5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The protean prion protein

      review-article
      *
      PLoS Biology
      Public Library of Science

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The prion protein, PrP, can adopt at least 2 conformations, the overwhelmingly prevalent cellular conformation (PrP C) and the scrapie conformation (PrP Sc). PrP C features a globular C-terminal domain containing 3 α-helices and a short β-sheet and a long flexible N-terminal tail whose exact conformation in vivo is not yet known and a metastable subdomain with β-strand propensity has been identified within it. The PrP Sc conformation is very rare and has the characteristics of an amyloid. Furthermore, PrP Sc is a prion, i.e., it is infectious. This involves 2 steps: (1) PrP Sc can template PrP C and coerce it to adopt the PrP Sc conformation and (2) PrP Sc can be transmitted between individuals, by oral, parenteral, and other routes and thus propagate as an infectious agent. However, this is a simplification: On the one hand, PrP Sc is not a single conformation, but rather, a set of alternative similar but distinct conformations. Furthermore, other amyloid conformations of PrP exist with different biochemical and propagative properties. In this issue of PLOS Biology, Asante and colleagues describe the first murine model of familial human prion disease and demonstrate the emergence and propagation of 2 PrP amyloid conformers. Of these, one causes neurodegeneration, whereas the other does not. With its many conformers, PrP is a truly protean protein.

          Abstract

          This Primer explores the implications of a study that describes the first murine model of familial human prion disease, demonstrating the emergence and propagation of two PrP amyloid conformers; of these, one causes neurodegeneration while the other does not. With its many conformers, PrP is a truly protean protein.

          Related collections

          Most cited references17

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Prion neuropathology follows the accumulation of alternate prion protein isoforms after infective titre has peaked

