Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(11): e0294465, 2023.
Article in English | MEDLINE | ID: mdl-37976283

ABSTRACT

Oligomers formed from monomers of the amyloid ß-protein (Aß) are thought to be central to the pathogenesis of Alzheimer's disease (AD). Unsurprisingly for a complex disease, current mouse models of AD fail to fully mimic the clinical disease in humans. Moreover, results obtained in a given mouse model are not always reproduced in a different model. Cellular prion protein (PrPC) is now an established receptor for Aß oligomers. However, studies of the Aß-PrPC interaction in different mouse models have yielded contradictory results. Here we performed a longitudinal study assessing a range of biochemical and histological features in the commonly used J20 and APP-PS1 mouse models. Our analysis demonstrated that PrPC ablation had no effect on amyloid accumulation or oligomer production. However, we found that APP-PS1 mice had higher levels of oligomers, that these could bind to recombinant PrPC, and were recognised by the OC antibody which distinguishes parallel, in register fibrils. On the other hand, J20 mice had a lower level of Aß oligomers, which did not interact with PrPC when tested in vitro and were OC-negative. These results suggest the two mouse models produce diverse Aß assemblies that could interact with different targets, highlighting the necessity to characterise the conformation of the Aß oligomers concomitantly with the toxic cascade elicited by them. Our results provide an explanation for the apparent contradictory results found in APP-PS1 mice and the J20 mouse line in regards to Aß toxicity mediated by PrPC.


Subject(s)
Alzheimer Disease , PrPC Proteins , Prions , Humans , Mice , Animals , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Prion Proteins/genetics , Longitudinal Studies , PrPC Proteins/genetics , PrPC Proteins/metabolism , Mice, Transgenic
3.
Biol Psychiatry ; 83(4): 358-368, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29331212

ABSTRACT

The initial report that cellular prion protein (PrPC) mediates toxicity of amyloid-ß species linked to Alzheimer's disease was initially treated with scepticism, but growing evidence supports this claim. That there is a high-affinity interaction is now clear, and its molecular basis is being unraveled, while recent studies have identified possible downstream toxic mechanisms. Determination of the clinical significance of such interactions between PrPC and disease-associated amyloid-ß species will require experimental medicine studies in humans. Trials of compounds that inhibit PrP-dependent amyloid-ß toxicity are commencing in humans, and although it is clear that only a fraction of Alzheimer's disease toxicity could be governed by PrPC, a partial, but still therapeutically useful, role in human disease may soon be testable.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Prion Proteins/metabolism , Animals , Humans
4.
EMBO Mol Med ; 10(1): 22-31, 2018 01.
Article in English | MEDLINE | ID: mdl-29113975

ABSTRACT

Intronic GGGGCC repeat expansions in C9orf72 are the most common known cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), which are characterised by degeneration of cortical and motor neurons, respectively. Repeat expansions have been proposed to cause disease by both the repeat RNA forming foci that sequester RNA-binding proteins and through toxic dipeptide repeat proteins generated by repeat-associated non-ATG translation. GGGGCC repeat RNA folds into a G-quadruplex secondary structure, and we investigated whether targeting this structure is a potential therapeutic strategy. We performed a screen that identified three structurally related small molecules that specifically stabilise GGGGCC repeat G-quadruplex RNA We investigated their effect in C9orf72 patient iPSC-derived motor and cortical neurons and show that they significantly reduce RNA foci burden and the levels of dipeptide repeat proteins. Furthermore, they also reduce dipeptide repeat proteins and improve survival in vivo, in GGGGCC repeat-expressing Drosophila Therefore, small molecules that target GGGGCC repeat G-quadruplexes can ameliorate the two key pathologies associated with C9orf72 FTD/ALS These data provide proof of principle that targeting GGGGCC repeat G-quadruplexes has therapeutic potential.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , C9orf72 Protein/genetics , Drug Discovery , Frontotemporal Dementia/drug therapy , G-Quadruplexes/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Amyotrophic Lateral Sclerosis/genetics , Animals , Drosophila , Frontotemporal Dementia/genetics , Humans , RNA/chemistry , RNA/genetics , Repetitive Sequences, Nucleic Acid/drug effects , Small Molecule Libraries/therapeutic use
5.
Open Biol ; 7(11)2017 11.
Article in English | MEDLINE | ID: mdl-29142106

ABSTRACT

Mammalian prions cause lethal neurodegenerative diseases such as Creutzfeldt-Jakob disease (CJD) and consist of multi-chain assemblies of misfolded cellular prion protein (PrPC). Ligands that bind to PrPC can inhibit prion propagation and neurotoxicity. Extensive prior work established that certain soluble assemblies of the Alzheimer's disease (AD)-associated amyloid ß-protein (Aß) can tightly bind to PrPC, and that this interaction may be relevant to their toxicity in AD. Here, we investigated whether such soluble Aß assemblies might, conversely, have an inhibitory effect on prion propagation. Using cellular models of prion infection and propagation and distinct Aß preparations, we found that the form of Aß assemblies which most avidly bound to PrP in vitro also inhibited prion infection and propagation. By contrast, forms of Aß which exhibit little or no binding to PrP were unable to attenuate prion propagation. These data suggest that soluble aggregates of Aß can compete with prions for binding to PrPC and emphasize the bidirectional nature of the interplay between Aß and PrPC in Alzheimer's and prion diseases. Such inhibitory effects of Aß on prion propagation may contribute to the apparent fall-off in the incidence of sporadic CJD at advanced age where cerebral Aß deposition is common.


Subject(s)
Amyloid beta-Peptides/metabolism , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , Amyloid beta-Peptides/toxicity , Animals , Cell Line , Mice , PrPSc Proteins/toxicity , Protein Binding
7.
Nature ; 525(7568): 247-50, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26354483

ABSTRACT

More than two hundred individuals developed Creutzfeldt-Jakob disease (CJD) worldwide as a result of treatment, typically in childhood, with human cadaveric pituitary-derived growth hormone contaminated with prions. Although such treatment ceased in 1985, iatrogenic CJD (iCJD) continues to emerge because of the prolonged incubation periods seen in human prion infections. Unexpectedly, in an autopsy study of eight individuals with iCJD, aged 36-51 years, in four we found moderate to severe grey matter and vascular amyloid-ß (Aß) pathology. The Aß deposition in the grey matter was typical of that seen in Alzheimer's disease and Aß in the blood vessel walls was characteristic of cerebral amyloid angiopathy and did not co-localize with prion protein deposition. None of these patients had pathogenic mutations, APOE ε4 or other high-risk alleles associated with early-onset Alzheimer's disease. Examination of a series of 116 patients with other prion diseases from a prospective observational cohort study showed minimal or no Aß pathology in cases of similar age range, or a decade older, without APOE ε4 risk alleles. We also analysed pituitary glands from individuals with Aß pathology and found marked Aß deposition in multiple cases. Experimental seeding of Aß pathology has been previously demonstrated in primates and transgenic mice by central nervous system or peripheral inoculation with Alzheimer's disease brain homogenate. The marked deposition of parenchymal and vascular Aß in these relatively young patients with iCJD, in contrast with other prion disease patients and population controls, is consistent with iatrogenic transmission of Aß pathology in addition to CJD and suggests that healthy exposed individuals may also be at risk of iatrogenic Alzheimer's disease and cerebral amyloid angiopathy. These findings should also prompt investigation of whether other known iatrogenic routes of prion transmission may also be relevant to Aß and other proteopathic seeds associated with neurodegenerative and other human diseases.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/etiology , Creutzfeldt-Jakob Syndrome/etiology , Drug Contamination , Human Growth Hormone/administration & dosage , Iatrogenic Disease , Adult , Alleles , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/analysis , Autopsy , Blood Vessels/metabolism , Blood Vessels/pathology , Case-Control Studies , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Creutzfeldt-Jakob Syndrome/complications , Creutzfeldt-Jakob Syndrome/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Gray Matter/metabolism , Gray Matter/pathology , Humans , Middle Aged , Prions/administration & dosage , Prions/metabolism , Risk Factors
8.
J Biol Chem ; 290(27): 17020-8, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25995455

ABSTRACT

The prion protein (PrP) has been implicated both in prion diseases such as Creutzfeldt-Jakob disease, where its monomeric cellular isoform (PrP(C)) is recruited into pathogenic self-propagating polymers of misfolded protein, and in Alzheimer disease, where PrP(C) may act as a receptor for synaptotoxic oligomeric forms of amyloid-ß (Aß). There has been considerable interest in identification of compounds that bind to PrP(C), stabilizing its native fold and thereby acting as pharmacological chaperones to block prion propagation and pathogenesis. However, compounds binding PrP(C) could also inhibit the binding of toxic Aß species and may have a role in treating Alzheimer disease, a highly prevalent dementia for which there are currently no disease-modifying treatments. However, the absence of a unitary, readily measurable, physiological function of PrP makes screening for ligands challenging, and the highly heterogeneous nature of Aß oligomer preparations makes conventional competition binding assays difficult to interpret. We have therefore developed a high-throughput screen that utilizes site-specifically fluorescently labeled protein to identify compounds that bind to PrP and inhibit both Aß binding and prion propagation. Following a screen of 1,200 approved drugs, we identified Chicago Sky Blue 6B as the first small molecule PrP ligand capable of inhibiting Aß binding, demonstrating the feasibility of development of drugs to block this interaction. The interaction of Chicago Sky Blue 6B was characterized by isothermal titration calorimetry, and its ability to inhibit Aß binding and reduce prion levels was established in cell-based assays.


Subject(s)
Amyloid beta-Peptides/metabolism , PrPC Proteins/metabolism , Protein Binding/drug effects , Trypan Blue/pharmacology , Amyloid beta-Peptides/genetics , Calorimetry , Drug Discovery , High-Throughput Screening Assays , Humans , PrPC Proteins/genetics
9.
Science ; 345(6201): 1192-1194, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25103406

ABSTRACT

An expanded GGGGCC repeat in C9orf72 is the most common genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis. A fundamental question is whether toxicity is driven by the repeat RNA itself and/or by dipeptide repeat proteins generated by repeat-associated, non-ATG translation. To address this question, we developed in vitro and in vivo models to dissect repeat RNA and dipeptide repeat protein toxicity. Expression of pure repeats, but not stop codon-interrupted "RNA-only" repeats in Drosophila caused adult-onset neurodegeneration. Thus, expanded repeats promoted neurodegeneration through dipeptide repeat proteins. Expression of individual dipeptide repeat proteins with a non-GGGGCC RNA sequence revealed that both poly-(glycine-arginine) and poly-(proline-arginine) proteins caused neurodegeneration. These findings are consistent with a dual toxicity mechanism, whereby both arginine-rich proteins and repeat RNA contribute to C9orf72-mediated neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA Repeat Expansion/genetics , Drosophila melanogaster/genetics , Frontotemporal Dementia/genetics , Proteins/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , C9orf72 Protein , Cell Line, Tumor , Dipeptides/metabolism , Disease Models, Animal , Escherichia coli , Frontotemporal Dementia/pathology , Humans , Neurons/metabolism , Neurons/pathology
10.
J Biol Chem ; 289(37): 25497-508, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25074940

ABSTRACT

The self-association of prion protein (PrP) is a critical step in the pathology of prion diseases. It is increasingly recognized that small non-fibrillar ß-sheet-rich oligomers of PrP may be of crucial importance in the prion disease process. Here, we characterize the structure of a well defined ß-sheet-rich oligomer, containing ∼12 PrP molecules, and often enclosing a central cavity, formed using full-length recombinant PrP. The N-terminal region of prion protein (residues 23-90) is required for the formation of this distinct oligomer; a truncated form comprising residues 91-231 forms a broad distribution of aggregated species. No infectivity or toxicity was found using cell and animal model systems. This study demonstrates that examination of the full repertoire of conformers and assembly states that can be accessed by PrP under specific experimental conditions should ideally be done using the full-length protein.


Subject(s)
Amyloid/chemistry , Prion Diseases/metabolism , Prions/chemistry , Protein Structure, Tertiary , Amyloid/metabolism , Humans , Hydrogen-Ion Concentration , Prion Diseases/pathology , Prions/metabolism , Protein Folding , Protein Structure, Secondary , Recombinant Proteins/chemistry
11.
J Neurosci ; 34(18): 6140-5, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24790184

ABSTRACT

Alzheimer's disease (AD) is associated with pathological assembly states of amyloid-ß protein (Aß). Aß-related synaptotoxicity can be blocked by anti-prion protein (PrP) antibodies, potentially allowing therapeutic targeting of this aspect of AD neuropathogenesis. Here, we show that intravascular administration of a high-affinity humanized anti-PrP antibody to rats can prevent the plasticity-disrupting effects induced by exposure to soluble AD brain extract. These results provide an in vivo proof of principle for such a therapeutic strategy.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Antibodies, Monoclonal/administration & dosage , CA1 Region, Hippocampal/drug effects , Long-Term Potentiation/drug effects , Peptide Fragments/pharmacology , Prions/immunology , Aged, 80 and over , Analysis of Variance , Animals , Biophysics , Drug Administration Routes , Electric Stimulation , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Prions/metabolism , Rats , Rats, Wistar , Temporal Lobe/chemistry , Temporal Lobe/metabolism
12.
Nat Commun ; 5: 3374, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24594908

ABSTRACT

NMDA-type glutamate receptors (NMDARs) are currently regarded as paramount in the potent and selective disruption of synaptic plasticity by Alzheimer's disease amyloid ß-protein (Aß). Non-NMDAR mechanisms remain relatively unexplored. Here we describe how Aß facilitates NMDAR-independent long-term depression of synaptic transmission in the hippocampus in vivo. Synthetic Aß and Aß in soluble extracts of Alzheimer's disease brain usurp endogenous acetylcholine muscarinic receptor-dependent long-term depression, to enable long-term depression that required metabotropic glutamate-5 receptors (mGlu5Rs). We also find that mGlu5Rs are essential for Aß-mediated inhibition of NMDAR-dependent long-term potentiation in vivo. Blocking Aß binding to cellular prion protein with antibodies prevents the facilitation of long-term depression. Our findings uncover an overarching role for Aß-PrP(C)-mGlu5R interplay in mediating both LTD facilitation and LTP inhibition, encompassing NMDAR-mediated processes that were previously considered primary.


Subject(s)
Amyloid beta-Peptides/metabolism , Long-Term Synaptic Depression/physiology , Receptor, Metabotropic Glutamate 5/metabolism , Animals , Hippocampus/metabolism , Male , Prions/metabolism , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5/genetics
13.
Nat Commun ; 4: 2416, 2013.
Article in English | MEDLINE | ID: mdl-24022506

ABSTRACT

Growing evidence suggests water-soluble, non-fibrillar forms of amyloid-ß protein (Aß) have important roles in Alzheimer's disease with toxicities mimicked by synthetic Aß(1-42). However, no defined toxic structures acting via specific receptors have been identified and roles of proposed receptors, such as prion protein (PrP), remain controversial. Here we quantify binding to PrP of Aß(1-42) after different durations of aggregation. We show PrP-binding and PrP-dependent inhibition of long-term potentiation (LTP) correlate with the presence of protofibrils. Globular oligomers bind less avidly to PrP and do not inhibit LTP, whereas fibrils inhibit LTP in a PrP-independent manner. That only certain transient Aß assemblies cause PrP-dependent toxicity explains conflicting reports regarding the involvement of PrP in Aß-induced impairments. We show that these protofibrils contain a defined nanotubular structure with a previously unidentified triple helical conformation. Blocking the formation of Aß nanotubes or their interaction with PrP might have a role in treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Nanotubes/toxicity , Prions/toxicity , Synapses/drug effects , Synapses/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/ultrastructure , Animals , Humans , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Time Factors , Tomography
14.
Sci Rep ; 2: 1016, 2012.
Article in English | MEDLINE | ID: mdl-23264878

ABSTRACT

Large expansions of a non-coding GGGGCC-repeat in the first intron of the C9orf72 gene are a common cause of both amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). G-rich sequences have a propensity for forming highly stable quadruplex structures in both RNA and DNA termed G-quadruplexes. G-quadruplexes have been shown to be involved in a range of processes including telomere stability and RNA transcription, splicing, translation and transport. Here we show using NMR and CD spectroscopy that the C9orf72 hexanucleotide expansion can form a stable G-quadruplex, which has profound implications for disease mechanism in ALS and FTD.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA Repeat Expansion , Frontotemporal Dementia/genetics , G-Quadruplexes , Proteins/genetics , Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein , DNA/genetics , Frontotemporal Dementia/metabolism , Genotype , Humans , Nuclear Magnetic Resonance, Biomolecular , Proteins/chemistry
15.
Nat Commun ; 2: 336, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21654636

ABSTRACT

A role for PrP in the toxic effect of oligomeric forms of Aß, implicated in Alzheimer's disease (AD), has been suggested but remains controversial. Here we show that PrP is required for the plasticity-impairing effects of ex vivo material from human AD brain and that standardized Aß-derived diffusible ligand (ADDL) preparations disrupt hippocampal synaptic plasticity in a PrP-dependent manner. We screened a panel of anti-PrP antibodies for their ability to disrupt the ADDL-PrP interaction. Antibodies directed to the principal PrP/Aß-binding site and to PrP helix-1, were able to block Aß binding to PrP suggesting that the toxic Aß species are of relatively high molecular mass and/or may bind multiple PrP molecules. Two representative and extensively characterized monoclonal antibodies directed to these regions, ICSM-35 and ICSM-18, were shown to block the Aß-mediated disruption of synaptic plasticity validating these antibodies as candidate therapeutics for AD either individually or in combination.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Models, Molecular , Neuronal Plasticity/physiology , Prions/metabolism , Synapses/metabolism , Synapses/physiology , Alzheimer Disease/immunology , Animals , Antibodies, Monoclonal , Chromatography, Gel , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Electrophysiology , Humans , Ligands , Long-Term Potentiation/physiology , Mice , Mice, Knockout , Microscopy, Electron , Ultracentrifugation
16.
J Neurosci ; 30(43): 14411-9, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20980598

ABSTRACT

Nonfibrillar, water-soluble low-molecular weight assemblies of the amyloid ß-protein (Aß) are believed to play an important role in Alzheimer's disease (AD). Aqueous extracts of human brain contain Aß assemblies that migrate on SDS-polyacrylamide gels and elute from size exclusion as dimers (∼8 kDa) and can block long-term potentiation and impair memory consolidation in the rat. Such species are detected specifically and sensitively in extracts of Alzheimer brain suggesting that SDS-stable dimers may be the basic building blocks of AD-associated synaptotoxic assemblies. Consequently, understanding the structure and properties of Aß dimers is of great interest. In the absence of sufficient brain-derived dimer to facilitate biophysical analysis, we generated synthetic dimers designed to mimic the natural species. For this, Aß(1-40) containing cysteine in place of serine 26 was used to produce disulphide cross-linked dimer, (AßS26C)2. Such dimers had no detectable secondary structure, produced an analytical ultracentrifugation profile consistent for an ∼8.6 kDa protein, and had no effect on hippocampal long-term potentiation (LTP). However, (AßS26C)2 aggregated more rapidly than either AßS26C or wild-type monomers and formed parastable ß-sheet rich, thioflavin T-positive, protofibril-like assemblies. Whereas wild-type Aß aggregated to form typical amyloid fibrils, the protofibril-like structures formed by (AßS26C)2 persisted for prolonged periods and potently inhibited LTP in mouse hippocampus. These data support the idea that Aß dimers may stabilize the formation of fibril intermediates by a process distinct from that available to Aß monomer and that higher molecular weight prefibrillar assemblies are the proximate mediators of Aß toxicity.


Subject(s)
Amyloid beta-Peptides/toxicity , Neurofibrillary Tangles/pathology , Neurotoxins/toxicity , Synapses/pathology , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Animals , Circular Dichroism , Cross-Linking Reagents , Dimerization , Drug Synergism , Electrophoresis, Polyacrylamide Gel , Electrophysiology , Light , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Molecular Sequence Data , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurotoxins/chemistry , Protein Conformation , Scattering, Radiation , Synapses/drug effects , Ultracentrifugation
17.
Proc Natl Acad Sci U S A ; 107(41): 17610-5, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20876144

ABSTRACT

In prion diseases, the misfolded protein aggregates are derived from cellular prion protein (PrP(C)). Numerous ligands have been reported to bind to human PrP(C) (huPrP), but none to the structured region with the affinity required for a pharmacological chaperone. Using equilibrium dialysis, we screened molecules previously suggested to interact with PrP to discriminate between those which did not interact with PrP, behaved as nonspecific polyionic aggregates or formed a genuine interaction. Those that bind could potentially act as pharmacological chaperones. Here we report that a cationic tetrapyrrole [Fe(III)-TMPyP], which displays potent antiprion activity, binds to the structured region of huPrP. Using a battery of biophysical techniques, we demonstrate that Fe(III)-TMPyP forms a 11 complex via the structured C terminus of huPrP with a K(d) of 4.5 ± 2 µM, which is in the range of its IC(50) for curing prion-infected cells of 1.6 ± 0.4 µM and the concentration required to inhibit protein-misfolding cyclic amplification. Therefore, this molecule tests the hypothesis that stabilization of huPrP(C), as a principle, could be used in the treatment of human prion disease. The identification of a binding site with a defined 3D structure opens up the possibility of designing small molecules that stabilize huPrP and prevent its conversion into the disease-associated form.


Subject(s)
Drug Discovery/methods , Models, Molecular , Prion Diseases/drug therapy , Prions/metabolism , Protein Binding , Tetrapyrroles/metabolism , Binding Sites/genetics , Biophysics/methods , Circular Dichroism , Humans , Magnetic Resonance Spectroscopy , Prions/chemistry , Protein Folding , Ultracentrifugation
18.
Infect Disord Drug Targets ; 9(1): 48-57, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19200015

ABSTRACT

Prions are unique in that the infectious particles contain no detectable nucleic acid and appear to consist of aggregated forms of misfolded cellular prion protein. Prions form distinct strains and can transmit disease between species. Whilst the molecular basis of prion diseases is beginning to be unravelled, much remains unknown including the atomic structure of the infectious and toxic species. In contrast, the structure and folding properties of the cellular prion protein are well characterised and, although its precise function remains enigmatic, constitutive knockout of protein expression in mice produces apparently healthy animals but which are fully resistant to prion infection. Furthermore, recent data show that neuronal knockout of the gene encoding for prion protein during established brain infection leads to reversal of pathology and behavioural deficits, giving hope that effective therapies could be designed. Stabilising the cellular form of the prion protein and preventing it from misfolding could be one way to slow or prevent prion formation. Immunotherapy of peripherally prion-infected mice with an antibody specific for cellular prion protein can prevent disease onset. However, a small molecule capable of curing prion infection in vivo has still to be discovered. Recent work has provided proof of principle that compounds which bind selectively to the cellular prion protein could act as therapeutics for prion diseases.


Subject(s)
PrPC Proteins/physiology , Prion Diseases/metabolism , Prions/pathogenicity , Animals , Humans , PrPC Proteins/genetics , PrPC Proteins/metabolism , Prion Diseases/genetics , Prion Diseases/prevention & control , Prions/genetics , Prions/metabolism
19.
Acta Crystallogr D Biol Crystallogr ; 64(Pt 3): 264-72, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18323621

ABSTRACT

The high photon flux at third-generation synchrotron sources can inflict significant primary radiation damage upon macromolecular crystals, even when the crystals are cryocooled. However, specific radiation-induced structural changes can be exploited for de novo phasing by an approach known as radiation damage-induced phasing (RIP). Here, RIP and single-wavelength anomalous dispersion (SAD) phasing were alternatively used to derive experimental phases to 1.2 A resolution for crystals of an alpha-helical 18-residue peptide, MINTS, which was derived from the neurotoxin apamin and the palladium-bound structure of which is now reported. Helix formation is induced by the binding of palladium (or copper) to two histidines spaced four residues apart, while two disulfide bonds tether the N-terminal helix to the C-terminal loop-like part of the peptide. Either RIP or SAD phasing of the palladium-bound and copper-bound forms of MINTS, which crystallized in different space groups, resulted in density maps of superb quality. Surprisingly, RIP phasing of the metal-bound complex structures of MINTS was a consequence of differential radiation damage, resting primarily on the reduction of the disulfide bonds in Pd-MINTS and on depletion of the metal sites in Cu-MINTS. Its miniprotein-like characteristics, versatile metal-binding properties and ease of crystallization suggest MINTS to be a convenient test specimen for methods development in crystallographic phasing based on either synchrotron or in-house X-ray diffraction data.


Subject(s)
Copper/chemistry , Lead/chemistry , Protein Structure, Secondary/drug effects , X-Ray Diffraction , Binding Sites , Crystallization , Crystallography, X-Ray , Macromolecular Substances , Mentha/chemistry , Models, Molecular , Radiation
20.
J Am Chem Soc ; 128(28): 9187-93, 2006 Jul 19.
Article in English | MEDLINE | ID: mdl-16834392

ABSTRACT

The design, synthesis, and characterization of a folded high-affinity metal-binding peptide is described. Based on the previously described folded peptide NTH-18, in which an alpha-helix was constrained through two disulfide bonds to a C-terminal extension of noncanonical secondary structure, a peptide (1) was designed to contain two histidine residues in positions 3 and 7. Air oxidation of 1 led to the formation of peptide 2, which contained two intramolecular disulfide bonds. The presence of the two histidines significantly destabilized the alpha-helical structure of 2 when compared to NTH-18. However, CD spectroscopy revealed that the addition of certain transition metal ions allowed the reformation of a stable alpha-helix. CD, NMR, and EPR spectroscopy as well as MALDI-TOF mass spectrometry indicated that 2 bound to Cu2+ to form a 1:1 complex via the imidazoles of the two histidine side chains. A glycine displacement assay revealed a dissociation constant for this complex of 5 nM at pH 8, which is the lowest reported value for a designed Cu2+-binding peptide. This peptide displayed more than 100-fold selectivity for Cu2+ over Zn2+, Ni2+, and Co2+. The 1.05 A crystal structure of the Cu(II)-complex of 2 revealed a square-pyramidal coordination geometry and confirmed that 2 bound to copper in an alpha-helical conformation via its two histidine side chains. The high affinity metal binding of peptide 2 demonstrates that metals can be used for the selective nucleation of alpha-helices.


Subject(s)
Copper/chemistry , Peptides/chemistry , Protein Folding , Models, Molecular , Protein Binding , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...