Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Neurol ; 13: 826676, 2022.
Article in English | MEDLINE | ID: mdl-35309588

ABSTRACT

Several variants of the TANK-Binding Kinase 1 (TBK1) gene have been associated with frontotemporal dementia - amyotrophic lateral sclerosis (FTD-ALS) spectrum diseases. Corticobasal syndrome (CBS) is characterized by asymmetric limb rigidity, dystonia or myoclonus, in association with speech or limb apraxia, cortical sensory deficit, and/or alien limb. It can result from a variety of underlying pathologies and although typically sporadic, it has been occasionally associated with MAPT and GRN variants. We describe here the proband of a family with multiple occurrences of FTD-ALS spectrum disease who developed an isolated right-sided primary asymmetric akinetic-rigid syndrome and subsequent speech and cognitive dysfunction associated with contralateral anterior temporal lobe atrophy on MRI and corresponding hypometabolism by FDG-PET. Genetic testing revealed a novel Lys694del variant of the TBK1 gene and Type A TDP-43 pathology in a predominantly frontotemporal distribution contralateral to the affected side. To our knowledge this is the first report of CBS as the initial expression of a TBK1 variant. This case emphasizes the importance of considering TBK1 genetic screening in patients with CBS, as this may be an underrepresented population on the spectrum of genetic FTD-ALS.

2.
Semin Neurol ; 42(6): 742-751, 2022 12.
Article in English | MEDLINE | ID: mdl-36623535

ABSTRACT

Dementia is broadly defined by DSM-V as cognitive decline from a previous level that impacts the patient's functioning at work or play. This broad definition does not provide information about the underlying disease process, an aspect of clinical care that is of increasing importance, as therapeutic development inches closer to effective disease-modifying treatments. The most common neurodegenerative dementias include Alzheimer's disease, dementia with Lewy bodies, frontotemporal dementia, and Parkinson's disease dementia. Although rare, the prion diseases constitute an important group of dementias that should be routinely considered in the evaluation. Over the last two decades, advances in neuroimaging, biomarker development, and neurogenetics have not only led to a better understanding of the biology of these diseases, but they have improved our awareness of less common clinical subtypes of dementia. As such, to best define the disease process, the evaluation of a patient with cognitive decline requires attention to a myriad of disease aspects, such as the primary symptom at onset (memory, language, visual perception, praxis, etc.), the age at onset (younger or older than 65 years), the rate of disease progression (weeks to months or years), the cognitive and behavioral profile (neuropsychological assessment), and involvement of physical findings. We present here three cases that highlight the decision-making process in the evaluation of patients with atypical presentations of dementia.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Frontotemporal Dementia , Parkinson Disease , Prion Diseases , Humans , Aged , Parkinson Disease/diagnosis , Alzheimer Disease/diagnosis , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/etiology
3.
Acta Neuropathol Commun ; 9(1): 158, 2021 09 26.
Article in English | MEDLINE | ID: mdl-34565488

ABSTRACT

Chronic wasting disease (CWD) is a cervid prion disease caused by the accumulation of an infectious misfolded conformer (PrPSc) of cellular prion protein (PrPC). It has been spreading rapidly in North America and also found in Asia and Europe. Although bovine spongiform encephalopathy (i.e. mad cow disease) is the only animal prion disease known to be zoonotic, the transmissibility of CWD to humans remains uncertain. Here we report the generation of the first CWD-derived infectious human PrPSc by elk CWD PrPSc-seeded conversion of PrPC in normal human brain homogenates using in vitro protein misfolding cyclic amplification (PMCA). Western blotting with human PrP selective antibody confirmed that the PMCA-generated protease-resistant PrPSc was derived from the human PrPC substrate. Two lines of humanized transgenic mice expressing human PrP with either Val or Met at the polymorphic codon 129 developed clinical prion disease following intracerebral inoculation with the PMCA-generated CWD-derived human PrPSc. Diseased mice exhibited distinct PrPSc patterns and neuropathological changes in the brain. Our study, using PMCA and animal bioassays, provides the first evidence that CWD PrPSc can cross the species barrier to convert human PrPC into infectious PrPSc that can produce bona fide prion disease when inoculated into humanized transgenic mice.


Subject(s)
Deer , PrPSc Proteins , Wasting Disease, Chronic , Zoonoses/pathology , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , PrPC Proteins
5.
Sci Rep ; 9(1): 8524, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31189938

ABSTRACT

To address the question of cross-talk between prion protein (PrP) and Alzheimer's disease (AD), we generated TgAD/GSS mice that develop amyloid-ß (Aß) plaques of AD and PrP (specifically mutated PrPA116V) plaques of Gerstmann-Sträussler-Scheinker disease (GSS) and compared plaque-related features in these mice to AD mice that express normal (TgAD), high (TgAD/HuPrP), or no (TgAD/PrP-/-) PrPC. In contrast to PrPC, PrPA116V weakly co-localized to Aß plaques, did not co-immunoprecipitate with Aß, and poorly bound to Aß in an ELISA-based binding assay. Despite the reduced association of PrPA116V with Aß, TgAD/GSS and TgAD/HuPrP mice that express comparable levels of PrPA116V and PrPC respectively, displayed similar increases in Aß plaque burden and steady state levels of Aß and its precursor APP compared with TgAD mice. Our Tg mouse lines also revealed a predominance of intracellular Aß plaques in mice lacking PrPC (TgAD/PrP-/-, TgAD/GSS) compared with an extracellular predominance in PrPC-expressing mice (TgAD, TgAD/HuPrP). Parallel studies in N2aAPPswe cells revealed a direct dependence on PrPC but not PrPA116V for exosome-related secretion of Aß. Overall, our findings are two-fold; they suggest that PrP expression augments Aß plaque production, at least in part by an indirect mechanism, perhaps by increasing steady state levels of APP, while they also provide support for a fundamental role of PrPC to bind to and deliver intraneuronal Aß to exosomes for secretion.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Gerstmann-Straussler-Scheinker Disease , Plaque, Amyloid , PrPC Proteins , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Gerstmann-Straussler-Scheinker Disease/genetics , Gerstmann-Straussler-Scheinker Disease/metabolism , Gerstmann-Straussler-Scheinker Disease/pathology , Mice , Mice, Transgenic , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , PrPC Proteins/genetics , PrPC Proteins/metabolism
6.
Mol Neurobiol ; 56(4): 2353-2361, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30027340

ABSTRACT

Our previous studies showed that intravenous immunoglobulin (IVIG) contained anti-Aß autoantibodies that might be able to treat Alzheimer's disease (AD). Recently, we identified and characterized naturally occurring autoantibodies against PrP from IVIG. Although autoantibodies in IVIG blocked PrP fibril formation and PrP neurotoxicity in vitro, it remained unknown whether IVIG could reduce amyloid plaque pathology in vivo and be used to effectively treat animals with prion diseases. In this study, we used Gerstmann-Sträussler-Scheinker (GSS)-Tg (PrP-A116V) transgenic mice to test IVIG efficacy since amyloid plaque formation played an important role in GSS pathogenesis. Here, we provided strong evidence that demonstrates how IVIG could significantly delay disease onset, elongate survival, and improve clinical phenotype in Tg (PrP-A116V) mice. Additionally, in treated animals, IVIG could markedly inhibit PrP amyloid plaque formation and attenuate neuronal apoptosis at the age of 120 days in mice. Our results indicate that IVIG may be a potential, effective therapeutic treatment for GSS and other prion diseases.


Subject(s)
Gerstmann-Straussler-Scheinker Disease/pathology , Immunoglobulins, Intravenous/pharmacology , Animals , Apoptosis/drug effects , Disease Models, Animal , Disease Progression , Kaplan-Meier Estimate , Mice, Transgenic , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/pathology , Phenotype , Plaque, Amyloid/pathology , Prion Proteins/metabolism
7.
Neurol Genet ; 4(4): e253, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30109268

ABSTRACT

OBJECTIVE: To describe the clinicopathologic, molecular, and transmissible characteristics of genetic prion disease in a young man carrying the PRNP-G114V variant. METHODS: We performed genetic, histologic, and molecular studies, combined with in vivo transmission studies and in vitro replication studies, to characterize this genetic prion disease. RESULTS: A 24-year-old American man of Polish descent developed progressive dementia, aphasia, and ataxia, leading to his death 5 years later. Histologic features included widespread spongiform degeneration, gliosis, and infrequent PrP plaque-like deposits within the cerebellum and putamen, best classifying this as a Creutzfeldt-Jakob disease (CJD) subtype. Molecular typing of proteinase K-resistant PrP (resPrPSc) revealed a mixture of type 1 (∼21 kDa) and type 2 (∼19 kDa) conformations with only 2, rather than the usual 3, PrPSc glycoforms. Brain homogenates from the proband failed to transmit prion disease to transgenic Tg(HuPrP) mice that overexpress human PrP and are typically susceptible to sporadic and genetic forms of CJD. When subjected to protein misfolding cyclic amplification, the PrPSc type 2 (∼19 kDa) was selectively amplified. CONCLUSIONS: The features of genetic CJDG114V suggest that residue 114 within the highly conserved palindromic region (113-AGAAAAGA-120) plays an important role in prion conformation and propagation.

8.
Int J Cell Biol ; 2013: 560421, 2013.
Article in English | MEDLINE | ID: mdl-24454378

ABSTRACT

Prion diseases are linked to the accumulation of a misfolded isoform (PrP(Sc)) of prion protein (PrP). Evidence suggests that lysosomes are degradation endpoints and sites of the accumulation of PrP(Sc). We questioned whether lysosomes participate in the early quality control of newly generated misfolded PrP. We found PrP carrying the disease-associated T182A mutation (Mut-PrP) was delivered to lysosomes in a Golgi-independent manner. Time-lapse live cell imaging revealed early formation and uptake of GFP-tagged Mut-PrP aggregates into LysoTracker labeled vesicles. Compared with Wt-PrP, Mut-PrP expression was associated with an elevation in several markers of the autophagy-lysosomal pathway, and it extensively colocalized with the autophagosome-specific marker, LC3B. In autophagy deficient (ATG5(-/-)) mouse embryonic fibroblasts, or in normal cells treated with the autophagy-inhibitor 3-MA, Mut-PrP colocalization with lysosomes was reduced to a similar extent. Additionally, 3-MA selectively impaired the degradation of insoluble Mut-PrP, resulting in an increase in protease-resistant PrP, whereas the induction of autophagy by rapamycin reduced it. These findings suggest that autophagy might function as a quality control mechanism to limit the accumulation of misfolded PrP that normally leads to the generation of PrP(Sc).

9.
J Neurosci ; 32(36): 12396-405, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22956830

ABSTRACT

Autophagy is a cell survival response to nutrient deprivation that delivers cellular components to lysosomes for digestion. In recent years, autophagy has also been shown to assist in the degradation of misfolded proteins linked to neurodegenerative disease (Ross and Poirier, 2004). In support of this, rapamycin, an autophagy inducer, improves the phenotype of several animal models of neurodegenerative disease. Our Tg(PrP-A116V) mice model Gerstmann-Sträussler-Scheinker disease (GSS), a genetic prion disease characterized by prominent ataxia and extracellular PrP amyloid plaque deposits in brain (Yang et al., 2009). To determine whether autophagy induction can mitigate the development of GSS, Tg(PrP-A116V) mice were chronically treated with 10 or 20 mg/kg rapamycin intraperitoneally thrice weekly, beginning at 6 weeks of age. We observed a dose-related delay in disease onset, a reduction in symptom severity, and an extension of survival in rapamycin-treated Tg(PrP-A116V) mice. Coincident with this response was an increase in the autophagy-specific marker LC3II, a reduction in insoluble PrP-A116V, and a near-complete absence of PrP amyloid plaques in the brain. An increase in glial cell apoptosis of unclear significance was also detected. These findings suggest autophagy induction enhances elimination of misfolded PrP before its accumulation in plaques. Because ataxia persisted in these mice despite the absence of plaque deposits, our findings also suggest that PrP plaque pathology, a histopathological marker for the diagnosis of GSS, is not essential for the GSS phenotype.


Subject(s)
Disease Models, Animal , Gerstmann-Straussler-Scheinker Disease/prevention & control , Plaque, Amyloid/prevention & control , Prions/antagonists & inhibitors , Sirolimus/therapeutic use , Animals , Female , Gerstmann-Straussler-Scheinker Disease/metabolism , Gerstmann-Straussler-Scheinker Disease/pathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Prions/metabolism , Random Allocation , Time Factors
10.
Ann Neurol ; 69(4): 712-20, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21416485

ABSTRACT

OBJECTIVE: To describe the Alzheimer disease (AD)-like clinical and pathological features, including marked neurofibrillary tangle (NFT) pathology, of a familial prion disease due to a rare nonsense mutation of the prion gene (PRNP). METHODS: Longitudinal clinical assessments were available for the proband and her mother. After death, both underwent neuropathological evaluation. PRNP was sequenced after failure to find immunopositive Aß deposits in the proband and the documentation of prion protein (PrP) immunopositive pathology. RESULTS: The proband presented at age 42 years with a 3-year history of progressive short-term memory impairment and depression. Neuropsychological testing found impaired memory performance, with relatively preserved attention and construction. She was diagnosed with AD and died at age 47 years. Neuropathologic evaluation revealed extensive limbic and neocortical NFT formation and neuritic plaques consistent with a Braak stage of VI. The NFTs were immunopositive, with multiple tau antibodies, and electron microscopy revealed paired helical filaments. However, the neuritic plaques were immunonegative for Aß, whereas immunostaining for PrP was positive. The mother of the proband had a similar presentation, including depression, and had been diagnosed clinically and pathologically as AD. Reevaluation of her brain tissue confirmed similar tau and PrP immunostaining findings. Genetic analysis revealed that both the proband and her mother had a rare PRNP mutation (Q160X) that resulted in the production of truncated PrP. INTERPRETATION: We suggest that PRNP mutations that result in a truncation of PrP lead to a prolonged clinical course consistent with a clinical diagnosis of AD and severe AD-like NFTs.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Codon, Nonsense , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/pathology , Prion Diseases/genetics , Prions/genetics , tau Proteins/genetics , Adult , Aged , Alzheimer Disease/diagnosis , Depressive Disorder, Major/genetics , Depressive Disorder, Major/pathology , Female , Glutamine , Humans , Memory Disorders/genetics , Memory Disorders/pathology , Memory, Short-Term , Middle Aged , Neuropsychological Tests , Phenotype , Prion Diseases/pathology , Prion Diseases/psychology , Prion Proteins , Prions/metabolism , Tyrosine , tau Proteins/metabolism
11.
J Biol Chem ; 286(9): 7490-5, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21209079

ABSTRACT

Prion diseases are infectious neurodegenerative disorders that affect humans and animals and that result from the conversion of normal prion protein (PrP(C)) into the misfolded prion protein (PrP(Sc)). Chronic wasting disease (CWD) is a prion disorder of increasing prevalence within the United States that affects a large population of wild and captive deer and elk. Determining the risk of transmission of CWD to humans is of utmost importance, considering that people can be infected by animal prions, resulting in new fatal diseases. To study the possibility that human PrP(C) can be converted into the misfolded form by CWD PrP(Sc), we performed experiments using the protein misfolding cyclic amplification technique, which mimics in vitro the process of prion replication. Our results show that cervid PrP(Sc) can induce the conversion of human PrP(C) but only after the CWD prion strain has been stabilized by successive passages in vitro or in vivo. Interestingly, the newly generated human PrP(Sc) exhibits a distinct biochemical pattern that differs from that of any of the currently known forms of human PrP(Sc). Our results also have profound implications for understanding the mechanisms of the prion species barrier and indicate that the transmission barrier is a dynamic process that depends on the strain and moreover the degree of adaptation of the strain. If our findings are corroborated by infectivity assays, they will imply that CWD prions have the potential to infect humans and that this ability progressively increases with CWD spreading.


Subject(s)
Deer , PrPSc Proteins/genetics , Wasting Disease, Chronic/genetics , Wasting Disease, Chronic/transmission , Zoonoses/transmission , Amyloidosis/epidemiology , Amyloidosis/genetics , Animals , Humans , In Vitro Techniques , Mice , Mice, Transgenic , PrPSc Proteins/metabolism , Risk Factors , Species Specificity , Wasting Disease, Chronic/epidemiology , Zoonoses/epidemiology
12.
J Geriatr Psychiatry Neurol ; 23(4): 277-98, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20938044

ABSTRACT

The prion diseases are a family of rare neurodegenerative disorders that result from the accumulation of a misfolded isoform of the prion protein (PrP), a normal constituent of the neuronal membrane. Five subtypes constitute the known human prion diseases; kuru, Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker syndrome (GSS), fatal insomnia (FI), and variant CJD (vCJD). These subtypes are distinguished, in part, by their clinical phenotype, but primarily by their associated brain histopathology. Evidence suggests these phenotypes are defined by differences in the pathogenic conformation of misfolded PrP. Although the vast majority of cases are sporadic, 10% to 15% result from an autosomal dominant mutation of the PrP gene (PRNP). General phenotype-genotype correlations can be made for the major subtypes of CJD, GSS, and FI. This paper will review some of the general background related to prion biology and detail the clinical and pathologic features of the major prion diseases, with a particular focus on the genetic aspects that result in prion disease or modification of its risk or phenotype.


Subject(s)
Brain/pathology , Prion Diseases/classification , Prion Diseases/genetics , Prion Diseases/pathology , Prions/genetics , Animals , Brain Stem/pathology , Cerebellum/pathology , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/pathology , Gerstmann-Straussler-Scheinker Disease/genetics , Gerstmann-Straussler-Scheinker Disease/pathology , Humans , Insomnia, Fatal Familial/genetics , Insomnia, Fatal Familial/pathology , Kuru/genetics , Kuru/pathology , Mutation , Phenotype , Prion Diseases/diagnosis , Prion Diseases/psychology , Prion Proteins , Risk Factors , Severity of Illness Index , Thalamus/pathology
13.
Genet Med ; 12(4): 187-95, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20216075

ABSTRACT

Prion diseases are a rare group of fatal neurodegenerative disorders of humans and animals that manifest primarily as progressive dementia and ataxia. Unique to these diseases is the prion, a misfolded isoform of the prion protein that can transmit disease from cell to cell or host to host by associating with, and transforming, normal prion protein into the misfolded isoform (the pathogenic scrapie-inducing form). Although the majority of cases occur on a sporadic basis, and rarely result from exposure to prions, such as mad cow disease, 10-15% are attributable to the presence of an autosomal dominant mutation of the prion protein gene (PRNP). Single base pair changes, or the insertion of one or more multiples of a 24 base pair repeat segment, make up the known sequence alterations of PRNP associated with genetic prion disease. The common polymorphic codon 129 of PRNP also plays an important and complex role in risk and phenotype of sporadic and genetic prion disease. This review will focus on the clinical and histopathologic features of the genetic prion diseases. Selected mutations will be highlighted as a way to illustrate general phenotype-genotype correlations.


Subject(s)
Mutation , Prion Diseases/genetics , Prions/genetics , Brain/metabolism , Brain/pathology , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Polymorphism, Genetic , Prion Diseases/classification , Prion Diseases/pathology , Prion Proteins
14.
J Biol Chem ; 285(12): 8967-75, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20086009

ABSTRACT

Prion diseases result from the accumulation of a misfolded isoform (PrP(Sc)) of the normal host prion protein (PrP(C)). PrP(Sc) propagates by templating its conformation onto resident PrP(C) to generate new PrP(Sc). Although the nature of the PrP(Sc)-PrP(C) complex is unresolved, certain segments or specific residues are thought to feature critically in its formation. The polymorphic residue 129 is one such site under considerable study. We combined transmission studies with a novel live cell yeast-based fluorescence resonance energy transfer (FRET) system that models the molecular association of PrP in a PrP(Sc)-like state, as a way to explore the role of residue 129 in this process. We show that a reduction in efficiency of prion transmission between donor PrP(Sc) and recipient PrP(C) that are mismatched at residue 129 correlates with a reduction in FRET between PrP-129M and PrP-129V in our yeast model. We further show that this effect depends on the different secondary structure propensities of Met and Val, rather than the specific amino acids. Finally, introduction of the disease-associated P101L mutation (mouse- equivalent) abolished FRET with wild-type mouse PrP, whereas mutant PrP-P101L displayed high FRET with homologous PrP-P101L, as long as residue 129 matched. These studies provide the first evidence for a physical alteration in the molecular association of PrP molecules differing in one or more residues, and they further predict that the different secondary structure propensities of Met and Val define the impaired association observed between PrP(Sc) and PrP(C) mismatched at residue 129.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , PrPC Proteins/chemistry , PrPSc Proteins/chemistry , Animals , Brain/metabolism , Cytosol/metabolism , Fungal Proteins/chemistry , Genotype , Mice , Mice, Transgenic , Protein Binding , Protein Conformation , Protein Folding , Protein Isoforms , Protein Structure, Secondary
15.
J Neurosci ; 29(32): 10072-80, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19675240

ABSTRACT

Gerstmann-Sträussler-Scheinker syndrome (GSS) is a genetic prion disease typified clinically by the development of progressive ataxia and dementia, and histopathologically by the presence of prion protein (PrP) amyloid plaques in the CNS, especially within the cerebellum. Several mutations of the PrP gene (PRNP) are associated with GSS, but only the P102L mutation has been convincingly modeled in transgenic (Tg) mice. To determine whether other mutations carry specific GSS phenotypic information, we constructed Tg mice that express PrP carrying the mouse homolog of the GSS-associated A117V mutation. Tg(A116V) mice express approximately six times the endogenous levels of PrP, develop progressive ataxia by approximately 140 d, and die by approximately 170 d. Compared with a mouse model of transmissible Creutzfeldt-Jakob disease (CJD), the ataxia of Tg(A116V) mice is more prominent, and the course of disease is more protracted, paralleling that observed in human disease. Neuropathology includes mild scattered vacuolation and prominent, mainly cerebellar localized, thioflavin S-positive PrP plaques comprised of full-length PrP(A116V). In some mice, more prominent vacuolation or a noncerebellar distribution of PrP plaques was evident, suggesting some variability in phenotype. The biophysical properties of PrP from Tg(A116V) mice and human GSS(A117V) revealed a similarly low fraction of insoluble PrP and a weakly protease-resistant approximately 13 kDa midspan PrP fragment, not observed in CJD. Overall, Tg(A116V) mice recapitulate many clinicopathologic features of GSS(A117V) that are distinct from CJD, supporting PrP(A116V) to carry specific phenotypic information. The occasional variation in histopathology they exhibit may shed light on a similar observation in human GSS(A117V).


Subject(s)
Disease Models, Animal , Gerstmann-Straussler-Scheinker Disease/genetics , Mutation, Missense , Prions/genetics , Animals , Ataxia/complications , Ataxia/genetics , Benzothiazoles , Blotting, Western , Brain/metabolism , Brain/pathology , Creutzfeldt-Jakob Syndrome/genetics , Gerstmann-Straussler-Scheinker Disease/complications , Gerstmann-Straussler-Scheinker Disease/mortality , Humans , Kaplan-Meier Estimate , Mice , Mice, Transgenic , Phenotype , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , Prion Proteins , Prions/metabolism , Thiazoles/metabolism , Time Factors
16.
J Biol Chem ; 283(49): 34021-8, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18826953

ABSTRACT

In prion disease, direct interaction between the cellular prion protein (PrP(C)) and its misfolded disease-associated conformer PrP(Sc) is a crucial, although poorly understood step promoting the formation of nascent PrP(Sc) and prion infectivity. Recently, we hypothesized that three regions of PrP (corresponding to amino acid residues 23-33, 98-110, and 136-158) interacting specifically and robustly with PrP(Sc), likely represent peptidic components of one flank of the prion replicative interface. In this study, we created epitope-tagged mouse PrP(C) molecules in which the PrP sequences 23-33, 98-110, and 136-158 were modified. These novel PrP molecules were individually expressed in the prion-infected neuroblastoma cell line (ScN2a) and the conversion of each mutated mouse PrP(C) substrate to PrP(Sc) compared with that of the epitope-tagged wild-type mouse PrP(C). Mutations within PrP 98-110, substituting all 4 wild-type lysine residues with alanine residues, prevented conversion to PrP(Sc). Furthermore, when residues within PrP 136-140 were collectively scrambled, changed to alanines, or amino acids at positions 136, 137, and 139 individually replaced by alanine, conversion to PrP(Sc) was similarly halted. However, other PrP molecules containing mutations within regions 23-33 and 101-104 were able to readily convert to PrP(Sc). These results suggest that PrP sequence comprising residues 98-110 and 136-140 not only participates in the specific binding interaction between PrP(C) and PrP(Sc), but also in the process leading to conversion of PrP(Sc)-sequestered PrP(C) into its disease-associated form.


Subject(s)
PrPC Proteins/chemistry , PrPSc Proteins/chemistry , Alanine/chemistry , Animals , Cell Line, Tumor , Epitopes/chemistry , Flow Cytometry , Mice , Mutation , Neuroblastoma/metabolism , Peptides/chemistry , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , Prions/chemistry , Protein Denaturation , Protein Folding , Transfection
17.
Ann Neurol ; 63(6): 697-708, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18571782

ABSTRACT

OBJECTIVE: To report a novel prion disease characterized by distinct histopathological and immunostaining features, and associated with an abnormal isoform of the prion protein (PrP) that, contrary to the common prion diseases, is predominantly sensitive to protease digestion. METHODS: Eleven subjects were investigated at the National Prion Disease Pathology Surveillance Center for clinical, histopathological, immunohistochemical, genotypical, and PrP characteristics. RESULTS: Patients presented with behavioral and psychiatric manifestations on average at 62 years, whereas mean disease duration was 20 months. The type of spongiform degeneration, the PrP immunostaining pattern, and the presence of microplaques distinguished these cases from those with known prion diseases. Typical protease-resistant PrP was undetectable in the cerebral neocortex with standard diagnostic procedures. After enrichment, abnormal PrP was detected at concentrations 16 times lower than common prion diseases; it included nearly 4 times less protease-resistant PrP, which formed a distinct electrophoretic profile. The subjects examined comprised about 3% of sporadic cases evaluated by the National Prion Disease Pathology Surveillance Center. Although several subjects had family histories of dementia, no mutations were found in the PrP gene open reading frame. INTERPRETATION: The distinct histopathological, PrP immunohistochemical, and physicochemical features, together with the homogeneous genotype, indicate that this is a previously unidentified type of disease involving the PrP, which we designated "protease-sensitive prionopathy" (or PSPr). Protease-sensitive prionopathy is not rare among prion diseases, and it may be even more prevalent than our data indicate because protease-sensitive prionopathy cases are likely also to be classified within the group of non-Alzheimer's dementias.


Subject(s)
Dementia/pathology , Dementia/physiopathology , Prion Diseases/pathology , Prion Diseases/physiopathology , Prions/analysis , Prions/chemistry , Age of Onset , Aged , Brain/metabolism , Brain/pathology , Brain/physiopathology , DNA Mutational Analysis , Dementia/etiology , Disease Progression , Female , Genetic Predisposition to Disease/genetics , Genetic Testing , Genotype , Humans , Immunohistochemistry , Incidence , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Male , Middle Aged , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/metabolism , Neurons/pathology , Peptide Hydrolases/metabolism , Prion Diseases/metabolism , Prions/genetics , Prions/metabolism
18.
J Virol ; 81(6): 2831-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17182694

ABSTRACT

Prion diseases are transmissible neurodegenerative diseases caused by a conformational isoform of the prion protein (PrP), a host-encoded cell surface sialoglycoprotein. Recent evidence suggests a cytosolic fraction of PrP (cyPrP) functions either as an initiating factor or toxic element of prion disease. When expressed in cultured cells, cyPrP acquires properties of the infectious conformation of PrP (PrP(Sc)), including insolubility, protease resistance, aggregation, and toxicity. Transgenic mice (2D1 and 1D4 lines) that coexpress cyPrP and PrP(C) exhibit focal cerebellar atrophy, scratching behavior, and gait abnormalities suggestive of prion disease, although they lack protease-resistant PrP. To determine if the coexpression of PrP(C) is necessary or inhibitory to the phenotype of these mice, we crossed Tg1D4(Prnp(+/+)) mice with PrP-ablated mice (TgPrnp(o/o)) to generate Tg1D4(Prnp(o/o)) mice and followed the development of disease and pathological phenotype. We found no difference in the onset of symptoms or the clinical or pathological phenotype of disease between Tg1D4(Prnp(+/+)) and Tg1D4(Prnp(o/o)) mice, suggesting that cyPrP and PrP(C) function independently in the disease state. Additionally, Tg1D4(Prnp(o/o)) mice were resistant to challenge with mouse-adapted scrapie (RML), suggesting cyPrP is inaccessible to PrP(Sc). We conclude that disease phenotype and cellular toxicity associated with the expression of cyPrP are independent of PrP(C) and the generation of typical prion disease.


Subject(s)
Cytosol/metabolism , Gene Expression , PrPSc Proteins/metabolism , Prions/metabolism , Animals , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Disease Progression , Injections, Intraventricular , Leupeptins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/pathology , Neuroblastoma/virology , PrPSc Proteins/genetics , PrPSc Proteins/pathogenicity , Prions/genetics , Transfection
19.
J Virol ; 80(17): 8521-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16912302

ABSTRACT

The prion diseases are transmissible neurodegenerative disorders linked to a pathogenic conformer (PrP(Sc)) of the normal prion protein (PrP(C)). Accumulation of PrP(Sc) occurs via a poorly defined process in which PrP(Sc) complexes with and converts endogenous PrP(C) to nascent PrP(Sc). Recent experiments have focused on the highly charged first alpha helix (H1) of PrP. It has been proposed that two putative asparagine-to-arginine intrahelical salt bridges stabilize H1 in PrP(C) yet form intermolecular ionic bonds with adjacent PrP molecules during conversion of PrP(C) to PrP(Sc) (M. P. Morrissey and E. I. Shakhnovich, Proc. Natl. Acad. Sci. USA 96:11293-11298, 1999). Subsequent work (J. O. Speare et al., J. Biol. Chem. 278:12522-12529, 2003 using a cell-free assay of PrP(Sc) conversion suggested that rather than promoting conversion, the salt bridges stabilize PrP(C) against it. However, the role of individual H1 charges in PrP(Sc) generation has not yet been investigated. To approach this question, we systematically reversed or neutralized each charged residue in H1 and tested the effect on conversion to PrP(Sc) in scrapie-infected murine neuroblastoma (ScN2a) cells. We find that replacements of charged H1 residues with like charges permit conversion, while charge reversals hinder it. Neutralization of charges in the N-terminal (amino acids 143 to 146) but not the C-terminal (amino acids 147 to 151) half of H1 permits conversion, while complete reversal of charge orientation of the putative salt bridges produces a nonconvertible PrP. Circular dichroism spectroscopy studies and confocal microscopy immunofluorescence localization studies indicated that charge substitutions did not alter the secondary structure or cell surface expression of PrP(C). These data support the necessity of specific charge orientations in H1 for a productive PrP(Sc)-PrP(C) complex.


Subject(s)
PrPC Proteins/chemistry , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , PrPSc Proteins/pathogenicity , Animals , Cell Line, Tumor , Circular Dichroism , Fluorescent Antibody Technique , Mice , Microscopy, Confocal , Mutation , Neuroblastoma , PrPC Proteins/genetics , PrPSc Proteins/chemistry , Prions/chemistry , Prions/genetics , Prions/metabolism
20.
Ann Neurol ; 58(6): 858-64, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16315279

ABSTRACT

The prion protein (PrP) is central to the prion diseases, although a role in other neurodegenerative diseases has been postulated. A common polymorphism (Met or Val) at codon 129 of the PrP gene (PRNP) features prominently in the risk and phenotype, of prion disease, and an abnormality in its distribution frequency may signal a role for PrP in other diseases. We conducted a case-control study to compare the PRNP codon 129 genotype distribution in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and primary progressive aphasia (PPA), including 281 AD, 256 ALS, 39 PPA, and 415 healthy control subjects. Statistical analysis was applied to determine the presence or absence of disease-specific genotype associations. The distribution of codon 129 genotypes was similar among healthy control, AD, and ALS subjects, although the heterozygous state was significantly overrepresented (age-adjusted odds ratio, 8.47) in PPA, a rare condition of unknown cause. Although these findings do not entirely exclude a role for PrP in AD or ALS, they do not support the codon 129 genotype as a risk factor for either disease. However, the strong association between heterozygosity and PPA raises new questions about its cause and the role of PrP in other neurodegenerative diseases.


Subject(s)
Aphasia, Primary Progressive/epidemiology , Aphasia, Primary Progressive/genetics , Prions/genetics , Aged , Aged, 80 and over , Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Amyotrophic Lateral Sclerosis/epidemiology , Amyotrophic Lateral Sclerosis/genetics , Case-Control Studies , Female , Genotype , Heterozygote , Humans , Male , Middle Aged , Phenotype , Prevalence
SELECTION OF CITATIONS
SEARCH DETAIL
...