Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
PLoS One ; 12(2): e0170503, 2017.
Article En | MEDLINE | ID: mdl-28207746

Resistance to proteolytic digestion has long been considered a defining trait of prions in tissues of organisms suffering from transmissible spongiform encephalopathies. Detection of proteinase K-resistant prion protein (PrPSc) still represents the diagnostic gold standard for prion diseases in humans, sheep and cattle. However, it has become increasingly apparent that the accumulation of PrPSc does not always accompany prion infections: high titers of prion infectivity can be reached also in the absence of protease resistant PrPSc. Here, we describe a structural basis for the phenomenon of protease-sensitive prion infectivity. We studied the effect on proteinase K (PK) resistance of the amino acid substitution Y169F, which removes a single oxygen atom from the ß2-α2 loop of the cellular prion protein (PrPC). When infected with RML or the 263K strain of prions, transgenic mice lacking wild-type (wt) PrPC but expressing MoPrP169F generated prion infectivity at levels comparable to wt mice. The newly generated MoPrP169F prions were biologically indistinguishable from those recovered from prion-infected wt mice, and elicited similar pathologies in vivo. Surprisingly, MoPrP169F prions showed greatly reduced PK resistance and density gradient analyses showed a significant reduction in high-density aggregates. Passage of MoPrP169F prions into mice expressing wt MoPrP led to full recovery of protease resistance, indicating that no strain shift had taken place. We conclude that a subtle structural variation in the ß2-α2 loop of PrPC affects the sensitivity of PrPSc to protease but does not impact prion replication and infectivity. With these findings a specific structural feature of PrPC can be linked to a physicochemical property of the corresponding PrPSc.


Brain/metabolism , Endopeptidase K/metabolism , PrPSc Proteins/metabolism , Prion Diseases/transmission , Prions/pathogenicity , Amino Acid Substitution , Animals , Brain/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed , Mutation/genetics , PrPSc Proteins/chemistry , PrPSc Proteins/genetics , Prion Diseases/pathology , Protein Conformation , Proteolysis
2.
PLoS Pathog ; 11(4): e1004808, 2015 Apr.
Article En | MEDLINE | ID: mdl-25875479

[This corrects the article DOI: 10.1371/journal.ppat.1004662.].

3.
PLoS One ; 10(2): e0117412, 2015.
Article En | MEDLINE | ID: mdl-25658480

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.


Cerebellum/pathology , PrPC Proteins/metabolism , Prion Diseases/metabolism , Prion Diseases/pathology , Unfolded Protein Response , Animals , Cerebellum/metabolism , Endoplasmic Reticulum Stress , Mice , Mice, Transgenic , PrPC Proteins/analysis , Prions
4.
PLoS Pathog ; 11(2): e1004662, 2015 Feb.
Article En | MEDLINE | ID: mdl-25710374

Prions induce lethal neurodegeneration and consist of PrPSc, an aggregated conformer of the cellular prion protein PrPC. Antibody-derived ligands to the globular domain of PrPC (collectively termed GDL) are also neurotoxic. Here we show that GDL and prion infections activate the same pathways. Firstly, both GDL and prion infection of cerebellar organotypic cultured slices (COCS) induced the production of reactive oxygen species (ROS). Accordingly, ROS scavenging, which counteracts GDL toxicity in vitro and in vivo, prolonged the lifespan of prion-infected mice and protected prion-infected COCS from neurodegeneration. Instead, neither glutamate receptor antagonists nor inhibitors of endoplasmic reticulum calcium channels abolished neurotoxicity in either model. Secondly, antibodies against the flexible tail (FT) of PrPC reduced neurotoxicity in both GDL-exposed and prion-infected COCS, suggesting that the FT executes toxicity in both paradigms. Thirdly, the PERK pathway of the unfolded protein response was activated in both models. Finally, 80% of transcriptionally downregulated genes overlapped between prion-infected and GDL-treated COCS. We conclude that GDL mimic the interaction of PrPSc with PrPC, thereby triggering the downstream events characteristic of prion infection.


Antibodies , PrPSc Proteins/immunology , Prion Diseases/chemically induced , Prion Diseases/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Animals , Antibodies/immunology , Antibodies/toxicity , Mice , Mice, Transgenic , PrPSc Proteins/genetics , Prion Diseases/genetics , Prion Diseases/pathology , Reactive Oxygen Species/immunology , Signal Transduction/genetics , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology
5.
Biochemistry ; 51(30): 5912-20, 2012 Jul 31.
Article En | MEDLINE | ID: mdl-22747506

The enzyme thiopurine S-methyltransferase (TPMT) is involved in the metabolism of thiopurine drugs used to treat acute lymphoblastic leukemia and inflammatory bowel disease. Thus far, at least 29 variants of the TPMT gene have been described, many of which encode proteins that have low enzyme activity and in some cases become more prone to aggregation and degradation. Here, the two naturally occurring variants, TPMT*2 (Ala80 → Pro) and TPMT*5 (Leu49 → Ser), were cloned and expressed in Escherichia coli. Far-UV circular dichroism spectroscopy showed that TPMT*2 was substantially destabilized whereas TPMT*5 showed much greater stability comparable to that of wild-type TPMT (TPMTwt). The extrinsic fluorescent molecule anilinonaphthalene sulfonate (ANS) was used to probe the tertiary structure during thermal denaturation. In contrast to TPMTwt, neither of the variants bound ANS to a large extent. To explore the morphology of the TPMT aggregates, we performed luminescent conjugated oligothiophene staining and showed fibril formation for TPMT*2 and TPMT*5. The differences in the flexibility of TPMTwt, TPMT*2, and TPMT*5 were evaluated in a limited proteolysis experiment to pinpoint stable regions. Even though there is only one amino acid difference between the analyzed TPMT variants, a clear disparity in the cleavage patterns was observed. TPMT*2 displays a protected region in the C-terminus, which differs from TPMTwt, whereas the protected regions in TPMT*5 are located mainly in the N-terminus close to the active site. In conclusion, this in vitro study, conducted to probe structural changes during unfolding of TPMT*2 and TPMT*5, demonstrates that the various causes of the low enzyme activity in vivo could be explained on a molecular level.


Alleles , Genetic Variation/genetics , Methyltransferases/chemistry , Methyltransferases/genetics , Amino Acid Sequence , Catalytic Domain/genetics , Enzyme Activation/genetics , Humans , Methyltransferases/metabolism , Molecular Sequence Data
6.
PLoS One ; 5(2): e9107, 2010 Feb 08.
Article En | MEDLINE | ID: mdl-20161712

The cellular form of the prion protein, PrP(C), undergoes extensive proteolysis at the alpha site (109K [see text]H110). Expression of non-cleavable PrP(C) mutants in transgenic mice correlates with neurotoxicity, suggesting that alpha-cleavage is important for PrP(C) physiology. To gain insights into the mechanisms of alpha-cleavage, we generated a library of PrP(C) mutants with mutations in the region neighbouring the alpha-cleavage site. The prevalence of C1, the carboxy adduct of alpha-cleavage, was determined for each mutant. In cell lines of disparate origin, C1 prevalence was unaffected by variations in charge and hydrophobicity of the region neighbouring the alpha-cleavage site, and by substitutions of the residues in the palindrome that flanks this site. Instead, alpha-cleavage was size-dependently impaired by deletions within the domain 106-119. Almost no cleavage was observed upon full deletion of this domain. These results suggest that alpha-cleavage is executed by an alpha-PrPase whose activity, despite surprisingly limited sequence specificity, is dependent on the size of the central region of PrP(C).


Mutation , Prions/genetics , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Glycosylation , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Knockout , Molecular Sequence Data , NIH 3T3 Cells , Peptide Hydrolases/metabolism , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPC Proteins/metabolism , PrPSc Proteins/genetics , PrPSc Proteins/metabolism , Prion Proteins , Prions/chemistry , Prions/metabolism , Sequence Homology, Amino Acid
...