Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Biochem Biophys Rep ; 34: 101446, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36923008

ABSTRACT

Synucleinopathies are characterized by the deposition of alpha-synuclein (α-syn) aggregates in brain tissue. Pathological α-syn aggregates propagate in a prion-like manner and display prion-like biochemical properties. Using RT-QuIC, we measured α-syn seeding activity from brains of Dementia with Lewy body (DLB) patients post autoclave. Here, we show that autoclaving at 121 °C removes one to two log10 of α-syn seeding activity but the remaining 50% seeding dose (SD50) is more than 107/mg tissue. DLB brain samples autoclaved at 132 °C still revealed an SD50 of approximately 106/mg tissue. Our data suggest that DLB α-syn seeds are incompletely inactivated by standard autoclave, thus highlighting the need for evaluating laboratory procedures that fully inactivate them.

2.
Nat Microbiol ; 6(1): 19-26, 2021 01.
Article in English | MEDLINE | ID: mdl-33139883

ABSTRACT

Gram-negative bacteria have a cell envelope that comprises an outer membrane (OM), a peptidoglycan (PG) layer and an inner membrane (IM)1. The OM and PG are load-bearing, selectively permeable structures that are stabilized by cooperative interactions between IM and OM proteins2,3. In Escherichia coli, Braun's lipoprotein (Lpp) forms the only covalent tether between the OM and PG and is crucial for cell envelope stability4; however, most other Gram-negative bacteria lack Lpp so it has been assumed that alternative mechanisms of OM stabilization are present5. We used a glycoproteomic analysis of PG to show that ß-barrel OM proteins are covalently attached to PG in several Gram-negative species, including Coxiella burnetii, Agrobacterium tumefaciens and Legionella pneumophila. In C. burnetii, we found that four different types of covalent attachments occur between OM proteins and PG, with tethering of the ß-barrel OM protein BbpA becoming most abundant in the stationary phase and tethering of the lipoprotein LimB similar throughout the cell cycle. Using a genetic approach, we demonstrate that the cell cycle-dependent tethering of BbpA is partly dependent on a developmentally regulated L,D-transpeptidase (Ldt). We use our findings to propose a model of Gram-negative cell envelope stabilization that includes cell cycle control and an expanded role for Ldts in covalently attaching surface proteins to PG.


Subject(s)
Agrobacterium tumefaciens/metabolism , Bacterial Outer Membrane Proteins/metabolism , Coxiella burnetii/metabolism , Escherichia coli/metabolism , Legionella pneumophila/metabolism , Peptidoglycan/metabolism , Cell Cycle/physiology , Cell Membrane/metabolism , Cell Wall/metabolism , Lipoproteins/metabolism , Molecular Dynamics Simulation , Peptidyl Transferases/metabolism , Protein Binding/physiology
3.
PLoS One ; 14(7): e0219457, 2019.
Article in English | MEDLINE | ID: mdl-31291644

ABSTRACT

Prion protein (PrPC) is a protease-sensitive and soluble cell surface glycoprotein expressed in almost all mammalian cell types. PrPSc, a protease-resistant and insoluble form of PrPC, is the causative agent of prion diseases, fatal and transmissible neurogenerative diseases of mammals. Prion infection is initiated via either ingestion or inoculation of PrPSc or when host PrPC stochastically refolds into PrPSc. In either instance, the early events that occur during prion infection remain poorly understood. We have used transgenic mice expressing mouse PrPC tagged with a unique antibody epitope to monitor the response of host PrPC to prion inoculation. Following intracranial inoculation of either prion-infected or uninfected brain homogenate, we show that host PrPC can accumulate both intra-axonally and within the myelin membrane of axons suggesting that it may play a role in axonal loss following brain injury. Moreover, in response to the inoculation host PrPC exhibits an increased insolubility and protease resistance similar to that of PrPSc, even in the absence of infectious prions. Thus, our results raise the possibility that damage to the brain may be one trigger by which PrPC stochastically refolds into pathogenic PrPSc leading to productive prion infection.


