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 312 毫秒
1.
The prion diseases occur following the conversion of the cellular prion protein (PrPC) into disease-related isoforms (PrPSc). In this study, the role of the glycosylphosphatidylinositol (GPI) anchor attached to PrPC in prion formation was examined using a cell painting technique. PrPSc formation in two prion-infected neuronal cell lines (ScGT1 and ScN2a cells) and in scrapie-infected primary cortical neurons was increased following the introduction of PrPC. In contrast, PrPC containing a GPI anchor from which the sialic acid had been removed (desialylated PrPC) was not converted to PrPSc. Furthermore, the presence of desialylated PrPC inhibited the production of PrPSc within prion-infected cortical neurons and ScGT1 and ScN2a cells. The membrane rafts surrounding desialylated PrPC contained greater amounts of sialylated gangliosides and cholesterol than membrane rafts surrounding PrPC. Desialylated PrPC was less sensitive to cholesterol depletion than PrPC and was not released from cells by treatment with glimepiride. The presence of desialylated PrPC in neurons caused the dissociation of cytoplasmic phospholipase A2 from PrP-containing membrane rafts and reduced the activation of cytoplasmic phospholipase A2. These findings show that the sialic acid moiety of the GPI attached to PrPC modifies local membrane microenvironments that are important in PrP-mediated cell signaling and PrPSc formation. These results suggest that pharmacological modification of GPI glycosylation might constitute a novel therapeutic approach to prion diseases.  相似文献   

2.
There is increasing interest in the role of the glycosylphosphatidylinositol (GPI) anchor attached to the cellular prion protein (PrPC). Since GPI anchors can alter protein targeting, trafficking and cell signaling, our recent study examined how the structure of the GPI anchor affected prion formation. PrPC containing a GPI anchor from which the sialic acid had been removed (desialylated PrPC) was not converted to PrPSc in prion-infected neuronal cell lines and in scrapie-infected primary cortical neurons. In uninfected neurons desialylated PrPC was associated with greater concentrations of gangliosides and cholesterol than PrPC. In addition, the targeting of desialylated PrPC to lipid rafts showed greater resistance to cholesterol depletion than PrPC. The presence of desialylated PrPC caused the dissociation of cytoplasmic phospholipase A2 (cPLA2) from PrP-containing lipid rafts, reduced the activation of cPLA2 and inhibited PrPSc production. We conclude that the sialic acid moiety of the GPI attached to PrPC modifies local membrane microenvironments that are important in PrP-mediated cell signaling and PrPSc formation.  相似文献   

3.
In prion diseases, the cellular form of the prion protein, PrPC, undergoes a conformational conversion to the infectious isoform, PrPSc. PrPC associates with lipid rafts through its glycosyl-phosphatidylinositol (GPI) anchor and a region in its N-terminal domain which also binds to heparan sulfate proteoglycans (HSPGs). We show that heparin displaces PrPC from rafts and promotes its endocytosis, suggesting that heparin competes with an endogenous raft-resident HSPG for binding to PrPC. We then utilised a transmembrane-anchored form of PrP (PrP-TM), which is targeted to rafts solely by its N-terminal domain, to show that both heparin and phosphatidylinositol-specific phospholipase C can inhibit its association with detergent-resistant rafts, implying that a GPI-anchored HSPG targets PrPC to rafts. Depletion of the major neuronal GPI-anchored HSPG, glypican-1, significantly reduced the raft association of PrP-TM and displaced PrPC from rafts, promoting its endocytosis. Glypican-1 and PrPC colocalised on the cell surface and both PrPC and PrPSc co-immunoprecipitated with glypican-1. Critically, treatment of scrapie-infected N2a cells with glypican-1 siRNA significantly reduced PrPSc formation. In contrast, depletion of glypican-1 did not alter the inhibitory effect of PrPC on the β-secretase cleavage of the Alzheimer''s amyloid precursor protein. These data indicate that glypican-1 is a novel cellular cofactor for prion conversion and we propose that it acts as a scaffold facilitating the interaction of PrPC and PrPSc in lipid rafts.  相似文献   

4.
The prion diseases occur following the conversion of the cellular prion protein (PrPC) into a disease-related isoform (PrPSc). In this study a cell painting technique was used to examine the role of the glycosylphosphatidylinositol (GPI) anchor attached to PrPC in prion formation. The introduction of PrPC to infected neuronal cells increased the cholesterol content of cell membranes, increased activation of cytoplasmic phospholipase A2 (cPLA2) and increased PrPSc formation. In contrast, PrPC with a monoacylated GPI anchor did not alter the amount of cholesterol in cell membranes, was not found within lipid rafts and did not activate cPLA2. Although monoacylated PrPC remains within cells for longer than native PrPC it was not converted to PrPSc. Moreover, the presence of monoacylated PrPC displaced cPLA2 from PrPSc-containing lipid rafts, reducing the activation of cPLA2 and PrPSc formation. We conclude that acylation of the GPI anchor attached to PrPC modifies the local membrane microenvironments that control some cell signaling pathways, the trafficking of PrPC and PrPSc formation. In addition, such observations raise the possibility that the pharmacological modification of GPI anchors might constitute a novel therapeutic approach to prion diseases.Key words: cholesterol, glycosylphosphatidylinositol, lipid rafts, membranes, phospholipase A2, prion, trafficA key event in the prion diseases is the conversion of a normal host protein (PrPC) into a disease-associated isoform (PrPSc).1 Although the presence of PrPC is essential for prion formation,2,3 not all cells that express PrPC are permissive for PrPSc replication. The reasons why some cells that express PrPC do not replicate PrPSc are not fully understood. Reports that the targeting of PrPC to specific membranes is required for efficient PrPSc formation4 indicate that the factors that affect the cellular targeting and intracellular trafficking of PrPC are critical in determining PrPSc replication.Our study examined the effects of the glycosylphosphatidylinositol (GPI) anchor that links the majority of PrPC molecules to cell membranes5 on PrPSc formation. Originally GPI anchors were seen as a simple method of attaching proteins to cell membranes. However, there is increasing interest in the role of GPI anchors in complex biological functions including the regulation of membrane composition, cell signaling and protein trafficking.6 To examine the role of the GPI anchor PrPC preparations were digested with phosphatidylinositol-phospholipase C (PI-PLC) (deacylated PrPC) or phospholipase A2 (PLA2) (monoacylated PrPC) (Fig. 1) and isolated by reverse phase chromatography. These digestions, coupled with a cell painting technique, allowed us to examine modifications of the GPI anchor that could not be achieved by genetic manipulation methods. Controversy surrounds the role of the GPI anchor in PrPSc formation; the seminal observation that transgenic mice producing anchorless PrPC produced large amounts of extracellular PrPSc,7 suggests that the GPI has little effect upon PrPSc replication. In contrast, a recent study showed that cells that produce anchorless PrPC were not permissive to PrPSc formation8 and in our study deacylated PrPC did not affect PrPSc production. Although at first glance these results appear contradictory, they may be explained by reference to the site of conversion of PrPC to PrPSc. Clearly anchorless PrPC can be converted to PrPSc in a process that occurs within the extracellular milieu. However, as anchorless PrPC is rapidly secreted from cells7 it has little contact with cell-associated PrPSc. Similarly we found that deacylated PrPC was fully soluble and did not readily associate with cells.Open in a separate windowFigure 1Phospholipase digestion of PrPC affects the acylation of the GPI anchor. Cartoon showing the putative GPI anchor attached to PrPC, monoacylated PrPC and deacylated PrPC. Glycan residues shown include inositol (Inos), mannose (Man), sialic acid (SA), galactose (Gal), N-acetyl galactosamine (GalNAc) and glucosamine (GlcN) as well as phosphate (P).Native PrPC is rapidly transferred to cells9 and we showed that the addition of PrPC caused a dose-dependent increase in the PrPSc content of all prion-infected cell lines tested. We used this cell painting technique to introduce monoacylated PrPC to recipient cells. Our paper describes three major observations; firstly that monoacylated PrPC behaves differently to native PrPC with regards to cellular distribution, intracellular trafficking and cell signaling; secondly, that monoacylated PrPC was not converted to PrPSc and thirdly, that monoacylated PrPC inhibited the conversion of endogenous PrPC to PrPSc.The presence of GPI anchors targets proteins including PrPC and PrPSc to specialized membrane micro-domains that are commonly called lipid rafts.10,11 Lipid rafts are patches of membranes that are highly enriched in cholesterol and sphingolipids and which are operationally defined by their insolubility in cold non-ionic detergents and floatation as low density membranes on sucrose density gradients. The importance of lipid rafts in prion diseases is based upon studies showing that treatment with cholesterol synthesis inhibitors reduced cellular cholesterol and the formation of PrPSc.12 Since the cholesterol content of cell membranes is critical for the formation of lipid rafts13 it is assumed that the integrity of these lipid rafts is necessary for efficient PrPSc formation. The presence of GPI-anchored proteins is thought to help lipid rafts form as the saturated fatty acids that are contained within GPI anchors facilitate the solubilisation of cholesterol in the membrane14 and the glycan component protect cholesterol from water.15 Thus the nature and number of the acyl chains contained within GPI anchors are factors that affect lipid raft formation (Fig. 2).Open in a separate windowFigure 2Acylation of PrPC affects the underlying cell membrane. Cartoon showing the proposed membranes surrounding native PrPC and monoacylated PrPC, including cholesterol (), lyso-phospholipids (), saturated phospholipids () and unsaturated phospholipids (). Monoacylated PrPC is not directed to lipid rafts and the membrane surrounding contains less cholesterol and more unsaturated phospholipids.We observed that the addition of native PrPC significantly increased the amount of cholesterol in cell membranes. This result was unexpected as the amount of cholesterol in cell membranes is tightly controlled by an esterification and hydrolysis cycle which releases cholesterol from stored cholesterol esters.16 The PrPC-induced increase in cholesterol was accompanied by a reduction in cholesterol esters suggesting that it was derived from the hydrolysis of cholesterol esters. Pharmacological inhibition of cholesterol ester hydrolysis not only blocked the PrPC-induced increase in cholesterol and the reduction in cholesterol esters, but also reduced the PrPC-induced increase in PrPSc formation. Collectively, these results indicate that cells respond to the introduction of PrPC by the hydrolysis of cholesterol esters; which provides cholesterol to stabilize PrPC within the lipid rafts that are necessary to facilitate PrPSc formation.The differences in the cellular distribution of PrPC and monoacylated PrPC were examined using neurons from Prnp knockout mice. While PrPC was targeted to lipid rafts, monoacylated PrPC was found predominantly within the normal (non-raft) cell membranes. Unlike native PrPC, monoacylated PrPC did not affect the cholesterol content of cell membranes; an observation that highlights the critical role of the presence of two saturated fatty acids contained within the GPI anchor to sequester cholesterol and precipitate the formation of lipid rafts.14 Critically, monoacylated PrPC was unable to solubilize cholesterol or precipitate lipid raft formation and was consequently found outside lipid rafts (Fig. 2).Since many raft-associated proteins including PrPC traffic within cells via specific pathways,17 we argued that monoacylated PrPC might undergo alternative trafficking pathways to those utilized by native PrPC. Our observations, that greater amounts of monoacylated PrPC than native PrPC were expressed at the cell surface, and that while most of the native PrPC was removed from these cells within 24 h, monoacylated PrPC remained in neurons for longer, are indicative of altered intracellular trafficking. It is possible that the cellular location and/or pathway(s) used by monoacylated PrPC may be physically segregated from PrPSc. This hypothesis would explain our observation that the addition of monoacylated PrPC to prion-infected cell lines did not increase PrPSc formation indicating that it was not converted to PrPSc.Since monoacylated phospholipids exist only transiently within cell membranes, they are rapidly reacylated by esterases,18 we wondered whether the monoacylated PrPC could also be reacylated to form the native, diacylated PrPC. We found no evidence that the monoacylated PrPC added to Prnp knockout neurons was reacylated suggesting that the enzymes involved in reacylation of membrane phospholipids do not recognize phosphatidylinositol when it is incorporated as part of the GPI anchor. In addition we were unable to detect monoacylated PrPC occurring naturally within Prnp wild-type neurons.While these theories explain why monoacylated PrPC was not readily converted to PrPSc; a more refined hypothesis is required to explain why monoacylated PrPC reduced PrPSc production. One possibility is that monoacylated PrPC is converted to monoacylated PrPSc which in turn acts as an inefficient template for PrPC to PrPSc conversion.19 It is also possible that monoacylated PrPC competes with endogenous PrPC for specific partner proteins involved in endocytosis. The depletion of these partner proteins could consequently alter the trafficking of endogenous PrPC and hence PrPSc formation. In our paper we explored the idea that the binding of monoacylated PrPC to PrPSc modifies the lipid rafts that are involved in PrPSc formation. Both the composition and function of lipid rafts is dynamic and controlled by an induced fit model.20 Since monoacylated PrPC does not sequester cholesterol, the membrane surrounding a complex between PrPSc and monoacylated PrPC might be expected to contain less cholesterol than membranes formed following the interaction between PrPSc and PrPC (Fig. 3). Thus, the binding of monoacylated PrPC to PrPSc may reduce the cholesterol content of local membranes to a level below that required for the conversion of PrPC to PrPSc. This hypothesis is consistent with observations that formation of PrPSc was affected by the lipid composition of membranes21 and that lipids were essential co-factors in prion formation.22Open in a separate windowFigure 3Monoacylated PrPC affects the capture of cPLA2 in PrPSc-containing lipid rafts. (A) Cartoon showing the proposed membranes surrounding PrPSc and PrPC including the capture of cPLA2 in lipid rafts that are dense in cholesterol () and saturated phospholipids (). (B) Cartoon showing the proposed interactions between PrPSc and monoacylated PrPC which reduces the solubility of membranes to cholesterol, increases the concentration of unsaturated phospholipids () and prevents the capture of cPLA2 into PrPSc-containing lipid rafts.Our studies raise the question “why are lipid rafts important in PrPSc formation?” Lipid rafts are enriched with signaling molecules and can act as domains in which the GPI anchors attached to PrPC can interact with cell signaling pathways.23 Although PrPC has been reported to interact with many cell signaling pathways we concentrated upon its effects on the activation of cPLA2, based upon studies showing that the activation of cPLA2 correlates strongly with the amounts of cholesterol and PrPSc,24 and that the inhibition of cPLA2 reduces PrPSc formation.25 These observations underpin the hypothesis that it is the clustering of GPI anchors attached to PrP proteins that leads to the activation of cPLA2. This hypothesis was tested by incubating Prnp knockout neurons with PrPC or monoacylated PrPC and then adding the anti-PrP mAb 4F2. We found that the cross-linkage of PrPC by mAb 4F2 caused the activation of cPLA2, whereas the cross-linkage of monoacylated PrPC by mAb 4F2 had no significant affect. The activation of cPLA2 is associated with multiple phosphorylation events and the translocation of cPLA2 to specific membranes.26 In scrapie-infected GT1 (ScGT1) cells most of the cPLA2 was found within lipid rafts consistent with reports that the activation of cPLA2 is dependent upon cholesterol-sensitive lipid rafts.27 More specifically, immunoprecipitation studies showed that cPLA2 was targeted to PrPSc-containing lipid rafts.24 Collectively, these observations suggest that PrPC binds to PrPSc in cholesterol-dense lipid rafts, where it activates the cPLA2 that facilitates the conversion of PrPC to PrPSc (Fig. 3).We found that the presence of monoacylated PrPC reduced the activation of cPLA2 within prion-infected cells. As cPLA2 can be activated by multiple different stimuli we sought to determine whether the inhibitory effect of monoacylated PrPC was stimulus specific. We reported that monoacylated PrPC did not affect the activation of cPLA2 by a phospholipase A2-activating peptide indicating that monoacylated PrPC did not have a direct inhibitory effect upon cPLA2. Rather we found that the addition of monoacylated PrPC to prion-infected cells caused the dissociation of some cPLA2 from PrPSc-containing lipid rafts. The targeting of cPLA2 to membranes containing their endogenous substrates can regulate cell signaling, including for the formation of second messengers such as platelet-activating factor that facilitate PrPSc formation.25 We propose that the binding of monoacylated PrPC to PrPSc changed the composition of the underlying membrane so that it no longer captured and activated cPLA2 (Fig. 3).In conclusion our study showed that the addition of monoacylated PrPC modified cell membranes thus reducing the activation of cPLA2 and PrPSc formation in prion-infected cells. We propose that the 2 acyl chains attached to the GPI anchor is a critical factor that facilitates the conversion to PrPC to PrPSc within cell membranes and that the presence of monoacylated PrPC disrupts lipid raft micro-domains that are essential for efficient PrPSc formation. Moreover, these results raise the possibility that targeting the GPI anchor attached to PrPC may reveal novel therapeutics/treatments for prion diseases.  相似文献   

5.

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   

6.
Conversion of prion protein (PrPC) into a pathological isoform (PrPSc) during prion infection occurs in lipid rafts and is dependent on cholesterol. Here, we show that prion infection increases the abundance of cholesterol transporter, ATP-binding cassette transporter type A1 (ATP-binding cassette transporter type A1), but reduces cholesterol efflux from neuronal cells leading to the accumulation of cellular cholesterol. Increased abundance of ABCA1 in prion disease was confirmed in prion-infected mice. Mechanistically, conversion of PrPC to the pathological isoform led to PrPSc accumulation in rafts, displacement of ABCA1 from rafts and the cell surface, and enhanced internalization of ABCA1. These effects were abolished with reversal of prion infection or by loading cells with cholesterol. Stimulation of ABCA1 expression with liver X receptor agonist or overexpression of heterologous ABCA1 reduced the conversion of prion protein into the pathological form upon infection. These findings demonstrate a reciprocal connection between prion infection and cellular cholesterol metabolism, which plays an important role in the pathogenesis of prion infection in neuronal cells.  相似文献   

7.
The prion protein (PrPC) is highly expressed within the nervous system. Similar to other GPI-anchored proteins, PrPC is found in lipid rafts, membrane domains enriched in cholesterol and sphingolipids. PrPC raft association, together with raft lipid composition, appears essential for the conversion of PrPC into the scrapie isoform PrPSc, and the development of prion disease. Controversial findings were reported on the nature of PrPC-containing rafts, as well as on the distribution of PrPC between rafts and non-raft membranes. We investigated PrPC/ganglioside relationships and their influence on PrPC localization in a neuronal cellular model, cerebellar granule cells. Our findings argue that in these cells at least two PrPC conformations coexist: in lipid rafts PrPC is present in the native folding (α-helical), stabilized by chemico-physical condition, while it is mainly present in other membrane compartments in a PrPSc-like conformation. We verified, by means of antibody reactivity and circular dichroism spectroscopy, that changes in lipid raft-ganglioside content alters PrPC conformation and interaction with lipid bilayers, without modifying PrPC distribution or cleavage. Our data provide new insights into the cellular mechanism of prion conversion and suggest that GM1-prion protein interaction at the cell surface could play a significant role in the mechanism predisposing to pathology.  相似文献   

8.
The cellular prion protein (PrPC) is an N-glycosylated GPI-anchored protein usually present in lipid rafts with numerous putative functions. When it changes its conformation to a pathological isoform (then referred to as PrPSc), it is an essential part of the prion, the agent causing fatal and transmissible neurodegenerative prion diseases. There is growing evidence that toxicity and neuronal damage on the one hand and propagation/infectivity on the other hand are two distinct processes of the disease and that the GPI-anchor attachment of PrPC and PrPSc plays an important role in protein localization and in neurotoxicity. Here we review how the signal sequence of the GPI-anchor matters in PrPC localization, how an altered cellular localization of PrPC or differences in GPI-anchor composition can affect prion infection, and we discuss through which mechanisms changes on the anchorage of PrPC can modify the disease process.  相似文献   

9.
《朊病毒》2013,7(4):350-353
Precisely how the accumulation of PrPSc causes the neuronal degeneration that leads to the clinical symptoms of prion diseases is poorly understood. Our recent paper showed that the clustering of specific glycosylphosphatidylinositol (GPI) anchors attached to PrP proteins triggered synapse damage in cultured neurons. First, we demonstrated that small, soluble PrPSc oligomers caused synapse damage via a GPI-dependent process. Our hypothesis, that the clustering of specific GPIs caused synapse damage, was supported by observations that cross-linkage of PrPC, either chemically or by monoclonal antibodies, also triggered synapse damage. Synapse damage was preceded by an increase in the cholesterol content of synapses and activation of cytoplasmic phospholipase A2 (cPLA2). The presence of a terminal sialic acid moiety, a rare modification of mammalian GPI anchors, was essential in the activation of cPLA2 and synapse damage induced by cross-linked PrPC. We conclude that the sialic acid modifies local membrane microenvironments (rafts) surrounding clustered PrP molecules resulting in aberrant activation of cPLA2 and synapse damage. A recent observation, that toxic amyloid-β assemblies cross-link PrPC, suggests that synapse damage in prion and Alzheimer diseases is mediated via a common molecular mechanism, and raises the possibility that the pharmacological modification of GPI anchors might constitute a novel therapeutic approach to these diseases.  相似文献   

10.
The key molecular event underlying prion diseases is the conversion of the monomeric and α-helical cellular form of the prion protein (PrPC) to the disease-associated state, which is aggregated and rich in β-sheet (PrPSc). The molecular details associated with the conversion of PrPC into PrPSc are not fully understood. The prion protein is attached to the cell membrane via a GPI lipid anchor and evidence suggests that the lipid environment plays an important role in prion conversion and propagation. We have previously shown that the interaction of the prion protein with anionic lipid membranes induces β-sheet structure and promotes prion aggregation, whereas zwitterionic membranes stabilize the α-helical form of the protein. Here, we report on the interaction of recombinant sheep prion protein with planar lipid membranes in real-time, using dual polarization interferometry (DPI). Using this technique, the simultaneous evaluation of multiple physical properties of PrP layers on membranes was achieved. The deposition of prion on membranes of POPC and POPC/POPS mixtures was studied. The properties of the resulting protein layers were found to depend on the lipid composition of the membranes. Denser and thicker protein deposits formed on lipid membranes containing POPS compared to those formed on POPC. DPI thus provides a further insight on the organization of PrP at the surface of lipid membranes.  相似文献   

11.
Precisely how the accumulation of PrPSc causes the neuronal degeneration that leads to the clinical symptoms of prion diseases is poorly understood. Our recent paper showed that the clustering of specific glycosylphosphatidylinositol (GPI) anchors attached to PrP proteins triggered synapse damage in cultured neurons. First, we demonstrated that small, soluble PrPSc oligomers caused synapse damage via a GPI-dependent process. Our hypothesis, that the clustering of specific GPIs caused synapse damage, was supported by observations that cross-linkage of PrPC, either chemically or by monoclonal antibodies, also triggered synapse damage. Synapse damage was preceded by an increase in the cholesterol content of synapses and activation of cytoplasmic phospholipase A2 (cPLA2). The presence of a terminal sialic acid moiety, a rare modification of mammalian GPI anchors, was essential in the activation of cPLA2 and synapse damage induced by cross-linked PrPC. We conclude that the sialic acid modifies local membrane microenvironments (rafts) surrounding clustered PrP molecules resulting in aberrant activation of cPLA2 and synapse damage. A recent observation, that toxic amyloid-β assemblies cross-link PrPC, suggests that synapse damage in prion and Alzheimer diseases is mediated via a common molecular mechanism, and raises the possibility that the pharmacological modification of GPI anchors might constitute a novel therapeutic approach to these diseases.  相似文献   

12.
Prion diseases are fatal neurodegenerative disorders caused by prion proteins (PrP). Infectious prions accumulate in the brain through a template-mediated conformational conversion of endogenous PrPC into alternately folded PrPSc. Immunoassays toward pre-clinical detection of infectious PrPSc have been confounded by low-level prion accumulation in non-neuronal tissue and the lack of PrPSc selective antibodies. We report a method to purify infectious PrPSc from biological tissues for use as an immunogen and sample enrichment for increased immunoassay sensitivity. Significant prion enrichment is accomplished by sucrose gradient centrifugation of infected tissue and isolation with detergent resistant membranes from lipid rafts (DRMs). At equivalent protein concentration a 50-fold increase in detectable PrPSc was observed in DRM fractions relative to crude brain by direct ELISA. Sequential purification steps result in increased specific infectivity (DRM >20-fold and purified DRM immunogen >40-fold) relative to 1% crude brain homogenate. Purification of PrPSc from DRM was accomplished using phosphotungstic acid protein precipitation after proteinase-K (PK) digestion followed by size exclusion chromatography to separate PK and residual protein fragments from larger prion aggregates. Immunization with purified PrPSc antigen was performed using wild-type (wt) and Prnp0/0 mice, both on Balb/cJ background. A robust immune response against PrPSc was observed in all inoculated Prnp0/0 mice resulting in antisera containing high-titer antibodies against prion protein. Antisera from these mice recognized both PrPC and PrPSc, while binding to other brain-derived protein was not observed. In contrast, the PrPSc inoculum was non-immunogenic in wt mice and antisera showed no reactivity with PrP or any other protein.Key words: prion, scrapie, Prnp0/0 mice, purification methodology, antibody, antisera, lipid-rafts, detergent resistant membranes, neuroscience, immunization, diagnostic  相似文献   

13.
Prion diseases occur following the conversion of the cellular prion protein (PrP(C)) into a disease related, protease-resistant isoform (PrP(Sc)). In these studies, a cell painting technique was used to introduce PrP(C) to prion-infected neuronal cell lines (ScGT1, ScN2a, or SMB cells). The addition of PrP(C) resulted in increased PrP(Sc) formation that was preceded by an increase in the cholesterol content of cell membranes and increased activation of cytoplasmic phospholipase A(2) (cPLA(2)). In contrast, although PrP(C) lacking one of the two acyl chains from its glycosylphosphatidylinositol (GPI) anchor (PrP(C)-G-lyso-PI) bound readily to cells, it did not alter the amount of cholesterol in cell membranes, was not found within detergent-resistant membranes (lipid rafts), and did not activate cPLA(2). It remained within cells for longer than PrP(C) with a conventional GPI anchor and was not converted to PrP(Sc). Moreover, the addition of high amounts of PrP(C)-G-lyso-PI displaced cPLA(2) from PrP(Sc)-containing lipid rafts, reduced the activation of cPLA(2), and reduced PrP(Sc) formation in all three cell lines. In addition, ScGT1 cells treated with PrP(C)-G-lyso-PI did not transmit infection following intracerebral injection to mice. We propose that that the chemical composition of the GPI anchor attached to PrP(C) modified the local membrane microenvironments that control cell signaling, the fate of PrP(C), and hence PrP(Sc) formation. In addition, our observations raise the possibility that pharmacological modification of GPI anchors might constitute a novel therapeutic approach to prion diseases.  相似文献   

14.
Prion diseases are fatal, neurodegenerative disorders in humans and animals and are characterized by the accumulation of an abnormally folded isoform of the cellular prion protein (PrPC), denoted PrPSc, which represents the major component of infectious scrapie prions. Characterization of the mechanism of conversion of PrPC into PrPSc and identification of the intracellular site where it occurs are among the most important questions in prion biology. Despite numerous efforts, both of these questions remain unsolved. We have quantitatively analyzed the distribution of PrPC and PrPSc and measured PrPSc levels in different infected neuronal cell lines in which protein trafficking has been selectively impaired. Our data exclude roles for both early and late endosomes and identify the endosomal recycling compartment as the likely site of prion conversion. These findings represent a fundamental step towards understanding the cellular mechanism of prion conversion and will allow the development of new therapeutic approaches for prion diseases.  相似文献   

15.
Transmissible spongiform encephalopathies are neurodegenerative diseases characterized by the accumulation of an abnormal isoform of the prion protein PrPSc. Its fragment 106-126 has been reported to maintain most of the pathological features of PrPSc, and a role in neurodegeneration has been proposed based on the modulation of membrane properties and channel formation. The ability of PrPSc to modulate membranes and/or form channels in membranes has not been clearly demonstrated; however, if these processes are important, peptide-membrane interactions would be a key feature in the toxicity of PrPSc. In this work, the interaction of PrP(106-126) with model membranes comprising typical lipid identities, as well as more specialized lipids such as phosphatidylserine and GM1 ganglioside, was examined using surface plasmon resonance and fluorescence methodologies. This comprehensive study examines different parameters relevant to characterization of peptide-membrane interactions, including membrane charge, viscosity, lipid composition, pH, and ionic strength. We report that PrP(106-126) has a low affinity for lipid membranes under physiological conditions without evidence of membrane disturbances. Membrane insertion and leakage occur only under conditions in which strong electrostatic interactions operate. These results support the hypothesis that the physiological prion protein PrPC mediates PrP(106-126) toxic effects in neuronal cells.  相似文献   

16.
Mammalian prions refold host glycosylphosphatidylinositol-anchored PrPC into β-sheet–rich PrPSc. PrPSc is rapidly truncated into a C-terminal PrP27-30 core that is stable for days in endolysosomes. The nature of cell-associated prions, their attachment to membranes and rafts, and their subcellular locations are poorly understood; live prion visualization has not previously been achieved. A key obstacle has been the inaccessibility of PrP27-30 epitopes. We overcame this hurdle by focusing on nascent full-length PrPSc rather than on its truncated PrP27-30 product. We show that N-terminal PrPSc epitopes are exposed in their physiological context and visualize, for the first time, PrPSc in living cells. PrPSc resides for hours in unexpected cell-surface, slow moving strings and webs, sheltered from endocytosis. Prion strings observed by light and scanning electron microscopy were thin, micrometer-long structures. They were firmly cell associated, resisted phosphatidylinositol-specific phospholipase C, aligned with raft markers, fluoresced with thioflavin, and were rapidly abolished by anti-prion glycans. Prion strings and webs are the first demonstration of membrane-anchored PrPSc amyloids.  相似文献   

17.
During prion infection, the normal, protease-sensitive conformation of prion protein (PrPC) is converted via seeded polymerization to an abnormal, infectious conformation with greatly increased protease-resistance (PrPSc). In vitro, protein misfolding cyclic amplification (PMCA) uses PrPSc in prion-infected brain homogenates as an initiating seed to convert PrPC and trigger the self-propagation of PrPSc over many cycles of amplification. While PMCA reactions produce high levels of protease-resistant PrP, the infectious titer is often lower than that of brain-derived PrPSc. More recently, PMCA techniques using bacterially derived recombinant PrP (rPrP) in the presence of lipid and RNA but in the absence of any starting PrPSc seed have been used to generate infectious prions that cause disease in wild-type mice with relatively short incubation times. These data suggest that lipid and/or RNA act as cofactors to facilitate the de novo formation of high levels of prion infectivity. Using rPrP purified by two different techniques, we generated a self-propagating protease-resistant rPrP molecule that, regardless of the amount of RNA and lipid used, had a molecular mass, protease resistance and insolubility similar to that of PrPSc. However, we were unable to detect prion infectivity in any of our reactions using either cell-culture or animal bioassays. These results demonstrate that the ability to self-propagate into a protease-resistant insoluble conformer is not unique to infectious PrP molecules. They suggest that the presence of RNA and lipid cofactors may facilitate the spontaneous refolding of PrP into an infectious form while also allowing the de novo formation of self-propagating, but non-infectious, rPrP-res.  相似文献   

18.
The cellular prion protein (PrPC), which is present ubiquitously in all mammalian neurons, is normally found to be linked to the cell membrane through a glycosylphosphatidylinositol (GPI) anchor. The conformational conversion of PrPC into misfolded and aggregated forms is associated with transmissible neurodegenerative diseases known as prion diseases. The importance of different misfolded conformations in prion diseases, and the mechanism by which prion aggregates induce neurotoxicity remain poorly understood. Multiple studies have been shown that the toxicity of misfolded prion protein is directly correlated with its ability to interact with and perturb membranes. This review describes the current progress toward understanding prion protein misfolding and aggregation, as well as the interaction of prion protein aggregates with lipid membrane.  相似文献   

19.
Molecules that inhibit the formation of an abnormal isoform of prion protein (PrPSc) in prion-infected cells are candidate therapeutic agents for prion diseases. Understanding how these molecules inhibit PrPSc formation provides logical basis for proper evaluation of their therapeutic potential. In this study, we extensively analyzed the effects of the anti-PrP monoclonal antibody (mAb) 44B1, pentosan polysulfate (PPS), chlorpromazine (CPZ) and U18666A on the intracellular dynamics of a cellular isoform of prion protein (PrPC) and PrPSc in prion-infected mouse neuroblastoma cells to re-evaluate the effects of those agents. MAb 44B1 and PPS rapidly reduced PrPSc levels without altering intracellular distribution of PrPSc. PPS did not change the distribution and levels of PrPC, whereas mAb 44B1 appeared to inhibit the trafficking of cell surface PrPC to organelles in the endocytic-recycling pathway that are thought to be one of the sites for PrPSc formation. In contrast, CPZ and U18666A initiated the redistribution of PrPSc from organelles in the endocytic-recycling pathway to late endosomes/lysosomes without apparent changes in the distribution of PrPC. The inhibition of lysosomal function by monensin or bafilomycin A1 after the occurrence of PrPSc redistribution by CPZ or U18666A partly antagonized PrPSc degradation, suggesting that the transfer of PrPSc to late endosomes/lysosomes, possibly via alteration of the membrane trafficking machinery of cells, leads to PrPSc degradation. This study revealed that precise analysis of the intracellular dynamics of PrPC and PrPSc provides important information for understanding the mechanism of anti-prion agents.  相似文献   

20.
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   

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