RESUMO
Integrin α5ß1 mediates cell adhesion to the extracellular matrix by binding fibronectin (Fn). Selectivity for Fn by α5ß1 is achieved through recognition of an RGD motif in the 10th type III Fn domain (Fn10) and the synergy site in the ninth type III Fn domain (Fn9). However, details of the interaction dynamics are unknown. Here, we compared synergy-site and Fn-truncation mutations for their α5ß1-binding affinities and stabilities. We also interrogated binding of the α5ß1 ectodomain headpiece fragment to Fn using hydrogen-deuterium exchange (HDX) mass spectrometry to probe binding sites and sites of integrin conformational change. Our results suggest the synergistic effect of Fn9 requires both specific residues and a folded domain. We found some residues considered important for synergy are required for stability. Additionally, we show decreases in fibronectin HDX are localized to a synergy peptide containing contacting residues in two ß-strands, an intervening loop in Fn9, and the RGD-containing loop in Fn10, indicative of binding sites. We also identified binding sites in the α5-subunit ß-propeller domain for the Fn9 synergy site and in the ß1-subunit ßI domain for Fn10 based on decreases in α5ß1 HDX. Interestingly, the dominant effect of Fn binding was an increase in α5ß1 deuterium exchange distributed over multiple sites that undergo changes in conformation or solvent accessibility and appear to be sites where energy is stored in the higher-energy, open-integrin conformation. Together, our results highlight regions important for α5ß1 binding to Fn and dynamics associated with this interaction.
Assuntos
Fibronectinas , Integrina alfa5beta1 , Domínios e Motivos de Interação entre Proteínas , Sítios de Ligação , Adesão Celular , Medição da Troca de Deutério , Fibronectinas/química , Fibronectinas/genética , Integrina alfa5beta1/química , Mutação , Oligopeptídeos/química , SolventesRESUMO
We show that the three conformational states of integrin α5ß1 have discrete free energies and define activation by measuring intrinsic affinities for ligand of each state and the equilibria linking them. The 5,000-fold higher affinity of the extended-open state than the bent-closed and extended-closed states demonstrates profound regulation of affinity. Free energy requirements for activation are defined with protein fragments and intact α5ß1 On the surface of K562 cells, α5ß1 is 99.8% bent-closed. Stabilization of the bent conformation by integrin transmembrane and cytoplasmic domains must be overcome by cellular energy input to stabilize extension. Following extension, headpiece opening is energetically favored. N-glycans and leg domains in each subunit that connect the ligand-binding head to the membrane repel or crowd one another and regulate conformational equilibria in favor of headpiece opening. The results suggest new principles for regulating signaling in the large class of receptors built from extracellular domains in tandem with single-span transmembrane domains.
Assuntos
Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Linhagem Celular , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , TermodinâmicaRESUMO
Integrins are heterodimeric transmembrane proteins able to connect cells with the micro-environment. They represent a family of receptors involved in almost all the hallmarks of cancer. Integrins recognizing the Arg-Gly-Asp (RGD) peptide in their natural extracellular matrix ligands have been particularly investigated as tumoral therapeutic targets. In the last 30â years, intense research has been dedicated to designing specific RGD-like ligands able to discriminate selectively the different RGD-recognizing integrins. Chemists' efforts have led to the proposition of modified peptide or peptidomimetic libraries to be used for tumor targeting and/or tumor imaging. Here we review, from the biological point of view, the rationale underlying the need to clearly delineate each RGD-integrin subtype by selective tools. We describe the complex roles of RGD-integrins (mainly the most studied αvß3 and α5ß1 integrins) in tumors, the steps towards selective ligands and the current usefulness of such ligands. Although the impact of integrins in cancer is well acknowledged, the biological characteristics of each integrin subtype in a specific tumor are far from being completely resolved. Selective ligands might help us to reconsider integrins as therapeutic targets in specific clinical settings.
Assuntos
Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Neoplasias/patologia , Oligopeptídeos/metabolismo , Animais , Humanos , Integrina alfa5beta1/química , Integrina alfaVbeta3/química , Ligantes , Neoplasias/diagnóstico , Neoplasias/metabolismo , Oligopeptídeos/química , Peptidomiméticos/química , Peptidomiméticos/metabolismo , Ligação ProteicaRESUMO
Cells regulate adhesion in response to internally generated and externally applied forces. Integrins connect the extracellular matrix to the cytoskeleton and provide cells with mechanical anchorages and signaling platforms. Here we show that cyclic forces applied to a fibronectin-integrin α5ß1 bond switch the bond from a short-lived state with 1 s lifetime to a long-lived state with 100 s lifetime. We term this phenomenon "cyclic mechanical reinforcement," as the bond strength remembers the history of force application and accumulates over repeated cycles, but does not require force to be sustained. Cyclic mechanical reinforcement strengthens the fibronectin-integrin α5ß1 bond through the RGD binding site of the ligand with the synergy binding site greatly facilitating the process. A flexible integrin hybrid domain is also important for cyclic mechanical reinforcement. Our results reveal a mechanical regulation of receptor-ligand interactions and identify a molecular mechanism for cell adhesion strengthening by cyclic forces.
Assuntos
Adesão Celular , Fibronectinas/química , Integrina alfa5beta1/química , Fenômenos Biomecânicos , Fibronectinas/fisiologia , Humanos , Integrina alfa5beta1/fisiologia , Molécula 1 de Adesão Intercelular/química , Molécula 1 de Adesão Intercelular/fisiologia , Células Jurkat , Membranas Artificiais , Microscopia de Força Atômica , Modelos Moleculares , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/fisiologia , Poliestirenos/química , Ligação ProteicaRESUMO
Cancer cell-matrix interactions have been shown to enhance cancer cell survival via the activation of pro-survival signaling pathways. These pathways are initiated at the site of interaction, i.e., integrins, and thus, their inhibition has been the target of therapeutic strategies. Individual roles for fibronectin-binding integrin subtypes αvß3 and α5ß1 have been shown for various cellular processes; however, a systematic comparison of their function in adhesion-dependent chemoresistance is lacking. Here, we utilize integrin subtype-specific peptidomimetics for αvß3 and α5ß1, both as blocking agents on fibronectin-coated surfaces and as surface-immobilized adhesion sites, in order to parse out their role in breast cancer cell survival. Block copolymer micelle nanolithography is utilized to immobilize peptidomimetics onto highly ordered gold nanoparticle arrays with biologically relevant interparticle spacings (35, 50, or 70 nm), thereby providing a platform for ascertaining the dependence of ligand spacing in chemoprotection. We show that several cellular properties-morphology, focal adhesion formation, and migration-are intricately linked to both the integrin subtype and their nanospacing. Importantly, we show that chemotherapeutic drug sensitivity is highly dependent on both parameters, with smaller ligand spacing generally hindering survival. Furthermore, we identify ligand type-specific patterns of drug sensitivity, with enhanced chemosurvival when cells engage αvß3 vs α5ß1 on fibronectin; however, this is heavily reliant on nanoscale spacing, as the opposite is observed when ligands are spaced at 70 nm. These data imply that even nanoscale alterations in extracellular matrix properties have profound effects on cancer cell survival and can thus inform future therapies and drug testing platforms.
Assuntos
Neoplasias da Mama/tratamento farmacológico , Adesão Celular/genética , Integrina alfa5beta1/genética , Integrina alfaVbeta3/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Fibronectinas/química , Fibronectinas/genética , Ouro/química , Humanos , Integrina alfa5beta1/química , Integrina alfaVbeta3/química , Ligantes , Nanopartículas Metálicas/química , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacosRESUMO
Dynamic single-molecule force spectroscopy was performed to monitor the unbinding of fibronectin with the proteoglycans syndecan-4 (SDC4) and decorin and to compare this with the unbinding characteristics of α5ß1-integrin. A single energy barrier was sufficient to describe the unbinding of both SDC4 and decorin from fibronectin, whereas two barriers were observed for the dissociation of α5ß1-integrin from fibronectin. The outer (high-affinity) barriers in the interactions of fibronectin with α5ß1-integrin and SDC4 are characterized by larger barrier heights and widths and slower dissociation rates than those of the inner (low-affinity) barriers in the interactions of fibronectin with α5ß1-integrin and decorin. These results indicate that SDC4 and (ultimately) α5ß1-integrin have the ability to withstand deformation in their interactions with fibronectin, whereas the decorin-fibronectin interaction is considerably more brittle.
Assuntos
Decorina/metabolismo , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Sindecana-4/metabolismo , Sítios de Ligação , Decorina/química , Fibronectinas/química , Humanos , Integrina alfa5beta1/química , Ligação Proteica , Sindecana-4/química , TermodinâmicaRESUMO
Whether ß1 integrin ectodomains visit conformational states similarly to ß2 and ß3 integrins has not been characterized. Furthermore, despite a wealth of activating and inhibitory antibodies to ß1 integrins, the conformational states that these antibodies stabilize, and the relation of these conformations to function, remain incompletely characterized. Using negative-stain electron microscopy, we show that the integrin α5ß1 ectodomain adopts extended-closed and extended-open conformations as well as a bent conformation. Antibodies SNAKA51, 8E3, N29, and 9EG7 bind to different domains in the α5 or ß1 legs, activate, and stabilize extended ectodomain conformations. Antibodies 12G10 and HUTS-4 bind to the ß1 ßI domain and hybrid domains, respectively, activate, and stabilize the open headpiece conformation. Antibody TS2/16 binds a similar epitope as 12G10, activates, and appears to stabilize an open ßI domain conformation without requiring extension or hybrid domain swing-out. mAb13 and SG/19 bind to the ßI domain and ßI-hybrid domain interface, respectively, inhibit, and stabilize the closed conformation of the headpiece. The effects of the antibodies on cell adhesion to fibronectin substrates suggest that the extended-open conformation of α5ß1 is adhesive and that the extended-closed and bent-closed conformations are nonadhesive. The functional effects and binding sites of antibodies and fibronectin were consistent with their ability in binding to α5ß1 on cell surfaces to cross-enhance or inhibit one another by competitive or noncompetitive (allosteric) mechanisms.
Assuntos
Integrina alfa5beta1/química , Escherichia coli , Fibronectinas/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Conformação Proteica , Relação Estrutura-AtividadeRESUMO
We previously demonstrated that Arg-Gly-Asp (RGD)-containing ligand-mimetic inhibitors of integrins are unable to dissociate pre-formed integrin-fibronectin complexes (IFCs). These observations suggested that amino acid residues involved in integrin-fibronectin binding become obscured in the ligand-occupied state. Because the epitopes of some function-blocking anti-integrin monoclonal antibodies (mAbs) lie near the ligand-binding pocket, it follows that the epitopes of these mAbs may become shielded in the ligand-occupied state. Here, we tested whether function-blocking mAbs directed against α5ß1 can interact with the integrin after it forms a complex with an RGD-containing fragment of fibronectin. We showed that the anti-α5 subunit mAbs JBS5, SNAKA52, 16, and P1D6 failed to disrupt IFCs and hence appeared unable to bind to the ligand-occupied state. In contrast, the allosteric anti-ß1 subunit mAbs 13, 4B4, and AIIB2 could dissociate IFCs and therefore were able to interact with the ligand-bound state. However, another class of function-blocking anti-ß1 mAbs, exemplified by Lia1/2, could not disrupt IFCs. This second class of mAbs was also distinguished from 13, 4B4, and AIIB2 by their ability to induce homotypic cell aggregation. Although the epitope of Lia1/2 was closely overlapping with those of 13, 4B4, and AIIB2, it appeared to lie closer to the ligand-binding pocket. A new model of the α5ß1-fibronectin complex supports our hypothesis that the epitopes of mAbs that fail to bind to the ligand-occupied state lie within, or very close to, the integrin-fibronectin interface. Importantly, our findings imply that the efficacy of some therapeutic anti-integrin mAbs could be limited by epitope masking.
Assuntos
Anticorpos Monoclonais Murinos/química , Epitopos/química , Fibronectinas/química , Integrina alfa5beta1/química , Modelos Moleculares , Oligopeptídeos/química , Regulação Alostérica/imunologia , Anticorpos Monoclonais Murinos/imunologia , Epitopos/genética , Epitopos/imunologia , Fibronectinas/genética , Fibronectinas/imunologia , Humanos , Integrina alfa5beta1/genética , Integrina alfa5beta1/imunologia , Células Jurkat , Oligopeptídeos/genética , Oligopeptídeos/imunologiaRESUMO
Integrin α5ß1 binds to an Arg-Gly-Asp (RGD) motif in its ligand fibronectin. We report high-resolution crystal structures of a four-domain α5ß1 headpiece fragment, alone or with RGD peptides soaked into crystals, and RGD peptide affinity measurements. The headpiece crystallizes in a closed conformation essentially identical to that seen previously for α5ß1 complexed with a Fab that allosterically inhibits ligand binding by stabilizing the closed conformation. Soaking experiments show that binding of cyclic RGD peptide with 20-fold higher affinity than a linear RGD peptide induces conformational change in the ß1-subunit ßI domain to a state that is intermediate between closed (low affinity) and open (high affinity). In contrast, binding of a linear RGD peptide induces no shape shifting. However, linear peptide binding induces shape shifting when Ca(2+) is depleted during soaking. Ca(2+) bound to the adjacent to metal ion-dependent adhesion site (ADMIDAS), at the locus of shape shifting, moves and decreases in occupancy, correlating with an increase in affinity for RGD measured when Ca(2+) is depleted. The results directly demonstrate that Ca(2+) binding to the ADMIDAS stabilizes integrins in the low-affinity, closed conformation. Comparisons in affinity between four-domain and six-domain headpiece constructs suggest that flexible integrin leg domains contribute to conformational equilibria. High-resolution views of the hybrid domain interface with the plexin-semaphorin-integrin (PSI) domain in different orientations show a ball-and-socket joint with a hybrid domain Arg side chain that rocks in a PSI domain socket lined with carbonyl oxygens.
Assuntos
Fibronectinas/metabolismo , Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Modelos Moleculares , Cálcio/metabolismo , Cristalização , Polarização de Fluorescência , Ligantes , Peptídeos Cíclicos/metabolismo , Conformação ProteicaRESUMO
The linker between the targeting moiety and the nanoparticle is often overlooked when engineering targeted drug delivery vehicles. We hypothesized that pH-triggered conformational changes of an elastin-like peptide (ELP) linker, with repeating VPGVG sequences, could alter the binding affinity of the well-established targeting moiety arginine-glycine-aspartic acid (RGD), which is known to enhance the delivery of nanoparticles to tumor cells via integrin overexpression. The pH change from blood (pH 7.4) to the tumor environment (pH 6) was used to elicit a conformational change in the ELP linker, as described by circular dichroism. Atomic force microscopy confirmed that RGD-ELP resulted in stronger adhesion to both MDA-MB-231 and HCC1806 breast cancer cells at pH 6 relative to pH 7.4. No change in adhesion force was measured as a function of pH for the non-neoplastic MCF-10A cell line and the nontargeting GDR-ELP peptide. This translated to significant binding and uptake of RGD-ELP modified liposomes at pH 6.0 relative to pH 7.4. These results indicate that the pH-triggered conformational structure of the ELP linker shifts RGD-mediated cancer cell targeting from non-active (pH 7.4) to active (pH 6). The reversible shift in ELP secondary structure may be used to engineer targeted drug delivery vehicles with tunable uptake.
Assuntos
Elastina/química , Oligopeptídeos/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Sequência de Aminoácidos , Adesão Celular , Linhagem Celular Tumoral , Humanos , Concentração de Íons de Hidrogênio , Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/química , Integrina alfaVbeta3/metabolismo , Lipossomos , Modelos Moleculares , Nanopartículas/química , Fragmentos de Peptídeos/toxicidade , Estrutura Secundária de Proteína , Transporte ProteicoRESUMO
Fibronectin (FN) is a major component of the extracellular matrix and functions in cell adhesion, cell spreading and cell migration. In the retina, FN is transiently expressed and assembled on astrocytes (ACs), which guide sprouting tip cells and deposit a provisional matrix for sprouting angiogenesis. The precise function of FN in retinal angiogenesis is largely unknown. Using genetic tools, we show that astrocytes are the major source of cellular FN during angiogenesis in the mouse retina. Deletion of astrocytic FN reduces radial endothelial migration during vascular plexus formation in a gene dose-dependent manner. This effect correlates with reduced VEGF receptor 2 and PI3K/AKT signalling, and can be mimicked by selectively inhibiting VEGF-A binding to FN through intraocular injection of blocking peptides. By contrast, AC-specific replacement of the integrin-binding RGD sequence with FN-RGE or endothelial deletion of itga5 shows little effect on migration and PI3K/AKT signalling, but impairs filopodial alignment along AC processes, suggesting that FN-integrin α5ß1 interaction is involved in filopodial adhesion to the astrocytic matrix. AC FN shares its VEGF-binding function and cell-surface distribution with heparan-sulfate (HS), and genetic deletion of both FN and HS together greatly enhances the migration defect, indicating a synergistic function of FN and HS in VEGF binding. We propose that in vivo the VEGF-binding properties of FN and HS promote directional tip cell migration, whereas FN integrin-binding functions to support filopodia adhesion to the astrocytic migration template.
Assuntos
Astrócitos/metabolismo , Fibronectinas/metabolismo , Integrinas/metabolismo , Neovascularização Fisiológica , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/metabolismo , Animais , Movimento Celular , Matriz Extracelular/metabolismo , Fibronectinas/deficiência , Fibronectinas/genética , Heparitina Sulfato/metabolismo , Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oligopeptídeos/química , Fosfatidilinositol 3-Quinases/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vasos Retinianos/inervação , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismoRESUMO
UNLABELLED: Ursodeoxycholic acid, which in vivo is converted to its taurine conjugate tauroursodeoxycholic acid (TUDC), is a mainstay for the treatment of cholestatic liver disease. Earlier work showed that TUDC exerts its choleretic properties in the perfused rat liver in an α5 ß1 integrin-mediated way. However, the molecular basis of TUDC-sensing in the liver is unknown. We herein show that TUDC (20 µmol/L) induces in perfused rat liver and human HepG2 cells the rapid appearance of the active conformation of the ß1 subunit of α5 ß1 integrins, followed by an activating phosphorylation of extracellular signal-regulated kinases. TUDC-induced kinase activation was no longer observed after ß1 integrin knockdown in isolated rat hepatocytes or in the presence of an integrin-antagonistic hexapeptide in perfused rat liver. TUDC-induced ß1 integrin activation occurred predominantly inside the hepatocyte and required TUDC uptake by way of the Na(+) /taurocholate cotransporting peptide. Molecular dynamics simulations of a 3D model of α5 ß1 integrin with TUDC bound revealed significant conformational changes within the head region that have been linked to integrin activation before. CONCLUSIONS: TUDC can directly activate intrahepatocytic ß1 integrins, which trigger signal transduction pathways toward choleresis. (HEPATOLOGY 2013).
Assuntos
Hepatócitos/metabolismo , Integrina alfa5beta1/metabolismo , Ácido Tauroquenodesoxicólico/metabolismo , Regulação Alostérica/fisiologia , Animais , Dimerização , Proteínas de Fluorescência Verde/genética , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Humanos , Integrina alfa5beta1/química , Integrina alfa5beta1/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Estrutura Secundária de Proteína/fisiologia , Estrutura Terciária de Proteína/fisiologia , RNA Interferente Pequeno/genética , Ratos , Ratos Wistar , Relação Estrutura-Atividade , Simportadores/genética , Simportadores/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , Ácido Taurocólico/metabolismo , Ácido Taurocólico/farmacologia , Ácido Ursodesoxicólico/farmacocinéticaRESUMO
Receptor targeted, PEGylated transfection agents can improve stability and delivery specificity of current cationic lipid and polymer based nonviral gene delivery vehicles, but their mode of transfection is poorly understood. We therefore investigated the transfection mechanisms of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) lipoplexes, branched polyethylenimine (bPEI) polyplexes, and bPEI encapsulated in either PEGylated (stealth) nontargeted liposomes or PR_b peptide (targeted to α5ß1 integrin) functionalized stealth liposomes in DLD-1 colorectal cancer cells in vitro with gene expression assays, flow cytometry and confocal microscopy. DOTAP/DOPE and PR_b functionalized stealth liposomes mediated higher gene expression compared to nontargeted stealth liposomes and bPEI. However DOTAP/DOPE was internalized slowly leading to lower levels of DNA uptake. In contrast, despite high internalization of bPEI polyplexes, gene expression levels were low as DNA was unable to escape from the endosomes. Nontargeted stealth liposomes also mediated low gene expression due to low amounts of DNA internalized and slow internalization kinetics. PR_b functionalized stealth liposomes struck an optimal balance among these transfection agents with efficient transfection arising from fast integrin mediated internalization kinetics, high amounts of DNA uptake, and endosomal escape. We found α5ß1 integrin to be a valuable target for gene delivery and that the caveolar endocytic pathway may offer an advantage to receptor targeted PEGylated transfection agents in DLD-1 cells.
Assuntos
Células Epiteliais/metabolismo , Técnicas de Transferência de Genes , Integrina alfa5beta1/metabolismo , Lipossomos/metabolismo , Transporte Biológico , Cavéolas/metabolismo , Linhagem Celular Tumoral , Colo/metabolismo , Colo/patologia , Endocitose , Endossomos/metabolismo , Células Epiteliais/patologia , Ácidos Graxos Monoinsaturados/química , Citometria de Fluxo , Expressão Gênica , Genes Reporter , Humanos , Integrina alfa5beta1/química , Lipossomos/síntese química , Luciferases/genética , Luciferases/metabolismo , Fosfatidiletanolaminas/química , Polietilenoimina/química , Compostos de Amônio Quaternário/químicaRESUMO
Fibronectin (FN) fibrillogenesis depends on the binding of FN to cellular receptors and subsequent unfolding of bound FN. Integrins αIIbß3, αvß3, and α5ß1 are known to assemble FN fibrils on platelets. In our study, we examined the contribution of these integrins to FN binding, unfolding, and assembly on platelets in suspension and adherent platelets in the presence or absence of agonists. Phorbol 12-myristate 13-acetate (PMA), but not adenosine diphosphate (ADP), induced binding of FN to platelets in suspension. In contrast, adherent platelets were able to deposit FN on their surfaces in the absence of agonists. ß3 integrins had a major impact on the interaction of FN on platelets. αvß3 showed a similar contribution to the binding of FN as αIIbß3 on PMA-stimulated platelets in suspension but had a lesser contribution to unfolding and deposition of FN on adherent platelets. α5ß1 also participated in the interaction of FN with platelets by mediating the unfolding and assembly of FN, but to a lesser extent than ß3 integrins. None of the distinct antibodies directed against one of the three integrins caused a complete inhibition of binding, unfolding, and assembly of FN by platelets. Thus, it is likely that αIIbß3, αvß3, and α5ß1 or another still unknown receptor can be substituted.
Assuntos
Plaquetas/química , Fibronectinas/química , Integrina alfa5beta1/química , Integrina alfaVbeta3/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Plaquetas/metabolismo , Adesão Celular , Fibronectinas/sangue , Transferência Ressonante de Energia de Fluorescência , Humanos , Integrina alfa5beta1/agonistas , Integrina alfa5beta1/sangue , Integrina alfaVbeta3/agonistas , Integrina alfaVbeta3/sangue , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/agonistas , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ligação Proteica , Conformação Proteica , Desnaturação Proteica , Mapeamento de Interação de Proteínas , Desdobramento de ProteínaRESUMO
We present a click chemistry-based molecular toolkit for the biofunctionalization of materials to selectively control integrin-mediated cell adhesion. To this end, α5ß1-selective RGD peptidomimetics were covalently immobilized on Ti-based materials, and the capacity to promote the selective binding of α5ß1 was evaluated using a solid-phase integrin binding assay. This functionalization strategy yielded surfaces with a nine-fold increased affinity for α5ß1, in comparison to control samples, and total selectivity against the binding of the closely related integrin αvß3. Moreover, our methodology allowed the screening of several phosphonic acid containing anchoring units to find the best spacer-anchor moiety required for establishing an efficient binding to titanium and to promote selective integrin binding. The integrin subtype specificity of these biofunctionalized surfaces was further examined in vitro by inducing selective adhesion of genetically modified fibroblasts, which express exclusively the α5ß1 integrin. The versatility of our molecular toolkit was proven by shifting the cellular specificity of the materials from α5ß1- to αvß3-expressing fibroblasts by using an αvß3-selective peptidomimetic as coating molecule. The results shown here represent the first functionalization of Ti-based materials with α5ß1- or αvß3-selective peptidomimetics that allow an unprecedented control to discriminate between α5ß1- and αvß3-mediated adhesions. The role of these two integrins in different biological events is still a matter of debate and is frequently discussed in literature. Thus, such bioactive titanium surfaces will be of great relevance for the study of integrin-mediated cell adhesion and the development of new biomaterials targeting specific cell types.
Assuntos
Integrina alfa5beta1/química , Integrina alfaVbeta3/química , Oligopeptídeos/química , Peptidomiméticos/química , Titânio , Animais , Materiais Biocompatíveis , Adesão Celular , Química Click , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Peptidomiméticos/farmacologia , Ligação ProteicaRESUMO
The α5ß1 integrin heterodimer is involved in many cellular processes and is an anti-cancer therapeutic target. Therefore, access to quantities of protein suitable for studies aimed at understanding its biological functions is important. To this end, a large-scale protein expression system, utilizing the recombinant baculovirus/SF9 insect cell expression system, was created to produce the extracellular domain of the α5ß1 integrin. An incorporated 8X-histidine tag enabled one-step nickel-column purification. Following sequence confirmation by LC-MS/MS, the conformation of the heterodimer was characterized by native dot blot and negative stain electron microscopy. Cellular transduction inhibition studies confirmed biological activity. The system allows expression and purification of α5ß1 integrin in quantities suitable for an array of different experiments including structural biology.
Assuntos
Clonagem Molecular/métodos , Integrina alfa5beta1/genética , Integrina alfa5beta1/isolamento & purificação , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Linhagem Celular , Cromatografia Líquida , Humanos , Insetos , Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Espectrometria de Massas em Tandem , Regulação para CimaRESUMO
Distinct lipid environments, including lipid rafts, are increasingly recognized as a crucial factor affecting membrane protein function in plasma membranes. Unfortunately, an understanding of their role in membrane protein activation and oligomerization has remained elusive due to the challenge of characterizing these often small and transient plasma membrane heterogeneities in live cells. To address this difficulty, we present an experimental model membrane platform based on polymer-supported lipid bilayers containing stable raft-mimicking domains (type I) and homogeneous cholesterol-lipid mixtures (type II) into which transmembrane proteins are incorporated (α(v)ß(3) and α(5)ß(1) integrins). These flexible lipid platforms enable the use of confocal fluorescence spectroscopy, including the photon counting histogram method, in tandem with epifluorescence microscopy to quantitatively probe the effect of the binding of native ligands from the extracellular matrix ligands (vitronectin and fibronectin for α(v)ß(3) and α(5)ß(1), respectively) on domain-specific protein sequestration and on protein oligomerization state. We found that both α(v)ß(3) and α(5)ß(1) sequester preferentially to nonraft domains in the absence of extracellular matrix ligands, but upon ligand addition, α(v)ß(3) sequesters strongly into raft-like domains and α(5)ß(1) loses preference for either raft-like or nonraft-like domains. A corresponding photon counting histogram analysis showed that integrins exist predominantly in a monomeric state. No change was detected in oligomerization state upon ligand binding in either type I or type II bilayers, but a moderate increase in oligomerization state was observed for increasing concentrations of cholesterol. The combined findings suggest a mechanism in which changes in integrin sequestering are caused by ligand-induced changes in integrin conformation and/or dynamics that affect integrin-lipid interactions without altering the integrin oligomerization state.
Assuntos
Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/química , Integrina alfaVbeta3/metabolismo , Bicamadas Lipídicas/metabolismo , Microdomínios da Membrana/metabolismo , Multimerização Proteica , Soluções Tampão , Colesterol/metabolismo , Humanos , Ligantes , Bicamadas Lipídicas/química , Fluidez de Membrana , Microdomínios da Membrana/química , Estrutura Quaternária de Proteína , Estrutura Terciária de ProteínaRESUMO
Engagement of α(5)ß(1)-integrin by fibronectin (FN) acutely enhances Cav1.2 channel (Ca(L)) current in rat arteriolar smooth muscle and human embryonic kidney cells (HEK293-T) expressing Ca(L). Using coimmunoprecipitation strategies, we show that coassociation of Ca(L) with α(5)- or ß(1)-integrin in HEK293-T cells is specific and depends on cell adhesion to FN. In rat arteriolar smooth muscle, coassociations between Ca(L) and α(5)ß(1)-integrin and between Ca(L) and phosphorylated c-Src are also revealed and enhanced by FN treatment. Using site-directed mutagenesis of Ca(L) heterologously expressed in HEK293-T cells, we identified two regions of Ca(L) required for these interactions: 1) COOH-terminal residues Ser(1901) and Tyr(2122), known to be phosphorylated by protein kinase A (PKA) and c-Src, respectively; and 2) two proline-rich domains (PRDs) near the middle of the COOH terminus. Immunofluorescence confocal imaging revealed a moderate degree of wild-type Ca(L) colocalization with ß(1)-integrin on the plasma membrane. Collectively, our results strongly suggest that 1) upon ligation by FN, Ca(L) associates with α(5)ß(1)-integrin in a macromolecular complex including PKA, c-Src, and potentially other protein kinases; 2) phosphorylation of Ca(L) at Y(2122) and/or S(1901) is required for association of Ca(L) with α(5)ß(1)-integrin; and 3) c-Src, via binding to PRDs that reside in the II-III linker region and/or the COOH terminus of Ca(L), mediates current potentiation following α(5)ß(1)-integrin engagement. These findings provide new evidence for how interactions between α(5)ß(1)-integrin and FN can modulate Ca(L) entry and consequently alter the physiological function of multiple types of excitable cells.
Assuntos
Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/genética , Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Integrina alfa5beta1/fisiologia , Músculo Liso Vascular/metabolismo , Animais , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/genética , Sinalização do Cálcio/efeitos dos fármacos , Adesão Celular/genética , Adesão Celular/fisiologia , Membrana Celular/química , Membrana Celular/ultraestrutura , Células Cultivadas , Células Epiteliais/química , Fibronectinas/genética , Fibronectinas/metabolismo , Fibronectinas/fisiologia , Células HEK293 , Humanos , Integrina alfa5beta1/química , Integrina alfa5beta1/genética , Microscopia Confocal , Músculo Liso Vascular/química , Músculo Liso Vascular/citologia , Mutagênese Sítio-Dirigida , Fosforilação/genética , Fosforilação/fisiologia , RatosRESUMO
While vital to platelet and leukocyte adhesion, the role of integrin affinity modulation in adherent cells remains controversial. In endothelial cells, atheroprone hemodynamics and oxidized lipoproteins drive an increase in the high affinity conformation of α5ß1 integrins in endothelial cells in vitro, and α5ß1 integrin inhibitors reduce proinflammatory endothelial activation to these stimuli in vitro and in vivo. However, the importance of α5ß1 integrin affinity modulation to endothelial phenotype remains unknown. We now show that endothelial cells (talin1 L325R) unable to induce high affinity integrins initially adhere and spread but show significant defects in nascent adhesion formation. In contrast, overall focal adhesion number, area, and composition in stably adherent cells are similar between talin1 wildtype and talin1 L325R endothelial cells. However, talin1 L325R endothelial cells fail to induce high affinity α5ß1 integrins, fibronectin deposition, and proinflammatory responses to atheroprone hemodynamics and oxidized lipoproteins. Inducing the high affinity conformation of α5ß1 integrins in talin1 L325R endothelial cells suggest that NF-κB activation and maximal fibronectin deposition require both integrin activation and other integrin-independent signaling. In endothelial-specific talin1 L325R mice, atheroprone hemodynamics fail to promote inflammation and macrophage recruitment, demonstrating a vital role for integrin activation in regulating endothelial phenotype.
Assuntos
Aterosclerose/metabolismo , Células Endoteliais/citologia , Integrina alfa5beta1/metabolismo , Talina/genética , Animais , Aterosclerose/genética , Adesão Celular , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Fibronectinas/metabolismo , Adesões Focais/metabolismo , Humanos , Integrina alfa5beta1/química , Camundongos , Mutação , NF-kappa B/metabolismo , Conformação Proteica , Transdução de SinaisRESUMO
Maspin is a serpin that has multiple effects on cell behavior, including inhibition of migration. How maspin mediates these diverse effects remains unclear, as it is devoid of protease inhibitory activity. We have previously shown that maspin rapidly inhibits the migration of vascular smooth muscle cells (VSMC), suggesting the involvement of direct interactions with cell surface proteins. Here, using immunofluorescence microscopy, we demonstrate that maspin binds specifically to the surface of VSMC in the dedifferentiated, but not the differentiated, phenotype. Ligand blotting of VSMC lysates revealed the presence of several maspin-binding proteins, with a protein of 150 kDa differentially expressed between the two VSMC phenotypes. Western blotting suggested that this protein was the beta1 integrin subunit, and subsequently both alpha3beta1 and alpha5beta1, but not alphavbeta3, were shown to associate with maspin by coimmunoprecipitation. Specific binding of these integrins was also observed using maspin-affinity chromatography, using HT1080 cell lysates. Direct binding of maspin to alpha5beta1 was confirmed using a recombinant alpha5beta1-Fc fusion protein. Using conformation-dependent anti-beta1 antibodies, maspin binding to VSMC was found to lead to a decrease in the activation status of the integrin. The functional involvement of alpha5beta1 in mediating the effect of maspin was established by the inhibition of migration of CHO cells overexpressing human alpha5 integrin, but not those lacking alpha5 expression. Our observations suggest that maspin engages in specific interactions with a limited number of integrins on VSMC, leading to their inactivation, and that these interactions are responsible for the effects of maspin in the pericellular environment.