Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 972
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 19(4): e1011309, 2023 04.
Article in English | MEDLINE | ID: mdl-37104170

ABSTRACT

Prion diseases, also known as transmissible spongiform encephalopathies, are rare, progressive, and fatal neurodegenerative disorders, which are caused by the accumulation of the misfolded cellular prion protein (PrPC). The resulting cytotoxic prion species, referred to as the scrapie prion isoform (PrPSc), assemble in aggregates and interfere with neuronal pathways, ultimately rendering neurons dysfunctional. As the prion protein physiologically interacts with redox-active metals, an altered redox balance within the cell can impact these interactions, which may lead to and facilitate further misfolding and aggregation. The initiation of misfolding and the aggregation processes will, in turn, induce microglial activation and neuroinflammation, which leads to an imbalance in cellular redox homeostasis and enhanced redox stress. Potential approaches for therapeutics target redox signalling, and this review illustrates the pathways involved in the above processes.


Subject(s)
Prion Diseases , Prions , Scrapie , Animals , Sheep , Prion Proteins/metabolism , Prions/metabolism , Scrapie/pathology , Oxidation-Reduction
2.
J Infect Dis ; 230(1): 161-171, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052723

ABSTRACT

BACKGROUND: Atypical/Nor98 scrapie (AS) is an idiopathic infectious prion disease affecting sheep and goats. Recent findings suggest that zoonotic prions from classical bovine spongiform encephalopathy (C-BSE) may copropagate with atypical/Nor98 prions in AS sheep brains. Investigating the risk AS poses to humans is crucial. METHODS: To assess the risk of sheep/goat-to-human transmission of AS, we serially inoculated brain tissue from field and laboratory isolates into transgenic mice overexpressing human prion protein (Met129 allele). We studied clinical outcomes as well as presence of prions in brains and spleens. RESULTS: No transmission occurred on the primary passage, with no clinical disease or pathological prion protein in brains and spleens. On subsequent passages, 1 isolate gradually adapted, manifesting as prions with a phenotype resembling those causing MM1-type sporadic Creutzfeldt-Jakob disease in humans. However, further characterization using in vivo and in vitro techniques confirmed both prion agents as different strains, revealing a case of phenotypic convergence. Importantly, no C-BSE prions emerged in these mice, especially in the spleen, which is more permissive than the brain for C-BSE cross-species transmission. CONCLUSIONS: The results obtained suggest a low zoonotic potential for AS. Rare adaptation may allow the emergence of prions phenotypically resembling those spontaneously forming in humans.


Subject(s)
Brain , Creutzfeldt-Jakob Syndrome , Goats , Mice, Transgenic , Prions , Scrapie , Zoonoses , Animals , Creutzfeldt-Jakob Syndrome/transmission , Creutzfeldt-Jakob Syndrome/pathology , Creutzfeldt-Jakob Syndrome/metabolism , Humans , Scrapie/transmission , Scrapie/pathology , Mice , Zoonoses/transmission , Brain/pathology , Brain/metabolism , Sheep , Cattle , Prions/metabolism , Phenotype , Spleen/pathology , Encephalopathy, Bovine Spongiform/transmission , Encephalopathy, Bovine Spongiform/pathology , Encephalopathy, Bovine Spongiform/metabolism , Goat Diseases/transmission , Goat Diseases/pathology , Disease Models, Animal
3.
J Infect Dis ; 227(12): 1386-1395, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36344485

ABSTRACT

BACKGROUND: Classic scrapie is a prion disease of sheep and goats that is associated with accumulation of abnormal prion protein (PrPSc) in the central nervous and lymphoid tissues. Chronic wasting disease (CWD) is the prion disease of cervids. This study was conducted to determine the susceptibility of white-tailed deer (WTD) to the classic scrapie agent. METHODS: We inoculated WTD (n = 5) by means of a concurrent oral/intranasal exposure with the classic scrapie agent from sheep or oronasally with the classic scrapie agent from goats (n = 6). RESULTS: All deer exposed to the agent of classic scrapie from sheep accumulated PrPSc. PrPSc was detected in lymphoid tissues at preclinical time points, and necropsies in deer 28 months after inoculation showed clinical signs, spongiform lesions, and widespread PrPSc in neural and lymphoid tissues. Western blots on samples from the brainstem, cerebellum, and lymph nodes of scrapie-infected WTD have a molecular profile similar to CWD and distinct from samples from the cerebral cortex, retina, or the original classic scrapie inoculum. There was no evidence of PrPSc in any of the WTD inoculated with classic scrapie prions from goats. CONCLUSIONS: WTD are susceptible to the agent of classic scrapie from sheep, and differentiation from CWD may be difficult.


