首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Prion disease research has opened up the “black-box” of neurodegeneration, defining a key role for protein misfolding wherein a predominantly alpha-helical precursor protein, PrPC, is converted to a disease-associated, β-sheet enriched isoform called PrPSc. In Alzheimer disease (AD) the Aβ peptide derived from the β-amyloid precuror protein APP folds in β-sheet amyloid. Early thoughts along the lines of overlap may have been on target,1 but were eclipsed by a simultaneous (but now anachronistic) controversy over the role of PrPSc in prion diseases.2,3 Nonetheless, as prion diseases such as Creutzfeldt-Jakob Disease (CJD) are themselves rare and can include an overt infectious mode of transmission, and as familial prion diseases and familial AD involve different genes, an observer might reasonably have concluded that prion research could occasionally catalyze ideas in AD, but could never provide concrete overlaps at the mechanistic level. Surprisingly, albeit a decade or three down the road, several prion/AD commonalities can be found within the contemporary literature. One important prion/AD overlap concerns seeded spread of Aβ aggregates by intracerebral inoculation much like prions,4 and, with a neuron-to-neuron ‘spreading’ also reported for pathologic forms of other misfolded proteins, Tau5,6 and α-synuclein in the case of Parkinson Disease.7,8 The concept of seeded spread has been discussed extensively elsewhere, sometimes under the rubric of “prionoids”9, and lies outside the scope of this particular review where we will focus upon PrPC. From this point the story can now be subdivided into four strands of investigation: (1) pathologic effects of Aβ can be mediated by binding to PrPC,10 (2) the positioning of endoproteolytic processing events of APP by pathologic (β-cleavage + γ-cleavage) and non-pathologic (α-cleavage + γ-cleavage) secretase pathways is paralleled by seemingly analogous α- and β-like cleavage of PrPC (Fig. 1) (3) similar lipid raft environments for PrPC and APP processing machinery,11-13 and perhaps in consequence, overlaps in repertoire of the PrPC and APP protein interactors (“interactomes”),14,15 and (4) rare kindreds with mixed AD and prion pathologies.16 Here we discuss confounds, consensus and conflict associated with parameters that apply to these experimental settings.  相似文献   

2.
The conversion of a cellular prion protein (PrPC) to its pathogenic isoform (PrPSc) is a critical event in the pathogenesis of prion diseases. Pathogenic conversion is usually associated with the oligomerization process; therefore, the conformational characteristics of the pre-oligomer state may provide insights into the conversion process. Previous studies indicate that PrPC is prone to oligomer formation at low pH, but the conformation of the pre-oligomer state remains unknown. In this study, we systematically analyzed the acid-induced conformational changes of PrPC and discovered a unique acid-induced molten globule state at pH 2.0 termed the “A-state.” We characterized the structure of the A-state using far/near-UV CD, 1-anilino-8-naphthalene sulfonate fluorescence, size exclusion chromatography, and NMR. Deuterium exchange experiments with NMR detection revealed its first unique structure ever reported thus far; i.e. the Strand 1-Helix 1-Strand 2 segment at the N terminus was preferentially unfolded, whereas the Helix 2-Helix 3 segment at the C terminus remained marginally stable. This conformational change could be triggered by the protonation of Asp144, Asp147, and Glu196, followed by disruption of key salt bridges in PrPC. Moreover, the initial population of the A-state at low pH (pH 2.0–5.0) was well correlated with the rate of the β-rich oligomer formation, suggesting that the A-state is the pre-oligomer state. Thus, the specific conformation of the A-state would provide crucial insights into the mechanisms of oligomerization and further pathogenic conversion as well as facilitating the design of novel medical chaperones for treating prion diseases.  相似文献   

3.
The conformational conversion of the cellular prion protein (PrPC) to the β-rich infectious isoform PrPSc is considered a critical and central feature in prion pathology. Although PrPSc is the critical component of the infectious agent, as proposed in the “protein-only” prion hypothesis, cellular components have been identified as important cofactors in triggering and enhancing the conversion of PrPC to proteinase K resistant PrPSc. A number of in vitro systems using various chemical and/or physical agents such as guanidine hydrochloride, urea, SDS, high temperature, and low pH, have been developed that cause PrPC conversion, their amplification, and amyloid fibril formation often under non-physiological conditions. In our ongoing efforts to look for endogenous and exogenous chemical mediators that might initiate, influence, or result in the natural conversion of PrPC to PrPSc, we discovered that lipopolysaccharide (LPS), a component of gram-negative bacterial membranes interacts with recombinant prion proteins and induces conversion to an isoform richer in β sheet at near physiological conditions as long as the LPS concentration remains above the critical micelle concentration (CMC). More significant was the LPS mediated conversion that was observed even at sub-molar ratios of LPS to recombinant ShPrP (90–232).  相似文献   

4.
Each known abnormal prion protein (PrPSc) is considered to have a specific range and therefore the ability to infect some species and not others. Consequently, some species have been assumed to be prion disease resistant as no successful natural or experimental challenge infections have been reported. This assumption suggested that, independent of the virulence of the PrPSc strain, normal prion protein (PrPC) from these ‘resistant’ species could not be induced to misfold. Numerous in vitro and in vivo studies trying to corroborate the unique properties of PrPSc have been undertaken. The results presented in the article “Rabbits are not resistant to prion infection” demonstrated that normal rabbit PrPC, which was considered to be resistant to prion disease, can be misfolded to PrPSc and subsequently used to infect and transmit a standard prion disease to leporids. Using the concept of species resistance to prion disease, we will discuss the mistake of attributing species specific prion disease resistance based purely on the absence of natural cases and incomplete in vivo challenges. The BSE epidemic was partially due to an underestimation of species barriers. To repeat this error would be unacceptable, especially if present knowledge and techniques can show a theoretical risk. Now that the myth of prion disease resistance has been refuted it is time to re-evaluate, using the new powerful tools available in modern prion laboratories, whether any other species could be at risk.  相似文献   

5.
While the conversion of PrPC into PrPSc in the transmissible form of prion disease requires a preexisting PrPSc seed, in genetic prion disease accumulation of disease related PrP could be associated with biochemical and metabolic modifications resulting from the designated PrP mutation. To investigate this possibility, we looked into the time related changes of PrP proteins in the brains of TgMHu2ME199K/wt mice, a line modeling for heterozygous genetic prion disease linked to the E200K PrP mutation. We found that while oligomeric entities of mutant E199KPrP exist at all ages, aggregates of wt PrP in the same brains presented only in advanced disease, indicating a late onset conversion process. We also show that most PK resistant PrP in TgMHu2ME199K mice is soluble and truncated (PrPST), a pathogenic form never before associated with prion disease. We next looked into brain samples from E200K patients and found that both PK resistant PrPs, PrPST as in TgMHu2ME199K mice, and “classical” PrPSc as in infectious prion diseases, coincide in the patient''s post mortem brains. We hypothesize that aberrant metabolism of mutant PrPs may result in the formation of previously unknown forms of the prion protein and that these may be central for the fatal outcome of the genetic prion condition.  相似文献   

6.
Different transmissible spongiform encephalopathy (TSE)-associated forms of prion protein (e.g. PrPSc) can vary markedly in ultrastructure and biochemical characteristics, but each is propagated in the host. PrPSc propagation involves conversion from its normal isoform, PrPC, by a seeded or templated polymerization mechanism. Such a mechanism is also the basis of the RT-QuIC and eQuIC prion assays which use recombinant PrP (rPrPSen) as a substrate. These ultrasensitive detection assays have been developed for TSE prions of several host species and sample tissues, but not for murine models which are central to TSE pathogenesis research. Here we have adapted RT-QuIC and eQuIC to various murine prions and evaluated how seeding activity depends on glycophosphatidylinositol (GPI) anchoring and the abundance of amyloid plaques and protease-resistant PrPSc (PrPRes). Scrapie brain dilutions up to 10−8 and 10−13 were detected by RT-QuIC and eQuIC, respectively. Comparisons of scrapie-affected wild-type mice and transgenic mice expressing GPI anchorless PrP showed that, although similar concentrations of seeding activity accumulated in brain, the heavily amyloid-laden anchorless mouse tissue seeded more rapid reactions. Next we compared seeding activities in the brains of mice with similar infectivity titers, but widely divergent PrPRes levels. For this purpose we compared the 263K and 139A scrapie strains in transgenic mice expressing P101L PrPC. Although the brains of 263K-affected mice had little immunoblot-detectable PrPRes, RT-QuIC indicated that seeding activity was comparable to that associated with a high-PrPRes strain, 139A. Thus, in this comparison, RT-QuIC seeding activity correlated more closely with infectivity than with PrPRes levels. We also found that eQuIC, which incorporates a PrPSc immunoprecipitation step, detected seeding activity in plasma from wild-type and anchorless PrP transgenic mice inoculated with 22L, 79A and/or RML scrapie strains. Overall, we conclude that these new mouse-adapted prion seeding assays detect diverse types of PrPSc.  相似文献   

7.
The prion protein (PrP) is implicated in the Transmissible Spongiform Encephalopathies (TSEs), which comprise a group of fatal neurodegenerative diseases affecting humans and other mammals. Conversion of cellular PrP (PrPC) into the scrapie form (PrPSc) is the hallmark of TSEs. Once formed, PrPSc aggregates and catalyzes PrPC misfolding into new PrPSc molecules. Although many compounds have been shown to inhibit the conversion process, so far there is no effective therapy for TSEs. Besides, most of the previously evaluated compounds failed in vivo due to poor pharmacokinetic profiles. In this work we propose a combined in vitro/in silico approach to screen for active anti-prion compounds presenting acceptable drugability and pharmacokinetic parameters. A diverse panel of aromatic compounds was screened in neuroblastoma cells persistently infected with PrPSc (ScN2a) for their ability to inhibit PK-resistant PrP (PrPRes) accumulation. From ∼200 compounds, 47 were effective in decreasing the accumulation of PrPRes in ScN2a cells. Pharmacokinetic and physicochemical properties were predicted in silico, allowing us to obtain estimates of relative blood brain barrier permeation and mutagenicity. MTT reduction assays showed that most of the active compounds were non cytotoxic. Compounds that cleared PrPRes from ScN2a cells, were non-toxic in the MTT assay, and presented a good pharmacokinetic profile were investigated for their ability to inhibit aggregation of an amyloidogenic PrP peptide fragment (PrP109–149). Molecular docking results provided structural models and binding affinities for the interaction between PrP and the most promising compounds. In summary, using this combined in vitro/in silico approach we have identified new small organic anti-scrapie compounds that decrease the accumulation of PrPRes in ScN2a cells, inhibit the aggregation of a PrP peptide, and possess pharmacokinetic characteristics that support their drugability. These compounds are attractive candidates for prion disease therapy.  相似文献   

8.

Background

Transmissible agents involved in prion diseases differ in their capacities to target different regions of the central nervous system and lymphoid tissues, which are also host-dependent.

Methodology/Principal Findings

Protease-resistant prion protein (PrPres) was analysed by Western blot in the spleen of transgenic mice (TgOvPrP4) that express the ovine prion protein under the control of the neuron-specific enolase promoter, after infection by intra-cerebral route with a variety of transmissible spongiform encephalopathies (TSEs) from cattle and small ruminants. Splenic PrPres was consistently detected in classical BSE and in most natural scrapie sources, the electrophoretic pattern showing similar features to that of cerebral PrPres. However splenic PrPres was not detected in L-type BSE and TME-in-cattle, or in the CH1641 experimental scrapie isolate, indicating that some TSE strains showed reduced splenotropism in the ovine transgenic mice. In contrast with CH1641, PrPres was also consistently detected in the spleen of mice infected with six natural “CH1641-like” scrapie isolates, but then showed clearly different molecular features from those identified in the brains (unglycosylated PrPres at ∼18 kDa with removal of the 12B2 epitope) of ovine transgenic mice or of sheep. These features included different cleavage of the main PrPres cleavage product (unglycosylated PrPres at ∼19 kDa with preservation of the 12B2 epitope) and absence of the additional C-terminally cleaved PrPres product (unglycosylated form at ∼14 kDa) that was detected in the brain.

Conclusion/Significance

Studies in a transgenic mouse model expressing the sheep prion protein revealed different capacities of ruminant prions to propagate in the spleen. They showed unexpected features in “CH1641-like” ovine scrapie suggesting that such isolates contain mixed conformers with distinct capacities to propagate in the brain or lymphoid tissues of these mice.  相似文献   

9.
Increasing evidence suggests that proteins exhibiting “prion-like” behavior cause distinct neurodegenerative diseases, including inherited, sporadic and acquired types. The conversion of cellular prion protein (PrPC) to its infectious protease resistant counterpart (PrPRes) is the essential feature of prion diseases. However, PrPC also performs important functions in transmembrane signaling, especially in neurodegenerative processes. Beta-amyloid (Aβ) synaptotoxicity and cognitive dysfunction in mouse models of Alzheimer disease are mediated by a PrPC-dependent pathway. Here we review how this pathway converges with proinflammatory cytokine signaling to activate membrane NADPH oxidase (NOX) and generate reactive oxygen species (ROS) leading to dynamic remodeling of the actin cytoskeleton. The NOX signaling pathway may also be integrated with those of other transmembrane receptors clustered in PrPC-enriched membrane domains. Such a signal convergence along the PrPC-NOX axis could explain the relevance of PrPC in a broad spectrum of neurodegenerative disorders, including neuroinflammatory-mediated alterations in synaptic function following traumatic brain injury. PrPC overexpression alone activates NOX and generates a local increase in ROS that initiates cofilin activation and formation of cofilin-saturated actin bundles (rods). Rods sequester cofilin from synaptic regions where it is required for plasticity associated with learning and memory. Rods can also interrupt vesicular transport by occluding the neurite within which they form. Through either or both mechanisms, rods may directly mediate the synaptic dysfunction that accompanies various neurodegenerative disorders.  相似文献   

10.
The development of antibodies effective in crossing the blood brain barrier (BBB), capable of accessing the cytosol of affected cells and with higher affinity for PrPSc would be of paramount importance in arresting disease progression in its late stage and treating individuals with prion diseases. Antibody-based therapy appears to be the most promising approach following the exciting report from White and colleagues, establishing the “proof-of-principle” for prion-immunotherapy. After passive transfer, anti-prion antibodies were shown to be very effective in curing peripheral but not central rodent prion disease, due to the fact that these anti-prion antibodies are relatively large molecules and cannot therefore cross the BBB. Here, we show that an anti-prion antibody derived from camel immunised with murine scrapie material adsorbed to immunomagnetic beads is able to prevent infection of susceptible N2a cells and cure chronically scrapie-infected neuroblastoma cultures. This antibody was also shown to transmigrate across the BBB and cross the plasma membrane of neurons to target cytosolic PrPC. In contrast, treatment with a conventional anti-prion antibody derived from mouse immunised with recombinant PrP protein was unable to prevent recurrence of PrPSc replication. Furthermore, our camelid antibody did not display any neurotoxic effects following treatment of susceptible N2a cells as evidenced by TUNEL staining. These findings demonstrate the potential use of anti-prion camelid antibodies for the treatment of prion and other related diseases via non-invasive means.  相似文献   

11.
Prion diseases are caused by the conversion of a cellular protein (PrPC) into a misfolded, aggregated isoform (PrPRes). Misfolding of recombinant PrPC in the absence of PrPRes template, cellular factors, denaturing agents, or at neutral pH has not been achieved. A number of studies indicate that dimerization of PrPC may be a key step in the aggregation process. In an effort to understand the molecular event that may activate misfolding of PrPC in more relevant physiological conditions, we tested if enforced dimerization of PrPC may induce a conformational change reminiscent of the conversion of PrPC to PrPRes. We used a well described inducible dimerization strategy whereby a chimeric PrPC composed of a modified FK506-binding protein (Fv) fused with PrPC and termed Fv-PrP is incubated in the presence of a monomeric FK506 or dimerizing AP20187 ligand. Addition of AP20187 but not FK506 to recombinant Fv-PrP (rFv-PrP) in physiological-like conditions resulted in a rapid conformational change characterized by an increase in β-sheet structure and simultaneous aggregation of the protein. Aggregates were partially resistant to proteinase K and induced the conversion of soluble rFv-PrP in serial seeding experiments. As judged from thioflavin T binding and electron microscopy, aggregates converted to amyloid fibers. Aggregates were toxic to cultured cells, whereas soluble rFv-PrP and amyloid fibers were harmless. This study strongly supports the proposition that dimerization of PrPC is a key pathological primary event in the conversion of PrPC and may initiate the pathogenesis of prion diseases.  相似文献   

12.
Misfolding and aggregation of proteins are common pathogenic mechanisms of a group of diseases called proteinopathies. The formation and spread of proteinaceous lesions within and between individuals were first described in prion diseases and proposed as the basis of their infectious nature. Recently, a similar “prion-like” mechanism of transmission has been proposed in other neurodegenerative diseases such as Alzheimer''s disease. We investigated if misfolding and aggregation of corrupted prion protein (PrPTSE) are always associated with horizontal transmission of disease. Knock-in transgenic mice (101LL) expressing mutant PrP (PrP-101L) that are susceptible to disease but do not develop any spontaneous neurological phenotype were inoculated with (i) brain extracts containing PrPTSE from healthy 101LL mice with PrP plaques in the corpus callosum or (ii) brain extracts from mice overexpressing PrP-101L with neurological disease, severe spongiform encephalopathy, and formation of proteinase K-resistant PrPTSE. In all instances, 101LL mice developed PrP plaques in the area of inoculation and vicinity in the absence of clinical disease or spongiform degeneration of the brain. Importantly, 101LL mice did not transmit disease on serial passage, ruling out the presence of subclinical infection. Thus, in both experimental models the formation of PrPTSE is not infectious. These results have implications for the interpretation of tests based on the detection of protein aggregates and suggest that de novo formation of PrPTSE in the host does not always result in a transmissible prion disease. In addition, these results question the validity of assuming that all diseases due to protein misfolding can be transmitted between individuals.  相似文献   

13.
Prion disease is caused by a single pathogenic protein (PrPSc), an abnormal conformer of the normal cellular prion protein PrPC. Depletion of PrPC in prion knockout mice makes them resistant to prion disease. Thus, gene silencing of the Prnp gene is a promising effective therapeutic approach. Here, we examined adeno-associated virus vector type 2 encoding a short hairpin RNA targeting Prnp mRNA (AAV2-PrP-shRNA) to suppress PrPC expression both in vitro and in vivo. AAV2-PrP-shRNA treatment suppressed PrP levels and prevented dendritic degeneration in RML-infected brain aggregate cultures. Infusion of AAV2-PrP-shRNA-eGFP into the thalamus of CD-1 mice showed that eGFP was transported to the cerebral cortex via anterograde transport and the overall PrPC levels were reduced by ∼70% within 4 weeks. For therapeutic purposes, we treated RML-infected CD-1 mice with AAV2-PrP-shRNA beginning at 50 days post inoculation. Although AAV2-PrP-shRNA focally suppressed PrPSc formation in the thalamic infusion site by ∼75%, it did not suppress PrPSc formation efficiently in other regions of the brain. Survival of mice was not extended compared to the untreated controls. Global suppression of PrPC in the brain is required for successful therapy of prion diseases.  相似文献   

14.
Corruption of cellular prion protein (PrPC) function(s) at the plasma membrane of neurons is at the root of prion diseases, such as Creutzfeldt-Jakob disease and its variant in humans, and Bovine Spongiform Encephalopathies, better known as mad cow disease, in cattle. The roles exerted by PrPC, however, remain poorly elucidated. With the perspective to grasp the molecular pathways of neurodegeneration occurring in prion diseases, and to identify therapeutic targets, achieving a better understanding of PrPC roles is a priority. Based on global approaches that compare the proteome and metabolome of the PrPC expressing 1C11 neuronal stem cell line to those of PrPnull-1C11 cells stably repressed for PrPC expression, we here unravel that PrPC contributes to the regulation of the energetic metabolism by orienting cells towards mitochondrial oxidative degradation of glucose. Through its coupling to cAMP/protein kinase A signaling, PrPC tones down the expression of the pyruvate dehydrogenase kinase 4 (PDK4). Such an event favors the transfer of pyruvate into mitochondria and its conversion into acetyl-CoA by the pyruvate dehydrogenase complex and, thereby, limits fatty acids β-oxidation and subsequent onset of oxidative stress conditions. The corruption of PrPC metabolic role by pathogenic prions PrPSc causes in the mouse hippocampus an imbalance between glucose oxidative degradation and fatty acids β-oxidation in a PDK4-dependent manner. The inhibition of PDK4 extends the survival of prion-infected mice, supporting that PrPSc-induced deregulation of PDK4 activity and subsequent metabolic derangements contribute to prion diseases. Our study posits PDK4 as a potential therapeutic target to fight against prion diseases.  相似文献   

15.
The “protein only” hypothesis states that the key phenomenon in prion pathogenesis is the conversion of the host protein (PrPC) into a β-sheet enriched polymeric and pathogenic conformer (PrPSc). However the region of PrP bearing the information for structural transfer is still controversial. In a recent report, we highlighted the role of the C terminal part i.e., the helixes H2 and H3, using mutation approaches on recombinant PrP. The H2H3 was shown to be the minimal region necessary to reproduce the oligomerization pattern of the full-length protein. The oligomers produced from isolated H2H3 domain presented the same structural characteristics as the oligomers formed from the full-length PrP. Combining other groups'' results, this paper further discusses the relative, direct or indirect role of different PrP regions in assembly. The H2H3 region represents the core of PrP oligomers and fibrils, whereas the N terminus could explain divergences among different aggregates. Finally this review evocates the possibility to separate the domain involved in prion information transference (i.e., prion replication) from the domain bearing the cytotoxicity properties.Key words: prion, H2H3, amyloid, domain of replication, unfolding, strain, polymer, fibersTransmissible spongiform encephalopathies (TSE), fatal neurodegenerative diseases affecting humans and other mammalians, induce in most cases loss of motor control and dementia. PrP is a protein physiologically present in parts of the animal kingdom (in mammals, birds, reptiles and fishes). According to the “protein-only” hypothesis,1,2 the key phenomenon in the pathogenesis is the conversion of the α-helix rich host-encoded PrP form (PrPC) into a pathogenic conformer (PrPSc) characterized by a higher content in β-sheet and a polymeric state. The conversion to an enriched β-sheet structure is supposed to be due to the modification—induced only by a PrPSc-like state acting as a template- of PrPC into the PrPSc conformer. This hypothesis was first proposed by Griffith in 19673 and revisited by Lansbury et al. in 1993.4 The prion hypothesis has now found increasing support from experimental evidence based on the synthetic production of β-sheeted recombinant PrP which shows pathogenic properties in a wide variety of physico-chemical conditions.57 However, the molecular basis of prion conversion remains unclear, especially the various structural landscape of the PrPSc, which is the basis of the strain phenomenon.8To understand the mechanisms of transfer of the structural information, two mains issues have to be addressed: (1) we need to understand which region(s) of the protein act as template for conversion and (2) what is the “pathogenic” state of this domain. In this review, we shall assume that the region bearing the infectious information for replication and the region responsible for polymerization are identical. However, the link between the propensity of a domain to form aggregates and the ability to contain the necessary information for prion replication is far from being trivial. Generally the formation of amyloid assemblies results from the aggregation of disordered peptides or in some cases from disordered regions of a folded protein.8 If we consider that prion replication is only supported by the globular part of PrP9 the currently available model involves the folded domain. Since all structural transitions need at least a partial unfolding and refolding process, pre-required structural events should be considered prior to the conversion process.  相似文献   

16.
Prions, the agents of transmissible spongiform encephalopathies, require the expression of prion protein (PrPC) to propagate disease. PrPC is converted into an abnormal insoluble form, PrPSc, that gains neurotoxic activity. Conversely, clinical manifestations of prion disease may occur either before or in the absence of PrPSc deposits, but the loss of normal PrPC function contribution for the etiology of these diseases is still debatable. Prion disease-associated mutations in PrPC represent one of the best models to understand the impact of PrPC loss-of-function. PrPC associates with various molecules and, in particular, the interaction of PrPC with laminin (Ln) modulates neuronal plasticity and memory formation. To assess the functional alterations associated with PrPC mutations, wild-type and mutated PrPC proteins were expressed in a neural cell line derived from a PrPC-null mouse. Treatment with the laminin γ1 chain peptide (Ln γ1), which mimics the Ln binding site for PrPC, increased intracellular calcium in cells expressing wild-type PrPC, whereas a significantly lower response was observed in cells expressing mutated PrPC molecules. The Ln γ1 did not promote process outgrowth or protect against staurosporine-induced cell death in cells expressing mutated PrPC molecules in contrast to cells expressing wild-type PrPC. The co-expression of wild-type PrPC with mutated PrPC molecules was able to rescue the Ln protective effects, indicating the lack of negative dominance of PrPC mutated molecules. These results indicate that PrPC mutations impair process outgrowth and survival mediated by Ln γ1 peptide in neural cells, which may contribute to the pathogenesis of genetic prion diseases.  相似文献   

17.
The cellular prion protein (PrPC) consists of a flexible N-terminal tail (FT, aa 23–128) hinged to a membrane-anchored globular domain (GD, aa 129–231). Ligation of the GD with antibodies induces rapid neurodegeneration, which is prevented by deletion or functional inactivation of the FT. Therefore, the FT is an allosteric effector of neurotoxicity. To explore its mechanism of action, we generated transgenic mice expressing the FT fused to a GPI anchor, but lacking the GD (PrPΔ141–225, or “FTgpi”). Here we report that FTgpi mice develop a progressive, inexorably lethal neurodegeneration morphologically and biochemically similar to that triggered by anti-GD antibodies. FTgpi was mostly retained in the endoplasmic reticulum, where it triggered a conspicuous unfolded protein response specifically activating the PERK pathway leading to phosphorylation of eIF2α and upregulation of CHOP ultimately leading to neurodegeration similar to what was observed in prion infection.  相似文献   

18.
19.
Prion diseases are a group of transmissible, invariably fatal neurodegenerative diseases that affect both humans and animals. According to the protein-only hypothesis, the infectious agent is a prion (proteinaceous infectious particle) that is composed primarily of PrPSc, the disease-associated isoform of the cellular prion protein, PrP. PrPSc arises from the conformational change of the normal, glycosylphosphatidylinositol (GPI)-anchored protein, PrPC. The mechanism by which this process occurs, however, remains enigmatic. Rabbits are one of a small number of mammalian species reported to be resistant to prion infection. Sequence analysis of rabbit PrP revealed that its C-terminal amino acids differ from those of PrP from other mammals and may affect the anchoring of rabbit PrP through its GPI anchor. Using a cell culture model, this study investigated the effect of the rabbit PrP-specific C-terminal amino acids on the addition of the GPI anchor to PrPC, PrPC localization, and PrPSc formation. The incorporation of rabbit-specific C-terminal PrP residues into mouse PrP did not affect the addition of a GPI anchor or the localization of PrP. However, these residues did inhibit PrPSc formation, suggesting that these rabbit-specific residues interfere with a C-terminal PrPSc interaction site.Prion diseases, traditionally known as transmissible spongiform encephalopathies (TSE), are a group of invariably fatal neurodegenerative diseases that affect both humans and animals. According to the protein-only hypothesis, an abnormal isoform of the host-encoded prion protein (PrPC), referred to as PrPSc, is the sole or major component of the infectious agent causing these diseases (33). These disorders affect a wide range of mammals and include diseases such as Creutzfeldt-Jakob disease (CJD), variant CJD, Gerstmann-Straüssler-Scheinker (GSS) syndrome, kuru, and fatal familial insomnia (FFI) in humans, scrapie in sheep and goats, chronic wasting disease (CWD) in cervids, and bovine spongiform encephalopathy (BSE) in cattle. The term “prion” was first used to describe the unique infectious agent and was derived from “proteinaceous infectious particle” to distinguish it from conventional pathogens such as bacteria and viruses (33).To date, rabbits are one of the few mammalian species reported to be resistant to prion infection. Rabbits do not develop clinical disease after inoculation with brain tissue from individuals affected by the human prion diseases CJD and kuru, or by a number of animal forms of the disease, including scrapie and transmissible mink encephalopathy (TME) (12). In addition, mouse neuroblastoma (MNB) cells overexpressing rabbit PrP are also resistant to prion infection (45). Evidence that rabbit cells per se have the correct cellular machinery to support prion propagation has come from studies using the rabbit kidney epithelial cell line RK13. Upon transfection with appropriate PrP-expressing transgenes, these cells are a highly efficient and robust model of prion infection (6, 25, 41, 43). RK13 cells do not have detectable levels of endogenous rabbit PrPC and are therefore ideal for studying exogenous PrPC and the propagation of prions from different species (6). Originally, it was shown that RK13 cells overexpressing ovine PrP became susceptible to infection with scrapie (43), and more recently, RK13 cells expressing rodent PrPC, from either the mouse or the bank vole, were readily infected by prions adapted to and propagated in these two species (6, 41). RK13 cells expressing human PrPC, however, were resistant to infection with human prions derived directly from a patient with sporadic CJD (25). Since RK13 cells overexpressing PrP are a well-established model of prion propagation, we can therefore conclude that while these cells apparently have the appropriate cellular machinery to support prion propagation, it may be a characteristic of the rabbit prion protein itself that results in the resistance of this species to prion infection. However, the loss of a cellular cofactor may also be a contributing factor.Analysis of the rabbit PrP amino acid sequence shows that it has all the features previously described for members of the PrP protein family, including an N-terminal signal peptide, an octapeptide repeat region, and a C-terminal signal sequence (26). While amino acid sequence comparison of both mouse and rabbit PrP species reveals 87% sequence homology, there are 22 amino acid differences between the two, and several of these reside in regions of PrP known to be important in PrPSc formation. In scrapie-infected MNB cells, the residues Gly99 and Met108 within the N terminus, Ser173 within the central region, and Ile214 within the C terminus of rabbit PrP were shown to inhibit PrPSc generation when incorporated into mouse PrP, suggesting that multiple amino acid residues in rabbit PrP inhibit PrPSc formation (45). Approximately one-third (9/33 residues in the immature sequence) of the amino acid difference between mouse and rabbit PrPs was shown to occur at the glycosylphosphatidylinositol (GPI) anchor attachment site (see Fig. S1 in the supplemental material). As yet, studies involving this region of rabbit PrP have not been performed. Therefore, this region of rabbit PrP may provide further insight into the resistance of rabbits to prion infection.GPI anchor addition occurs via a transamination reaction in the endoplasmic reticulum (ER) following cleavage of the C-terminal signal sequence (39). There is no consensus sequence with which to identify the C-terminal cleavage site, but there are three key C-terminal elements: (i) the cleavage site, or ω site, where the GPI anchor attaches to the COOH group of the ω amino acid; (ii) a hydrophilic spacer region of 8 to 12 amino acids (ω + 1 up to ω + 10); and (iii) a hydrophobic region of 10 to 20 amino acids (ω + 11 onwards) (9). Analysis of known GPI-anchored proteins has given rise to sequence motifs in the C-terminal signal peptide allowing the prediction of the ω site of proteins. Due to the complexity of experimentally determining the ω site of GPI-anchored proteins, relatively few of the many known GPI-anchored proteins have had their ω sites determined (36 of 340 proteins in 2008) (32) The ω site of hamster PrP was determined experimentally to be at amino acid 231 (34) and is predicted to be at the same site for PrPs from all mammals, based on amino acid sequence comparison. Amino acid substitutions near the ω site of mouse PrP revealed that mouse PrP has an ω site at residue 230 (17). It was also shown that single amino acid substitutions at and near the ω site of mouse PrP affect the anchoring and conversion efficiency of PrP (17). It is therefore possible that the amino acids at the C terminus and within the GPI anchor signal sequence of rabbit PrP lead to the resistance to prion infection.To date, no protein structures containing a GPI anchor have been determined by X-ray crystallography, and although the nuclear magnetic resonance (NMR) structures of mouse and rabbit PrP have been solved, they do not contain any structural information for the residues immediately preceding the GPI anchor. We therefore created a mutant mouse PrP model containing rabbit PrP-specific amino acids at the ω site to investigate whether these residues are involved in rabbit resistance to prion infection. Here we demonstrate that the GPI anchor attachment site is an important site that controls the ability of PrP to be converted into PrPSc and that residues ω and ω + 1 of PrP are important modulators of this pathogenic process.  相似文献   

20.

Background

A hallmark of the prion diseases is the conversion of the host-encoded cellular prion protein (PrPC) into a disease related, alternatively folded isoform (PrPSc). The accumulation of PrPSc within the brain is associated with synapse loss and ultimately neuronal death. Novel therapeutics are desperately required to treat neurodegenerative diseases including the prion diseases.

Principal Findings

Treatment with glimepiride, a sulphonylurea approved for the treatment of diabetes mellitus, induced the release of PrPC from the surface of prion-infected neuronal cells. The cell surface is a site where PrPC molecules may be converted to PrPSc and glimepiride treatment reduced PrPSc formation in three prion infected neuronal cell lines (ScN2a, SMB and ScGT1 cells). Glimepiride also protected cortical and hippocampal neurones against the toxic effects of the prion-derived peptide PrP82–146. Glimepiride treatment significantly reduce both the amount of PrP82–146 that bound to neurones and PrP82–146 induced activation of cytoplasmic phospholipase A2 (cPLA2) and the production of prostaglandin E2 that is associated with neuronal injury in prion diseases. Our results are consistent with reports that glimepiride activates an endogenous glycosylphosphatidylinositol (GPI)-phospholipase C which reduced PrPC expression at the surface of neuronal cells. The effects of glimepiride were reproduced by treatment of cells with phosphatidylinositol-phospholipase C (PI-PLC) and were reversed by co-incubation with p-chloromercuriphenylsulphonate, an inhibitor of endogenous GPI-PLC.

Conclusions

Collectively, these results indicate that glimepiride may be a novel treatment to reduce PrPSc formation and neuronal damage in prion diseases.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号