Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Protein Expr Purif ; 122: 64-71, 2016 06.
Article in English | MEDLINE | ID: mdl-26923061

ABSTRACT

Matrix metalloproteinases (MMPs) are crucial proteases in maintaining the health and integrity of many tissues, however their dysregulation often facilitates disease progression. In disease states these remodeling and repair functions support, for example, metastasis of cancer by both loosening the matrix around tumors to enable cellular invasion and by affecting proliferation and apoptosis, and they promote degradation of biological restorations by weakening the substrate to which the restoration is attached. As such, MMPs are important therapeutic targets. MMP-8 participates in cancer, arthritis, asthma and failure of dental fillings. MMP-8 differs from other MMPs in that it has an insertion that enlarges its active site. To elucidate the unique features of MMP-8 and develop selective inhibitors to this therapeutic target, a stable and active form of the enzyme is needed. MMP-8 has been difficult to express at high yield in a soluble, active form. Typically recombinant MMPs accumulate in inclusion bodies and complex methods are applied to refold and purify protein in acceptable yield. Presented here is a streamlined approach to produce in Escherichia coli a soluble, active, stable MMP-8 fusion protein in high yield. This fusion shows much greater retention of activity when stored refrigerated without glycerol. A variant of this construct that contains the metal binding claMP Tag was also examined to demonstrate the ability to use this tag with a metalloprotein. SDS-PAGE, densitometry, mass spectrometry, circular dichroism spectroscopy and an activity assay were used to analyze the chemical integrity and function of the enzyme.


Subject(s)
Escherichia coli/genetics , Matrix Metalloproteinase 8/genetics , Catalytic Domain , Cloning, Molecular/methods , Genetic Engineering , Genetic Vectors/genetics , Humans , Matrix Metalloproteinase 8/chemistry , Matrix Metalloproteinase 8/isolation & purification , Matrix Metalloproteinase 8/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Solubility , Thioredoxins/chemistry , Thioredoxins/genetics , Thioredoxins/isolation & purification , Thioredoxins/metabolism
2.
J Dent Res ; 93(12): 1243-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25190266

ABSTRACT

In the United States, composites accounted for nearly 70% of the 173.2 million composite and amalgam restorations placed in 2006 (Kingman et al., 2012), and it is likely that the use of composite will continue to increase as dentists phase out dental amalgam. This trend is not, however, without consequences. The failure rate of composite restorations is double that of amalgam (Ferracane, 2013). Composite restorations accumulate more biofilm, experience more secondary decay, and require more frequent replacement. In vivo biodegradation of the adhesive bond at the composite-tooth interface is a major contributor to the cascade of events leading to restoration failure. Binding by proteins, particularly gp340, from the salivary pellicle leads to biofilm attachment, which accelerates degradation of the interfacial bond and demineralization of the tooth by recruiting the pioneer bacterium Streptococcus mutans to the surface. Bacterial production of lactic acid lowers the pH of the oral microenvironment, erodes hydroxyapatite in enamel and dentin, and promotes hydrolysis of the adhesive. Secreted esterases further hydrolyze the adhesive polymer, exposing the soft underlying collagenous dentinal matrix and allowing further infiltration by the pathogenic biofilm. Manifold approaches are being pursued to increase the longevity of composite dental restorations based on the major contributing factors responsible for degradation. The key material and biological components and the interactions involved in the destructive processes, including recent advances in understanding the structural and molecular basis of biofilm recruitment, are described in this review. Innovative strategies to mitigate these pathogenic effects and slow deterioration are discussed.


Subject(s)
Biofilms , Composite Resins/chemistry , Dental Materials/chemistry , Dental Restoration Failure , Salivary Proteins and Peptides/pharmacokinetics , Streptococcus mutans/physiology , Tooth/microbiology , Bacterial Adhesion/physiology , Dental Bonding , Dental Pellicle/microbiology , Humans , Tooth/metabolism , Tooth Demineralization/microbiology
3.
J Dent Res ; 90(12): 1434-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21960682

ABSTRACT

When adhesives and/or composites are bonded to the tooth, water in the environment can interfere with proper interface formation. Formation of water blisters and phase separation at the adhesive/dentin interface have appeared as new types of bond defects. To better understand this problem, we determined the near-equilibrium partition of the hydrophobic/hydrophilic components when exposed to over-wet environments. Model methacrylate-based adhesives were mixed with different amounts of water to yield well-separated aqueous and resin phases. It was found that less than 0.1% BisGMA but nearly one-third of the HEMA diffused into the aqueous phase, leaving the remaining resin phase relatively hydrophobic. A partial phase diagram was created for the ternary BisGMA/HEMA/water system. All the experimental phase partitioning data were plotted, and the points lay on a binodal curve that separated the single-phase region from the two-phase region. We obtained the 3 tie lines by connecting the 2 points of each conjugate pair of the phase partitioning data from the 3 sets of tripartite mixtures. Information about solubility, water miscibility, distribution ratio, and phase partitioning behavior could be obtained quantitatively. This type of phase diagram will provide a more thorough understanding of current adhesive performance and elucidate directions for further improvement.


Subject(s)
Dentin-Bonding Agents/chemistry , Models, Chemical , Bisphenol A-Glycidyl Methacrylate/analysis , Hydrophobic and Hydrophilic Interactions , Methacrylates/analysis , Solubility , Water/analysis
4.
Biochemistry ; 40(36): 10782-91, 2001 Sep 11.
Article in English | MEDLINE | ID: mdl-11535053

ABSTRACT

MIP-1beta, a member of the chemokine family of proteins, tightly binds the receptor CCR5 as part of its natural function in the immune response, and in doing so also blocks the ability of many strains of HIV to enter the cell. The single most important MIP-1beta residue known to contribute to its interaction with the receptor is Phe13, which when mutated reduces the ability of MIP-1beta to bind to CCR5 by more than 1000-fold. To obtain a structural understanding of the dramatic effect of the absence of Phe13 in MIP-1beta, we used multidimensional heteronuclear NMR to determine the three-dimensional structure of the MIP-1beta F13A variant. We had previously shown that, unlike the wild-type protein which has been shown to be a tight dimer, the F13A mutant is monomeric even at high concentrations [Laurence, J. S., Blanpain, C., Burgner, J. W., Parmentier, M., and LiWang, P. J. (2000) Biochemistry 39, 3401-3409], leading to significant changes in the NMR spectra of F13A and the wild-type protein. We have obtained a total of 940 structural restraints for MIP-1beta F13A, and have calculated a family of structures having a backbone rmsd from the average of 0.55 A (residues 12-67). A structural comparison of the F13A mutant with a fully active monomeric variant, P8A, shows that despite some differences in the (1)H-(15)N HSQC spectra the two are nearly identical in NOE distance restraints and in backbone conformation. A comparison of F13A with the wild-type protein shows largely the same fold, although differences exist in the N-terminal and loop regions for which the loss of the dimer in F13A can mainly account. A dynamics comparison confirms greater flexibility in F13A than in the wild-type protein in regions of dimer contact in the wild-type protein. In an analysis to determine if the large functional effect resulting from the loss of Phe13 is due to the local side chain change or due to more global structural changes, we conclude that local effects predominate. This suggests that a strategy for designing tight binding anti-CCR5 therapeutics should include a Phe-like component.


Subject(s)
Macrophage Inflammatory Proteins/chemistry , Macrophage Inflammatory Proteins/metabolism , Receptors, CCR5/chemistry , Receptors, CCR5/metabolism , Amino Acid Substitution , Binding Sites , Calorimetry , Chemokine CCL4 , Cloning, Molecular , Dimerization , Escherichia coli , Genetic Variation , Humans , Hydrogen Bonding , Macrophage Inflammatory Proteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Structure, Secondary
5.
Biochemistry ; 40(16): 4990-9, 2001 Apr 24.
Article in English | MEDLINE | ID: mdl-11305915

ABSTRACT

Chemokines direct immune cells toward sites of infection by establishing a gradient across the extracellular matrix of the tissue. This gradient is thought to be stabilized by ligation of chemokines to sulfated polysaccharides known as glycosaminoglycans (GAGs) that are found on the surface of endothelial and other cells as well as in the tissue matrix. GAGs interact with chemokines and in some cases cause them to aggregate. The interaction between cell surface GAGs and chemokines has also been postulated to play a role in the anti-HIV activity of some chemokines, including MIP-1beta. Since many proteins interact with GAGs by utilizing basic residues, we mutated R18, K45, R46, and K48 in MIP-1beta to investigate the role of these residues in GAG binding and CCR5 function. We find that no single amino acid substitution alone has a dramatic effect on heparin binding, although change at R46 has a moderate effect. However, binding to heparin is completely abrogated in a mutant (K45A/R46A/K48A) in which the entire "40's loop" has been neutralized. A functional study of these mutants reveals that the charged residues in this 40's loop, particularly K48 and R46, are critical mediators of MIP-1beta binding to its receptor CCR5. However, despite the partially overlapping function of the residues in the 40's loop in binding to both CCR5 and heparin, the presence of cell surface sugars does not appear to be necessary for the ability of MIP-1beta to function on its receptor CCR5, as enzymatic removal of GAGs from cells results in little effect on MIP-1beta activity. Because the means by which the chemokine gradient transmits information to the recruited cells is not well defined, we also mutated the basic residues in MIP(9), a truncated form of MIP-1beta that is impaired in its ability to dimerize, to probe whether the quaternary structure of this chemokine influences its ability to bind heparin. None of the truncated variants bound as well as the full-length proteins containing the same mutation, suggesting that the MIP-1beta dimer participates in heparin binding.


Subject(s)
Amino Acids/chemistry , Glycosaminoglycans/metabolism , Macrophage Inflammatory Proteins/chemistry , Macrophage Inflammatory Proteins/metabolism , Receptors, CCR5/metabolism , Amino Acids/genetics , Animals , Binding, Competitive/genetics , CHO Cells , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Chemokine CCL4 , Chromatography, Agarose , Cricetinae , Heparin/metabolism , Macrophage Inflammatory Proteins/genetics , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Protein Binding/genetics , Protein Structure, Quaternary/genetics , Receptors, CCR5/biosynthesis , Receptors, CCR5/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility
6.
Biochemistry ; 39(12): 3401-9, 2000 Mar 28.
Article in English | MEDLINE | ID: mdl-10727234

ABSTRACT

The reported structures of many CC chemokines show a conserved dimer interface along their N-terminal region, raising the possibility that the quaternary arrangement of these small immune proteins might influence their function. We have produced and analyzed several mutants of MIP-1 beta having a range of dimer K(d) values in order to determine the significance of dimerization in receptor binding and cellular activation. NMR and analytical ultracentrifugation were used to analyze the oligomeric state of the mutants. Functional relevance was determined by receptor binding affinity and the ability to invoke intracellular calcium release from CHO cells transfected with the MIP-1 beta receptor CCR5. The monomeric N-terminally truncated mutant MIP(9) was able to bind the CCR5 receptor with a K(i) of 600 pM but displayed weak agonistic properties, while the monomeric mutant P8A still retained the ability to tightly bind (K(i) = 480 pM) and to activate (EC(50) = 12 nM) the receptor. These data suggest that the MIP-1 beta dimer is not required for CCR5 binding or activation. In addition, we identified Phe13, the residue immediately following the conserved CC motif in MIP-1 beta, as a key determinant for binding to CCR5. Replacement of Phe13 by Tyr, Leu, Lys, and Ala showed the aromatic side chain to be important for both binding to CCR5 and chemokine dimerization.


Subject(s)
Chemokines, CC/chemistry , Chemokines, CC/metabolism , Macrophage Inflammatory Proteins/chemistry , Macrophage Inflammatory Proteins/metabolism , Phenylalanine/metabolism , Receptors, Chemokine/metabolism , Alanine/genetics , Animals , Binding Sites/genetics , CHO Cells , Chemokine CCL4 , Chemokines, CC/genetics , Chemokines, CC/physiology , Cricetinae , Dimerization , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/physiology , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/genetics , Phenylalanine/genetics , Point Mutation , Receptors, CCR5/metabolism , Signal Transduction/genetics , Transfection , Ultracentrifugation
7.
Biochemistry ; 37(26): 9346-54, 1998 Jun 30.
Article in English | MEDLINE | ID: mdl-9649315

ABSTRACT

Chemokines (chemotactic cytokines) are a family of immune system proteins, several of which have been shown to block human immunodeficiency virus (HIV) infection in various cell types. While the solved structures of most chemokines reveal protein dimers, evidence has accumulated for the biological activity of individual chemokine monomers, and a debate has arisen regarding the biological role of the chemokine dimer. Concurrent with this debate, several N-terminal truncations and modifications in the CC subfamily of chemokines have been shown to have functional significance, in many cases antagonizing their respective receptors and in some cases retaining the ability to block HIV entry to the cell. As the dimer interface of CC chemokines is located at their N-terminus, a structural study of N-terminally truncated chemokines will address the effect that this type of mutation has on the dimer-monomer equilibrium. We have studied the structural consequences of N-terminal truncation in macrophage inflammatory protein 1 beta (MIP-1 beta), a CC chemokine that has been shown to block HIV infection. Examination of nuclear magnetic resonance (NMR) spectra of a series of N-terminally truncated MIP-1 beta variants reveals that these proteins possess a range of ability to dimerize. A mutant beginning at amino acid Asp6 [termed MIP(6)] has near wild-type dimer properties, while further truncation results in weakened dimer affinity. The mutant MIP(9) (beginning with amino acid Thr9) has been found to exist solely as a folded monomer. Relaxation measurements yield a rotational correlation time of 8.6 +/- 0.1 ns for wild-type MIP-1 beta and 4.5 +/- 0.1 ns for the MIP(9) mutant, consistent with a wild-type dimer and a fully monomeric MIP(9) variant. The presence of physiological salt concentration drastically changes the monomer-dimer equilibrium for both wild-type and most mutant proteins, heavily favoring the dimeric form of the protein. These results have implications for structure-function analysis of existing chemokine mutants as well as for the larger debate regarding the biological existence and activity of the chemokine dimer.


Subject(s)
Macrophage Inflammatory Proteins/chemistry , Macrophage Inflammatory Proteins/genetics , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Amino Acid Sequence , Chemokine CCL4 , Chemokines, CC/chemistry , Chemokines, CC/genetics , Chemokines, CC/metabolism , Dimerization , Humans , Macrophage Inflammatory Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/metabolism , Solutions
SELECTION OF CITATIONS
SEARCH DETAIL
...