Subject(s)
Deer , Prion Diseases , Scrapie , Wasting Disease, Chronic , Animals , Sheep , Scrapie/metabolism , Scrapie/pathology , Deer/metabolism , Prion Diseases/metabolism , Prion Diseases/veterinary , PrPSc Proteins/metabolism , Wasting Disease, Chronic/metabolism , Goats/metabolism
4.
Cell Tissue Res ; 392(1): 113-133, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35796874

ABSTRACT

Prion diseases are a group of inevitably fatal neurodegenerative disorders affecting numerous mammalian species, including humans. The existence of heritable phenotypes of disease in the natural host suggested that prions exist as distinct strains. Transmission of sheep scrapie to rodent models accelerated prion research, resulting in the isolation and characterization of numerous strains with distinct characteristics. These strains are grouped into categories based on the incubation period of disease in different strains of mice and also by how stable the strain properties were upon serial passage. These classical studies defined the host and agent parameters that affected strain properties, and, prior to the advent of the prion hypothesis, strain properties were hypothesized to be the result of mutations in a nucleic acid genome of a conventional pathogen. The development of the prion hypothesis challenged the paradigm of infectious agents, and, initially, the existence of strains was difficult to reconcile with a protein-only agent. In the decades since, much evidence has revealed how a protein-only infectious agent can perform complex biological functions. The prevailing hypothesis is that strain-specific conformations of PrPSc encode prion strain diversity. This hypothesis can provide a mechanism to explain the observed strain-specific differences in incubation period of disease, biochemical properties of PrPSc, tissue tropism, and subcellular patterns of pathology. This hypothesis also explains how prion strains mutate, evolve, and adapt to new species. These concepts are applicable to prion-like diseases such as Parkinson's and Alzheimer's disease, where evidence of strain diversity is beginning to emerge.


Subject(s)
Prion Diseases , Prions , Scrapie , Humans , Animals , Sheep , Scrapie/pathology , Phenotype , Mutation , Prion Diseases/genetics , Mammals
5.
Mol Biol Rep ; 50(2): 1743-1752, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36446981

ABSTRACT

Transmissible spongiform encephalopathies (TSEs) or prion diseases consist of a broad range of fatal neurological disorders affecting humans and animals. Contrary to Watson and Crick's 'central dogma', prion diseases are caused by a protein, devoid of DNA involvement. Herein, we briefly review various cellular and biological aspects of prions and prion pathogenesis focusing mainly on historical milestones, biosynthesis, degradation, structure-function of cellular and scrapie forms of prions .


Subject(s)
Prion Diseases , Prions , Scrapie , Animals , Sheep , Humans , Scrapie/genetics , Scrapie/metabolism , Scrapie/pathology , Prions/genetics , Prion Diseases/genetics , Prion Diseases/metabolism , Prion Diseases/pathology
6.
PLoS Pathog ; 16(5): e1008581, 2020 05.
Article in English | MEDLINE | ID: mdl-32421750

ABSTRACT

Prions are unorthodox infectious agents that replicate by templating misfolded conformations of a host-encoded glycoprotein, collectively termed PrPSc. Prion diseases are invariably fatal and currently incurable, but oral drugs that can prolong incubation times in prion-infected mice have been developed. Here, we tested the efficacy of combination therapy with two such drugs, IND24 and Anle138b, in scrapie-infected mice. The results indicate that combination therapy was no more effective than either IND24 or Anle138b monotherapy in prolonging scrapie incubation times. Moreover, combination therapy induced the formation of a new prion strain that is specifically resistant to the combination regimen but susceptible to Anle138b. To our knowledge, this is the first report of a pathogen with specific resistance to combination therapy despite being susceptible to monotherapy. Our findings also suggest that combination therapy may be a less effective strategy for treating prions than conventional pathogens.


Subject(s)
Benzodioxoles/pharmacology , PrPSc Proteins/metabolism , Pyrazoles/pharmacology , Scrapie/drug therapy , Animals , Drug Therapy, Combination , Mice , PrPSc Proteins/pathogenicity , Scrapie/metabolism , Scrapie/pathology
7.
Int J Mol Sci ; 23(12)2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35742934

ABSTRACT

Prion diseases are a group of devastating neurodegenerative disorders, which include Creutzfeldt-Jakob disease (CJD) in humans, and scrapie and bovine spongiform encephalopathy (BSE) in animals [...].


Subject(s)
Creutzfeldt-Jakob Syndrome , Encephalopathy, Bovine Spongiform , Prion Diseases , Prions , Scrapie , Animals , Brain/metabolism , Cattle , Creutzfeldt-Jakob Syndrome/etiology , Creutzfeldt-Jakob Syndrome/pathology , Encephalopathy, Bovine Spongiform/pathology , Prion Diseases/etiology , Prion Diseases/pathology , Prions/metabolism , Scrapie/pathology , Sheep
8.
J Gen Virol ; 102(12)2021 12.
Article in English | MEDLINE | ID: mdl-34904943

ABSTRACT

Prion diseases are fatal and infectious neurodegenerative diseases in humans and other mammals caused by templated misfolding of the endogenous prion protein (PrP). Although there is currently no vaccine or therapy against prion disease, several classes of small-molecule compounds have been shown to increase disease-free incubation time in prion-infected mice. An apparent obstacle to effective anti-prion therapy is the emergence of drug-resistant strains during static therapy with either single compounds or multi-drug combination regimens. Here, we treated scrapie-infected mice with dynamic regimens that alternate between different classes of anti-prion drugs. The results show that alternating regimens containing various combinations of Anle138b, IND24 and IND116135 reduce the incidence of combination drug resistance, but do not significantly increase long-term disease-free survival compared to monotherapy. Furthermore, the alternating regimens induced regional vacuolation profiles resembling those generated by a single component of the alternating regimen, suggesting the emergence of strain dominance.


Subject(s)
Drug Resistance/drug effects , Prions/antagonists & inhibitors , Scrapie/drug therapy , Animals , Brain/pathology , Disease Models, Animal , Disease-Free Survival , Drug Therapy, Combination , Infectious Disease Incubation Period , Mice , Prions/drug effects , Scrapie/mortality , Scrapie/pathology
9.
Int J Mol Sci ; 22(9)2021 May 04.
Article in English | MEDLINE | ID: mdl-34064393

ABSTRACT

Although it is not yet universally accepted that all neurodegenerative diseases (NDs) are prion disorders, there is little disagreement that Alzheimer's disease (AD), Parkinson's disease, frontotemporal dementia (FTD), and other NDs are a consequence of protein misfolding, aggregation, and spread. This widely accepted perspective arose from the prion hypothesis, which resulted from investigations on scrapie, a common transmissible disease of sheep and goats. The prion hypothesis argued that the causative infectious agent of scrapie was a novel proteinaceous pathogen devoid of functional nucleic acids and distinct from viruses, viroids, and bacteria. At the time, it seemed impossible that an infectious agent like the one causing scrapie could replicate and exist as diverse microbiological strains without nucleic acids. However, aggregates of a misfolded host-encoded protein, designated the prion protein (PrP), were shown to be the cause of scrapie as well as Creutzfeldt-Jakob disease (CJD) and Gerstmann-Sträussler-Scheinker syndrome (GSS), which are similar NDs in humans. This review discusses historical research on diseases caused by PrP misfolding, emphasizing principles of pathogenesis that were later found to be core features of other NDs. For example, the discovery that familial prion diseases can be caused by mutations in PrP was important for understanding prion replication and disease susceptibility not only for rare PrP diseases but also for far more common NDs involving other proteins. We compare diseases caused by misfolding and aggregation of APP-derived Aß peptides, tau, and α-synuclein with PrP prion disorders and argue for the classification of NDs caused by misfolding of these proteins as prion diseases. Deciphering the molecular pathogenesis of NDs as prion-mediated has provided new approaches for finding therapies for these intractable, invariably fatal disorders and has revolutionized the field.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Prion Proteins/genetics , Scrapie/genetics , alpha-Synuclein/genetics , tau Proteins/genetics , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Creutzfeldt-Jakob Syndrome/etiology , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Frontotemporal Dementia/etiology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Gene Expression , Gerstmann-Straussler-Scheinker Disease/etiology , Gerstmann-Straussler-Scheinker Disease/genetics , Gerstmann-Straussler-Scheinker Disease/metabolism , Gerstmann-Straussler-Scheinker Disease/pathology , Humans , Mice , Mutation , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Prion Proteins/chemistry , Prion Proteins/metabolism , Prions , Protein Folding , Scrapie/etiology , Scrapie/metabolism , Scrapie/pathology , Sheep , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , tau Proteins/chemistry , tau Proteins/metabolism
10.
Lab Invest ; 100(1): 52-63, 2020 01.
Article in English | MEDLINE | ID: mdl-31477795

ABSTRACT

Autophagy appears to play a role in the etiology and progress of misfolded protein disorders. Although this process is dysregulated in prion diseases, it is unknown whether this impairment is a cause or a consequence of prion neuropathology. The study of autophagy during the progress of the disease could elucidate its role. For this purpose, we have investigated its regulation at different stages of the disease in Tg338 mice, a transgenic murine model that overexpresses the highly susceptible ovine VRQ prion protein allele. Mice were intracerebrally inoculated with mouse-adapted classical scrapie and euthanized at the preclinical and clinical stages of the disease. Regulation of autophagy was investigated analyzing the distribution of LC3-B and p62 proteins by immunohistochemistry. Moreover, the expression of genes involved in autophagy regulation was quantified by real-time PCR. LC3-B and p62 proteins were downregulated and upregulated, respectively, in the central nervous system of infected mice with clinical signs of scrapie. Accumulation of p62 correlated with scrapie-related lesions, suggesting an impairment of autophagy in highly prion-affected areas. In addition, Gas5 (growth arrest-specific 5), Atg5 (autophagy-related 5), and Fbxw7 (F-box and WD repeat domain containing 7) transcripts were downregulated in mesencephalon and cervical spinal cord of the same group of animals. The impairment of autophagic machinery seems to be part of the pathological process of scrapie, but only during the late stage of prion infection. Similarities between Tg338 mice and the natural ovine disease make them a reliable in vivo model to study prion infection and autophagy side by side.


Subject(s)
Autophagy , Disease Models, Animal , Scrapie/metabolism , Animals , Brain/metabolism , Brain/pathology , Cervical Cord/metabolism , Mice, Transgenic , RNA, Messenger/metabolism , RNA, Untranslated/metabolism , Scrapie/etiology , Scrapie/pathology , Sheep
11.
Neurobiol Dis ; 144: 105057, 2020 10.
Article in English | MEDLINE | ID: mdl-32829029

ABSTRACT

Tau aggregates consisting of hyperphosphorylated tau fibrils are associated with many neurodegenerative diseases, including Alzheimer's disease, Pick's disease, frontotemporal dementia, and progressive supranuclear palsy. Tau may contribute to the pathogenesis of these diseases, collectively referred to as tauopathies. In human genetic prion diseases, tau aggregates are detected in association with amyloid plaques consisting of prion protein (PrP). However, the role of abnormal tau aggregates in PrP amyloid disease remains unclear. Previously we inoculated scrapie prions into transgenic mice expressing human tau, mouse tau, glycophosphatidylinositol (GPI) anchored PrP, and anchorless PrP. These mice developed both spongiform vacuolar pathology and PrP amyloid pathology, and human tau was detected near PrP amyloid plaques. However, the presence of human tau did not alter the disease tempo or prion-induced neuropathology. In the present study, we tested mice which more closely modeled familial human prion disease. These mice expressed human tau but lacked both mouse tau and GPI-anchored PrP. However, they did produce anchorless PrP, resulting in perivascular PrP amyloid plaques, i.e. cerebral amyloid angiopathy (CAA), without spongiform degeneration. Typical of PrP amyloid disease, the clinical course was very slow in this model. Nevertheless, the accumulation of aggregated, phosphorylated human tau and its association with PrP amyloid plaques failed to alter the timing or course of the clinical disease observed. These data suggest that human tau does not contribute to the pathogenesis of mouse PrP amyloid brain disease and raise the possibility that tau may also not be pathogenic in human PrP amyloid disease.


Subject(s)
Brain/metabolism , Cerebral Amyloid Angiopathy/metabolism , Plaque, Amyloid/metabolism , Prion Proteins/metabolism , Protein Aggregates , Scrapie/metabolism , tau Proteins/metabolism , Animals , Brain/pathology , Cerebral Amyloid Angiopathy/pathology , Disease Progression , Humans , Mice , Mice, Transgenic , Phosphorylation , Plaque, Amyloid/pathology , Scrapie/pathology , tau Proteins/genetics
12.
PLoS Pathog ; 14(11): e1007424, 2018 11.
Article in English | MEDLINE | ID: mdl-30496289

ABSTRACT

Transmissible spongiform encephalopathies (TSEs) are caused by the prion, which consists essentially of PrPSc, an aggregated, conformationally modified form of the cellular prion protein (PrPC). Although TSEs can be experimentally transmitted by intracerebral inoculation, most instances of infection in the field occur through extracerebral routes. The epidemics of kuru and variant Creutzfeldt-Jakob disease were caused by dietary exposure to prions, and parenteral administration of prion-contaminated hormones has caused hundreds of iatrogenic TSEs. In all these instances, the development of postexposure prophylaxis relies on understanding of how prions propagate from the site of entry to the brain. While much evidence points to lymphoreticular invasion followed by retrograde transfer through peripheral nerves, prions are present in the blood and may conceivably cross the blood-brain barrier directly. Here we have addressed the role of the blood-brain barrier (BBB) in prion disease propagation using Pdgfbret/ret mice which possess a highly permeable BBB. We found that Pdgfbret/ret mice have a similar prion disease incubation time as their littermate controls regardless of the route of prion transmission. These surprising results indicate that BBB permeability is irrelevant to the initiation of prion disease, even when prions are administered parenterally.


Subject(s)
Blood-Brain Barrier/metabolism , Prion Diseases/metabolism , Prions/metabolism , Animals , Biological Transport , Brain/blood supply , Brain/pathology , Cattle , Creutzfeldt-Jakob Syndrome/pathology , Disease Models, Animal , Encephalopathy, Bovine Spongiform/pathology , Humans , Mice , Prion Diseases/transmission , Prion Proteins/metabolism , Prions/pathogenicity , Scrapie/pathology
13.
Metabolomics ; 16(6): 72, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32533504

ABSTRACT

INTRODUCTION: Prion disease is a form of neurodegenerative disease caused by the misfolding and aggregation of cellular prion protein (PrPC). The neurotoxicity of the misfolded form of prion protein, PrPSc still remains understudied. Here we try to investigate this issue using a metabolomics approach. OBJECTIVES: The intention was to identify and quantify the small-in-size and water-soluble metabolites extracted from mice brains infected with the Rocky Mountain Laboratory isolate of mouse-adapted scrapie prions (RML) and track changes in these metabolites during disease evolution. METHODS: A total of 73 mice were inoculated with RML prions or normal brain homogenate control; brains were harvested at 30, 60, 90, 120 and 150 days post-inoculation (dpi). We devised a high-efficiency metabolite extraction method and used nuclear magnetic resonance spectroscopy to identify and quantify 50 metabolites in the brain extracts. Data were analyzed using multivariate approach. RESULTS: Brain metabolome profiles of RML infected animals displayed continuous changes throughout the course of disease. Among the analyzed metabolites, the most noteworthy changes included increases in myo-inositol and glutamine as well as decreases in 4-aminobutyrate, acetate, aspartate and taurine. CONCLUSION: We report a novel metabolite extraction method for lipid-rich tissue. As all the major metabolites are identifiable and quantifiable by magnetic resonance spectroscopy, this study suggests that tracking of neurochemical profiles could be effective in monitoring the progression of neurodegenerative diseases and useful for assessing the efficacy of candidate therapeutics.


Subject(s)
Metabolomics/methods , Prions/metabolism , Scrapie/metabolism , Animals , Brain/metabolism , Disease Progression , Female , Male , Metabolome/physiology , Mice , Mice, Inbred Strains , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Prions/chemistry , Scrapie/pathology
14.
Med Microbiol Immunol ; 209(1): 81-94, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31720785

ABSTRACT

Activation of complement system in central nervous system (CNS) of the patients suffering from prion diseases or animal models infected with prion agents experimentally is reported repeatedly, but which pathways are involved in the complement system during prion infection is not well documented. Here, we evaluated the level of complement factor B (CFB), which is the key factor that triggers alterative pathway (AP) of complement in the brain tissues of scrapie-infected mice with various methodologies. We found that the levels of mRNA and protein of CFB significantly increased in the brain tissues of scrapie-infected mice. Morphologically, the increased CFB-specific signal overlapped with the elevated C3 signal in brain sections of scrapie-infected mice, meanwhile overlapped with damaged neurons and activated microglia, but not with the proliferative astrocytes. Additionally, the level of complement factor P (CFP), the key positive regulator of AP, also increased remarkably in the brain tissues of infected mice. The transcriptional levels of CD55 and CD46, two negative regulators of AP, decreased without significance in brain tissues of scrapie-infected mice at the terminal stage. However, the mRNA and protein levels of CFH, another negative regulator of AP, increased. Through the dynamic analyses of the expressions of CFB, CFP, and CFH in brain sections of 139A-infected mice, which were collected at different time-points during incubation period, illustrated time-dependent increase levels of each factor during the incubation period of scrapie infection. Taken together, our data here demonstrate that the AP of complement cascade is activated in the CNS microenvironment during prion infection.


Subject(s)
Brain/immunology , Complement Pathway, Alternative/immunology , Complement System Proteins/immunology , Scrapie/immunology , Animals , Biomarkers , Brain/metabolism , Brain/pathology , Complement C3/immunology , Complement C3/metabolism , Complement System Proteins/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression , Genes, Reporter , Immunohistochemistry , Mice , Microglia/metabolism , Neurons/metabolism , PrPSc Proteins/immunology , PrPSc Proteins/metabolism , Scrapie/metabolism , Scrapie/pathology
15.
Vet Pathol ; 57(1): 90-107, 2020 01.
Article in English | MEDLINE | ID: mdl-31331254

ABSTRACT

Mammalian transmissible spongiform encephalopathies (TSEs) display marked activation of astrocytes and microglia that precedes neuronal loss. Investigation of clinical parallels between TSEs and other neurodegenerative protein misfolding diseases, such as Alzheimer's disease, has revealed similar patterns of neuroinflammatory responses to the accumulation of self-propagating amyloids. The contribution of glial activation to the progression of protein misfolding diseases is incompletely understood, with evidence for mediation of both protective and deleterious effects. Glial populations are heterogeneously distributed throughout the brain and capable of dynamic transitions along a spectrum of functional activation states between pro- and antiinflammatory polarization extremes. Using a murine model of Rocky Mountain Laboratory scrapie, the neuroinflammatory response to prion infection was characterized by evaluating glial activation across 15 brain regions over time and correlating it to traditional markers of prion neuropathology, including vacuolation and PrPSc deposition. Quantitative immunohistochemistry was used to evaluate glial expression of iNOS and Arg1, markers of classical and alternative glial activation, respectively. The results indicate progressive upregulation of iNOS in microglia and a mixed astrocytic profile featuring iNOS expression in white matter tracts and detection of Arg1-positive populations throughout the brain. These data establish a temporospatial lesion profile for this prion infection model and demonstrate evidence of multiple glial activation states.


Subject(s)
Inflammation/pathology , Nitric Oxide Synthase Type II/metabolism , PrPSc Proteins/metabolism , Prion Diseases/pathology , Prions/metabolism , Scrapie/pathology , Animals , Arginase/metabolism , Astrocytes/pathology , Brain/pathology , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microglia/pathology , Neuroglia/pathology , Up-Regulation
16.
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
17.
J Biol Chem ; 293(21): 8020-8031, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29636413

ABSTRACT

A central step in the pathogenesis of prion diseases is the conformational transition of the cellular prion protein (PrPC) into the scrapie isoform, denoted PrPSc Studies in transgenic mice have indicated that this conversion requires a direct interaction between PrPC and PrPSc; however, insights into the underlying mechanisms are still missing. Interestingly, only a subfraction of PrPC is converted in scrapie-infected cells, suggesting that not all PrPC species are suitable substrates for the conversion. On the basis of the observation that PrPC can form homodimers under physiological conditions with the internal hydrophobic domain (HD) serving as a putative dimerization domain, we wondered whether PrP dimerization is involved in the formation of neurotoxic and/or infectious PrP conformers. Here, we analyzed the possible impact on dimerization of pathogenic mutations in the HD that induce a spontaneous neurodegenerative disease in transgenic mice. Similarly to wildtype (WT) PrPC, the neurotoxic variant PrP(AV3) formed homodimers as well as heterodimers with WTPrPC Notably, forced PrP dimerization via an intermolecular disulfide bond did not interfere with its maturation and intracellular trafficking. Covalently linked PrP dimers were complex glycosylated, GPI-anchored, and sorted to the outer leaflet of the plasma membrane. However, forced PrPC dimerization completely blocked its conversion into PrPSc in chronically scrapie-infected mouse neuroblastoma cells. Moreover, PrPC dimers had a dominant-negative inhibition effect on the conversion of monomeric PrPC Our findings suggest that PrPC monomers are the major substrates for PrPSc propagation and that it may be possible to halt prion formation by stabilizing PrPC dimers.


Subject(s)
Neuroblastoma/prevention & control , Prion Proteins/chemistry , Prion Proteins/metabolism , Protein Multimerization , Scrapie/prevention & control , Animals , HeLa Cells , Humans , Mice , Mice, Transgenic , Neuroblastoma/pathology , Protein Transport , Scrapie/pathology , Tumor Cells, Cultured
18.
J Biol Chem ; 293(33): 12730-12740, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29934306

ABSTRACT

Prions are composed solely of the pathological isoform (PrPSc) of the normal cellular prion protein (PrPC). Identification of different PrPSc structures is crucially important for understanding prion biology because the pathogenic properties of prions are hypothesized to be encoded in the structures of PrPSc However, these structures remain yet to be identified, because of the incompatibility of PrPSc with conventional high-resolution structural analysis methods. Previously, we reported that the region between the first and the second α-helix (H1∼H2) of PrPC might cooperate with the more C-terminal side region for efficient interactions with PrPSc From this starting point, we created a series of PrP variants with two cysteine substitutions (C;C-PrP) forming a disulfide-crosslink between H1∼H2 and the distal region of the third helix (Ctrm). We then assessed the conversion capabilities of the C;C-PrP variants in N2a cells infected with mouse-adapted scrapie prions (22L-ScN2a). Specifically, Cys substitutions at residues 165, 166, or 168 in H1∼H2 were combined with cysteine scanning along Ctrm residues 220-229. We found that C;C-PrPs are expressed normally with glycosylation patterns and subcellular localization similar to WT PrP, albeit differing in expression levels. Interestingly, some C;C-PrPs converted to protease-resistant isoforms in the 22L-ScN2a cells, but not in Fukuoka1 prion-infected cells. Crosslink patterns of convertible C;C-PrPs indicated a positional change of H1∼H2 toward Ctrm in PrPSc-induced conformational conversion. Given the properties of the C;C-PrPs reported here, we propose that these PrP variants may be useful tools for investigating prion strain-specific structures and structure-phenotype relationships of PrPSc.


Subject(s)
Cross-Linking Reagents/chemistry , Disulfides/chemistry , Neuroblastoma/pathology , PrPSc Proteins/metabolism , Prions/pathogenicity , Protein Conformation , Scrapie/pathology , Amino Acid Sequence , Animals , Humans , Mice , Neuroblastoma/metabolism , PrPSc Proteins/chemistry , Prions/chemistry , Prions/genetics , Scrapie/metabolism , Sequence Homology , Tumor Cells, Cultured
19.
J Virol ; 92(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29769333

ABSTRACT

Microglial cells in the central nervous system play important roles in neurodevelopment and resistance to infection, yet microglia can become neurotoxic under some conditions. An early event during prion infection is the activation of microglia and astrocytes in the brain prior to damage or death of neurons. Previous prion disease studies using two different strategies to manipulate signaling through the microglial receptor CSF-1R reported contrary effects on survival from prion disease. However, in these studies, reductions of microglial numbers and function were variable, thus confounding interpretation of the results. In the present work, we used oral treatment with a potent inhibitor of CSF-1R, PLX5622, to eliminate 78 to 90% of microglia from cortex early during the course of prion infection. Oral drug treatment early after infection with the RML scrapie strain significantly accelerated vacuolation, astrogliosis, and deposition of disease-associated prion protein. Furthermore, drug-treated mice had advanced clinical disease requiring euthanasia 31 days earlier than untreated control mice. Similarly, PLX5622 treatment during the preclinical phase at 80 days postinfection with RML scrapie also accelerated disease and resulted in euthanasia of mice 33 days earlier than infected controls. PLX5622 also accelerated clinical disease after infection with scrapie strains ME7 and 22L. Thus, microglia are critical in host defense during prion disease. The early accumulation of PrPSc in the absence of microglia suggested that microglia may function by clearing PrPSc, resulting in longer survival.IMPORTANCE Microglia contribute to many aspects of health and disease. When activated, microglia can be beneficial by repairing damage in the central nervous system (CNS) or they can turn harmful by becoming neurotoxic. In prion and prionlike diseases, the involvement of microglia in disease is unclear. Previous studies suggest that microglia can either speed up or slow down disease. In this study, we infected mice with prions and depleted microglia from the brains of mice using PLX5622, an effective CSF-1R tyrosine kinase inhibitor. Microglia were markedly reduced in brains, and prion disease was accelerated, so that mice needed to be euthanized 20 to 33 days earlier than infected control mice due to advanced clinical disease. Similar results occurred when mice were treated with PLX5622 at 80 days after infection, which was just prior to the start of clinical signs. Thus, microglia are important for removing prions, and the disease is faster when microglia are depleted.


Subject(s)
Microglia/cytology , Microglia/drug effects , Organic Chemicals/adverse effects , PrPSc Proteins/metabolism , Scrapie/metabolism , Administration, Oral , Animals , Apoptosis , Disease Models, Animal , Female , Male , Mice , Microglia/metabolism , Microglia/pathology , Organic Chemicals/administration & dosage , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Scrapie/chemically induced , Scrapie/pathology , Severity of Illness Index
20.
Proc Natl Acad Sci U S A ; 113(48): 13851-13856, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27849581

ABSTRACT

Recombinant C-terminally truncated prion protein PrP23-144 (which corresponds to the Y145Stop PrP variant associated with a Gerstmann-Sträussler-Scheinker-like prion disease) spontaneously forms amyloid fibrils with a parallel in-register ß-sheet architecture and ß-sheet core mapping to residues ∼112-139. Here we report that mice (both tga20 and wild type) inoculated with a murine (moPrP23-144) version of these fibrils develop clinical prion disease with a 100% attack rate. Remarkably, even though fibrils in the inoculum lack the entire C-terminal domain of PrP, brains of clinically sick mice accumulate longer proteinase K-resistant (PrPres) fragments of ∼17-32 kDa, similar to those observed in classical scrapie strains. Shorter, Gerstmann-Sträussler-Scheinker-like PrPres fragments are also present. The evidence that moPrP23-144 amyloid fibrils generated in the absence of any cofactors are bona fide prions provides a strong support for the protein-only hypothesis of prion diseases in its pure form, arguing against the notion that nonproteinaceous cofactors are obligatory structural components of all infectious prions. Furthermore, our finding that a relatively short ß-sheet core of PrP23-144 fibrils (residues ∼112-139) with a parallel in-register organization of ß-strands is capable of seeding the conversion of full-length prion protein to the infectious form has important implications for the ongoing debate regarding structural aspects of prion protein conversion and molecular architecture of mammalian prions.


Subject(s)
Amyloid/chemistry , Gerstmann-Straussler-Scheinker Disease/genetics , Prion Diseases/genetics , Prion Proteins/chemistry , Amyloid/adverse effects , Amyloid/genetics , Animals , Gerstmann-Straussler-Scheinker Disease/etiology , Gerstmann-Straussler-Scheinker Disease/pathology , Humans , Mice , Prion Diseases/etiology , Prion Diseases/pathology , Prion Proteins/adverse effects , Prion Proteins/genetics , Protein Conformation, beta-Strand/genetics , Scrapie/genetics , Scrapie/pathology , Spectroscopy, Fourier Transform Infrared
SELECTION OF CITATIONS
SEARCH DETAIL