Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2217: 3-15, 2021.
Article in English | MEDLINE | ID: mdl-33215372

ABSTRACT

Soluble ligand and conformation-dependent antibody binding assay of integrins on the cell surface is an effective approach to evaluate the activation status of integrins in live cells. The ligands or antibodies are usually labeled with biotin or a fluorescent dye and incubated with integrin-expressing cells in suspension. The cell-bound ligands and antibodies are then detected by flow cytometry. Here we describe the detailed protocols of soluble ligand or antibody binding assay for αIIbß3, αVß3, α5ß1, and αLß2 integrins that are transiently or stably expressed in the model cell lines such as HEK293 or CHO-k1 cells.


Subject(s)
Biological Assay , Integrin alphaVbeta3/chemistry , Lymphocyte Function-Associated Antigen-1/chemistry , Platelet Membrane Glycoprotein IIb/chemistry , Receptors, Vitronectin/chemistry , Staining and Labeling/methods , Animals , Antibodies/chemistry , Antibodies/metabolism , CHO Cells , Cell Adhesion , Cricetulus , Flow Cytometry , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Intercellular Adhesion Molecule-1 , Ligands , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Plasmids/chemistry , Plasmids/metabolism , Platelet Membrane Glycoprotein IIb/genetics , Platelet Membrane Glycoprotein IIb/metabolism , Protein Binding , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
2.
Structure ; 27(9): 1443-1451.e6, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31353240

ABSTRACT

Targeting both integrins αVß3 and α5ß1 simultaneously appears to be more effective in cancer therapy than targeting each one alone. The structural requirements for bispecific binding of ligand to integrins have not been fully elucidated. RGD-containing knottin 2.5F binds selectively to αVß3 and α5ß1, whereas knottin 2.5D is αVß3 specific. To elucidate the structural basis of this selectivity, we determined the structures of 2.5F and 2.5D as apo proteins and in complex with αVß3, and compared their interactions with integrins using molecular dynamics simulations. These studies show that 2.5D engages αVß3 by an induced fit, but conformational selection of a flexible RGD loop accounts for high-affinity selective binding of 2.5F to both integrins. The contrasting binding of the highly flexible low-affinity linear RGD peptides to multiple integrins suggests that a "Goldilocks zone" of conformational flexibility of the RGD loop in 2.5F underlies its selective binding promiscuity to integrins.


Subject(s)
Cystine-Knot Miniproteins/metabolism , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Receptors, Vitronectin/chemistry , Receptors, Vitronectin/metabolism , Binding Sites , Humans , Integrin alphaVbeta3/genetics , K562 Cells , Models, Molecular , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation , Receptors, Vitronectin/genetics
3.
ACS Comb Sci ; 21(8): 598-607, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31269394

ABSTRACT

We report the identification of high-affinity and selectivity integrin α5ß1-binding bicyclic peptides via "designed random libraries", that is, the screening of libraries comprising the universal integrin-binding sequence Arg-Gly-Asp (RGD) in the first loop in combination with a randomized sequence (XXX) in the second loop. Screening of first-generation libraries for α5ß1-binding peptides yielded a triple-digit nanomolar bicyclic α5ß1-binder (CT3RGDcT3AYGCT3, IC50 = 406 nM). Next-generation libraries were designed by partially varying the structure of the strongest first-generation lead inhibitor and screened for improved affinities and selectivities for this receptor. In this way, we identified three high-affinity α5ß1-binders (CT3RGDcT3AYJCT3, J = d-Leu, IC50 = 90 nM; CT3RGDcT3AYaCT3, IC50 = 156 nM; CT3RGDcT3AWGCT3, IC50 = 173 nM), of which one even showed a higher α5ß1-affinity than the 32 amino acid benchmark peptide knottin-RGD (IC50 = 114 nM). Affinity for α5ß1-integrin was confirmed by SPFS analysis showing a Kd of 4.1 nM for Cy5-labeled RGD-bicycle CT3RGDcT3AYJCT3 (J = d-Leu) and a somewhat higher Kd (9.0 nM) for Cy5-labeled knottin-RGD. The α5ß1-bicycles, for example, CT3RGDcT3AYJCT3 (J = d-Leu), showed excellent selectivities over αvß5 (IC50 ratio α5ß1/αvß5 between <0.009 and 0.039) and acceptable selectivities over αvß3 (IC50 ratios α5ß1/αvß3 between 0.090 and 0.157). In vitro staining of adipose-derived stem cells with Cy5-labeled peptides using confocal microscopy revealed strong binding of the α5ß1-selective bicycle CT3RGDcT3AWGCT3 to integrins in their natural environment, illustrating the high potential of these RGD bicycles as markers for α5ß1-integrin expression.


Subject(s)
Oligopeptides/analysis , Peptide Library , Receptors, Vitronectin/chemistry , Combinatorial Chemistry Techniques , Oligopeptides/chemical synthesis
4.
ChemMedChem ; 14(14): 1315-1320, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31207080

ABSTRACT

Up to 45 % of deaths in developed nations can be attributed to chronic fibroproliferative diseases, highlighting the need for effective therapies. The RGD (Arg-Gly-Asp) integrin αvß1 was recently investigated for its role in fibrotic disease, and thus warrants therapeutic targeting. Herein we describe the identification of non-RGD hit small-molecule αvß1 inhibitors. We show that αvß1 activity is embedded in a range of published α4ß1 (VLA-4) ligands; we also demonstrate how a non-RGD integrin inhibitor (of α4ß1 in this case) was converted into a potent non-zwitterionic RGD integrin inhibitor (of αvß1 in this case). We designed urea ligands with excellent selectivity over α4ß1 and the other αv integrins (αvß3, αvß5, αvß6, αvß8). In silico docking models and density functional theory (DFT) calculations aided the discovery of the lead urea series.


Subject(s)
Phenylalanine/analogs & derivatives , Receptors, Vitronectin/antagonists & inhibitors , Urea/analogs & derivatives , Animals , Binding Sites , Drug Design , Drug Stability , Humans , Ligands , Liver/metabolism , Male , Phenylalanine/chemical synthesis , Phenylalanine/metabolism , Rats, Sprague-Dawley , Receptors, Vitronectin/chemistry , Receptors, Vitronectin/metabolism , Urea/chemical synthesis , Urea/metabolism
5.
Biomed Mater ; 14(3): 035009, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30630151

ABSTRACT

Biomaterial design in tissue engineering aims to identify appropriate cellular microenvironments in which cells can grow and guide new tissue formation. Despite the large diversity of synthetic polymers available for regenerative medicine, most of them fail to fully match the functional properties of their native counterparts. In contrast, the few biological alternatives employed as biomaterials lack the versatility that chemical synthesis can offer. Herein, we studied the HUVEC adhesion and proliferation properties of elastin-like recombinamers (ELRs) that were covalently functionalized with each three high-affinity and selectivity α v ß 3- and α 5 ß 1-binding bicyclic RGD peptides. Next to the bicycles, ELRs were also functionalized with various integrin-binding benchmark peptides, i.e. knottin-RGD, cyclo-[KRGDf] and GRGDS, allowing for better classification of the obtained results. Covalent functionalization with the RGD peptides, as validated by MALDI-TOF analysis, guarantees flexibility and minimal steric hindrance for interactions with cellular integrins. In addition to the covalently modified RGD-ELRs, we also synthesized another benchmark ELR comprising RGD as part of the backbone. HUVEC adhesion and proliferation analysis using the PicoGreen® assay revealed a higher short-term adhesion and proliferative capacity of cells on ELR surfaces functionalized with high affinity, integrin-binding bicyclic RGD-peptides compared with the ELRs containing RGD in the backbone.


