Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pept Sci ; 29(4): e3460, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36285908

RESUMO

Semaphorin-3A (Sema-3A) is a chemorepellant protein with various biological functions, including kidney development. It interacts with a protein complex consisting of the receptors neuropilin-1 (NRP-1) and plexin-A1. After acute kidney injury, Sema-3A is overexpressed and secreted, leading to a loss of kidney function. The development of peptide inhibitors is a promising approach to modulate the interaction of Sema-3A with its receptor NRP-1. Few interaction points between these binding partners are known. However, an immunoglobulin-like domain-derived peptide of Sema-3A has shown a positive effect on cell proliferation. To specify these interactions between the peptide inhibitor and the Sema-3A-NRP-1 system, the peptides were modified with the photoactivatable amino acids 4-benzoyl-l-phenylalanine or photo-l-leucine by solid-phase peptide synthesis. Activity was tested by an enzyme-linked immunosorbent-based binding assay, and crosslinking experiments were analyzed by Western blot and mass spectrometry, demonstrating a specific binding site of the peptide at Sema-3A. The observed signals for Sema-3A-peptide interaction were found in a defined area of the Sema domain, which was also demonstrated to be involved in NRP-1 binding. The presented data identified the interaction site for further development of therapeutic peptides to treat acute kidney injury by blocking the Sema-3A-NRP-1 interaction.


Assuntos
Injúria Renal Aguda , Semaforina-3A , Humanos , Semaforina-3A/metabolismo , Peptídeos , Neuropilina-1
2.
ACS Cent Sci ; 7(2): 345-354, 2021 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-33655072

RESUMO

The maintenance of therapeutic glycoproteins within the circulatory system is associated, in large part, with the integrity of sialic acids as terminal sugars on the glycans. Glycoprotein desialylation, either by spontaneous cleavage or through host sialidases, leads to protein clearance, mainly through the liver. Thus, the installation of minimally modified sialic acids that are hydrolysis-resistant yet biologically equivalent should lead to increased circulatory half-lives and improved pharmacokinetic profiles. Here we describe the chemoenzymatic synthesis of CMP-sialic acid sugar donors bearing fluorine atoms at the 7-position, starting from the corresponding 4-deoxy-4-fluoro-N-acetylhexosamine precursors. For the derivative with natural stereochemistry we observe efficient glycosyl transfer by sialyltransferases, along with improved stability of the resultant 7-fluorosialosides toward spontaneous hydrolysis (3- to 5-fold) and toward cleavage by GH33 sialidases (40- to 250-fold). Taking advantage of the rapid transfer of 7-fluorosialic acid by sialyltransferases, we engineered the O-glycan of Interferon α-2b and the N-glycans of the therapeutic glycoprotein α1-antitrypsin. Studies of the uptake of the glyco-engineered α1-antitrypsin by HepG2 liver cells demonstrated the bioequivalence of 7-fluorosialic acid to sialic acid in suppressing interaction with liver cell lectins. In vivo pharmacokinetic studies reveal enhanced half-life of the protein decorated with 7-fluorosialic acid relative to unmodified sialic acid in the murine circulatory system. 7-Fluorosialylation therefore offers considerable promise as a means of prolonging circulatory half-lives of glycoproteins and may pave the way toward biobetters for therapeutic use.

3.
Glycobiology ; 29(10): 715-725, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31264681

RESUMO

CXCL14, chemokine (C-X-C motif) ligand 14, is a novel highly conserved chemokine with unique features. Despite exhibiting the typical chemokine fold, it has a very short N-terminus of just two amino acid residues responsible for chemokine receptor activation. CXCL14 actively participates in homeostatic immune surveillance of skin and mucosae, is linked to metabolic disorders and fibrotic lung diseases and possesses strong anti-angiogenic properties in early tumor development. In this work, we investigated the interaction of CXCL14 with various glycosaminoglycans (GAGs) by nuclear magnetic resonance spectroscopy, microscale thermophoresis, analytical heparin (HE) affinity chromatography and in silico approaches to understand the molecular basis of GAG-binding. We observed different GAG-binding modes specific for the GAG type used in the study. In particular, the CXCL14 epitope for HE suggests a binding pose distinguishable from the ones of the other GAGs investigated (hyaluronic acid, chondroitin sulfate-A/C, -D, dermatan sulfate). This observation is also supported by computational methods that included molecular docking, molecular dynamics and free energy calculations. Based on our results, we suggest that distinct GAG sulfation patterns confer specificity beyond simple electrostatic interactions usually considered to represent the driving forces in protein-GAG interactions. The CXCL14-GAG system represents a promising approach to investigate the specificity of GAG-protein interactions, which represents an important topic for developing the rational approaches to novel strategies in regenerative medicine.


