Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Amyloid ; 28(3): 158-167, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33583280

ABSTRACT

More than 30 proteins and peptides have been found to form amyloid fibrils in human diseases. Fibrils formed by transthyretin (TTR) are associated with ATTR amyloidosis, affecting many vital organs, including the heart and peripheral nervous system. Congo red staining is the gold standard method for detection of amyloid deposits in tissue. However, Congo red staining and amyloid typing methods such as immunofluorescence labelling are limited to relatively large deposits. Detection of small ATTR deposits present at an early stage of the disease could enable timely treatment and prevent severe tissue damage. In this study, we developed an enhanced ATTR amyloid detection method that uses functionalised protein nanofibrils. Using this method, we achieved sensitive detection of monomeric TTR in a microplate immunoassay and immunofluorescence labelling of ex vivo tissue from two patients containing ATTR aggregates. The system's utility was confirmed on sections from a patient with AA amyloidosis and liver sections from inflamed mouse. These results suggest that the detection system constitutes important new technology for highly sensitive detection of microscopic amounts of ATTR amyloid deposited in tissue.


Subject(s)
Amyloid Neuropathies, Familial , Amyloidosis , Amyloid , Amyloid Neuropathies, Familial/diagnosis , Amyloid Neuropathies, Familial/genetics , Amyloidogenic Proteins , Animals , Humans , Mice , Prealbumin/genetics , Serum Amyloid A Protein
2.
J Biol Chem ; 294(41): 14966-14977, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31416835

ABSTRACT

Concerns over the environment are a central driver for designing cell-free enzymatic cascade reactions that synthesize non-petrol-based commodity compounds. An often-suggested strategy that would demonstrate the economic competitiveness of this technology is recycling of valuable enzymes through their immobilization. For this purpose, amyloid nanofibrils are an ideal scaffold to realize chemistry-free covalent enzyme immobilization on a material that offers a large surface area. However, in most instances, only single enzyme-functionalized amyloid fibrils have so far been studied. To embark on the next stage, here we displayed xylanase A, ß-xylosidase, and an aldose sugar dehydrogenase on Sup35(1-61) nanofibrils to convert beechwood xylan to xylonolactone. We characterized this enzymatic cascade by measuring the time-dependent accumulation of xylose, xylooligomers, and xylonolactone. Furthermore, we studied the effects of relative enzyme concentrations, pH, temperature, and agitation on product formation. Our investigations revealed that a modular cascade with a mixture of xylanase and ß-xylosidase, followed by product removal and separate oxidation of xylose with the aldose sugar dehydrogenase, is more productive than an enzyme mix containing all of these enzymes together. Moreover, we found that the nanofibril-coupled enzymes do not lose activity compared with their native state. These findings provide proof of concept of the feasibility of functionalized Sup35(1-61) fibrils as a molecular scaffold for biocatalytic cascades consisting of reusable enzymes that can be used in biotechnology.


Subject(s)
Amyloid/chemistry , Biocatalysis , Nanostructures/chemistry , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Protein Aggregates , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Biotechnology , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Kinetics , Models, Molecular , Oxidation-Reduction , Protein Structure, Secondary , Xylose/metabolism
3.
Front Aging Neurosci ; 11: 64, 2019.
Article in English | MEDLINE | ID: mdl-30967771

ABSTRACT

Different strategies for treatment and prevention of Alzheimer's disease (AD) are currently under investigation, including passive immunization with anti-amyloid ß (anti-Aß) monoclonal antibodies (mAbs). Here, we investigate the therapeutic potential of a novel type of Aß-targeting agent based on an affibody molecule with fundamentally different properties to mAbs. We generated a therapeutic candidate, denoted ZSYM73-albumin-binding domain (ABD; 16.8 kDa), by genetic linkage of the dimeric ZSYM73 affibody for sequestering of monomeric Aß-peptides and an ABD for extension of its in vivo half-life. Amyloid precursor protein (APP)/PS1 transgenic AD mice were administered with ZSYM73-ABD, followed by behavioral examination and immunohistochemistry. Results demonstrated rescued cognitive functions and significantly lower amyloid burden in the treated animals compared to controls. No toxicological symptoms or immunology-related side-effects were observed. To our knowledge, this is the first reported in vivo investigation of a systemically delivered scaffold protein against monomeric Aß, demonstrating a therapeutic potential for prevention of AD.

4.
J Alzheimers Dis ; 66(3): 1053-1064, 2018.
Article in English | MEDLINE | ID: mdl-30372682

ABSTRACT

Aggregation and deposition of misfolded amyloid-ß (Aß) peptide in the brain is central to Alzheimer's disease (AD). Oligomeric, protofibrillar, and fibrillar forms of Aß are believed to be neurotoxic and cause neurodegeneration in AD, but the toxicity mechanisms are not well understood and may involve Aß-interacting molecular partners. In a previous study, we identified potential Aß42 protofibrillar-binding proteins in serum and cerebrospinal fluid (CSF) using an engineered version of Aß42 (Aß42CC) that forms protofibrils, but not fibrils. Here we studied binding of proteins to Aß42 fibrils in AD and non-AD CSF and compared these with protofibrillar Aß42CC-binding partners. Aß42 fibrils sequestered 2.4-fold more proteins than Aß42CC protofibrils. Proteins with selective binding to fibrillar aggregates with low nanomolar affinity were identified. We also found that protofibrillar and fibrillar Aß-binding proteins represent distinct functional categories. Aß42CC protofibrils triggered interactions with proteins involved in catalytic activities, like transferases and oxidoreductases, while Aß42 fibrils were more likely involved in binding to proteoglycans, growth factors and neuron-associated proteins, e.g., neurexin-1, -2, and -3. Interestingly, 10 brain-enriched proteins were identified among the fibril-binding proteins, while protofibril-extracted proteins had more general expression patterns. Both types of Aß aggregates bound several extracellular proteins. Additionally, we list a set of CSF proteins that might have potential to discriminate between AD and non-AD CSF samples. The results may be of relevance both for biomarker studies and for studies of Aß-related toxicity mechanisms.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid/cerebrospinal fluid , Aged , Female , Humans , Male , Mass Spectrometry , Middle Aged , Peptide Fragments/cerebrospinal fluid , Protein Binding
5.
ACS Nano ; 12(9): 9363-9371, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30207696

ABSTRACT

Amyloid nanofibrils are excellent scaffolds for designable materials that can be endowed with biotechnologically relevant functions. However, most of all excellent ideas and concepts that have been reported in the literature might never see real-world implementation in biotechnological applications. One bottleneck is the large-scale production of these materials. In this paper, we present an attempt to create a generic and scalable platform for producing ready-to-use functionalized nanofibrils directly from a eukaryotic organism. As a model material, we assembled Sup35(1-61) amyloid nanofibrils from Saccharomyces cerevisiae decorated with the Z-domain dimer, which has a high affinity toward antibody molecules. To this end, Komagataella pastoris was engineered by inserting gene copies of Sup35(1-61) and the protein chimera Sup35(1-61)-ZZ into the genome. This strain has the capability to constantly secrete amyloidogenic proteins into the extracellular medium, where the mature functionalized fibrils form, with a production yield of 35 mg/L culture. Another striking feature of this strategy is that the separation of the fibril material from the cells requires only centrifugation and resuspension in saline water. The fast production rates, minimal hands-on time, and high stability of the assembled material are some highlights that make the direct assembly of functionalized fibrils in the extracellular medium an alternative to production methods that are not suitable for large-scale production of designed amyloids.


Subject(s)
Nanofibers/chemistry , Peptide Termination Factors/biosynthesis , Pichia/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Models, Molecular , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Pichia/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
6.
PLoS One ; 13(4): e0196250, 2018.
Article in English | MEDLINE | ID: mdl-29684061

ABSTRACT

Enzymatic functionalization of cross-ß structured protein nanofibrils has hitherto resulted in a severe reduction of the catalytic efficiency of high turnover biocatalysts. It has been speculated that steric restrictions and mass transport pose limits on the attached enzymes, but detailed kinetics analyzing this have not yet been reported. For a more comprehensive understanding, we studied protein nanofibrils endowed with TEM1, a ß-lactamase from Escherichia coli. The packing density of TEM1 along the fibrils was controlled by co-fibrillation; in other words, the N-terminal ureidosuccinate transporter Ure2(1-80) from Saccharomyces cerevisiae was simultaneously aggregated with the chimeric proteins TEM1-Ure2(1-80). The mature fibrils were trapped in a column, and the rate of ampicillin hydrolysis was recorded using a continuous substrate flow. The turnover rate was plotted as a function of substrate molecules available per enzyme per second, which demonstrated that an elevated substrate availability counteracts mass transport limitations. To analyze this data set, we derived a kinetic model, which makes it possible to easily characterize and compare enzymes packed in columns. The functional TEM1 nanofibrils possess 80% of the catalytic turnover rate compared to free TEM1 in solution. Altogether, we have created protein nanofibrils that can effectively hydrolyze ß-lactam antibiotic contaminations and provided a groundwork strategy for other highly functional enzymatic nanofibrils.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Bioreactors , Enzymes, Immobilized , Glutathione Peroxidase/metabolism , Nanofibers , Prions/metabolism , Saccharomyces cerevisiae Proteins/metabolism , beta-Lactamases/metabolism , Ampicillin/metabolism , Ampicillin/pharmacokinetics , Anti-Bacterial Agents/metabolism , Biocatalysis , Biodegradation, Environmental , Bioreactors/microbiology , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Escherichia coli/enzymology , Glutathione Peroxidase/chemistry , Hydrolysis , Kinetics , Nanofibers/chemistry , Prions/chemistry , Protein Multimerization , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry
7.
Sci Rep ; 7(1): 5949, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28729665

ABSTRACT

Protofibrils of the 42 amino acids long amyloid-ß peptide are transient pre-fibrillar intermediates in the process of peptide aggregation into amyloid plaques and are thought to play a critical role in the pathology of Alzheimer's disease. Hence, there is a need for research reagents and potential diagnostic reagents for detection and imaging of such aggregates. Here we describe an in vitro selection of Affibody molecules that bind to protofibrils of Aß42cc, which is a stable engineered mimic of wild type Aß42 protofibrils. Several binders were identified that bind Aß42cc protofibrils with low nanomolar affinities, and which also recognize wild type Aß42 protofibrils. Dimeric head-to-tail fusion proteins with subnanomolar binding affinities, and very slow dissociation off-rates, were also constructed. A mapping of the chemical properties of the side chains onto the Affibody scaffold surface reveals three distinct adjacent surface areas of positively charged surface, nonpolar surface and a polar surface, which presumably match a corresponding surface epitope on the protofibrils. The results demonstrate that the engineered Aß42cc is a suitable antigen for directed evolution of affinity reagents with specificity for wild type Aß42 protofibrils.


Subject(s)
Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Protein Aggregates , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Cell Surface Display Techniques , Kinetics , Peptide Fragments/chemistry , Protein Binding , Protein Multimerization , Recombinant Fusion Proteins/chemistry
8.
Biotechnol J ; 12(6)2017 Jun.
Article in English | MEDLINE | ID: mdl-28371185

ABSTRACT

Elevated performance of instruments and electronic devices is frequently attained through miniaturization of the involved components, which increases the number of functional units in a given volume. Analogously, to conquer the limitations of materials used for the purification of monoclonal antibodies and for the sensitivity of immunoassays, the support for capturing antibodies requires miniaturization. A suitable scaffold for this purpose are cross-ß structured protein nanofibrils, as they offer a superior surface area over volume ratio and because manipulation can be implemented genetically. To display the antibody binding Z-domain dimers (ZZ) along the surface of the fibrils and grant maximal accessibility to the functional units, the N-terminal fragments of the fibrillating translation release factor Sup35 or ureidosuccinate transporter Ure2, both from Saccharomyces cerevisae, are simultaneously fibrillated with the chimeric-proteins Sup35-ZZ and ZZ-Ure2, respectively. Optimization of the fibril composition yields a binding capacity of 1.8 mg antibody per mg fibril, which is a binding capacity that is almost 20-fold higher, compared to the commercially available affinity medium gold standard, protein A sepharose. This study lifts the craft of nanofibril functionalization to the next level, and offers a universal framework to improve biomaterials that rely on the display of functional proteins or enzymes.


Subject(s)
Antibodies, Monoclonal/metabolism , Glutathione Peroxidase/metabolism , Peptide Termination Factors/metabolism , Prions/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Antibodies, Monoclonal/isolation & purification , Protein Binding , Protein Structure, Tertiary
9.
Mol Microbiol ; 102(4): 593-610, 2016 11.
Article in English | MEDLINE | ID: mdl-27507539

ABSTRACT

Three pathogenic species of the genus Yersinia assemble adhesive fimbriae via the FGL-chaperone/usher pathway. Closely related Y. pestis and Y. pseudotuberculosis elaborate the pH6 antigen (Psa), which mediates bacterial attachment to alveolar cells of the lung. Y. enterocolitica, instead, assembles the homologous fimbriae Myf of unknown function. Here, we discovered that Myf, like Psa, specifically recognizes ß1-3- or ß1-4-linked galactose in glycosphingolipids, but completely lacks affinity for phosphatidylcholine, the main receptor for Psa in alveolar cells. The crystal structure of a subunit of Psa (PsaA) complexed with choline together with mutagenesis experiments revealed that PsaA has four phosphatidylcholine binding pockets that enable super-high-avidity binding of Psa-fibres to cell membranes. The pockets are arranged as six tyrosine residues, which are all missing in the MyfA subunit of Myf. Conversely, the crystal structure of the MyfA-galactose complex revealed that the galactose-binding site is more extended in MyfA, enabling tighter binding to lactosyl moieties. Our results suggest that during evolution, Psa has acquired a tyrosine-rich surface that enables it to bind to phosphatidylcholine and mediate adhesion of Y. pestis/pseudotuberculosis to alveolar cells, whereas Myf has specialized as a carbohydrate-binding adhesin, facilitating the attachment of Y. enterocolitica to intestinal cells.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Fimbriae, Bacterial/metabolism , Yersinia/metabolism , Adhesins, Bacterial/metabolism , Amino Acid Sequence , Antigens, Bacterial/genetics , Antigens, Bacterial/ultrastructure , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/ultrastructure , Binding Sites , Fimbriae Proteins/metabolism , Molecular Chaperones/metabolism , Tropism/genetics , Virulence/genetics , Yersinia enterocolitica/metabolism , Yersinia pestis/metabolism , Yersinia pseudotuberculosis/metabolism
10.
Biotechnol J ; 10(11): 1707-18, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26184787

ABSTRACT

The amyloid hypothesis suggests that accumulation of amyloid ß (Aß) peptides in the brain is involved in development of Alzheimer's disease. We previously generated a small dimeric affinity protein that inhibited Aß aggregation by sequestering the aggregation prone parts of the peptide. The affinity protein is originally based on the Affibody scaffold, but is evolved to a distinct interaction mechanism involving complex structural rearrangement in both the Aß peptide and the affinity proteins upon binding. The aim of this study was to decrease the size of the dimeric affinity protein and significantly improve its affinity for the Aß peptide to increase its potential as a future therapeutic agent. We combined a rational design approach with combinatorial protein engineering to generate two different affinity maturation libraries. The libraries were displayed on staphylococcal cells and high-affinity Aß-binding molecules were isolated using flow-cytometric sorting. The best performing candidate binds Aß with a KD value of around 300 pM, corresponding to a 50-fold improvement in affinity relative to the first-generation binder. The new dimeric Affibody molecule was shown to capture Aß1-42 peptides from spiked E. coli lysate. Altogether, our results demonstrate successful engineering of this complex binder for increased affinity to the Aß peptide.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Cell Surface Display Techniques/methods , Protein Engineering/methods , Recombinant Fusion Proteins/metabolism , Combinatorial Chemistry Techniques , Escherichia coli/metabolism , Flow Cytometry , Models, Molecular , Peptide Library , Protein Binding , Recombinant Fusion Proteins/chemistry , Staphylococcus/chemistry , Staphylococcus/metabolism
11.
Angew Chem Int Ed Engl ; 54(30): 8837-40, 2015 Jul 20.
Article in English | MEDLINE | ID: mdl-26119103

ABSTRACT

Conversion of the intrinsically disordered protein α-synuclein (α-syn) into amyloid aggregates is a key process in Parkinson's disease. The sequence region 35-59 contains ß-strand segments ß1 and ß2 of α-syn amyloid fibril models and most disease-related mutations. ß1 and ß2 frequently engage in transient interactions in monomeric α-syn. The consequences of ß1-ß2 contacts are evaluated by disulfide engineering, biophysical techniques, and cell viability assays. The double-cysteine mutant α-synCC, with a disulfide linking ß1 and ß2, is aggregation-incompetent and inhibits aggregation and toxicity of wild-type α-syn. We show that α-syn delays the aggregation of amyloid-ß peptide and islet amyloid polypeptide involved in Alzheimer's disease and type 2 diabetes, an effect enhanced in the α-synCC mutant. Tertiary interactions in the ß1-ß2 region of α-syn interfere with the nucleation of amyloid formation, suggesting promotion of such interactions as a potential therapeutic approach.


Subject(s)
Amyloid/metabolism , Protein Aggregates , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Amyloid/chemistry , Amyloid/ultrastructure , Humans , Models, Molecular , Protein Folding , Protein Structure, Secondary , alpha-Synuclein/ultrastructure
12.
Chembiochem ; 16(3): 411-4, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25557164

ABSTRACT

Amyloidogenic proteins share a propensity to convert to the ß-structure-rich amyloid state that is associated with the progression of several protein-misfolding disorders. Here we show that a single engineered ß-hairpin-binding protein, the ß-wrapin AS10, binds monomers of three different amyloidogenic proteins, that is, amyloid-ß peptide, α-synuclein, and islet amyloid polypeptide, with sub-micromolar affinity. AS10 binding inhibits the aggregation and toxicity of all three proteins. The results demonstrate common conformational preferences and related binding sites in a subset of the amyloidogenic proteins. These commonalities enable the generation of multispecific monomer-binding agents.


Subject(s)
Amyloidogenic Proteins/metabolism , Recombinant Proteins/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins/chemistry , Binding Sites , Humans , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/metabolism , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Recombinant Proteins/chemistry , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism
13.
ACS Chem Biol ; 10(3): 766-74, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25469473

ABSTRACT

The progressive neurodegeneration in Alzheimer's disease is believed to be linked to the presence of prefibrillar aggregates of the amyloid-ß (Aß) peptide in the brain. The exact role of these aggregates in the disease pathology is, however, still an open question. Any mechanism by which oligomeric Aß may cause damage to neuronal cells must, in one way or another, involve interactions with other molecules. Here, we identify proteins in human serum and cerebrospinal fluid that bind to stable protofibrils formed by an engineered variant of Aß42 (Aß42CC). We find that the protofibrils attract a substantial number of protein binding partners. Many of the 101 identified proteins are involved in lipid transport and metabolism, the complement system, or in hemostasis. Binding of representative proteins from all of these groups with micromolar affinity was confirmed using surface plasmon resonance. In addition, binding of apolipoprotein E to the protofibrils with nanomolar affinity was demonstrated. We also find that aggregation of Aß enhances protein binding, as lower amounts of proteins bind monomeric Aß. Proteins that bind to Aß protofibrils might contribute to biological effects in which these aggregates are involved. Our results therefore suggest that an improved understanding of the mechanisms by which Aß causes cytotoxicity and neurodegeneration might be gained from studies carried out in biologically relevant matrices in which Aß-binding proteins are present.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Apolipoproteins E/chemistry , Blood Proteins/chemistry , Cerebrospinal Fluid Proteins/chemistry , Peptide Fragments/chemistry , Aged , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Apolipoproteins E/isolation & purification , Blood Proteins/isolation & purification , Cerebrospinal Fluid Proteins/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression , Humans , Kinetics , Male , Middle Aged , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Aggregates , Protein Binding , Protein Engineering , Protein Interaction Mapping , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface Plasmon Resonance
14.
Angew Chem Int Ed Engl ; 53(47): 12756-60, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25256598

ABSTRACT

Oligomeric and protofibrillar aggregates formed by the amyloid-ß peptide (Aß) are believed to be involved in the pathology of Alzheimer's disease. Central to Alzheimer pathology is also the fact that the longer Aß42 peptide is more prone to aggregation than the more prevalent Aß40 . Detailed structural studies of Aß oligomers and protofibrils have been impeded by aggregate heterogeneity and instability. We previously engineered a variant of Aß that forms stable protofibrils and here we use solid-state NMR spectroscopy and molecular modeling to derive a structural model of these. NMR data are consistent with packing of residues 16 to 42 of Aß protomers into hexameric barrel-like oligomers within the protofibril. The core of the oligomers consists of all residues of the central and C-terminal hydrophobic regions of Aß, and hairpin loops extend from the core. The model accounts for why Aß42 forms oligomers and protofibrils more easily than Aß40 .


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Amyloid/chemical synthesis , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular
15.
J Phys Chem Lett ; 5(3): 607-14, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-26276617

ABSTRACT

Amyloid fibrils with cross-ß spine basic architectures are prevalent and stable forms of peptides and proteins. Recent research has provided significant contributions to our understanding of the mechanisms of fibril formation and to the surprising diversity and persistence of structural polymorphism in amyloid fibrils. There have also been successful demonstrations of how molecules can be engineered to inhibit unwanted amyloid formation by different mechanisms. Future research in these areas will include investigations of mechanisms for primary nucleation and the structure of oligomeric intermediates, the general role of secondary nucleation events (autocatalysis), elucidation of the mechanisms and implications of preservation of structural morphology in amyloid propagation, and research into the largely unexplored phenomenon of cross-seeding, by which amyloid fibrils of one species induce the formation of amyloid by another species.

16.
Protein Eng Des Sel ; 26(10): 635-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23924760

ABSTRACT

Affibody molecules are engineered binding proteins, in which the three-helix bundle motif of the Z domain derived from protein A is used as a scaffold for sequence variation. We used phage display to select Affibody binders to staphylococcal protein A itself. The best binder, called ZpA963, binds with similar affinity and kinetics to the five homologous E, D, A, B and C domains of protein A, and to a five-domain protein A construct with an average dissociation constant, K(D), of ~20 nM. The structure of ZpA963 in complex with the Z domain shows that it interacts with a surface on Z that is identical in the five protein A domains, which explains the multi-domain affinity. This property allows for high-affinity binding by dimeric Affibody molecules that simultaneously engage two protein A domains in a complex. We studied two ZpA963 dimers in which the subunits were linked by a C-terminal disulfide in a symmetric dimer or head-to-tail in a fusion protein, respectively. The dimers both bind protein A with high affinity, very slow off-rates and with saturation-dependent kinetics that can be understood in terms of dimer binding to multiple sites. The head-to-tail (ZpA963)2htt dimer binds with an off-rate of k(off) ≤ 5 × 10(-6) s(-1) and an estimated K(D) ≤ 16 pM. The results illustrate how dimers of selected monomer binding proteins can provide an efficient route for engineering of high-affinity binders to targets that contain multiple homologous domains or repeated structural units.


Subject(s)
Protein Engineering , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Staphylococcal Protein A/metabolism , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Peptide Library , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics
17.
PLoS One ; 8(7): e66101, 2013.
Article in English | MEDLINE | ID: mdl-23843949

ABSTRACT

Structural and biochemical studies of the aggregation of the amyloid-ß peptide (Aß) are important to understand the mechanisms of Alzheimer's disease, but research is complicated by aggregate inhomogeneity and instability. We previously engineered a hairpin form of Aß called Aßcc, which forms stable protofibrils that do not convert into amyloid fibrils. Here we provide a detailed characterization of Aß42cc protofibrils. Like wild type Aß they appear as smooth rod-like particles with a diameter of 3.1 (±0.2) nm and typical lengths in the range 60 to 220 nm when observed by atomic force microscopy. Non-perturbing analytical ultracentrifugation and nanoparticle tracking analyses are consistent with such rod-like protofibrils. Aß42cc protofibrils bind the ANS dye indicating that they, like other toxic protein aggregates, expose hydrophobic surface. Assays with the OC/A11 pair of oligomer specific antibodies put Aß42cc protofibrils into the same class of species as fibrillar oligomers of wild type Aß. Aß42cc protofibrils may be used to extract binding proteins in biological fluids and apolipoprotein E is readily detected as a binder in human serum. Finally, Aß42cc protofibrils act to attenuate spontaneous synaptic activity in mouse hippocampal neurons. The experiments indicate considerable structural and chemical similarities between protofibrils formed by Aß42cc and aggregates of wild type Aß42. We suggest that Aß42cc protofibrils may be used in research and applications that require stable preparations of protofibrillar Aß.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/pharmacology , Apolipoproteins E/chemistry , Hippocampus/drug effects , Neurons/drug effects , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/ultrastructure , Animals , Binding Sites , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/cytology , Hippocampus/physiology , Humans , Mice , Microscopy, Atomic Force , Molecular Mimicry , Neurons/cytology , Neurons/physiology , Particle Size , Peptide Fragments/ultrastructure , Protein Binding , Protein Engineering , Protein Structure, Secondary , Synapses , Synaptic Transmission
18.
Biotechnol J ; 8(1): 139-45, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22987778

