Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Acta Neuropathol Commun ; 9(1): 145, 2021 08 28.
Article in English | MEDLINE | ID: mdl-34454616

ABSTRACT

Treatment with human pituitary-derived growth hormone (hGH) was responsible for a significant proportion of iatrogenic Creutzfeldt-Jakob disease (iCJD) cases. France and the UK experienced the largest case numbers of hGH-iCJD, with 122 and 81 cases respectively. Differences in the frequency of the three PRNP codon 129 polymorphisms (MM, MV and VV) and the estimated incubation periods associated with each of these genotypes in the French and the UK hGH-iCJD cohorts led to the suggestion that the prion strains responsible for these two hGH-iCJD cohorts were different. In this study, we characterized the prion strains responsible for hGH-iCJD cases originating from UK (n = 11) and France (n = 11) using human PrP expressing mouse models. The cases included PRNP MM, MV and VV genotypes from both countries. UK and French sporadic CJD (sCJD) cases were included as controls. The prion strains identified following inoculation with hGH-iCJD homogenates corresponded to the two most frequently observed sCJD prion strains (M1CJD and V2CJD). However, in clear contradiction to the initial hypothesis, the prion strains that were identified in the UK and the French hGH-iCJD cases were not radically different. In the vast majority of the cases originating from both countries, the V2CJD strain or a mixture of M1CJD + V2CJD strains were identified. These data strongly support the contention that the differences in the epidemiological and genetic profiles observed in the UK and France hGH-iCJD cohorts cannot be attributed only to the transmission of different prion strains.


Subject(s)
Creutzfeldt-Jakob Syndrome/epidemiology , Creutzfeldt-Jakob Syndrome/pathology , Encephalopathy, Bovine Spongiform/epidemiology , Encephalopathy, Bovine Spongiform/pathology , Human Growth Hormone/adverse effects , PrPSc Proteins/adverse effects , Adult , Animals , Cohort Studies , Creutzfeldt-Jakob Syndrome/transmission , Encephalopathy, Bovine Spongiform/transmission , Female , France/epidemiology , Human Growth Hormone/administration & dosage , Humans , Male , Mice , Mice, Transgenic , Middle Aged , PrPSc Proteins/administration & dosage , PrPSc Proteins/isolation & purification , United Kingdom/epidemiology
2.
Elife ; 102021 04 14.
Article in English | MEDLINE | ID: mdl-33851575

ABSTRACT

Background: Fatal Familial Insomnia (FFI) is a genetic prion disease caused by the D178N mutation in the prion protein gene (PRNP) in coupling phase with methionine at PRNP 129. In 2017, we have shown that the olfactory mucosa (OM) collected from FFI patients contained traces of PrPSc detectable by Protein Misfolding Cyclic Amplification (PMCA). Methods: In this work, we have challenged PMCA-generated products obtained from OM and brain homogenate of FFI patients in BvPrP-Tg407 transgenic mice expressing the bank vole prion protein to test their ability to induce prion pathology. Results: All inoculated mice developed mild spongiform changes, astroglial activation, and PrPSc deposition mainly affecting the thalamus. However, their neuropathological alterations were different from those found in the brain of BvPrP-Tg407 mice injected with raw FFI brain homogenate. Conclusions: Although with some experimental constraints, we show that PrPSc present in OM of FFI patients is potentially infectious. Funding: This work was supported in part by the Italian Ministry of Health (GR-2013-02355724 and Ricerca Corrente), MJFF, ALZ, Alzheimer's Research UK and the Weston Brain Institute (BAND2015), and Euronanomed III (SPEEDY) to FM; by the Spanish Ministerio de Economía y Competitividad (grant AGL2016-78054-R [AEI/FEDER, UE]) to JMT and JCE; AM-M was supported by a fellowship from the INIA (FPI-SGIT-2015-02).


Subject(s)
Insomnia, Fatal Familial/etiology , Olfactory Mucosa/chemistry , PrPSc Proteins/administration & dosage , Animals , Humans , Mice , Mice, Transgenic
3.
Acta Neuropathol Commun ; 9(1): 17, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33509294

ABSTRACT

Accumulation of misfolded host proteins is central to neuropathogenesis of numerous human brain diseases including prion and prion-like diseases. Neurons of retina are also affected by these diseases. Previously, our group and others found that prion-induced retinal damage to photoreceptor cells in mice and humans resembled pathology of human retinitis pigmentosa caused by mutations in retinal proteins. Here, using confocal, epifluorescent and electron microscopy we followed deposition of disease-associated prion protein (PrPSc) and its association with damage to critical retinal structures following intracerebral prion inoculation. The earliest time and place of retinal PrPSc deposition was 67 days post-inoculation (dpi) on the inner segment (IS) of cone photoreceptors. At 104 and 118 dpi, PrPSc was associated with the base of cilia and swollen cone inner segments, suggesting ciliopathy as a pathogenic mechanism. By 118 dpi, PrPSc was deposited in both rods and cones which showed rootlet damage in the IS, and photoreceptor cell death was indicated by thinning of the outer nuclear layer. In the outer plexiform layer (OPL) in uninfected mice, normal host PrP (PrPC) was mainly associated with cone bipolar cell processes, but in infected mice, at 118 dpi, PrPSc was detected on cone and rod bipolar cell dendrites extending into ribbon synapses. Loss of ribbon synapses in cone pedicles and rod spherules in the OPL was observed to precede destruction of most rods and cones over the next 2-3 weeks. However, bipolar cells and horizontal cells were less damaged, indicating high selectivity among neurons for injury by prions. PrPSc deposition in cone and rod inner segments and on the bipolar cell processes participating in ribbon synapses appear to be critical early events leading to damage and death of photoreceptors after prion infection. These mechanisms may also occur in human retinitis pigmentosa and prion-like diseases, such as AD.


Subject(s)
Photoreceptor Connecting Cilium/metabolism , PrPSc Proteins/metabolism , Retinal Bipolar Cells/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Photoreceptor Cell Inner Segment/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Cell Death , Disease Progression , Mice , Microscopy, Confocal , Microscopy, Electron , Microscopy, Fluorescence , Photoreceptor Connecting Cilium/pathology , Photoreceptor Connecting Cilium/ultrastructure , PrPSc Proteins/administration & dosage , Retinal Bipolar Cells/pathology , Retinal Bipolar Cells/ultrastructure , Retinal Cone Photoreceptor Cells/pathology , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Photoreceptor Cell Inner Segment/pathology , Retinal Photoreceptor Cell Inner Segment/ultrastructure , Retinal Photoreceptor Cell Outer Segment/pathology , Retinal Photoreceptor Cell Outer Segment/ultrastructure , Retinal Rod Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/ultrastructure , Scrapie/metabolism , Scrapie/pathology
4.
PLoS One ; 15(12): e0243009, 2020.
Article in English | MEDLINE | ID: mdl-33270721

ABSTRACT

Scrapie, a prion disease of sheep, is highly resistant to conventional deactivation. Numerous methods to deactivate scrapie have been tested in laboratory animal models, and adequate autoclave treatment can reduce or remove the infectivity of some classical scrapie strains depending on the heating parameters used. In this study, we autoclaved brain homogenate from a sheep with US scrapie strain 13-7 for 30 minutes at 121°C. Genetically susceptible VRQ/ARQ sheep were orally inoculated with 3 grams of the autoclaved brain homogenate. For comparison, a second group of sheep was inoculated with a non-autoclaved brain homogenate. Rectal biopsies were used to assess antemortem scrapie disease progression throughout the study. Five out of ten (5/10) sheep that received autoclaved inoculum ultimately developed scrapie after an experimental endpoint of 72 months. These sheep had a mean incubation period of 26.99 months. Two out of five (2/5) positive sheep had detectable PrPSc in antemortem rectal biopsies, and two (2/5) other sheep had PrPSc in postmortem rectal tissue. A single sheep (1/5) was positive for scrapie in the CNS, small intestine, and retropharyngeal lymph node but had negative rectal tissue. All of the sheep (10/10) that received non-autoclaved inoculum developed scrapie with a mean incubation period of 20.2 months and had positive rectal biopsies at the earliest timepoint (14.7 months post-inoculation). These results demonstrate that sheep are orally susceptible to US derived classical scrapie strain 13-7 after autoclave treatment at 121°C for 30 minutes. Differences in incubation periods and time interval to first positive rectal biopsies indicate a partial reduction in infectivity titers for the autoclaved inoculum group.


Subject(s)
PrPSc Proteins/administration & dosage , Scrapie/transmission , Sterilization/methods , Administration, Oral , Animals , Brain/metabolism , Brain/pathology , Infectious Disease Incubation Period , PrPSc Proteins/chemistry , PrPSc Proteins/pathogenicity , Scrapie/mortality , Scrapie/pathology , Sheep/genetics
5.
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
6.
mBio ; 6(5): e01419-15, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26396245

ABSTRACT

UNLABELLED: Aggregation of misfolded host proteins in the central nervous system is believed to be important in the pathogenic process in several neurodegenerative diseases of humans, including prion diseases, Alzheimer's disease, and Parkinson's disease. In these diseases, protein misfolding and aggregation appear to expand through a process of seeded polymerization. Prion diseases occur in both humans and animals and are experimentally transmissible orally or by injection, thus providing a controllable model of other neurodegenerative protein misfolding diseases. In rodents and ruminants, prion disease has a slow course, lasting months to years. Although prion infectivity has been detected in brain tissue at 3 to 4 weeks postinfection (p.i.), the details of early prion replication in the brain are not well understood. Here we studied the localization and quantitation of PrPSc generation in vivo starting at 30 min postmicroinjection of scrapie into the brain. In C57BL mice at 3 days p.i., generation of new PrPSc was detected by immunohistochemistry and immunoblot assays, and at 7 days p.i., new generation was confirmed by real-time quaking-induced conversion assay. The main site of new PrPSc generation was near the outer basement membrane of small and medium blood vessels. The finding and localization of replication at this site so early after injection have not been reported previously. This predominantly perivascular location suggested that structural components of the blood vessel basement membrane or perivascular astrocytes might act as cofactors in the initial generation of PrPSc. The location of PrPSc replication at the basement membrane also implies a role for the brain interstitial fluid drainage in the early infection process. IMPORTANCE: Neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and prion diseases, of humans are characterized by misfolding and aggregation of certain proteins, resulting in the destruction of brain tissue. In these diseases, the damage process spreads progressively within the central nervous system, but only prion diseases are known to be transmissible between individuals. Here we used microinjection of infectious prion protein (PrPSc) into the mouse brain to model early events of iatrogenic prion transmission via surgical instruments or tissue grafts. At 3 and 7 days postinjection, we detected the generation of new PrPSc, mostly on the outer walls of blood vessels near the injection site. This location and very early replication were surprising and unique. Perivascular prion replication suggested the transport of injected PrPSc via brain interstitial fluid to the basement membranes of blood vessels, where interactions with possible cofactors made by astrocytes or endothelia might facilitate the earliest cycles of prion infection.


Subject(s)
Blood Vessels/pathology , PrPSc Proteins/administration & dosage , PrPSc Proteins/analysis , Scrapie/pathology , Animals , Chemistry Techniques, Analytical , Disease Models, Animal , Immunoblotting , Immunohistochemistry , Mice, Inbred C57BL , Microinjections , Time Factors
7.
Hybridoma (Larchmt) ; 31(5): 314-24, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23098297

ABSTRACT

Prions, or infectious proteins, cause a class of uniformly fatal neurodegenerative diseases. Prions are composed solely of an aberrantly folded isoform (PrP(Sc)) of a normal cellular protein (PrP(C)). Shared sequence identity of PrP(Sc) with PrP(C) has limited the detection sensitivity of immunochemical assays, as antibodies specific for the disease-causing PrP(Sc) isoform have not been developed. Here we report the generation of three new monoclonal antibodies (MAbs) to PrP, which were isolated following immunization of Prnp(0/0) Balb/cJ mice with highly purified PrP(Sc) isolated from brain lipid rafts. Epitope mapping using synthetic PrP peptides revealed that the three MAbs bind different epitopes of PrP. The DRM1-31 MAb has a conformational epitope at the proposed binding site for the putative prion conversion co-factor "protein X." The DRM1-60 MAb binds a single linear epitope localized to the ß2-α2 loop region of PrP, whereas DRM2-118 binds an epitope that includes sequences within the octarepeat region and near the site of N-terminal truncation of PrP(Sc) by proteinase K. Our novel anti-PrP MAbs with defined PrP epitopes may be useful in deciphering the conformational conversion of PrP(C) into PrP(Sc).


Subject(s)
Antibodies, Monoclonal/chemistry , Epitope Mapping , Epitopes/chemistry , Peptides/chemical synthesis , PrPSc Proteins/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , Binding Sites, Antibody , Blotting, Western , Brain Chemistry , Cricetinae , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Humans , Immunization , Membrane Microdomains/chemistry , Mesocricetus , Mice , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Peptides/immunology , PrPSc Proteins/administration & dosage , PrPSc Proteins/immunology , PrPSc Proteins/isolation & purification , Protein Structure, Secondary
8.
Jpn J Infect Dis ; 65(1): 37-44, 2012.
Article in English | MEDLINE | ID: mdl-22274156

ABSTRACT

The pathologic disease-associated prion protein (PrP(Sc)) has been shown to be expressed in the central nervous system of Holstein cattle inoculated intracerebrally with 3 sources of classical bovine spongiform encephalopathy (BSE) isolates. Several regions of the brain and spinal cord were analyzed for PrP(Sc) expression by immunohistochemical and Western blotting analyses. Animals euthanized at 10 months post-inoculation (mpi) showed PrP(Sc) deposits in the brainstem and thalamus, but no vacuolation; this suggested that the BSE agent might exhibit area-dependent tropism in the brain. At 16 and 18 mpi, a small amount of vacuolation was detected in the brainstem and thalamus, but not in the cerebral cortices. At 20 to 24 mpi, when clinical symptoms were apparent, heavy PrP(Sc) deposits were evident throughout the brain and spinal cord. The mean time to the appearance of clinical symptoms was 19.7 mpi, and the mean survival time was 22.7 mpi. These findings show that PrP(Sc) accumulation was detected approximately 10 months before the clinical symptoms of BSE became apparent. In addition, the 3 sources of BSE prion induced no detectable differences in the clinical signs, incubation periods, neuroanatomical location of vacuoles, or distribution and pattern of PrP(Sc) depositions in the brain.


Subject(s)
Brain Stem/pathology , Encephalopathy, Bovine Spongiform/pathology , PrPSc Proteins/metabolism , Spinal Cord/pathology , Animals , Blotting, Western , Brain Stem/metabolism , Cattle , Encephalopathy, Bovine Spongiform/metabolism , Female , Immunohistochemistry , PrPSc Proteins/administration & dosage , PrPSc Proteins/analysis , Spinal Cord/metabolism , Thalamus/metabolism , Thalamus/pathology , Time Factors , Vacuoles/metabolism , Vacuoles/pathology
9.
Vet Res ; 42: 107, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21988781

ABSTRACT

Interspecies transmission studies afford the opportunity to better understand the potential host range and origins of prion diseases. The purpose of this experiment was to determine susceptibility of white-tailed deer to the agent of scrapie after intracerebral inoculation and to compare clinical signs and lesions to those reported for chronic wasting disease (CWD). Deer (n = 5) were inoculated with 1 mL of a 10% (wt/vol) brain homogenate derived from a sheep clinically affected with scrapie. A non-inoculated deer was maintained as a negative control. Deer were observed daily for clinical signs of disease and euthanized and necropsied when unequivocal signs of scrapie were noted. One animal died 7 months post inoculation (pi) due to intercurrent disease. Examinations of brain tissue for the presence of the disease-associated abnormal prion protein (PrP(Sc)) by western blot (WB) and immunohistochemistry (IHC) were negative whereas IHC of lymphoid tissues was positive. Deer necropsied at 15-22 months pi were positive for scrapie by IHC and WB. Deer necropsied after 20 months pi had clinical signs of depression and progressive weight loss. Tissues with PrP(Sc) immunoreactivity included brain (at levels of cerebrum, hippocampus, colliculus, cerebellum, and brainstem), trigeminal ganglion, neurohypophysis, retina, spinal cord, and various lymphoid tissues including tonsil, retropharyngeal and mesenteric lymph nodes, Peyer's patches, and spleen. This work demonstrates for the first time that white-tailed deer are susceptible to sheep scrapie by intracerebral inoculation. To further test the susceptibility of white-tailed deer to scrapie these experiments will be repeated with a more natural route of inoculation.


Subject(s)
Brain/pathology , Deer , PrPSc Proteins/metabolism , Scrapie/pathology , Wasting Disease, Chronic/pathology , Animals , Blotting, Western/veterinary , Disease Susceptibility/pathology , PrPSc Proteins/administration & dosage , Scrapie/etiology , Wasting Disease, Chronic/etiology
10.
FASEB J ; 25(8): 2792-803, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21555356

ABSTRACT

Prion diseases are infectious neurodegenerative disorders associated with the misfolded prion protein (PrP(Sc)), which appears to be the sole component of the infectious agent (termed prion). To produce disease, prions have to be absorbed into the body and reach sufficient quantities in the brain. Very little is known about the biological mechanisms controlling the initial fate of prions. Here, we studied the systemic pharmacokinetics and biodistribution of PrP(Sc) in vivo. After an intravenous injection of highly purified radiolabeled or native unlabeled PrP(Sc), the protein was eliminated rapidly from the serum (half-life of 3.24 h), mostly through tissue uptake. The quantity of intact PrP(Sc) reaching the brain was ∼ 0.2% of the injected dose per gram of brain tissue (ID/g). The highest levels were found in liver (∼ 20% ID/g), spleen (∼ 13% ID/g), and kidney (∼ 7.4% ID/g). Cell surface PrP(C) does not appear to play a role in PrP(Sc) pharmacokinetics, since the infectious protein distributed similarly in wild-type and PrP-null mice. To measure tissue uptake kinetics and biodistribution accurately, vascular space in tissues was measured with radioactively labeled albumin coinjected with radioactively labeled PrP(Sc). Our results provide a fundamental pharmacokinetic characterization of PrP(Sc) in vivo, which may be relevant to estimate tissue risks and mechanisms of prion neuroinvasion and to identify novel therapeutic strategies.


Subject(s)
PrPSc Proteins/metabolism , Prion Diseases/metabolism , Animals , Biological Transport, Active , Brain/metabolism , Half-Life , Injections, Intravenous , Metabolic Clearance Rate , Mice , PrPSc Proteins/administration & dosage , PrPSc Proteins/blood , PrPSc Proteins/pharmacokinetics , Prion Diseases/blood , Protein Stability , Scrapie/metabolism , Tissue Distribution
11.
Methods ; 53(3): 208-13, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21176786

ABSTRACT

The infectivity associated with prion disease sets it apart from a large group of late-onset neurodegenerative disorders that shares the characteristics of protein aggregation and neurodegeneration. The unconventional infectious agent, PrP(Sc), is an aberrantly folded form of the normal prion protein (PrP(C)) and the PrP(C)-to-PrP(Sc) conversion is a critical pathogenic step in prion disease. Using the Protein Misfolding Cyclic Amplification technique, we converted folded bacterially expressed recombinant PrP into a proteinase K-resistant and aggregated conformation (rPrP-res) in the presence of anionic lipid and RNA molecules. Moreover, high prion infectivity was demonstrated by intracerebral inoculation of rPrP-res into wild-type mice, which caused prion disease with a short incubation period. The establishment of the in vitro recombinant PrP conversion assay makes it feasible for us to explore the molecular basis behind the intriguing properties associated with prion infectivity.


Subject(s)
Escherichia coli/metabolism , PrPC Proteins/chemistry , PrPSc Proteins/chemistry , Recombinant Proteins/chemistry , Animals , Brain/pathology , Cloning, Molecular/methods , Disease Models, Animal , Endopeptidase K/metabolism , Female , Mice , Phosphatidylglycerols/chemistry , PrPC Proteins/administration & dosage , PrPC Proteins/biosynthesis , PrPSc Proteins/administration & dosage , PrPSc Proteins/biosynthesis , Prion Diseases/pathology , Protein Folding , RNA/chemistry , Recombinant Proteins/administration & dosage , Recombinant Proteins/biosynthesis , Sonication
12.
Int J Mol Med ; 26(6): 845-51, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21042778

ABSTRACT

Transmissible spongiform encephalopathies (TSE) are caused by dietary oral exposure to infectious prion proteins (PrPSc); however, the mechanism behind the uptake of PrPSc in the intestines is poorly understood. In addition, epidemiological studies of BSE showed that most cattle are exposed to the agents in the first 6 months of life, during the suckling and weaning periods. In the present study, to elucidate the enteric invasion mechanism of prions and to investigate the age-dependent transmission mechanism suggested by epidemiological studies, wild-type and SCID mice were orally administered brain homogenate from scrapie (Tsukuba 1)-infected mice during the suckling and weaning stages, before being analyzed histopathologically. PrPSc was found to be incorporated into the villous columnar epithelial cells and was also detected in the villous lacteal of 15-day-old suckling mice. However, no such uptake of PrPSc was observed in the weaned mice at 25-days-old. Four different strains of mice were tested. There was no mouse strain difference in the frequency of PrPSc positive columnar epithelial cells. In addition, the uptake of PrPSc in suckling SCID mice lacking maternal antibodies was significantly lower than that in the wild-type suckling mice, and the uptake of PrPSc was enhanced by dilution with purified IgG. In the present study, it was suggested that the weaning period and maternal immunoglobulin are important risk factors for the oral transmission of PrPSc.


Subject(s)
Immunoglobulins/metabolism , Intestinal Mucosa/metabolism , PrPSc Proteins/metabolism , Prion Diseases/metabolism , Administration, Oral , Age Factors , Animals , Animals, Suckling , Brain Chemistry , Cattle , Disease Transmission, Infectious , Host-Pathogen Interactions , Immunoglobulins/immunology , Immunohistochemistry , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , PrPSc Proteins/administration & dosage , PrPSc Proteins/immunology , Prion Diseases/etiology , Prion Diseases/immunology
13.
J Immunol ; 183(8): 5199-207, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19786551

ABSTRACT

Following peripheral exposure, many transmissible spongiform encephalopathy (TSE) agents accumulate first in lymphoid tissues before spreading to the CNS (termed neuroinvasion) where they cause neurodegeneration. Early TSE agent accumulation upon follicular dendritic cells (FDCs) in lymphoid follicles appears critical for efficient neuroinvasion. Most clinical cases of variant Creutzfeldt-Jakob disease have occurred in young adults, although the reasons behind this apparent age-related susceptibility are uncertain. Host age has a significant influence on immune function. As FDC status and immune complex trapping is reduced in aged mice (600 days old), we hypothesized that this aging-related decline in FDC function might impair TSE pathogenesis. We show that coincident with the effects of host age on FDC status, the early TSE agent accumulation in the spleens of aged mice was significantly impaired. Furthermore, following peripheral exposure, none of the aged mice developed clinical TSE disease during their lifespans, although most mice displayed histopathological signs of TSE disease in their brains. Our data imply that the reduced status of FDCs in aged mice significantly impairs the early TSE agent accumulation in lymphoid tissues and subsequent neuroinvasion. Furthermore, the inefficient neuroinvasion in aged individuals may lead to significant levels of subclinical TSE disease in the population.


Subject(s)
Aging/physiology , Brain/immunology , Dendritic Cells, Follicular/immunology , PrPSc Proteins/pathogenicity , Prion Diseases/immunology , Sympathetic Nervous System/immunology , Administration, Oral , Age Factors , Animals , Brain/pathology , Complement C4/immunology , Complement C4/metabolism , Dendritic Cells, Follicular/metabolism , Dendritic Cells, Follicular/pathology , Disease Models, Animal , Disease Susceptibility , Mice , Mice, Inbred C57BL , PrPSc Proteins/administration & dosage , Prion Diseases/pathology , Spleen/immunology , Spleen/innervation , Spleen/pathology , Sympathetic Nervous System/pathology
14.
Protein Pept Lett ; 16(3): 247-55, 2009.
Article in English | MEDLINE | ID: mdl-19275737

ABSTRACT

Transmissible spongiform encephalopathies (TSEs) are characterized by the accumulation of a protease-resistant abnormal isoform of the prion protein (PrPSc), which is converted from the cellular isoform of the prion protein (PrPC). In the oral transmission of prion protein, PrPSc can invade a host body through the intestinal tract. There is only limited information available on how the infectious agent passes through one or several biological barriers before it can finally reach the brain. After oral administration, PrPSc withstands the digestive process and may be incorporated by microfold (M) cells or villous columnar epithelial cells in the intestine. After entry into the intestinal epithelium, PrPSc accumulates and is amplified in follicular dendritic cells (FDCs) within Peyer's patches and other isolated lymphoid follicles possibly by an interaction with dendritic cells or macrophages. Following accumulation in gut-associated lymphoid tissues, PrPSc is thought to move to the enteric nervous systems (ENS) by an interaction with FDCs or dendritic cells. As a result of neuroinvasion into the ENS, PrPSc spreads to the central nervous system. In addition, an epidemiological study suggested that most bovine spongiform encephalopathy cases had been exposed to the agent in the first 6 months of life. Developments of the intestinal defense and immune system may be involved in the susceptibility to infection.


Subject(s)
Intestinal Mucosa/metabolism , PrPSc Proteins/pharmacokinetics , Prion Diseases/transmission , Animals , Humans , Intestines/immunology , Intestines/pathology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , PrPSc Proteins/administration & dosage , Prion Diseases/epidemiology
15.
Acta Neuropathol ; 117(2): 175-84, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19107494

ABSTRACT

The olfactory system has been implicated in the pathogenesis of transmissible spongiform encephalopathies (TSEs). To examine this issue and identify the pattern of TSE agent spread after intranasal administration, we inoculated a high-infectious dose of neurotropic scrapie strain 263K into the nasal cavity of Syrian hamsters. All animals allowed to survive became symptomatic with a mean incubation period of 162.4 days. Analysis at different time points revealed deposition of the pathological prion protein (PrP(TSE)) in nasal-associated lymphoid tissues in the absence of brain involvement from 80 days post-infection (50% of the incubation period). Olfactory-related structures and brainstem nuclei were involved from 100 days post-inoculation (62% of the incubation period) when animals were still asymptomatic. Intriguingly, vagal or trigeminal nuclei were identified as early sites of PrP(TSE) deposition in some pre-symptomatic animals. These findings indicate that the 263K scrapie agent is unable to effectively spread from the olfactory neuroepithelium to the olfactory-related structures and that, after intranasal inoculation, neuroinvasion occurs through olfactory-unrelated pathways.


Subject(s)
Brain Chemistry , PrPSc Proteins/pathogenicity , Scrapie/metabolism , Scrapie/pathology , Administration, Intranasal , Animals , Brain/pathology , Cricetinae , Immunohistochemistry , Lymphoid Tissue/chemistry , Lymphoid Tissue/pathology , Mesocricetus , Nasal Cavity/chemistry , Neurons/chemistry , PrPSc Proteins/administration & dosage , PrPSc Proteins/analysis
16.
J Neurosci Res ; 86(12): 2753-62, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18478553

ABSTRACT

Prion diseases are fatal neurodegenerative disorders characterized by long incubation periods. To investigate whether concurrent diseases can modify the clinical outcome of prion-affected subjects, we tested the effect of viral infection on the binding and internalization of PrP(Sc), essential steps of prion propagation. To this effect, we added scrapie brain homogenate or purified PrP(Sc) to fibroblasts previously infected with minute virus of mice (MVM), a mouse parvovirus. We show here that the rate of incorporation of PrP(Sc) into MVM-infected cells was significantly higher than that observed for naïve cells. Immunostaining of cells and immunoblotting of subcellular fractions using antibodies recognizing PrP and LysoTracker, a lysosomal marker, revealed that in both control and MVM-infected cells the incorporated PrP(Sc) was associated mostly with lysosomes. Interestingly, flotation gradient analysis revealed that the majority of the PrP(Sc) internalized into MVM-infected cells shifted toward raft-containing low-density fractions. Concomitantly, the MVM-infected cells demonstrated increased levels of the glycosphingolipid GM1 (an essential raft lipid component) throughout the gradient and a shift in caveolin 1 (a raft protein marker) toward lighter membrane fractions compared with noninfected cells. Our results suggest that the effect of viral infection on membrane lipid composition may promote the incorporation of exogenous PrP(Sc) into rafts. Importantly, membrane rafts are believed to be the conversion site of PrP(C) to PrP(Sc); therefore, the association of exogenous PrP(Sc) with such membrane microdomains may facilitate prion infection.


Subject(s)
Cell Membrane/metabolism , Cell Membrane/virology , Membrane Lipids/metabolism , Minute Virus of Mice/physiology , PrPSc Proteins/metabolism , Animals , Brain/metabolism , Brain/virology , Cell Membrane/drug effects , Cells, Cultured , Cricetinae , Membrane Lipids/physiology , Mesocricetus , Mice , PrPSc Proteins/administration & dosage , Prion Diseases/metabolism , Prion Diseases/virology
17.
Can J Vet Res ; 72(1): 63-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18214164

ABSTRACT

Scrapie, a transmissible spongiform encephalopathy (TSE), is a naturally occurring fatal neurodegenerative disease of sheep and goats. This study documents survival periods, pathological findings, and the presence of abnormal prion protein (PrP(Sc)) in genetically susceptible sheep inoculated with scrapie agent. Suffolk lambs (AA/RR/QQ at codons 136, 154, and 171, respectively) aged 4 mo were injected by the intralingual (IL) or intracerebral (IC) route with an inoculum prepared from a pool of scrapie-affected US sheep brains. The animals were euthanized when advanced clinical signs of scrapie were observed. Spongiform lesions in the brain and PrPsc deposits in the central nervous system (CNS) and lymphoid tissues were detected by immunohistochemical and Western blot (WB) testing in all the sheep with clinical prion disease. The mean survival period was 18.3 mo for the sheep inoculated by the IL route and 17.6 mo for those inoculated by the IC route. Since the IC method is occasionally associated with anesthesia-induced complications, intracranial hematoma, and CNS infections, and the IL method is very efficient, it may be more humane to use the latter. However, before this method can be recommended for inoculation of TSE agents, research needs to show that other TSE agents can also transmit disease via the tongue.


Subject(s)
Brain/pathology , PrPSc Proteins/administration & dosage , Scrapie/pathology , Scrapie/transmission , Tongue/pathology , Administration, Oral , Animals , Female , Genetic Predisposition to Disease , Injections/veterinary , Male , PrPSc Proteins/isolation & purification , Scrapie/genetics , Scrapie/mortality , Sheep , Survival Analysis
18.
J Immunol ; 179(11): 7758-66, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18025222

ABSTRACT

Following oral exposure, some transmissible spongiform encephalopathy (TSE) agents accumulate first upon follicular dendritic cells (DCs) in the GALT. Studies in mice have shown that TSE agent accumulation in the GALT, in particular the Peyer's patches, is obligatory for the efficient transmission of disease to the brain. However, the mechanism through which TSE agents are initially conveyed from the gut lumen to the GALT is not known. Studies have implicated migratory hemopoietic DCs in this process, but direct demonstration of their involvement in vivo is lacking. In this study, we have investigated the contribution of CD11c(+) DCs in scrapie agent neuroinvasion through use of CD11c-diptheria toxin receptor-transgenic mice in which CD11c(+) DCs can be specifically and transiently depleted. Using two distinct scrapie agent strains (ME7 and 139A scrapie agents), we show that when CD11c(+) DCs were transiently depleted in the GALT and spleen before oral exposure, early agent accumulation in these tissues was blocked. In addition, CD11c(+) cell depletion reduced susceptibility to oral scrapie challenge indicating that TSE agent neuroinvasion from the GALT was impaired. In conclusion, these data demonstrate that migratory CD11c(+) DCs play a key role in the translocation of the scrapie agent from the gut lumen to the GALT from which neuroinvasion subsequently occurs.


Subject(s)
CD11c Antigen/immunology , Intestines/immunology , PrPSc Proteins/immunology , Administration, Oral , Animals , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/pathology , Immunity, Mucosal/drug effects , Immunity, Mucosal/immunology , Intestines/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peyer's Patches/immunology , Peyer's Patches/pathology , PrPSc Proteins/administration & dosage , Spleen/immunology , Spleen/pathology
19.
Biosci Biotechnol Biochem ; 71(10): 2565-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17928684

ABSTRACT

Prions, infectious agents causing transmissible spongiform encephalopathy, retain infectivity even after undergoing routine sterilization processes. We found that MSK103 protease, identified in our previous study, effectively reduces infectivity and the level of misfolded isoform of the prion protein in scrapie-infected brain homogenates in the presence of SDS. The treatment therefore can be applied to the decontamination of thermolabile instruments.


Subject(s)
Bacillus/enzymology , Peptide Hydrolases/pharmacology , PrPSc Proteins/metabolism , Sodium Dodecyl Sulfate/pharmacology , Sterilization/methods , Animals , Drug Synergism , Hydrogen-Ion Concentration , Injections, Intraventricular , Mice , Mice, Transgenic , Peptide Hydrolases/isolation & purification , PrPSc Proteins/administration & dosage , Protein Denaturation , Protein Isoforms , Temperature , Time Factors
20.
BMC Vet Res ; 3: 20, 2007 Aug 28.
Article in English | MEDLINE | ID: mdl-17725818

ABSTRACT

BACKGROUND: Active surveillance for transmissible spongiform encephalopathies in small ruminants has been an EU regulatory requirement since 2002. A number of European countries have subsequently reported cases of atypical scrapie, similar to previously published cases from Norway, which have pathological and molecular features distinct from classical scrapie. Most cases have occurred singly in flocks, associated with genotypes considered to be more resistant to classical disease. Experimental transmissibility of such isolates has been reported in certain ovinised transgenic mice, but has not previously been reported in the natural host. Information on the transmissibility of this agent is vital to ensuring that disease control measures are effective and proportionate. RESULTS: This report presents the successful experimental transmission, in 378 days, of atypical scrapie to a recipient sheep of homologous genotype with preservation of the pathological and molecular characteristics of the donor. This isolate also transmitted to ovinised transgenic mice (Tg338) with a murine phenotype indistinguishable from that of Nor 98. CONCLUSION: This result strengthens the opinion that these cases result from a distinct strain of scrapie agent, which is potentially transmissible in the natural host under field conditions.


Subject(s)
Brain/metabolism , Disease Outbreaks/veterinary , PrPSc Proteins/metabolism , Scrapie/transmission , Animals , Blotting, Western/veterinary , Brain/pathology , Europe/epidemiology , European Union , Genetic Predisposition to Disease , Genotype , Immunohistochemistry/veterinary , PrPSc Proteins/administration & dosage , Scrapie/epidemiology , Scrapie/genetics , Scrapie/metabolism , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...