Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 20(9): e1012517, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39264912

RESUMEN

The cellular prion protein, PrPC, has been postulated to function as a receptor for α-synuclein, potentially facilitating cell-to-cell spreading and/or toxicity of α-synuclein aggregates in neurodegenerative disorders such as Parkinson's disease. Previously, we generated the "Salt (S)" and "No Salt (NS)" strains of α-synuclein aggregates that cause distinct pathological phenotypes in M83 transgenic mice overexpressing A53T-mutant human α-synuclein. To test the hypothesis that PrPC facilitates the propagation of α-synuclein aggregates, we produced M83 mice that either express or do not express PrPC. Following intracerebral inoculation with the S or NS strain, the absence of PrPC in M83 mice did not prevent disease development and had minimal influence on α-synuclein strain-specified attributes such as the extent of cerebral α-synuclein deposition, selective targeting of specific brain regions and cell types, the morphology of induced α-synuclein deposits, and the structural fingerprints of protease-resistant α-synuclein aggregates. Likewise, there were no appreciable differences in disease manifestation between PrPC-expressing and PrPC-lacking M83 mice following intraperitoneal inoculation of the S strain. Interestingly, intraperitoneal inoculation with the NS strain resulted in two distinct disease phenotypes, indicative of α-synuclein strain evolution, but this was also independent of PrPC expression. Overall, these results suggest that PrPC plays at most a minor role in the propagation, neuroinvasion, and evolution of α-synuclein strains in mice that express A53T-mutant human α-synuclein. Thus, other putative receptors or cell-to-cell propagation mechanisms may have a larger effect on the spread of α-synuclein aggregates during disease.


Asunto(s)
Sinucleinopatías , alfa-Sinucleína , Animales , Humanos , Ratones , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Ratones Transgénicos , Proteínas PrPC/metabolismo , Proteínas PrPC/genética , Sinucleinopatías/metabolismo , Sinucleinopatías/patología
2.
PLoS Pathog ; 20(9): e1012538, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39255320

RESUMEN

In prion diseases, the species barrier limits the transmission of prions from one species to another. However, cross-species prion transmission is remarkably efficient in bank voles, and this phenomenon is mediated by the bank vole prion protein (BVPrP). The molecular determinants of BVPrP's ability to function as a universal prion acceptor remain incompletely defined. Building on our finding that cultured cells expressing BVPrP can replicate both mouse and hamster prion strains, we systematically identified key residues in BVPrP that permit cross-species prion replication. We found that residues N155 and N170 of BVPrP, which are absent in mouse PrP but present in hamster PrP, are critical for cross-species prion replication. Additionally, BVPrP residues V112, I139, and M205, which are absent in hamster PrP but present in mouse PrP, are also required to enable replication of both mouse and hamster prions. Unexpectedly, we found that residues E227 and S230 near the C-terminus of BVPrP severely restrict prion accumulation following cross-species prion challenge, suggesting that they may have evolved to counteract the inherent propensity of BVPrP to misfold. PrP variants with an enhanced ability to replicate both mouse and hamster prions displayed accelerated spontaneous aggregation kinetics in vitro. These findings suggest that BVPrP's unusual properties are governed by a key set of amino acids and that the enhanced misfolding propensity of BVPrP may enable cross-species prion replication.


Asunto(s)
Arvicolinae , Enfermedades por Prión , Animales , Ratones , Cricetinae , Enfermedades por Prión/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/transmisión , Proteínas Priónicas/metabolismo , Proteínas Priónicas/genética , Especificidad de la Especie , Priones/metabolismo
3.
Front Neurosci ; 18: 1436262, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161653

RESUMEN

Intra- or extracellular aggregates of proteins are central pathogenic features in most neurodegenerative disorders. The accumulation of such proteins in diseased brains is believed to be the end-stage of a stepwise aggregation of misfolded monomers to insoluble cross-ß fibrils via a series of differently sized soluble oligomers/protofibrils. Several studies have shown how α-synuclein, amyloid-ß, tau and other amyloidogenic proteins can act as nucleating particles and thereby share properties with misfolded forms, or strains, of the prion protein. Although the roles of different protein assemblies in the respective aggregation cascades remain unclear, oligomers/protofibrils are considered key pathogenic species. Numerous observations have demonstrated their neurotoxic effects and a growing number of studies have indicated that they also possess seeding properties, enabling their propagation within cellular networks in the nervous system. The seeding behavior of oligomers differs between the proteins and is also affected by various factors, such as size, shape and epitope presentation. Here, we are providing an overview of the current state of knowledge with respect to the "prion-like" behavior of soluble oligomers for several of the amyloidogenic proteins involved in neurodegenerative diseases. In addition to providing new insight into pathogenic mechanisms, research in this field is leading to novel diagnostic and therapeutic opportunities for neurodegenerative diseases.

4.
J Clin Invest ; 134(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39087478

RESUMEN

Most cases of human prion disease arise due to spontaneous misfolding of WT or mutant prion protein, yet recapitulating this event in animal models has proven challenging. It remains unclear whether spontaneous prion generation can occur within the mouse lifespan in the absence of protein overexpression and how disease-causing mutations affect prion strain properties. To address these issues, we generated knockin mice that express the misfolding-prone bank vole prion protein (BVPrP). While mice expressing WT BVPrP (I109 variant) remained free from neurological disease, a subset of mice expressing BVPrP with mutations (D178N or E200K) causing genetic prion disease developed progressive neurological illness. Brains from spontaneously ill knockin mice contained prion disease-specific neuropathological changes as well as atypical protease-resistant BVPrP. Moreover, brain extracts from spontaneously ill D178N- or E200K-mutant BVPrP-knockin mice exhibited prion seeding activity and transmitted disease to mice expressing WT BVPrP. Surprisingly, the properties of the D178N- and E200K-mutant prions appeared identical before and after transmission, suggesting that both mutations guide the formation of a similar atypical prion strain. These findings imply that knockin mice expressing mutant BVPrP spontaneously develop a bona fide prion disease and that mutations causing prion diseases may share a uniform initial mechanism of action.


Asunto(s)
Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Ratones Transgénicos , Enfermedades por Prión , Proteínas Priónicas , Animales , Ratones , Enfermedades por Prión/genética , Enfermedades por Prión/patología , Enfermedades por Prión/metabolismo , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Mutación Missense , Humanos , Arvicolinae/genética , Arvicolinae/metabolismo , Sustitución de Aminoácidos , Priones/genética , Priones/metabolismo , Pliegue de Proteína
5.
PLoS Pathog ; 20(4): e1012087, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38557815

RESUMEN

Prion diseases uniquely manifest in three distinct forms: inherited, sporadic, and infectious. Wild-type prions are responsible for the sporadic and infectious versions, while mutant prions cause inherited variants like fatal familial insomnia (FFI) and familial Creutzfeldt-Jakob disease (fCJD). Although some drugs can prolong prion incubation times up to four-fold in rodent models of infectious prion diseases, no effective treatments for FFI and fCJD have been found. In this study, we evaluated the efficacy of various anti-prion drugs on newly-developed knock-in mouse models for FFI and fCJD. These models express bank vole prion protein (PrP) with the pathogenic D178N and E200K mutations. We applied various drug regimens known to be highly effective against wild-type prions in vivo as well as a brain-penetrant compound that inhibits mutant PrPSc propagation in vitro. None of the regimens tested (Anle138b, IND24, Anle138b + IND24, cellulose ether, and PSCMA) significantly extended disease-free survival or prevented mutant PrPSc accumulation in either knock-in mouse model, despite their ability to induce strain adaptation of mutant prions. Our results show that anti-prion drugs originally developed to treat infectious prion diseases do not necessarily work for inherited prion diseases, and that the recombinant sPMCA is not a reliable platform for identifying compounds that target mutant prions. This work underscores the need to develop therapies and validate screening assays specifically for mutant prions, as well as anti-prion strategies that are not strain-dependent.


Asunto(s)
Síndrome de Creutzfeldt-Jakob , Enfermedades por Prión , Priones , Animales , Ratones , Priones/metabolismo , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Síndrome de Creutzfeldt-Jakob/tratamiento farmacológico , Síndrome de Creutzfeldt-Jakob/genética , Síndrome de Creutzfeldt-Jakob/metabolismo , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Encéfalo/patología , Arvicolinae/metabolismo
6.
Nat Commun ; 15(1): 375, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38195640

RESUMEN

Selective autophagy is an essential process to maintain cellular homeostasis through the constant recycling of damaged or superfluous components. Over a dozen selective autophagy pathways mediate the degradation of diverse cellular substrates, but whether these pathways can influence one another remains unknown. We address this question using pexophagy, the autophagic degradation of peroxisomes, as a model. We show in cells that upregulated pexophagy impairs the selective autophagy of both mitochondria and protein aggregates by exhausting the autophagy initiation factor, ULK1. We confirm this finding in cell models of the pexophagy-mediated form of Zellweger Spectrum Disorder, a disease characterized by peroxisome dysfunction. Further, we extend the generalizability of limited selective autophagy by determining that increased protein aggregate degradation reciprocally reduces pexophagy using cell models of Parkinson's Disease and Huntington's Disease. Our findings suggest that the degradative capacity of selective autophagy can become limited by an increase in one substrate.


Asunto(s)
Enfermedad de Huntington , Enfermedad de Parkinson , Humanos , Macroautofagia/genética , Autofagia/genética , Enfermedad de Huntington/genética , Mitocondrias/genética , Enfermedad de Parkinson/genética
7.
bioRxiv ; 2023 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-37808761

RESUMEN

Prion diseases uniquely manifest in three distinct forms: inherited, sporadic, and infectious. Wild-type prions are responsible for the sporadic and infectious versions, while mutant prions cause inherited variants like fatal familial insomnia (FFI) and familial Creutzfeldt-Jakob disease (fCJD). Although some drugs can prolong prion incubation times up to four-fold in rodent models of infectious prion diseases, no effective treatments for FFI and fCJD have been found. In this study, we evaluated the efficacy of various anti-prion drugs on newly-developed knock-in mouse models for FFI and fCJD. These models express bank vole prion protein (PrP) with the pathogenic D178N and E200K mutations. We applied various drug regimens known to be highly effective against wild-type prions in vivo as well as a brain-penetrant compound that inhibits mutant PrP Sc propagation in vitro . None of the regimens tested (Anle138b, IND24, Anle138b + IND24, cellulose ether, and PSCMA) significantly extended disease-free survival or prevented mutant PrP Sc accumulation in either knock-in mouse model, despite their ability to induce strain adaptation of mutant prions. Paradoxically, the combination of Anle138b and IND24 appeared to accelerate disease by 16% and 26% in kiBVI E200K and kiBVI D178N mice, respectively, and accelerated the aggregation of mutant PrP molecules in vitro . Our results show that anti-prion drugs originally developed to treat infectious prion diseases do not necessarily work for inherited prion diseases, and that the recombinant sPMCA is not a reliable platform for identifying compounds that target mutant prions. This work underscores the need to develop therapies and validate screening assays specifically for mutant prions.

8.
Acta Neuropathol Commun ; 11(1): 72, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-37138318

RESUMEN

Unique strains of α-synuclein aggregates have been postulated to underlie the spectrum of clinical and pathological presentations seen across the synucleinopathies. Whereas multiple system atrophy (MSA) is associated with a predominance of oligodendroglial α-synuclein inclusions, α-synuclein aggregates in Parkinson's disease (PD) preferentially accumulate in neurons. The G51D mutation in the SNCA gene encoding α-synuclein causes an aggressive, early-onset form of PD that exhibits clinical and neuropathological traits reminiscent of both PD and MSA. To assess the strain characteristics of G51D PD α-synuclein aggregates, we performed propagation studies in M83 transgenic mice by intracerebrally inoculating patient brain extracts. The properties of the induced α-synuclein aggregates in the brains of injected mice were examined using immunohistochemistry, a conformational stability assay, and by performing α-synuclein seed amplification assays. Unlike MSA-injected mice, which developed a progressive motor phenotype, G51D PD-inoculated animals remained free of overt neurological illness for up to 18 months post-inoculation. However, a subclinical synucleinopathy was present in G51D PD-inoculated mice, characterized by the accumulation of α-synuclein aggregates in restricted regions of the brain. The induced α-synuclein aggregates in G51D PD-injected mice exhibited distinct properties in a seed amplification assay and were much more stable than those present in mice injected with MSA extract, which mirrored the differences observed between human MSA and G51D PD brain samples. These results suggest that the G51D SNCA mutation specifies the formation of a slowly propagating α-synuclein strain that more closely resembles α-synuclein aggregates associated with PD than MSA.


Asunto(s)
Atrofia de Múltiples Sistemas , Enfermedad de Parkinson , Sinucleinopatías , Humanos , Ratones , Animales , alfa-Sinucleína/genética , alfa-Sinucleína/química , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Atrofia de Múltiples Sistemas/genética , Atrofia de Múltiples Sistemas/patología , Mutación/genética , Sinucleinopatías/genética , Ratones Transgénicos
9.
Sci Rep ; 13(1): 2337, 2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36759538

RESUMEN

The neuroendocrine peptide somatostatin (SST) has long been thought of as influencing the deposition of the amyloid ß peptide (Aß) in Alzheimer's disease (AD). Missing have been in vivo data in a relevant Aß amyloidosis model. Here we crossed AppNL-F/NL-F mice with Sst-deficient mice to assess if and how the presence of Sst influences pathological hallmarks of Aß amyloidosis. We found that Sst had no influence on whole brain neprilysin transcript, protein or activity levels, an observation that cannot be accounted for by a compensatory upregulation of the Sst paralog, cortistatin (Cort), that we observed in 15-month-old Sst-deficient mice. Sst-deficiency led to a subtle but significant increase in the density of cortical Aß amyloid plaques. Follow-on western blot analyses of whole brain extracts indicated that Sst interferes with early steps of Aß assembly that manifest in the appearance of SDS-stable smears of 55-150 kDa in Sst null brain samples. As expected, no effect of Sst on tau steady-state levels or its phosphorylation were observed. Results from this study are easier reconciled with an emerging body of data that point toward Sst affecting Aß amyloid plaque formation through direct interference with Aß aggregation rather than through its effects on neprilysin expression.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Ratones , Animales , Péptidos beta-Amiloides/metabolismo , Placa Amiloide/patología , Neprilisina/genética , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/patología , Somatostatina/metabolismo , Ratones Transgénicos , Modelos Animales de Enfermedad
10.
J Mol Biol ; 435(12): 168011, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-36792008

RESUMEN

The synucleinopathies, which include Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy, are a class of human neurodegenerative disorders unified by the presence of α-synuclein aggregates in the brain. Considerable clinical and pathological heterogeneity exists within and among the individual synucleinopathies. A potential explanation for this variability is the existence of distinct conformational strains of α-synuclein aggregates that cause different disease manifestations. Like prion strains, α-synuclein strains can be delineated based on their structural architecture, with structural differences among α-synuclein aggregates leading to unique biochemical attributes and neuropathological properties in humans and animal models. Bolstered by recent high-resolution structural data from patient brain-derived material, it has now been firmly established that there are conformational differences among α-synuclein aggregates from different human synucleinopathies. Moreover, recombinant α-synuclein can be polymerized into several structurally distinct aggregates that exhibit unique pathological properties. In this review, we outline the evidence supporting the existence of α-synuclein strains and highlight how they can act as drivers of phenotypic heterogeneity in the human synucleinopathies.


Asunto(s)
Sinucleinopatías , alfa-Sinucleína , Animales , Humanos , alfa-Sinucleína/química , Encéfalo/metabolismo , Atrofia de Múltiples Sistemas/genética , Atrofia de Múltiples Sistemas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Sinucleinopatías/metabolismo , Sinucleinopatías/patología , Conformación Proteica
11.
Cell Tissue Res ; 392(1): 63-80, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35581386

RESUMEN

For over three decades, cultured cells have been a useful tool for dissecting the molecular details of prion replication and the identification of candidate therapeutics for prion disease. A major issue limiting the translatability of these studies has been the inability to reliably propagate disease-relevant, non-mouse strains of prions in cells relevant to prion pathogenesis. In recent years, fueled by advances in gene editing technology, it has become possible to propagate prions from hamsters, cervids, and sheep in immortalized cell lines originating from the central nervous system. In particular, the use of CRISPR-Cas9-mediated gene editing to generate versions of prion-permissive cell lines that lack endogenous PrP expression has provided a blank canvas upon which re-expression of PrP leads to species-matched susceptibility to prion infection. When coupled with the ability to propagate prions in cells or organoids derived from stem cells, these next-generation cellular models should provide an ideal paradigm for identifying small molecules and other biological therapeutics capable of interfering with prion replication in animal and human prion disorders. In this review, we summarize recent advances that have widened the spectrum of prion strains that can be propagated in cultured cells and cutting-edge tissue-based models.


Asunto(s)
Ciervos , Enfermedades por Prión , Priones , Cricetinae , Animales , Humanos , Ovinos/genética , Priones/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Células Cultivadas , Línea Celular , Edición Génica , Ciervos/genética , Ciervos/metabolismo
12.
J Neurochem ; 165(2): 230-245, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36511154

RESUMEN

The bank vole (BV) prion protein (PrP) can function as a universal acceptor of prions. However, the molecular details of BVPrP's promiscuity for replicating a diverse range of prion strains remain obscure. To develop a cultured cell paradigm capable of interrogating the unique properties of BVPrP, we generated monoclonal lines of CAD5 cells lacking endogenous PrP but stably expressing either hamster (Ha), mouse (Mo), or BVPrP (M109 or I109 polymorphic variants) and then challenged them with various strains of mouse or hamster prions. Cells expressing BVPrP were susceptible to both mouse and hamster prions, whereas cells expressing MoPrP or HaPrP could only be infected with species-matched prions. Propagation of mouse and hamster prions in cells expressing BVPrP resulted in strain adaptation in several instances, as evidenced by alterations in conformational stability, glycosylation, susceptibility to anti-prion small molecules, and the inability of BVPrP-adapted mouse prion strains to infect cells expressing MoPrP. Interestingly, cells expressing BVPrP containing the G127V prion gene variant, identified in individuals resistant to kuru, were unable to become infected with prions. Moreover, the G127V polymorphic variant impeded the spontaneous aggregation of recombinant BVPrP. These results demonstrate that BVPrP can facilitate cross-species prion replication in cultured cells and that a single amino acid change can override the prion-permissive nature of BVPrP. This cellular paradigm will be useful for dissecting the molecular features of BVPrP that allow it to function as a universal prion acceptor.


Asunto(s)
Enfermedades por Prión , Priones , Cricetinae , Animales , Priones/genética , Priones/metabolismo , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Enfermedades por Prión/genética , Arvicolinae/genética , Arvicolinae/metabolismo , Células Cultivadas
13.
Transl Neurodegener ; 11(1): 7, 2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35125105

RESUMEN

BACKGROUND: Multiple system atrophy (MSA) is a neurodegenerative condition characterized by variable combinations of parkinsonism, autonomic failure, cerebellar ataxia and pyramidal features. Although the distribution of synucleinopathy correlates with the predominant clinical features, the burden of pathology does not fully explain observed differences in clinical presentation and rate of disease progression. We hypothesized that the clinical heterogeneity in MSA is a consequence of variability in the seeding activity of α-synuclein both between different patients and between different brain regions. METHODS: The reliable detection of α-synuclein seeding activity derived from MSA using cell-free amplification assays remains challenging. Therefore, we conducted a systematic evaluation of 168 different reaction buffers, using an array of pH and salts, seeded with fully characterized brain homogenates from one MSA and one PD patient. We then validated the two conditions that conferred the optimal ability to discriminate between PD- and MSA-derived samples in a larger cohort of 40 neuropathologically confirmed cases, including 15 MSA. Finally, in a subset of brains, we conducted the first multi-region analysis of seeding behaviour in MSA. RESULTS: Using our novel buffer conditions, we show that the physicochemical factors that govern the in vitro amplification of α-synuclein can be tailored to generate strain-specific reaction buffers that can be used to reliably study the seeding capacity from MSA-derived α-synuclein. Using this novel approach, we were able to sub-categorize the 15 MSA brains into 3 groups: high, intermediate and low seeders. To further demonstrate heterogeneity in α-synuclein seeding in MSA, we conducted a comprehensive multi-regional evaluation of α-synuclein seeding in 13 different regions from 2 high seeders, 2 intermediate seeders and 2 low seeders. CONCLUSIONS: We have identified unexpected differences in seed-competent α-synuclein across a cohort of neuropathologically comparable MSA brains. Furthermore, our work has revealed a substantial heterogeneity in seeding activity, driven by the PBS-soluble α-synuclein, between different brain regions of a given individual that goes beyond immunohistochemical observations. Our observations pave the way for future subclassification of MSA, which exceeds conventional clinical and neuropathological phenotyping and considers the structural and biochemical heterogeneity of α-synuclein present. Finally, our methods provide an experimental framework for the development of vitally needed, rapid and sensitive diagnostic assays for MSA.


Asunto(s)
Atrofia de Múltiples Sistemas , Trastornos Parkinsonianos , Sinucleinopatías , Encéfalo/metabolismo , Humanos , Atrofia de Múltiples Sistemas/diagnóstico , Atrofia de Múltiples Sistemas/patología , Trastornos Parkinsonianos/patología , Sinucleinopatías/diagnóstico , alfa-Sinucleína/metabolismo
14.
Brain Commun ; 3(4): fcab247, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34761222

RESUMEN

The accumulation of aggregated alpha-synuclein (α-syn) in Parkinson's disease, dementia with Lewy bodies and multiple system atrophy is thought to involve a common prion-like mechanism, whereby misfolded α-syn provides a conformational template for further accumulation of pathological α-syn. We tested whether silencing α-syn gene expression could reduce native non-aggregated α-syn substrate and thereby disrupt the propagation of pathological α-syn initiated by seeding with synucleinopathy-affected mouse brain homogenates. Unilateral intracerebral injections of adeno-associated virus serotype-1 encoding microRNA targeting the α-syn gene reduced the extent and severity of both the α-syn pathology and motor deficits. Importantly, a moderate 50% reduction in α-syn was sufficient to prevent the spread of α-syn pathology to distal brain regions. Our study combines behavioural, immunohistochemical and biochemical data that strongly support α-syn knockdown gene therapy for synucleinopathies.

15.
PLoS One ; 16(11): e0258682, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34847154

RESUMEN

The prion protein (PrP) is best known for its ability to cause fatal neurodegenerative diseases in humans and animals. Here, we revisited its molecular environment in the brain using a well-developed affinity-capture mass spectrometry workflow that offers robust relative quantitation. The analysis confirmed many previously reported interactions. It also pointed toward a profound enrichment of Na,K-ATPases (NKAs) in proximity to cellular PrP (PrPC). Follow-on work validated the interaction, demonstrated partial co-localization of the ATP1A1 and PrPC, and revealed that cells exposed to cardiac glycoside (CG) inhibitors of NKAs exhibit correlated changes to the steady-state levels of both proteins. Moreover, the presence of PrPC was observed to promote the ion uptake activity of NKAs in a human co-culture paradigm of differentiated neurons and glia cells, and in mouse neuroblastoma cells. Consistent with this finding, changes in the expression of 5'-nucleotidase that manifest in wild-type cells in response to CG exposure can also be observed in untreated PrPC-deficient cells. Finally, the endoproteolytic cleavage of the glial fibrillary acidic protein, a hallmark of late-stage prion disease, can also be induced by CGs, raising the prospect that a loss of NKA activity may contribute to the pathobiology of prion diseases.


Asunto(s)
Proteínas Priónicas/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , 5'-Nucleotidasa/metabolismo , Animales , Encéfalo/metabolismo , Calpaína/metabolismo , Glicósidos Cardíacos/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Ratones , Modelos Biológicos , Proteínas Priónicas/deficiencia , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/metabolismo , Reproducibilidad de los Resultados
16.
J Biol Chem ; 297(3): 101073, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34390689

RESUMEN

The study of prions and the discovery of candidate therapeutics for prion disease have been facilitated by the ability of prions to replicate in cultured cells. Paradigms in which prion proteins from different species are expressed in cells with low or no expression of endogenous prion protein (PrP) have expanded the range of prion strains that can be propagated. In these systems, cells stably expressing a PrP of interest are typically generated via coexpression of a selectable marker and treatment with an antibiotic. Here, we report the unexpected discovery that the aminoglycoside G418 (Geneticin) interferes with the ability of stably transfected cultured cells to become infected with prions. In G418-resistant lines of N2a or CAD5 cells, the presence of G418 reduced levels of protease-resistant PrP following challenge with the RML or 22L strains of mouse prions. G418 also interfered with the infection of cells expressing hamster PrP with the 263K strain of hamster prions. Interestingly, G418 had minimal to no effect on protease-resistant PrP levels in cells with established prion infection, arguing that G418 selectively interferes with de novo prion infection. As G418 treatment had no discernible effect on cellular PrP levels or its localization, this suggests that G418 may specifically target prion assemblies or processes involved in the earliest stages of prion infection.


Asunto(s)
Gentamicinas/farmacología , Proteínas Priónicas/efectos de los fármacos , Priones/antagonistas & inhibidores , Aminoglicósidos/metabolismo , Aminoglicósidos/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Gentamicinas/metabolismo , Ratones , Proteínas PrPC/efectos de los fármacos , Proteínas PrPC/metabolismo , Proteínas PrPSc/efectos de los fármacos , Proteínas PrPSc/metabolismo , Enfermedades por Prión/prevención & control , Proteínas Priónicas/metabolismo , Priones/metabolismo , Inhibidores de la Síntesis de la Proteína
17.
Acta Neuropathol Commun ; 9(1): 83, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33971978

RESUMEN

When injected into genetically modified mice, aggregates of the amyloid-ß (Aß) peptide from the brains of Alzheimer's disease (AD) patients or transgenic AD mouse models seed cerebral Aß deposition in a prion-like fashion. Within the brain, Aß exists as a pool of distinct C-terminal variants with lengths ranging from 37 to 43 amino acids, yet the relative contribution of individual C-terminal Aß variants to the seeding behavior of Aß aggregates remains unknown. Here, we have investigated the relative seeding activities of Aß aggregates composed exclusively of recombinant Aß38, Aß40, Aß42, or Aß43. Cerebral Aß42 levels were not increased in AppNL-F knock-in mice injected with Aß38 or Aß40 aggregates and were only increased in a subset of mice injected with Aß42 aggregates. In contrast, significant accumulation of Aß42 was observed in the brains of all mice inoculated with Aß43 aggregates, and the extent of Aß42 induction was comparable to that in mice injected with brain-derived Aß seeds. Mice inoculated with Aß43 aggregates exhibited a distinct pattern of cerebral Aß pathology compared to mice injected with brain-derived Aß aggregates, suggesting that recombinant Aß43 may polymerize into a unique strain. Our results indicate that aggregates containing longer Aß C-terminal variants are more potent inducers of cerebral Aß deposition and highlight the potential role of Aß43 seeds as a crucial factor in the initial stages of Aß pathology in AD.


Asunto(s)
Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Priones/genética , Priones/metabolismo , Agregado de Proteínas/fisiología , Animales , Secuencia de Bases , Encéfalo/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
18.
Sci Rep ; 11(1): 4058, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33603091

RESUMEN

Unlike variant Creutzfeldt-Jakob disease prions, sporadic Creutzfeldt-Jakob disease prions have been shown to be difficult to amplify in vitro by protein misfolding cyclic amplification (PMCA). We assessed PMCA of pathological prion protein (PrPTSE) from 14 human sCJD brain samples in 3 substrates: 2 from transgenic mice expressing human prion protein (PrP) with either methionine (M) or valine (V) at position 129, and 1 from bank voles. Brain extracts representing the 5 major clinicopathological sCJD subtypes (MM1/MV1, MM2, MV2, VV1, and VV2) all triggered seeded PrPTSE amplification during serial PMCA with strong seed- and substrate-dependence. Remarkably, bank vole PrP substrate allowed the propagation of all sCJD subtypes with preservation of the initial molecular PrPTSE type. In contrast, PMCA in human PrP substrates was accompanied by a PrPTSE molecular shift during heterologous (M/V129) PMCA reactions, with increased permissiveness of V129 PrP substrate to in vitro sCJD prion amplification compared to M129 PrP substrate. Combining PMCA amplification sensitivities with PrPTSE electrophoretic profiles obtained in the different substrates confirmed the classification of 4 distinct major sCJD prion strains (M1, M2, V1, and V2). Finally, the level of sensitivity required to detect VV2 sCJD prions in cerebrospinal fluid was achieved.


Asunto(s)
Síndrome de Creutzfeldt-Jakob/metabolismo , Priones/metabolismo , Animales , Arvicolinae/metabolismo , Humanos , Ratones , Ratones Transgénicos , Proteínas Priónicas/metabolismo , Pliegue de Proteína , Deficiencias en la Proteostasis/metabolismo
19.
Acta Neuropathol ; 142(1): 17-39, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32743745

RESUMEN

Reminiscent of the human prion diseases, there is considerable clinical and pathological variability in Alzheimer's disease, the most common human neurodegenerative condition. As in prion disorders, protein misfolding and aggregation is a hallmark feature of Alzheimer's disease, where the initiating event is thought to be the self-assembly of Aß peptide into aggregates that deposit in the central nervous system. Emerging evidence suggests that Aß, similar to the prion protein, can polymerize into a conformationally diverse spectrum of aggregate strains both in vitro and within the brain. Moreover, certain types of Aß aggregates exhibit key hallmarks of prion strains including divergent biochemical attributes and the ability to induce distinct pathological phenotypes when intracerebrally injected into mouse models. In this review, we discuss the evidence demonstrating that Aß can assemble into distinct strains of aggregates and how such strains may be primary drivers of the phenotypic heterogeneity in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Enfermedad de Alzheimer/psicología , Animales , Humanos , Fenotipo , Enfermedades por Prión , Proteínas Priónicas/genética
20.
Prog Mol Biol Transl Sci ; 175: 179-211, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32958232

RESUMEN

The transmission of prions between species is typically an inefficient process due to the species barrier, which represents incompatibility between prion seed and substrate molecules. Bank voles (Myodes glareolus) are an exception to this rule, as they are susceptible to a diverse range of prion strains from many different animal species. In particular, bank voles can be efficiently infected with most types of human prions and have played a critical role in validating variably protease-sensitive prionopathy (VPSPr) and certain forms of Gerstmann-Sträussler-Scheinker (GSS) disease as bona fide prion disorders rather than non-transmissible proteinopathies. The bank vole prion protein (BVPrP) confers a "universal prion acceptor" phenotype when expressed in mice and when used as a substrate for in vitro prion amplification assays, indicating that the unique prion transmission properties of bank voles are mediated by BVPrP. Over-expression of BVPrP in mice can also promote the spontaneous development of prion disease, indicating that BVPrP is intrinsically prone to both spontaneous and template-directed misfolding. Here, we discuss the utility of bank voles and BVPrP for prion research and how they have provided new tools for establishing rapid animal bioassays, modeling spontaneous prion disease, standardizing prion diagnostics, and understanding the molecular basis of the species barrier.


Asunto(s)
Arvicolinae/fisiología , Enfermedades por Prión/patología , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Enfermedades por Prión/transmisión , Priones/química , Priones/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA