Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 242
Filter
1.
J Mol Biol ; 436(11): 168576, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38641239

ABSTRACT

Prions, the misfolding form of prion proteins, are contagious proteinaceous macromolecules. Recent studies have shown that infectious prion fibrils formed in the brain and non-infectious fibrils formed from recombinant prion protein in a partially denaturing condition have distinct structures. The amyloid core of the in vitro-prepared non-infectious fibrils starts at about residue 160, while that of infectious prion fibrils formed in the brain involves a longer sequence (residues ∼90-230) of structural conversion. The C-terminal truncated prion protein PrP(23-144) can form infectious fibrils under certain conditions and cause disease in animals. In this study, we used cryogenic electron microscopy (cryo-EM) to resolve the structure of hamster sHaPrP(23-144) fibrils prepared at pH 3.7. This 2.88 Å cryo-EM structure has an amyloid core covering residues 94-144. It comprises two protofilaments, each containing five ß-strands arranged as a long hairpin plus an N-terminal ß-strand. This N-terminal ß-strand resides in a positively charged cluster region (named PCC2; sequence 96-111), which interacts with the turn region of the opposite protofilaments' hairpin to stabilize the fibril structure. Interestingly, this sHaPrP(23-144) fibril structure differs from a recently reported structure formed by the human or mouse counterpart at pH 6.5. Moreover, sHaPrP(23-144) fibrils have many structural features in common with infectious prions. Whether this structure is infectious remains to be determined. More importantly, the sHaPrP(23-144) structure is different from the sHaPrP(108-144) fibrils prepared in the same fibrillization buffer, indicating that the N-terminal disordered region, possibly the positively charged cluster, influences the misfolding pathway of the prion protein.


Subject(s)
Amyloid , Cryoelectron Microscopy , Prion Proteins , Protein Folding , Cryoelectron Microscopy/methods , Animals , Amyloid/chemistry , Amyloid/metabolism , Amyloid/ultrastructure , Cricetinae , Prion Proteins/chemistry , Prion Proteins/metabolism , Prion Proteins/genetics , Models, Molecular , Prions/chemistry , Prions/metabolism , Prions/ultrastructure , Protein Conformation
2.
Mol Cell ; 81(21): 4540-4551.e6, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34433091

ABSTRACT

Within the extensive range of self-propagating pathologic protein aggregates of mammals, prions are the most clearly infectious (e.g., ∼109 lethal doses per milligram). The structures of such lethal assemblies of PrP molecules have been poorly understood. Here we report a near-atomic core structure of a brain-derived, fully infectious prion (263K strain). Cryo-electron microscopy showed amyloid fibrils assembled with parallel in-register intermolecular ß sheets. Each monomer provides one rung of the ordered fibril core, with N-linked glycans and glycolipid anchors projecting outward. Thus, single monomers form the templating surface for incoming monomers at fibril ends, where prion growth occurs. Comparison to another prion strain (aRML) revealed major differences in fibril morphology but, like 263K, an asymmetric fibril cross-section without paired protofilaments. These findings provide structural insights into prion propagation, strains, species barriers, and membrane pathogenesis. This structure also helps frame considerations of factors influencing the relative transmissibility of other pathologic amyloids.


Subject(s)
Brain/metabolism , Cryoelectron Microscopy/methods , Polysaccharides/chemistry , Prions/chemistry , Prions/ultrastructure , Amyloid/chemistry , Animals , Glycolipids/chemistry , Humans , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Mice , Phenotype , Prion Proteins/chemistry , Protein Binding , Protein Structure, Secondary , Thermodynamics
3.
Chem Rev ; 121(13): 8285-8307, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34137605

ABSTRACT

This review will focus on the process of amyloid-type protein aggregation. Amyloid fibrils are an important hallmark of protein misfolding diseases and therefore have been investigated for decades. Only recently, however, atomic or near-atomic resolution structures have been elucidated from various in vitro and ex vivo obtained fibrils. In parallel, the process of fibril formation has been studied in vitro under highly artificial but comparatively reproducible conditions. The review starts with a summary of what is known and speculated from artificial in vitro amyloid-type protein aggregation experiments. A partially hypothetic fibril selection model will be described that may be suitable to explain why amyloid fibrils look the way they do, in particular, why at least all so far reported high resolution cryo-electron microscopy obtained fibril structures are in register, parallel, cross-ß-sheet fibrils that mostly consist of two protofilaments twisted around each other. An intrinsic feature of the model is the prion-like nature of all amyloid assemblies. Transferring the model from the in vitro point of view to the in vivo situation is not straightforward, highly hypothetic, and leaves many open questions that need to be addressed in the future.


Subject(s)
Amyloid/chemistry , Amyloidogenic Proteins/chemistry , Prions/chemistry , Protein Aggregates , Amyloid/ultrastructure , Amyloidogenic Proteins/ultrastructure , Animals , Cryoelectron Microscopy , Humans , Prions/ultrastructure
4.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513942

ABSTRACT

Stress granules are ribonucleoprotein assemblies that form in response to cellular stress. Many of the RNA-binding proteins found in stress granule proteomes contain prion-like domains (PrLDs), which are low-complexity sequences that compositionally resemble yeast prion domains. Mutations in some of these PrLDs have been implicated in neurodegenerative diseases, including amyotrophic lateral sclerosis and frontotemporal dementia, and are associated with persistent stress granule accumulation. While both stress granules and prions are macromolecular assemblies, they differ in both their physical properties and complexity. Prion aggregates are highly stable homopolymeric solids, while stress granules are complex dynamic biomolecular condensates driven by multivalent homotypic and heterotypic interactions. Here, we use stress granules and yeast prions as a paradigm to examine how distinct sequence and compositional features of PrLDs contribute to different types of PrLD-containing assemblies.


Subject(s)
Cytoplasmic Granules/genetics , Organelles/genetics , Prion Proteins/genetics , Prions/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Cytoplasmic Granules/ultrastructure , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Organelles/ultrastructure , Prion Proteins/ultrastructure , Prions/ultrastructure , Protein Domains/genetics , Proteome/genetics , RNA-Binding Proteins , Saccharomyces cerevisiae/genetics
5.
FEBS J ; 288(9): 2956-2969, 2021 05.
Article in English | MEDLINE | ID: mdl-33124131

ABSTRACT

The eukaryotic translation elongation factor 1Bγ (eEF1Bγ) is an atypical member of the glutathione transferase (GST) superfamily. Contrary to more classical GSTs having a role in toxic compound detoxification, eEF1Bγ is suggested to act as a scaffold protein, anchoring the elongation factor complex EF1B to the endoplasmic reticulum. In this study, we show that eEF1Bγ from the basidiomycete Phanerochaete chrysosporium is fully active as a glutathione transferase in vitro and undergoes conformational changes upon binding of oxidized glutathione. Using real-time analyses of biomolecular interactions, we show that GSSG allows eEF1Bγ to physically interact with other GSTs from the Ure2p class, opening new perspectives for a better understanding of the role of eEF1Bγ in cellular oxidative stress response.


Subject(s)
Glutathione Peroxidase/genetics , Oxidative Stress/genetics , Peptide Elongation Factor 1/ultrastructure , Phanerochaete/genetics , Prions/genetics , Saccharomyces cerevisiae Proteins/genetics , Amino Acid Sequence/genetics , Animals , Crystallography, X-Ray , DNA-Binding Proteins/genetics , DNA-Binding Proteins/ultrastructure , Glutathione/genetics , Glutathione Disulfide/genetics , Glutathione Peroxidase/ultrastructure , Glutathione Transferase/genetics , Humans , Mice , Peptide Elongation Factor 1/genetics , Phanerochaete/ultrastructure , Prions/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/ultrastructure
6.
Commun Biol ; 3(1): 402, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32728168

ABSTRACT

Prion diseases, a group of incurable, lethal neurodegenerative disorders of mammals including humans, are caused by prions, assemblies of misfolded host prion protein (PrP). A single point mutation (G127V) in human PrP prevents prion disease, however the structural basis for its protective effect remains unknown. Here we show that the mutation alters and constrains the PrP backbone conformation preceding the PrP ß-sheet, stabilising PrP dimer interactions by increasing intermolecular hydrogen bonding. It also markedly changes the solution dynamics of the ß2-α2 loop, a region of PrP structure implicated in prion transmission and cross-species susceptibility. Both of these structural changes may affect access to protein conformers susceptible to prion formation and explain its profound effect on prion disease.


Subject(s)
Prion Diseases/genetics , Prion Proteins/genetics , Prions/genetics , Protein Conformation , Animals , Humans , Point Mutation/genetics , Prion Diseases/pathology , Prion Proteins/ultrastructure , Prions/ultrastructure , Protein Conformation, beta-Strand/genetics
7.
Sci Rep ; 9(1): 19305, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31848406

ABSTRACT

A conformational change from normal prion protein(PrPC) to abnormal prion protein(PrPSC) induces fatal neurodegenerative diseases. Acidic pH is well-known factors involved in the conformational change. Because the protonation of H187 is strongly linked to the change in PrP stability, we examined the charged residues R156, E196, and D202 around H187. Interestingly, there have been reports on pathological mutants, such as H187R, E196A, and D202N. In this study, we focused on how an acidic pH and pathological mutants disrupt this electrostatic network and how this broken network destabilizes PrP structure. To do so, we performed a temperature-based replica-exchange molecular dynamics (T-REMD) simulation using a cumulative 252 µs simulation time. We measured the distance between amino acids comprising four salt bridges (R156-E196/D202 and H187-E196/D202). Our results showed that the spatial configuration of the electrostatic network was significantly altered by an acidic pH and mutations. The structural alteration in the electrostatic network increased the RMSF value around the first helix (H1). Thus, the structural stability of H1, which is anchored to the H2-H3 bundle, was decreased. It induces separation of R156 from the electrostatic network. Analysis of the anchoring energy also shows that two salt-bridges (R156-E196/D202) are critical for PrP stability.


Subject(s)
Prion Proteins/chemistry , Prions/chemistry , Protein Conformation , Static Electricity , Amino Acids/chemistry , Amino Acids/metabolism , Animals , Humans , Hydrogen-Ion Concentration , Molecular Dynamics Simulation , Prion Proteins/ultrastructure , Prions/ultrastructure , Protons
8.
PLoS Pathog ; 15(7): e1007864, 2019 07.
Article in English | MEDLINE | ID: mdl-31295325

ABSTRACT

Prions are unusual protein assemblies that propagate their conformationally-encoded information in absence of nucleic acids. The first prion identified, the scrapie isoform (PrPSc) of the cellular prion protein (PrPC), caused epidemic and epizootic episodes [1]. Most aggregates of other misfolding-prone proteins are amyloids, often arranged in a Parallel-In-Register-ß-Sheet (PIRIBS) [2] or ß-solenoid conformations [3]. Similar folding models have also been proposed for PrPSc, although none of these have been confirmed experimentally. Recent cryo-electron microscopy (cryo-EM) and X-ray fiber-diffraction studies provided evidence that PrPSc is structured as a 4-rung ß-solenoid (4RßS) [4, 5]. Here, we combined different experimental data and computational techniques to build the first physically-plausible, atomic resolution model of mouse PrPSc, based on the 4RßS architecture. The stability of this new PrPSc model, as assessed by Molecular Dynamics (MD) simulations, was found to be comparable to that of the prion forming domain of Het-s, a naturally-occurring ß-solenoid. Importantly, the 4RßS arrangement allowed the first simulation of the sequence of events underlying PrPC conversion into PrPSc. This study provides the most updated, experimentally-driven and physically-coherent model of PrPSc, together with an unprecedented reconstruction of the mechanism underlying the self-catalytic propagation of prions.


Subject(s)
PrPSc Proteins/chemistry , PrPSc Proteins/pathogenicity , Prions/chemistry , Prions/pathogenicity , Animals , Cryoelectron Microscopy , Mice , Models, Molecular , Molecular Dynamics Simulation , PrPC Proteins , PrPSc Proteins/ultrastructure , Prions/ultrastructure , Protein Conformation , Protein Structure, Quaternary
9.
Sci Rep ; 9(1): 376, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30675000

ABSTRACT

Seeded polymerisation of proteins forming amyloid fibres and their spread in tissues has been implicated in the pathogenesis of multiple neurodegenerative diseases: so called "prion-like" mechanisms. While ex vivo mammalian prions, composed of multichain assemblies of misfolded host-encoded prion protein (PrP), act as lethal infectious agents, PrP amyloid fibrils produced in vitro generally do not. The high-resolution structure of authentic infectious prions and the structural basis of prion strain diversity remain unknown. Here we use cryo-electron microscopy and atomic force microscopy to examine the structure of highly infectious PrP rods isolated from mouse brain in comparison to non-infectious recombinant PrP fibrils generated in vitro. Non-infectious recombinant PrP fibrils are 10 nm wide single fibres, with a double helical repeating substructure displaying small variations in adhesive force interactions across their width. In contrast, infectious PrP rods are 20 nm wide and contain two fibres, each with a double helical repeating substructure, separated by a central gap of 8-10 nm in width. This gap contains an irregularly structured material whose adhesive force properties are strikingly different to that of the fibres, suggestive of a distinct composition. The structure of the infectious PrP rods, which cause lethal neurodegeneration, readily differentiates them from all other protein assemblies so far characterised in other neurodegenerative diseases.


Subject(s)
Amyloid/chemistry , Prion Proteins/chemistry , Prions/chemistry , Amyloid/ultrastructure , Animals , Mammals , Microscopy, Atomic Force , Prions/ultrastructure , Protein Conformation , Protein Folding , Recombinant Proteins , Structure-Activity Relationship
10.
FEMS Yeast Res ; 18(6)2018 09 01.
Article in English | MEDLINE | ID: mdl-29846554

ABSTRACT

Yeast prions are self-templating protein-based mechanisms of inheritance whose conformational changes lead to the acquisition of diverse new phenotypes. The best studied of these is the prion domain (NM) of Sup35, which forms an amyloid that can adopt several distinct conformations (strains) that confer distinct phenotypes when introduced into cells that do not carry the prion. Here, we investigate the structure of NM fibrils templated into the prion conformation with cellular lysates. Our electron microscopy studies reveal that NM fibrils that confer either a strong or a weak prion phenotype are both mixtures of thin and thick fibrils that result from differences in packing of the M domain. Strong NM fibrils have more thin fibrils and weak NM fibrils have more thick fibrils. Interestingly, both mass per length and solid state NMR reveal that the thin and thick fibrils have different underlying molecular structures in the prion strain variants that do not interconvert.


Subject(s)
Amyloid/genetics , Amyloid/ultrastructure , Prions/genetics , Prions/ultrastructure , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Amyloid/metabolism , Microscopy, Electron, Scanning , Nuclear Magnetic Resonance, Biomolecular , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Phenotype , Prions/metabolism , Protein Domains , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Species Specificity
11.
J Struct Biol ; 201(1): 5-14, 2018 01.
Article in English | MEDLINE | ID: mdl-29078994

ABSTRACT

Prion and some other incurable human neurodegenerative diseases are associated with misfolding of specific proteins, followed by the formation of amyloids. Despite the widespread usage of the transmission electron and of the atomic force microscopy for studing such amyloids, many related methodological issues still have not been studied until now. Here, we consider one of the first amyloids found in Saccharomyces cerevisiae yeast, i.e. Sup35NMp, to study the adsorption of monomeric protein and its fibrils on the surface of mica, silica, gold and on formvar film. Comparison of linear characteristics of these units calculated by processing of images obtained by the atomic force, transmission and scanning electron microscopy was carried out. The minimal number of measurements of fibril diameters to obtain the values in a given confidence interval were determined. We investigated the film formed by monomeric protein on mica surface, which veiled some morphology features of fibrils. Besides, we revealed that parts of the Sup35NMp excluded from the fibril core can form a wide "coat". The length of the protein forming the core of the fibrils was estimated.


Subject(s)
Amyloid/chemistry , Peptide Termination Factors/chemistry , Prions/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Adsorption , Aluminum Silicates/chemistry , Amyloid/ultrastructure , Gold/chemistry , Microscopy, Atomic Force/methods , Microscopy, Electron, Scanning/methods , Microscopy, Electron, Transmission/methods , Peptide Termination Factors/ultrastructure , Prions/ultrastructure , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/ultrastructure , Silicon Dioxide/chemistry , Surface Properties
12.
J Phys Chem B ; 121(19): 5058-5071, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28459565

ABSTRACT

A fragment of the human prion protein spanning residues 106-126 (PrP106-126) recapitulates many essential properties of the disease-causing protein such as amyloidogenicity and cytotoxicity. PrP106-126 has an amphipathic characteristic that resembles many antimicrobial peptides (AMPs). Therefore, the toxic effect of PrP106-126 could arise from a direct association of monomeric peptides with the membrane matrix. Several experimental approaches are employed to scrutinize the impacts of monomeric PrP106-126 on model lipid membranes. Porous defects in planar bilayers are observed by using solution atomic force microscopy. Adding cholesterol does not impede defect formation. A force spectroscopy experiment shows that PrP106-126 reduces Young's modulus of planar lipid bilayers. We use Raman microspectroscopy to study the effect of PrP106-126 on lipid atomic vibrational dynamics. For phosphatidylcholine lipids, PrP106-126 disorders the intrachain conformation, while the interchain interaction is not altered; for phosphatidylethanolamine lipids, PrP106-126 increases the interchain interaction, while the intrachain conformational order remains similar. We explain the observed differences by considering different modes of peptide insertion. Finally, electron paramagnetic resonance spectroscopy shows that PrP106-126 progressively decreases the orientational order of lipid acyl chains in magnetically aligned bicelles. Together, our experimental data support the proposition that monomeric PrP106-126 can disrupt lipid membranes by using similar mechanisms found in AMPs.


Subject(s)
Microscopy, Atomic Force , Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Prions/chemistry , Prions/ultrastructure , Electron Spin Resonance Spectroscopy , Humans , Particle Size , Spectrum Analysis, Raman , Surface Properties
13.
Int Rev Cell Mol Biol ; 329: 277-301, 2017.
Article in English | MEDLINE | ID: mdl-28109330

ABSTRACT

Prion diseases, such as Creutzfeldt-Jakob disease in humans, bovine spongiform encephalopathy in cattle, chronic wasting disease in cervids (i.e., deer, elk, moose, and reindeer), and sheep scrapie, are caused by the misfolding of the cellular prion protein (PrPC) into a disease-causing conformer (PrPSc). PrPC is a normal, GPI-anchored protein that is expressed on the surface of neurons and other cell types. The structure of PrPC is well understood, based on studies of recombinant PrP, which closely mimics the structure of native PrPC. In contrast, PrPSc is prone to aggregate into a variety of quaternary structures, such as oligomers, amorphous aggregates, and amyloid fibrils. The propensity of PrPSc to assemble into these diverse forms of aggregates is also responsible for our limited knowledge about its structure. Then again, the repeating nature of certain regular PrPSc aggregates has allowed (lower resolution) insights into the structure of the infectious conformer, establishing a four-rung ß-solenoid structure as a key element of its architecture.


Subject(s)
Mammals/metabolism , Prions/chemistry , Protein Aggregates , Amyloid/chemistry , Animals , Crystallization , Humans , Models, Molecular , Prions/metabolism , Prions/ultrastructure
14.
J Cell Biol ; 211(1): 145-58, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26438827

ABSTRACT

Prions consist of misfolded proteins that have adopted an infectious amyloid conformation. In vivo, prion biogenesis is intimately associated with the protein quality control machinery. Using electron tomography, we probed the effects of the heat shock protein Hsp70 chaperone system on the structure of a model yeast [PSI+] prion in situ. Individual Hsp70 deletions shift the balance between fibril assembly and disassembly, resulting in a variable shell of nonfibrillar, but still immobile, aggregates at the surface of the [PSI+] prion deposits. Both Hsp104 (an Hsp100 disaggregase) and Sse1 (the major yeast form of Hsp110) were localized to this surface shell of [PSI+] deposits in the deletion mutants. Elevation of Hsp104 expression promoted the appearance of this novel, nonfibrillar form of the prion aggregate. Moreover, Sse1 was found to regulate prion fibril length. Our studies reveal a key role for Sse1 (Hsp110), in cooperation with Hsp104, in regulating the length and assembly state of [PSI+] prion fibrils in vivo.


Subject(s)
Heat-Shock Proteins/metabolism , Prions/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , HSP110 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Prions/ultrastructure , Protein Aggregates
15.
J Biol Chem ; 290(35): 21510-22, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26175152

ABSTRACT

Human prion diseases can have acquired, sporadic, or genetic origins, each of which results in the conversion of prion protein (PrP) to transmissible, pathological forms. The genetic prion disease Gerstmann-Straussler-Scheinker syndrome can arise from point mutations of prolines 102 or 105. However, the structural effects of these two prolines, and mutations thereof, on PrP misfolding are not well understood. Here, we provide evidence that individual mutations of Pro-102 or Pro-105 to noncyclic aliphatic residues such as the Gerstmann-Straussler-Scheinker-linked leucines can promote the in vitro formation of PrP amyloid with extended protease-resistant cores reminiscent of infectious prions. This effect was enhanced by additional charge-neutralizing mutations of four nearby lysine residues comprising the so-called central lysine cluster. Substitution of these proline and lysine residues accelerated PrP conversion such that spontaneous amyloid formation was no longer slower than scrapie-seeded amyloid formation. Thus, Pro-102 and Pro-105, as well as the lysines in the central lysine cluster, impede amyloid formation by PrP, implicating these residues as key structural modulators in the conversion of PrP to disease-associated types of amyloid.


Subject(s)
Amyloid/metabolism , Lysine/metabolism , Prions/chemistry , Prions/metabolism , Proline/metabolism , Amyloid/ultrastructure , Animals , Brain/metabolism , Brain/pathology , Conserved Sequence , Cricetinae , Endopeptidase K/metabolism , Humans , Kinetics , Mice , Molecular Sequence Data , Mutant Proteins/chemistry , Mutation , Negative Staining , PrPSc Proteins/metabolism , Prions/ultrastructure , Protein Denaturation , Protein Structure, Secondary , Scrapie/metabolism , Sequence Alignment , Spectroscopy, Fourier Transform Infrared , Structure-Activity Relationship
16.
Biochim Biophys Acta ; 1848(10 Pt A): 2422-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26215743

ABSTRACT

PrP 106-126 conserves the pathogenic and physicochemical properties of the Scrapie isoform of the prion protein. PrP 106-126 and other amyloidal proteins are capable of inducing ion permeability through cell membranes, and this property may represent the common primary mechanism of pathogenesis in the amyloid-related degenerative diseases. However, for many amyloidal proteins, despite numerous phenomenological observations of their interactions with membranes, it has been difficult to determine the molecular mechanisms by which the proteins cause ion permeability. One approach that has not been undertaken is the kinetic study of protein-membrane interactions. We found that the reaction time constant of the interaction between PrP 106-126 and membranes is suitable for such studies. The kinetic experiment with giant lipid vesicles showed that the membrane area first increased by peptide binding but then decreased. The membrane area decrease was coincidental with appearance of extramembranous aggregates including lipid molecules. Sometimes, the membrane area would increase again followed by another decrease. The kinetic experiment with small vesicles was monitored by circular dichroism for peptide conformation changes. The results are consistent with a molecular simulation following a simple set of well-defined rules. We deduced that at the molecular level the formation of peptide amyloids incorporated lipid molecules as part of the aggregates. Most importantly the amyloid aggregates desorbed from the lipid bilayer, consistent with the macroscopic phenomena observed with giant vesicles. Thus we conclude that the main effect of membrane-mediated amyloid formation is extraction of lipid molecules from the membrane. We discuss the likelihood of this effect on membrane ion permeability.


Subject(s)
Amyloid/chemical synthesis , Amyloid/ultrastructure , Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Phospholipids/chemistry , Prions/chemistry , Prions/ultrastructure , Unilamellar Liposomes/chemistry , Kinetics
17.
Tsitologiia ; 57(2): 144-52, 2015.
Article in Russian | MEDLINE | ID: mdl-26035972

ABSTRACT

We have studied the steady-state transmembrane current induced by amyloid and amyloid-like peptides in lipid bilayers in the presence of dipole modifiers. It has been shown that the addition of dipole modifier, phloretin, to the membrane bathing solutions leads to an increase in the multichannel activity of amyloid beta-peptide fragment 25-35, [Gly35]-amyloid beta-peptide fragment 25--35, prion protein fragment 106-126 and amyloid-like peptides myr-BASP1 (1--13), myr-BASP1(1--19) and GAP-43(1--40). We have found that the effect of phloretin is not the result of dipole potential changes due to adsorption of this modifier on the membrane. Using the various fragments of amyloid beta-peptide, presenilin, prion protein and neuronal proteins BASP1 and GAP-43 allowes to conclude that the steady-state peptide-induced transmembrane current in the case of addition of phloretin is due to the electrostatic interaction between the positively charged channel-forming agents and negatively charged dipole modifier. The results obtained by electron microscopy have demonstrated that this interaction increases degree of peptide oligomerization.


Subject(s)
Amyloid beta-Peptides/ultrastructure , GAP-43 Protein/ultrastructure , Membrane Proteins/ultrastructure , Nerve Tissue Proteins/ultrastructure , Peptide Fragments/ultrastructure , Prions/ultrastructure , Repressor Proteins/ultrastructure , Amyloid beta-Peptides/drug effects , GAP-43 Protein/drug effects , Humans , Lipid Bilayers/chemical synthesis , Lipid Bilayers/metabolism , Membrane Potentials , Membrane Proteins/drug effects , Microscopy, Electron , Nerve Tissue Proteins/drug effects , Peptide Fragments/drug effects , Phloretin/pharmacology , Prions/drug effects , Repressor Proteins/drug effects , Static Electricity
18.
Proteins ; 83(10): 1751-65, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26018750

ABSTRACT

The amyloid conversion is a massive detrimental modification affecting several proteins upon specific physical or chemical stimuli characterizing a plethora of diseases. In many cases, the amyloidogenic stimuli induce specific structural features to the protein conferring the propensity to misfold and form amyloid deposits. The investigation of mutants, structurally similar to their native isoform but inherently prone to amyloid conversion, may be a viable strategy to elucidate the structural features connected with amyloidogenesis. In this article, we present a computational protocol based on the combination of molecular dynamics (MD) and grid-based approaches suited for the pairwise comparison of closely related protein structures. This method was applied on the cellular prion protein (PrP(C)) as a case study and, in particular, addressed to the quali/quantification of the structural features conferred by either E200K mutations and treatment with CaCl(2), both able to induce the scrapie conversion of PrP. Several schemes of comparison were developed and applied to this case study, and made up suitable of application to other protein systems. At this purpose an in-house python codes has been implemented that, together with the parallelization of the GRID force fields program, will spread the applicability of the proposed computational procedure.


Subject(s)
Computational Biology/methods , Molecular Dynamics Simulation , Prions/chemistry , Prions/ultrastructure , Cluster Analysis , Structural Homology, Protein , Surface Properties
19.
Sci Rep ; 5: 10101, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25960067

ABSTRACT

Prion diseases are lethal, infectious diseases associated with prion protein (PrP) misfolding. A large number of mammals are susceptible to both sporadic and acquired prion diseases. Although PrP is highly conserved and ubiquitously expressed in all mammals, not all species exhibit prion disease. By employing full length recombinant PrP from five known prion susceptible species (human, cattle, cat, mouse and hamster) and two species considered to be prion resistant (pig and dog) the amyloidogenicity of these PrPs has been delineated. All the mammalian PrPs, even from resistant species, were swiftly converted from the native state to amyloid-like structure when subjected to a native condition conversion assay. The PrPs displayed amyloidotypic tinctorial and ultrastructural hallmarks. Self-seeded conversion of the PrPs displayed significantly decreased lag phases demonstrating that nucleation dependent polymerization is a dominating mechanism in the fibrillation process. Fibrils from Aß1-40, Aß1-42, Lysozyme, Insulin and Transthyretin did not accelerate conversion of HuPrP whereas fibrils from HuPrP90-231 and HuPrP121-231 as well as full length PrPs of all PrPs efficiently seeded conversion showing specificity of the assay requiring the C-terminal PrP sequence. Our findings have implications for PrP misfolding and could have ramifications in the context of prion resistant species and silent carriers.


Subject(s)
Amyloid/chemistry , Mammals/metabolism , Prions/chemistry , Amino Acid Sequence , Animals , Benzothiazoles , Circular Dichroism , Humans , Kinetics , Molecular Sequence Data , Negative Staining , Prions/isolation & purification , Prions/ultrastructure , Protein Aggregates , Protein Folding , Species Specificity , Thiazoles/metabolism , Time Factors
20.
Sci Rep ; 5: 10062, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25950908

ABSTRACT

Mammalian prions exist as multiple strains which produce characteristic and highly reproducible phenotypes in defined hosts. How this strain diversity is encoded by a protein-only agent remains one of the most interesting and challenging questions in biology with wide relevance to understanding other diseases involving the aggregation or polymerisation of misfolded host proteins. Progress in understanding mammalian prion strains has however been severely limited by the complexity and variability of the methods used for their isolation from infected tissue and no high resolution structures have yet been reported. Using high-throughput cell-based prion bioassay to re-examine prion purification from first principles we now report the isolation of prion strains to exceptional levels of purity from small quantities of infected brain and demonstrate faithful retention of biological and biochemical strain properties. The method's effectiveness and simplicity should facilitate its wide application and expedite structural studies of prions.


Subject(s)
Brain/metabolism , Prions/isolation & purification , Prions/metabolism , Animals , Cricetinae , Humans , Mice , Prions/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...