Subject(s)
PrPC Proteins/genetics , PrPSc Proteins/genetics , Prion Diseases/genetics , Prion Proteins/genetics , Animals , Brain/metabolism , Brain/pathology , Epitopes/genetics , Epitopes/immunology , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/metabolism , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , PrPC Proteins/chemistry , PrPSc Proteins/chemistry , Prion Diseases/pathology
4.
Personal Neurosci ; 2: e10, 2019.
Article in English | MEDLINE | ID: mdl-32435745

ABSTRACT

Psychologists have identified multiple different forms of conflict, such as information processing conflict and goal conflict. As such, there is a need to examine the similarities and differences in neurology between each form of conflict. To address this, we conducted a comprehensive electroencephalogram (EEG) analysis of Shadli, Glue, McIntosh, and McNaughton's calibrated stop-signal task (SST) goal-conflict task. Specifically, we examined changes in scalp-wide current source density (CSD) power and coherence across a wide range of frequency bands during the calibrated SST (n = 34). We assessed differences in EEG between the high and low goal-conflict conditions using hierarchical analyses of variance (ANOVAs). We also related goal-conflict EEG to trait anxiety, neuroticism, Behavioural Inhibition System (BIS)-anxiety and revised BIS (rBIS) using regression analyses. We found that changes in CSD power during goal conflict were limited to increased midfrontocentral theta. Conversely, coherence increased across 23 scalp-wide theta region pairs and one frontal delta region pair. Finally, scalp-wide theta significantly predicted trait neuroticism but not trait anxiety, BIS-anxiety or rBIS. We conclude that goal conflict involves increased midfrontocentral CSD theta power and scalp-wide theta-dominated coherence. Therefore, compared with information processing conflict, goal conflict displays a similar EEG power profile of midfrontocentral theta but a much wider coherence profile. Furthermore, the increases in theta during goal conflict are the characteristic of BIS-driven activity. Therefore, future research should confirm whether these goal-conflict effects are driven by the BIS by examining whether the effects are attenuated by anxiolytic drugs. Overall, we have identified a unique network of goal-conflict EEG during the calibrated SST.

5.
Biochim Biophys Acta Proteins Proteom ; 1866(11): 1174-1180, 2018 11.
Article in English | MEDLINE | ID: mdl-30282615

ABSTRACT

Prions represent a class of universally fatal and transmissible neurodegenerative disorders that affect humans and other mammals. The prion agent contains a pathologically aggregated form of the host prion protein that can transmit infectivity without any bacterial or viral component and is thus difficult to inactivate using disinfection protocols designed for infectious microorganisms. Methods for prion inactivation include treatment with acids, bases, detergents, bleach, prolonged autoclaving and incineration. During these procedures, the sample is often either destroyed or damaged such that further analysis for research purposes is compromised. In this study we show that a straightforward denaturation and in-gel protease digestion protocol used to prepare prion-infected samples for mass spectroscopy leads to the loss of at least 7 logs of prion infectivity, yielding a final product that fails to transmit prion disease in vivo. We further show that the resultant sample remains suitable for mass spectrometry-based protein identifications. Thus, the procedure described can be used to prepare prion-infected samples for mass spectrometry analysis with greatly reduced biosafety concerns.


Subject(s)
Mass Spectrometry , PrPSc Proteins/chemistry , Animals , Cricetinae , Detergents/pharmacology , Disinfection , Mice , PrPSc Proteins/isolation & purification , Protein Denaturation
6.
Int J Psychophysiol ; 120: 108-117, 2017 10.
Article in English | MEDLINE | ID: mdl-28739481

ABSTRACT

Band power linked to lower and upper alpha (i.e. 8-10Hz; 10-12Hz) and lower and upper beta (i.e. 12-20Hz; 20-30Hz) were examined during response related stages, including anticipation, response execution (RE), response inhibition (RI) and post response recovery (PRR). Group and individual data from 34 participants were considered. The participant's objective was to press a response key immediately following 4 non-repeating, single integer odd digits. These were presented amongst a continuous stream of digits and Xs. Electroencephalogram (EEG) signals were recorded from 32 electrodes (pooled to 12 regions). In the group analyses, participant EEG response was compared to baseline revealing that upper alpha desynchronised during anticipation, RE and RI; lower beta during anticipation and RE; and upper beta just RE. Upper alpha desynchronisation during rapid, unplanned RI is novel. Also, upper alpha and lower/upper beta synchronised during PRR. For upper alpha, we speculate this indexes brief cortical deactivation; for beta we propose this indexes response set maintenance. Lastly, lower alpha fluctuations correlated negatively with RT, indexing neural efficiency. Individual analyses involved calculation of the proportion of individuals displaying the typical RE and PRR trends; these were not reflected by all participants. The former was displayed individually by the largest proportion in upper alpha recorded left fronto-centrally; the latter was most reliably displayed individually in lower beta recorded mid centro-parietally. Therefore, group analyses identified typical alpha and beta synchronisation/desynchronisation trends, whilst individual analyses identified their degree of representation in single participants. Attention is drawn to the clinical relevance of this issue.


Subject(s)
Alpha Rhythm/physiology , Beta Rhythm/physiology , Brain Waves/physiology , Cerebral Cortex/physiology , Movement/physiology , Adolescent , Adult , Analysis of Variance , Brain Mapping , Electroencephalography , Female , Follow-Up Studies , Humans , Inhibition, Psychological , Male , Middle Aged , Psychomotor Performance/physiology , Reaction Time/physiology , Young Adult
7.
Clin Case Rep ; 5(6): 744-747, 2017 06.
Article in English | MEDLINE | ID: mdl-28588802

ABSTRACT

Adenosine is increasingly used to assess for dormant conduction following pulmonary vein isolation during atrial fibrillation ablation. While the half-life of adenosine is typically short and side effects transient, operators should be aware of more serious, lasting adverse reactions including anaphylaxis and bronchospasm.

8.
J Virol ; 91(18)2017 09 15.
Article in English | MEDLINE | ID: mdl-28659480

ABSTRACT

Mitochondria are crucial to proper neuronal function and overall brain health. Mitochondrial dysfunction within the brain has been observed in many neurodegenerative diseases, including prion disease. Several markers of decreased mitochondrial activity during prion infection have been reported, yet the bioenergetic respiratory status of mitochondria from prion-infected animals is unknown. Here we show that clinically ill transgenic mice overexpressing hamster prion protein (Tg7) infected with the hamster prion strain 263K suffer from a severe deficit in mitochondrial oxygen consumption in response to the respiratory complex II substrate succinate. Characterization of the mitochondrial proteome of purified brain mitochondria from infected and uninfected Tg7 mice showed significant differences in the relative abundance of key mitochondrial electron transport proteins in 263K-infected animals relative to that in controls. Our results suggest that at clinical stages of prion infection, dysregulation of respiratory chain proteins may lead to impairment of mitochondrial respiration in the brain.IMPORTANCE Mitochondrial dysfunction is present in most major neurodegenerative diseases, and some studies have suggested that mitochondrial processes may be altered during prion disease. Here we show that hamster prion-infected transgenic mice overexpressing the hamster prion protein (Tg7 mice) suffer from mitochondrial respiratory deficits. Tg7 mice infected with the 263K hamster prion strain have little or no signs of mitochondrial dysfunction at the disease midpoint but suffer from a severe deficit in mitochondrial respiration at the clinical phase of disease. A proteomic analysis of the isolated brain mitochondria from clinically affected animals showed that several proteins involved in electron transport, mitochondrial dynamics, and mitochondrial protein synthesis were dysregulated. These results suggest that mitochondrial dysfunction, possibly exacerbated by prion protein overexpression, occurs at late stages during 263K prion disease and that this dysfunction may be the result of dysregulation of mitochondrial proteins.


Subject(s)
Brain/pathology , Cell Respiration , Mitochondria/metabolism , Prion Diseases/pathology , Animals , Disease Models, Animal , Electron Transport , Mice, Transgenic , Mitochondria/chemistry , Oxygen/metabolism , Proteome/analysis
9.
Sci Rep ; 7: 41556, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28148964

ABSTRACT

Cellular prion protein (PrPC) is a mammalian glycoprotein which is usually found anchored to the plasma membrane via a glycophosphatidylinositol (GPI) anchor. PrPC misfolds to a pathogenic isoform PrPSc, the causative agent of neurodegenerative prion diseases. The precise function of PrPC remains elusive but may depend upon its cellular localization. Here we show that PrPC is present in brain mitochondria from 6-12 week old wild-type and transgenic mice in the absence of disease. Mitochondrial PrPC was fully processed with mature N-linked glycans and did not require the GPI anchor for localization. Protease treatment of purified mitochondria suggested that mitochondrial PrPC exists as a transmembrane isoform with the C-terminus facing the mitochondrial matrix and the N-terminus facing the intermembrane space. Taken together, our data suggest that PrPC can be found in mitochondria in the absence of disease, old age, mutation, or overexpression and that PrPC may affect mitochondrial function.


Subject(s)
Mitochondria/metabolism , PrPC Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Chromatography, Liquid , Electron Transport Complex IV/metabolism , Glycosylation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Polysaccharides/metabolism , PrPC Proteins/chemistry , Prion Diseases/metabolism , Protein Binding , Tandem Mass Spectrometry
10.
J Proteome Res ; 15(12): 4518-4531, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27597180

ABSTRACT

Aggregated and protease-resistant mammalian prion protein (PrPSc) is the primary protein component of infectious prions. Enriched PrPSc preparations are often used to study the mechanisms that underly prion disease. However, most enrichment procedures are relatively nonspecific and tend to yield significant amounts of non-PrPSc components including various proteins that could confound functional and structural studies. It is thus important to identify these proteins and assess their potential relevance to prion pathogenesis. Following proteinase K treatment and phosphotungstic acid precipitation of brain homogenate, we have used mass spectrometry to analyze the protein content of PrPSc isolated from prion-infected mice, multiple cases of sporadic Creutzfeldt-Jakob disease (sCJD), and human growth hormone associated cases of iatrogenic CJD (iCJD). Creatine kinase was the primary protein contaminant in all PrPSc samples, while many of the other proteins identified were also found in non-CJD controls, which suggests that they are not CJD specific. Interestingly, the Alzheimer's disease associated peptide amyloid ß 1-42 (Aß1-42) was identified in the majority of the sCJD cases as well as non-CJD age-matched controls, while apoliprotein E was found in greater abundance in the sCJD cases. By contrast, while some of the iCJD cases showed evidence of higher molecular weight Aß oligomers, monomeric Aß1-42 peptide was not detected by immunoblot, and only one case had significant levels of apolipoprotein E. Our data are consistent with the age-associated deposition of Aß1-42 in older sporadic CJD and non-CJD patients and suggest that both apolipoprotein E and Aß1-42 abundance can differ depending upon the type of CJD.


Subject(s)
Amyloid beta-Peptides/analysis , Apolipoproteins E/analysis , Creutzfeldt-Jakob Syndrome/classification , Peptide Fragments/analysis , Prion Proteins/analysis , Adult , Age Factors , Aged , Animals , Female , Humans , Male , Mice , Middle Aged , Prion Proteins/isolation & purification
13.
PLoS Pathog ; 12(2): e1005416, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26840342

ABSTRACT

Sporadic Creutzfeldt-Jakob disease (sCJD) is the most prevalent of the human prion diseases, which are fatal and transmissible neurodegenerative diseases caused by the infectious prion protein (PrP(Sc)). The origin of sCJD is unknown, although the initiating event is thought to be the stochastic misfolding of endogenous prion protein (PrP(C)) into infectious PrP(Sc). By contrast, human growth hormone-associated cases of iatrogenic CJD (iCJD) in the United Kingdom (UK) are associated with exposure to an exogenous source of PrP(Sc). In both forms of CJD, heterozygosity at residue 129 for methionine (M) or valine (V) in the prion protein gene may affect disease phenotype, onset and progression. However, the relative contribution of each PrP(C) allotype to PrP(Sc) in heterozygous cases of CJD is unknown. Using mass spectrometry, we determined that the relative abundance of PrP(Sc) with M or V at residue 129 in brain specimens from MV cases of sCJD was highly variable. This result is consistent with PrP(C) containing an M or V at residue 129 having a similar propensity to misfold into PrP(Sc) thus causing sCJD. By contrast, PrP(Sc) with V at residue 129 predominated in the majority of the UK human growth hormone associated iCJD cases, consistent with exposure to infectious PrP(Sc) containing V at residue 129. In both types of CJD, the PrP(Sc) allotype ratio had no correlation with CJD type, age at clinical onset, or disease duration. Therefore, factors other than PrP(Sc) allotype abundance must influence the clinical progression and phenotype of heterozygous cases of CJD.


Subject(s)
Creutzfeldt-Jakob Syndrome/genetics , PrPC Proteins/genetics , PrPSc Proteins/genetics , Adult , Aged , Brain/pathology , Brain Chemistry , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Female , Humans , Iatrogenic Disease , Male , Methionine/genetics , Middle Aged , Phenotype , PrPC Proteins/chemistry , PrPC Proteins/metabolism , PrPSc Proteins/chemistry , PrPSc Proteins/metabolism , Recombinant Proteins , Valine/genetics
14.
Expert Rev Proteomics ; 12(2): 171-84, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25795148

ABSTRACT

Prion diseases are a heterogeneous class of fatal neurodegenerative disorders associated with misfolding of host cellular prion protein (PrP(C)) into a pathological isoform, termed PrP(Sc). Prion diseases affect various mammals, including humans, and effective treatments are not available. Prion diseases are distinguished from other protein misfolding disorders - such as Alzheimer's or Parkinson's disease - in that they are infectious. Prion diseases occur sporadically without any known exposure to infected material, and hereditary cases resulting from rare mutations in the prion protein have also been documented. The mechanistic underpinnings of prion and other neurodegenerative disorders remain poorly understood. Various proteomics techniques have been instrumental in early PrP(Sc) detection, biomarker discovery, elucidation of PrP(Sc) structure and mapping of biochemical pathways affected by pathogenesis. Moving forward, proteomics approaches will likely become more integrated into the clinical and research settings for the rapid diagnosis and characterization of prion pathogenesis.


Subject(s)
Prion Diseases/metabolism , Prions/chemistry , Animals , Biomarkers , Humans , Prion Diseases/diagnosis , Prions/metabolism , Proteomics/methods
15.
J Proteome Res ; 13(11): 4620-34, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25140793

ABSTRACT

Prion diseases are a heterogeneous group of neurodegenerative disorders affecting various mammals including humans. Prion diseases are characterized by a misfolding of the host-encoded prion protein (PrP(C)) into a pathological isoform termed PrP(Sc). In wild-type mice, PrP(C) is attached to the plasma membrane by a glycosylphosphatidylinositol (GPI) anchor and PrP(Sc) typically accumulates in diffuse nonamyloid deposits with gray matter spongiosis. By contrast, when mice lacking the GPI anchor are infected with the same prion inoculum, PrP(Sc) accumulates in dense perivascular amyloid plaques with little or no gray matter spongiosis. In order to evaluate whether different host biochemical pathways were implicated in these two phenotypically distinct prion disease models, we utilized a proteomics approach. In both models, infected mice displayed evidence of a neuroinflammatory response and complement activation. Proteins involved in cell death and calcium homeostasis were also identified in both phenotypes. However, mitochondrial pathways of apoptosis were implicated only in the nonamyloid form, whereas metal binding and synaptic vesicle transport were more disrupted in the amyloid phenotype. Thus, following infection with a single prion strain, PrP(C) anchoring to the plasma membrane correlated not only with the type of PrP(Sc) deposition but also with unique biochemical pathways associated with pathogenesis.


Subject(s)
Amyloid/metabolism , Phenotype , Prion Diseases/metabolism , Prion Diseases/physiopathology , Proteomics/methods , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Calcium/metabolism , Cell Membrane/metabolism , Chromatography, Liquid , Homeostasis/genetics , Homeostasis/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Tandem Mass Spectrometry
16.
PLoS One ; 8(7): e71081, 2013.
Article in English | MEDLINE | ID: mdl-23936256

ABSTRACT

During prion infection, the normal, protease-sensitive conformation of prion protein (PrP(C)) is converted via seeded polymerization to an abnormal, infectious conformation with greatly increased protease-resistance (PrP(Sc)). In vitro, protein misfolding cyclic amplification (PMCA) uses PrP(Sc) in prion-infected brain homogenates as an initiating seed to convert PrP(C) and trigger the self-propagation of PrP(Sc) over many cycles of amplification. While PMCA reactions produce high levels of protease-resistant PrP, the infectious titer is often lower than that of brain-derived PrP(Sc). More recently, PMCA techniques using bacterially derived recombinant PrP (rPrP) in the presence of lipid and RNA but in the absence of any starting PrP(Sc) seed have been used to generate infectious prions that cause disease in wild-type mice with relatively short incubation times. These data suggest that lipid and/or RNA act as cofactors to facilitate the de novo formation of high levels of prion infectivity. Using rPrP purified by two different techniques, we generated a self-propagating protease-resistant rPrP molecule that, regardless of the amount of RNA and lipid used, had a molecular mass, protease resistance and insolubility similar to that of PrP(Sc). However, we were unable to detect prion infectivity in any of our reactions using either cell-culture or animal bioassays. These results demonstrate that the ability to self-propagate into a protease-resistant insoluble conformer is not unique to infectious PrP molecules. They suggest that the presence of RNA and lipid cofactors may facilitate the spontaneous refolding of PrP into an infectious form while also allowing the de novo formation of self-propagating, but non-infectious, rPrP-res.


Subject(s)
Lipids/chemistry , Prions/chemistry , Protein Refolding , RNA/chemistry , Recombinant Proteins/chemistry , Animals , Brain/metabolism , Brain/pathology , Cell Line , Detergents/chemistry , Female , Mice , PrPC Proteins/chemistry , PrPC Proteins/metabolism , PrPC Proteins/ultrastructure , PrPSc Proteins/chemistry , PrPSc Proteins/metabolism , PrPSc Proteins/ultrastructure , Prions/metabolism , Prions/ultrastructure , Proteolysis , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Solubility
17.
Immunity ; 38(4): 705-16, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23499490

ABSTRACT

La Crosse virus (LACV), a zoonotic Bunyavirus, is a major cause of pediatric viral encephalitis in the United States. A hallmark of neurological diseases caused by LACV and other encephalitic viruses is the induction of neuronal cell death. Innate immune responses have been implicated in neuronal damage, but no mechanism has been elucidated. By using in vitro studies in primary neurons and in vivo studies in mice, we have shown that LACV infection induced the RNA helicase, RIG-I, and mitochondrial antiviral signaling protein (MAVS) signaling pathway, resulting in upregulation of the sterile alpha and TIR-containing motif 1 (SARM1), an adaptor molecule that we found to be directly involved in neuronal damage. SARM1-mediated cell death was associated with induced oxidative stress response and mitochondrial damage. These studies provide an innate-immune signaling mechanism for virus-induced neuronal death and reveal potential targets for development of therapeutics to treat encephalitic viral infections.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Armadillo Domain Proteins/metabolism , Cytoskeletal Proteins/metabolism , Encephalitis, California/immunology , La Crosse virus/immunology , Mitochondria/metabolism , Neurons/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Armadillo Domain Proteins/genetics , Cells, Cultured , Cytoskeletal Proteins/genetics , Encephalitis, California/complications , Encephalitis, California/drug therapy , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Neurons/virology , Oxidative Stress , Primary Cell Culture , Signal Transduction/immunology , Up-Regulation
18.
J Virol ; 86(21): 11763-78, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22915801

ABSTRACT

Mammalian prions are thought to consist of misfolded aggregates (protease-resistant isoform of the prion protein [PrP(res)]) of the cellular prion protein (PrP(C)). Transmissible spongiform encephalopathy (TSE) can be induced in animals inoculated with recombinant PrP (rPrP) amyloid fibrils lacking mammalian posttranslational modifications, but this induction is inefficient in hamsters or transgenic mice overexpressing glycosylphosphatidylinositol (GPI)-anchored PrP(C). Here we show that TSE can be initiated by inoculation of misfolded rPrP into mice that express wild-type (wt) levels of PrP(C) and that synthetic prion strain propagation and selection can be affected by GPI anchoring of the host's PrP(C). To create prions de novo, we fibrillized mouse rPrP in the absence of molecular cofactors, generating fibrils with a PrP(res)-like protease-resistant banding profile. These fibrils induced the formation of PrP(res) deposits in transgenic mice coexpressing wt and GPI-anchorless PrP(C) (wt/GPI(-)) at a combined level comparable to that of PrP(C) expression in wt mice. Secondary passage into mice expressing wt, GPI(-), or wt plus GPI(-) PrP(C) induced TSE disease with novel clinical, histopathological, and biochemical phenotypes. Contrary to laboratory-adapted mouse scrapie strains, the synthetic prion agents exhibited a preference for conversion of GPI(-) PrP(C) and, in one case, caused disease only in GPI(-) mice. Our data show that novel TSE agents can be generated de novo solely from purified mouse rPrP after amplification in mice coexpressing normal levels of wt and anchorless PrP(C). These observations provide insight into the minimal elements required to create prions in vitro and suggest that the PrP(C) GPI anchor can modulate the propagation of synthetic TSE strains.


Subject(s)
Prions/genetics , Prions/isolation & purification , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prions/pathogenicity , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
19.
Int J Psychophysiol ; 85(2): 135-44, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22732350

ABSTRACT

Previous research has revealed that EEG theta oscillations are affected during goal conflict processing. This is consistent with the behavioural inhibition system (BIS) theory of anxiety (Gray & McNaughton, 2000). However, studies have not attempted to relate these BIS-related theta effects to BIS personality measures. Confirmation of such an association would provide further support for BIS theory, especially as it relates to trait differences. EEG was measured (32 electrodes) from extreme groups (low/high trait BIS) engaged in a target detection task. Goal conflicts were introduced throughout the task. Results show that the two groups did not differ in behavioural performance. The major EEG result was that a stepwise discriminant analysis indicated discrimination by 6 variables derived from coherence and power, with 5 of the 6 in the theta range as predicted by BIS theory and one in the beta range. Also, across the whole sample, EEG theta coherence increased at a variety of regions during primary goal conflict and showed a general increase during response execution; EEG theta power, in contrast, was primarily reactive to response execution. This is the first study to reveal a three-way relationship between the induction of goal conflict, the induction of theta power and coherence, and differentiation by psychometrically-defined low/high BIS status.


Subject(s)
Brain/physiology , Conflict, Psychological , Goals , Inhibition, Psychological , Monitoring, Physiologic , Theta Rhythm/physiology , Adolescent , Adult , Analysis of Variance , Brain Mapping , Discriminant Analysis , Electroencephalography , Female , Humans , Longitudinal Studies , Male , Mathematics , Middle Aged , Neuropsychological Tests , Reaction Time/physiology , Young Adult
20.
Biochemistry ; 51(22): 4600-8, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22591453

ABSTRACT

Fibril dissociation is necessary for efficient conversion of normal prion protein to its misfolded state and continued propagation into amyloid. Recent studies have revealed that conversion occurs along the endocytic pathway. To improve our understanding of the dissociation process, we have investigated the effect of low pH on the stability of recombinant prion fibrils. We show that under conditions that mimic the endocytic environment, amyloid fibrils made from full-length prion protein dissociate both laterally and axially to form protofilaments. Approximately 5% of the protofilaments are short enough to be considered soluble and contain ~100-300 monomers per structure; these also retain the biophysical characteristics of the filaments. We propose that protonation of His residues and charge repulsion in the N-terminal domain trigger fibril dissociation. Our data suggest that lysosomes and late endosomes are competent milieus for propagating the misfolded state not only by destabilizing the normal prion protein but also by accelerating the dissociation of fibrils into smaller structures that may act as seeds.


Subject(s)
Amyloid/chemistry , Amyloid/metabolism , Prions/chemistry , Prions/metabolism , Amyloid/ultrastructure , Animals , Circular Dichroism , Cricetinae , Endocytosis , Hydrogen-Ion Concentration , Mesocricetus , Prions/ultrastructure , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility , Spectroscopy, Fourier Transform Infrared
SELECTION OF CITATIONS
SEARCH DETAIL
...