Assuntos
Quimiocinas CXC/metabolismo , Epitopos/genética , Glicosaminoglicanos/metabolismo , Heparina/metabolismo , Sítios de Ligação/genética , Quimiocinas CXC/química , Quimiocinas CXC/genética , Sulfatos de Condroitina/química , Sulfatos de Condroitina/genética , Dermatan Sulfato/química , Dermatan Sulfato/genética , Epitopos/química , Glicosaminoglicanos/química , Glicosaminoglicanos/genética , Heparina/genética , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/genética , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica/genética , Dobramento de Proteína
4.
Glycobiology ; 29(7): 588-598, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30976781

RESUMO

Polysialyltransferases (polySTs) are glycosyltransferases that synthesize polymers of sialic acid found in vertebrates and some bacterial pathogens. Bacterial polySTs have utility in the modification of therapeutic proteins to improve serum half-life, and the potential for tissue engineering. PolySTs are membrane-associated proteins and as recombinant proteins suffer from inherently low solubility, low expression levels and poor thermal stability. To improve their physicochemical and biochemical properties, we applied a directed evolution approach using a FACS-based ultrahigh-throughput assay as a simple, robust and reliable screening method. We were able to enrich a large mutant library and, in combination with plate-based high-throughput secondary screening, we discovered mutants with increased enzymatic activity and improved stability compared to the wildtype enzyme. This work presents a powerful strategy for the screening of directed evolution libraries of bacterial polySTs to identify better catalysts for in vitro polysialylation of therapeutics.


Assuntos
Evolução Molecular Direcionada , Sialiltransferases/genética , Sialiltransferases/metabolismo , Biocatálise , Estabilidade Enzimática , Escherichia coli/enzimologia , Escherichia coli/genética , Biblioteca Gênica , Ensaios de Triagem em Larga Escala , Mutação , Sialiltransferases/química , Solubilidade
5.
Biopolymers ; 109(10): e23103, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29377072

RESUMO

The chemokine interleukin-8 (IL-8, CXCL8) plays an important role in inflammatory processes and consecutive wound healing. It recruits primarily neutrophils to infection sites and stimulates their degranulation and phagocytosis in effector cells. IL-8 binds glycosaminoglycans (GAGs), a class of complex linear anionic polysaccharides often organized into diversely sulfated micro-domains, that enriches the protein concentration locally and so facilitate the formation of stable concentration gradients. In this study, we applied experimental and computational techniques to investigate the binding of wild type and truncated IL-8 variants to natural and chemically modified GAGs to gain further insight into the IL-8/GAG interaction. Circular dichroism spectroscopy of IL-8 variants did not reveal major structural changes upon GAG binding. Heparin affinity chromatography clearly demonstrates that gradual truncation of the C-terminal helix leads to decreasing affinities. Similarly, surface plasmon resonance indicates participation of both IL-8 termini in GAG binding, which strength is dependent on GAG sulfation degree. Molecular modeling suggests that C-terminal truncation of IL-8 weakens the interaction with GAGs by an alteration of IL-8 GAG binding site. Our study provides more detailed understanding of the IL-8/GAG interaction and contributes to the data of potential use for the development of biomedical implications in tissue regeneration.


Assuntos
Glicosaminoglicanos/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Mutação/genética , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Heparina/química , Interleucina-8/química , Ligação Proteica , Receptores de Interleucina-8A , Regeneração , Termodinâmica
6.
Sci Rep ; 7(1): 5842, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28724897

RESUMO

Polysialic acid (polySia) is a homopolymeric saccharide that is associated with some neuroinvasive pathogens and is found on selective cell types in their eukaryotic host. The presence of a polySia capsule on these bacterial pathogens helps with resistance to phagocytosis, cationic microbial peptides and bactericidal antibody production. The biosynthesis of bacterial polySia is catalysed by a single polysialyltransferase (PST) transferring sialic acid from a nucleotide-activated donor to a lipid-linked acceptor oligosaccharide. Here we present the X-ray structure of the bacterial PST from Mannheimia haemolytica serotype A2, thereby defining the architecture of this class of enzymes representing the GT38 family. The structure reveals a prominent electropositive groove between the two Rossmann-like domains forming the GT-B fold that is suitable for binding of polySia chain products. Complex structures of PST with a sugar donor analogue and an acceptor mimetic combined with kinetic studies of PST active site mutants provide insight into the principles of substrate binding and catalysis. Our results are the basis for a molecular understanding of polySia biosynthesis in bacteria and might assist the production of polysialylated therapeutic reagents and the development of novel antibiotics.


Assuntos
Cápsulas Bacterianas/metabolismo , Mannheimia haemolytica/enzimologia , Ácidos Siálicos/biossíntese , Sialiltransferases/química , Sítios de Ligação , Biocatálise , Cristalografia por Raios X , Fondaparinux , Cinética , Nucleotídeos/metabolismo , Domínios Proteicos , Ácidos Siálicos/química , Açúcares/metabolismo
7.
PLoS Pathog ; 12(12): e1006067, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27973583

RESUMO

In recent years, there has been a growing interest in teichoic acids as targets for antibiotic drug design against major clinical pathogens such as Staphylococcus aureus, reflecting the disquieting increase in antibiotic resistance and the historical success of bacterial cell wall components as drug targets. It is now becoming clear that ß-O-GlcNAcylation of S. aureus wall teichoic acids plays a major role in both pathogenicity and antibiotic resistance. Here we present the first structure of S. aureus TarS, the enzyme responsible for polyribitol phosphate ß-O-GlcNAcylation. Using a divide and conquer strategy, we obtained crystal structures of various TarS constructs, mapping high resolution overlapping N-terminal and C-terminal structures onto a lower resolution full-length structure that resulted in a high resolution view of the entire enzyme. Using the N-terminal structure that encapsulates the catalytic domain, we furthermore captured several snapshots of TarS, including the native structure, the UDP-GlcNAc donor complex, and the UDP product complex. These structures along with structure-guided mutants allowed us to elucidate various catalytic features and identify key active site residues and catalytic loop rearrangements that provide a valuable platform for anti-MRSA drug design. We furthermore observed for the first time the presence of a trimerization domain composed of stacked carbohydrate binding modules, commonly observed in starch active enzymes, but adapted here for a poly sugar-phosphate glycosyltransferase.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Glicosiltransferases/química , Glicosiltransferases/metabolismo , Staphylococcus aureus Resistente à Meticilina/metabolismo , Parede Celular , Cromatografia Líquida , Cristalografia por Raios X , Espectrometria de Massas , Resistência a Meticilina/fisiologia , Staphylococcus aureus Resistente à Meticilina/química , Estabilidade Proteica , Estrutura Quaternária de Proteína , Ácidos Teicoicos/metabolismo
9.
Glycobiology ; 26(11): 1209-1221, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27496764

RESUMO

The stromal cell-derived factor 1α (CXCL12) belongs to the CXC chemokine family and plays an important role in tissue regeneration and the recruitment of stem cells. Here, a stable chemotactic gradient is essential that is formed by the interaction of CXCL12 with the extracellular matrix. Binding properties of CXCL12 to naturally occurring glycosaminoglycans (GAGs) as well as to the artificial highly sulfated hyaluronic acid (HA) are investigated by using a combination of NMR spectroscopy, molecular modeling and molecular dynamics simulations. Our results demonstrate a preferred protein binding for the sulfated GAGs heparin (HE) and highly sulfated HA. Furthermore, we could demonstrate that the orientation of the sulfate is crucial for binding. All sulfated GAGs interact with the CXCL12 GAG-binding motif (K24-H25-L26-K27-R41-K43-R47), where K27 and R41 represent the anchor points. Furthermore, differences could be observed in the second interaction interface of CXCL12: both HE and highly sulfated HA interfere with the receptor-binding motif, while chondroitin sulfate binds different amino acids in close proximity to this motif. CXCL12 does not interact with HA, which was directly demonstrated by NMR spectroscopy and molecular modeling and explained by the lack of sulfate groups of the HA molecule.


Assuntos
Quimiocina CXCL12/química , Glicosaminoglicanos/química , Sítios de Ligação , Configuração de Carboidratos , Humanos , Modelos Moleculares
10.
J Control Release ; 224: 59-68, 2016 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-26763375

RESUMO

Cell fate decisions in many physiological processes, including embryogenesis, stem cell niche homeostasis and wound healing, are regulated by secretion of small signaling proteins, called cytokines, from source cells to their neighbors or into the environment. Concentration level and steepness of the resulting paracrine gradients elicit different cell responses, including proliferation, differentiation or chemotaxis. For an in-depth analysis of underlying mechanisms, in vitro models are required to mimic in vivo cytokine gradients. We set up a microparticle-based system to establish short-range cytokine gradients in a three-dimensional extracellular matrix context. To provide native binding sites for cytokines, agarose microparticles were functionalized with different glycosaminoglycans (GAG). After protein was loaded onto microparticles, its slow release was quantified by confocal microscopy and fluorescence correlation spectroscopy. Besides the model protein lysozyme, SDF-1 was used as a relevant chemokine for hematopoietic stem and progenitor cell (HSPC) chemotaxis. For both proteins we found gradients ranging up to 50µm from the microparticle surface and concentrations in the order of nM to pM in dependence on loading concentration and affinity modulation by the GAG functionalization. Directed chemotactic migration of cells from a hematopoietic cell line (FDCPmix) and primary murine HSPC (Sca-1(+) CD150(+) CD48(-)) toward the SDF-1-laden microparticles proved functional short-range gradients in a two-dimensional and three-dimensional setting over time periods of many hours. The approach has the potential to be applied to other cytokines mimicking paracrine cell-cell interactions in vitro.


Assuntos
Citocinas/metabolismo , Comunicação Parácrina , Algoritmos , Animais , Comunicação Celular , Linhagem Celular , Quimiocina CXCL12/administração & dosagem , Quimiotaxia/efeitos dos fármacos , Matriz Extracelular/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Muramidase/análise , Nanopartículas , Células-Tronco/efeitos dos fármacos
11.
FEBS Open Bio ; 5: 579-93, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26258013

RESUMO

In the search for effective therapeutic strategies, protein-based biologicals are under intense development. While monoclonal antibodies represent the majority of these drugs, other innovative approaches are exploring the use of scaffold proteins for the creation of binding molecules with tailor-made properties. Ubiquitin is especially suited for this strategy due to several key characteristics. Ubiquitin is a natural serum protein, 100% conserved across the mammalian class and possesses high thermal, structural and proteolytic stability. Because of its small size and lack of posttranslational modifications, it can be easily produced in Escherichia coli. In this work we provide evidence that ubiquitin is safe as tested experimentally in vivo. In contrast to previously published results, we show that, in our hands, ubiquitin does not act as a functional ligand of the chemokine receptor CXCR4. Cellular assays based on different signaling pathways of the receptor were conducted with the natural agonist SDF-1 as a benchmark. In none of the assays could a response to ubiquitin treatment be elicited. Furthermore, intravenous application to mice at high concentrations did not induce any detectable effect on cytokine levels or hematological parameters.

12.
Nat Struct Mol Biol ; 22(8): 627-35, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26192331

RESUMO

Sialyltransferases of the mammalian ST8Sia family catalyze oligo- and polysialylation of surface-localized glycoproteins and glycolipids through transfer of sialic acids from CMP-sialic acid to the nonreducing ends of sialic acid acceptors. The crystal structure of human ST8SiaIII at 1.85-Å resolution presented here is, to our knowledge, the first solved structure of a polysialyltransferase from any species, and it reveals a cluster of polysialyltransferase-specific structural motifs that collectively provide an extended electropositive surface groove for binding of oligo-polysialic acid chain products. The ternary complex of ST8SiaIII with a donor sugar analog and a sulfated glycan acceptor identified with a sialyltransferase glycan array provides insight into the residues involved in substrate binding, specificity and sialyl transfer.


Assuntos
Estrutura Terciária de Proteína , Ácidos Siálicos/metabolismo , Sialiltransferases/química , Sialiltransferases/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Células Cultivadas , Cromatografia em Camada Fina , Cristalografia por Raios X , Eletroforese em Gel de Poliacrilamida , Glicosilação , Humanos , Cinética , Espectrometria de Massas/métodos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Moléculas de Adesão de Célula Nervosa/química , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Polissacarídeos/química , Polissacarídeos/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Ácidos Siálicos/química , Sialiltransferases/genética
13.
Proc Natl Acad Sci U S A ; 112(6): E576-85, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25624472

RESUMO

Unique to Gram-positive bacteria, wall teichoic acids are anionic glycopolymers cross-stitched to a thick layer of peptidoglycan. The polyol phosphate subunits of these glycopolymers are decorated with GlcNAc sugars that are involved in phage binding, genetic exchange, host antibody response, resistance, and virulence. The search for the enzymes responsible for GlcNAcylation in Staphylococcus aureus has recently identified TarM and TarS with respective α- and ß-(1-4) glycosyltransferase activities. The stereochemistry of the GlcNAc attachment is important in balancing biological processes, such that the interplay of TarM and TarS is likely important for bacterial pathogenicity and survival. Here we present the crystal structure of TarM in an unusual ternary-like complex consisting of a polymeric acceptor substrate analog, UDP from a hydrolyzed donor, and an α-glyceryl-GlcNAc product formed in situ. These structures support an internal nucleophilic substitution-like mechanism, lend new mechanistic insight into the glycosylation of glycopolymers, and reveal a trimerization domain with a likely role in acceptor substrate scaffolding.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Parede Celular/enzimologia , Glicosiltransferases/metabolismo , Modelos Moleculares , Staphylococcus aureus/enzimologia , Ácidos Teicoicos/metabolismo , Proteínas de Bactérias/genética , Clonagem Molecular , Cristalização , Estabilidade Enzimática , Glicosiltransferases/química , Glicosiltransferases/genética , Espectrometria de Massas , Metais/análise , Ressonância Magnética Nuclear Biomolecular , Polimerização , Conformação Proteica
14.
ACS Appl Mater Interfaces ; 6(8): 5891-9, 2014 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24646074

RESUMO

Preparation of smart materials by coatings of established surfaces with biomolecules will lead to the next generation of functionalized biomaterials. Rejection of implants is still a major problem in medical applications but masking the implant material with protein coatings is a promising approach. These layers not only disguise the material but also equip it with a certain biological function. The anti-inflammatory chemokine stromal cell-derived factor 1α (SDF-1α) is well suited to take over this function, because it efficiently attracts stem cells and promotes their differentiation and proliferation. At least the initial stem cell homing requires the formation of a concentration gradient. Thus, a reliable and robust release mechanism of SDF-1α from the material is essential. Several proteases, most notably matrix metalloproteinases, are upregulated during inflammation, which, in principle, can be exploited for a tightly controlled release of SDF-1α. Herein, we present the covalent immobilization of M-[S4V]-SDF-1α on novel biodegradable polymer films, which consist of heterobifunctional poly(ethylene glycol) and oligolactide-based functionalized macromers. A peptidic linker with a trimeric matrix metalloproteinase 9 (MMP-9) cleavage site (MCS) was used as connection and the linkage between the three components was achieved by combination of expressed protein ligation and Cu(I) catalyzed azide/alkyne cycloaddition. The MCS was used for MMP-9 mediated release of M-[S4V]-SDF-1α from the biomaterial and the released SDF-1α derivative was biologically active and induced strong cell migration, which demonstrates the great potential of this system.


Assuntos
Quimiocina CXCL12/química , Materiais Revestidos Biocompatíveis/química , Metaloproteinase 9 da Matriz/metabolismo , Polímeros/química , Linhagem Celular , Movimento Celular , Quimiocina CXCL12/metabolismo , Materiais Revestidos Biocompatíveis/metabolismo , Humanos , Metaloproteinase 9 da Matriz/química , Células Estromais/citologia , Células Estromais/metabolismo
15.
Methods Mol Biol ; 1047: 103-18, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23943481

RESUMO

In order to link structural features on a molecular level to the function of chemokines, site-specific modification strategies are strongly required. These can be used to incorporate fluorescent dyes and/or physical probes to allow investigations in a wide range of biological and physical techniques, e.g., nuclear magnetic resonance (NMR) spectroscopy, fluorescence microscopy, fluorescence resonance energy transfer (FRET), or fluorescence correlation spectroscopy (FCS). Only a limited number of functional groups within the 20 canonical amino acids allow ligation strategies that can be helpful to introduce novel functionalities, which in turn expand the scope of chemoselective and orthogonal reactivity of (semi)synthetic chemokines. In the present chapter we mainly focus on the fabulous history of native chemical ligation (NCL) and provide a general protocol for the preparation of C-terminally modified SDF-1α including tips and tricks for practical work. We believe that this protocol can be easily adapted to other chemokines and many proteins in general.


Assuntos
Quimiocinas/química , Quimiocinas/genética , Expressão Gênica , Quimiocinas/metabolismo , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Fragmentos de Peptídeos/síntese química , Redobramento de Proteína , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Técnicas de Síntese em Fase Sólida/métodos
17.
J Control Release ; 162(1): 68-75, 2012 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-22634073

RESUMO

The CXC chemokine stromal cell-derived factor-1α (SDF-1α, CXCL12) has been proven to recruit CXCR4 positive stem and progenitor cells of different sources to defected heart sites, with significant clinical benefits. However, the rapid proteolytic inactivation by inflammation-related proteases, inaccurate drug delivery or inappropriate local concentrations belong to the largest disadvantages for feasible application. Herein, we present a switchable, biased-like SDF-1α variant, AAV-[S4V]-SDF-1α, whose distinct activity is coupled to the inflammation-associated presence of dipeptidylpeptidase-4 (DPP-4), which cleaves an alanine-alanine dipeptide from the precursor. We decorated starPEG-heparin hydrogels with our novel SDF-1α variant and tested them for immobilization efficiency, time-dependent protein release as well as mobilization of early endothelial progenitor cells (eEPCs) in vitro. We found higher migration rates compared to conventional SDF-1α. In summary, we provide a conceptual work on cooperative effects of enzymatically activatable SDF-1α and starPEG-heparin hydrogels.


Assuntos
Quimiocina CXCL12/administração & dosagem , Quimiocina CXCL12/farmacologia , Células Endoteliais/efeitos dos fármacos , Heparina/química , Hidrogéis/química , Polietilenoglicóis/química , Células-Tronco/efeitos dos fármacos , Animais , Células COS , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CXCL12/metabolismo , Chlorocebus aethiops , Cromatografia de Afinidade , Dipeptidil Peptidase 4/metabolismo , Células Endoteliais/citologia , Heparina/metabolismo , Humanos , Hidrogéis/metabolismo , Células Jurkat , Metaloproteinase 9 da Matriz/metabolismo , Elastase Pancreática/metabolismo , Polietilenoglicóis/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Células-Tronco/citologia , Alicerces Teciduais/química
18.
Glycobiology ; 22(1): 134-45, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21873605

RESUMO

The interactions between glycosaminoglycans (GAGs), important components of the extracellular matrix, and proteins such as growth factors and chemokines play critical roles in cellular regulation processes. Therefore, the design of GAG derivatives for the development of innovative materials with bio-like properties in terms of their interaction with regulatory proteins is of great interest for tissue engineering and regenerative medicine. Previous work on the chemokine interleukin-8 (IL-8) has focused on its interaction with heparin and heparan sulfate, which regulate chemokine function. However, the extracellular matrix contains other GAGs, such as hyaluronic acid (HA), dermatan sulfate (DS) and chondroitin sulfate (CS), which have so far not been characterized in terms of their distinct molecular recognition properties towards IL-8 in relation to their length and sulfation patterns. NMR and molecular modeling have been in great part the methods of choice to study the structural and recognition properties of GAGs and their protein complexes. However, separately these methods have challenges to cope with the high degree of similarity and flexibility that GAGs exhibit. In this work, we combine fluorescence spectroscopy, NMR experiments, docking and molecular dynamics simulations to study the configurational and recognition properties of IL-8 towards a series of HA and CS derivatives and DS. We analyze the effects of GAG length and sulfation patterns in binding strength and specificity, and the influence of GAG binding on IL-8 dimer formation. Our results highlight the importance of combining experimental and theoretical approaches to obtain a better understanding of the molecular recognition properties of GAG-protein systems.


Assuntos
Sulfatos de Condroitina/química , Dermatan Sulfato/química , Ácido Hialurônico/química , Interleucina-8/química , Simulação de Dinâmica Molecular , Motivos de Aminoácidos , Sítios de Ligação , Configuração de Carboidratos , Humanos , Ligação de Hidrogênio , Espectroscopia de Ressonância Magnética , Ligação Proteica , Multimerização Proteica , Espectrometria de Fluorescência , Termodinâmica , Titulometria
19.
J Pept Sci ; 16(10): 568-74, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20862724

RESUMO

SDF1α plays an important role in the regeneration of injured tissue after ischemia or stroke by inducing the migration of progenitor cells. In order to study the function of this therapeutically relevant chemokine site-specific protein labelling is of great interest. However, modification of SDF1α is complicated because of its complex tertiary structure. Here, we describe the first site-specific fluorescent modification of SDF1α by EPL. We recombinantly expressed SDF1α (1-49) by intein-mediated protein expression. The C-terminal peptide SDF1α (50-68) was synthesised by SPPS and selectively labelled with carboxyfluorescein at Lys(56). In a cell migration assay, M-[K(56)(CF)]SDF1α showed a clear potency to induce chemotaxis of human T-cell leukaemia cells. Microscopic analysis on HEK293 cells transfected with the CXCR4 revealed specific binding of the fluorescent ligand. Furthermore, receptor-induced internalisation of the ligand could be visualised. These results show that site-specific modification of SDF1α yields in a biologically functional molecule that allows the characterisation of CXCR4 production of cells on a molecular level.


Assuntos
Quimiocina CXCL12 , Endocitose/fisiologia , Fluoresceínas/química , Corantes Fluorescentes/química , Sequência de Aminoácidos , Linhagem Celular , Movimento Celular , Quimiocina CXCL12/química , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Humanos , Dados de Sequência Molecular , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
20.
Biopolymers ; 94(6): 771-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20564039

RESUMO

Selective modification of proteins is an important tool to study their function. However, it is still challenging to identify the best position to avoid a loss of activity. By using a 6-nitroveratryl (Nvoc)-modification approach, we facilitate the identification of a potential modification site as Nvoc can be removed in situ by UV irradiation and accordingly allows directly the comparison of the biological activity of the modified and the unmodified protein derived from the same precursor. As a test system, we used stromal cell-derived factor-1 (SDF-1), which is involved in a wide range of physiological functions, mainly hematopoiesis and embryonic organ development. This chemokine is a potential candidate in regenerative medicine because of its capability to attract stem cells to distinct localizations. First, we synthesized the wildtype and the Nvoc-modified C-terminal segments SDF-1(50-68) and studied their secondary structure formation by circular dichroism spectroscopy. By using the intein-mediated purification with a affinity chitin binding tag system, we then expressed the peptide thioester M-[A(49)]-SDF-1(1-49)-MESNA recombinantly, in which the valine at position 49 was replaced by a more suitable alanine residue to allow improved cleavage and ligation. After ligation and refolding, the biological activity was proven in a cell-based inositol phosphate accumulation assay prior and after Nvoc removal, which showed that neither the alanine 49 nor the attached Nvoc group impair the activity of the analog. The study shows that lysine 56 is a potential site to introduce labels site-specifically in SDF-1.


Assuntos
Quimiocina CXCL12/química , Mapeamento de Peptídeos , Peptídeos/química , Animais , Benzaldeídos/química , Células COS , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Chlorocebus aethiops , Humanos , Peptídeos/farmacologia , Medicina Regenerativa/métodos , Células-Tronco/metabolismo , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...