Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 605
Filter
1.
Mol Cell ; 72(3): 426-443.e12, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30401430

ABSTRACT

Protein phase separation by low-complexity, intrinsically disordered domains generates membraneless organelles and links to neurodegeneration. Cellular prion protein (PrPC) contains such domains, causes spongiform degeneration, and is a receptor for Alzheimer's amyloid-ß oligomers (Aßo). Here, we show that PrPC separates as a liquid phase, in which α-helical Thr become unfolded. At the cell surface, PrPC Lys residues interact with Aßo to create a hydrogel containing immobile Aßo and relatively mobile PrPC. The Aßo/PrP hydrogel has a well-defined stoichiometry and dissociates with excess Aßo. NMR studies of hydrogel PrPC reveal a distinct α-helical conformation for natively unfolded amino-terminal Gly and Ala residues. Aßo/PrP hydrogel traps signal-transducing mGluR5 on the plasma membrane. Recombinant PrPC extracts endogenous Aßo from human Alzheimer's soluble brain lysates into hydrogel, and a PrPC antagonist releases Aßo from endogenous brain hydrogel. Thus, coupled phase and conformational transitions of PrPC are driven by Aß species from Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/physiology , PrPC Proteins/chemistry , PrPC Proteins/physiology , Alzheimer Disease/metabolism , Animals , Brain , COS Cells , Cell Line , Cell Membrane , Chlorocebus aethiops , HEK293 Cells , Humans , Hydrogels , Magnetic Resonance Imaging/methods , Molecular Conformation , Neurons , Prions/chemistry , Prions/physiology , Protein Binding , Receptor, Metabotropic Glutamate 5 , Signal Transduction
2.
Proc Natl Acad Sci U S A ; 120(1): e2209815120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36574660

ABSTRACT

The cellular prion protein (PrPC) converts to alternatively folded pathogenic conformations (PrPSc) in prion infections and binds neurotoxic oligomers formed by amyloid-ß α-synuclein, and tau. ß-Endoproteolysis, which splits PrPC into N- and C-terminal fragments (N2 and C2, respectively), is of interest because a protease-resistant, C2-sized fragment (C2Sc) accumulates in the brain during prion infections, seemingly comprising the majority of PrPSc at disease endpoint in mice. However, candidates for the underlying proteolytic mechanism(s) remain unconfirmed in vivo. Here, a cell-based screen of protease inhibitors unexpectedly linked type II membrane proteins of the S9B serine peptidase subfamily to PrPC ß-cleavage. Overexpression experiments in cells and assays with recombinant proteins confirmed that fibroblast activation protein (FAP) and its paralog, dipeptidyl peptidase-4 (DPP4), cleave directly at multiple sites within PrPC's N-terminal domain. For wild-type mouse and human PrPC substrates expressed in cells, the rank orders of activity were human FAP ~ mouse FAP > mouse DPP4 > human DPP4 and human FAP > mouse FAP > mouse DPP4 >> human DPP4, respectively. C2 levels relative to total PrPC were reduced in several tissues from FAP-null mice, and, while knockout of DPP4 lacked an analogous effect, the combined DPP4/FAP inhibitor linagliptin, but not the FAP-specific inhibitor SP-13786, reduced C2Sc and total PrPSc levels in two murine cell-based models of prion infections. Thus, the net activity of the S9B peptidases FAP and DPP4 and their cognate inhibitors/modulators affect the physiology and pathogenic potential of PrPC.


Subject(s)
PrPC Proteins , Prion Diseases , Prions , Mice , Animals , Humans , Prion Proteins/genetics , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Prions/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Peptide Hydrolases , Fibroblasts/metabolism , Prion Diseases/metabolism , PrPC Proteins/chemistry
3.
J Biol Chem ; 299(2): 102823, 2023 02.
Article in English | MEDLINE | ID: mdl-36565989

ABSTRACT

The prion protein (PrPC) is subjected to several conserved endoproteolytic events producing bioactive fragments that are of increasing interest for their physiological functions and their implication in the pathogenesis of prion diseases and other neurodegenerative diseases. However, systematic and comprehensive investigations on the full spectrum of PrPC proteoforms have been hampered by the lack of methods able to identify all PrPC-derived proteoforms. Building on previous knowledge of PrPC endoproteolytic processing, we thus developed an optimized Western blot assay able to obtain the maximum information about PrPC constitutive processing and the relative abundance of PrPC proteoforms in a complex biological sample. This approach led to the concurrent identification of the whole spectrum of known endoproteolytic-derived PrPC proteoforms in brain homogenates, including C-terminal, N-terminal and, most importantly, shed PrPC-derived fragments. Endoproteolytic processing of PrPC was remarkably similar in the brain of widely used wild type and transgenic rodent models, with α-cleavage-derived C1 representing the most abundant proteoform and ADAM10-mediated shedding being an unexpectedly prominent proteolytic event. Interestingly, the relative amount of shed PrPC was higher in WT mice than in most other models. Our results indicate that constitutive endoproteolytic processing of PrPC is not affected by PrPC overexpression or host factors other than PrPC but can be impacted by PrPC primary structure. Finally, this method represents a crucial step in gaining insight into pathophysiological roles, biomarker suitability, and therapeutic potential of shed PrPC and for a comprehensive appraisal of PrPC proteoforms in therapies, drug screening, or in the progression of neurodegenerative diseases.


Subject(s)
Blotting, Western , Peptide Fragments , PrPC Proteins , Proteolysis , Animals , Mice , Blotting, Western/methods , Prion Diseases/metabolism , Prion Diseases/pathology , Prion Diseases/physiopathology , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPC Proteins/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Brain/metabolism
4.
Cell ; 137(6): 994-6, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524502

ABSTRACT

Inactivation of mahogunin, an E3 ubiquitin ligase, causes a spongiform encephalopathy resembling prion disease. Chakrabarti and Hegde (2009) now report that prion proteins with aberrant topologies inactivate mahogunin, providing a plausible explanation for certain aspects of prion pathology.


Subject(s)
PrPC Proteins/metabolism , Prion Diseases/metabolism , Animals , Humans , Mice , PrPC Proteins/chemistry , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
5.
J Biol Chem ; 295(30): 10420-10433, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32513872

ABSTRACT

The causative factors underlying conformational conversion of cellular prion protein (PrPC) into its infectious counterpart (PrPSc) during prion infection remain undetermined, in part because of a lack of monoclonal antibodies (mAbs) that can distinguish these conformational isoforms. Here we show that the anti-PrP mAb PRC7 recognizes an epitope that is shielded from detection when glycans are attached to Asn-196. We observed that whereas PrPC is predisposed to full glycosylation and is therefore refractory to PRC7 detection, prion infection leads to diminished PrPSc glycosylation at Asn-196, resulting in an unshielded PRC7 epitope that is amenable to mAb recognition upon renaturation. Detection of PRC7-reactive PrPSc in experimental and natural infections with various mouse-adapted scrapie strains and with prions causing deer and elk chronic wasting disease and transmissible mink encephalopathy uncovered that incomplete PrPSc glycosylation is a consistent feature of prion pathogenesis. We also show that interrogating the conformational properties of the PRC7 epitope affords a direct means of distinguishing different prion strains. Because the specificity of our approach for prion detection and strain discrimination relies on the extent to which N-linked glycosylation shields or unshields PrP epitopes from antibody recognition, it dispenses with the requirement for additional standard manipulations to distinguish PrPSc from PrPC, including evaluation of protease resistance. Our findings not only highlight an innovative and facile strategy for prion detection and strain differentiation, but are also consistent with a mechanism of prion replication in which structural instability of incompletely glycosylated PrP contributes to the conformational conversion of PrPC to PrPSc.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Epitopes/chemistry , PrPC Proteins/chemistry , Animals , Cell Line , Epitopes/metabolism , Glycosylation , PrPC Proteins/metabolism , Rabbits
6.
J Biol Chem ; 295(22): 7789-7798, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32321762

ABSTRACT

Prion diseases are transmissible, lethal neurodegenerative disorders caused by accumulation of the aggregated scrapie form of the prion protein (PrPSc) after conversion of the cellular prion protein (PrPC). The glycosylphosphatidylinositol (GPI) anchor of PrPC is involved in prion disease pathogenesis, and especially sialic acid in a GPI side chain reportedly affects PrPC conversion. Thus, it is important to define the location and structure of the GPI anchor in human PrPC Moreover, the sialic acid linkage type in the GPI side chain has not been determined for any GPI-anchored protein. Here we report GPI glycan structures of human PrPC isolated from human brains and from brains of a knock-in mouse model in which the mouse prion protein (Prnp) gene was replaced with the human PRNP gene. LC-electrospray ionization-MS analysis of human PrPC from both biological sources indicated that Gly229 is the ω site in PrPC to which GPI is attached. Gly229 in human PrPC does not correspond to Ser231, the previously reported ω site of Syrian hamster PrPC We found that ∼41% and 28% of GPI anchors in human PrPCs from human and knock-in mouse brains, respectively, have N-acetylneuraminic acid in the side chain. Using a sialic acid linkage-specific alkylamidation method to discriminate α2,3 linkage from α2,6 linkage, we found that N-acetylneuraminic acid in PrPC's GPI side chain is linked to galactose through an α2,3 linkage. In summary, we report the GPI glycan structure of human PrPC, including the ω-site amino acid for GPI attachment and the sialic acid linkage type.