Subject(s)
Biocompatible Materials/chemistry , Cell Adhesion/drug effects , Elastin/chemistry , Genetic Engineering/methods , Integrin alphaVbeta3/chemistry , Oligopeptides/chemistry , Receptors, Vitronectin/chemistry , Cell Proliferation , Cells, Cultured , Human Umbilical Vein Endothelial Cells , Humans , Peptides/chemistry , Polymers/chemistry , Protein Binding , Regenerative Medicine , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Engineering
6.
Eur J Pharmacol ; 842: 239-247, 2019 Jan 05.
Article in English | MEDLINE | ID: mdl-30389632

ABSTRACT

Compound 8 is a selective αvß1 small molecule inhibitor that has been used in pre-clinical studies to identify and characterise the αvß1 integrin as a potential target in fibrotic disease. In this study we further investigated the selectivity and pharmacokinetics of compound 8 to determine a link between the levels of αvß1 engagement required to achieve in vivo pharmacodynamic efficacy. The selectivity of compound 8 for the arginyl-glycinyl-aspartic acid and ß1 integrins was measured using purified integrin protein preparations in radioligand binding studies with both labelled ([3H]compound 8) and unlabelled versions. The pharmacokinetic profile of compound 8 was completed in in vitro blood protein binding assays and in in vivo studies using male C57BL/6 mouse following i.v. dosing. The high selectivity of compound 8 for αvß1 over the other αv integrins was confirmed, however a reduced selectivity was demonstrated for the ß1 integrin family, with high affinity observed for α4ß1 (comparable to αvß1), moderate affinity for α2ß1, α3ß1 and α8ß1, and low affinity for α5ß1 and α9ß1. Compound 8 was shown to be cleared quickly from the blood with a short half-life of 0.5 h. In conclusion, the data in this study suggest that compound 8 has the potential to engage a number of integrins in vivo beyond αvß1, that raises a degree of uncertainty regarding its mechanism of action in models of fibrotic disease.


Subject(s)
Oligopeptides/chemistry , Peptidomimetics/pharmacology , Receptors, Vitronectin/metabolism , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Peptidomimetics/chemistry , Peptidomimetics/metabolism , Peptidomimetics/pharmacokinetics , Receptors, Vitronectin/chemistry
7.
BMC Biol ; 16(1): 92, 2018 08 17.
Article in English | MEDLINE | ID: mdl-30119679

ABSTRACT

BACKGROUND: Increased activity of the receptor tyrosine kinase Tie2 has been implicated in the promotion of pathological angiogenesis. This activity is mainly mediated through angiopoietin (Ang)1- and Ang2-dependent activation of integrins by Tie2, rendering the Ang/Tie2/integrin axis an attractive putative target for cancer therapeutics. RESULTS: To target this axis, we developed single domain, non-immunoglobulin high-affinity bi-specific protein inhibitors against both Tie2 and αvß3 integrin. We have previously engineered the Ang2-binding domain of Tie2 (Ang2-BD) as a Tie2 inhibitor. Here, we engineered an exposed loop in Ang2-BD to generate variants that include an integrin-binding Arg-Gly-Asp (RGD) motif and used flow cytometry screening of a yeast-displayed Ang2-BD RGD loop library to identify the integrin antagonists. The bi-specific antagonists targeting both Tie2 and αvß3 integrin inhibited adhesion and proliferation of endothelial cells cultured together with the αvß3 integrin ligand vitronectin, as well as endothelial cell invasion and tube formation. The bi-specific reagents inhibited downstream signaling by Tie2 intracellularly in response to its agonist Ang1 more effectively than the wild-type Ang2 BD that binds Tie2 alone. CONCLUSIONS: Collectively, this study-the first to describe inhibitors targeting all the known functions resulting from Tie2/integrin αvß3 cross-talk-has created new tools for studying Tie2- and integrin αvß3-dependent molecular pathways and provides the basis for the rational and combinatorial engineering of ligand-Tie2 and ligand-integrin αvß3 receptor interactions. Given the roles of these pathways in cancer angiogenesis and metastasis, this proof of principle study paves the route to create novel Tie2/integrin αvß3-targeting proteins for clinical use as imaging and therapeutic agents.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neovascularization, Physiologic/genetics , Receptor, TIE-2/antagonists & inhibitors , Receptors, Vitronectin/genetics , Ribonuclease, Pancreatic/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Animals , Mice , Receptor, TIE-2/chemistry , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Receptors, Vitronectin/chemistry , Receptors, Vitronectin/metabolism , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism
8.
J Fluoresc ; 26(1): 241-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26527222

ABSTRACT

Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome results in respiratory obstruction and severe lung inflammation. Critical characteristics of ALI are alveolar edema, infiltration of leukocytes (neutrophils and monocytes), release of pro-inflammatory cytokines and chemokines into broncho-alveolar lavage fluid, and activation of integrin receptors. The purpose of the study was to demonstrate non-invasive detection of lung inflammation using integrin receptor targeted fluorescence liposomes. An inflammation similar to that observed in ALI was elicited in rodents by intra-tracheal instillation of interleukin-1beta (IL-1beta). Cyclic arginine glycine-(D)-aspartic acid-peptide (cRGD-peptide) grafted fluorescence liposomes were administered to ALI induced male Sprague-Dawley rats for targeting lung integrin receptors. Near-infrared fluorescence imaging (NIRFI) was applied for visualization and quantitation of lung inflammation. NIRFI signals were correlated with inflammatory cellular and biochemical markers of lungs. A positive correlation was observed between NIRF signals and lung inflammation markers. Compared to control group, an intense NIRF signal was observed in ALI induced rats in the window 6-24 h post-IL-1beta instillation. Interaction of integrin receptors with targeted liposomes was assumed to contribute to intense NIRF signal. RT-PCR studies showed an elevated lung expression of alphavbeta5 integrin receptors, 12 h post-IL-1beta instillation. In vitro studies demonstrated integrin receptor specificity of targeted liposomes. These targeted liposomes showed binding to alphavbeta5 integrin receptors expressed on alveolar cells. Non-invasive detection of lung inflammation was demonstrated using a combination of integrin receptor targeting and NIRFI.


Subject(s)
Acute Lung Injury/diagnosis , Acute Lung Injury/metabolism , Fluorescence , Liposomes/chemistry , Pneumonia/diagnosis , Pneumonia/metabolism , Animals , Cell Line , Humans , Integrins/genetics , Integrins/metabolism , Male , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Receptors, Vitronectin/chemistry , Receptors, Vitronectin/metabolism
9.
Chemistry ; 22(2): 681-93, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26548575

ABSTRACT

The critical role of integrins in tumor progression and metastasis has stimulated intense efforts to identify pharmacological agents that can modulate integrin function. In recent years, αv ß3 and αv ß5 integrin antagonists were demonstrated to be effective in blocking tumor progression. RGDechi-hCit, a chimeric peptide containing a cyclic RGD motif linked to an echistatin C-terminal fragment, is able to recognize selectively αv ß3 integrin both in vitro and in vivo. High-resolution molecular details of the selective αv ß3 recognition of the peptide are certainly required, nonetheless RGDechi-hCit internalization limited the use of classical in cell NMR experiments. To overcome such limitations, we used WM266 isolated cellular membranes to accomplish a detailed NMR interaction study that, combined with a computational analysis, provides significant structural insights into αv ß3 molecular recognition by RGDechi-hCit. Remarkably, on the basis of the identified molecular determinants, we design a RGDechi-hCit mutant that is selective for αv ß5 integrin.


Subject(s)
Cell Membrane/chemistry , Integrin alphaVbeta3/chemistry , Magnetic Resonance Spectroscopy , Oligopeptides/chemistry , Peptides/chemistry , Receptors, Vitronectin/chemistry , Cell Membrane/metabolism , Computers, Molecular , Integrin alphaVbeta3/metabolism , Intercellular Signaling Peptides and Proteins , Ligands , Peptides/metabolism , Receptors, Vitronectin/metabolism
10.
J Biol Chem ; 290(1): 259-71, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25398877

ABSTRACT

Integrins are activated by signaling from inside the cell (inside-out signaling) through global conformational changes of integrins. We recently discovered that fractalkine activates integrins in the absence of CX3CR1 through the direct binding of fractalkine to a ligand-binding site in the integrin headpiece (site 2) that is distinct from the classical RGD-binding site (site 1). We propose that fractalkine binding to the newly identified site 2 induces activation of site 1 though conformational changes (in an allosteric mechanism). We reasoned that site 2-mediated activation of integrins is not limited to fractalkine. Human secreted phospholipase A2 type IIA (sPLA2-IIA), a proinflammatory protein, binds to integrins αvß3 and α4ß1 (site 1), and this interaction initiates a signaling pathway that leads to cell proliferation and inflammation. Human sPLA2-IIA does not bind to M-type receptor very well. Here we describe that sPLA2-IIA directly activated purified soluble integrin αvß3 and transmembrane αvß3 on the cell surface. This activation did not require catalytic activity or M-type receptor. Docking simulation predicted that sPLA2-IIA binds to site 2 in the closed-headpiece of αvß3. A peptide from site 2 of integrin ß1 specifically bound to sPLA2-IIA and suppressed sPLA2-IIA-induced integrin activation. This suggests that sPLA2-IIA activates αvß3 through binding to site 2. sPLA2-IIA also activated integrins α4ß1 and α5ß1 in a site 2-mediated manner. We recently identified small compounds that bind to sPLA2-IIA and suppress integrin-sPLA2-IIA interaction (e.g. compound 21 (Cmpd21)). Cmpd21 effectively suppressed sPLA2-IIA-induced integrin activation. These results define a novel mechanism of proinflammatory action of sPLA2-IIA through integrin activation.


Subject(s)
Group II Phospholipases A2/chemistry , Integrin alpha4beta1/chemistry , Integrin alphaVbeta3/chemistry , Receptors, Vitronectin/chemistry , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cricetulus , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Group II Phospholipases A2/antagonists & inhibitors , Group II Phospholipases A2/genetics , Group II Phospholipases A2/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , K562 Cells , Models, Molecular , Molecular Docking Simulation , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Signal Transduction
11.
PLoS One ; 9(5): e96372, 2014.
Article in English | MEDLINE | ID: mdl-24789099

ABSTRACT

The chemokine domain of fractalkine (FKN-CD) binds to the classical RGD-binding site of αvß3 and that the resulting ternary complex formation (integrin-FKN-CX3CR1) is critical for CX3CR1 signaling and FKN-induced integrin activation. However, only certain cell types express CX3CR1. Here we studied if FKN-CD can activate integrins in the absence of CX3CR1. We describe that WT FKN-CD activated recombinant soluble αvß3 in cell-free conditions, but the integrin-binding defective mutant of FKN-CD (K36E/R37E) did not. This suggests that FKN-CD can activate αvß3 in the absence of CX3CR1 through the direct binding of FKN-CD to αvß3. WT FKN-CD activated αvß3 on CX3CR1-negative cells (K562 and CHO) but K36E/R37E did not, suggesting that FKN-CD can activate integrin at the cellular levels in a manner similar to that in cell-free conditions. We hypothesized that FKN-CD enhances ligand binding to the classical RGD-binding site (site 1) through binding to a second binding site (site 2) that is distinct from site 1 in αvß3. To identify the possible second FKN-CD binding site we performed docking simulation of αvß3-FKN-CD interaction using αvß3 with a closed inactive conformation as a target. The simulation predicted a potential FKN-CD-binding site in inactive αvß3 (site 2), which is located at a crevice between αv and ß3 on the opposite side of site 1 in the αvß3 headpiece. We studied if FKN-CD really binds to site 2 using a peptide that is predicted to interact with FKN-CD in site 2. Notably the peptide specifically bound to FKN-CD and effectively suppressed integrin activation by FKN-CD. This suggests that FKN-CD actually binds to site 2, and this leads to integrin activation. We obtained very similar results in α4ß1 and α5ß1. The FKN binding to site 2 and resulting integrin activation may be a novel mechanism of integrin activation and of FKN signaling.


Subject(s)
Chemokine CX3CL1/metabolism , Integrin alphaVbeta3/metabolism , Oligopeptides/metabolism , Receptors, Chemokine/metabolism , ADAM Proteins/chemistry , ADAM Proteins/genetics , ADAM Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , CHO Cells , CX3C Chemokine Receptor 1 , Chemokine CX3CL1/chemistry , Chemokine CX3CL1/genetics , Cricetinae , Cricetulus , Humans , Integrin alpha4beta1/chemistry , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/genetics , K562 Cells , Ligands , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Mutation , Oligopeptides/chemistry , Oligopeptides/genetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Chemokine/genetics , Receptors, Vitronectin/chemistry , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , U937 Cells
12.
Protein Expr Purif ; 89(2): 225-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23583935

ABSTRACT

A recombinant integrin expression system has been created for the large-scale production of αVß5 integrin extracellular domains that take advantage of Fos and Jun dimerization for expression in bacterial, insect, and mammalian cells. This utilizes an all-in-one vector, pQE-TriSystem, with molecular machinery for parallel expression without the need of additional subcloning. Optimal expression in HEK293 cells was determined by a time course analysis. The heterodimer was purified in a one-step nickel column purification scheme, and the sequence and functional state were confirmed by mass spectrometry and inhibition assays, respectively. The yields of αVß5 integrin obtained are in quantities suitable for multiple applications including structural biology and functional assays.


Subject(s)
Receptors, Vitronectin/genetics , Receptors, Vitronectin/isolation & purification , Amino Acid Sequence , Animals , Gene Expression , Genetic Vectors/genetics , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Plasmids/genetics , Protein Conformation , Protein Folding , Protein Multimerization , Receptors, Vitronectin/chemistry , Receptors, Vitronectin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
13.
Biochem Biophys Res Commun ; 422(2): 207-12, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22560905

ABSTRACT

The human CD23 protein binds to αvß3 and αvß5 integrins. The integrins recognize a short tripeptide motif of arg-lys-cys (RKC) in CD23, and peptides containing this motif inhibit the binding of CD23 to B cells and monocytes; neither fibronectin, nor vitronectin, which contain arg-gly-asp motifs, inhibit binding of RKC-containing peptides to cells. RKC-containing peptides derived from CD23 show dose-dependent, biphasic binding profiles to both αvß3 and αvß5 that are cation-independent but sensitive to high chloride ion concentrations. Substitution of one basic residue in the RKC motif with alanine reduces but does not abolish integrin binding or the ability of peptides to stimulate pre-B cell growth or cytokine release by monocytes. Substitution of both basic residues abolishes both integrin binding and biological activity of CD23-derived peptides. These features indicate that binding of RKC-containing peptides to αv integrins has clearly distinct characteristics to those for binding of RGD-containing ligands.


Subject(s)
Integrin alphaV/metabolism , Integrin alphaVbeta3/metabolism , Receptors, IgE/metabolism , Receptors, Vitronectin/metabolism , Amino Acid Sequence , Amino Acid Substitution , Cell Line , Humans , Integrin alphaV/chemistry , Integrin alphaVbeta3/chemistry , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Receptors, IgE/chemistry , Receptors, IgE/genetics , Receptors, Vitronectin/chemistry , Surface Plasmon Resonance
14.
Chemistry ; 18(20): 6195-207, 2012 May 14.
Article in English | MEDLINE | ID: mdl-22517378

ABSTRACT

The synthesis of eight bifunctional diketopiperazine (DKP) scaffolds is described; these were formally derived from 2,3-diaminopropionic acid and aspartic acid (DKP-1-DKP-7) or glutamic acid (DKP-8) and feature an amine and a carboxylic acid functional group. The scaffolds differ in the configuration at the two stereocenters and the substitution at the diketopiperazinic nitrogen atoms. The bifunctional diketopiperazines were introduced into eight cyclic peptidomimetics containing the Arg-Gly-Asp (RGD) sequence. The resulting RGD peptidomimetics were screened for their ability to inhibit biotinylated vitronectin binding to the purified integrins α(v)ß(3) and α(v)ß(5), which are involved in tumor angiogenesis. Nanomolar IC(50) values were obtained for the RGD peptidomimetics derived from trans DKP scaffolds (DKP-2-DKP-8). Conformational studies of the cyclic RGD peptidomimetics by (1)H NMR spectroscopy experiments (VT-NMR and NOESY spectroscopy) in aqueous solution and Monte Carlo/Stochastic Dynamics (MC/SD) simulations revealed that the highest affinity ligands display well-defined preferred conformations featuring intramolecular hydrogen-bonded turn motifs and an extended arrangement of the RGD sequence [Cß(Arg)-Cß(Asp) average distance ≥8.8 Å]. Docking studies were performed, starting from the representative conformations obtained from the MC/SD simulations and taking as a reference model the crystal structure of the extracellular segment of integrin α(v)ß(3) complexed with the cyclic pentapeptide, Cilengitide. The highest affinity ligands produced top-ranked poses conserving all the important interactions of the X-ray complex.


Subject(s)
Diketopiperazines/chemistry , Models, Molecular , Oligopeptides/chemistry , Peptides, Cyclic/chemistry , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Integrins/metabolism , Ligands , Molecular Conformation , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Receptors, Vitronectin/chemistry , Receptors, Vitronectin/metabolism
17.
Sci Transl Med ; 2(45): 45ra60, 2010 Aug 18.
Article in English | MEDLINE | ID: mdl-20720217

ABSTRACT

Engineered biointerfaces covered with biomimetic motifs, including short bioadhesive ligands, are a promising material-based strategy for tissue repair in regenerative medicine. Potentially useful coating molecules are ligands for the integrins, major extracellular matrix receptors that require both ligand binding and nanoscale clustering for maximal signaling efficiency. We prepared coatings consisting of well-defined multimer constructs with a precise number of recombinant fragments of fibronectin (monomer, dimer, tetramer, and pentamer) to assess how nanoscale ligand clustering affects integrin binding, stem cell responses, tissue healing, and biomaterial integration. Clinical-grade titanium was grafted with polymer brushes that presented monomers, dimers, trimers, or pentamers of the alpha(5)beta(1) integrin-specific fibronectin III (7 to 10) domain (FNIII(7-10)). Coatings consisting of trimers and pentamers enhanced integrin-mediated adhesion in vitro, osteogenic signaling, and differentiation in human mesenchymal stem cells more than did surfaces presenting monomers and dimers. Furthermore, ligand clustering promoted bone formation and functional integration of the implant into bone in rat tibiae. This study establishes that a material-based strategy in which implants are coated with clustered bioadhesive ligands can promote robust implant-tissue integration.


Subject(s)
Biocompatible Materials/pharmacology , Fibronectins/metabolism , Receptors, Vitronectin/metabolism , Wound Healing/drug effects , Animals , Binding Sites , Fibronectins/chemistry , Humans , Implants, Experimental , Ligands , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Nanostructures/chemistry , Osseointegration/drug effects , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Receptors, Vitronectin/chemistry , Substrate Specificity
18.
Adv Exp Med Biol ; 664: 123-31, 2010.
Article in English | MEDLINE | ID: mdl-20238010

ABSTRACT

The apical plasma membrane domain of retinal pigment epithelial (RPE) cells in the eye faces the outer segment portions of rods and cones and the interphotoreceptor matrix in the subretinal space. Two important receptor-mediated interactions between the apical surface of the retinal pigment epithelium (RPE) and adjacent photoreceptors are adhesion ensuring outer segment alignment and diurnal phagocytosis of shed outer segment fragments contributing to outer segment renewal. Both depend on the apical distribution of the integrin family adhesion receptor alphavbeta5 as lack of alphavbeta5 in mice causes weakened retinal adhesion and asynchronous phagocytosis. With age, lack of alphavbeta5 leads to accumulation of harmful lipofuscin in the RPE and to vision loss. Here, we discuss three different possible mechanisms that could generate the exclusive apical distribution of alphavbeta5 integrin receptors in the RPE. (1) alphavbeta5 could be apical in the RPE because RPE attachment to neural retina generally or alphavbeta5 ligands specifically in the subretinal space stabilize apical but not basolateral alphavbeta5 surface receptors. (2) alphavbeta5 could be apical in the RPE because it resides in a complex with other components of the phagocytic machinery that assembles at the apical, phagocytic surface of the RPE. (3) alphavbeta5 could be apical due to mechanisms intrinsic to this receptor protein and specifically to its beta5 integrin subunit.


Subject(s)
Cell Polarity , Receptors, Vitronectin/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Adhesion , Eye Proteins/metabolism , Humans , Mice , Models, Biological , Phagocytosis , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Vitronectin/chemistry , c-Mer Tyrosine Kinase
19.
Amino Acids ; 38(1): 329-37, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19267182

ABSTRACT

3-Aza-6,8-dioxabicyclo[3.2.1]octane-based amino acids as reverse turn inducers have been introduced into cyclic peptidomimetics containing the RGD or DGR retro-sequence, in order to achieve a stereochemical scanning of the binding capability of the resulting molecules towards alpha(v)beta(3) and alpha(v)beta(5) integrins, resulting in retro-inverso DGR peptides as micromolar ligands. A comparative analysis between the conformational preferences of 4 and of its isomer 3, having the opposite RGD sequence, was reported with respect to the binding activity, giving insight into the factors affecting the preferential binding of 4 to the alpha(v)beta(5) integrin.


Subject(s)
Peptides, Cyclic/chemistry , Receptors, Vitronectin/chemistry , Female , Humans , Kinetics , Ligands , Peptides, Cyclic/agonists , Peptides, Cyclic/chemical synthesis , Placenta/chemistry , Placenta/metabolism , Pregnancy , Protein Binding , Receptors, Vitronectin/metabolism
20.
Mini Rev Med Chem ; 9(12): 1439-46, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19929817

ABSTRACT

Integrins are a large family of dimeric receptors composed by alpha and beta subunits that, once bound to extra-cellular matrix (ECM) proteins, regulate a variety of cellular processes such as cell motility, migration, and proliferation. The integrins transduce signals from inside-out and outside-in the cell, thus representing the cellular link to the external environment. For these properties, integrin activation has been involved in pathological processes like tumor growth and metastasis formation. Recent advances in the elucidation of the crystallographic structures of the alphavbeta3 and alphaIIbeta3 integrins are promoting studies focused to the search of small molecule antagonists that can block the integrin binding to ECM and inhibit the biological effects exerted by these receptors. In this review we will focus on small molecule antagonists of alphavbeta3 and alphavbeta5 integrin as tools for cancer therapy while other integrins will only be briefly mentioned. Cilengitide (cyclic peptidic alphavbeta3 and alphavbeta5 antagonist) is currently in clinical trials for anti cancer therapy. Combination of integrin alphavbeta3 antagonists and other traditional therapeutic approaches may represent a future strategy to inhibit tumor growth and metastasis spreading.


Subject(s)
Integrins/antagonists & inhibitors , Neoplasms/drug therapy , Anoikis , Humans , Indoles/chemistry , Indoles/pharmacology , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Integrins/chemistry , Integrins/metabolism , Peptides/chemistry , Peptides/pharmacology , Receptors, Vitronectin/antagonists & inhibitors , Receptors, Vitronectin/chemistry , Receptors, Vitronectin/metabolism , Signal Transduction , Snake Venoms/chemistry , Snake Venoms/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL