Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.488
Filter
1.
ACS Biomater Sci Eng ; 10(5): 3017-3028, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38655791

ABSTRACT

Macroporous cryogels are attractive scaffolds for biomedical applications, such as biomolecular immobilization, diagnostic sensing, and tissue engineering. In this study, thiol-reactive redox-responsive cryogels with a porous structure are prepared using photopolymerization of a pyridyl disulfide poly(ethylene glycol) methacrylate (PDS-PEG-MA) monomer. Reactive cryogels are produced using PDS-PEG-MA and hydrophilic poly(ethylene glycol) methyl ether methacrylate (PEGMEMA) monomers, along with a PEG-based cross-linker and photoinitiator. Functionalization of cryogels using a fluorescent dye via the disulfide-thiol exchange reactions is demonstrated, followed by release under reducing conditions. For ligand-mediated protein immobilization, first, thiol-containing biotin or mannose is conjugated onto the cryogels. Subsequently, fluorescent dye-labeled proteins streptavidin and concanavalin A (ConA) are immobilized via ligand-mediated conjugation. Furthermore, we demonstrate that the mannose-decorated cryogel could capture ConA selectively from a mixture of lectins. The efficiency of protein immobilization could be easily tuned by changing the ratio of the thiol-sensitive moiety in the scaffold. Finally, an integrin-binding cell adhesive peptide is attached to cryogels to achieve successful attachment, and the on-demand detachment of integrin-receptor-rich fibroblast cells is demonstrated. Redox-responsive cryogels can serve as potential scaffolds for a variety of biomedical applications because of their facile synthesis and modification.


Subject(s)
Cryogels , Oxidation-Reduction , Polyethylene Glycols , Cryogels/chemistry , Polyethylene Glycols/chemistry , Animals , Concanavalin A/chemistry , Concanavalin A/metabolism , Methacrylates/chemistry , Mice , Mannose/chemistry , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Sulfhydryl Compounds/chemistry , Streptavidin/chemistry , Streptavidin/metabolism , Proteins/chemistry , Proteins/metabolism , Biotin/chemistry , Biotin/metabolism , Biotin/analogs & derivatives , Porosity
2.
Biomater Sci ; 12(11): 2841-2864, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38683585

ABSTRACT

Polymer-based biomaterials have received a lot of attention due to their biomedical, agricultural, and industrial potential. Soluble protein-polymer bioconjugates, immobilized proteins, and encapsulated proteins have been shown to tune enzymatic activity, improved pharmacokinetic ability, increased chemical and thermal stability, stimuli responsiveness, and introduced protein recovery. Controlled polymerization techniques, increased protein-polymer attachment techniques, improved polymer surface grafting techniques, controlled polymersome self-assembly, and sophisticated characterization methods have been utilized for the development of well-defined polymer-based biomaterials. In this review we aim to provide a brief account of the field, compare these methods for engineering biomaterials, provide future directions for the field, and highlight impacts of these forms of bioconjugation.


Subject(s)
Polymers , Polymers/chemistry , Biocompatible Materials/chemistry , Immobilized Proteins/chemistry , Proteins/chemistry , Humans , Protein Stability , Animals
3.
Nanotechnology ; 35(32)2024 May 21.
Article in English | MEDLINE | ID: mdl-38688249

ABSTRACT

Dealing with bone defects is a significant challenge to global health. Electrospinning in bone tissue engineering has emerged as a solution to this problem. In this study, we designed a PVDF-b-PTFE block copolymer by incorporating TFE, which induced a phase shift in PVDF fromαtoß, thereby enhancing the piezoelectric effect. Utilizing the electrospinning process, we not only converted the material into a film with a significant surface area and high porosity but also intensified the piezoelectric effect. Then we used polydopamine to immobilize BMP-2 onto PVDF-b-PTFE electrospun nanofibrous membranes, achieving a controlled release of BMP-2. The scaffold's characters were examined using SEM and XRD. To assess its osteogenic effectsin vitro, we monitored the proliferation of MC3T3-E1 cells on the fibers, conducted ARS staining, and measured the expression of osteogenic genes.In vivo, bone regeneration effects were analyzed through micro-CT scanning and HE staining. ELISA assays confirmed that the sustained release of BMP-2 can be maintained for at least 28 d. SEM images and CCK-8 results demonstrated enhanced cell viability and improved adhesion in the experimental group. Furthermore, the experimental group exhibited more calcium nodules and higher expression levels of osteogenic genes, including COL-I, OCN, and RUNX2. HE staining and micro-CT scans revealed enhanced bone tissue regeneration in the defective area of the PDB group. Through extensive experimentation, we evaluated the scaffold's effectiveness in augmenting osteoblast proliferation and differentiation. This study emphasized the potential of piezoelectric PVDF-b-PTFE nanofibrous membranes with controlled BMP-2 release as a promising approach for bone tissue engineering, providing a viable solution for addressing bone defects.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Regeneration , Indoles , Nanofibers , Osteogenesis , Polymers , Tissue Engineering , Tissue Scaffolds , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/metabolism , Nanofibers/chemistry , Bone Regeneration/drug effects , Animals , Mice , Indoles/chemistry , Indoles/pharmacology , Polymers/chemistry , Polymers/pharmacology , Tissue Engineering/methods , Osteogenesis/drug effects , Tissue Scaffolds/chemistry , Cell Proliferation/drug effects , Cell Line , Immobilized Proteins/pharmacology , Immobilized Proteins/chemistry , Cell Survival/drug effects
4.
J Chromatogr A ; 1722: 464902, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38636150

ABSTRACT

Although immobilized metal ion affinity chromatography (IMAC) is one of the most effective methods for purifying his-tagged proteins, it has limitations such as expensive commercial resins and non-specific binding of unwanted proteins to the nickel immobilized on the resin. In this study, biocompatible chitosan and porous chitosan membranes as alternative resins were synthesized for protein immobilization and purification, but finally porous chitosan membrane was selected due to its higher porosity and consequently higher nickel adsorption. Once the membrane was functionalized with nickel ions and its metal adsorption confirmed by EDS and ICP methods, it was used to immobilize and purify recombinant ß-NGF as a protein model with his-tag tail in batch-fashion. Protein binding and purification were also approved by FTIR and UV-Vis spectroscopy and SDS-PAGE technique. Our results indicated that the protein of interest could bind to the nickel-functionalized porous chitosan membrane with high efficiency at pH=7. Furthermore, for protein purification, the pH value of 6 and an imidazole concentration of 750 mM were suggested for the final elution buffer. In conclusion, nickel-functionalized porous chitosan membrane could be a suitable alternative to IMAC for low cost and specific protein immobilization and purification.


Subject(s)
Chitosan , Chromatography, Affinity , Histidine , Membranes, Artificial , Nickel , Nickel/chemistry , Chitosan/chemistry , Chromatography, Affinity/methods , Histidine/chemistry , Porosity , Adsorption , Immobilized Proteins/chemistry , Hydrogen-Ion Concentration , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
5.
Colloids Surf B Biointerfaces ; 236: 113818, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38417347

ABSTRACT

The feasibility of immobilized protein-based biodetection relies critically on the activity of the immobilized proteins as well as the biocompatibility of the protein surface. Although many protein immobilization strategies have been developed with satisfied detection readout signals. Non-specific interactions caused by the protein-coating surface are still of great concern since they often interfere with or affect the reliability of detection. Herein, we developed a highly efficient G protein-coupled receptor (GPCR) immobilization method by the combination of polyethylene glycol (PEG) with a self-labeling enzyme-catalyzed reaction. The immobilization relies on the covalent interaction between the fusion tag of a target GPCR (kinase domain of epidermal growth factor receptor, EGFR) and its covalent inhibitor ibrutinib, which is modified on PEGylated silica gels. Two types of GPCRs, N-methyl-D-aspartate 2 A receptor (NMDAR2A) and endothelin A receptor (ETAR), were used as examples to realize protein immobilization. The GPCR modified gels and the affinity columns packed with them have been extensively characterized, in terms of non-specific adsorptions, retention factor (k'), half peak width (W1/2), tailing factor (Tf), theoretical plates (N), and association and dissociation constants of the ligands with the receptors. The immobilized GPCRs with reduced non-specific interactions and enhanced fouling resistance, salt tolerance, and chromatographic performance were clearly observed. We believe it is the first work to introduce PEGylation in GPCR immobilization and provide comprehensive proof-of-concept studies to illustrate the improved antifouling property, salt tolerance, and chromatographic performance. This method could be generally applicable in other immobilized protein-based technology for reliable biodetection.


Subject(s)
Receptors, G-Protein-Coupled , Salt Tolerance , Reproducibility of Results , Receptors, G-Protein-Coupled/metabolism , Immobilized Proteins/chemistry , Gels
6.
Acta Biomater ; 179: 371-384, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38382829

ABSTRACT

Endovascular stenting is a safer alternative to open surgery for use in treating cerebral arterial stenosis and significantly reduces the recurrence of ischemic stroke, but the widely used bare-metal stents (BMSs) often result in in-stent restenosis (ISR). Although evidence suggests that drug-eluting stents are superior to BMSs in the short term, their long-term performances remain unknown. Herein, we propose a potential vascular stent modified by immobilizing clickable chemerin 15 (C15) peptides on the stent surface to suppress coagulation and restenosis. Various characterization techniques and an animal model were used to evaluate the surface properties of the modified stents and their effects on endothelial injury, platelet adhesion, and inflammation. The C15-immobilized stent could prevent restenosis by minimizing endothelial injury, promoting physiological healing, restraining the platelet-leukocyte-related inflammatory response, and inhibiting vascular smooth muscle cell proliferation and migration. Furthermore, in vivo studies demonstrated that the C15-immobilized stent mitigated inflammation, suppressed neointimal hyperplasia, and accelerated endothelial restoration. The use of surface-modified, anti-inflammatory, endothelium-friendly stents may be of benefit to patients with arterial stenosis. STATEMENT OF SIGNIFICANCE: Endovascular stenting is increasingly used for cerebral arterial stenosis treatment, aiming to prevent and treat ischemic stroke. But an important accompanying complication is in-stent restenosis (ISR). Persistent inflammation has been established as a hallmark of ISR and anti-inflammation strategies in stent modification proved effective. Chemerin 15, an inflammatory resolution mediator with 15-aa peptide, was active at picomolar through cell surface receptor, no need to permeate cell membrane and involved in resolution of inflammation by inhibiting inflammatory cells adhesion, modulating macrophage polarization into protective phenotype, and reducing inflammatory factors release. The implications of this study are that C15 immobilized stent favors inflammation resolution and rapid re-endothelialization, and exhibits an inhibitory role of restenosis. As such, it helps the decreased incidence of ISR.


Subject(s)
Chemokines , Hyperplasia , Neointima , Stents , Animals , Chemokines/metabolism , Humans , Neointima/pathology , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Intercellular Signaling Peptides and Proteins/pharmacology , Peptides/pharmacology , Peptides/chemistry , Mice , Cell Proliferation/drug effects , Wound Healing/drug effects , Immobilized Proteins/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects
7.
Molecules ; 28(12)2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37375186

ABSTRACT

Biological organisms rely on proteins to perform the majority of their functions. Most protein functions are based on their physical motions (conformational changes), which can be described as transitions between different conformational states in a multidimensional free-energy landscape. A comprehensive understanding of this free-energy landscape is therefore of paramount importance for understanding the biological functions of proteins. Protein dynamics includes both equilibrium and nonequilibrium motions, which typically exhibit a wide range of characteristic length and time scales. The relative probabilities of various conformational states in the energy landscape, the energy barriers between them, their dependence on external parameters such as force and temperature, and their connection to the protein function remain largely unknown for most proteins. In this paper, we present a multimolecule approach in which the proteins are immobilized at well-defined locations on Au substrates using an atomic force microscope (AFM)-based patterning method called nanografting. This method enables precise control over the protein location and orientation on the substrate, as well as the creation of biologically active protein ensembles that self-assemble into well-defined nanoscale regions (protein patches) on the gold substrate. We performed AFM-force compression and fluorescence experiments on these protein patches and measured the fundamental dynamical parameters such as protein stiffness, elastic modulus, and transition energies between distinct conformational states. Our results provide new insights into the processes that govern protein dynamics and its connection to protein function.


Subject(s)
Immobilized Proteins , Proteins , Microscopy, Atomic Force , Proteins/chemistry , Mechanical Phenomena , Microscopy, Fluorescence
8.
Int J Mol Sci ; 24(7)2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37047243

ABSTRACT

Actinomycin is a family of chromogenic lactone peptides that differ in their peptide portions of the molecule. An antimicrobial peptide, actinomycin X2 (Ac.X2), was produced through the fermentation of a Streptomyces cyaneofuscatus strain. Immobilization of Ac.X2 onto a prepared silk fibroin (SF) film was done through a carbodiimide reaction. The physical properties of immobilized Ac.X2 (antimicrobial films, AMFs) were analyzed by ATR-FTIR, SEM, AFM, and WCA. The findings from an in vitro study showed that AMFs had a more broad-spectrum antibacterial activity against both S. aureus and E. coli compared with free Ac.X2, which showed no apparent strong effect against E. coli. These AMFs showed a suitable degradation rate, good hemocompatibility, and reduced cytotoxicity in the biocompatibility assay. The results of in vivo bacterially infected wound healing experiments indicated that wound inflammation was prevented by AMFs, which promoted wound repair and improved the wound microenvironment. This study revealed that Ac.X2 transformation is a potential candidate for skin wound healing.


Subject(s)
Antimicrobial Peptides , Dactinomycin , Fibroins , Immobilized Proteins , Wound Healing , Dactinomycin/chemistry , Dactinomycin/pharmacology , Fibroins/chemistry , Fibroins/pharmacology , Immobilized Proteins/chemistry , Immobilized Proteins/pharmacology , Wound Healing/drug effects , Streptomyces/metabolism , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Spectroscopy, Fourier Transform Infrared , Microscopy, Atomic Force , Fermentation , Materials Testing , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Animals , Rats , Male , Rats, Sprague-Dawley
9.
Langmuir ; 39(10): 3720-3728, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36857653

ABSTRACT

Proteins adsorbed to gold nanoparticles (AuNPs) form bioconjugates and are critical to many emerging technologies for drug delivery, diagnostics, therapies, and other biomedical applications. A thorough understanding of the interaction between the immobilized protein and AuNP is essential for the bioconjugate to perform as designed. Here, we explore a correlation between the number of solvent-accessible thiol groups on a protein and the protein desorption rate from the AuNP surface in the presence of a competing protein. The chemical modification of human serum albumin (HSA) was carried out to install additional free thiols using Traut's reagent and create a library of HSA analogues by tailoring the molar excess of the Traut's reagent. We pre-adsorbed HSA variants onto the AuNP surface, and the resulting bioconjugates were then exposed to IgG antibody, and protein exchange was monitored as a function of time. We found that the rate of HSA displacement from the AuNP correlated with the experimentally measured number of accessible free thiol groups. Additionally, bioconjugates were synthesized using thiolated analogues of bovine serum albumin (BSA) and suspended in serum as a model for a complex sample matrix. Similarly, desorption rates with serum proteins were modulated with solvent-accessible thiols on the immobilized protein. These results further highlight the key role of Au-S bonds in the formation of protein-AuNP conjugates and provide a pathway to systematically control the number of free thiols on a protein, enabling the controlled release of protein from the surface of AuNP.


Subject(s)
Metal Nanoparticles , Serum Albumin , Humans , Gold/chemistry , Metal Nanoparticles/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Human , Solvents , Sulfhydryl Compounds , Immobilized Proteins
10.
Anal Chem ; 95(13): 5643-5651, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36939216

ABSTRACT

Protein immobilization is of utmost importance in many areas, where various proteins are used for selective detection of target compounds. Despite the importance given to determine the amount of immobilized protein, there is no simple method that allows direct, noninvasive detection. In this work, a method based on pH transition, occurring during change of solution ionic strength, was developed. The method utilized the ionic character of the immobilized protein while implementing biologically compatible buffers. Five different proteins, namely, glucose oxidase, horseradish peroxidase, bovine serum albumin, lysozyme, and protein A, were immobilized in different amounts on a porous polymeric matrix, and their pH transition was measured using lactate buffer of various concentrations and pH values. A linear correlation was found between the amount of immobilized protein and the amplitude of the pH transition, allowing the detection down to 2 nmol of immobilized protein. By changing the buffer concentration and pH, the sensitivity of the method could be tailored. Criteria based on the symmetry of the pH transition peak have been developed to determine if a particular measurement is within a linear range. In addition, a mathematical model was developed enabling prediction of pH transition profiles based solely on the protein amino acid sequence, the buffer pKa value(s), and the amount of immobilized protein.Hence, it can be used to design pH transition method experiments to achieve the required sensitivity for a target sample. Since the proposed method is noninvasive, it can be routinely applied during optimization of the immobilization protocol, for quality control, and also as an in-process monitoring tool.


Subject(s)
Glucose Oxidase , Serum Albumin, Bovine , Glucose Oxidase/metabolism , Horseradish Peroxidase/chemistry , Serum Albumin, Bovine/chemistry , Immobilized Proteins , Enzymes, Immobilized/chemistry , Hydrogen-Ion Concentration
11.
J Biotechnol ; 364: 31-39, 2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36702256

ABSTRACT

One of the key metrics in the design of biosensors is the presence of an effective capture layer. Surface-immobilized proteins (especially as a part of antibody-antigen combinations) are the most commonly used capture ligands in biosensors. The surface coverage of these proteins in flow-based biosensors are affected by both the linker chemistry used to attach them as well as the microchannel geometry. We used streptavidin as a model protein to compare glutaraldehyde, EDC-NHS, sulfo-SMCC and sulfo-NHS-biotin as linkers inside straight, serpentine and square-wave microchannel geometries. We found that straight microchannels achieve the highest degree of protein immobilization compared to serpentine and square-wave microchannels, irrespective of the linker chemistry used. We also showed that for a given microchannel geometry, sulfo-NHS-biotin leads to the highest immobilization of streptavidin among all the linkers.


Subject(s)
Biosensing Techniques , Proteins , Streptavidin/metabolism , Biotin/chemistry , Immobilized Proteins , Lab-On-A-Chip Devices
12.
Biomater Adv ; 141: 213092, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36191539

ABSTRACT

Functional surfaces that enable both spatial and temporal control of biomolecules immobilization have attracted enormous attention for various fields including smart biointerface materials, high-throughput bioarrays, and fundamental research in the biosciences. Here, a flexible and promising method was presented for regulating the spatiotemporal arrangement of multiple biomolecules by constructing the topographically and chemically diverse polymer brushes patterned surfaces. A series of polymer brushes patterned surfaces, including antifouling brushes patterned surface, epoxy-presenting brushes patterned surface without and with antifouling background layer, were fabricated to control the spatial distribution of protein and cell adhesion through specific and nonspecific means. The fluorescence measurements demonstrated the effectiveness of spatially regulating the density of surface-immobilized protein through controlling the areal thickness of the poly (glycidyl methacrylate) (PGMA) brush patterns, leading to various complex patterns featuring well-defined biomolecule concentration gradients. Furthermore, a multiplexed surface bearing epoxy groups and azido groups with various areal densities was fabricated for regulating the spatiotemporal arrangement of different proteins, enabling binary biomolecules patterns with higher degrees of functionality and complexity. The presented strategy for the spatiotemporal control of biomolecules immobilization would boost the development of dynamic and multifunctional biosystems.


Subject(s)
Immobilized Proteins , Polymers , Cell Adhesion , Polymers/chemistry
13.
N Biotechnol ; 72: 114-121, 2022 Dec 25.
Article in English | MEDLINE | ID: mdl-36307012

ABSTRACT

In Escherichia coli, acyl carrier protein (ACP) is posttranslationally converted into its active holo-ACP form via covalent linkage of 4'-phosphopantetheine (4'-PP) to residue serine-36. We found that the long flexible 4'-PP arm could react chemoselectively with the iodoacetyl group introduced on solid supports with high efficiency under mild conditions. Based on this finding, we developed site-selective immobilisation of proteins via the active holo-ACP fusion tag, independently of the physicochemical properties of the protein of interest. Furthermore, the molecular ratios of co-immobilised proteins can be manipulated because the tethering process is predominantly directed by the molar concentrations of diverse holo-ACP fusions during co-immobilisation. Conveniently tuning the molecular ratios of co-immobilised proteins allows their cooperation, leading to a highly productive multi-protein co-immobilisation system. Kinetic studies of enzymes demonstrated that α-amylase (Amy) and methyl parathion hydrolase (MPH) immobilised via active tag holo-ACP had higher catalytic efficiency (kcat/Km) in comparison with their corresponding counterparts immobilised via the sulfhydryl groups (-SH) of these proteins. The immobilised holo-ACP-Amy also presented higher thermostability compared with free Amy. The enhanced α-amylase thermostability upon immobilisation via holo-ACP renders it more suitable for industrial application.


Subject(s)
Acyl Carrier Protein , Pantetheine , Kinetics , Pantetheine/chemistry , Pantetheine/metabolism , Acyl Carrier Protein/chemistry , Acyl Carrier Protein/metabolism , Escherichia coli/metabolism , alpha-Amylases/metabolism , Immobilized Proteins/metabolism
14.
J Immunol ; 209(6): 1180-1188, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35977795

ABSTRACT

The structure of wild-type pentameric C-reactive protein (CRP) is stabilized by two calcium ions that are required for the binding of CRP to its ligand phosphocholine. CRP in its structurally altered pentameric conformations also binds to proteins that are denatured and aggregated by immobilization on microtiter plates; however, the identity of the ligand on immobilized proteins remains unknown. We tested the hypotheses that immobilization of proteins generated an amyloid-like structure and that amyloid-like structure was the ligand for structurally altered pentameric CRP. We found that the Abs to amyloid-ß peptide 1-42 (Aß) reacted with immobilized proteins, indicating that some immobilized proteins express an Aß epitope. Accordingly, four different CRP mutants capable of binding to immobilized proteins were constructed, and their binding to fluid-phase Aß was determined. All CRP mutants bound to fluid-phase Aß, suggesting that Aß is a ligand for structurally altered pentameric CRP. In addition, the interaction between CRP mutants and Aß prevented the formation of Aß fibrils. The growth of Aß fibrils was also halted when CRP mutants were added to growing fibrils. Biochemical analyses of CRP mutants revealed altered topology of the Ca2+-binding site, suggesting a role of this region of CRP in binding to Aß. Combined with previous reports that structurally altered pentameric CRP is generated in vivo, we conclude that CRP is a dual pattern recognition molecule and an antiamyloidogenic protein. These findings have implications for Alzheimer's and other neurodegenerative diseases caused by amyloidosis and for the diseases caused by the deposition of otherwise fluid-phase proteins.


Subject(s)
C-Reactive Protein , Phosphorylcholine , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , C-Reactive Protein/metabolism , Calcium/metabolism , Epitopes , Immobilized Proteins , Ligands , Peptide Fragments
15.
Biointerphases ; 17(2): 021005, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35477241

ABSTRACT

Biomaterials used for blood contacting devices are inherently thrombogenic. Antithrombotic agents can be used as surface modifiers on biomaterials to reduce thrombus formation on the surface and to maintain device efficacy. For quality control and to assess the effectiveness of immobilization strategies, it is necessary to quantify the surface-immobilized antithrombotic agent directly. There are limited methods that allow direct quantification on device surfaces such as catheters. In this study, an enzyme immunoassay (EIA) has been developed to measure the density of a synthetic antithrombin-heparin (ATH) covalent complex immobilized on a catheter surface. The distribution of the immobilized ATH was further characterized by an immunohistochemical assay. This analyte-specific EIA is relatively simple and has high throughput, thus providing a tool for quantitative analysis of biomaterial surface modifications. These methods may be further modified to evaluate plasma proteins adsorbed and immobilized on various biomaterial surfaces of complex shapes, with a range of bioactive functionalities, as well as to assess conformational changes of proteins using specific antibodies.


Subject(s)
Heparin , Membrane Proteins , Antithrombins/chemistry , Biocompatible Materials , Fibrinolytic Agents , Heparin/chemistry , Immobilized Proteins , Surface Properties
16.
Nat Commun ; 13(1): 341, 2022 01 17.
Article in English | MEDLINE | ID: mdl-35039540

ABSTRACT

Robust regulatory signals in the cell often depend on interactions between short linear motifs (SLiMs) and globular proteins. Many of these interactions are poorly characterized because the binding proteins cannot be produced in the amounts needed for traditional methods. To address this problem, we developed a single-molecule off-rate (SMOR) assay based on microscopy of fluorescent ligand binding to immobilized protein partners. We used it to characterize substrate binding to the Anaphase-Promoting Complex/Cyclosome (APC/C), a ubiquitin ligase that triggers chromosome segregation. We find that SLiMs in APC/C substrates (the D box and KEN box) display distinct affinities and specificities for the substrate-binding subunits of the APC/C, and we show that multiple SLiMs in a substrate generate a high-affinity multivalent interaction. The remarkably adaptable substrate-binding mechanisms of the APC/C have the potential to govern the order of substrate destruction in mitosis.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/chemistry , Anaphase-Promoting Complex-Cyclosome/metabolism , Saccharomyces cerevisiae/metabolism , Single Molecule Imaging , Amino Acid Motifs , Amino Acid Sequence , Anisotropy , Humans , Immobilized Proteins/metabolism , Ligands , Peptides/chemistry , Peptides/metabolism , Protein Binding , Proteolysis , Substrate Specificity
17.
Cells ; 10(12)2021 12 10.
Article in English | MEDLINE | ID: mdl-34944004

ABSTRACT

The hedgehog (Hh) and Wnt pathways, crucial for the embryonic development and stem cell proliferation of Metazoa, have long been known to have similarities that argue for their common evolutionary origin. A surprising additional similarity of the two pathways came with the discovery that WIF1 proteins are involved in the regulation of both the Wnt and Hh pathways. Originally, WIF1 (Wnt Inhibitory Factor 1) was identified as a Wnt antagonist of vertebrates, but subsequent studies have shown that in Drosophila, the WIF1 ortholog serves primarily to control the distribution of Hh. In the present, work we have characterized the interaction of the human WIF1 protein with human sonic hedgehog (Shh) using Surface Plasmon Resonance spectroscopy and reporter assays monitoring the signaling activity of human Shh. Our studies have shown that human WIF1 protein binds human Shh with high affinity and inhibits its signaling activity efficiently. Our observation that the human WIF1 protein is a potent antagonist of human Shh suggests that the known tumor suppressor activity of WIF1 may not be ascribed only to its role as a Wnt inhibitor.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hedgehog Proteins/antagonists & inhibitors , Animals , Cell Line , Hedgehog Proteins/metabolism , Humans , Immobilized Proteins/metabolism , Kinetics , Mice , NIH 3T3 Cells , Protein Binding , Signal Transduction
18.
Nucleic Acids Res ; 49(19): 11211-11223, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34614173

ABSTRACT

Binding and unbinding of transcription factors to DNA are kinetically controlled to regulate the transcriptional outcome. Control of the release of the transcription factor NF-κB from DNA is achieved through accelerated dissociation by the inhibitor protein IκBα. Using single-molecule FRET, we observed a continuum of conformations of NF-κB in free and DNA-bound states interconverting on the subseconds to minutes timescale, comparable to in vivo binding on the seconds timescale, suggesting that structural dynamics directly control binding kinetics. Much of the DNA-bound NF-κB is partially bound, allowing IκBα invasion to facilitate DNA dissociation. IκBα induces a locked conformation where the DNA-binding domains of NF-κB are too far apart to bind DNA, whereas a loss-of-function IκBα mutant retains the NF-κB conformational ensemble. Overall, our results suggest a novel mechanism with a continuum of binding modes for controlling association and dissociation of transcription factors.


Subject(s)
DNA/genetics , Interferons/genetics , NF-KappaB Inhibitor alpha/genetics , Transcription Factor RelA/genetics , Transcription, Genetic , Animals , Avidin/chemistry , Binding Sites , Biotin/chemistry , DNA/metabolism , Fluorescence Resonance Energy Transfer , Gene Expression Regulation , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/genetics , Immobilized Proteins/metabolism , Interferons/chemistry , Interferons/metabolism , Inverted Repeat Sequences , Mice , Molecular Dynamics Simulation , NF-KappaB Inhibitor alpha/chemistry , NF-KappaB Inhibitor alpha/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Single Molecule Imaging/methods , Transcription Factor RelA/chemistry , Transcription Factor RelA/metabolism
19.
Molecules ; 26(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34684775

ABSTRACT

Carbohydrate-protein conjugates have diverse applications. They have been used clinically as vaccines against bacterial infection and have been developed for high-throughput assays to elucidate the ligand specificities of glycan-binding proteins (GBPs) and antibodies. Here, we report an effective process that combines highly efficient chemoenzymatic synthesis of carbohydrates, production of carbohydrate-bovine serum albumin (glycan-BSA) conjugates using a squarate linker, and convenient immobilization of the resulting neoglycoproteins on carboxylate-coated fluorescent magnetic beads for the development of a suspension multiplex array platform. A glycan-BSA-bead array containing BSA and 50 glycan-BSA conjugates with tuned glycan valency was generated. The binding profiles of six plant lectins with binding preference towards Gal and/or GalNAc, as well as human galectin-3 and galectin-8, were readily obtained. Our results provide useful information to understand the multivalent glycan-binding properties of human galectins. The neoglycoprotein-immobilized fluorescent magnetic bead suspension multiplex array is a robust and flexible platform for rapid analysis of glycan and GBP interactions and will find broad applications.


Subject(s)
Galectins/metabolism , Protein Array Analysis/methods , Blood Proteins/chemistry , Blood Proteins/metabolism , Carbohydrate Conformation , Carbohydrate Sequence , Fluorescent Dyes , Galectins/chemistry , Glycation End Products, Advanced , Glycoproteins , Humans , Immobilized Proteins , Magnetic Phenomena , Plant Lectins/chemistry , Plant Lectins/metabolism , Polysaccharides/chemistry , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serum Albumin , Serum Albumin, Bovine , Glycated Serum Albumin
20.
ACS Appl Mater Interfaces ; 13(39): 46406-46420, 2021 Oct 06.
Article in English | MEDLINE | ID: mdl-34569225

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease associated with amyloid-ß (Aß) deposition, leading to neurotoxicity (oxidative stress and neuroinflammation) and gut microbiota imbalance. Resveratrol (Res) has neuroprotective properties, but its bioavailability in vivo is very low. Herein, we developed a small Res-selenium-peptide nanocomposite to enable the application of Res for eliminating Aß aggregate-induced neurotoxicity and mitigating gut microbiota disorder in aluminum chloride (AlCl3) and d-galactose(d-gal)-induced AD model mice. Res functional selenium nanoparticles (Res@SeNPs) (8 ± 0.34 nm) were prepared first, after which the surface of Res@SeNPs was decorated with a blood-brain barrier transport peptide (TGN peptide) to generate Res-selenium-peptide nanocomposites (TGN-Res@SeNPs) (14 ± 0.12 nm). Oral administration of TGN-Res@SeNPs improves cognitive disorder through (1) interacting with Aß and decreasing Aß aggregation, effectively inhibiting Aß deposition in the hippocampus; (2) decreasing Aß-induced reactive oxygen species (ROS) and increasing activity of antioxidation enzymes in PC12 cells and in vivo; (3) down-regulating Aß-induced neuroinflammation via the nuclear factor kappa B/mitogen-activated protein kinase/Akt signal pathway in BV-2 cells and in vivo; and (4) alleviating gut microbiota disorder, particularly with respect to oxidative stress and inflammatory-related bacteria such as Alistipes, Helicobacter, Rikenella, Desulfovibrio, and Faecalibaculum. Thus, we anticipate that Res-selenium-peptide nanocomposites will offer a new potential strategy for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Drug Carriers/chemistry , Nanocomposites/chemistry , Neuroprotective Agents/therapeutic use , Resveratrol/therapeutic use , Administration, Oral , Aluminum Chloride , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/metabolism , Animals , Bacteria/drug effects , Drug Carriers/administration & dosage , Drug Carriers/toxicity , Galactose , Gastrointestinal Microbiome/drug effects , Immobilized Proteins/administration & dosage , Immobilized Proteins/chemistry , Immobilized Proteins/toxicity , Male , Memory/drug effects , Mice, Inbred ICR , Multifunctional Nanoparticles/administration & dosage , Multifunctional Nanoparticles/chemistry , Multifunctional Nanoparticles/toxicity , Nanocomposites/administration & dosage , Nanocomposites/toxicity , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Oxidative Stress/drug effects , PC12 Cells , Peptide Fragments/metabolism , Peptides/administration & dosage , Peptides/chemistry , Peptides/toxicity , Protein Multimerization/drug effects , Rats , Resveratrol/administration & dosage , Resveratrol/chemistry , Selenium/administration & dosage , Selenium/chemistry , Selenium/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...