          Prions are infectious agents causing lethal neurodegenerative diseases, such as Creutzfeldt–Jakob disease in humans, and scrapie and bovine spongiform encephalopathy in animals, following prolonged asymptomatic incubation periods1. In humans, these can span decades and are followed by a rapidly progressive, often stereotypic, clinical phase, which is invariably fatal1 2. In inbred lines of laboratory mice, incubation periods span months and yet are highly reproducible for a defined line with clinical onsets spanning only a few days. In studies of mice intracerebrally infected with the Rocky Mountain Laboratory (RML) prion strain, we have previously demonstrated that prion propagation in the brain involves two distinct phases3. In phase 1, prions propagate exponentially, not rate limited by cellular prion protein (PrPC) concentration, rapidly rising by ~106-fold to reach a maximal prion titre, which is also independent of PrPC concentration over the range we studied. This is followed by a plateau phase (phase 2), which determines time to clinical onset of disease, the duration of which is inversely proportional to PrPC concentration. Determination of prion titres at the beginning and end of the plateau in FVB/N mice by mouse end-point titration bioassay showed indistinguishable levels of infectivity3. Prion propagation and neurotoxicity can therefore be uncoupled, arguing that prions are not themselves directly neurotoxic. We hypothesized that once prion propagation saturates, there is a mechanistic switch from autocatalytic production of infectivity (phase 1) to a toxic pathway where prions act as a catalytic surface for the production of toxic species in a process now linearly dependent on PrPC concentration3. The rate of production of such toxic PrP species (previously designated PrPL for lethal4) would determine the time to exceed local toxic thresholds and subsequent clinical onset. According to the widely accepted protein-only hypothesis5, prions are thought to comprise multi-chain forms of misfolded, host-encoded, PrPC, referred to as PrPSc (refs 6, 7) and are now thought to propagate by an autocatalytic process of seeded fibrilization and fission8 9 10. It is increasingly recognized that such processes may be involved in the pathogenesis of commoner neurodegenerative conditions, including Alzheimer’s and Parkinson’s diseases11. The multi-chain forms of misfolded prion protein thought to comprise infectious prions have been generally referred to as PrPSc and originally defined by their relative protease-resistance (with characteristic band shift on western blotting due to deletion of an amino-terminal fragment following treatment with proteinase K (PK)) and detergent insolubility as compared with normal PrPC (refs 7, 12). However, it is becoming clear that there are multiple disease-related forms of PrP, some of which are protease sensitive13 14. Indeed, such protease-sensitive disease-related PrP may constitute the majority of infectivity in some prion isolates14. These species remain poorly defined in physical terms and an internationally agreed, even provisional, nomenclature is lacking. As PrPSc is by definition protease resistant, we do not favour the term protease-sensitive PrPSc (sPrPSc) (ref. 15). Here we simply use the term ‘PK-sensitive disease-related PrP’ to refer to the ensemble of PrP species seen in prion infection but which do not meet the biochemical definition of ‘classical’ PrPSc. In our previous studies of the kinetics of prion propagation, we determined the brain prion titre at defined intervals during the incubation period in four types of mice with different PrPC expression levels3. We have now proceeded to measure changes in disease-related PrP isoforms in the brain in mice in both the original3 and repeat time-course studies. We have developed an assay to measure total PrP. This must comprise PrPC and all disease-related isoforms (which are the sum of PK-sensitive and PK-resistant (classical PrPSc) isoforms). By subtraction of levels of classical PK-resistant PrPSc, any rise would represent the ensemble of PK-sensitive disease-related PrP, assuming PrPC levels are unchanged during infection. Prnp expression is known to be unaltered during prion infection16 17. A recent study reported an apparent fall in PrPC concentration towards the end of the incubation period18; if confirmed, this would suggest such a subtraction method would underestimate the rise in levels of PK-sensitive disease-related PrP late in phase 2. We have then sought to determine how changes in levels of disease-related isoforms related to the mechanistic phases we have described and to the development of neuropathology. Results Levels of total PrP and classical PrPSc during prion infection In our previous study3, we intracerebrally inoculated large groups of the following mice with RML mouse prions (30 μl 1% brain homogenate containing 105.8 intracerebral LD50 units) and groups of five to six mice were killed at multiple defined time points or at the onset of clinical disease: inbred FVB/N mice (Prnp +/+ ; wild-type PrPC expression level); Prnp-null mice19 (Prnp o/o; no PrPC expression; FVB/N background); hemizygous Prnp-null mice (Prnp +/o; 50% wild-type PrPC expression; FVB/N background) and Tg20 transgenic mice20 (~8-fold wild-type PrPC expression level; FVB/N background). Prion titre in mouse brain was determined by scrapie cell assay (SCA) or SCA in end-point format (scrapie cell endpoint assay (SCEPA)) for low-titre samples as previously reported21 (Fig. 1). Prions were undetectable in Prnp o/o mice, which are unable to propagate prions22, after 10 days, indicating clearance of the inoculum by this time and allowing interpretation of de novo produced prions in the other mouse lines3. In our earlier work, we had anticipated that prion titres would principally rise late in the incubation period such that relatively few timed culls were early in the incubation period3. We have since repeated the original study with many more timed culls, particularly in phase 1. The infectivity curves obtained were closely similar to our earlier results (Fig. 2). We then proceeded to measure changes in disease-related PrP isoforms in the brain in these mice to determine how these related to the mechanistic phases we have described and to the development of neuropathology. Although our ultimate aim is to isolate and structurally characterize the putative toxic PrP species (PrPL), at this stage we do not know whether toxicity is mediated by a single defined species of high specific toxicity, or, at the other extreme, relates to an ensemble of diverse species with a generic toxicity. It appears that infectious prion strains constitute a cloud or quasispecies10 23 24, and it is possible that infectious and toxic PrP populations overlap. Given these uncertainties and the lack of physical characterization of disease-related PrP species, we modified for our purposes an enzyme-linked immunosorbent assay (ELISA) that is capable of detecting all PrP chains following their denaturation with heat and SDS14 25 26. This assay therefore provides a measure of total PrP present in brain homogenates and must necessarily comprise the sum of normal PrPC and disease-related PrP generated during the infection. The latter would comprise both classical PK-resistant PrPSc and PK-sensitive disease-related PrP isoforms13 14 26. As Prnp expression is unaltered16 17 and PrPC concentration does not rise18 during prion infection, the rise in total PrP represents the rise in levels of total disease-related PrP (including PK-resistant and PK-sensitive forms). Levels of classical PK-resistant PrPSc were also determined, thus enabling estimation of PK-sensitive disease-related PrP isoform levels during the incubation period (Fig. 1). Remarkably, total PrP levels, although starting at different levels in the three mouse lines due to their different PrPC expression levels, were essentially unchanged throughout phase 1 in wild-type, Prnp +/o and Tg20 mice, a period during which titres rose in each case by ~106-fold (Fig. 1). When prion titre had reached plateau (marking the start of phase 2), total PrP levels then rose in a linear manner throughout phase 2 with a gradient approximately proportional to PrPC expression level (Fig. 1). In agreement with previous studies, classical PrPSc represents only a small fraction of total PrP levels at onset of clinical disease and the majority of this is detected towards the end of phase 2 (Figs 1 and 3). Notably, in all three lines of mice, clinical onset appears to ensue only once a closely similar level of total disease-related PrP in the brain is reached (Figs 1 and 3). These data suggest that a particular PrP isoform(s) that is generated in phase 2 must reach a critical threshold level to elicit neurotoxic effects, irrespective of the level of PrPC expressed in the brain. Correlation with onset of neuropathological changes We then sought to correlate changes in prion titre and disease-related PrP isoforms with onset of neuropathological changes. To allow a systematic neuropathological analysis, we repeated the original time course using only the wild-type FVB mice and performed timed culls with a considerably increased number of time points (at 5-day intervals from post-inoculation day 5 to day 90, then at days 125 and 140) to prepare suitable fixed brain tissues for histology and immunohistochemistry. All neuropathological scoring was performed blind to sample identity. Prion titres were also determined again at the same time points to ensure comparability with the original experiment (Fig. 3). Prion titre had reached plateau level by day 75. Spongiform vacuolation of the neuropil is a key specific histological characteristic of prion disease. It appears to correlate with early disease activity and indeed early spongiosis is completely reversible on targeted knockout of PrPC expression in neuroinvasive RML prion infection of FVB mice27. Uniform (12/12 mice), albeit mild, spongiosis was only seen at post-infection day 85 and was restricted to the thalamus and hippocampus. As expected for the RML prion strain, spongiosis then extended to the basal ganglia by day 90 and had become of moderate severity and involved virtually all grey matter areas by day 140 (Figs 4 and 5). No spongiform vacuolation was detected in any mice before post-infection day 65 when small subtle vacuoles in the hippocampus and thalamus were seen in 4/12 animals on haematoxylin and eosin-stained sections (Fig. 5). The uniform appearance of specific spongiform pathology only in phase 2 is consistent with a lack of direct neurotoxicity of infectious prions and the hypothesized production of toxic species via a distinct pathway once prion propagation has saturated. PrP immunohistochemistry using anti-PrP monoclonal antibody ICSM35 revealed evidence of residual inoculum in some animals from day 5 to 30, but without synaptic PrP deposition that is characteristic of prion infection. Very subtle occasional synaptic deposits28 were seen in some mice from day 35 to 50; consistent deposits were seen in the thalamus and cortex in all mice from day 55 (Fig. 5). Whether these subtle PrP deposits seen in phase 1 simply represent infectious prions is unknown. Microglia were assessed using antibody Iba1 and the earliest reproducible activation was seen at day 35 in the thalamus in some animals. Activation was however well established in all animals by day 65. Synaptophysin immunoreactivity was assessed to detect synaptic loss. The first clear reduction of synaptic density and granular normality was detected at day 125 (Fig. 5). Discussion The basis of neurotoxicity in prion neurodegeneration, its relationship to the prolonged clinically silent incubation periods and the remarkable synchronicity of clinical onset have remained obscure. Although classical PrPSc rises towards the end of the incubation period and has been considered a candidate neurotoxic species, the well-documented existence of subclinical carrier states of prion infection, where conventional mice with normal PrPC expression live a typical lifespan despite harbouring levels of classical PrPSc and prion titres similar to those of mice with end-stage clinical disease4 29 30 31 32 33, argues against this. These findings are consistent with the general model of prion propagation and neurotoxicity in which prion infectivity and toxicity are mediated by different PrP species, which are the product of distinct pathways3 10. According to this model, production of neurotoxic species is triggered when the exponential phase (phase 1) of prion propagation saturates (following depletion of a key co-factor or occupancy of available replication sites). The fact that this explosive rise in prion titre is not rate limited by PrPC concentration is consistent with the absence of a rise of total PrP during phase 1 arguing that infectious prions comprise a small minority of available PrP. In phase 2, there is a linear rise in PK-sensitive disease-related PrP isoforms, a mechanistically distinct process rate limited by, and directly proportional to, PrPC concentration. Clinical onset occurs at a similar level of PK-sensitive disease-related PrP isoforms irrespective of PrPC expression level (Fig. 6). The classical neuropathological features of prion disease, indicative of the presence of neurotoxic species, are only detected once phase 2 is well established. The precise temporal relationship between production of such neurotoxic species, reaching critical concentrations at critical sites, and onset of neuropathology cannot be defined by these experiments, and will ultimately require isolation of PrPL species and direct studies of neurotoxicity. However, such isolation and physical characterization of neurotoxic species, as with other neurodegenerative diseases, is likely to prove technically demanding. Such species may be transitory and inherently labile. In addition, as discussed above, it is unknown whether the majority of the PK-sensitive disease-related PrP isoforms constitute generic toxic material or whether at the other extreme a low abundance specific sub-species of defined structure and receptor interaction with high specific toxicity constitutes the hypothetical PrPL. An essential prerequisite will be the development of a robust in vitro assay for such neurotoxic activity with a sufficient dynamic range to facilitate fractionation and isolation of such species. These data also suggest that the term PrPSc, often used synonymously with infectivity, should be restricted to material as classically biochemically defined7 12. Such classical PrPSc only constitutes a small (variable) fraction of total disease-related PrP isoforms and its proportional contribution to both infectivity and neurotoxicity remains unclear. All the common neurodegenerative diseases involve accumulation of aggregated misfolded host proteins and it is increasingly proposed that seeded protein aggregation (or ‘prion-like mechanisms’) may be involved in their pathogenesis and/or spread of pathology throughout the neuraxis. Experimental prion disease offers major advantages in studying neurodegenerative processes, because prion diseases naturally affect many mammalian species, including laboratory rodents, avoiding the inherent limitations of mouse models of other neurodegenerative diseases and, crucially, because of the predictable incubation periods and tools established to determine prion strain properties and accurately measure titre1. If comparable tools can be developed for the commoner neurodegenerative conditions such as Alzheimer’s and Parkinson’s diseases, it will be interesting to investigate whether similar mechanistic relationships between seeding activity and neurotoxicity are also involved. Methods All experimental procedures involving prions were carried out in microbiological containment level 3 facilities with strict adherence to safety protocols. Prion inoculation of mice Work with mice was performed under licence granted by the UK Home Office and conformed to the University College London institutional and ARRIVE guidelines. RML prion inoculum (I6200) was prepared and titrated14, and mice (female 6–9 weeks old) were intracerebrally inoculated and monitored as described previously3. Groups of 5–12 mice were killed at multiple defined time points or at onset of clinical disease. Brains were removed and divided sagittally with half-frozen at −70 °C and half-fixed in 10% (v/v) formal buffered saline. Brain homogenates (10% (w/v)) were prepared from frozen specimens in sterile Dulbecco’s PBS lacking Ca2+ and Mg2+ ions (D-PBS) and stored as aliquots at −70 °C. Prion bioassay PK1 cells (sub-clone PK1/2), a line derived from N2a cells that are highly susceptible to RML prions21 were routinely grown into OFCS medium (Opti-MEM, containing 10% FCS; 100 U ml−1 penicillin and 100 μg ml−1 streptomycin; Invitrogen, UK) using 15 cm Petri dishes. Prion infectivity was assayed using two modified automated protocols based on the original published methods21, the SCEPA and the automated SCA (ASCA). The SCEPA is capable of detecting sample infectivity in a range down to ~103 tissue culture infectious units (TCIU)  ml−1 of 10% (w/v) brain homogenate21, while the ASCA is typically used to detect infectivity above 104.5 TCIU ml−1 in 10% (w/v) brain homogenate. The day before infection, PK1/2 cells were seeded into 96-well plates (18,000 cells in 260 μl OFCS; Costar flat bottom 96-well plates; Corning, UK) and kept at 37 °C in a 5% CO2 incubator. The following day, 10 μl of brain homogenate appropriately diluted in OFCS was applied to the cells. A serial dilution of a reference 10% (w/v) RML brain homogenate (maximal titre of 107.7 TCIU ml−1 determined by SCEPA) was applied in parallel. Typically, 10% (w/v) brain homogenate was used in the range of 1 × 10−3 to 1 × 10−5 dilution for SCEPA and 3 × 10−4 to 3 × 10−6 dilution for ASCA. Three days after infection, an automated platform (Biomek FX liquid handling robot; Beckman Coulter) was used to split cells. For the SCEPA protocol, cells were split into 96-well plates containing fresh OFCS at ratios of 1:3 on days 3, 5 and 7, followed by splitting 1:8 at days 10, 13, 17 and 21 post infection, while the ASCA was split at a ratio of 1:8 on days 3, 6, 10 and 14 post infection. ELISpot plates (96-well– polyvinylidene difluoride membrane plates; Millipore, UK) were prepared from the SCEPA assays on days 17 and 21 post infection and on days 10 and 14 post infection for the ASCA assays (25,000 cells per well). The plates were vacuum drained and dried at 50 °C after which 60 μl of 1 μg ml−1 PK (Roche, UK) in lysis buffer (50 mM Tris HCl, pH 8, containing 150 mM NaCl, 0.5% (w/v) sodium deoxycholate and 0.5% (v/v) Triton X-100) was added to each well and incubated for 60 min at 40 °C. The plates were washed (2 × 160 μl PBS) after which 120 μl of 3 M guanidinium thiocyanate prepared in 10 mM Tris HCl, pH 8.0, was added to each well for 20 min. The wells were washed (7 × 160 μl PBS) and 150 μl of Superblock dry blend blocking buffer (Perbio, UK) was added to each well and incubated for 1 h. Following vacuum removal of Superblock, each well was incubated with 0.55 μg ml−1 anti-PrP monoclonal antibody ICSM18 (D-Gen Ltd, London) prepared in TBST (10 mM Tris HCl, pH 8, 150 mM NaCl, 0.1% (v/v) Tween 20) containing 1% (w/v) non-fat dry milk for 1 h. After washing (5 × 160 μl TBST), wells were incubated for 1 h with 60 μl goat anti-mouse alkaline phosphatase-conjugated anti-IgG1 (Southern Biotechnology Associates, USA) diluted 1:8,000 in TBST-1% (w/v) non-fat dry milk. Following washing (5 × 160 μl TBST), wells were incubated for 35 min with 50 μl AP dye (Bio-Rad, USA). The plates were then washed twice with water, dried and stored at −20 °C. Spot counts (reporting PK-resistant PrP-positive cells) were determined with a Zeiss KS Elispot system (Stemi 2000-C stereo microscope equipped with a Hitachi HV-C20A colour camera, a KL 1,500 LCD scanner and Wellscan software from Imaging Associates, Oxfordshire, UK). Normal interval regression was used to calculate mean and s.e.m. for the groups that contained points below assay sensitivity cut-off. If all samples in a timed group were below assay sensitivity cut-off, the group was excluded from further analyses. ELISA for total PrP PrP levels in brain homogenate were determined by ELISA as previously described14 26, but with adaptations. A pooled preparation of 10% (w/v) terminal RML mouse brain homogenate (I8700) of known prion infectivity titre26 was diluted into D-PBS. These samples were used to generate standard curves of fluorescence versus I8700 concentration, and were processed in parallel with unknown samples. PrP levels in unknown samples were expressed relative to the I8700 reference preparation. The protein concentration of 10% (w/v) brain homogenate was determined using the Bicinchoninic acid assay kit (Thermo Scientific, Rockford, IL, USA) according to the manufacturer’s instructions and aliquots subsequently adjusted with D-PBS to give a final protein concentration of 7 mg ml−1. Twenty-microlitre aliquots of these samples were treated with an equal volume of either D-PBS alone or D-PBS containing 50 μg ml−1 PK and incubated at 37 °C for 1 h. This concentration of PK (at a ratio of 1:140 total brain protein by weight) was used to ensure complete digestion of PrPC in overexpressing Tg20 transgenic mice and to retain only the most PK-resistant abnormal PrP conformers classically designated as PrPSc 7. After addition of 1 μl 100 mM 4-(2-aminoethyl)-benzenesulphonyl fluoride prepared in water and 1 μl 20% (w/v) SDS prepared in water, the samples were heated at 100 °C for 10 min. Twenty microlitres of the denatured samples were then diluted into ELISA capture buffer (50 mM Tris/HCl, pH 8.4, containing 2% (v/v) Triton X-100, 2% (w/v) sodium lauroylsarcosine and 2% (w/v) BSA (fraction V, protease free, Sigma)) between 30 and 150 times for analysis. The use of multiple dilutions ensured that fluorescence measurements generated by unknown samples could be correlated with the linear range of the standard curves of I8700. Microtitre plates (Microlon 96W, Greiner Bio-One) were coated overnight with anti-PrP monoclonal antibody ICSM18 (D-Gen Ltd) diluted in carbonate coating buffer (250 ng antibody per well). The wells were blocked using Superblock T20 (PBS) blocking buffer (Thermo Scientific) and incubated with constant agitation for 1 h at 37 °C. Aliquots (50 μl) of the diluted samples were transferred into the wells of the plates and incubated with constant agitation at 37 °C for 1 h. Wells were washed with 3 × 300 μl of PBS containing 0.05% (v/v) Tween 20 using an automated microplate washer, followed by the addition of 100 μl of PBS containing 1% (v/v) Tween 20 and 1 μg ml−1 biotinylated anti-PrP monoclonal antibody ICSM 35 (D-Gen Ltd). Following incubation at 37 °C for 15 min with constant agitation, wells were washed as detailed above, followed by the addition of 100 μl of PBS containing 1% (v/v) Tween 20 and a dilution of streptavidin–horseradish-peroxidase conjugate (1:10,000 dilution; Thermo Scientific). After incubation at 37 °C for 15 min with constant agitation, wells were washed with 4 × 300 μl of PBS containing 0.05% (v/v) Tween 20. Wells were developed using QuantaBlu Fluorogenic Peroxidase substrate kit according to the manufacturer’s instructions (Thermo Scientific). Fluorescence was measured on a Tecan infinite M200 spectra image microplate reader (λex=313 nm, λem=398 nm). Neuropathology and immunohistochemistry Brain fixed in 10% buffered formol-saline was immersed in 98% formic acid for 1 hour and paraffin wax embedded. Serial sections (4-μm-thick) were pretreated by boiling for 10 min in a low ionic strength buffer (2.1 mM Tris, 1.3 mM EDTA, 1.1 mM sodium citrate, pH 7.8) before exposure to 98% formic acid for 5 min. Abnormal PrP accumulation was examined using anti-PrP monoclonal antibody ICSM 35 (D-Gen Ltd). Microglial activation was examined using an ionized calcium binding adaptor molecule 1 (Iba1) antibody (Wako Pure Chemical Industries, Ltd) and synaptophysin was examined using rabbit polyclonal antibodies (Z66, Life Technologies). All immunohistochemistry was performed on automated immunohistochemistry staining machines (Ventana Medical Systems Inc., Tucson, Arizona) using proprietary secondary detection reagents (Ventana Medical Systems Inc.) before development with 3′3-diaminobenzedine tetrachloride as the chromogen34. Conventional methods were used for Harris haematoxylin and eosin staining. Appropriate positive and negative controls were used throughout. Photographs were taken on an ImageView digital camera and composed with Adobe Photoshop. Author contributions M.K.S., H.A.-D., B.S., M.W.D.O., A.R.-L., S.L. and C.S. performed the work. J.M.L. and S.B analysed the neuropathology. M.K.S., A.R.C., J.D.F.W. and J.C. designed the study and analysed the data. J.C. drafted the manuscript. All authors discussed the results and commented on the manuscript. Additional information How to cite this article: Sandberg, M. K. et al. Prion neuropathology follows the accumulation of alternate prion protein isoforms after infective titre has peaked. Nat. Commun. 5:4347 doi: 10.1038/ncomms5347 (2014).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Parallel in-register intermolecular β-sheet architectures for prion-seeded prion protein (PrP) amyloids.

            Structures of the infectious form of prion protein (e.g. PrP(Sc) or PrP-Scrapie) remain poorly defined. The prevalent structural models of PrP(Sc) retain most of the native α-helices of the normal, noninfectious prion protein, cellular prion protein (PrP(C)), but evidence is accumulating that these helices are absent in PrP(Sc) amyloid. Moreover, recombinant PrP(C) can form amyloid fibrils in vitro that have parallel in-register intermolecular β-sheet architectures in the domains originally occupied by helices 2 and 3. Here, we provide solid-state NMR evidence that the latter is also true of initially prion-seeded recombinant PrP amyloids formed in the absence of denaturants. These results, in the context of a primarily β-sheet structure, led us to build detailed models of PrP amyloid based on parallel in-register architectures, fibrillar shapes and dimensions, and other available experimentally derived conformational constraints. Molecular dynamics simulations of PrP(90-231) octameric segments suggested that such linear fibrils, which are consistent with many features of PrP(Sc) fibrils, can have stable parallel in-register β-sheet cores. These simulations revealed that the C-terminal residues ∼124-227 more readily adopt stable tightly packed structures than the N-terminal residues ∼90-123 in the absence of cofactors. Variations in the placement of turns and loops that link the β-sheets could give rise to distinct prion strains capable of faithful template-driven propagation. Moreover, our modeling suggests that single PrP monomers can comprise the entire cross-section of fibrils that have previously been assumed to be pairs of laterally associated protofilaments. Together, these insights provide a new basis for deciphering mammalian prion structures. © 2014 by The American Society for Biochemistry and Molecular Biology, Inc.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Accumulation of prion protein in the brain that is not associated with transmissible disease.

