Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
1.
Int J Mol Sci ; 21(9)2020 May 02.
Article in English | MEDLINE | ID: mdl-32370224

ABSTRACT

Neuroinflammation has been correlated with the progress of neurodegeneration in many neuropathologies. Although glial cells have traditionally been considered to be protective, the concept of them as neurotoxic cells has recently emerged. Thus, a major unsolved question is the exact role of astroglia and microglia in neurodegenerative disorders. On the other hand, it is well known that glucocorticoids are the first choice to regulate inflammation and, consequently, neuroglial inflammatory activity. The objective of this study was to determine how chronic dexamethasone treatment influences the host immune response and to characterize the beneficial or detrimental role of glial cells. To date, this has not been examined using a natural neurodegenerative model of scrapie. With this aim, immunohistochemical expression of glial markers, prion protein accumulation, histopathological lesions and clinical evolution were compared with those in a control group. The results demonstrated how the complex interaction between glial populations failed to compensate for brain damage in natural conditions, emphasizing the need for using natural models. Additionally, the data showed that modulation of neuroinflammation by anti-inflammatory drugs might become a research focus as a potential therapeutic target for prion diseases, similar to that considered previously for other neurodegenerative disorders classified as prion-like diseases.


Subject(s)
Astrocytes/drug effects , Dexamethasone/pharmacology , Microglia/drug effects , Neuroglia/drug effects , Scrapie/physiopathology , Animals , Anti-Inflammatory Agents/pharmacology , Astrocytes/cytology , Astrocytes/metabolism , Female , Kaplan-Meier Estimate , Microglia/cytology , Microglia/metabolism , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/physiopathology , Neuroglia/metabolism , Prion Proteins/metabolism , Scrapie/diagnosis , Scrapie/metabolism , Sheep
2.
Brain Pathol ; 29(2): 248-262, 2019 03.
Article in English | MEDLINE | ID: mdl-30588682

ABSTRACT

Transmissible spongiform encephalopathies (TSEs) or prion diseases of animals notably include scrapie in small ruminants, chronic wasting disease (CWD) in cervids and classical bovine spongiform encephalopathy (C-BSE). As the transmission barrier phenomenon naturally limits the propagation of prions from one species to another, and the lack of epidemiological evidence for an association with human prion diseases, the zoonotic potential of these diseases was for a long time considered negligible. However, in 1996, C-BSE was recognized as the cause of a new human prion disease, variant Creutzfeldt-Jakob disease (vCJD), which triggered an unprecedented public health crisis in Europe. Large-scale epidemio-surveillance programs for scrapie and C-BSE that were implemented in the EU after the BSE crisis revealed that the distribution and prevalence of prion diseases in the ruminant population had previously been underestimated. They also led to the recognition of new forms of TSEs (named atypical) in cattle and small ruminants and to the recent identification of CWD in Europe. At this stage, the characterization of the strain diversity and zoonotic abilities associated with animal prion diseases remains largely incomplete. However, transmission experiments in nonhuman primates and transgenic mice expressing human PrP clearly indicate that classical scrapie, and certain forms of atypical BSE (L-BSE) or CWD may have the potential to infect humans. The remaining uncertainties about the origins and relationships between animal prion diseases emphasize the importance of the measures implemented to limit human exposure to these potentially zoonotic agents, and of continued surveillance for both animal and human prion diseases.


Subject(s)
Prion Diseases/genetics , Prion Diseases/metabolism , Prion Diseases/physiopathology , Animals , Brain/metabolism , Cattle , Creutzfeldt-Jakob Syndrome/epidemiology , Creutzfeldt-Jakob Syndrome/physiopathology , Encephalopathy, Bovine Spongiform/epidemiology , Encephalopathy, Bovine Spongiform/physiopathology , Humans , Prions/metabolism , Scrapie/epidemiology , Scrapie/physiopathology
3.
J Gen Virol ; 98(6): 1563-1569, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28613153

ABSTRACT

Proteinase-activated receptor 2 (PAR2) is suspected to modulate the pathogenesis of various neurodegenerative conditions. We previously described delayed onset of clinical symptoms and prolonged survival of PAR2-deficient mice after intracerebral inoculation with prions. Here we report the results from a refined blinded study that aimed to investigate the effects of PAR2 deletion on scrapie pathogenesis after peripheral infection. This study failed to confirm that PAR2 deficiency impacts on the length of the incubation period, with PAR2-/- and PAR2+/+ littermates developing scrapie at the same time. To clarify the discrepancy between the two observations, we repeated the intracerebral inoculation study while utilizing our refined protocol, which aimed to limit possible sources of experimental bias. The study again failed to confirm the significant effect of PAR2 expression on the course of prion infection. Our report emphasizes and discusses the importance of unbiased experimental design and the selection of proper genetic controls when using genetically altered animal models for prion pathogenesis studies.


Subject(s)
Receptor, PAR-2/metabolism , Scrapie/physiopathology , Animals , Disease Models, Animal , Female , Mice , Mice, Knockout , Receptor, PAR-2/deficiency
4.
J Virol ; 91(10)2017 05 15.
Article in English | MEDLINE | ID: mdl-28275192

ABSTRACT

After oral exposure, the early replication of certain prion strains upon stromal cell-derived follicular dendritic cells (FDC) in the Peyer's patches in the small intestine is essential for the efficient spread of disease to the brain. However, little is known of how prions are initially conveyed from the gut lumen to establish infection on FDC. Our previous data suggest that mononuclear phagocytes such as CD11c+ conventional dendritic cells play an important role in the initial propagation of prions from the gut lumen into Peyer's patches. However, whether these cells conveyed orally acquired prions toward FDC within Peyer's patches was not known. The chemokine CXCL13 is expressed by FDC and follicular stromal cells and modulates the homing of CXCR5-expressing cells toward the FDC-containing B cell follicles. Here, novel compound transgenic mice were created in which a CXCR5 deficiency was specifically restricted to CD11c+ cells. These mice were used to determine whether CXCR5-expressing conventional dendritic cells propagate prions toward FDC after oral exposure. Our data show that in the specific absence of CXCR5-expressing conventional dendritic cells the early accumulation of prions upon FDC in Peyer's patches and the spleen was impaired, and disease susceptibility significantly reduced. These data suggest that CXCR5-expressing conventional dendritic cells play an important role in the efficient propagation of orally administered prions toward FDC within Peyer's patches in order to establish host infection.IMPORTANCE Many natural prion diseases are acquired by oral consumption of contaminated food or pasture. Once the prions reach the brain they cause extensive neurodegeneration, which ultimately leads to death. In order for the prions to efficiently spread from the gut to the brain, they first replicate upon follicular dendritic cells within intestinal Peyer's patches. How the prions are first delivered to follicular dendritic cells to establish infection was unknown. Understanding this process is important since treatments which prevent prions from infecting follicular dendritic cells can block their spread to the brain. We created mice in which mobile conventional dendritic cells were unable to migrate toward follicular dendritic cells. In these mice the early accumulation of prions on follicular dendritic cells was impaired and oral prion disease susceptibility was reduced. This suggests that prions exploit conventional dendritic cells to facilitate their initial delivery toward follicular dendritic cells to establish host infection.


Subject(s)
Dendritic Cells, Follicular/immunology , Encephalopathy, Bovine Spongiform/immunology , Encephalopathy, Bovine Spongiform/physiopathology , Gene Expression , Prions/pathogenicity , Receptors, CXCR5/genetics , Animals , Brain/pathology , Cattle , Chemokine CXCL13/genetics , Dendritic Cells/pathology , Dendritic Cells/physiology , Dendritic Cells, Follicular/pathology , Disease Susceptibility , Intestine, Small/immunology , Intestine, Small/pathology , Mice , Mice, Transgenic , Peyer's Patches/immunology , Peyer's Patches/pathology , Prions/physiology , Scrapie/physiopathology , Spleen/immunology , Spleen/pathology
5.
Acta Neuropathol Commun ; 3: 21, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25853789

ABSTRACT

INTRODUCTION: Scrapie and bovine spongiform encephalopathy (BSE) are transmissible spongiform encephalopathies (TSEs) which naturally affect small and large ruminants respectively. However, small ruminants, which are susceptible to BSE under experimental conditions, have been exposed to the same or similar contaminated food additives as cattle. To date two natural cases of BSE in small ruminants have been reported. As a result surveillance projects, combined with appropriate control measures, have been established throughout the European Union (EU) to minimize the overall incidence of small ruminant TSEs. Although BSE can be differentiated from classical scrapie (subsequently referred to as scrapie) if appropriate discriminatory tests are applied, the value of these tests in BSE/scrapie co-infection scenarios has not been evaluated fully. Mouse bioassay is regarded as the gold standard regarding differentiation of distinct TSE strains and has been used as to resolve TSE cases were laboratory tests produced equivocal results. However, the ability of this method to discriminate TSE strains when they co-exist has not been examined systematically. To address this issue we prepared in vitro mixtures of ovine BSE and scrapie and used them to challenge RIII, C57BL/6 and VM mice. RESULTS: Disease phenotype analysis in all three mouse lines indicated that most phenotypic parameters (attack rates, incubation periods, lesion profiles and Western blots) were compatible with scrapie phenotypes as were immunohistochemistry (IHC) data from RIII and C57BL/6 mice. However, in VM mice that were challenged with BSE/scrapie mixtures a single BSE-associated IHC feature was identified, indicating the existence of BSE in animals where the scrapie phenotype was dominant. CONCLUSIONS: We conclude that wild type mouse bioassay is of limited value in detecting BSE in the presence of scrapie particularly if the latter is in relative excess.


Subject(s)
Biological Assay/methods , Mice, Inbred Strains , Phenotype , Prion Diseases/physiopathology , Scrapie/physiopathology , Species Specificity , Animals , Blotting, Western , Cattle , Immunohistochemistry , Mice , Mice, Inbred C57BL , Prion Diseases/metabolism , Scrapie/metabolism
6.
Can J Vet Res ; 79(1): 68-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25673912

ABSTRACT

Meadow voles (Microtus pennsylvanicus) are permissive to chronic wasting disease (CWD) infection, but their susceptibility to other transmissible spongiform encephalopathies (TSEs) is poorly characterized. In this initial study, we intracerebrally challenged 6 meadow voles with 2 isolates of sheep scrapie. Three meadow voles acquired a TSE after the scrapie challenge and an extended incubation period. The glycoform profile of proteinase K-resistant prion protein (PrP(res)) in scrapie-sick voles remained similar to the sheep inocula, but differed from that of voles clinically affected by CWD. Vacuolization patterns and disease-associated prion protein (PrP(Sc)) deposition were generally similar in all scrapie-affected voles, except in the hippocampus, where PrP(Sc) staining varied markedly among the animals. Our results demonstrate that meadow voles can acquire a TSE after intracerebral scrapie challenge and that this species could therefore prove useful for characterizing scrapie isolates.


Les campagnols des champs (Microtus pennsylvanicus) sont permissifs à l'infection par l'agent de la maladie débilitante chronique (MDC), mais leur susceptibilité aux autres encéphalopathies spongiformes transmissibles (EST) est peu caractérisée. Dans cette première étude, six campagnols ont été inoculés par voie intracérébrale avec deux isolats de l'agent de la tremblante du mouton. Trois campagnols ont présenté une EST suite à l'inoculation de l'agent de la tremblante après une période d'incubation prolongée. Le profil glycoforme de la protéine prion résistante à la protéinase K (PrPres) chez les campagnols atteints demeura similaire à celui de l'inoculum ovin, mais différait de celui des campagnols affectés cliniquement de MDC. Les patrons de vacuolisation et le dépôt de protéine prion associée à la maladie (PrPSc) étaient généralement similaires chez tous les campagnols affectés de tremblante, à l'exception de l'hippocampe, où la coloration de PrPSc variait de façon marquée parmi les animaux. Ces résultats démontrent que les campagnols peuvent souffrir d'EST après inoculation intracérébrale de l'agent de la tremblante et que cette espèce pourrait s'avérer utile pour caractériser les isolats de l'agent de la tremblante.(Traduit par Docteur Serge Messier).


Subject(s)
Arvicolinae/metabolism , Brain/metabolism , Disease Models, Animal , Prions/metabolism , Scrapie/metabolism , Animals , Arvicolinae/physiology , Biomarkers/metabolism , Brain/pathology , Phenotype , Scrapie/pathology , Scrapie/physiopathology , Sheep , Wasting Disease, Chronic/metabolism
7.
Prion ; 8(5): 339-43, 2014.
Article in English | MEDLINE | ID: mdl-25495671

ABSTRACT

Previous studies have demonstrated that Shadoo (Sho), a GPI-linked glycoprotein encoded by the Sprn gene with a membrane localization similar to PrP(C), is reduced in the brains of rodents with terminal prion disease. To determine the functional significance of Sho in prion disease pathogenesis, Sho-deficient mice were generated by gene targeting. Sho knockout and control wild-type (WT) mice were infected with themouse-adapted scrapie strains 22L or RML. No significant differences in survival, the incubation period of prion disease or other disease features were observed between Sho mutant and WT mice. In this model of prion disease, Sho removal had no effect on disease pathogenesis.


Subject(s)
Nerve Tissue Proteins/physiology , Scrapie/physiopathology , Animals , GPI-Linked Proteins , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Scrapie/genetics , Survival Rate
8.
J Clin Invest ; 124(2): 847-58, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24430187

ABSTRACT

The symptoms of prion infection can take years or decades to manifest following the initial exposure. Molecular markers of prion disease include accumulation of the misfolded prion protein (PrPSc), which is derived from its cellular precursor (PrPC), as well as downregulation of the PrP-like Shadoo (Sho) glycoprotein. Given the overlapping cellular environments for PrPC and Sho, we inferred that PrPC levels might also be altered as part of a host response during prion infection. Using rodent models, we found that, in addition to changes in PrPC glycosylation and proteolytic processing, net reductions in PrPC occur in a wide range of prion diseases, including sheep scrapie, human Creutzfeldt-Jakob disease, and cervid chronic wasting disease. The reduction in PrPC results in decreased prion replication, as measured by the protein misfolding cyclic amplification technique for generating PrPSc in vitro. While PrPC downregulation is not discernible in animals with unusually short incubation periods and high PrPC expression, slowly evolving prion infections exhibit downregulation of the PrPC substrate required for new PrPSc synthesis and as a receptor for pathogenic signaling. Our data reveal PrPC downregulation as a previously unappreciated element of disease pathogenesis that defines the extensive, presymptomatic period for many prion strains.


Subject(s)
Prion Diseases/physiopathology , Animals , Arvicolinae , Brain/metabolism , Cell Line , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/physiopathology , Disease Progression , Down-Regulation , Glycosylation , Humans , Mesocricetus , Mice , Mice, Transgenic , PrPC Proteins/chemistry , PrPSc Proteins/chemistry , Prion Diseases/metabolism , Protein Isoforms/chemistry , Scrapie/metabolism , Scrapie/physiopathology , Signal Transduction , Time Factors , Wasting Disease, Chronic
9.
BMC Genomics ; 15: 59, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24450868

ABSTRACT

BACKGROUND: Prion diseases are characterized by the accumulation of the pathogenic PrPSc protein, mainly in the brain and the lymphoreticular system. Although prions multiply/accumulate in the lymph nodes without any detectable pathology, transcriptional changes in this tissue may reflect biological processes that contribute to the molecular pathogenesis of prion diseases. Little is known about the molecular processes that occur in the lymphoreticular system in early and late stages of prion disease. We performed a microarray-based study to identify genes that are differentially expressed at different disease stages in the mesenteric lymph node of sheep naturally infected with scrapie. Oligo DNA microarrays were used to identify gene-expression profiles in the early/middle (preclinical) and late (clinical) stages of the disease. RESULTS: In the clinical stage of the disease, we detected 105 genes that were differentially expressed (≥2-fold change in expression). Of these, 43 were upregulated and 62 downregulated as compared with age-matched negative controls. Fewer genes (50) were differentially expressed in the preclinical stage of the disease. Gene Ontology enrichment analysis revealed that the differentially expressed genes were largely associated with the following terms: glycoprotein, extracellular region, disulfide bond, cell cycle and extracellular matrix. Moreover, some of the annotated genes could be grouped into 3 specific signaling pathways: focal adhesion, PPAR signaling and ECM-receptor interaction. We discuss the relationship between the observed gene expression profiles and PrPSc deposition and the potential involvement in the pathogenesis of scrapie of 7 specific differentially expressed genes whose expression levels were confirmed by real time-PCR. CONCLUSIONS: The present findings identify new genes that may be involved in the pathogenesis of natural scrapie infection in the lymphoreticular system, and confirm previous reports describing scrapie-induced alterations in the expression of genes involved in protein misfolding, angiogenesis and the oxidative stress response. Further studies will be necessary to determine the role of these genes in prion replication, dissemination and in the response of the organism to this disease.


Subject(s)
Gene Expression Profiling/veterinary , Gene Expression Regulation , Lymph Nodes/metabolism , Scrapie/physiopathology , Sheep/genetics , Sheep/metabolism , Animals , Cluster Analysis , Down-Regulation , Focal Adhesions/genetics , Oligonucleotide Array Sequence Analysis , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Prions/genetics , Prions/metabolism , Receptors, Cytoadhesin/genetics , Receptors, Cytoadhesin/metabolism , Scrapie/metabolism , Scrapie/pathology , Up-Regulation
10.
Acta Biochim Biophys Sin (Shanghai) ; 45(6): 485-93, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23583976

ABSTRACT

The infectious agent of the transmissible spongiform encephalopathies, or prion diseases, has been the center of intense debate for decades. Years of studies have provided overwhelming evidence to support the prion hypothesis that posits a protein conformal infectious agent is responsible for the transmissibility of the disease. The recent studies that generate prion infectivity with purified bacterially expressed recombinant prion protein not only provides convincing evidence supporting the core of the prion hypothesis, that a pathogenic conformer of host prion protein is able to seed the conversion of its normal counterpart to the likeness of itself resulting in the replication of the pathogenic conformer and occurrence of disease, they also indicate the importance of cofactors, particularly lipid or lipid-like molecules, in forming the protein conformation-based infectious agent. This article reviews the literature regarding the chemical nature of the infectious agent and the potential contribution from lipid molecules to prion infectivity, and discusses the important remaining questions in this research area.


Subject(s)
Lipids/physiology , Scrapie/physiopathology , Animals , Humans , Prions/chemistry , Prions/metabolism , Prions/radiation effects , Protein Conformation , Radiation, Ionizing , Scrapie/metabolism , Ultraviolet Rays
11.
Proc Natl Acad Sci U S A ; 109(34): 13733-8, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22869751

ABSTRACT

Copper influences the pathogenesis of prion disease, but whether it is beneficial or detrimental remains controversial. Copper homeostasis is also essential for normal physiology, as highlighted by the spectrum of diseases caused by disruption of the copper transporting enzymes ATP7A and ATP7B. Here, by using a forward genetics approach in mice, we describe the isolation of three alleles of Atp7a, each with different phenotypic consequences. The mildest of the three, Atp7a(brown), was insufficient to cause lethality in hemizygotes or mottling of the coat in heterozygotes, but did lead to coat hypopigmentation and reduced copper content in the brains of hemizygous males. When challenged with Rocky Mountain Laboratory scrapie, the onset of prion disease was delayed in Atp7a(brown) mice, and significantly less proteinase-resistant prion protein was found in the brains of moribund Atp7a(brown) mice compared with WT littermates. Our results establish that ATP7A-mediated copper homeostasis is important for the formation of pathogenic proteinase-resistant prion protein.


Subject(s)
Adenosine Triphosphatases/genetics , Cation Transport Proteins/genetics , Copper/metabolism , Mutation , Scrapie/genetics , Alleles , Animals , Copper-Transporting ATPases , Ethylnitrosourea/pharmacology , Homeostasis , Male , Menkes Kinky Hair Syndrome/genetics , Mice , Mice, Inbred C57BL , Mutagenesis , Phenotype , Pigmentation , Prions/metabolism , Scrapie/physiopathology
12.
Vet Microbiol ; 159(1-2): 23-32, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22482719

ABSTRACT

Neuronal loss is one of the characteristics of scrapie neuropathology. Previous analysis of brains from sheep naturally infected with scrapie that were in a terminal stage did not detect a clear induction of apoptosis, although molecular changes were evidenced. As neuronal death could be occurring early in scrapie, we developed a neuropathological and gene expression study of sheep infected with scrapie in a presymptomatic stage. The histopathology, immunolabelling of PrP(Sc), Bax and activated caspase-3, and the analysis of the expression of 7 genes involved in the regulation of the mitochondrial pathway of apoptosis were investigated in the following 4 central nervous system areas: medulla oblongata, diencephalon, frontal cortex and cerebellum. Moreover, TUNEL and NeuN immunolabelling was performed in the medulla oblongata. The PrP(Sc) immunolabelling in the four areas, as well as a neuropil spongiform change, were more evident in the terminal stage than in presymptomatic animals. Cytoplasmic Bax immunostaining was observed in the presymptomatic medulla oblongata. In contrast to symptomatic animals, the immunostaining was not extended to the hypothalamus, indicating the progression of Bax induction during the course of the disease. Although neither caspase-3 immunostaining nor the TUNEL technique detected neurons with apoptosis, NeuN-immunolabelled cell counting determined that presymptomatic animals have already suffered neuronal loss in a lower or equal degree than symptomatic animals. Finally, the gene expression profiles indicated that the mitochondrial pathway of apoptosis was activated with higher intensity in presymptomatic animals than in symptomatic sheep and confirmed the implication of genes such as BAX or AIF in the disease.


Subject(s)
Apoptosis/genetics , Brain/pathology , Gene Expression Regulation , Scrapie/pathology , Animals , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Biomarkers/metabolism , Brain/metabolism , Brain/physiopathology , Caspase 3/genetics , Caspase 3/metabolism , Female , Gene Expression Profiling , In Situ Nick-End Labeling , Neurons/pathology , Scrapie/genetics , Scrapie/metabolism , Scrapie/physiopathology , Sheep , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
J Virol ; 86(1): 25-35, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22031932

ABSTRACT

Infections with variant Creutzfeldt-Jakob disease (vCJD) have almost exclusively occurred in young patients, but the reasons for this age distribution are uncertain. Our data suggest that the pathogenesis of many peripherally acquired transmissible spongiform encephalopathy (TSE) agents is less efficient in aged individuals. Four vCJD cases linked to transfusion of vCJD-contaminated blood or blood products have been described. Three cases occurred in elderly patients, implying that intravenous exposure is more efficient in aged individuals than other peripheral routes. To test this hypothesis, young (6 to 8 weeks old) and aged (600 days old) mice were injected intravenously with a TSE agent. In aged and young mice, the intravenous route was more efficient than other peripheral routes of TSE agent exposure. However, in aged mice, disease pathogenesis was significantly reduced. Although most aged mice failed to develop clinical disease during their life spans, many showed histopathological signs of TSE disease in their brains. Thus, the effects of age on intravenous TSE pathogenesis may lead to significant levels of subclinical disease in the population. After peripheral exposure, many TSE agents accumulate upon follicular dendritic cells (FDCs) in lymphoid tissues before they infect the brain. In aged spleens, PrP(C) expression and TSE agent accumulation upon FDCs were reduced. Furthermore, the splenic marginal zone microarchitecture was substantially disturbed, adversely affecting the delivery of immune complexes to FDCs. This study is the first to suggest that the effects of aging on the microarchitecture and the function of the splenic marginal zone significantly influence the pathogenesis of an important pathogen.


Subject(s)
Aging/metabolism , Complement System Proteins/metabolism , Scrapie/pathology , Scrapie/physiopathology , Spleen/metabolism , Age Factors , Aged, 80 and over , Aging/pathology , Animals , Biological Transport , Dendritic Cells, Follicular/metabolism , Dendritic Cells, Follicular/pathology , Dendritic Cells, Follicular/virology , Disease Models, Animal , Female , Host-Pathogen Interactions , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PrPSc Proteins/genetics , PrPSc Proteins/metabolism , Scrapie/metabolism , Scrapie/virology , Spleen/pathology , Spleen/virology
14.
Vet Q ; 31(3): 115-28, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22029882

ABSTRACT

Transmissible spongiform encephalopathies (TSEs) or prion diseases are unique disorders that are not caused by infectious micro-organisms (bacteria or fungi), viruses or parasites, but rather seem to be the result of an infectious protein. TSEs are comprised of fatal neurodegenerative disorders affecting both human and animals. Prion diseases cause sponge-like degeneration of neuronal tissue and include (among others) Creutzfeldt-Jacob disease in humans, bovine spongiform encephalopathy (BSE) in cattle and scrapie in sheep. TSEs are characterized by the formation and accumulation of transmissible (infectious) disease-associated protease-resistant prion protein (PrP(Sc)), mainly in tissues of the central nervous system. The exact molecular processes behind the conversion of PrP(C) into PrP(Sc) are not clearly understood. Correlations between prion protein polymorphisms and disease have been found, however in what way these polymorphisms influence the conversion processes remains an enigma; is stabilization or destabilization of the prion protein the basis for a higher conversion propensity? Apart from the disease-associated polymorphisms of the prion protein, the molecular processes underlying conversion are not understood. There are some notions as to which regions of the prion protein are involved in refolding of PrP(C) into PrP(Sc) and where the most drastic structural changes take place. Direct interactions between PrP(C) molecules and/or PrP(Sc) are likely at the basis of conversion, however which specific amino acid domains are involved and to what extent these domains contribute to conversion resistance/sensitivity of the prion protein or the species barrier is still unknown.


Subject(s)
Prion Diseases/veterinary , Prions , Animals , Cattle , Creutzfeldt-Jakob Syndrome/genetics , Humans , Polymorphism, Genetic , Prion Diseases/classification , Prion Diseases/drug therapy , Prion Diseases/epidemiology , Prions/drug effects , Prions/genetics , Prions/metabolism , Scrapie/physiopathology , Sheep
15.
Cell Mol Neurobiol ; 31(7): 999-1008, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21516351

ABSTRACT

We first verified that a single chain Fv fragment against prion protein (anti-PrP scFv) was secreted by HEK293T cells and prevented prion replication in infected cells. We then stably expressed anti-PrP scFv in brain-engraftable murine microglial cells and intracerebrally injected these cells into mice before or after infection with prions. Interestingly, the injection before or at an early time point after infection attenuated the infection marginally but significantly prolonged survival times of the mice. These suggest that the ex vivo gene transfer of anti-PrP scFvs using brain-engraftable cells could be a possible immunotherapeutic approach against prion diseases.


Subject(s)
Brain/cytology , Microglia/physiology , Microglia/transplantation , Prions/immunology , Scrapie/physiopathology , Single-Chain Antibodies/immunology , Animals , Cell Line , Genetic Vectors , HEK293 Cells , Humans , Mice , Microglia/cytology , Prions/pathogenicity , Prions/physiology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Scrapie/therapy , Single-Chain Antibodies/genetics , Single-Chain Antibodies/therapeutic use , Survival Rate
16.
J Virol ; 85(4): 1484-94, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21123371

ABSTRACT

In nature prion diseases are usually transmitted by extracerebral prion infection, but clinical disease results only after invasion of the central nervous system (CNS). Prion protein (PrP), a host-encoded glycosylphosphatidylinositol (GPI)-anchored membrane glycoprotein, is necessary for prion infection and disease. Here, we investigated the role of the anchoring of PrP on prion neuroinvasion by studying various inoculation routes in mice expressing either anchored or anchorless PrP. In control mice with anchored PrP, intracerebral or sciatic nerve inoculation resulted in rapid CNS neuroinvasion and clinical disease (154 to 156 days), and after tongue, ocular, intravenous, or intraperitoneal inoculation, CNS neuroinvasion was only slightly slower (193 to 231 days). In contrast, in anchorless PrP mice, these routes resulted in slow and infrequent CNS neuroinvasion. Only intracerebral inoculation caused brain PrPres, a protease-resistant isoform of PrP, and disease in both types of mice. Thus, anchored PrP was an essential component for the rapid neural spread and CNS neuroinvasion of prion infection.


Subject(s)
Cell Membrane/metabolism , Central Nervous System/physiopathology , Prions/metabolism , Prions/pathogenicity , Scrapie/physiopathology , Animals , Brain/metabolism , Central Nervous System/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , PrPSc Proteins/metabolism , Prion Diseases/metabolism , Prion Diseases/physiopathology , Sciatic Nerve/metabolism , Scrapie/metabolism , Spinal Cord/metabolism , Tongue/metabolism
17.
Article in Chinese | MEDLINE | ID: mdl-21186515

ABSTRACT

UNLABELLED: OBJECTIVE To study the potential interaction between PrP protein. METHODS: The supernatant of health and scrapie-infected hamsters' brain homogenate was prepared, while various recombinant 14-3-3beta or PrP proteins were purified. The possible molecular interaction between 14-3-3beta proteins and PrP was tested by pull-down and immunoprecipitation assays. RESULTS: Both native PrP(c) and its protease-resistant isoform (PrP(Sc)) formed complexes with 14-3-3beta. The full-length recombinant 14-3-3beta proteins interacted with PrP. The domain responsible for interacting 14-3-3beta was located at N-terminal of 14-3-3beta (residues 1 to 38). CONCLUSION: The studies of the association of PrP with 14-3-3beta may further provide insight into a potential role of 14-3-3beta in the biological function of PrP and the pathogenesis of prion disease.


Subject(s)
14-3-3 Proteins/metabolism , PrPSc Proteins/metabolism , Prions/metabolism , Animals , Binding Sites , Brain Chemistry , Cricetinae , Endopeptidases/metabolism , Prion Diseases/pathology , Scrapie/physiopathology
18.
PLoS One ; 5(10)2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20957200

ABSTRACT

Fatal neurodegenerative prion diseases are caused by the transmissible PrP(Sc) prion agent whose initial replication after peripheral inoculation takes place in follicular dendritic cells present in germinal centers of lymphoid organs. However, prion replication also occurs in lymphoid cells. To assess the role of the hematopoietic compartment in neuroinvasion and prion replication, we generated chimeric mice, on a uniform congenic C57/BL6J background, by bone marrow replacement with hematopoietic cells expressing different levels of PrP protein. Nine different types of chimeric mice were inoculated intraperitoneally either with the lymphotropic Rocky Mountain Laboratory (RML) strain or the non lymphotropic ME-7 scrapie strain, at different doses. Here, we clearly demonstrate that overexpression of PrP by the hematopoietic system, or the lack of PrP expression by the bone marrow derived cells, does not change the incubation time period of the disease, even when the mice are infected at limiting doses. We conclude that the hematopoietic compartment is more or less permissive to prion replication, both for RML and ME-7, but does not play a role in neuroinvasion.


Subject(s)
Bone Marrow/physiopathology , Prions/physiology , Scrapie/physiopathology , Animals , Mice , Mice, Inbred C57BL , PrPC Proteins/metabolism
19.
Neuroscience ; 165(3): 662-74, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-19879926

ABSTRACT

We previously detailed how intrahippocampal inoculation of C57BL/6J mice with murine modified scrapie (ME7) leads to chronic neurodegeneration (Cunningham C, Deacon R, Wells H, Boche D, Waters S, Diniz CP, Scott H, Rawlins JN, Perry VH (2003) Eur J Neurosci 17:2147-2155.). Our characterization of the ME7-model is based on inoculation of this murine modified scrapie agent into C57BL/6J mice from Harlan laboratories. This agent in the C57BL/6J host generates a disease that spans a 24-week time course. The hippocampal pathology shows progressive misfolded prion (PrP(Sc)) deposition, astrogliosis and leads to behavioural dysfunction underpinned by the early synaptic loss that precedes neuronal death. The Harlan C57BL/6J, although widely used as a wild type mouse, are a sub-strain harbouring a spontaneous deletion of alpha-synuclein with the full description C57BL/6JOlaHsd. Recently alpha-synuclein has been shown to ameliorate the synaptic loss in a mouse model lacking the synaptic chaperone CSP-alpha. This opens a potential confound of the ME7-model, particularly with respect to the signature synaptic loss that underpin the physiological and behavioural dysfunction. To investigate if this strain-selective loss of a candidate disease modifier impacts on signature ME7 pathology, we compared cohorts of C57BL/6JOlaHsd (alpha-synuclein negative) with the founder strain from Charles Rivers (C57BL/6JCrl, alpha-synuclein positive). There were subtle changes in behaviour when comparing control animals from the two sub-strains indicating potentially significant consequences for studies assuming neurobiogical identity of both strains. However, there was no evidence that the absence of alpha-synuclein modifies disease. Indeed, accumulation of PrP(Sc), synaptic loss and the behavioural dysfunction associated with the ME7-agent was the same in both genetic backgrounds. Our data suggest that alpha-synuclein deficiency does not contribute to the compartment specific processes that give rise to prion disease mediated synaptotoxicity and neurodegeneration.


Subject(s)
Disease Progression , Scrapie/physiopathology , alpha-Synuclein/deficiency , Animals , Behavior, Animal/physiology , Cohort Studies , Disease Models, Animal , Female , Hippocampus/pathology , Hippocampus/physiopathology , Mice , Mice, Inbred C57BL , PrPSc Proteins/metabolism , Random Allocation , Scrapie/pathology , Species Specificity , Synapses/pathology , Time Factors , alpha-Synuclein/metabolism
20.
J Anim Sci ; 88(2): 428-34, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19897640

ABSTRACT

The association of the prion protein (PrP) gene with susceptibility to scrapie has formed the basis of selection programs aimed at eradicating the disease from sheep populations. Animals are genotyped for the PrP gene and those with the less susceptible genotypes are selected. The objectives of this study were to determine the effectiveness of predicting PrP genotypes by using information from relatives and to investigate the association of the PrP genotype with lamb performance traits in Suffolk sheep. Data were obtained from a scrapie-affected flock maintained in Scotland. A total of 643 were animals genotyped at codon 171 of the PrP gene with 2 alleles, R and Q. The genotypes of these animals were used to predict the genotypes of 5,173 nongenotyped animals in the same flock using segregation analysis. The genotype of nongenotyped animals was predicted from the probabilities for each possible genotype; further, an overall index for each animal was calculated to reflect the accuracy of prediction. Association analyses of the PrP gene (using animals with both known and inferred genotypes) with BW at birth, at weaning (56 d), and at 150 d, and for backfat and muscle depths at 150 d of age were carried out. A linear mixed model with random direct and maternal additive genetic effects, maternal permanent and temporary environmental effects, and year of birth was tested, and the most appropriate model was used for each trait. The expected number of Q alleles carried (from 0 to 2) by each animal was calculated and used in the model as a linear and quadratic covariate to test for associations with possible additive and dominance PrP gene effects, respectively. Results showed that the genotypes of relatively few animals (235) were inferred with certainty (compared with the 5,173 nongenotyped animals). Approximately 25% of the 5,173 predicted genotypes were inferred with a genotype probability index of 50% and greater. There was no significant association of the PrP gene with any of the performance traits studied (there were no significant additive or dominance effects). Such was the case whether data on animals with known or with both known and predicted genotypes were considered. It can be concluded that selection for PrP-resistant alleles in Suffolk sheep is unlikely to affect performance directly.


Subject(s)
Prions/genetics , Scrapie/genetics , Sheep/growth & development , Alleles , Animals , Animals, Newborn/growth & development , Female , Genes/genetics , Genetic Association Studies/veterinary , Genetic Predisposition to Disease , Genotype , Male , Phenotype , Scrapie/physiopathology , Sheep/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...