Subject(s)
Glycosylphosphatidylinositols , N-Acetylneuraminic Acid , PrPC Proteins , Prion Proteins , Animals , Carbohydrate Conformation , Glycosylphosphatidylinositols/chemistry , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Humans , Male , Mesocricetus , Mice , Mice, Knockout , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/genetics , N-Acetylneuraminic Acid/metabolism , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPC Proteins/metabolism , Prion Diseases/genetics , Prion Diseases/metabolism , Prion Diseases/pathology , Prion Proteins/chemistry , Prion Proteins/genetics , Prion Proteins/metabolism
7.
Lab Invest ; 101(10): 1327-1330, 2021 10.
Article in English | MEDLINE | ID: mdl-34253850

ABSTRACT

Five sporadic Creutzfeldt-Jakob disease (CJD) strains have been identified to date, based on differences in clinicopathological features of the patients, the biochemical properties of abnormal prion proteins, and transmission properties. Recent advances in our knowledge about iatrogenic transmission of sporadic CJD have raised the possibility that the infectivity of sporadic CJD strains through peripheral routes is different from that of intracranial infection. To test this possibility, here we assessed systematically the infectivity of sporadic CJD strains through the peripheral route for the first time using a mouse model expressing human prion protein. Although the infectivity of the V2 and M1 sporadic CJD strains is almost the same in intracerebral transmission studies, the V2 strain infected more efficiently than the M1 strain through the peripheral route. The other sporadic CJD strains examined lacked infectivity. Of note, both the V2 and M1 strains showed preference for mice with the valine homozygosity at the PRNP polymorphic codon. These results indicate that the V2 strain is the most infectious sporadic CJD strain for infection through peripheral routes. In addition, these findings raise the possibility that individuals with the valine homozygosity at the PRNP polymorphic codon might have higher risks of infection through peripheral routes compared with the methionine homozygotes. Thus, preventive measures against the transmission of the V2 sporadic CJD strain will be important for the eradication of iatrogenic CJD transmission through peripheral routes.


Subject(s)
Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/transmission , Animals , Brain Chemistry , Creutzfeldt-Jakob Syndrome/classification , Humans , Mice , Mice, Transgenic , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPC Proteins/metabolism
8.
Nucleic Acids Res ; 47(18): 9934-9949, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31504787

ABSTRACT

The pathogenicity of Mycobacterium tuberculosis depends upon its ability to catabolize host cholesterol. Upregulation of the methylcitrate cycle (MCC) is required to assimilate and detoxify propionyl-CoA, a cholesterol degradation product. The transcription of key genes prpC and prpD in MCC is activated by MtPrpR, a member of a family of prokaryotic transcription factors whose structures and modes of action have not been clearly defined. We show that MtPrpR has a novel overall structure and directly binds to CoA or short-chain acyl-CoA derivatives to form a homotetramer that covers the binding cavity and locks CoA tightly inside the protein. The regulation of this process involves a [4Fe4S] cluster located close to the CoA-binding cavity on a neighboring chain. Mutations in the [4Fe4S] cluster binding residues rendered MtPrpR incapable of regulating MCC gene transcription. The structure of MtPrpR without the [4Fe4S] cluster-binding region shows a conformational change that prohibits CoA binding. The stability of this cluster means it is unlikely a redox sensor but may function by sensing ambient iron levels. These results provide mechanistic insights into this family of critical transcription factors who share similar structures and regulate gene transcription using a combination of acyl-CoAs and [4Fe4S] cluster.


Subject(s)
Acyl Coenzyme A/chemistry , Bacterial Proteins/chemistry , Mycobacterium tuberculosis/genetics , Transcription Factors/chemistry , Acyl Coenzyme A/genetics , Bacterial Proteins/genetics , Cholesterol/genetics , Gene Expression Regulation, Bacterial/genetics , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/pathogenicity , PrPC Proteins/chemistry , PrPC Proteins/genetics , Transcription Factors/genetics , Tuberculosis/genetics , Tuberculosis/microbiology
9.
Int J Mol Sci ; 22(6)2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33806892

ABSTRACT

Prions are infectious agents causing prion diseases, which include Creutzfeldt-Jakob disease (CJD) in humans. Several cases have been reported to be transmitted through medical instruments that were used for preclinical CJD patients, raising public health concerns on iatrogenic transmissions of the disease. Since preclinical CJD patients are currently difficult to identify, medical instruments need to be adequately sterilized so as not to transmit the disease. In this study, we investigated the sterilizing activity of two oxidizing agents, ozone gas and vaporized hydrogen peroxide, against prions fixed on stainless steel wires using a mouse bioassay. Mice intracerebrally implanted with prion-contaminated stainless steel wires treated with ozone gas or vaporized hydrogen peroxide developed prion disease later than those implanted with control prion-contaminated stainless steel wires, indicating that ozone gas and vaporized hydrogen peroxide could reduce prion infectivity on wires. Incubation times were further elongated in mice implanted with prion-contaminated stainless steel wires treated with ozone gas-mixed vaporized hydrogen peroxide, indicating that ozone gas mixed with vaporized hydrogen peroxide reduces prions on these wires more potently than ozone gas or vaporized hydrogen peroxide. These results suggest that ozone gas mixed with vaporized hydrogen peroxide might be more useful for prion sterilization than ozone gas or vaporized hydrogen peroxide alone.