ABSTRACT

We have previously generated an affibody molecule for the disease-associated amyloid beta (Aß) peptide, which has been shown to inhibit the formation of various Aß aggregates and revert the neurotoxicity of Aß in a fruit fly model of Alzheimer's disease. In this study, we have investigated a new bacterial display system for combinatorial protein engineering of the Aß-binder as a head-to-tail dimeric construct for future optimization efforts, e.g. affinity maturation. Using the bacterial display platform, we have: (i) demonstrated functional expression of the dimeric binder on the cell surface, (ii) determined the affinity and investigated the pH sensitivity of the interaction, (iii) demonstrated the importance of an intramolecular disulfide bond through selections from a cell-displayed combinatorial library, as well as (iv) investigated the effects from rational truncation of the N-terminal part of the affibody molecule on surface expression level and Aß binding. Overall, the detailed engineering and characterization of this promising Aß-specific affibody molecule have yielded valuable insights concerning its unusual binding mechanism. The results also demonstrated that our bacterial display system is a suitable technology for future protein engineering and characterization efforts of homo- or heterodimeric affinity proteins.


Subject(s)
Amyloid beta-Peptides/metabolism , Protein Engineering/methods , Recombinant Fusion Proteins/metabolism , Staphylococcus/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Biotechnology , Cell Membrane/metabolism , Flow Cytometry , Humans , Models, Molecular , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Staphylococcus/chemistry , Staphylococcus/cytology , Staphylococcus/genetics
19.
J Mol Biol ; 421(4-5): 441-65, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22244855

ABSTRACT

Amyloid is aggregated protein in the form of insoluble fibrils. Amyloid deposition in human tissue-amyloidosis-is associated with a number of diseases including all common dementias and type II diabetes. Considerable progress has been made to understand the mechanisms leading to amyloid formation. It is, however, not yet clear by which mechanisms amyloid and protein aggregates formed on the path to amyloid are cytotoxic. Strategies to prevent protein aggregation and amyloid formation are nevertheless, in many cases, promising and even successful. This review covers research on intervention of amyloidosis and highlights several examples of how inhibition of protein aggregation and amyloid formation has been achieved in practice. For instance, rational design can provide drugs that stabilize a native folded state of a protein, protein engineering can provide new binding proteins that sequester monomeric peptides from aggregation, small molecules and peptides can be designed to block aggregation or direct it into non-cytotoxic paths, and monoclonal antibodies have been developed for therapies towards neurodegenerative diseases based on inhibition of amyloid formation and clearance.


Subject(s)
Amyloid/antagonists & inhibitors , Amyloid/metabolism , Amyloid/toxicity , Amyloidosis/pathology , Amyloidosis/physiopathology , Amyloidosis/therapy , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy , Protein Conformation , Protein Denaturation
20.
J Chem Theory Comput ; 8(10): 3871-9, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-26593028

ABSTRACT

Backbone torsional strain has been implicated as a cause of rate enhancement in a class of autoprocessing proteins performing proteolysis and protein splicing. In the autoproteolytic protein domain SEA, folding and proteolytic activity have experimentally been shown to be coupled with about 7 kcal/mol of folding free energy available for catalysis. Here, we have examined the catalytic strategy of SEA with molecular dynamics simulations, potential of mean force free energy profiles, and B3LYP/6-311G(d,p) density functional calculations. A quantitative estimate of the free energy stored as protein strain (about 8 kcal/mol), that is available for catalyzing the cleavage reaction, is obtained and found to be in excellent agreement with thermodynamic and kinetic data. It is further shown that there is strong coupling between folding and reaction coordinates leading to reactant state destabilization in the direction of folding and transition state stabilization along the reaction coordinate. This situation is different from the preorganized active site model in that the fully folded transition state stabilizing structure is not realized until the reaction barrier is surmounted.

SELECTION OF CITATIONS
SEARCH DETAIL
...