Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Acta Crystallogr D Struct Biol ; 76(Pt 9): 889-898, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32876064

ABSTRACT

Cancer is one of the leading causes of mortality in humans, and recent work has focused on the area of immuno-oncology, in which the immune system is used to specifically target cancerous cells. Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) is an emerging therapeutic target in human cancers owing to its role in degrading cyclic GMP-AMP (cGAMP), an agonist of the stimulator of interferon genes (STING). The available structures of ENPP1 are of the mouse enzyme, and no structures are available with anything other than native nucleotides. Here, the first X-ray crystal structures of the human ENPP1 enzyme in an apo form, with bound nucleotides and with two known inhibitors are presented. The availability of these structures and a robust crystallization system will allow the development of structure-based drug-design campaigns against this attractive cancer therapeutic target.


Subject(s)
Enzyme Inhibitors , Membrane Proteins/agonists , Neoplasms/enzymology , Phosphoric Diester Hydrolases , Pyrophosphatases , Enzyme Inhibitors/chemistry , Humans , Phosphoric Diester Hydrolases/chemistry , Protein Binding , Protein Conformation , Pyrophosphatases/chemistry
2.
Struct Dyn ; 6(6): 064701, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31768400

ABSTRACT

The WD40-repeat protein WDR5 scaffolds various epigenetic writers and is a critical component of the mammalian SET/MLL histone methyltransferase complex. Dysregulation of the MLL1 catalytic function is associated with mixed-lineage leukemia, and antagonism of the WDR5-MLL1 interaction by small molecules has been proposed as a therapeutic strategy for MLL-rearranged cancers. Small molecule binders of the "WIN" site of WDR5 that cause displacement from chromatin have been additionally implicated to be of broader use in cancer treatment. In this study, a fragment screen with Surface Plasmon Resonance (SPR) was used to identify a highly ligand-efficient imidazole-containing compound that is bound in the WIN site. The subsequent medicinal chemistry campaign-guided by a suite of high-resolution cocrystal structures with WDR5-progressed the initial hit to a low micromolar binder. One outcome from this study is a moiety that substitutes well for the side chain of arginine; a tripeptide containing one such substitution was resolved in a high resolution structure (1.5 Å) with a binding mode analogous to the native tripeptide. SPR furthermore indicates a similar residence time (k d = ∼0.06 s-1) for these two analogs. This novel scaffold therefore represents a possible means to overcome the potential permeability issues of WDR5 ligands that possess highly basic groups like guanidine. The series reported here furthers the understanding of the WDR5 WIN site and functions as a starting point for the development of more potent WDR5 inhibitors that may serve as cancer therapeutics.

3.
FEMS Microbiol Lett ; 365(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-30052925

ABSTRACT

Gram-positive bacteria utilise class A sortases to coat the surface of their cells with a diversity of proteins that facilitate interactions with their environment and play fundamental roles in cell physiology and virulence. A putative sortase A gene was identified in the genome of the poorly studied meat spoilage bacterium Brochothrix thermosphacta. To understand how this bacterium mediates interactions with its environment, an N-terminal truncated, His-tagged variant of this protein (His6-BtSrtA) was expressed and purified. Catalytic activity of recombinant His6-BtSrtA was investigated, including sorting motif recognition of target proteins and bioconjugation activity. Further, the B. thermosphacta genome was examined for the presence of sortase A (SrtA) protein substrates. His6-BtSrtA readily formed intermediate complexes with LPXTG-tagged proteins. Although the reaction was inefficient, nucleophilic attack of the resultant thioacyl intermediates by tri-glycine was observed. Genome examination identified 11 potential SrtA substrates, two of which contained protein domains associated with adherence of pathogens to host extracellular matrix proteins and cells, suggesting the B. thermosphacta SrtA may be indirectly involved in its attachment to meat surfaces. Thus, further work in this area could provide crucial insight into molecular mechanisms involved in the colonisation of meat by B. thermosphacta.


Subject(s)
Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Brochothrix/enzymology , Cysteine Endopeptidases/chemistry , Amino Acid Sequence , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Brochothrix/chemistry , Brochothrix/genetics , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Genome, Bacterial , Protein Domains , Substrate Specificity
4.
J Biol Chem ; 291(24): 12641-12657, 2016 Jun 10.
Article in English | MEDLINE | ID: mdl-27036939

ABSTRACT

CXCR4 is a G protein-coupled receptor with excellent potential as a therapeutic target for a range of clinical conditions, including stem cell mobilization, cancer prognosis and treatment, fibrosis therapy, and HIV infection. We report here the development of a fully human single-domain antibody-like scaffold termed an "i-body," the engineering of which produces an i-body library possessing a long complementarity determining region binding loop, and the isolation and characterization of a panel of i-bodies with activity against human CXCR4. The CXCR4-specific i-bodies show antagonistic activity in a range of in vitro and in vivo assays, including inhibition of HIV infection, cell migration, and leukocyte recruitment but, importantly, not the mobilization of hematopoietic stem cells. Epitope mapping of the three CXCR4 i-bodies AM3-114, AM4-272, and AM3-523 revealed binding deep in the binding pocket of the receptor.


Subject(s)
Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/immunology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/pharmacology , Animals , Antibody Specificity/immunology , Binding Sites/immunology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/immunology , Cells, Cultured , Crystallography, X-Ray , Epitope Mapping , HEK293 Cells , HIV Infections/immunology , HIV Infections/prevention & control , HL-60 Cells , Humans , Jurkat Cells , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Models, Molecular , Protein Binding/immunology , Protein Domains , Receptors, CXCR4/metabolism , Single-Domain Antibodies/chemistry , Surface Plasmon Resonance
5.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 7): 1505-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26143922

ABSTRACT

The assembly and anchorage of various pathogenic proteins on the surface of Gram-positive bacteria is mediated by the sortase family of enzymes. These cysteine transpeptidases catalyze a unique sorting signal motif located at the C-terminus of their target substrate and promote the covalent attachment of these proteins onto an amino nucleophile located on another protein or on the bacterial cell wall. Each of the six distinct classes of sortases displays a unique biological role, with sequential activation of multiple sortases often observed in many Gram-positive bacteria to decorate their peptidoglycans. Less is known about the members of the class D family of sortases (SrtD), but they have a suggested role in spore formation in an oxygen-limiting environment. Here, the crystal structure of the SrtD enzyme from Clostridium perfringens was determined at 1.99 Šresolution. Comparative analysis of the C. perfringens SrtD structure reveals the typical eight-stranded ß-barrel fold observed in all other known sortases, along with the conserved catalytic triad consisting of cysteine, histidine and arginine residues. Biochemical approaches further reveal the specifics of the SrtD catalytic activity in vitro, with a significant preference for the LPQTGS sorting motif. Additionally, the catalytic activity of SrtD is most efficient at 316 K and can be further improved in the presence of magnesium cations. Since C. perfringens spores are heat-resistant and lead to foodborne illnesses, characterization of the spore-promoting sortase SrtD may lead to the development of new antimicrobial agents.


Subject(s)
Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Clostridium perfringens/enzymology , Cysteine Endopeptidases/chemistry , Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Clostridium perfringens/chemistry , Clostridium perfringens/metabolism , Crystallography, X-Ray , Cysteine Endopeptidases/metabolism , Dynamic Light Scattering , Metals/metabolism , Models, Molecular , Protein Conformation
6.
Biochem Biophys Res Commun ; 453(3): 449-54, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25281534

ABSTRACT

Self association of the amyloid-ß (Aß42) peptide into oligomers, high molecular weight forms, fibrils and ultimately neuritic plaques, has been correlated with progressive cognitive decline in Alzheimer's disease. Thus, insights into the drivers of the aggregation pathway have the capacity to significantly contribute to our understanding of disease mechanism. Functional assays and a three-dimensional crystal structure of the P3 amyloidogenic region 18-41 of Aß were used to identify residues important in self-association and to design novel non-aggregating variants of the peptide. Biophysical studies (gel filtration, SDS-PAGE, dynamic light scattering, thioflavin T assay, and electron microscopy) demonstrate that in contrast to wild type Aß these targeted mutations lose the ability to self-associate. Loss of aggregation also correlates with reduced neuronal toxicity. Our results highlight residues and regions of the Aß peptide important for future targeting agents aimed at the amelioration of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Peptides/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Benzothiazoles , Microscopy, Electron, Scanning , Mutation , PC12 Cells , Peptides/chemistry , Rats , Thiazoles/metabolism
7.
Org Biomol Chem ; 12(17): 2675-85, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24643508

ABSTRACT

The Sortase A (SrtA) enzyme from Staphylococcus aureus catalyses covalent attachment of protein substrates to pentaglycine cross-bridges in the Gram positive bacterial cell wall. In vitro SrtA-mediated protein ligation is now an important protein engineering tool for conjugation of substrates containing the LPXTGX peptide recognition sequence to oligo-glycine nucleophiles. In order to explore the use of alternative nucleophiles in this system, five different rhodamine-labelled compounds, with N-terminal nucleophilic amino acids, triglycine, glycine, and lysine, or N-terminal non-amino acid nucleophiles ethylenediamine and cadaverine, were synthesized. These compounds were tested for their relative abilities to function as nucleophiles in SrtA-mediated bioconjugation reactions. N-Terminal triglycine, glycine and ethylenediamine were all efficient in labelling a range of LPETGG containing recombinant antibody and scaffold proteins and peptides, while reduced activity was observed for the other nucleophiles across the range of proteins and peptides studied. Expansion of the range of available nucleophiles which can be utilised in SrtA-mediated bioconjugation expands the range of potential applications for this technology. As a demonstration of the utility of this system, SrtA coupling was used to conjugate the triglycine rhodamine-labelled nucleophile to the C-terminus of an Im7 scaffold protein displaying Aß, a neurologically important peptide implicated in Alzheimer's disease. Purified, labelled protein showed Aß-specific targeting to mammalian neuronal cells. Demonstration of targeting neuronal cells with a chimeric protein illustrates the power of this system, and suggests that SrtA-mediated direct cell-surface labelling and visualisation is an achievable goal.


Subject(s)
Aminoacyltransferases/metabolism , Amyloid beta-Peptides/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Embryo, Mammalian/metabolism , Neurons/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Staphylococcus aureus/enzymology , Amino Acid Sequence , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Ethylenediamines/metabolism , Fluorescent Antibody Technique , Immunoglobulin Fab Fragments/metabolism , Mice , Molecular Sequence Data , Neurons/cytology , Oligopeptides/metabolism , Protein Engineering , Recombinant Proteins/metabolism , Single-Chain Antibodies/metabolism , Spectrometry, Mass, Electrospray Ionization
8.
Proteins ; 81(10): 1748-58, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23609990

ABSTRACT

Alzheimer's disease is the most common form of dementia in humans and is related to the accumulation of the amyloid-ß (Aß) peptide and its interaction with metals (Cu, Fe, and Zn) in the brain. Crystallographic structural information about Aß peptide deposits and the details of the metal-binding site is limited owing to the heterogeneous nature of aggregation states formed by the peptide. Here, we present a crystal structure of Aß residues 1-16 fused to the N-terminus of the Escherichia coli immunity protein Im7, and stabilized with the fragment antigen binding fragment of the anti-Aß N-terminal antibody WO2. The structure demonstrates that Aß residues 10-16, which are not in complex with the antibody, adopt a mixture of local polyproline II-helix and turn type conformations, enhancing cooperativity between the two adjacent histidine residues His13 and His14. Furthermore, this relatively rigid region of Aß (residues, 10-16) appear as an almost independent unit available for trapping metal ions and provides a rationale for the His13-metal-His14 coordination in the Aß1-16 fragment implicated in Aß metal binding. This novel structure, therefore, has the potential to provide a foundation for investigating the effect of metal ion binding to Aß and illustrates a potential target for the development of future Alzheimer's disease therapeutics aimed at stabilizing the N-terminal monomer structure, in particular residues His13 and His14, and preventing Aß metal-binding-induced neurotoxicity.


Subject(s)
Amyloid beta-Peptides/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Escherichia coli , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Models, Molecular , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
Methods Mol Biol ; 911: 27-36, 2012.
Article in English | MEDLINE | ID: mdl-22886244

ABSTRACT

Immunoglobulin new antigen receptors (IgNARs) from sharks are a distinct class of immune receptors, consisting of homodimers with no associated light chains. Antigen binding is encapsulated within single VNAR immunoglobulin domains of 13-14 kDa in size. This small size and single domain format means that they exhibit considerable stability and are readily produced in heterologous protein expression systems. In this chapter, I describe the history and discovery of IgNARs, the development of VNAR biotechnology, and highlight important factors in VNAR protein production.


Subject(s)
Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/chemistry , Receptors, Antigen/chemistry , Single-Domain Antibodies/chemistry , Animals , Antibodies, Monoclonal, Humanized , Antibody Affinity/immunology , History, 20th Century , History, 21st Century , Humans , Receptors, Antigen/history , Recombinant Proteins/chemistry , Recombinant Proteins/classification , Recombinant Proteins/history , Recombinant Proteins/isolation & purification , Sharks/immunology , Single-Domain Antibodies/classification , Single-Domain Antibodies/history , Single-Domain Antibodies/isolation & purification
10.
Biotechnol Bioeng ; 109(6): 1461-70, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22170409

ABSTRACT

Sortase-mediated protein ligation is a biological covalent conjugation system developed from the enzymatic cell wall display mechanism found in Staphylococcus aureus. This three-component system requires: (i) purified Sortase A (SrtA) enzyme; (ii) a substrate containing the LPXTG peptide recognition sequence; and (iii) an oligo-glycine acceptor molecule. We describe cloning of the single-chain antibody sc528, which binds to the extracellular domain of the epidermal growth factor receptor (EGFR), from the parental monoclonal antibody and incorporation of a LPETGG tag sequence. Utilizing recombinant SrtA, we demonstrate successful incorporation of biotin from GGG-biotin onto sc528. EGFR is an important cancer target and is over-expressed in human tumor tissues and cancer lines, such as the A431 epithelial carcinoma cells. SrtA-biotinylated sc528 specifically bound EGFR expressed on A431 cells, but not negative control lines. Similarly, when sc528 was labeled with fluorescein we observed antigen-specific labeling. The ability to introduce functionality into recombinant antibodies in a controlled, site-specific manner has applications in experimental, diagnostic, and potentially clinical settings. For example, we demonstrate addition of all three reaction components in situ within a biosensor flow cell, resulting in oriented covalent capture and presentation of sc528, and determination of precise affinities for the antibody-receptor interaction.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , ErbB Receptors/antagonists & inhibitors , Single-Chain Antibodies/metabolism , Staining and Labeling/methods , Aminoacyltransferases/genetics , Antibodies, Blocking/genetics , Antibodies, Blocking/immunology , Antibodies, Blocking/metabolism , Bacterial Proteins/genetics , Biotin/metabolism , Cell Line, Tumor , Cysteine Endopeptidases/genetics , ErbB Receptors/immunology , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
11.
Proteins ; 79(4): 1306-17, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21322055

ABSTRACT

One method of laboratory- or field-based testing for anthrax is detection of Bacillus anthracis spores by high-affinity, high specificity binding reagents. From a pool of monoclonal antibodies, we selected one such candidate (A4D11) with high affinity for tBclA, a truncated version of the B. anthracis exosporium protein BclA. Kinetic analysis utilising both standard and kinetic titration on a Biacore biosensor indicated antibody affinities in the 300 pM range for recombinant tBclA, and the A4D11 antibody was also re-formatted into scFv configuration with no loss of affinity. However, assays against B. anthracis and related Bacilli species showed limited binding of intact spores as well as significant cross-reactivity between species. These results were rationalized by determination of the three-dimensional crystallographic structure of the scFv-tBclA complex. A4D11 binds the side of the tBclA trimer, contacting a face of the antigen normally packed against adjacent trimers within the exosporium structure; this inter-spore interface is highly conserved between Bacilli species. Our results indicate the difficulty of generating a high-affinity antibody to differentiate between the highly conserved spore structures of closely related species, but suggest the possibility of future structure-based antibody design for this difficult target.


Subject(s)
Antibodies, Bacterial/chemistry , Antibodies, Monoclonal/chemistry , Bacillus anthracis/immunology , Bacterial Proteins/immunology , Membrane Glycoproteins/immunology , Single-Chain Antibodies/chemistry , Amino Acid Sequence , Animals , Antibodies, Bacterial/immunology , Antibodies, Bacterial/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Bacillus anthracis/chemistry , Bacterial Proteins/metabolism , Biosensing Techniques , Crystallography, X-Ray , Hybridomas , Membrane Glycoproteins/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism
12.
J Neurosci ; 31(4): 1419-26, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21273426

ABSTRACT

Alzheimer's disease is a progressive neurodegenerative disorder associated with the presence of amyloid-ß (Aß) peptide fibrillar plaques in the brain. However, current evidence suggests that soluble nonfibrillar Aß oligomers may be the major drivers of Aß-mediated synaptic dysfunction. Structural information on these Aß species has been very limited because of their noncrystalline and unstable nature. Here, we describe a crystal structure of amylogenic residues 18-41 of the Aß peptide (equivalent to the p3 α/γ-secretase fragment of amyloid precursor protein) presented within the CDR3 loop region of a shark Ig new antigen receptor (IgNAR) single variable domain antibody. The predominant oligomeric species is a tightly associated Aß dimer, with paired dimers forming a tetramer in the crystal caged within four IgNAR domains, preventing uncontrolled amyloid formation. Our structure correlates with independently observed features of small nonfibrillar Aß oligomers and reveals conserved elements consistent with residues and motifs predicted as critical in Aß folding and oligomerization, thus potentially providing a model system for nonfibrillar oligomer formation in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/chemistry , Peptide Fragments/chemistry , Recombinant Fusion Proteins/chemistry , Amyloid beta-Peptides/genetics , Animals , Crystallography, X-Ray , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Models, Molecular , Mutation , Peptide Fragments/genetics , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Antigen/chemistry , Receptors, Antigen/genetics , Recombinant Fusion Proteins/genetics , Sharks
13.
Anal Biochem ; 385(2): 346-57, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19073134

ABSTRACT

Antibody generation by phage display and related in vitro display technologies routinely yields large panels of clones detected in primary end-point screenings such as enzyme-linked immunosorbent assay (ELISA). However, for the development of clinical lead candidates, rapid determination of secondary characteristics such as kinetics and thermodynamics is of nearly equal importance. Surface plasmon resonance-based biosensors are ideal tools for carrying out such high-throughput secondary screenings, allowing preliminary but confident ranking and identification of lead clones. A key feature of these assays is the stable and reversible capture of antibody fragments from crude samples leading to high-resolution kinetic analysis of library outputs. Here we exploit the high-affinity interaction between the naturally occurring nuclease domain of E. coli colicin E7 (DNaseE7) and its cognate partner, the immunity protein 7 (Im7), to develop a ligand capture system suitable for accurate kinetic ranking of library clones. We demonstrate generic applicability for a range of antibody formats: scFv antibodies, diabodies, antigen binding fragments (Fabs), and shark V(NAR) single domain antibodies. The system is adaptable and reproducible, with comparable results achieved for both the Biacore T100 and ProteOn XPR36 array biosensors.


Subject(s)
Antibodies/chemistry , Biosensing Techniques/methods , Carrier Proteins/chemistry , Colicins/chemistry , Escherichia coli Proteins/chemistry , Immunoconjugates/chemistry , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Kinetics
14.
Curr Opin Pharmacol ; 8(5): 609-15, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18619558

ABSTRACT

Protein scaffolds represent a new generation of universal binding frameworks for use as future immunopharmaceuticals to complement the expanding repertoire of therapeutic monoclonal antibodies. Here, we review recent literature describing advances in protein scaffold development, including efforts to engineer the minimal immunoglobulin-based binding-domain and molecular library design. Several diverse protein folds are currently under development on the basis of modular construction, a strategy also observed in families of naturally evolved immune receptors. We describe potential therapeutic and intracellular applications where scaffold-specific features provide distinct advantages for targeting of non-conventional antigens and comment on the scientific progress and validation of several designed scaffolds in the voyage towards first-in-human trials.


Subject(s)
Protein Engineering/trends , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Animals , Antibodies/chemistry , Antibodies/pharmacology , Camelus , Humans , Immunoglobulins/chemistry , Immunoglobulins/pharmacology , Lampreys
15.
Proteins ; 71(1): 119-30, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17932913

ABSTRACT

Mimotopes mimic the three-dimensional topology of an antigen epitope, and are frequently recognized by antibodies with affinities comparable to those obtained for the original antibody-antigen interaction. Peptides and anti-idiotypic antibodies are two classes of protein mimotopes that mimic the topology (but not necessarily the sequence) of the parental antigen. In this study, we combine these two classes by selecting mimotopes based on single domain IgNAR antibodies, which display exceptionally long CDR3 loop regions (analogous to a constrained peptide library) presented in the context of an immunoglobulin framework with adjacent and supporting CDR1 loops. By screening an in vitro phage-display library of IgNAR variable domains (V(NAR)s) against the target antigen monoclonal antibody MAb5G8, we obtained four potential mimotopes. MAb5G8 targets a linear tripeptide epitope (AYP) in the flexible signal sequence of the Plasmodium falciparum Apical Membrane Antigen-1 (AMA1), and this or similar motifs were detected in the CDR loops of all four V(NAR)s. The V(NAR)s, 1-A-2, -7, -11, and -14, were demonstrated to bind specifically to this paratope by competition studies with an artificial peptide and all showed enhanced affinities (3-46 nM) compared to the parental antigen (175 nM). Crystallographic studies of recombinant proteins 1-A-7 and 1-A-11 showed that the SYP motifs on these V(NAR)s presented at the tip of the exposed CDR3 loops, ideally positioned within bulge-like structures to make contact with the MAb5G8 antibody. These loops, in particular in 1-A-11, were further stabilized by inter- and intra- loop disulphide bridges, hydrogen bonds, electrostatic interactions, and aromatic residue packing. We rationalize the higher affinity of the V(NAR)s compared to the parental antigen by suggesting that adjacent CDR1 and framework residues contribute to binding affinity, through interactions with other CDR regions on the antibody, though of course definitive support of this hypothesis will rely on co-crystallographic studies. Alternatively, the selection of mimotopes from a large (<4 x 10(8)) constrained library may have allowed selection of variants with even more favorable epitope topologies than present in the original antigenic structure, illustrating the power of in vivo selection of mimotopes from phage-displayed molecular libraries.


Subject(s)
Antibodies, Monoclonal , Complementarity Determining Regions/immunology , Epitopes , Immunoglobulins , Animals , Binding Sites , Complementarity Determining Regions/chemistry , Mice , Molecular Mimicry , Plasmodium falciparum , Protein Binding , Sharks
16.
Biotechnol Bioeng ; 99(4): 774-82, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-17879302

ABSTRACT

Spores of Bacillus anthracis, the causative agent of anthrax, are enclosed by an exosporium, which consists of a basal layer surrounded by a nap of hair-like filaments. The major structural component of the filaments is called BclA, which comprises a central collagen-like region (CLR) and a globular C-terminal domain. Here, the entire CLR coding sequence of BclA was removed, and the resulting protein (tBclA) produced in Escherichia coli. The crystallographic structure of tBclA was determined to 1.35 A resolution, and consists of an all-beta structure with a TNF-like jelly fold topology (12 beta-strands which form 2 beta-sheets of five strands each) consistent with previous studies on wild-type BclA. These globular domains are tightly packed into trimeric structures (surface shape complementarity; S (c) = 0.83), demonstrating that formation of the core structure of BclA is independent of the anchoring collagen-like region. A polyclonal antibody raised against tBclA recognized B. anthracis spores directly, and showed little cross-reactivity (<10%) with the spores of the closely related species Bacillus cereus and Bacillus thuringiensis, when compared to two other polyclonal antibodies raised against B. anthracis spore extracts and inactivated spores. The tBclA protein was used to purify a pool of specific antibodies from bovine colostrum whey samples from cows inoculated with the Sterne strain anthrax vaccine, which also showed reactivity with B. anthracis spores. Together, these results demonstrate that tBclA provides a safer and more effective way to the production and purification of antibodies with high binding affinity for B. anthracis spores. Biotechnol. Bioeng. 2008;99: 774-782. (c) 2007 Wiley Periodicals, Inc.


Subject(s)
Bacillus anthracis/physiology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/ultrastructure , Protein Engineering/methods , Animals , Cattle , Crystallography , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spores, Bacterial/physiology
17.
Structure ; 15(11): 1452-66, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17997971

ABSTRACT

Apical membrane antigen 1 (AMA1) is essential for invasion of erythrocytes and hepatocytes by Plasmodium parasites and is a leading malarial vaccine candidate. Although conventional antibodies to AMA1 can prevent such invasion, extensive polymorphisms within surface-exposed loops may limit the ability of these AMA1-induced antibodies to protect against all parasite genotypes. Using an AMA1-specific IgNAR single-variable-domain antibody, we performed targeted mutagenesis and selection against AMA1 from three P. falciparum strains. We present cocrystal structures of two antibody-AMA1 complexes which reveal extended IgNAR CDR3 loops penetrating deep into a hydrophobic cleft on the antigen surface and contacting residues conserved across parasite species. Comparison of a series of affinity-enhancing mutations allowed dissection of their relative contributions to binding kinetics and correlation with inhibition of erythrocyte invasion. These findings provide insights into mechanisms of single-domain antibody binding, and may enable design of reagents targeting otherwise cryptic epitopes in pathogen antigens.


Subject(s)
Antibodies, Protozoan/chemistry , Antigens, Protozoan/chemistry , Immunoglobulin Variable Region/chemistry , Membrane Proteins/chemistry , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Antibody Affinity , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Base Sequence , Binding Sites , Hydrophobic and Hydrophilic Interactions , Immunoglobulin Variable Region/immunology , Immunoglobulin Variable Region/metabolism , Kinetics , Malaria, Falciparum/immunology , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Library , Protein Structure, Tertiary , Protozoan Proteins/metabolism , Surface Plasmon Resonance
18.
Immunol Lett ; 107(2): 163-8, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17069896

ABSTRACT

We have employed a novel mutagenesis system, which utilizes an error-prone RNA dependent RNA polymerase from Qbeta bacteriophage, to create a diverse library of single domain antibody fragments based on the shark IgNAR antibody isotype. Coupling of these randomly mutated mRNA templates directly to the translating ribosome allowed in vitro selection of affinity matured variants showing enhanced binding to target, the apical membrane antigen 1 (AMA1) from Plasmodium falciparum. One mutation mapping to the IgNAR CDR1 loop was not readily additive to other changes, a result explained by structural analysis of aromatic interactions linking the CDR1, CDR3, and Ig framework regions. This combination appeared also to be counter-selected in experiments, suggesting that in vitro affinity maturation is additionally capable of discriminating against incorrectly produced protein variants. Interestingly, a further mutation was directed to a position in the IgNAR heavy loop 4 which is also specifically targeted during the in vivo shark response to antigen, providing a correlation between natural processes and laboratory-based affinity maturation systems.


Subject(s)
Antibodies/immunology , Antibody Affinity/genetics , Immunoglobulin Variable Region/immunology , Immunoglobulins/immunology , Sharks/immunology , Animals , Antibodies/chemistry , Antibodies/genetics , Antigens, Protozoan/immunology , Biosensing Techniques , Immunoglobulin Variable Region/genetics , Immunoglobulins/chemistry , Immunoglobulins/genetics , Membrane Proteins/immunology , Mutagenesis, Site-Directed/methods , Mutation , Peptide Library , Protein Conformation , Protozoan Proteins/immunology , Q beta Replicase/chemistry , RNA, Messenger/chemistry , RNA, Messenger/genetics , Ribosomes/metabolism
19.
J Immunol Methods ; 315(1-2): 171-84, 2006 Aug 31.
Article in English | MEDLINE | ID: mdl-16962608

ABSTRACT

Immunoglobulin new antigen receptors (IgNARs) are unique single domain antibodies found in the serum of sharks. The individual variable (VNAR) domains bind antigen independently and are candidates for the smallest antibody-based immune recognition units (approximately 13 kDa). Here, we first isolated and sequenced the cDNA of a mature IgNAR antibody from the spotted wobbegong shark (Orectolobus maculatus) and confirmed the independent nature of the VNAR domains by dynamic light scattering. Second, we asked which of the reported antibody fragment dimerisation strategies could be applied to VNAR domains to produce small bivalent proteins with high functional affinity (avidity). In contrast to single chain Fv (scFv) fragments, separate IgNARs could not be linked into a tandem single chain format, with the resulting proteins exhibited only monovalent binding due solely to interaction of the N-terminal domain with antigen. Similarly, incorporation of C-terminal helix-turn-helix (dhlx) motifs, while resulting in efficiently dimerised protein, resulted in only a modest enhancement of affinity, probably due to an insufficiently long hinge region linking the antibody to the dhlx motif. Finally, generation of mutants containing half-cystine residues at the VNAR C-terminus produced dimeric recombinant proteins exhibiting high functional affinity for the target antigens, but at the cost of 50-fold decreased protein expression levels. This study demonstrates the potential for construction of bivalent or bispecific IgNAR-based binding reagents of relatively small size (approximately 26 kDa), equivalent to a monovalent antibody Fv fragment, for formulation into future diagnostic and therapeutic formats.


Subject(s)
Immunoglobulin Fragments/immunology , Immunoglobulin Variable Region/immunology , Sharks/immunology , Amino Acid Sequence , Animals , Antibody Affinity , Antibody Specificity , Dimerization , Disulfides/chemistry , Helix-Turn-Helix Motifs/immunology , Immunoglobulin Fragments/genetics , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Models, Molecular , Molecular Sequence Data , Open Reading Frames , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Sharks/genetics , Solubility
20.
Protein Eng Des Sel ; 19(5): 231-44, 2006 May.
Article in English | MEDLINE | ID: mdl-16549402

ABSTRACT

Protein scaffolds derived from non-immunoglobulin sources are increasingly being adapted and engineered to provide unique binding molecules with a diverse range of targeting specificities. The ColE7 immunity protein (Im7) from Escherichia coli is potentially one such molecule, as it combines the advantages of (i) small size, (ii) stability conferred by a conserved four anti-parallel alpha-helical framework and (iii) availability of variable surface loops evolved to inactivate members of the DNase family of bacterial toxins, forming one of the tightest known protein-protein interactions. Here we describe initial cloning and protein expression of Im7 and its cognate partner the 15 kDa DNase domain of the colicin E7. Both proteins were produced efficiently in E.coli, and their in vitro binding interactions were validated using ELISA and biosensor. In order to assess the capacity of the Im7 protein to accommodate extensive loop region modifications, we performed extensive molecular modelling and constructed a series of loop graft variants, based on transfer of the extended CDR3 loop from the IgG1b12 antibody, which targets the gp120 antigen from HIV-1. Loop grafting in various configurations resulted in chimeric proteins exhibiting retention of the underlying framework conformation, as measured using far-UV circular dichroism spectroscopy. Importantly, there was low but measurable transfer of antigen-specific affinity. Finally, to validate Im7 as a selectable scaffold for the generation of molecular libraries, we displayed Im7 as a gene 3 fusion protein on the surface of fd bacteriophages, the most common library display format. The fusion was successfully detected using an anti-Im7 rabbit polyclonal antibody, and the recombinant phage specifically recognized the immobilized DNase. Thus, Im7 scaffold is an ideal protein display scaffold for the future generation and for the selection of libraries of novel binding proteins.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Protein Engineering , Amino Acid Sequence , Bacteriophage M13/genetics , Bacteriophage M13/metabolism , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...