Subject(s)
Hydrogen Peroxide/chemistry , Ozone/chemistry , Prions , Stainless Steel , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Hydrogen Peroxide/pharmacology , Mice , Ozone/pharmacology , PrPC Proteins/antagonists & inhibitors , PrPC Proteins/chemistry , Prion Diseases/etiology , Prion Diseases/prevention & control , Stainless Steel/chemistry
10.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830321

ABSTRACT

Conformational conversion of the cellular isoform of prion protein, PrPC, into the abnormally folded, amyloidogenic isoform, PrPSc, is an underlying pathogenic mechanism in prion diseases. The diseases manifest as sporadic, hereditary, and acquired disorders. Etiological mechanisms driving the conversion of PrPC into PrPSc are unknown in sporadic prion diseases, while prion infection and specific mutations in the PrP gene are known to cause the conversion of PrPC into PrPSc in acquired and hereditary prion diseases, respectively. We recently reported that a neurotropic strain of influenza A virus (IAV) induced the conversion of PrPC into PrPSc as well as formation of infectious prions in mouse neuroblastoma cells after infection, suggesting the causative role of the neuronal infection of IAV in sporadic prion diseases. Here, we discuss the conversion mechanism of PrPC into PrPSc in different types of prion diseases, by presenting our findings of the IAV infection-induced conversion of PrPC into PrPSc and by reviewing the so far reported transgenic animal models of hereditary prion diseases and the reverse genetic studies, which have revealed the structure-function relationship for PrPC to convert into PrPSc after prion infection.


Subject(s)
Creutzfeldt-Jakob Syndrome/genetics , Gerstmann-Straussler-Scheinker Disease/genetics , Influenza, Human/genetics , Insomnia, Fatal Familial/genetics , PrPC Proteins/genetics , PrPSc Proteins/genetics , Prion Proteins/genetics , Animals , Cell Line, Tumor , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Creutzfeldt-Jakob Syndrome/virology , Gerstmann-Straussler-Scheinker Disease/metabolism , Gerstmann-Straussler-Scheinker Disease/pathology , Gerstmann-Straussler-Scheinker Disease/virology , Humans , Influenza A virus/genetics , Influenza A virus/growth & development , Influenza A virus/pathogenicity , Influenza, Human/metabolism , Influenza, Human/pathology , Influenza, Human/virology , Insomnia, Fatal Familial/metabolism , Insomnia, Fatal Familial/pathology , Insomnia, Fatal Familial/virology , Mice , Mice, Transgenic , Mutation , Neurons/metabolism , Neurons/pathology , Neurons/virology , PrPC Proteins/chemistry , PrPC Proteins/metabolism , PrPSc Proteins/chemistry , PrPSc Proteins/metabolism , Prion Proteins/chemistry , Prion Proteins/metabolism , Protein Conformation , Reverse Genetics/methods
11.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525334

ABSTRACT

Prion diseases are progressive and transmissive neurodegenerative diseases. The conformational conversion of normal cellular prion protein (PrPC) into abnormal pathogenic prion protein (PrPSc) is critical for its infection and pathogenesis. PrPC possesses the ability to bind to various neurometals, including copper, zinc, iron, and manganese. Moreover, increasing evidence suggests that PrPC plays essential roles in the maintenance of homeostasis of these neurometals in the synapse. In addition, trace metals are critical determinants of the conformational change and toxicity of PrPC. Here, we review our studies and other new findings that inform the current understanding of the links between trace elements and physiological functions of PrPC and the neurotoxicity of PrPSc.