              Prion diseases or transmissible spongiform encephalopathies are characterized histopathologically by the accumulation of prion protein (PrP) ranging from diffuse deposits to amyloid plaques. Moreover, pathologic PrP isoforms (PrP(Sc)) are detected by immunoblot analysis and used both as diagnostic markers of disease and as indicators of the presence of infectivity in tissues. It is not known which forms of PrP are associated with infectivity. To address this question, we performed bioassays using human brain extracts from two cases with phenotypically distinct forms of familial prion disease (Gerstmann-Sträussler-Scheinker P102L). Both cases had PrP accumulations in the brain, but each had different PrP(Sc) isoforms. Only one of the brains had spongiform degeneration. Tissue from this case transmitted disease efficiently to transgenic mice (Tg PrP101LL), resulting in spongiform encephalopathy. In contrast, inoculation of tissue from the case with no spongiform degeneration resulted in almost complete absence of disease transmission but elicited striking PrP-amyloid deposition in several recipient mouse brains. Brains of these mice failed to transmit any neurological disease on passage, but PrP-amyloid deposition was again observed in the brains of recipient mice. These data suggest the possible isolation of an infectious agent that promotes PrP amyloidogenesis in the absence of a spongiform encephalopathy. Alternatively, the infectious agent may be rendered nonpathogenic by sequestration in amyloid plaques, or PrP amyloid can seed amyloid accumulation in the brain, causing a proteinopathy that is unrelated to prion disease. Formation of PrP amyloid may therefore not necessarily be a reliable marker of transmissible spongiform encephalopathy infectivity.
                Bookmark

                Author and article information

                Journal
                PLoS Biol
                PLoS Biol
                plos
                plosbiol
                PLoS Biology
                Public Library of Science (San Francisco, CA USA )
                1544-9173
                1545-7885
                25 June 2020
                June 2020
                25 June 2020
                : 18
                : 6
                : e3000754
                Affiliations
                [001]CIMUS Biomedical Research Institute & Department of Medical Sciences, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
                Author notes

                The author has declared that no competing interests exist.

                Author information
                http://orcid.org/0000-0003-1954-4975
                Article
                PBIOLOGY-D-20-01027
                10.1371/journal.pbio.3000754
                7316225
                32584805
                09265a73-09d3-4748-9b40-2471b6cad9bf
                © 2020 Jesús R. Requena

                This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                Page count
                Figures: 2, Tables: 0, Pages: 8
                Funding
                This work was funded by the Spanish Ministry of Industry and Competitiveness (grant BFU2017-86692-P), partially supported with EU FEDER funds. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Primer
                Biology and Life Sciences
                Zoology
                Animal Diseases
                Animal Prion Diseases
                Medicine and Health Sciences
                Infectious Diseases
                Prion Diseases
                Animal Prion Diseases
                Medicine and Health Sciences
                Infectious Diseases
                Zoonoses
                Prion Diseases
                Animal Prion Diseases
                Medicine and Health Sciences
                Infectious Diseases
                Prion Diseases
                Medicine and Health Sciences
                Infectious Diseases
                Zoonoses
                Prion Diseases
                Research and Analysis Methods
                Animal Studies
                Experimental Organism Systems
                Model Organisms
                Mouse Models
                Research and Analysis Methods
                Model Organisms
                Mouse Models
                Research and Analysis Methods
                Animal Studies
                Experimental Organism Systems
                Animal Models
                Mouse Models
                Biology and Life Sciences
                Zoology
                Animal Diseases
                Animal Prion Diseases
                Scrapie
                Medicine and Health Sciences
                Infectious Diseases
                Prion Diseases
                Animal Prion Diseases
                Scrapie
                Medicine and Health Sciences
                Infectious Diseases
                Zoonoses
                Prion Diseases
                Animal Prion Diseases
                Scrapie
                Medicine and Health Sciences
                Neurology
                Brain Diseases
                Medicine and Health Sciences
                Neurology
                Neurodegenerative Diseases
                Biology and Life Sciences
                Biochemistry
                Proteins
                Amyloid Proteins
                Research and Analysis Methods
                Animal Studies
                Animal Models of Disease

                Life sciences
                Life sciences

                Comments

                Comment on this article