Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 253
Filter
1.
J Phys Chem B ; 128(16): 3929-3936, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38619541

ABSTRACT

Yersinia pestis, the causative agent of plague, is capable of evading the human immune system response by recruiting the plasma circulating vitronectin proteins, which act as a shield and avoid its lysis. Vitronectin recruitment is mediated by its interaction with the bacterial transmembrane protein Ail, protruding from the Y. pestis outer membrane. By using all-atom long-scale molecular dynamic simulations of Ail embedded in a realistic model of the bacterial membrane, we have shown that vitronectin forms a stable complex, mediated by interactions between the disordered moieties of the two proteins. The main amino acids driving the complexation have also been evidenced, thus favoring the possible rational design of specific peptides which, by inhibiting vitronectin recruitment, could act as original antibacterial agents.


Subject(s)
Bacterial Outer Membrane Proteins , Molecular Dynamics Simulation , Vitronectin , Vitronectin/chemistry , Vitronectin/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Humans , Yersinia pestis/chemistry , Yersinia pestis/metabolism , Virulence Factors/chemistry , Virulence Factors/metabolism , Protein Domains , Protein Binding
2.
ACS Appl Mater Interfaces ; 15(23): 27560-27567, 2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37276244

ABSTRACT

Peptide sequence periodicity is a simple design tool that can be used to generate functional peptide-based surface coatings. De novo-designed peptide N3-PEG-VK16 is characterized by a hydrophobic periodicity of two that avidly binds to native polystyrene priming its surface for subsequent targeted functionalization via chemical ligation. The peptidic portion of N3-PEG-VK16 is responsible for surface binding, converting polystyrene's hydrophobic surface into a wettable and electrostatically charged environment that facilitates cell attachment. Native polystyrene surfaces are coated by simple peptide adsorption from an aqueous buffered solution, and the resulting primed surface is easily functionalized by cycloaddition chemistry. Herein, we show that ligating a vitronectin-derived peptide to primed polystyrene surfaces enables adhesion, expansion, long-term culture, and phenotype maintenance of human induced pluripotent stem cells. To demonstrate scope, we also show that additional functional ligands can be used, for example, nerve growth factor protein, to control neurite outgrowth.


Subject(s)
Induced Pluripotent Stem Cells , Polystyrenes , Humans , Polystyrenes/chemistry , Cell Adhesion , Peptides/pharmacology , Vitronectin/chemistry , Surface Properties
3.
J Biol Chem ; 298(12): 102652, 2022 12.
Article in English | MEDLINE | ID: mdl-36444882

ABSTRACT

The serpin plasminogen activator inhibitor 1 (PAI-1) spontaneously undergoes a massive structural change from a metastable and active conformation, with a solvent-accessible reactive center loop (RCL), to a stable, inactive, or latent conformation, with the RCL inserted into the central ß-sheet. Physiologically, conversion to the latent state is regulated by the binding of vitronectin, which hinders the latency transition rate approximately twofold. The molecular mechanisms leading to this rate change are unclear. Here, we investigated the effects of vitronectin on the PAI-1 latency transition using all-atom path sampling simulations in explicit solvent. In simulated latency transitions of free PAI-1, the RCL is quite mobile as is the gate, the region that impedes RCL access to the central ß-sheet. This mobility allows the formation of a transient salt bridge that facilitates the transition; this finding rationalizes existing mutagenesis results. Vitronectin binding reduces RCL and gate mobility by allosterically rigidifying structural elements over 40 Å away from the binding site, thus blocking transition to the latent conformation. The effects of vitronectin are propagated by a network of dynamically correlated residues including a number of conserved sites that were previously identified as important for PAI-1 stability. Simulations also revealed a transient pocket populated only in the vitronectin-bound state, corresponding to a cryptic drug-binding site identified by crystallography. Overall, these results shed new light on PAI-1 latency transition regulation by vitronectin and illustrate the potential of path sampling simulations for understanding functional protein conformational changes and for facilitating drug discovery.


Subject(s)
Plasminogen Activator Inhibitor 1 , Vitronectin , Plasminogen Activator Inhibitor 1/metabolism , Vitronectin/chemistry , Models, Molecular , Protein Conformation , Solvents
4.
Biophys J ; 121(20): 3896-3906, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36056555

ABSTRACT

The adaptability of proteins to their work environments is fundamental for cellular life. Here, we describe how the hemopexin-like domain of the multifunctional blood glycoprotein vitronectin binds Ca2+ to adapt to excursions of temperature and shear stress. Using X-ray crystallography, molecular dynamics simulations, NMR, and differential scanning fluorimetry, we describe how Ca2+ and its flexible hydration shell enable the protein to perform conformational changes that relay beyond the calcium-binding site and alter the number of polar contacts to enhance conformational stability. By means of mutagenesis, we identify key residues that cooperate with Ca2+ to promote protein stability, and we show that calcium association confers protection against shear stress, a property that is advantageous for proteins that circulate in the vasculature, like vitronectin.


Subject(s)
Calcium , Vitronectin , Calcium/metabolism , Vitronectin/chemistry , Vitronectin/metabolism , Protein Binding , Hemopexin/metabolism , Binding Sites , Crystallography, X-Ray , Protein Conformation
5.
PLoS One ; 17(1): e0262183, 2022.
Article in English | MEDLINE | ID: mdl-34986165

ABSTRACT

PURPOSE: Spondyloarthritis (SpA) is a systemic inflammatory arthritis mediated mainly by interleukin (IL)-17. The vitronectin-derived bioactive peptide, VnP-16, exerts an anti-osteoporotic effect via ß1 and αvß3 integrin signaling. SpA is associated with an increased risk of osteoporosis, and we investigated the effect of VnP-16 in mice with SpA. METHODS: SpA was induced by curdlan in SKG ZAP-70W163C mice, which were treated with vehicle, celecoxib, VnP-16, or VnP-16+celecoxib. The clinical score, arthritis score, spondylitis score, and proinflammatory cytokine expression of the spine were evaluated by immunohistochemical staining. Type 17 helper T cell (Th17) and regulatory T cell (Treg) differentiation in the spleen was evaluated by flow cytometry and in the spine by confocal staining. Splenocyte expression of signal transducer and activator of transcription (STAT) 3 and pSTAT3 was evaluated by in vitro Western blotting. RESULTS: The clinical score was significantly reduced in the VnP16+celecoxib group. The arthritis and spondylitis scores were significantly lower in the VnP-16 and VnP16+celecoxib groups than the vehicle group. In the spine, the levels of IL-1ß, IL-6, tumor necrosis factor-α, and IL-17 expression were reduced and Th17/Treg imbalance was regulated in the VnP-16 alone and VnP-16+celecoxib groups. Flow cytometry of splenocytes showed increased polarization of Tregs in the VnP-16+celecoxib group. In vitro, VnP-16 suppressed pSTAT3. CONCLUSIONS: VnP-16 plus celecoxib prevented SpA progression in a mouse model by regulating the Th17/Treg imbalance and suppressing the expression of proinflammatory cytokines.


Subject(s)
Celecoxib/administration & dosage , Peptides/administration & dosage , Spondylarthritis/drug therapy , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Vitronectin/chemistry , beta-Glucans/adverse effects , Animals , Celecoxib/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Gene Expression Regulation/drug effects , Humans , Integrin alphaVbeta3/metabolism , Integrin beta1/metabolism , Mice , Peptides/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Spleen/immunology , Spondylarthritis/chemically induced , Spondylarthritis/genetics , Spondylarthritis/immunology
6.
Acta Biochim Pol ; 68(4): 515-525, 2021 Nov 14.
Article in English | MEDLINE | ID: mdl-34773933

ABSTRACT

Although many atypical proteinaceous cell wall components that belong to a group of multitasking, "moonlighting" proteins, have been repeatedly identified in numerous pathogenic microorganisms, their novel extracellular functions and secretion mechanisms remain largely unrecognized. In Candida albicans, one of the most common fungal pathogens in humans, phosphoglycerate mutase (Gpm1) - a cytoplasmic enzyme involved in the glycolysis pathway - has been shown to occur on the cell surface and has been identified as a potentially important virulence factor. In this study, we demonstrated tight binding of C. albicans Gpm1 to the candidal cell surface, thus suggesting that the readsorption of soluble Gpm1 from the external environment could be a likely mechanism leading to the presence of this moonlighting protein on the pathogen surface. Several putative Gpm1-binding receptors on the yeast surface were identified. The affinities of Gpm1 to human vitronectin (VTR) and fibronectin (FN) were characterized with surface plasmon resonance measurements, and the dissociation constants of the complexes formed were determined to be in the order of 10-8 M. The internal Gpm1 sequence motifs, directly interacting with VTR (aa 116-158) and FN (aa 138-175) were mapped using chemical crosslinking and mass spectrometry. Synthetic peptides with matching sequences significantly inhibited formation of the Gpm1-VTR and Gpm1-FN complexes. A molecular model of the Gpm1-VTR complex was developed. These results provide the first structural insights into the adhesin function of candidal surface-exposed Gpm1.


Subject(s)
Candida albicans/metabolism , Extracellular Matrix Proteins/metabolism , Fungal Proteins/metabolism , Phosphoglycerate Mutase/metabolism , Candida albicans/chemistry , Cell Membrane/metabolism , Cell Wall/metabolism , Extracellular Matrix Proteins/chemistry , Fibronectins/chemistry , Fibronectins/metabolism , Fungal Proteins/chemistry , Humans , Models, Molecular , Phosphoglycerate Mutase/chemistry , Protein Binding , Surface Plasmon Resonance/methods , Virulence Factors/metabolism , Vitronectin/chemistry , Vitronectin/metabolism
7.
Sci Rep ; 11(1): 12023, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103584

ABSTRACT

Vitronectin (VN) is a glycoprotein found in extracellular matrix and blood. Collagen, a major extracellular matrix component in mammals, is degraded by cathepsin K (CatK), which is essential for bone resorption under acidic conditions. The relationship between VN and cathepsins has been unclear. We discovered that VN promoted collagen fibril formation and inhibited CatK activity, and observed its activation in vitro. VN accelerated collagen fibril formation at neutral pH. Collagen fibers formed with VN were in close contact with each other and appeared as scattered flat masses in scanning electron microscopy images. VN formed collagen fibers with high acid solubility and significantly inhibited CatK; the IC50 was 8.1-16.6 nM and competitive, almost the same as those of human and porcine VNs. VN inhibited the autoprocessing of inactive pro-CatK from active CatK. DeN-glycosylation of VN attenuated the inhibitory effects of CatK and its autoprocessing by VN, but had little effect on acid solubilization of collagen and VN degradation via CatK. CatK inhibition is an attractive treatment approach for osteoporosis and osteoarthritis. These findings suggest that glycosylated VN is a potential biological candidate for CatK inhibition and may help to understand the molecular mechanisms of tissue re-modeling.


Subject(s)
Cathepsin K/chemistry , Collagen/chemistry , Vitronectin/chemistry , Animals , Glycosylation , Humans , Swine
8.
ACS Appl Mater Interfaces ; 13(7): 8006-8014, 2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33590757

ABSTRACT

Hydrogels formed from peptide self-assembly are a class of materials that are being explored for their utility in tissue engineering, drug and cell delivery, two- and three-dimensional cell culture, and as adjuvants in surgical procedures. Most self-assembled peptide gels can be syringe-injected in vivo to facilitate the local delivery of payloads, including cells, directly to the targeted tissue. Herein, we report that highly positively charged peptide gels are inherently toxic to cells, which would seem to limit their utility. However, adding media containing fetal bovine serum, a common culture supplement, directly transforms these toxic gels into cytocompatible materials capable of sustaining cell viability even in the absence of added nutrients. Multistage mass spectrometry showed that at least 40 serum proteins can absorb to a gel's surface through electrostatic attraction ameliorating its toxicity. Further, cell-based studies employing model gels having only bovine serum albumin, fetuin-A, or vitronectin absorbed to the gel surface showed that single protein additives can also be effective depending on the identity of the cell line. Separate studies employing these model gels showed that the mechanism(s) responsible for mitigating apoptosis involve both the pacification of gel surface charge and adsorbed protein-mediated cell signaling events that activate both the PI3/Akt and MAPK/ERK pathways which are known to facilitate resistance to stress-induced apoptosis and overall cell survival.


Subject(s)
Hydrogels/pharmacology , Peptides/pharmacology , Serum Albumin, Bovine/antagonists & inhibitors , Vitronectin/antagonists & inhibitors , alpha-2-HS-Glycoprotein/antagonists & inhibitors , Adsorption , Cell Survival/drug effects , Cells, Cultured , Gels/chemical synthesis , Gels/chemistry , Gels/pharmacology , Humans , Hydrogels/chemical synthesis , Hydrogels/chemistry , Particle Size , Peptides/chemical synthesis , Peptides/chemistry , Serum Albumin, Bovine/chemistry , Surface Properties , Vitronectin/chemistry , alpha-2-HS-Glycoprotein/chemistry , alpha-2-HS-Glycoprotein/isolation & purification
9.
Exp Mol Med ; 53(2): 210-222, 2021 02.
Article in English | MEDLINE | ID: mdl-33526813

ABSTRACT

Osteoarthritis is characterized by structural alteration of joints. Fibrosis of the synovial tissue is often detected and considered one of the main causes of joint stiffness and pain. In our earlier proteomic study, increased levels of vitronectin (VTN) fragment (amino acids 381-397) were observed in the serum of osteoarthritis patients. In this work, the affinity of this fragment for integrins and its putative role in TGF-ß1 activation were investigated. A competition study determined the interaction of VTN(381-397 a.a.) with αVß6 integrin. Subsequently, the presence of αVß6 integrin was substantiated on primary human fibroblast-like synoviocytes (FLSs) by western blot and flow cytometry. By immunohistochemistry, ß6 was detected in synovial membranes, and its expression showed a correlation with tissue fibrosis. Moreover, ß6 expression was increased under TGF-ß1 stimulation; hence, a TGF-ß bioassay was applied. We observed that αVß6 could mediate TGF-ß1 bioavailability and that VTN(381-397 a.a.) could prevent TGF-ß1 activation by interacting with αVß6 in human FLSs and increased α-SMA. Finally, we analyzed serum samples from healthy controls and patients with osteoarthritis and other rheumatic diseases by nano-LC/Chip MS-MS, confirming the increased expression of VTN(381-397 a.a.) in osteoarthritis as well as in lupus erythematosus and systemic sclerosis. These findings corroborate our previous observations concerning the overexpression of VTN(381-397 a.a.) in osteoarthritis but also in other rheumatic diseases. This fragment interacts with αVß6 integrin, a receptor whose expression is increased in FLSs from the osteoarthritic synovial membrane and that can mediate the activation of the TGF-ß1 precursor in human FLSs.


Subject(s)
Antigens, Neoplasm/metabolism , Integrins/metabolism , Osteoarthritis/complications , Protein Interaction Domains and Motifs , Synovitis/etiology , Synovitis/metabolism , Transforming Growth Factor beta1/metabolism , Vitronectin/metabolism , Aged , Antigens, Neoplasm/genetics , Biomarkers , Chromatography, Liquid , Disease Susceptibility , Female , Humans , Immunohistochemistry , Immunophenotyping , Inflammation Mediators/metabolism , Integrins/genetics , Male , Middle Aged , Osteoarthritis/etiology , Osteoarthritis/pathology , Peptides/chemistry , Peptides/metabolism , Protein Binding , Proteomics/methods , Synoviocytes/metabolism , Synoviocytes/pathology , Synovitis/blood , Synovitis/pathology , Tandem Mass Spectrometry , Vitronectin/chemistry
10.
Molecules ; 25(22)2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33182593

ABSTRACT

Multivalent interactions frequently occur in biological systems and typically provide higher binding affinity and selectivity in target recognition than when only monovalent interactions are operative. Thus, taking inspiration by nature, bivalent or multivalent nucleic acid aptamers recognizing a specific biological target have been extensively studied in the last decades. Indeed, oligonucleotide-based aptamers are suitable building blocks for the development of highly efficient multivalent systems since they can be easily modified and assembled exploiting proper connecting linkers of different nature. Thus, substantial research efforts have been put in the construction of dimeric/multimeric versions of effective aptamers with various degrees of success in target binding affinity or therapeutic activity enhancement. The present review summarizes recent advances in the design and development of dimeric and multimeric DNA-based aptamers, including those forming G-quadruplex (G4) structures, recognizing different key proteins in relevant pathological processes. Most of the designed constructs have shown improved performance in terms of binding affinity or therapeutic activity as anti-inflammatory, antiviral, anticoagulant, and anticancer agents and their number is certainly bound to grow in the next future.


Subject(s)
Aptamers, Nucleotide/chemistry , G-Quadruplexes , Phosphoproteins/chemistry , RNA-Binding Proteins/chemistry , Anti-Inflammatory Agents/chemistry , Anticoagulants/chemistry , Antineoplastic Agents/chemistry , Antiviral Agents/chemistry , CD3 Complex/chemistry , Cell Adhesion Molecules/chemistry , DNA/chemistry , Dimerization , Humans , Immunoglobulin M/chemistry , Protein Structure, Secondary , Proto-Oncogene Proteins c-met/chemistry , Pyrrolidines/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Receptors, Antigen, T-Cell/chemistry , Vascular Endothelial Growth Factor A/metabolism , Vitronectin/chemistry , Nucleolin
11.
Int J Mol Sci ; 21(21)2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33105833

ABSTRACT

Significant amounts of enolase-a cytosolic enzyme involved in the glycolysis pathway-are exposed on the cell surface of Candida yeast. It has been hypothesized that this exposed enolase form contributes to infection-related phenomena such as fungal adhesion to human tissues, and the activation of fibrinolysis and extracellular matrix degradation. The aim of the present study was to characterize, in structural terms, the protein-protein interactions underlying these moonlighting functions of enolase. The tight binding of human vitronectin, fibronectin and plasminogen by purified C. albicans and C. tropicalis enolases was quantitatively analyzed by surface plasmon resonance measurements, and the dissociation constants of the formed complexes were determined to be in the 10-7-10-8 M range. In contrast, the binding of human proteins by the S.cerevisiae enzyme was much weaker. The chemical cross-linking method was used to map the sites on enolase molecules that come into direct contact with human proteins. An internal motif 235DKAGYKGKVGIAMDVASSEFYKDGK259 in C. albicans enolase was suggested to contribute to the binding of all three human proteins tested. Models for these interactions were developed and revealed the sites on the enolase molecule that bind human proteins, extensively overlap for these ligands, and are well-separated from the catalytic activity center.


Subject(s)
Fibronectins/metabolism , Phosphopyruvate Hydratase/metabolism , Plasminogen/metabolism , Vitronectin/metabolism , Amino Acid Motifs , Antibodies/metabolism , Binding, Competitive , Candida albicans/enzymology , Candida tropicalis/enzymology , Cytosol/enzymology , Fibronectins/chemistry , Host-Pathogen Interactions/physiology , Humans , Immobilized Proteins/metabolism , Models, Molecular , Phosphopyruvate Hydratase/chemistry , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/immunology , Plasminogen/chemistry , Vitronectin/chemistry
12.
Proc Natl Acad Sci U S A ; 117(31): 18504-18510, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32699145

ABSTRACT

The human blood protein vitronectin (Vn) is a major component of the abnormal deposits associated with age-related macular degeneration, Alzheimer's disease, and many other age-related disorders. Its accumulation with lipids and hydroxyapatite (HAP) has been demonstrated, but the precise mechanism for deposit formation remains unknown. Using a combination of solution and solid-state NMR experiments, cosedimentation assays, differential scanning fluorimetry (DSF), and binding energy calculations, we demonstrate that Vn is capable of binding both soluble ionic calcium and crystalline HAP, with high affinity and chemical specificity. Calcium ions bind preferentially at an external site, at the top of the hemopexin-like (HX) domain, with a group of four Asp carboxylate groups. The same external site is also implicated in HAP binding. Moreover, Vn acquires thermal stability upon association with either calcium ions or crystalline HAP. The data point to a mechanism whereby Vn plays an active role in orchestrating calcified deposit formation. They provide a platform for understanding the pathogenesis of macular degeneration and other related degenerative disorders, and the normal functions of Vn, especially those related to bone resorption.


Subject(s)
Calcium/metabolism , Durapatite/metabolism , Macular Degeneration/metabolism , Vitronectin/metabolism , Binding Sites , Calcium/chemistry , Durapatite/chemistry , Humans , Protein Binding , Vitronectin/chemistry
13.
J Biol Chem ; 295(29): 10008-10022, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32499371

ABSTRACT

Staphylococcus aureus is an important bacterial pathogen that can cause a wide spectrum of diseases in humans and other animals. S. aureus expresses a variety of virulence factors that promote infection with this pathogen. These include cell-surface proteins that mediate adherence of the bacterial cells to host extracellular matrix components, such as fibronectin and fibrinogen. Here, using immunoblotting, ELISA, and surface plasmon resonance analysis, we report that the iron-regulated surface determinant B (IsdB) protein, besides being involved in heme transport, plays a novel role as a receptor for the plasma and extracellular matrix protein vitronectin (Vn). Vn-binding activity was expressed by staphylococcal strains grown under iron starvation conditions when Isd proteins are expressed. Recombinant IsdB bound Vn dose dependently and specifically. Both near-iron transporter motifs NEAT1 and NEAT2 of IsdB individually bound Vn in a saturable manner, with KD values in the range of 16-18 nm Binding of Vn to IsdB was specifically blocked by heparin and reduced at high ionic strength. Furthermore, IsdB-expressing bacterial cells bound significantly higher amounts of Vn from human plasma than did an isdB mutant. Adherence to and invasion of epithelial and endothelial cells by IsdB-expressing S. aureus cells was promoted by Vn, and an αvß3 integrin-blocking mAb or cilengitide inhibited adherence and invasion by staphylococci, suggesting that Vn acts as a bridge between IsdB and host αvß3 integrin.


Subject(s)
Cation Transport Proteins/chemistry , Staphylococcus aureus/chemistry , Vitronectin/chemistry , Cation Transport Proteins/metabolism , Humans , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Protein Binding , Staphylococcus aureus/metabolism , Vitronectin/metabolism
14.
Colloids Surf B Biointerfaces ; 193: 111031, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32408257

ABSTRACT

Cardiovascular diseases are the leading cause of death around the world according to the World Health Organization. In-stent restenosis is an inflammatory response of the immune system to endovascular stent implantation in atherosclerotic patients. Biocompatible and biodegradable polymers are of great interest in this field in order to limit the side effects of stent treatments. Poly([R,S]-3,3-dimethylmalic acid) (PDMMLA) is a new biodegradable statistical polyester which presents promising properties as a stent coating. In this work, we studied by dynamic tensiometry, the adhesion of extracellular matrix proteins (bovine serum albumin, fibronectin, fibrinogen, and vitronectin) and plasma membrane proteoglycan (syndecan-4) on three PDMMLA derivatives with different hydrophilicity levels. The results show that proteins have different adhesion profiles and affinity on these surfaces. They show similar behavior on the most hydrophilic surface, making hydrophilic, ionic and hydrogen type bonds. Then we compared each protein's individual profile to that of a mixture of all studied proteins. The comparison shows that vitronectin and syndecan-4 are the quantitatively dominating proteins adsorbed by specific interactions. Based on the results from previous studies, this work allowed us to identify the most important PDMMLA surface as a promising biomaterial for bioactive stent-coating.


Subject(s)
Malates/chemistry , Polymers/chemistry , Adsorption , Animals , Cattle , Fibrinogen/chemistry , Fibronectins/chemistry , Molecular Structure , Particle Size , Polymers/chemical synthesis , Serum Albumin, Bovine/chemistry , Surface Tension , Syndecan-4/chemistry , Vitronectin/chemistry
15.
Protein Sci ; 29(2): 494-508, 2020 02.
Article in English | MEDLINE | ID: mdl-31682300

ABSTRACT

The serine protease inhibitor, plasminogen activator inhibitor Type-1 (PAI-1) is a metastable protein that undergoes an unusual transition to an inactive conformation with a short half-life of only 1-2 hr. Circulating PAI-1 is bound to a cofactor vitronectin, which stabilizes PAI-1 by slowing this latency conversion. A well-characterized PAI-1-binding site on vitronectin is located within the somatomedin B (SMB) domain, corresponding to the first 44 residues of the protein. Another PAI-1 recognition site has been identified with an engineered form of vitronectin lacking the SMB domain, yet retaining PAI-1 binding capacity (Schar, Blouse, Minor, Peterson. J Biol Chem. 2008;283:28487-28496). This additional binding site is hypothesized to lie within an intrinsically disordered domain (IDD) of vitronectin. To localize the putative binding site, we constructed a truncated form of vitronectin containing 71 amino acids from the N-terminus, including the SMB domain and an additional 24 amino acids from the IDD region. This portion of the IDD is rich in acidic amino acids, which are hypothesized to be complementary to several basic residues identified within an extensive vitronectin-binding site mapped on PAI-1 (Schar, Jensen, Christensen, Blouse, Andreasen, Peterson. J Biol Chem. 2008;283:10297-10309). Steady-state and stopped-flow fluorescence measurements demonstrate that the truncated form of vitronectin exhibits the same rapid biphasic association as full-length vitronectin and that the IDD hosts the elusive second PAI-1 binding site that lies external to the SMB domain of vitronectin.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Plasminogen Activator Inhibitor 1/chemistry , Vitronectin/chemistry , Binding Sites , Humans , Models, Molecular , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/isolation & purification , Spectrometry, Fluorescence
16.
Biochemistry ; 58(51): 5117-5134, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31793295

ABSTRACT

Small-angle neutron scattering (SANS) measurements were pursued to study human vitronectin, a protein found in tissues and the circulation that regulates cell adhesion/migration and proteolytic cascades that govern hemostasis and pericellular proteolysis. Many of these functions occur via interactions with its binding partner, plasminogen activator inhibitor-1 (PAI-1), the chief inhibitor of proteases that lyse and activate plasminogen. We focused on a region of vitronectin that remains uncharacterized from previous X-ray scattering, nuclear magnetic resonance, and computational modeling approaches and which we propose is involved in binding to PAI-1. This region, which bridges the N-terminal somatomedin B (SMB) domain with a large central ß-propeller domain of vitronectin, appears unstructured and has characteristics of an intrinsically disordered domain (IDD). The effect of osmolytes was evaluated using circular dichroism and SANS to explore the potential of the IDD to undergo a disorder-to-order transition. The results suggest that the IDD favors a more ordered structure under osmotic pressure; SANS shows a smaller radius of gyration (Rg) and a more compact fold of the IDD upon addition of osmolytes. To test whether PAI-1 binding is also coupled to folding within the IDD structure, a set of SANS experiments with contrast variation were performed on the complex of PAI-1 with a vitronectin fragment corresponding to the N-terminal 130 amino acids (denoted the SMB-IDD because it contains the SMB domain and IDD in linear sequence). Analysis of the SANS data using the Ensemble Optimization Method confirms that the SMB-IDD adopts a more compact configuration when bound to PAI-1. Calculated structures for the PAI-1:SMB-IDD complex suggest that the IDD provides an interaction surface outside of the primary PAI-1-binding site located within the SMB domain; this binding is proposed to lead to the assembly of higher-order structures of vitronectin and PAI-1 commonly found in tissues.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Vitronectin/chemistry , Vitronectin/metabolism , Models, Molecular , Protein Binding , Protein Domains
17.
Nanoscale ; 11(40): 18806-18824, 2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31595922

ABSTRACT

The application of lipid-based nanoparticle (LNP) delivery systems remains a popular strategy for the systemic delivery of gene therapies to specific disease targets, including solid tumors. It is now well acknowledged that upon systemic administration, biomolecules from blood will adsorb onto nanoparticles' surfaces, forming a "protein corona", affording nanoparticles a "biological identity" on top of their "synthetic identity". Detailed analysis of nanoparticle protein corona is gradually revealing the "missing link" between nanoparticle chemical properties and the biological identity. Nevertheless, the discovery of nanoparticle protein corona's impact on tumor delivery is limited. In this study, we demonstrate that protein corona can be manipulated by formulation composition and particle surface charge changes, and a single lipid switch could switch the nanoparticle protein corona profile. The protein corona composition differences had a profound impact on cell transfection, in vivo biodistribution as well as tumor-specific delivery efficiency. Nanoparticles with apolipoprotein-rich corona showed better delivery to hepatocellular carcinoma (HepG2) as compared to those with vitronectin-rich corona. In addition, we found that, the PEG conjugated lipid chain length and PEG amount in LNPs were key factors to consider in successful RNA interference therapy for solid tumors.


Subject(s)
Apolipoproteins , Carcinoma, Hepatocellular/drug therapy , Drug Delivery Systems , Lipids , Liver Neoplasms/drug therapy , Nanoparticles , Oligonucleotides , Transfection , Vitronectin , Animals , Apolipoproteins/chemistry , Apolipoproteins/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Female , Hep G2 Cells , Humans , Lipids/chemistry , Lipids/pharmacology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Oligonucleotides/chemistry , Oligonucleotides/pharmacology , Vitronectin/chemistry , Vitronectin/pharmacology , Xenograft Model Antitumor Assays
18.
Sci Adv ; 5(9): eaax5068, 2019 09.
Article in English | MEDLINE | ID: mdl-31535027

ABSTRACT

Vitronectin (Vn) is a major component of blood that controls many processes central to human biology. It is a drug target and a key factor in cell and tissue engineering applications, but despite long-standing efforts, little is known about the molecular basis for its functions. Here, we define the domain organization of Vn, report the crystal structure of its carboxyl-terminal domain, and show that it harbors the binding site for the Yersinia pestis outer membrane protein Ail, which recruits Vn to the bacterial cell surface to evade human host defenses. Vn forms a single four-bladed ß/α-propeller that serves as a hub for multiple functions. The structure explains key features of native Vn and provides a blueprint for understanding and targeting this essential human protein.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Virulence Factors/metabolism , Vitronectin/metabolism , Yersinia pestis/metabolism , Amino Acid Sequence , Bacterial Outer Membrane Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Protein Binding , Protein Conformation , Sequence Homology , Virulence Factors/chemistry , Vitronectin/chemistry
19.
Biomater Sci ; 7(10): 4345-4362, 2019 Sep 24.
Article in English | MEDLINE | ID: mdl-31411209

ABSTRACT

Recombinant vitronectin-grafted hydrogels were developed by adjusting surface charge of the hydrogels with grafting of poly-l-lysine for optimal culture of human embryonic stem cells (hESCs) under xeno- and feeder-free culture conditions, with elasticity regulated by crosslinking time (10-30 kPa), in contrast to conventional recombinant vitronectin coating dishes, which have a fixed stiff surface (3 GPa). hESCs proliferated on the hydrogels for over 10 passages and differentiated into the cells derived from three germ layers indicating the maintenance of pluripotency. hESCs on the hydrogels differentiated into cardiomyocytes under xeno-free culture conditions with much higher efficiency (80% of cTnT+ cells) than those on conventional recombinant vitronectin or Matrigel-coating dishes just only after 12 days of induction. It is important to have an optimal design of cell culture biomaterials where biological cues (recombinant vitronectin) and physical cues (optimal elasticity) are combined for high differentiation of hESCs into specific cell lineages, such as cardiomyocytes, under xeno-free and feeder-free culture conditions.


Subject(s)
Human Embryonic Stem Cells/cytology , Hydrogels/chemistry , Vitronectin/chemistry , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Human Embryonic Stem Cells/metabolism , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...