Subject(s)
Copper/metabolism , Iron/metabolism , Manganese/metabolism , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , Prion Diseases/metabolism , Zinc/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Cations, Divalent , Homeostasis , Humans , Neurons/metabolism , Neurons/pathology , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPSc Proteins/chemistry , PrPSc Proteins/genetics , Prion Diseases/genetics , Prion Diseases/pathology , Protein Binding , Synapses/metabolism , Synapses/pathology , Synaptic Transmission
12.
Proc Natl Acad Sci U S A ; 114(36): 9617-9622, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827331

ABSTRACT

In transmissible spongiform encephalopathies (TSEs), which are lethal neurodegenerative diseases that affect humans and a wide range of other mammalian species, the normal "cellular" prion protein ([Formula: see text]) is transformed into amyloid aggregates representing the "scrapie form" of the protein ([Formula: see text]). Continued research on this system is of keen interest, since new information on the physiological function of [Formula: see text] in healthy organisms is emerging, as well as new data on the mechanism of the transformation of [Formula: see text] to [Formula: see text] In this paper we used two different approaches: a combination of the well-tempered ensemble (WTE) and parallel tempering (PT) schemes and metadynamics (MetaD) to characterize the conformational free-energy surface of [Formula: see text] The focus of the data analysis was on an 11-residue polypeptide segment in mouse [Formula: see text](121-231) that includes the [Formula: see text]2-[Formula: see text]2 loop of residues 167-170, for which a correlation between structure and susceptibility to prion disease has previously been described. This study includes wild-type mouse [Formula: see text] and a variant with the single-residue replacement Y169A. The resulting detailed conformational landscapes complement in an integrative manner the available experimental data on [Formula: see text], providing quantitative insights into the nature of the structural transition-related function of the [Formula: see text]2-[Formula: see text]2 loop.


Subject(s)
PrPC Proteins/chemistry , Amino Acid Substitution , Animals , Humans , Mice , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , PrPC Proteins/genetics , PrPC Proteins/pathogenicity , Prion Diseases/etiology , Prion Diseases/genetics , Prion Diseases/metabolism , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand
13.
Proc Natl Acad Sci U S A ; 114(5): 1141-1146, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096357

ABSTRACT

Adaptation of prions to new species is thought to reflect the capacity of the host-encoded cellular form of the prion protein (PrPC) to selectively propagate optimized prion conformations from larger ensembles generated in the species of origin. Here we describe an alternate replicative process, termed nonadaptive prion amplification (NAPA), in which dominant conformers bypass this requirement during particular interspecies transmissions. To model susceptibility of horses to prions, we produced transgenic (Tg) mice expressing cognate PrPC Although disease transmission to only a subset of infected TgEq indicated a significant barrier to EqPrPC conversion, the resulting horse prions unexpectedly failed to cause disease upon further passage to TgEq. TgD expressing deer PrPC was similarly refractory to deer prions from diseased TgD infected with mink prions. In both cases, the resulting prions transmitted to mice expressing PrPC from the species of prion origin, demonstrating that transmission barrier eradication of the originating prions was ephemeral and adaptation superficial in TgEq and TgD. Horse prions produced in vitro by protein misfolding cyclic amplification of mouse prions using horse PrPC also failed to infect TgEq but retained tropism for wild-type mice. Concordant patterns of neuropathology and prion deposition in susceptible mice infected with NAPA prions and the corresponding prion of origin confirmed preservation of strain properties. The comparable responses of both prion types to guanidine hydrochloride denaturation indicated this occurs because NAPA precludes selection of novel prion conformations. Our findings provide insights into mechanisms regulating interspecies prion transmission and a framework to reconcile puzzling epidemiological features of certain prion disorders.


Subject(s)
Host Specificity/physiology , PrPC Proteins/physiology , Prion Diseases/transmission , Prion Diseases/veterinary , Prions/physiology , Animals , Deer , Guanidine/pharmacology , Horses , Mice , Mice, Inbred C57BL , PrPC Proteins/chemistry , PrPC Proteins/genetics , Prions/chemistry , Protein Conformation , Protein Denaturation , Rabbits , Sheep , Species Specificity , Structure-Activity Relationship
14.
Int J Mol Sci ; 21(19)2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33019683

ABSTRACT

Alzheimer's disease (AD) is the most prevalent form of dementia and soluble amyloid ß (Aß) oligomers are thought to play a critical role in AD pathogenesis. Cellular prion protein (PrPC) is a high-affinity receptor for Aß oligomers and mediates some of their toxic effects. The N-terminal region of PrPC can interact with Aß, particularly the region encompassing residues 95-110. In this study, we identified a soluble and unstructured prion-derived peptide (PrP107-120) that is external to this region of the sequence and was found to successfully reduce the mitochondrial impairment, intracellular ROS generation and cytosolic Ca2+ uptake induced by oligomeric Aß42 ADDLs in neuroblastoma SH-SY5Y cells. PrP107-120 was also found to rescue SH-SY5Y cells from Aß42 ADDL internalization. The peptide did not change the structure and aggregation pathway of Aß42 ADDLs, did not show co-localization with Aß42 ADDLs in the cells and showed a partial colocalization with the endogenous cellular PrPC. As a sequence region that is not involved in Aß binding but in PrP self-recognition, the peptide was suggested to protect against the toxicity of Aß42 oligomers by interfering with cellular PrPC and/or activating a signaling that protected the cells. These results strongly suggest that PrP107-120 has therapeutic potential for AD.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Calcium/metabolism , Neurons/drug effects , Peptide Fragments/antagonists & inhibitors , Peptides/pharmacology , PrPC Proteins/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Humans , Ion Transport , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Models, Biological , Neurons/metabolism , Neurons/pathology , Peptide Fragments/toxicity , Peptides/chemistry , PrPC Proteins/metabolism , Protein Binding , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Solubility
15.
Int J Mol Sci ; 21(19)2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33019549

ABSTRACT

Conformational conversion of the cellular prion protein, PrPC, into the abnormally folded isoform, PrPSc, is a key pathogenic event in prion diseases. However, the exact conversion mechanism remains largely unknown. Transgenic mice expressing PrP with a deletion of the central residues 91-106 were generated in the absence of endogenous PrPC, designated Tg(PrP∆91-106)/Prnp0/0 mice and intracerebrally inoculated with various prions. Tg(PrP∆91-106)/Prnp0/0 mice were resistant to RML, 22L and FK-1 prions, neither producing PrPSc∆91-106 or prions in the brain nor developing disease after inoculation. However, they remained marginally susceptible to bovine spongiform encephalopathy (BSE) prions, developing disease after elongated incubation times and accumulating PrPSc∆91-106 and prions in the brain after inoculation with BSE prions. Recombinant PrP∆91-104 converted into PrPSc∆91-104 after incubation with BSE-PrPSc-prions but not with RML- and 22L-PrPSc-prions, in a protein misfolding cyclic amplification assay. However, digitonin and heparin stimulated the conversion of PrP∆91-104 into PrPSc∆91-104 even after incubation with RML- and 22L-PrPSc-prions. These results suggest that residues 91-106 or 91-104 of PrPC are crucially involved in prion pathogenesis in a strain-dependent manner and may play a similar role to digitonin and heparin in the conversion of PrPC into PrPSc.


Subject(s)
Encephalopathy, Bovine Spongiform/genetics , PrPC Proteins/genetics , PrPSc Proteins/genetics , Proteostasis Deficiencies/genetics , Scrapie/genetics , Sequence Deletion , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Base Sequence , Brain/metabolism , Brain/pathology , Cattle , Cloning, Molecular , Disease Susceptibility , Encephalopathy, Bovine Spongiform/metabolism , Encephalopathy, Bovine Spongiform/pathology , Gene Expression , Injections, Intraventricular , Mice , Mice, Transgenic , PrPC Proteins/chemistry , PrPC Proteins/metabolism , PrPSc Proteins/administration & dosage , PrPSc Proteins/chemistry , PrPSc Proteins/metabolism , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Scrapie/metabolism , Scrapie/pathology , Species Specificity
16.
Lab Invest ; 99(11): 1741-1748, 2019 11.
Article in English | MEDLINE | ID: mdl-31249376

ABSTRACT

Prion diseases are transmissible neurological disorders associated with the presence of abnormal, disease-related prion protein (PrPD). The detection of PrPD in the brain is the only definitive diagnostic evidence of prion disease and its identification in body fluids and peripheral tissues are valuable for pre-mortem diagnosis. Protein misfolding cyclic amplification (PMCA) is a technique able to detect minute amount of PrPD and is based on the conversion of normal or cellular PrP (PrPC) to newly formed PrPD, sustained by a self-templating mechanism. Several animal prions have been efficiently amplified by PMCA, but limited results have been obtained with human prions with the exception of variant-Creutzfeldt-Jakob-disease (vCJD). Since the total or partial absence of glycans on PrPC has been shown to affect PMCA efficiency in animal prion studies, we attempted to enhance the amplification of four major sporadic-CJD (sCJD) subtypes (MM1, MM2, VV1, and VV2) and vCJD by single round PMCA using partially or totally unglycosylated PrPC as substrates. The amplification efficiency of all tested sCJD subtypes underwent a strong increase, inversely correlated to the degree of PrPC glycosylation and directly related to the matching of the PrP polymorphic 129 M/V genotype between seed and substrate. This effect was particularly significant in sCJDMM2 and sCJDVV2 allowing the detection of PK-resistant PrPD (resPrPD) in sCJDMM2 and sCJDVV2 brains at dilutions of 6 × 107 and 3 × 106. vCJD, at variance with the tested sCJD subtypes, showed the best amplification with partially deglycosylated PrPC substrate reaching a resPrPD detectability at up to 3 × 1016 brain dilution. The differential effect of substrate PrPC glycosylations suggests subtype-dependent PrPC-PrPD interactions, strongly affected by the PrPC glycans. The enhanced PMCA prion amplification efficiency achieved with unglycosylated PrPC substrates may allow for the developing of a sensitive, non-invasive, diagnostic test for the different CJD subtypes based on body fluids or easily-accessible-peripheral tissues.


Subject(s)
Prion Diseases/diagnosis , Prion Diseases/metabolism , Prion Proteins/metabolism , Prions/metabolism , Animals , Brain/metabolism , Creutzfeldt-Jakob Syndrome/diagnosis , Creutzfeldt-Jakob Syndrome/metabolism , Encephalopathy, Bovine Spongiform/diagnosis , Encephalopathy, Bovine Spongiform/metabolism , Glycosylation , Humans , Mice , Mice, Transgenic , Phenotype , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPC Proteins/metabolism , Prion Diseases/genetics , Prion Proteins/chemistry , Prion Proteins/genetics , Prions/chemistry , Protein Folding
17.
J Virol ; 92(1)2018 01 01.
Article in English | MEDLINE | ID: mdl-29046443

ABSTRACT

Conformational conversion of the cellular isoform of prion protein, PrPC, into the abnormally folded, amyloidogenic isoform, PrPSc, is a key pathogenic event in prion diseases, including Creutzfeldt-Jakob disease in humans and scrapie and bovine spongiform encephalopathy (BSE) in animals. We previously reported that the octapeptide repeat (OR) region could be dispensable for converting PrPC into PrPSc after infection with RML prions. We demonstrated that mice transgenically expressing mouse PrP with deletion of the OR region on the PrP knockout background, designated Tg(PrPΔOR)/Prnp0/0 mice, did not show reduced susceptibility to RML scrapie prions, with abundant accumulation of PrPScΔOR in their brains. We show here that Tg(PrPΔOR)/Prnp0/0 mice were highly resistant to BSE prions, developing the disease with markedly elongated incubation times after infection with BSE prions. The conversion of PrPΔOR into PrPScΔOR was markedly delayed in their brains. These results suggest that the OR region may have a crucial role in the conversion of PrPC into PrPSc after infection with BSE prions. However, Tg(PrPΔOR)/Prnp0/0 mice remained susceptible to RML and 22L scrapie prions, developing the disease without elongated incubation times after infection with RML and 22L prions. PrPScΔOR accumulated only slightly less in the brains of RML- or 22L-infected Tg(PrPΔOR)/Prnp0/0 mice than PrPSc in control wild-type mice. Taken together, these results indicate that the OR region of PrPC could play a differential role in the pathogenesis of BSE prions and RML or 22L scrapie prions.IMPORTANCE Structure-function relationship studies of PrPC conformational conversion into PrPSc are worthwhile to understand the mechanism of the conversion of PrPC into PrPSc We show here that, by inoculating Tg(PrPΔOR)/Prnp0/0 mice with the three different strains of RML, 22L, and BSE prions, the OR region could play a differential role in the conversion of PrPC into PrPSc after infection with RML or 22L scrapie prions and BSE prions. PrPΔOR was efficiently converted into PrPScΔOR after infection with RML and 22L prions. However, the conversion of PrPΔOR into PrPScΔOR was markedly delayed after infection with BSE prions. Further investigation into the role of the OR region in the conversion of PrPC into PrPSc after infection with BSE prions might be helpful for understanding the pathogenesis of BSE prions.


Subject(s)
Disease Susceptibility , Encephalopathy, Bovine Spongiform/physiopathology , PrPC Proteins/chemistry , PrPC Proteins/physiology , Prion Diseases/physiopathology , Prions/pathogenicity , Animals , Brain/pathology , Cattle , Encephalopathy, Bovine Spongiform/prevention & control , Humans , Mice , Mice, Transgenic , Oligopeptides/chemistry , Oligopeptides/genetics , PrPC Proteins/genetics , Prion Diseases/prevention & control , Prions/chemistry , Prions/genetics , Sequence Deletion
18.
PLoS Pathog ; 13(11): e1006716, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29131852

ABSTRACT

One of the characteristics of prions is their ability to infect some species but not others and prion resistant species have been of special interest because of their potential in deciphering the determinants for susceptibility. Previously, we developed different in vitro and in vivo models to assess the susceptibility of species that were erroneously considered resistant to prion infection, such as members of the Leporidae and Equidae families. Here we undertake in vitro and in vivo approaches to understand the unresolved low prion susceptibility of canids. Studies based on the amino acid sequence of the canine prion protein (PrP), together with a structural analysis in silico, identified unique key amino acids whose characteristics could orchestrate its high resistance to prion disease. Cell- and brain-based PMCA studies were performed highlighting the relevance of the D163 amino acid in proneness to protein misfolding. This was also investigated by the generation of a novel transgenic mouse model carrying this substitution and these mice showed complete resistance to disease despite intracerebral challenge with three different mouse prion strains (RML, 22L and 301C) known to cause disease in wild-type mice. These findings suggest that dog D163 amino acid is primarily, if not totally, responsible for the prion resistance of canids.


Subject(s)
Canidae/immunology , PrPC Proteins/chemistry , Prion Diseases/veterinary , Amino Acid Sequence , Animals , Antelopes , Brain/pathology , Cats , Cattle , Chiroptera , Deer , Disease Resistance , Dogs , Encephalopathy, Bovine Spongiform/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , PrPC Proteins/ultrastructure , Prion Diseases/immunology , Protein Folding , Protein Structure, Quaternary , Rabbits , Sequence Alignment , Sheep , Static Electricity , Xenarthra
19.
J Pept Sci ; 25(10): e3216, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31713950

ABSTRACT

Deciphering the pathophysiologic events in prion diseases is challenging, and the role of posttranslational modifications (PTMs) such as glypidation and glycosylation remains elusive due to the lack of homogeneous protein preparations. So far, experimental studies have been limited in directly analyzing the earliest events of the conformational change of cellular prion protein (PrPC ) into scrapie prion protein (PrPSc ) that further propagates PrPC misfolding and aggregation at the cellular membrane, the initial site of prion infection, and PrP misfolding, by a lack of suitably modified PrP variants. PTMs of PrP, especially attachment of the glycosylphosphatidylinositol (GPI) anchor, have been shown to be crucially involved in the PrPSc formation. To this end, semisynthesis offers a unique possibility to understand PrP behavior invitro and invivo as it provides access to defined site-selectively modified PrP variants. This approach relies on the production and chemoselective linkage of peptide segments, amenable to chemical modifications, with recombinantly produced protein segments. In this article, advances in understanding PrP conversion using semisynthesis as a tool to obtain homogeneous posttranslationally modified PrP will be discussed.


Subject(s)
PrPC Proteins/chemical synthesis , PrPSc Proteins/chemical synthesis , Protein Folding , Protein Processing, Post-Translational , Animals , Humans , PrPC Proteins/chemistry , PrPC Proteins/metabolism , PrPSc Proteins/chemistry , PrPSc Proteins/metabolism , Prion Diseases/metabolism , Prion Diseases/pathology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology
20.
Nature ; 501(7465): 102-6, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23903654

ABSTRACT

Prion infections cause lethal neurodegeneration. This process requires the cellular prion protein (PrP(C); ref. 1), which contains a globular domain hinged to a long amino-proximal flexible tail. Here we describe rapid neurotoxicity in mice and cerebellar organotypic cultured slices exposed to ligands targeting the α1 and α3 helices of the PrP(C) globular domain. Ligands included seven distinct monoclonal antibodies, monovalent Fab1 fragments and recombinant single-chain variable fragment miniantibodies. Similar to prion infections, the toxicity of globular domain ligands required neuronal PrP(C), was exacerbated by PrP(C) overexpression, was associated with calpain activation and was antagonized by calpain inhibitors. Neurodegeneration was accompanied by a burst of reactive oxygen species, and was suppressed by antioxidants. Furthermore, genetic ablation of the superoxide-producing enzyme NOX2 (also known as CYBB) protected mice from globular domain ligand toxicity. We also found that neurotoxicity was prevented by deletions of the octapeptide repeats within the flexible tail. These deletions did not appreciably compromise globular domain antibody binding, suggesting that the flexible tail is required to transmit toxic signals that originate from the globular domain and trigger oxidative stress and calpain activation. Supporting this view, various octapeptide ligands were not only innocuous to both cerebellar organotypic cultured slices and mice, but also prevented the toxicity of globular domain ligands while not interfering with their binding. We conclude that PrP(C) consists of two functionally distinct modules, with the globular domain and the flexible tail exerting regulatory and executive functions, respectively. Octapeptide ligands also prolonged the life of mice expressing the toxic PrP(C) mutant, PrP(Δ94-134), indicating that the flexible tail mediates toxicity in two distinct PrP(C)-related conditions. Flexible tail-mediated toxicity may conceivably play a role in further prion pathologies, such as familial Creutzfeldt-Jakob disease in humans bearing supernumerary octapeptides.


Subject(s)
Antibodies/immunology , Antibodies/toxicity , Pliability , Prions/chemistry , Prions/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/toxicity , Binding Sites, Antibody , Calpain/metabolism , Cerebellum , Creutzfeldt-Jakob Syndrome/metabolism , Cross-Linking Reagents , Epitope Mapping , Female , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/toxicity , In Vitro Techniques , Ligands , Male , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Neurodegenerative Diseases/metabolism , Oxidative Stress , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPC Proteins/immunology , Prions/genetics , Reactive Oxygen Species/metabolism , Sequence Deletion/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL