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 250 毫秒
1.
Interactions between normal, protease-sensitive prion protein (PrP-sen or PrP(C)) and its protease-resistant isoform (PrP-res or PrP(Sc)) are critical in transmissible spongiform encephalopathy (TSE) diseases. To investigate the propagation of PrP-res between cells we tested whether PrP-res in scrapie brain microsomes can induce the conversion of PrP-sen to PrP-res if the PrP-sen is bound to uninfected raft membranes. Surprisingly, no conversion was observed unless the microsomal and raft membranes were fused or PrP-sen was released from raft membranes. These results suggest that the propagation of infection between cells requires transfer of PrP-res into the membranes of the recipient cell. To assess potential cofactors in PrP conversion, we used cell-free PrP conversion assays to show that heparan sulphate can stimulate PrP-res formation, supporting the idea that endogenous sulphated glycosaminoglycans can act as important cofactors or modulators of PrP-res formation in vivo. In an effort to develop therapeutics, the antimalarial drug quinacrine was identified as an inhibitor of PrP-res formation in scrapie-infected cell cultures. Confirmation of the latter result by others has led to the initiation of human clinical trials as a treatment for Creutzfeldt-Jakob disease. PrP-res formation can also be inhibited using a variety of other types of small molecule, specific synthetic PrP peptides, and an antiserum directed at the C-terminus of PrP-sen. The latter results help to localize the sites of interaction between PrP-sen and PrP-res. Disruption of those interactions with antibodies or peptidomimetic drugs may be an attractive therapeutic strategy. The likelihood that PrP-res inhibitors can rid TSE-infected tissues of PrP-res would presumably be enhanced if PrP-res formation were reversible. However, our attempts to measure dissociation of PrP-sen from PrP-res have failed under non-denaturing conditions. Finally, we have attempted to induce the spontaneous conversion of PrP-sen into PrP-res using low concentrations of detergents. A conformational conversion from alpha-helical monomers into high-beta-sheet aggregates and fibrils was induced by low concentrations of the detergent sarkosyl; however, the aggregates had neither infectivity nor the characteristic protease-resistance ofPrP-res.  相似文献   

2.
Prion protein (PrP) is usually attached to membranes by a glycosylphosphatidylinositol-anchor that associates with detergent-resistant membranes (DRMs), or rafts. To model the molecular processes that might occur during the initial infection of cells with exogenous transmissible spongiform encephalopathy (TSE) agents, we examined the effect of membrane association on the conversion of the normal protease-sensitive PrP isoform (PrP-sen) to the protease-resistant isoform (PrP-res). A cell-free conversion reaction approximating physiological conditions was used, which contained purified DRMs as a source of PrP-sen and brain microsomes from scrapie-infected mice as a source of PrP-res. Interestingly, DRM-associated PrP-sen was not converted to PrP-res until the PrP-sen was either released from DRMs by treatment with phosphatidylinositol-specific phospholipase C (PI-PLC), or the combined membrane fractions were treated with the membrane-fusing agent polyethylene glycol (PEG). PEG-assisted conversion was optimal at pH 6--7, and acid pre-treating the DRMs was not sufficient to permit conversion without PI-PLC or PEG, arguing against late endosomes/lysosomes as primary compartments for PrP conversion. These observations raise the possibility that generation of new PrP-res during TSE infection requires (i) removal of PrP-sen from target cells; (ii) an exchange of membranes between cells; or (iii) insertion of incoming PrP-res into the raft domains of recipient cells.  相似文献   

3.
Transmissible spongiform encephalopathies are accompanied by the accumulation of a pathologic isoform of a host-encoded protein, termed prion protein (PrP). Despite the widespread distribution of the cellular isoform of PrP (protease-sensitive PrP; PrP-sen), the disease-associated isoform (protease-resistant PrP; PrP-res) appears to be primarily restricted to cells of the nervous and lymphoreticular systems. In order to study why scrapie infection appears to be restricted to certain cells, we followed acute and persistent PrP-res formation upon exposure of cells to different scrapie agents. We found that, independent of the cell type and scrapie strain, initial PrP-res formation occurred rapidly in cells. However, sustained generation of PrP-res and persistent infection did not necessarily follow acute PrP-res formation. Persistent PrP-res formation and scrapie infection was restricted to one cell line inoculated with the mouse scrapie strain 22L. In contrast to cells that did not become scrapie-infected, the level of PrP-res in the 22L-infected cells rapidly increased in the absence of a concomitant increase in the number of PrP-res-producing cells. Furthermore, the protein banding pattern of PrP-res in these cells changed over time as the cells became chronically infected. Thus, our results suggest that the events leading to the initial formation of PrP-res may differ from those required for sustained PrP-res formation and infection. This may, at least in part, explain the observation that not all PrP-sen-expressing cells appear to support transmissible spongiform encephalopathy agent replication.  相似文献   

4.
Previous studies have indicated that scrapie infection results in the accumulation of a proteinase K-resistant form of an endogenous brain protein generally referred to as prion protein (PrP). The molecular nature of the scrapie-associated modification of PrP accounting for proteinase K resistance is not known. As an approach to understanding the cellular events associated with the PrP modification in brain tissue, we sought to identify proteinase K-resistant PrP (PrP-res) in scrapie-infected neuroblastoma cells in vitro and to compare properties of PrP-res with those of its normal proteinase K-sensitive homolog, PrP-sen. PrP-res was detected by immunoblot in scrapie-infected but not uninfected neuroblastoma clones. Densitometry of immunoblots indicated that there was two- to threefold more PrP-res than PrP-sen in one infected clone. Metabolic labeling and membrane immunofluorescence experiments indicated that PrP-sen was located on the cell surface and could be removed from intact cells by phosphatidylinositol-specific phospholipase C and proteases. In contrast, PrP-res was not removed after reaction with these enzymes. Thus, either the scrapie-associated PrP-res was not on the cell surface or it was there in a form that is resistant to these hydrolytic enzymes. Attempts to detect intracellular PrP-res by immunofluorescent staining of fixed and permeabilized cells revealed that PrP was present in discrete perinuclear Golgi-like structures. However, the staining pattern was similar in both scrapie-infected and uninfected clones, and thus the intracellular staining may have represented only PrP-sen. Analysis of scrapie infectivity in cells treated with extracellular phospholipase, proteinase K, and trypsin indicated that, like PrP-res, the scrapie agent was not removed from the infected cells by any of these enzymes.  相似文献   

5.
Transmissible spongiform encephalopathies (TSEs) are neurological diseases that are associated with the conversion of the normal host-encoded prion protein (PrP-sen) to an abnormal protease-resistant form, PrP-res. Transmission of the TSE agent from one species to another is usually inefficient and accompanied by a prolonged incubation time. Species barriers to infection by the TSE agent are of particular importance given the apparent transmission of bovine spongiform encephalopathy to humans. Among the few animal species that appear to be resistant to infection by the TSE agent are rabbits. They survive challenge with the human kuru and Creutzfeldt-Jakob agents as well as with scrapie agent isolated from sheep or mice. Species barriers to the TSE agent are strongly influenced by the PrP amino acid sequence of both the donor and recipient animals. Here we show that rabbit PrP-sen does not form PrP-res in murine tissue culture cells persistently infected with the mouse-adapted scrapie agent. Unlike other TSE species barriers that have been studied, critical amino acid residues that inhibit PrP-res formation are located throughout the rabbit PrP sequence. Our results suggest that the resistance of rabbits to infection by the TSE agent is due to multiple rabbit PrP-specific amino acid residues that result in a PrP structure that is unable to refold to the abnormal isoform associated with disease.  相似文献   

6.
Prion protein (PrP) is usually bound to membranes by a glycosylphosphatidylinositol (GPI) anchor that associates with detergent-resistant membranes, or rafts. To examine the effect of membrane association on the interaction between the normal protease-sensitive PrP isoform (PrP-sen) and the protease-resistant isoform (PrP-res), a model system was employed using PrP-sen reconstituted into sphingolipid-cholesterol-rich raft-like liposomes (SCRLs). Both full-length (GPI(+)) and GPI anchor-deficient (GPI(-)) PrP-sen produced in fibroblasts stably associated with SCRLs. The latter, alternative mode of membrane association was not detectably altered by glycosylation and was markedly reduced by deletion of residues 34-94. The SCRL-associated PrP molecules were not removed by treatments with either high salt or carbonate buffer. However, only GPI(+) PrP-sen resisted extraction with cold Triton X-100. PrP-sen association with SCRLs was pH-independent. PrP-sen was also one of a small subset of phosphatidylinositol-specific phospholipase C (PI-PLC)-released proteins from fibroblast cells found to bind SCRLs. A cell-free conversion assay was used to measure the interaction of SCRL-bound PrP-sen with exogenous PrP-res as contained in microsomes. SCRL-bound GPI(+) PrP-sen was not converted to PrP-res until PI-PLC was added to the reaction or the combined membrane fractions were treated with the membrane-fusing agent polyethylene glycol (PEG). In contrast, SCRL-bound GPI(-) PrP-sen was converted to PrP-res without PI-PLC or PEG treatment. Thus, of the two forms of raft membrane association by PrP-sen, only the GPI anchor-directed form resists conversion induced by exogenous PrP-res.  相似文献   

7.
In view of the effectiveness of antimalaria drugs inhibiting abnormal protease-resistant prion protein (PrP-res) formation in scrapie agent-infected cells, we tested other antimalarial compounds for similar activity. Mefloquine (MF), a quinoline antimalaria drug, was the most active compound tested against RML and 22L mouse scrapie agent-infected cells, with 50% inhibitory concentrations of approximately 0.5 and approximately 1.2 microM, respectively. However, MF administered to mice did not delay the onset of intraperitoneally inoculated scrapie agent, the result previously observed with quinacrine. While most anti-scrapie agent compounds inhibit PrP-res formation in vitro, many PrP-res inhibitors have no activity in vivo. This underscores the importance of testing promising candidates in vivo.  相似文献   

8.
The scrapie prion protein (PrPSc) is derived from a cellular isoform (PrPC) that acquires protease resistance posttranslationally. We have used several different experimental approaches in attempts to reconstitute in vitro the processes leading to protease-resistant PrPSc molecules. In the first study, we performed mixing experiments by adding mouse PrP 27-30 (MoPrP27-30), the protease-resistant core of PrPSc, to PrPC and then incubating the mixture to investigate the possibility of heterodimer formation as a first step in prion replication. We used epitopically tagged PrP molecules, synthesized in murine neuroblastoma (N2a) cells transfected with the chimeric mouse/Syrian hamster MHM2 PrP construct, which are recognized by the Syrian hamster-specific monoclonal antibody 3F4. After as long as 24 h of incubation, the reaction mixture was assayed for heterodimeric intermediates of MHM2 PrPC and MoPrPSc and for protease-resistant 3F4-reactive PrP. We were unable to identify any aggregates of MHM2 PrPC and MoPrPSc on immunoblots; furthermore, we did not observe de novo formation of protease-resistant MHM2 PrP. In a second study, MoPrPC was metabolically radiolabeled in scrapie prion-infected N2a cultured cells, and then the cell extract was homogenized and incubated under various conditions to allow for the formation of protease-resistant MoPrPSc. We observed no radiolabeled MoPrPSc by immunoprecipitation after as long as 24 h of in vitro incubation. In a third approach, Syrian hamster PrP (SHaPrP) was synthesized in a cell-free translation system supplemented with microsomal membranes derived from either normal or scrapie prion-infected cultured cells. We found that all SHaPrP species translocated across microsomal membranes from scrapie prion-infected cells were protease sensitive in the presence of detergents and displayed the same topology as those generated by microsomes from normal cells or from dog pancreas. We also studied PrP molecules that encode the codon 102 mutation that causes the rare human prion disease Gerstmann-Str?ussler-Scheinker (GSS) syndrome. On the basis of our data, GSSPrP appears to yield topological forms similar to those of the wild-type PrP when processed by either normal or scrapie prion-derived microsomes.  相似文献   

9.
Scrapie prion infectivity can be enriched from hamster brain homogenates by using limited proteolysis and detergent extraction. Purified fractions contain both scrapie infectivity and the protein PrP 27-30, which is aggregated in the form of prion rods. During purification, PrP 27-30 is produced from a larger membrane protein, PrPSc, by limited proteolysis with proteinase K. Brain homogenates from scrapie-infected hamsters do not contain prion rods prior to exposure to detergents and proteases. To determine whether both detergent extraction and limited proteolysis are required for the formation of prion rods, microsomal membranes were prepared from infected brains in the presence of protease inhibitors. The isolated membranes were then detergent extracted as well as protease digested to evaluate the effects of these treatments on the formation of prion rods. Neither detergent (2% Sarkosyl) extraction nor limited proteinase K digestion of scrapie microsomes produced recognizable prion amyloid rods. Only after combining detergent extraction with limited proteolysis were numerous prion rods observed. Rod formation was influenced by the protease concentration, the specificity of the protease, and the duration of digestion. Rod formation also depended upon the detergent; some combinations of protease and detergent did not produce prion amyloid rods. Similar results were obtained with purified PrPSc fractions prepared by repeated detergent extractions in the presence of protease inhibitors. These fractions contained amorphous structures but not rods; however, prion rods were produced upon conversion of PrPSc to PrP 27-30 by limited proteolysis. We conclude that the formation of prion amyloid rods in vitro requires both detergent extraction and limited proteolysis. In vivo, amyloid filaments found in the brains of animals with scrapie resemble prion rods in their width and their labeling with prion protein (PrP) antisera; however, filaments are typically longer than rods. Whether limited proteolysis and some process equivalent to detergent extraction are required for amyloid filament formation in vivo remains to be established.  相似文献   

10.
Normal cellular and abnormal disease-associated forms of prion protein (PrP) contain a C-terminal glycophosphatidyl-inositol (GPI) membrane anchor. The importance of the GPI membrane anchor in prion diseases is unclear but there are data to suggest that it both is and is not required for abnormal prion protein formation and prion infection. Utilizing an in vitro model of prion infection we have recently demonstrated that, while the GPI anchor is not essential for the formation of abnormal prion protein in a cell, it is necessary for the establishment of persistent prion infection. In combination with previously published data, our results suggest that GPI anchored PrP is important in the amplification and spread of prion infectivity from cell to cell.Key words: prion, GPI anchor, PrP, prion spread, scrapieIn transmissible spongiform encephalopathies (TSE or prion diseases) such as sheep scrapie, bovine spongiform encephalopathy and human Creutzfeldt-Jakob disease, normally soluble and protease-sensitive prion protein (PrP-sen or PrPC) is converted to an abnormal, insoluble and protease-resistant form termed PrP-res or PrPSc. PrP-res/PrPSc is believed to be the main component of the prion, the infectious agent of the TSE/prion diseases. Its precursor, PrP-sen, is anchored to the cell surface at the C-terminus by a co-translationally added glycophosphatidyl-inositol (GPI) membrane anchor which can be cleaved by the enzyme phosphatidyl-inositol specific phospholipase (PIPLC). The GPI anchor is also present in PrP-res, but is inaccessible to PIPLC digestion suggesting that conformational changes in PrP associated with PrP-res formation have blocked the PIPLC cleavage site.1 Although the GPI anchor is present in both PrP-sen and PrP-res, its precise role in TSE diseases remains unclear primarily because there are data to suggest that it both is and is not necessary for PrP-res formation and prion infection.In tissue culture cells infected with mouse scrapie, PrP-res formation occurs at the cell surface and/or along the endocytic pathway24 and may be dependent upon the membrane environment of PrP-sen. For example, localization via the GPI anchor to caveolae-like domains favors PrP-res formation5 while substitution of the GPI anchor addition site with carboxy termini favoring transmembrane anchored PrP-sen inhibits formation of PrP-res.5,6 Other studies have shown that localization of both PrP-sen and PrP-res to lipid rafts, cholesterol and sphingolipid rich membrane microdomains where GPI anchored proteins can be located, is important in PrP-res formation.69However, there are also data which suggest that such localization is not necessarily essential for PrP-res formation. Anchorless PrP-sen isolated from cells by immunoprecipitation or wild-type PrP-sen purified by immunoaffinity column followed by cation exchange chromatography are efficiently converted into PrP-res in cell-free systems.10,11 Furthermore, recombinant PrP-sen derived from E. coli, which has no membrane anchor or glycosylation, can be induced to form protease-resistant PrP in vitro when reacted with prion-infected brain homogenates.1214 Finally, in at least one instance, protease-resistant recombinant PrP-res generated in the absence of infected brain homogenate was reported to cause disease when inoculated into transgenic mice.15The data concerning the role of the PrP-sen GPI anchor in susceptibility to TSE infection are similarly contradictory. Transgenic mice expressing anchorless mouse PrP-sen are susceptible to infection with mouse scrapie and accumulate both PrP-res and prion infectivity.16 Thus, the GPI anchor is clearly not needed for PrP-res formation or productive TSE infection in vivo. However, we recently published data demonstrating that, in vitro, anchored PrP-sen is in fact required to persistently infect cells.17 Given that anchorless PrP-sen is not present on the cell surface but is released into the cell medium, we speculated that the differences between the in vitro and in vivo data were related to the location of PrP-res formation. In the mice expressing anchorless PrP-sen, environments conducive to PrP-res formation are present in certain areas of the complex extracellular milieu of the brain where anchorless, secreted PrP-sen can accumulate and come into contact with PrP-res from the infectious inoculum. Since similar environments are missing in vitro, any PrP-res formation in cells expressing anchorless PrP-sen must be cell-associated. While this explanation addresses how extracellular PrP-res could be generated in an unusual transgenic mouse model of TSE infection, it does not really help to define how the GPI anchor is involved in normal prion infection of a cell.As with other infectious organisms such as viruses, TSE infection can be roughly divided into three steps: uptake, replication and spread. Over the last several years, data derived from new techniques as well as new cell lines susceptible to prion infection have increased our knowledge of some of the basic events that occur during each of these steps. In order to try to tease out the role of the GPI anchor in normal TSE pathogenesis, it is therefore useful to consider the process of TSE infection of a cell and how the GPI anchor might be involved in each stage.In a conventional viral infection, binding and uptake of the virus is essential to establish infection. Studying PrP-res uptake has been complicated by the lack of an antibody that can specifically distinguish PrP-res from PrP-sen in live cells and by the difficulty of detecting the input PrP-res from the PrP-res made de novo by the cell. Recently, however, several groups have been able to study PrP-res uptake using input PrP-res that was either fluorescently labeled1820 or tagged with the epitope to the monoclonal antibody 3F4,21 or cell lines that express little or no PrP-sen.19,2123 The data show that PrP-res uptake is independent of scrapie strain or cell type but is influenced by the PrP-res microenvironment as well as PrP-res aggregate size.21 Importantly, these studies demonstrated that PrP-sen expression was not required.19,2123 Given these data, it is clear that GPI anchored PrP-sen is not involved in the initial uptake of PrP-res into the cell.The next stage of prion infection involves replication of infectivity which is typically assayed by following cellular PrP-res formation. Once again, however, the issue of how to distinguish PrP-res in the inoculum from newly formed PrP-res in the cells has made it difficult to study the early stages of prion replication. To overcome this difficulty, we developed a murine tissue culture system that utilizes cells expressing mouse PrP-sen tagged with the epitope to the 3F4 antibody (Mo3F4 PrP-sen).24 Wild-type mouse PrP does not have this epitope. As a result, following exposure to an infected mouse brain homogenate, de novo PrP-res formation can be followed by assaying for 3F4 positive PrP-res. Our studies showed that there were two stages of PrP-res formation: (1) an initial acute burst within the first 96 hours post-infection that was cell-type and scrapie strain independent and, (2) persistent PrP-res formation (i.e., formation of PrP-res over multiple cell passages) that was dependent on cell-type and scrapie strain and associated with long-term infection.24 Acute PrP-res formation did not necessarily lead to persistent PrP-res formation suggesting that other cell-specific factors or processes are needed for PrP-res formation to persist.24When cells expressing Mo3F4 PrP-sen without the GPI anchor (Mo3F4 GPI-PrP-sen) were exposed to mouse scrapie infected brain homogenates, GPI negative, 3F4 positive PrP-res (Mo3F4 GPI-PrP-res) was detected within 96 hours indicating that acute PrP-res formation had occurred.17 Thus, despite the fact that Mo3F4 GPI-PrP-sen is not expressed on the cell surface16 (Fig. 1A), it was still available for conversion to PrP-res. These results are consistent with data from cell-free systems and demonstrate that, at least acutely, membrane anchored PrP is not necessary for PrP-res formation in a cell.Open in a separate windowFigure 1Persistent infection of cells in vitro requires the expression of GPI-anchored cell surface PrP-sen. PrP knockout cells (CF10)21 were transduced with 3F4 epitope tagged mouse PrP-sen (Mo3F4), 3F4 epitope tagged mouse PrP-sen without the GPI anchor (Mo3F4 GPI-), or Mo3F4 GPI-PrP-sen plus wild-type, GPI anchored mouse PrP-sen (MoPrP). The cells were then exposed to the mouse scrapie strain 22L and passaged. (A) The presence of 3F4 epitope tagged, cell surface mouse PrP-sen was assayed by FACS analysis of fixed, non-permeabilized cells. CF10 cells expressing the following mouse PrP-sen molecules were assayed: Mo3F4 (solid line); Mo3F4 GPI (dashed line); Mo3F4 GPI + MoPrP (dotted and dashed line); Mo3F4 GPI + MoPrP infected with 22L scrapie (dotted line). Only cells expressing Mo3F4 PrP-sen were positive for cell surface, 3F4 epitope tagged PrP. (B) Persistent infection was analyzed by inoculating the cells intracranially into transgenic mice overexpressing MoPrP (Tga20 mice). Only cells expressing anchored mouse PrP-sen were susceptible to scrapie infection. Cells expressing anchorless mouse PrP-sen did not contain detectable infectivity in either the cells or the cellular supernatant (data not shown). Data in (B) are adapted from McNally 2009.17In terms of persistent PrP-res formation, however, our data suggest that the GPI anchor is important. Despite an initial burst of PrP-res formation within the first 96 hours post-infection, Mo3F4 GPI-PrP-res was not observed following passage of the cells nor did the cells become infected. This effect was not due either to resistance of the cells to scrapie infection or to an inability of the scrapie strain used to infect cells. When the same cells expressed anchored Mo3F4 PrP-sen and were exposed to the same mouse scrapie strain, both acute and persistent PrP-res formation were detected and the cells were persistently infected with scrapie (Fig. 1B).17 Taken together, these data demonstrate that cells expressing anchorless PrP-sen do not support persistent PrP-res formation. Furthermore, the data strongly suggest that GPI-anchored PrP-sen is required during the transition from acute to persistent scrapie infection. In support of this hypothesis, the resistance of cells expressing Mo3F4 GPI-PrP-sen to persistent prion infection could be overcome if wild-type GPI anchored PrP-sen was co-expressed in the same cell. When both forms of PrP-sen were expressed, anchored and anchorless forms of PrP-res were made and the cells became persistently infected (Fig. 1B).17 Thus, the data suggest that GPI anchored PrP is necessary to establish prion infection within a cell.How could GPI membrane anchored PrP be involved in the establishment and maintenance of persistent prion infection? Several studies have suggested that the GPI anchor is needed to localize PrP-sen to specific membrane environments where PrP-res formation is favored.58 However, if this localization was essential for PrP-res formation, GPI-PrP-sen would presumably never form PrP-res. Lacking the GPI anchor, it would not be in the correct membrane environment to support conversion. As a result, neither acute nor persistent prion infection could occur. This is obviously not the case. Transgenic mice expressing only anchorless PrP-sen generate PrP-res and can be infected with scrapie even though (1) flotation gradients showed that anchorless PrP-sen was not in the same membrane environment as anchored PrP-sen and, (2) flow cytometry analysis demonstrated that anchorless PrP-sen was not present on the cell surface.16 Thus, the GPI anchor is not needed to target PrP-sen to a conversion friendly membrane environment.Consistent with the idea that the GPI anchor is not essential for PrP-res formation, in our studies anchorless PrP-sen could form PrP-res in cells acutely infected with scrapie despite the fact that it is processed differently than anchored PrP-sen, is not present on the cell surface (Fig. 1A), and is secreted.17 Persistent formation of anchorless PrP-res only occurred when both anchored and anchorless forms of PrP were expressed in the same cell.17 For this to happen both types of PrP must share a cellular compartment where PrP-res formation occurs, presumably either on the cell surface or in a specific location along the endocytic pathway2,3 such as the endosomal recycling compartment.4 Analysis of infected and uninfected cells co-expressing Mo3F4 GPI-PrP-sen and wild-type PrP-sen demonstrated that Mo3F4 GPI-PrP-sen was not present on the cell surface (Fig. 1A). Thus, it is unlikely that GPI-PrP-res formation is occurring on the cell surface. We speculate that the anchored form of PrP-res encounters anchorless PrP-sen along either a secretory or endocytic pathway, allowing for the formation of anchorless PrP-res. Regardless of the precise location, the in vitro and in vivo data strongly suggest that the role of the anchor in persistent prion infection is not simply to localize PrP-sen to an environment compatible with PrP-res formation.However, the data are consistent with the idea that GPI anchored PrP is absolutely essential for the establishment of persistent infection in vitro. This is likely related to the spread of infectivity within a culture that is necessary for maintaining a persistent infection over time. Evidence suggests that PrP-res can be transferred between cells in a variety of ways including mother-daughter cell division,25 cell-to-cell contact26,27 and exosomes.28 Tunneling nanotubes have also been hypothesized to be involved in intercellular prion spread19 and recent data suggest that spread can occur via these structures.20 Any of these processes could involve the cell-to-cell transfer of PrP-res in membrane containing particles as has been observed in cell-free7 and cell-based systems.29 If cell-to-cell contact were required, for example via simple physical proximity or perhaps tunneling nanotubes,19,20 then the conversion of cell surface PrP-sen on the naïve cell by cell surface PrP-res on the infected cell would transfer infection to the naïve cell. In this instance, GPI membrane anchored, cell surface PrP-sen would be essential as it would allow for PrP-res formation on the cell surface. If spread is via cell division, then GPI-anchored, cell surface PrP-sen would be important for its role as a precursor to PrP-res formation.2 In this instance, cell surface PrP-sen would be an essential intermediate in the continuous formation of PrP-res necessary for the accumulation and amplification of PrP-res within the cell. It would also help to cycle PrP between the cell surface and intracellular compartments where PrP-res can be formed.4 In either case, GPI-anchored PrP-sen would facilitate the accumulation of intracellular PrP-res to high enough levels to maintain both persistent infection in the mother cell and enable the transfer of organelles containing sufficient PrP-res to initiate infection in the daughter cell. Thus, we would suggest that efficient spread of infectivity requires not just the passive transfer of PrP-res from cell-to-cell but the concurrent initiation of conversion and amplification of PrP-res via cell surface, GPI anchored PrP-sen.In vivo, several lines of evidence suggest that the spread of scrapie infectivity also requires de novo PrP-res formation in the recipient cell and not simply transfer of PrP-res from one cell to another. For example, when neurografts from PrP expressing mice were placed in the brains of PrP knockout mice and the mice were challenged intracranially with scrapie, the graft showed scrapie pathology, but the surrounding tissue did not.30 Furthermore, PrP-res from the graft migrated to the host tissue demonstrating that simple transfer of PrP-res was not sufficient and that PrP-sen expression was required for the spread of scrapie pathology.30 In fact, these mice did not develop scrapie pathology following peripheral infection even when peripheral lymphoid tissues were reconstituted with PrP-sen expressing cells.31 Even though PrP-sen expressing cells were present in both the brain and spleen, in order for infectivity to spread from the lymphoreticular system to the central nervous system PrP-sen expression was also required in an intermediate tissue such as peripheral nerve.31,32 Given that PrP-res uptake and trafficking do not require PrP-sen, the most obvious explanation for the requirement of PrP-sen in contiguous tissues is that de novo PrP-res formation in naïve cells is necessary for (1) infectivity to move from cell to cell within a tissue and, (2) infectivity to move from tissue to tissue.Another study demonstrated that peripheral expression of heterologous mouse PrP significantly increased the incubation time and actually prevented clinical disease in the majority of transgenic mice expressing hamster PrP in neurons of the brain.33 Once again, if simple transfer and uptake of PrP-res were sufficient for spread, the presence of heterologous PrP molecules should not interfere because cellular uptake of PrP-res is independent of PrP-sen expression.19,2123 Clinical disease in these mice was likely prevented by the heterologous PrP molecule interfering with conversion of PrP-sen to PrP-res suggesting that prevention of de novo PrP-res formation inhibits spread of PrP-res and infectivity. These in vivo data, when combined with our recent in vitro data,17 provide evidence to support the importance of cell surface, and by extension GPI-anchored, PrP in the spread of prion infection.Our data demonstrate that the GPI anchor plays a role in the establishment of persistent scrapie infection in vitro. In our tissue culture system,21 as well as others where spread of infectivity by cell to cell contact appears to be limited,25,34 the role of GPI anchored PrP-sen would be to amplify PrP-res to enable the efficient transfer of infectivity from mother to daughter cell. In cell systems where spread of prion infectivity may require cell to cell contact,26,27 we propose that the role of GPI anchored PrP-sen is to facilitate the spread of prion infection via a chain of conversion from cell-to-cell, a “domino” type spread of infection that has been previously hypothesized.35,36In vivo, such a mechanism might explain why neuroinvasion does not necessarily require axonal transport32,37,38 and can occur independently of the axonal neurofilament machinery.39 It would likely vary with cell type27 and be most important in areas where infectivity is transferred from the periphery to the nervous system as well as in areas where cell division may be limited. It is also possible, if the location of PrP-res formation differs for different scrapie strains,40 that the relative importance of a domino-like spread of infectivity in vivo would vary with the scrapie strain.Of course, spread of infectivity via a “wave” of GPI anchored, PrP mediated conversion would not preclude the spread of infectivity by other intracellular means such as axonal transport (reviewed in ref. 41). Furthermore, spread of infectivity may still also occur extracellularly such as in the unique case of mice which express anchorless PrP-sen,16 where our in vitro data would suggest that the cells themselves are not infected. In such a case, spread would require neither GPI anchored PrP-sen nor amplification of PrP-res in cells but would likely occur via other means such as blood41 or interstitial fluid flow.42  相似文献   

11.
Inhibition of the accumulation of protease-resistant prion protein (PrP-res) is a prime strategy in the development of potential transmissible spongiform encephalopathy (TSE) therapeutics. Here we show that curcumin (diferoylmethane), a major component of the spice turmeric, potently inhibits PrP-res accumulation in scrapie agent-infected neuroblastoma cells (50% inhibitory concentration, approximately 10 nM) and partially inhibits the cell-free conversion of PrP to PrP-res. In vivo studies showed that dietary administration of curcumin had no significant effect on the onset of scrapie in hamsters. Nonetheless, other studies have shown that curcumin is nontoxic and can penetrate the brain, properties that give curcumin advantages over inhibitors previously identified as potential prophylactic and/or therapeutic anti-TSE compounds.  相似文献   

12.
Rough-surfaced and light and heavy smooth-surfaced microsomes were isolated from rat brain by means of discontinuous sucrose gradient centrifugation. Electron microscopically, the rough-surfaced microsomes were characterized by vesicles with ribosomes and the light and heavy smooth-surfaced microsomes by fairly homogeneous membrane features without ribosomes. The rough-surfaced microsomal membranes were distinguished by the absence of glycolipids, such as ganglioside, cerebroside, and sulfatide. Cerebroside was exclusively recovered in the light smooth-surfaced microsomal membranes. Ganglioside and Na,K-ATPase were contained in larger amounts in the heavy smooth-surfaced microsomal membranes than in the light smooth-surfaced microsomal membranes in terms of protein content. Among the three submicrosomal membranes, cholesterol and phospholipid were found in the largest amounts in the light smooth-surfaced microsomal membranes, where the molar ratio of cerebroside-cholesterol-phospholipid was about 1:10:10. The membranes of rough- and smooth-surfaced microsomes were very similar in regards to the composition of phospholipid classes, although the fatty acid composition of the former contained a greater proportion of unsaturated fatty acids than that of the latter. When the membrane proteins were analyzed by sodium dodecyl sulfate gel electrophoresis, some differences were observed between the light and heavy smooth-surfaced microsomal membranes.  相似文献   

13.
Mutations within a host cellular protein, PrP, have been associated with disease in the transmissible spongiform encephalopathies. Murine neuroblastoma cells persistently infected with mouse scrapie accumulate protease-resistant PrP (PrP-res), the abnormal form of PrP associated with disease in the transmissible spongiform encephalopathies. These cells provide a controlled system in which to study the molecular interactions which are important in the formation of PrP-res. We have expressed recombinant PrP molecules in mouse scrapie-infected murine neuroblastoma cells and assayed the effect of these heterologous PrP genes on the formation and accumulation of PrP-res. The results demonstrate that expression of heterologous PrP molecules which differ from the endogenous PrP by as little as one amino acid can profoundly interfere with the overall accumulation of PrP-res. The data suggest that precise interactions between homologous PrP molecules are important in PrP-res accumulation and that heterologous PrP molecules can block these interactions.  相似文献   

14.
1. Sonication of bovine liver microsomes completely solubilized the membrane-bound lysophospholipase II (EC 3.1.1.5). Co-chromatography with purified 125I-labelled lysophospholipase indicated that the enzyme was solubilized from microsomes in a lipid-free state. 2. In the presence of residual microsomal membranes, the solubilized lysophospholipase could only be partly degraded by trypsin (EC 3.4.21.4). Therefore, trypsin could not be used to study the transmembrane disposition of lysophospholipase in intact microsomes. 3. Chymotrypsin (EC 3.4.21.1) destroyed the solubilized lysophospholipase activity, even in the presence of residual microsomal membranes. 4. Lysophospholipase in intact microsomal vesicles was resistant to chymotrypsin digestion. 5. When microsomal vesicles were made leaky with lysophosphatidylcholine, chymotrypsin destroyed more than 95% of the lysophospholipase activity. 6. It is concluded from these experiments that at least the active center of lysophospholipase is located at the luminal side of the bovine liver microsomal membrane.  相似文献   

15.
《朊病毒》2013,7(3):134-138
In transmissible spongiform encephalopathies (TSE or prion diseases) such as sheep scrapie, bovine spongiform encephalopathy and human Creutzfeldt-Jakob disease, normally soluble and protease-sensitive prion protein (PrP-sen or PrPC) is converted to an abnormal, insoluble and protease-resistant form termed PrP-res or PrPSc. PrP-res/PrPSc is believed to be the main component of the prion, the infectious agent of the TSE/prion diseases. Its precursor, PrP-sen, is anchored to the cell surface at the C-terminus by a co-translationally added glycophosphatidyl-inositol (GPI) membrane anchor which can be cleaved by the enzyme phosphatidyl-inositol specific phospholipase (PIPLC). The GPI anchor is also present in PrP-res, but is inaccessible to PIPLC digestion suggesting that conformational changes in PrP associated with PrP-res formation have blocked the PIPLC cleavage site. Although the GPI anchor is present in both PrP-sen and PrP-res, its precise role in TSE diseases remains unclear primarily because there are data to suggest that it both is and is not necessary for PrP-res formation and prion infection.  相似文献   

16.
Transmissible spongiform encephalopathy (TSE) diseases are characterized by the accumulation in brain of an abnormal protease-resistant form of the host-encoded prion protein (PrP), PrP-res. PrP-res conformation differs among TSE agents derived from various sources, and these conformational differences are thought to influence the biological characteristics of these agents. In this study, we introduced deletions into the flexible N-terminal region of PrP (residues 34-124) and investigated the effect of this region on the conformation of PrP-res generated in an in vitro cell-free conversion assay. PrP deleted from residues 34 to 99 generated 12-16-kDa protease-resistant bands with intact C termini but variable N termini. The variable N termini were the result of exposure of new protease cleavage sites in PrP-res between residues 130 and 157, suggesting that these new cleavage sites were caused by alterations in the conformation of the PrP-res generated. Similarly truncated 12-16-kDa PrP bands were also identified in brain homogenates from mice infected with mouse-passaged hamster scrapie as well as in the cell-free conversion assay using conditions that mimicked the hamster/mouse species barrier to infection. Thus, by its effects on PrP-res conformation, the flexible N-terminal region of PrP seemed to influence TSE pathogenesis and cross-species TSE transmission.  相似文献   

17.
A comparative study of proteins adsorbed on outer surface of microsomal membranes was carried out. Electrophoretic differences between endoplasmic reticulum proteins from liver and brain cells were revealed. These differences were not observed in the presence of sodium dodecyl sulphate. Proteins of brain microsomes are shown to bind in vitro with membranes of brain endoplasmic reticulum to a higher extent than with liver microsomal membranes.  相似文献   

18.
Amphotericin B delays the onset of clinical symptoms in hamsters infected with scrapie agent strain 263K. Here we show that accumulation of a scrapie-specific isoform of the prion protein (PrP-res) and agent replication were delayed early in amphotericin B-treated animals. By 8 weeks postinfection, only untreated animals exhibited clinical symptoms of scrapie infection whereas PrP-res levels and titers were similar in treated and untreated animals. This suggests that although PrP-res accumulation and agent replication are linked, they are not the sole factors required for the onset of clinical disease.  相似文献   

19.
D Lazard  N Tal  M Rubinstein  M Khen  D Lancet  K Zupko 《Biochemistry》1990,29(32):7433-7440
Two major transmembranal polypeptides of bovine olfactory epithelium were identified by SDS electrophoretic analysis of Triton X-114 solubilized membranes. Both polypeptides were present in large amounts in membranes of the olfactory epithelium but were barely detectable in membranes of the nasal respiratory epithelium. Both polypeptides are enriched in the deciliated epithelium as compared with isolated cilia. One of them is a glycoprotein with an apparent molecular mass of 56 kDa (gp56); the other is an unglycosylated protein with an apparent molecular mass of 52 kDa (p52). Sequence analysis of peptides obtained by CNBr cleavage of purified gp56 indicates that it is highly homologous to UDP-glucuronosyl transferase (UDPGT). Parallel analysis shows that p52 is highly homologous to cytochrome P-450 sequences of the IIA subfamily. This protein is assigned the name P-450olf2. Polyclonal antibodies were raised against synthetic peptides corresponding to gp56 and p52 peptide sequences. Immunoblots with these antibodies reveal the following properties of gp56 and p52: (1) they are enriched in the microsomal fraction of the bovine olfactory epithelium; (2) they are possibly specific to the olfactory epithelium, as we could not detect reactivity in microsomes derived from respiratory epithelium or lung, and only a very small amount of basal reactivity was seen with liver microsomes; (3) cross-reacting proteins exist in microsomes derived from the rat olfactory epithelium. These results are consistent with a mechanism whereby the microsomal enzymes are involved in odorant modification and clearance from the nasal tissue.  相似文献   

20.
Abstract— The fraction that sediments between 2 × 105 g -min and 6 × 106 g -min from dilute dispersions of rat brain in 0.32 m -sucrose is a microsomal fraction with very little contamination by myelin. A crude microsomal fraction prepared in the same way from rat spinal cord contains more myelin than microsomes. Centrifugation of the crude microsomal fraction in 0.85 m -sucrose gave a floating fraction, an infranatant fraction (purified microsomes) and a small pellet. The purified microsomes contained very little myelin as judged by electron microscopy and polyacrylamide gel electrophoresis. The lipid composition resembled that of spinal cord myelin except that the purified microsomes contained relatively less cholesterol and ethanolamine plasmalogens. The content of galactolipids was much greater in spinal cord microsomes than in brain microsomes. The spinal cord CDP-ethanol-amine:diglyceride ethanolaminephosphotransferase activity (EC 2.7.8.1) was concentrated in the purified microsomes.
A spinal cord myelin fraction isolated from the 2 × 105 g -min pellet was quite pure as judged by electron microscopy, enzyme activities and polyacrylamide gel electrophoresis. No NADPH-cyto-chrome c reductase activity (EC 1.6.2.3) could be detected in the purified myelin. The ethanolaminephosphotransferase specific activity was about 5% of that found in the purified microsomal fraction. The protein content was 25% by weight for spinal cord myelin and 31% for brain myelin. Of the total spinal cord 2',3'-cyclic nucleotide-3'-phosphohydrolase activity, 16% was lost from the crude myelin during purification, 21% was recovered in the purified myelin, and 11% was found in the floating fraction from the crude microsomes. The purified myelin and microsomal fractions from spinal cord were relatively pure. Additional myelin was recovered in the floating fraction from the crude microsomes.  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