Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mov Disord ; 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38946200

RESUMO

Various forms of Parkinson's disease, including its common sporadic form, are characterized by prominent α-synuclein (αSyn) aggregation in affected brain regions. However, the role of αSyn in the pathogenesis and evolution of the disease remains unclear, despite vast research efforts of more than a quarter century. A better understanding of the role of αSyn, either primary or secondary, is critical for developing disease-modifying therapies. Previous attempts to hone this research have been challenged by experimental limitations, but recent technological advances may facilitate progress. The Scientific Issues Committee of the International Parkinson and Movement Disorder Society (MDS) charged a panel of experts in the field to discuss current scientific priorities and identify research strategies with potential for a breakthrough. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.

2.
bioRxiv ; 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-39026799

RESUMO

Multiple system atrophy (MSA) and Parkinson's disease (PD) are caused by misfolded α-synuclein spreading throughout the central nervous system. While familial PD is linked to several point mutations in α-synuclein, there are no known mutations associated with MSA. Our previous work investigating differences in α-synuclein misfolding between the two disorders showed that the familial PD mutation E46K inhibits replication of MSA prions both in vitro and in vivo , providing key evidence to support the hypothesis that α-synuclein adopts unique strains in patients. Here, to further interrogate α-synuclein misfolding, we engineered a panel of cell lines harboring both PD-linked and novel mutations designed to identify key residues that facilitate α-synuclein misfolding in MSA. These data were paired with in silico analyses using Maestro software to predict the effect of each mutation on the ability of α-synuclein to misfold into one of the reported MSA cryo-electron microscopy conformations. In many cases, our modeling accurately identified mutations that facilitated or inhibited MSA replication. However, Maestro was occasionally unable to predict the effect of a mutation on MSA propagation in vitro , demonstrating the challenge of using computational tools to investigate intrinsically disordered proteins. Finally, we used our cellular models to determine the mechanism underlying the E46K-driven inhibition of MSA replication, finding that the E46/K80 salt bridge is necessary to support α-synuclein misfolding. Overall, our studies use a structure-based approach to investigate α-synuclein misfolding, resulting in the creation of a powerful panel of cell lines that can be used to interrogate MSA strain biology.

3.
J Parkinsons Dis ; 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-39031387

RESUMO

 Pre-formed fibrils (PFFs) made from recombinant α-synuclein are broadly used throughout the field in cellular and animal models of Parkinson's disease. However, their ability to successfully recapitulate disease biology is a controversial topic. In this article, two researchers debate this issue with Amanda Woerman taking the view that PFFs are a model of synucleinopathy but not Parkinson's disease, while Kelvin Luk defends their use as an important tool in the field.


Parkinson's disease patients develop accumulations of misfolded proteins in the brain called Lewy bodies. An important protein found in Lewy bodies is α-synuclein, which aggregates into large fibrils that are thought to have toxic effects on neurons. To study the effect of α-synuclein fibrils, many scientists use pre-formed fibrils (PFFs), which are α-synuclein fibrils made in the lab under a variety of conditions. In this article, two researchers will debate the use of PFFs as a model of Parkinson's disease. Amanda Woerman discusses the data suggesting why PFFs are an inadequate tool for understanding Parkinson's disease. Kelvin Luk discusses the data supporting the use of PFFs as a Parkinson's disease model.

4.
Trends Neurosci ; 47(7): 538-550, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38806297

RESUMO

Prion diseases are a group of neurodegenerative disorders caused by misfolding of proteins into pathogenic conformations that self-template to spread disease. Although this mechanism is largely associated with the prion protein (PrP) in classical prion diseases, a growing literature indicates that other proteins, including α-synuclein, rely on a similar disease mechanism. Notably, α-synuclein misfolds into distinct conformations, or strains, that cause discrete clinical disorders including multiple system atrophy (MSA) and Parkinson's disease (PD). Because the recognized similarities between PrP and α-synuclein are increasing, this review article draws from research on PrP to identify the host and strain factors that impact disease pathogenesis, predominantly in rodent models, and focuses on key considerations for future research on α-synuclein prions.


Assuntos
Doenças Priônicas , alfa-Sinucleína , alfa-Sinucleína/metabolismo , Animais , Humanos , Doenças Priônicas/metabolismo , Príons/metabolismo
6.
bioRxiv ; 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37502998

RESUMO

The recent discovery by cryo-electron microscopy that the neuropatho-logical hallmarks of different tauopathies, including Alzheimer's disease, corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP), are caused by unique misfolded conformations of the protein tau is among the most profound developments in neurodegenerative disease research. To capitalize on these discoveries for therapeutic development, one must achieve in vitro replication of tau fibrils that adopt the rep-resentative tauopathy disease folds - a grand challenge. To understand whether the commonly used, but imperfect, fragment of the tau pro-tein, K18, is capable of inducing specific protein folds, fibril seeds derived from CBD- and PSP-infected biosensor cells expressing K18, were used to achieve cell-free assembly of naïve, recombinant 4R tau into fibrils without the addition of any cofactors. Using Double Electron Electron Resonance (DEER) spectroscopy, we discovered that cell-passaged patho-logical seeds generate heterogeneous fibrils that are distinct between the CBD and PSP lysate-seeded fibrils, and are also unique from heparin-induced tau fibril populations. Moreover, the lysate-seeded fibrils contain a characteristic sub-population that resembles either the CBD or PSP disease fold, corresponding with the respective starting patient sam-ple. These findings indicate that CBD and PSP patient-derived fibrils retain strain properties after passaging through K18 reporter cells.

7.
PLoS Pathog ; 18(12): e1010956, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36454879

RESUMO

In multiple system atrophy (MSA), the α-synuclein protein misfolds into a self-templating prion conformation that spreads throughout the brain, leading to progressive neurodegeneration. While the E46K mutation in α-synuclein causes familial Parkinson's disease (PD), we previously discovered that this mutation blocks in vitro propagation of MSA prions. Recent studies by others indicate that α-synuclein adopts a misfolded conformation in MSA in which a Greek key motif is stabilized by an intramolecular salt bridge between residues E46 and K80. Hypothesizing that the E46K mutation impedes salt bridge formation and, therefore, exerts a selective pressure that can modulate α-synuclein strain propagation, we asked whether three distinct α-synuclein prion strains could propagate in TgM47+/- mice, which express human α-synuclein with the E46K mutation. Following intracranial injection of these strains, TgM47+/- mice were resistant to MSA prion transmission, whereas recombinant E46K preformed fibrils (PFFs) transmitted neurological disease to mice and induced the formation of phosphorylated α-synuclein neuropathology. In contrast, heterotypic seeding following wild-type (WT) PFF-inoculation resulted in preclinical α-synuclein prion propagation. Moreover, when we inoculated TgM20+/- mice, which express WT human α-synuclein, with E46K PFFs, we observed delayed transmission kinetics with an incomplete attack rate. These findings suggest that the E46K mutation constrains the number of α-synuclein prion conformations that can propagate in TgM47+/- mice, expanding our understanding of the selective pressures that impact α-synuclein prion replication.


Assuntos
Atrofia de Múltiplos Sistemas , Príons , Humanos , Camundongos , Animais , alfa-Sinucleína/genética , Príons/genética , Camundongos Transgênicos , Mutação
8.
Acta Neuropathol ; 144(4): 677-690, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36018376

RESUMO

In multiple system atrophy (MSA), the protein α-synuclein misfolds into a prion conformation that self-templates and causes progressive neurodegeneration. While many point mutations in the α-synuclein gene, SNCA, have been identified as the cause of heritable Parkinson's disease (PD), none have been identified as causing MSA. To examine whether MSA prions can transmit disease to mice expressing wild-type (WT) human α-synuclein, we inoculated transgenic (Tg) mice denoted TgM20+/- with brain homogenates prepared from six different deceased MSA patients. All six samples transmitted CNS disease to the mice, with an average incubation period of ~ 280 days. Interestingly, TgM20+/- female mice developed disease > 60 days earlier than their male counterparts. Brains from terminal mice contained phosphorylated α-synuclein throughout the hindbrain, consistent with the distribution of α-synuclein inclusions in MSA patients. In addition, using our α-syn-YFP cell lines, we detected α-synuclein prions in brain homogenates prepared from terminal mice that retained MSA strain properties. To our knowledge, the studies described here are the first to show that MSA prions transmit neurological disease to mice expressing WT SNCA and that the rate of transmission is sex dependent. By comparison, TgM20+/- mice inoculated with WT preformed fibrils (PFFs) developed severe neurological disease in ~ 210 days and exhibited robust α-synuclein neuropathology in both limbic regions and the hindbrain. Brain homogenates from these animals exhibited biological activities that are distinct from those found in MSA-inoculated mice when tested in the α-syn-YFP cell lines. Differences between brains from MSA-inoculated and WT PFF-inoculated mice potentially argue that α-synuclein prions from MSA patients are distinct from the PFF inocula and that PFFs do not replicate MSA strain biology.


Assuntos
Atrofia de Múltiplos Sistemas , Príons , Animais , Feminino , Humanos , Corpos de Inclusão/patologia , Masculino , Camundongos , Camundongos Transgênicos , Atrofia de Múltiplos Sistemas/patologia , Príons/genética , Príons/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
9.
Acta Neuropathol ; 143(3): 311-330, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35122113

RESUMO

Synucleinopathies are a group of clinically and neuropathologically distinct protein misfolding diseases caused by unique α-synuclein conformations, or strains. While multiple atomic resolution cryo-electron microscopy structures of α-synuclein fibrils are now deposited in Protein Data Bank, significant gaps in the biological consequences arising from each conformation have yet to be unraveled. Mutations in the α-synuclein gene (SNCA), cofactors, and the solvation environment contribute to the formation and maintenance of each disease-causing strain. This review highlights the impact of each of these factors on α-synuclein misfolding and discusses the implications of the resulting structural variability on therapeutic development.


Assuntos
Deficiências na Proteostase , Sinucleinopatias , Biologia , Microscopia Crioeletrônica , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
10.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35115402

RESUMO

The α-synuclein protein can adopt several different conformations that cause neurodegeneration. Different α-synuclein conformers cause at least three distinct α-synucleinopathies: multiple system atrophy (MSA), dementia with Lewy bodies (DLB), and Parkinson's disease (PD). In earlier studies, we transmitted MSA to transgenic (Tg) mice and cultured HEK cells both expressing mutant α-synuclein (A53T) but not to cells expressing α-synuclein (E46K). Now, we report that DLB is caused by a strain of α-synuclein prions that is distinct from MSA. Using cultured HEK cells expressing mutant α-synuclein (E46K), we found that DLB prions could be transmitted to these HEK cells. Our results argue that a third strain of α-synuclein prions likely causes PD, but further studies are needed to identify cells and/or Tg mice that express a mutant α-synuclein protein that is permissive for PD prion replication. Our findings suggest that other α-synuclein mutants should give further insights into α-synuclein prion replication, strain formation, and disease pathogenesis, all of which are likely required to discover effective drugs for the treatment of PD as well as the other α-synucleinopathies.


Assuntos
Demência/metabolismo , Doença por Corpos de Lewy/metabolismo , Atrofia de Múltiplos Sistemas/metabolismo , Príons/metabolismo , alfa-Sinucleína/metabolismo , Idoso , Linhagem Celular , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/metabolismo , Sinucleinopatias/metabolismo
11.
Neurobiol Dis ; 164: 105625, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35026401

RESUMO

In several neurodegenerative disorders, proteins that typically exhibit an α-helical structure misfold into an amyloid conformation rich in ß-sheet content. Through a self-templating mechanism, these amyloids are able to induce additional protein misfolding, facilitating their propagation throughout the central nervous system. This disease mechanism was originally identified for the prion protein (PrP), which misfolds into PrPSc in a number of disorders, including variant Creutzfeldt-Jakob disease (vCJD) and bovine spongiform encephalopathy (BSE). More recently, the prion mechanism of disease was expanded to include other proteins that rely on this self-templating mechanism to cause progressive degeneration, including α-synuclein misfolding in Parkinson's disease (PD). Several studies now suggest that PD patients can be subcategorized based on where in the body misfolded α-synuclein originates, either the brain or the gut, similar to patients developing sporadic CJD or vCJD. In this review, we discuss the human and animal model data indicating that α-synuclein and PrPSc misfolding originates in the gut in body-first PD and vCJD, and summarize the data identifying the role of the autonomic nervous system in the gut-brain axis of both diseases.


Assuntos
Encéfalo/metabolismo , Síndrome de Creutzfeldt-Jakob/metabolismo , Doença de Parkinson/metabolismo , Animais , Encéfalo/patologia , Síndrome de Creutzfeldt-Jakob/patologia , Modelos Animais de Doenças , Humanos , Neurônios/metabolismo , Neurônios/patologia , Doença de Parkinson/patologia , Proteínas Priônicas/metabolismo
12.
Acta Neuropathol Commun ; 9(1): 118, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34187585

RESUMO

Traumatic brain injury (TBI) constitutes one of the strongest environmental risk factors for several progressive neurodegenerative disorders of cognitive impairment and dementia that are characterized by the pathological accumulation of hyperphosphorylated tau (p-Tau). It has been questioned whether mouse closed-head TBI models can replicate human TBI-associated tauopathy. We conducted longitudinal histopathological characterization of a mouse closed head TBI model, with a focus on pathological features reported in human TBI-associated tauopathy. Male C57BL/6 J mice were subjected to once daily TBI for 5 consecutive days using a weight drop paradigm. Histological analyses (AT8, TDP-43, pTDP-43, NeuN, GFAP, Iba-1, MBP, SMI-312, Prussian blue, IgG, ßAPP, alpha-synuclein) were conducted at 1 week, 4 weeks, and 24 weeks after rTBI and compared to sham operated controls. We conducted a systematic review of the literature for mouse models of closed-head injury focusing on studies referencing tau protein assessment. At 1-week post rTBI, p-Tau accumulation was restricted to the corpus callosum and perivascular spaces adjacent to the superior longitudinal fissure. Progressive p-Tau accumulation was observed in the superficial layers of the cerebral cortex, as well as in mammillary bodies and cortical perivascular, subpial, and periventricular locations at 4 to 24 weeks after rTBI. Associated cortical histopathologies included microvascular injury, neuroaxonal rarefaction, astroglial and microglial activation, and cytoplasmatic localization of TDP-43 and pTDP-43. In our systematic review, less than 1% of mouse studies (25/3756) reported p-Tau using immunostaining, of which only 3 (0.08%) reported perivascular p-Tau, which is considered a defining feature of chronic traumatic encephalopathy. Commonly reported associated pathologies included neuronal loss (23%), axonal loss (43%), microglial activation and astrogliosis (50%, each), and beta amyloid deposition (29%). Our novel model, supported by systematic review of the literature, indicates progressive tau pathology after closed head murine TBI, highlighting the suitability of mouse models to replicate pertinent human histopathology.


Assuntos
Lesões Encefálicas Traumáticas/patologia , Encefalopatia Traumática Crônica/patologia , Modelos Animais de Doenças , Tauopatias/patologia , Animais , Traumatismos Cranianos Fechados , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
13.
Neuron ; 109(10): 1675-1691.e9, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-33848474

RESUMO

Tau aggregates contribute to neurodegenerative diseases, including frontotemporal dementia and Alzheimer's disease (AD). Although RNA promotes tau aggregation in vitro, whether tau aggregates in cells contain RNA is unknown. We demonstrate, in cell culture and mouse brains, that cytosolic and nuclear tau aggregates contain RNA with enrichment for small nuclear RNAs (snRNAs) and small nucleolar RNAs (snoRNAs). Nuclear tau aggregates colocalize with and alter the composition, dynamics, and organization of nuclear speckles, membraneless organelles involved in pre-mRNA splicing. Moreover, several nuclear speckle components, including SRRM2, mislocalize to cytosolic tau aggregates in cells, mouse brains, and brains of individuals with AD, frontotemporal dementia (FTD), and corticobasal degeneration (CBD). Consistent with these alterations, we observe that the presence of tau aggregates is sufficient to alter pre-mRNA splicing. This work identifies tau alteration of nuclear speckles as a feature of tau aggregation that may contribute to the pathology of tau aggregates.


Assuntos
Doença de Alzheimer/metabolismo , Núcleo Celular/metabolismo , RNA Nucleolar Pequeno/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Núcleo Celular/ultraestrutura , Citosol/metabolismo , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Transporte Proteico , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo
15.
Acta Neuropathol ; 142(1): 73-86, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-32440702

RESUMO

Synucleinopathies are a group of neurodegenerative disorders caused by the misfolding and self-templating of the protein α-synuclein, or the formation of α-synuclein prions. Each disorder differs by age of onset, presenting clinical symptoms, α-synuclein inclusion morphology, and neuropathological distribution. Explaining this disease-specific variability, the strain hypothesis postulates that each prion disease is encoded by a distinct conformation of the misfolded protein, and therefore, each synucleinopathy is caused by a unique α-synuclein structure. This review discusses the current data supporting the role of α-synuclein strains in disease heterogeneity. Several in vitro and in vivo models exist for evaluating strain behavior, however, as the focus of this article is to compare strains across synucleinopathy patients, our discussion predominantly focuses on the two models most commonly used for this purpose: the α-syn140*A53T-YFP cell line and the TgM83+/- mouse model. Here we define each strain based on biochemical stability, ability to propagate in α-syn140-YFP cell lines, and incubation period, inclusion morphology and distribution, and neurological signs in TgM83+/- mice.


Assuntos
Doenças Neurodegenerativas/genética , Sinucleinopatias/genética , alfa-Sinucleína/genética , Animais , Linhagem Celular , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos
16.
PLoS Pathog ; 16(2): e1008222, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32017806

RESUMO

Multiple system atrophy (MSA), a progressive neurodegenerative disease characterized by autonomic dysfunction and motor impairment, is caused by the self-templated misfolding of the protein α-synuclein. With no treatment currently available, we sought to characterize the spread of α-synuclein in a transgenic mouse model of MSA prion propagation to support drug discovery programs for synucleinopathies. Brain homogenates from MSA patient samples or mouse-passaged MSA were inoculated either by standard freehand injection or stereotactically into TgM83+/- mice, which express human α-synuclein with the A53T mutation. Following disease onset, brains from the mice were tested for biologically active α-synuclein prions using a cell-based assay and examined for α-synuclein neuropathology. Inoculation studies using homogenates prepared from brain regions lacking detectable α-synuclein neuropathology transmitted neurological disease to mice. Terminal animals contained similar concentrations of α-synuclein prions; however, a time-course study where mice were terminated every five days through disease progression revealed that the kinetics of α-synuclein prion replication in the mice were variable. Stereotactic inoculation into the thalamus reduced variability in disease onset in the mice, although incubation times were consistent with standard inoculations. Using human samples with and without neuropathological lesions, we observed that α-synuclein prion formation precedes neuropathology in the brain, suggesting that disease in patients is not limited to brain regions containing neuropathological lesions.


Assuntos
Encéfalo/metabolismo , Atrofia de Múltiplos Sistemas/metabolismo , Mutação Puntual , alfa-Sinucleína/metabolismo , Animais , Encéfalo/patologia , Feminino , Humanos , Cinética , Masculino , Camundongos , Camundongos Transgênicos , Atrofia de Múltiplos Sistemas/genética , Atrofia de Múltiplos Sistemas/patologia , Príons/genética , Príons/metabolismo , alfa-Sinucleína/genética
17.
Sci Rep ; 9(1): 16238, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31700063

RESUMO

Protein interactions of Tau are of interest in efforts to decipher pathogenesis in Alzheimer's disease, a subset of frontotemporal dementias, and other tauopathies. We CRISPR-Cas9 edited two human cell lines to generate broadly adaptable models for neurodegeneration research. We applied the system to inducibly express balanced levels of 3-repeat and 4-repeat wild-type or P301L mutant Tau. Following 12-h induction, quantitative mass spectrometry revealed the Parkinson's disease-causing protein DJ-1 and non-muscle myosins as Tau interactors whose binding to Tau was profoundly influenced by the presence or absence of the P301L mutation. The presence of wild-type Tau stabilized non-muscle myosins at higher steady-state levels. Strikingly, in human differentiated co-cultures of neuronal and glial cells, the preferential interaction of non-muscle myosins to wild-type Tau depended on myosin ATPase activity. Consistently, transgenic P301L Tau mice exhibited reduced phosphorylation of regulatory myosin light chains known to activate this ATPase. The direct link of Tau to non-muscle myosins corroborates independently proposed roles of Tau in maintaining dendritic spines and mitochondrial fission biology, two subcellular niches affected early in tauopathies.


Assuntos
Adenosina Trifosfatases/metabolismo , Sistemas CRISPR-Cas/genética , Engenharia Celular , Mutação , Neurônios/metabolismo , Mapeamento de Interação de Proteínas , Proteínas tau/metabolismo , Astrócitos/citologia , Técnicas de Cocultura , Humanos , Miosinas/metabolismo , Neurônios/citologia , Ligação Proteica , Proteínas tau/genética
18.
Sci Transl Med ; 11(490)2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31043574

RESUMO

The hallmarks of Alzheimer's disease (AD) are the accumulation of Aß plaques and neurofibrillary tangles composed of hyperphosphorylated tau. We developed sensitive cellular assays using human embryonic kidney-293T cells to quantify intracellular self-propagating conformers of Aß in brain samples from patients with AD or other neurodegenerative diseases. Postmortem brain tissue from patients with AD had measurable amounts of pathological Aß conformers. Individuals over 80 years of age had the lowest amounts of prion-like Aß and phosphorylated tau. Unexpectedly, the longevity-dependent decrease in self-propagating tau conformers occurred in spite of increasing amounts of total insoluble tau. When corrected for the abundance of insoluble tau, the ability of postmortem AD brain homogenates to induce misfolded tau in the cellular assays showed an exponential decrease with longevity, with a half-life of about one decade over the age range of 37 to 99 years. Thus, our findings demonstrate an inverse correlation between longevity in patients with AD and the abundance of pathological tau conformers. Our cellular assays can be applied to patient selection for clinical studies and the development of new drugs and diagnostics for AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Longevidade , Príons/metabolismo , Proteínas tau/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento , Doença de Alzheimer/complicações , Animais , Apolipoproteína E4/genética , Modelos Animais de Doenças , Genótipo , Gliose/complicações , Gliose/patologia , Células HEK293 , Humanos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fenótipo , Fosforilação , Placa Amiloide/complicações , Placa Amiloide/patologia , Isoformas de Proteínas/metabolismo
19.
Acta Neuropathol ; 137(3): 437-454, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30690664

RESUMO

Previously, we reported that intracranial inoculation of brain homogenate from multiple system atrophy (MSA) patient samples produces neurological disease in the transgenic (Tg) mouse model TgM83+/-, which uses the prion protein promoter to express human α-synuclein harboring the A53T mutation found in familial Parkinson's disease (PD). In our studies, we inoculated MSA and control patient samples into Tg mice constructed using a P1 artificial chromosome to express wild-type (WT), A30P, and A53T human α-synuclein on a mouse α-synuclein knockout background [Tg(SNCA+/+)Nbm, Tg(SNCA*A30P+/+)Nbm, and Tg(SNCA*A53T+/+)Nbm]. In contrast to studies using TgM83+/- mice, motor deficits were not observed by 330-400 days in any of the Tg(SNCA)Nbm mice after inoculation with MSA brain homogenates. However, using a cell-based bioassay to measure α-synuclein prions, we found brain homogenates from Tg(SNCA*A53T+/+)Nbm mice inoculated with MSA patient samples contained α-synuclein prions, whereas control mice did not. Moreover, these α-synuclein aggregates retained the biological and biochemical characteristics of the α-synuclein prions in MSA patient samples. Intriguingly, Tg(SNCA*A53T+/+)Nbm mice developed α-synuclein pathology in neurons and astrocytes throughout the limbic system. This finding is in contrast to MSA-inoculated TgM83+/- mice, which develop exclusively neuronal α-synuclein aggregates in the hindbrain that cause motor deficits with advanced disease. In a crossover experiment, we inoculated TgM83+/- mice with brain homogenate from two MSA patient samples or one control sample first inoculated, or passaged, in Tg(SNCA*A53T+/+)Nbm animals. Additionally, we performed the reverse experiment by inoculating Tg(SNCA*A53T+/+)Nbm mice with brain homogenate from the same two MSA samples and one control sample first passaged in TgM83+/- animals. The TgM83+/- mice inoculated with mouse-passaged MSA developed motor dysfunction and α-synuclein prions, whereas the mouse-passaged control sample had no effect. Similarly, the mouse-passaged MSA samples induced α-synuclein prion formation in Tg(SNCA*A53T+/+)Nbm mice, but the mouse-passaged control sample did not. The confirmed transmission of α-synuclein prions to a second synucleinopathy model and the ability to propagate prions between two distinct mouse lines while retaining strain-specific properties provides compelling evidence that MSA is a prion disease.


Assuntos
Atrofia de Múltiplos Sistemas/patologia , Doenças Priônicas/patologia , Doenças Priônicas/transmissão , Príons/metabolismo , alfa-Sinucleína/metabolismo , Animais , Humanos , Camundongos , Camundongos Transgênicos
20.
Artigo em Inglês | MEDLINE | ID: mdl-28213437

RESUMO

Multiple system atrophy (MSA) is a rapidly progressive neurodegenerative disease arising from the misfolding and accumulation of the protein α-synuclein in oligodendrocytes, where it forms glial cytoplasmic inclusions (GCIs). Several years of studying synthetic α-synuclein fibrils has provided critical insight into the ability of α-synuclein to template endogenous protein misfolding, giving rise to fibrillar structures capable of propagating from cell to cell. However, more recent studies with MSA-derived α-synuclein aggregates have shown that they have a similar ability to undergo template-directed propagation, like PrP prions. Almost 20 years after α-synuclein was discovered as the primary component of GCIs, α-synuclein aggregates isolated from MSA patient samples were shown to infect cultured mammalian cells and also to transmit neurological disease to transgenic mice. These findings argue that α-synuclein becomes a prion in MSA patients. In this review, we discuss the in vitro and in vivo data supporting the recent classification of MSA as a prion disease.


Assuntos
Atrofia de Múltiplos Sistemas/etiologia , Doenças Priônicas/complicações , Príons/fisiologia , alfa-Sinucleína/fisiologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Doenças Priônicas/transmissão , Príons/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...