex containing one PABP molecule and two Paip2 molecules. Significantly, only the central Paip2 fragment, which binds with high affinity to the PABP RRM region, inhibits PABP binding to poly(A) RNA and translation.


Subject(s)
Carrier Proteins/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Binding Sites , Binding, Competitive , Blotting, Western , Carrier Proteins/chemistry , Genetic Vectors , Humans , Kinetics , Models, Theoretical , Molecular Sequence Data , Mutation , Peptide Initiation Factors/metabolism , Poly(A)-Binding Proteins , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Time Factors
9.
J Cell Physiol ; 188(2): 211-22, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11424088

ABSTRACT

One of the differences between fetal and adult skin healing is the unique ability of fetal wounds to heal without contracture and scar formation. Studies have shown that the ratio between the three isoforms of TGFbeta is different in adult and fetal wounds. Thus, we analyzed the capacity of adult and fetal human skin fibroblasts to contract collagen gels after stimulation with TGFbeta isoforms. In control medium, fetal fibroblasts had a contractile capacity similar to that of adult fibroblasts. However, the growth capacity of fetal fibroblasts was completely inhibited, in contrast to adult fibroblasts. When cells were treated with TGFbeta, fetal fibroblasts showed an inhibition of their contractile capacity whereas adult fibroblasts further contracted gels. The contractile response was similar for all isoforms of TGFbeta although TGFbeta3 always had the strongest effect. We considered that the regulation of cell contractile capacity by TGFbeta may be dependent on receptor expression for this cytokine, on myofibroblast differentiation of the cells, or in cell links with matrix. Since TGFbeta receptor analysis did not show differences in receptor affinity, we studied the expression of alpha-smooth muscle (SM) actin, a fibroblast contractile marker and of three integrins, the cell surface receptors specific of the attachment of the fibroblasts with collagen matrix. We observed that the expression of alpha-SM actin and alpha3 and beta1 integrin subunits was increased when TGFbeta was added to the medium of adult fibroblasts whereas the levels of the alpha1 and alpha2 subunits were unchanged. In contrast, fetal fibroblasts treated with TGFbeta showed a decrease of alpha1, alpha2, and beta1 integrin expression but no change in alpha3 integrin and in alpha-SM actin expression. These results indicate that intrinsic differences between fetal and adult fibroblasts might explain their opposite responses to TGFbeta stimuli. The variations in their alpha-SM actin and integrin expression patterns represent potentially important mechanisms used by fetal fibroblasts to regulate their response to cytokines, and likely contribute to the resultant differences in the quality of wound repair.


Subject(s)
Fetus/cytology , Fibroblasts/cytology , Skin/cytology , Wound Healing/physiology , Actins/analysis , Adult , Age Factors , Antigens, CD/analysis , Blotting, Western , Cicatrix/pathology , Fibroblasts/chemistry , Fibroblasts/drug effects , Flow Cytometry , Humans , Integrin alpha1 , Integrin alpha2 , Integrin alpha3 , Integrin beta1/analysis , Integrins/analysis , Iodine Radioisotopes , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Transforming Growth Factor beta2 , Transforming Growth Factor beta3
10.
J Biol Chem ; 276(29): 27237-45, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11356827

ABSTRACT

Transforming growth factor-beta (TGF-beta) internalization was studied by monitoring the uptake of (125)I-TGF-beta1 in Mv1Lu cells, which endogenously express TGF-beta receptors types I (RI), II (RII), and III (RIII), and 293 cells transfected with RI and RII. At 37 degrees C internalization occurred rapidly, within 10 min of ligand addition. Internalization was optimal in 293 cells expressing both RI and RII. Internalization was prevented by phenylarsine oxide, a nonspecific inhibitor of receptor internalization, but was not affected by reagents that interfere with clathrin-mediated endocytosis such as monodansylcadaverine, K44A dynamin, and inhibitors of endosomal acidification. Electron microscopic examination of Mv1Lu cells treated with (125)I- TGF-beta1 at 37 degrees C indicated that internalization occurred via a noncoated vesicular mechanism. Internalization was prevented by prebinding cells with TGF-beta1 at 4 degrees C for 2 h prior to switching the cells to 37 degrees C. This was attributed to a loss of receptor binding, as indicated by a rapid decrease in the amount of TGF-beta1 bound to the cell surface at 37 degrees C and by a reduction in the labeling intensities of RI and RII in (125)I-TGF-beta1-cross-linking experiments. Mv1Lu or 293 (RI+RII) cells, prebound with TGF-beta1 at 4 degrees C and subsequently stripped of ligand by an acid wash, nevertheless initiated a signaling response upon transfer to 37 degrees C, suggesting that prebinding promotes formation of stable RI.RII complexes that can signal independently of ligand.


Subject(s)
Clathrin/metabolism , Endocytosis , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Line , Humans , Ligands , Microscopy, Electron , Mink , Protein Binding , Signal Transduction
11.
J Biol Chem ; 276(32): 29632-43, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11382746

ABSTRACT

Mature transforming growth factor-beta (TGF-beta) is proteolytically derived from the C terminus of a precursor protein. Latency-associated protein (LAP), the N-terminal remnant of the TGF-beta precursor, is able to bind and neutralize TGF-beta. Mature TGF-beta exerts its activity by binding and complexing members of two subfamilies of receptors, the type I and II receptors. In addition to these signaling receptors, TGF-beta can also interact with an accessory receptor termed the type III receptor. Using a surface plasmon resonance-based biosensor (BIAcore), we determined the mechanisms of interaction of four binding proteins (LAP, the type II and III receptor ectodomains (EDs), and a type II receptor ED/Fc chimera) with three TGF-beta isoforms, and we quantified their related kinetic parameters. Using global fitting based on a numerical integration data analysis method, we demonstrated that LAP and the type II receptor/Fc chimera interacted with the TGF-beta isoforms with a 1:1 stoichiometry. In contrast, the type II ED interactions with TGF-beta were best fit by a kinetic model assuming the presence of two independent binding sites on the ligand molecule. We also showed that the type III ED bound two TGF-beta molecules. Further experiments revealed that LAP was able to block the interactions of TGF-beta with the two EDs, but that the two EDs did not compete or cooperate with each other. Together, these results strongly support the existence of a cell-surface complex consisting of one type III receptor, two TGF-beta molecules, and four type II receptors, prior to the recruitment of the type I receptor for signal transduction. Additionally, our results indicate that the apparent dissociation rate constants are more predictive of the neutralizing potency of these TGF-beta-binding proteins (LAP, the type II and III receptor EDs, and the type II receptor/Fc chimera) than the apparent equilibrium constants.


Subject(s)
Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/metabolism , Animals , Binding Sites , Epithelial Cells/metabolism , Kinetics , Lung/cytology , Mink , Models, Chemical , Models, Theoretical , Protein Binding , Protein Isoforms , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Proteoglycans/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Surface Plasmon Resonance , Thermodynamics , Time Factors
12.
J Biol Chem ; 276(27): 24627-37, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11323414

ABSTRACT

Transforming growth factor beta (TGF-beta) signals through three high affinity cell surface receptors, TGF-beta type I, type II, and type III receptors. The type III receptor, also known as betaglycan, binds to the type II receptor and is thought to act solely by "presenting" the TGF-beta ligand to the type II receptor. The short cytoplasmic domain of the type III receptor is thought to have no role in TGF-beta signaling because deletion of this domain has no effect on association with the type II receptor, or with the presentation role of the type III receptor. Here we demonstrate that the cytoplasmic domains of the type III and type II receptors interact specifically in a manner dependent on the kinase activity of the type II receptor and the ability of the type II receptor to autophosphorylate. This interaction results in the phosphorylation of the cytoplasmic domain of the type III receptor by the type II receptor. The type III receptor with the cytoplasmic domain deleted is able to bind TGF-beta, to bind the type II receptor, and to enhance TGF-beta binding to the type II receptor but is unable to enhance TGF-beta2 signaling, determining that the cytoplasmic domain is essential for some functions of the type III receptor. The type III receptor functions by selectively binding the autophosphorylated type II receptor via its cytoplasmic domain, thus promoting the preferential formation of a complex between the autophosphorylated type II receptor and the type I receptor and then dissociating from this active signaling complex. These studies, for the first time, elucidate important functional roles of the cytoplasmic domain of the type III receptor and demonstrate that these roles are essential for regulating TGF-beta signaling.


Subject(s)
Activin Receptors, Type I , Cytoplasm/metabolism , Proteoglycans/physiology , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction , Transforming Growth Factor beta/physiology , Animals , COS Cells , Models, Molecular , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Structure-Activity Relationship
13.
Biochem Biophys Res Commun ; 275(2): 631-6, 2000 Aug 28.
Article in English | MEDLINE | ID: mdl-10964715

ABSTRACT

EGFRvIII is the most common deletion variant of the epidermal growth factor receptor and is found in cancers of the brain, breast, ovary, and lung. The complete absence of the receptor in healthy tissues makes it an ideal tumor marker. We sought to design a peptide ligand against EGFRvIII for development as a diagnostic imaging agent. We used the concept of hydropathic complementarity to search for sequences whose amino acid sidechains display a reciprocal pattern of hydropathicity to those of the deletion junction of EGFRvIII. The resulting peptide (PEPHC1) was synthesized and tested for binding to EGFRvIII and EGFR. In in vitro assays, PEPHC1 bound the recombinant EGFRvIII extracellular domain or full-length EGFRvIII solubilized from cell membranes in preference to native EGFR. These results demonstrate the utility of hydropathic complementarity as a basis for the design of highly specific ligands that may prove useful as tumor-targeting agents.


Subject(s)
ErbB Receptors/genetics , Peptides/chemistry , 3T3 Cells , Amino Acid Sequence , Animals , DNA, Complementary , Mice , Molecular Sequence Data , Peptides/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
14.
Biochemistry ; 39(31): 9466-76, 2000 Aug 08.
Article in English | MEDLINE | ID: mdl-10924142

ABSTRACT

Transforming growth factor alpha (TGF-alpha), epidermal growth factor (EGF), and related factors mediate their biological effects by binding to the extracellular domain of the EGF receptor, which leads to activation of the receptor's cytoplasmic tyrosine kinase activity. Much remains to be determined, however, about the detailed molecular mechanism involved in this ligand-induced receptor activation. The determination of the binding mechanism and the related thermodynamic and kinetic parameters are of prime importance. To do so, we have used a surface plasmon resonance-based biosensor (the BIAcore) that allows the real-time recording of the interaction between TGF-alpha and the extracellular domain of the EGF receptor. By immobilizing different biotinylated derivatives of TGF-alpha on the sensor chip surface, we demonstrated that the N-terminus of TGF-alpha is not directly involved in receptor binding. By optimizing experimental conditions and interpreting the biosensor results by several data analysis methods, we were able to show that the data do not fit a simple binding model. Through global analysis of the data using a numerical integration method, we tested several binding mechanisms for the TGF-alpha/EGF receptor interaction and found that a conformational change model best fits the biosensor data. Our results, combined with other analyses, strongly support a receptor activation mechanism in which ligand binding results in a conformation-driven exposure of a dimerization site on the receptor.


Subject(s)
ErbB Receptors/chemistry , ErbB Receptors/metabolism , Extracellular Space/chemistry , Extracellular Space/metabolism , Transforming Growth Factor alpha/chemistry , Transforming Growth Factor alpha/metabolism , Binding, Competitive , Biotinylation/methods , Computer Simulation , Humans , Iodine Radioisotopes/metabolism , Kinetics , Models, Chemical , Protein Conformation , Protein Structure, Tertiary , Streptavidin/chemistry , Streptavidin/metabolism , Surface Plasmon Resonance , Thermodynamics , Transforming Growth Factor alpha/isolation & purification
15.
Exp Cell Res ; 258(1): 121-34, 2000 Jul 10.
Article in English | MEDLINE | ID: mdl-10912794

ABSTRACT

Transforming growth factor-beta (TGF-beta) signaling requires the functional interaction of two distinct receptors, type I (RI) and type II (RII), at the cell surface. Exposure of cells to TGF-beta results in receptor internalization and down-regulation (Zwaagstra et al., 1999, Exp. Cell Res. 252, 352362); however, little is known about the subsequent fate of RI or RII. In this study the cellular distribution of RI was examined in cells before and after treatment with ligand. RI was localized by immunocytochemistry and confocal microscopy using two polyclonal antisera directed against two different epitopes, one in the C-terminal region and one in the N-terminal region of the cytoplasmic domain. The majority of RI molecules in untreated MvlLu and A549 cells were found to be intracellular. Treatment of MvlLu and A549 cells with 100 pM TGF-beta1 for 24 h at 37 degrees C caused a redistribution of surface RI on MvlLu cells, as evidenced by surface RI aggregation. Unexpectedly, this TGF-beta1 treatment also caused redistribution and accumulation of intracellular RI in and around the nucleus for both MvlLu and A549 cells. Nuclear accumulation of RI was also promoted independently of ligand receptor activation by treatment of MvlLu cells with olomoucine, an agent that results in growth arrest. The capacity of RI to localize in the nucleus was confirmed by microscopic examination of 293 cells transiently expressing RI fused to green fluorescent protein (RI-GFP). Olomoucine treatment of these cells resulted in the movement of RI-GFP into the nucleus. Our results indicate that growth arrest alters intracellular transport/routing of RI and may indicate that RI functions not only at the cell surface but inside the cell as well.


Subject(s)
Activin Receptors, Type I , Cell Cycle/physiology , Cell Nucleus/physiology , Protein Serine-Threonine Kinases/physiology , Receptors, Transforming Growth Factor beta/physiology , Animals , Cell Cycle/drug effects , Cell Line , Cell Membrane/physiology , Cyclin-Dependent Kinases/antagonists & inhibitors , Endoplasmic Reticulum/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Golgi Apparatus/physiology , Green Fluorescent Proteins , Humans , Kinetin , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Lung Neoplasms , Mink , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , Purines/pharmacology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/analysis , Receptors, Transforming Growth Factor beta/genetics , Recombinant Fusion Proteins/biosynthesis , Respiratory Mucosa , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured
16.
J Biol Chem ; 275(35): 26748-53, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10840042

ABSTRACT

Epidermal growth factor (EGF) and transforming growth factor-alpha (TGFalpha) are mitogenic hormones that exert their activity primarily by binding to the EGF receptor, also known as ErbB-1. We have recently characterized a set of EGF/TGFalpha chimeric molecules with similar high affinity for ErbB-1 as EGF and TGFalpha and shown that three of these chimeras induce mitogenic cell stimulation at already a 10-fold lower concentration than their wild-type counterparts (Lenferink, A. E., Kramer, R. H., van Vugt, M. J., Königswieser, M., DiFiore, P. P., van Zoelen, E. J., and van de Poll, M. L. (1997) Biochem. J. 327, 859-865). In the present study we show that these so-called superagonistic chimeras do not differ from EGF and TGFalpha in their ability to induce ErbB-1 tyrosine phosphorylation but are considerably more potent in activation of mitogen-activated protein kinase phosphorylation. Direct cell binding studies and analysis of ligand-receptor interaction by surface plasmon resonance measurements revealed that both the association rate constant (k(on)) and the dissociation rate constant (k(off)) of these superagonists is 3-5-fold higher in comparison with the wild-type ligands and nonsuperagonistic chimeras. These data indicate that the dynamic on and off rate constants for receptor binding may be more specific parameters for determining the mitogenic activity of peptide hormones than their constants for equilibrium receptor binding.


Subject(s)
Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , 3T3 Cells , Animals , Epidermal Growth Factor/metabolism , Humans , Kinetics , Mice , Mitogen-Activated Protein Kinases/metabolism , Protein Binding , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Surface Plasmon Resonance
17.
Protein Eng ; 13(3): 143-7, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10775655

ABSTRACT

A heteronuclear NMR study of human transforming growth factor alpha (TGFalpha) in complex with the epidermal growth factor receptor extracellular domain (EGFR-ED) provided an effective method for delineating the relative contributions of the residues of the ligand to its affinity for the receptor. In conjunction with previously obtained mutagenesis data, these results indicate that while a large number of residues are involved in complex formation and make up the binding interface, a small subset contribute most of the binding energy. They also show that while the residues which contribute to receptor binding are localized on one face of the molecule, the specific residues that play the major role in the affinity of TGFalpha in the complex are in two distinct regions of TGFalpha. This suggests that two small functional epitopes each composed of two residues exist within a larger structural epitope presented on the binding face. These results give the most detailed picture to date of the receptor binding determinants and yield further insight into the formation of the ligand-receptor complex.


Subject(s)
ErbB Receptors/chemistry , ErbB Receptors/metabolism , Transforming Growth Factor alpha/chemistry , Transforming Growth Factor alpha/metabolism , Amino Acid Sequence , Binding Sites , Epitopes/chemistry , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Conformation
18.
Exp Cell Res ; 252(2): 352-62, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10527625

ABSTRACT

The types I, II, and III receptors (RI, RII, RIII) for transforming growth factor-beta (TGF-beta) become down-regulated in response to ligand, presumably via their internalization from the cell surface. This report examines the down-regulation of full-length RI, RII, and RIII in cells endogenously or transiently expressing these receptors. Down-regulation occurred rapidly (within 2 h after TGF-beta1 treatment at 37 degrees C) and showed a dose response, between 10 and 200 pM TGF-beta1, in cells expressing RI, RII, and RIII (Mv1lu and A549 cells). A comparison between Mv1Lu and mutant cell derivatives R-1B (lacking RI) or DR-26 (lacking RII) indicated that all three receptors were necessary for efficient down-regulation. Down-regulation experiments, utilizing TGF-beta-treated 293 cells transiently expressing different combinations of these receptors indicated that neither RII or RIII were down-regulated when expressed alone and that RI was required for maximal down-regulation of RII. RII and RIII were partially down-regulated when these receptors were coexpressed in the absence of RI (in R-1B and 293 cells). Surprisingly, TGF-beta receptors were partially down-regulated in Mv1Lu, A549, and 293 cells treated with TGF-beta1 at 4 degrees C. Microscopic examination of 293 cells coexpressing RI fused to green fluorescent protein (RI-GFP) and RII indicated that, after treatment with TGF-beta1 at 4 degrees C, RI-GFP formed aggregates at the cell surface at this temperature. RI-GFP was not detected at the surface of these cells after TGF-beta1 treatment at 37 degrees C. Our results suggest a two phase mechanism for TGF-beta1 receptor down-regulation involving receptor modulation (aggregation) at the cell surface and internalization.


Subject(s)
Cell Membrane/physiology , Down-Regulation , Receptor Aggregation/physiology , Receptors, Transforming Growth Factor beta/physiology , Animals , Cell Line
19.
Growth Factors ; 17(1): 63-73, 1999.
Article in English | MEDLINE | ID: mdl-10495963

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a multi-functional regulator of cell growth and differentiation. Three distinct isoforms of TGF-beta exist having similar, but not identical actions. TGF-beta 1, but not TGF-beta 2, binds to T beta RII and also to endoglin, a cell surface protein abundant on endothelial cells. In contrast, the affinity constant of TGF-beta 2 for alpha 2-macroglobulin is 10-fold greater than that of TGF-beta 1. TGF-beta 2 also binds better than TGF-beta 1 to a glycosyl phosphatidylinositol (GPI)-linked binding protein expressed on vascular endothelial cells. Using chimeric TGF-beta molecules, in which selected regions of TGF-beta 1 have been exchanged for the corresponding region of TGF-beta 2, we demonstrate here that amino acids 92-95 or 94-98 of TGF-beta determine isoform specific binding to endoglin. In contrast, exchange of only amino acids 95 and 98 did not alter TGF-beta specificity. Isoform specific binding to a GPI-linked protein on EJG endothelial cells was modulated by a region containing amino acids 40-68, although exchange of only amino acids 40-47 did not confer isoform specific binding. Significantly, the 92-98 region also modulates binding of TGF-beta to the type II receptor whereas isoform specific binding to alpha 2-macroglobulin requires concerted exchange of amino acids 45 and 47. Taken together, these results show that at least three different functional domains are important modulators of TGF-beta interaction with binding proteins and receptors.


Subject(s)
Endothelium, Vascular/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , alpha-Macroglobulins/metabolism , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cells, Cultured , Cricetinae , Endothelium, Vascular/cytology , Humans , Models, Molecular , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Receptor, Transforming Growth Factor-beta Type II , Sequence Homology, Amino Acid , Structure-Activity Relationship , Transforming Growth Factor beta/chemistry
20.
FEBS Lett ; 456(1): 79-84, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10452534

ABSTRACT

Binding surfaces of the type II transforming growth factor (TGF)-beta receptor extracellular domain (TbetaRII-ECD) are mapped by combining scanning-deletion mutagenesis results with knowledge-based modeling of the ectodomain structure. Of the 17 deletion mutants produced within the core binding domain of TbetaRII-ECD, only three retained binding to TGF-beta. Comparative modeling based on the crystal structure of the activin type II receptor extracellular domain (ActRII-ECD) indicates that the TbetaRII mutants which retain TGF-beta binding are deleted in some of the loops connecting the beta-strands in the TbetaRII-ECD model. Interpretation of the mutagenesis data within the structural framework of the ectodomain model allows for the prediction of potential binding sites at the surface of TbetaRII-ECD.


Subject(s)
Computer Simulation , Models, Molecular , Mutagenesis , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Artificial Intelligence , Binding Sites , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Glycosylation , Humans , Molecular Sequence Data , Protein Conformation , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Sequence Deletion , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...