Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
3.
Mol Neurodegener ; 16(1): 63, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34503546

RESUMO

BACKGROUND: The misfolding of host-encoded proteins into pathological prion conformations is a defining characteristic of many neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, and Lewy body dementia. A current area of intense study is the way in which the pathological deposition of these proteins might influence each other, as various combinations of co-pathology between prion-capable proteins are associated with exacerbation of disease. A spectrum of pathological, genetic and biochemical evidence provides credence to the notion that amyloid ß (Aß) accumulation can induce and promote α-synuclein pathology, driving neurodegeneration. METHODS: To assess the interplay between α-synuclein and Aß on protein aggregation kinetics, we crossed mice expressing human α-synuclein (M20) with APPswe/PS1dE9 transgenic mice (L85) to generate M20/L85 mice. We then injected α-synuclein preformed fibrils (PFFs) unilaterally into the hippocampus of 6-month-old mice, harvesting 2 or 4 months later. RESULTS: Immunohistochemical analysis of M20/L85 mice revealed that pre-existing Aß plaques exacerbate the spread and deposition of induced α-synuclein pathology. This process was associated with increased neuroinflammation. Unexpectedly, the injection of α-synuclein PFFs in L85 mice enhanced the deposition of Aß; whereas the level of Aß deposition in M20/L85 bigenic mice, injected with α-synuclein PFFs, did not differ from that of mice injected with PBS. CONCLUSIONS: These studies reveal novel and unexpected interplays between α-synuclein pathology, Aß and neuroinflammation in mice that recapitulate the pathology of Alzheimer's disease and Lewy body dementia.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Demência/metabolismo , Modelos Animais de Doenças , Agregação Patológica de Proteínas , alfa-Sinucleína/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Cruzamentos Genéticos , Demência/patologia , Gliose/metabolismo , Gliose/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Injeções , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doenças Neuroinflamatórias , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Príons/química , Agregados Proteicos , Proteínas Recombinantes/metabolismo , alfa-Sinucleína/toxicidade
4.
Neurosci Lett ; 732: 135017, 2020 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-32371157

RESUMO

Pathologic intracellular inclusions formed from polymers of misfolded α-synuclein (αsyn) protein define a group of neurodegenerative diseases termed synucleinopathies which includes Parkinson's disease (PD). Prion-like recruitment of endogenous cellular αsyn has been demonstrated to occur in animal models of synucleinopathy, whereby misfolded αsyn can induce further pathologic αsyn inclusions to form through a prion-like mechanism. It has been suggested that misfolded αsyn may assume differing conformations which lead to varied clinical and pathological manifestations of disease; this phenomenon bears similarities to that of prion strains whereby the same misfolded protein can produce unique diseases. It is unclear what factors influence the development of unique αsyn strains, however post-translational modifications (PTMs) such as phosphorylation and truncation that are present in misfolded αsyn in disease may play a role due to their modulation of biochemical and structural αsyn properties. Herein, we investigate the prion-like properties of misfolded αsyn polymers containing either phosphomimetic (S129E) αsyn, 5 different major carboxy (C)-truncated forms of αsyn (1-115, 1-119, 1-122, 1-125, and 1-129 αsyn), or a mixture of these PTM containing αsyn forms compared to full-length (FL) αsyn in HEK293T cells and M83 transgenic mice overexpressing A53T αsyn. It is demonstrated that upon peripheral intramuscular injection of these C-truncated or S129E αsyn polymers into M83 mice, prion-like progression and time to disease onset in this mouse model is elongated when any of these PTMs are present, demonstrating that common modifications to the C-terminus of αsyn present in disease modulates the prion-like seeding properties of αsyn.


Assuntos
Sinucleinopatias/metabolismo , alfa-Sinucleína/metabolismo , Animais , Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Neurônios , Doença de Parkinson/metabolismo , Fosforilação , Príons , Processamento de Proteína Pós-Traducional , Análise de Sobrevida , Sinucleinopatias/patologia
5.
Acta Neuropathol Commun ; 7(1): 142, 2019 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-31477175

RESUMO

The protein α-synuclein (αsyn) forms pathologic aggregates in a number of neurodegenerative diseases including Lewy body dementia (LBD) and Parkinson's disease (PD). It is unclear why diseases such as LBD may develop widespread αsyn pathology, while in Alzheimer's disease with amygdala restricted Lewy bodies (AD/ALB) the αsyn aggregates remain localized. The amygdala contains αsyn aggregates in both LBD and in AD/ALB; to understand why αsyn pathology continues to progress in LBD but not in AD/ALB, tissue from the amygdala and other regions were obtained from 14 cases of LBD, 9 cases of AD/ALB, and 4 controls for immunohistochemical and biochemical characterization. Utilizing a panel of previously characterized αsyn antibodies, numerous unique pathologies differentiating LBD and AD/ALB were revealed; particularly the presence of dense neuropil αsyn aggregates, astrocytic αsyn, and αsyn-containing dystrophic neurites within senile plaques. Within LBD, these unique pathologies were predominantly present within the amygdala. Biochemically, the amygdala in LBD prominently contained specific carboxy-truncated forms of αsyn which are highly prone to aggregate, suggesting that the amygdala may be prone to initiate development of αsyn pathology. Similar to carboxy-truncated αsyn, it was demonstrated herein that the presence of aggregation prone A53T αsyn is sufficient to drive misfolding of wild-type αsyn in human disease. Overall, this study identifies within the amygdala in LBD the presence of unique strain-like variation in αsyn pathology that may be a determinant of disease progression.


Assuntos
Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/patologia , alfa-Sinucleína/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Progressão da Doença , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Camundongos Transgênicos , Pessoa de Meia-Idade , Neurônios/metabolismo , Neurônios/patologia
6.
Acta Neuropathol Commun ; 7(1): 80, 2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-31109378

RESUMO

α-synuclein (αS) is the major component of several types of brain pathological inclusions that define neurodegenerative diseases termed synucleinopathies. Central nervous system (CNS) inoculation studies using either in vitro polymerized αS fibrils or in vivo derived lysates containing αS aggregates to induce the progressive spread of αS inclusion pathology in animal disease models have supported the notion that αS mediated progressive neurodegeneration can occur by a prion-like mechanism. We have previously shown that neonatal brain inoculation with preformed αS fibrils in hemizygous M20+/- transgenic mice expressing wild type human αS and to a lesser extent in non-transgenic mice can result in a concentration-dependent progressive induction of CNS αS pathology. Recent studies using brain lysates from patients with multiple system atrophy (MSA), characterized by αS inclusion pathology in oligodendrocytes, indicate that these may be uniquely potent at inducing αS pathology with prion-like strain specificity. We demonstrate here that brain lysates from MSA patients, but not control individuals, can induce αS pathology following neonatal brain inoculation in transgenic mice expressing A53T human αS (M83 line), but not in transgenic expressing wild type human αS (M20 line) or non-transgenic mice within the timeframe of the study design. Further, we show that neuroanatomical and immunohistochemical properties of the pathology induced by MSA brain lysates is very similar to what is produced by the neonatal brain injection of preformed human αS fibrils in hemizygous M83+/- transgenic mice. Collectively, these findings reinforce the idea that the intrinsic traits of the M83 mouse model dominates over any putative prion-like strain properties of MSA αS seeds that can induce pathology.


Assuntos
Encéfalo/patologia , Atrofia de Múltiplos Sistemas/patologia , Sinucleinopatias/patologia , alfa-Sinucleína/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Pessoa de Meia-Idade , Atrofia de Múltiplos Sistemas/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Sinucleinopatias/metabolismo
7.
Lab Invest ; 99(7): 982-992, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30737468

RESUMO

Synucleinopathies are a group of neurodegenerative diseases characterized by the accumulation of insoluble, aggregated α-synuclein (αS) pathological inclusions. Multiple system atrophy (MSA) presents with extensive oligodendroglial αS pathology and additional more limited neuronal inclusions while most of the other synucleinopathies, such as Parkinson's disease and dementia with Lewy bodies (DLB), develop αS pathology primarily in neuronal cell populations. αS biochemical alterations specific to MSA have been described but thorough examination of these unique and disease-specific protein deposits is further warranted especially given recent findings implicating the prion-like nature of synucleinopathies perhaps with distinct strain-like properties. Taking advantage of an extensive panel of antibodies that target a wide range of epitopes within αS, we investigated the distinct properties of the various types of αS inclusion present in MSA brains with comparison to DLB. Brain biochemical fractionation followed by immunoblotting revealed that the immunoreactive profiles were significantly more consistent for DLB than for MSA. Furthermore, epitope-specific immunohistochemistry varied greatly between different types of MSA αS inclusions and even within different brain regions of individual MSA brains. These studies highlight the importance of using a battery of antibodies for adequate appreciation of the various pathology in this distinct synucleinopathy. In addition, it can be posited that if the spread of pathology in MSA undergoes prion-like mechanisms, "strains" of αS aggregated conformers must be inherently unstable and readily mutable, perhaps resulting in a more stochastic progression process.


Assuntos
Corpos de Inclusão/metabolismo , Atrofia de Múltiplos Sistemas/metabolismo , alfa-Sinucleína/metabolismo , Encéfalo/patologia , Humanos , Atrofia de Múltiplos Sistemas/patologia
8.
Neurosci Lett ; 661: 114-120, 2017 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-28964772

RESUMO

Neurofilaments are a major component of the axonal cytoskeleton in neurons and have been implicated in a number of neurodegenerative diseases due to their presence within characteristic pathological inclusions. Their contributions to these diseases are not yet fully understood, but previous studies investigated the effects of ablating the obligate subunit of neurofilaments, low molecular mass neurofilament subunit (NFL), on disease phenotypes in transgenic mouse models of Alzheimer's disease and tauopathy. Here, we tested the effects of ablating NFL in α-synuclein M83 transgenic mice expressing the human pathogenic A53T mutation, by breeding them onto an NFL null background. The induction and spread of α-synuclein inclusion pathology was triggered by the injection of preformed α-synuclein fibrils into the gastrocnemius muscle or hippocampus in M83 versus M83/NFL null mice. We observed no difference in the post-injection time to motor-impairment and paralysis endpoint or amount and distribution of α-synuclein inclusion pathology in the muscle injected M83 and M83/NFL null mice. Hippocampal injected M83/NFL null mice displayed subtle region-specific differences in the amount of α-synuclein inclusions however, pathology was observed in the same regions as the M83 mice. Overall, we observed only minor differences in the induction and transmission of α-synuclein pathology in these induced models of synucleinopathy in the presence or absence of NFL. This suggests that NFL and neurofilaments do not play a major role in influencing the induction and transmission of α-synuclein aggregation.


Assuntos
Filamentos Intermediários/metabolismo , Neurônios/metabolismo , Príons/metabolismo , alfa-Sinucleína/metabolismo , Animais , Encéfalo/metabolismo , Corpos de Inclusão/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Doenças Neurodegenerativas/patologia
9.
Hum Mol Genet ; 26(24): 4906-4915, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29036344

RESUMO

Parkinson's disease (PD) is one of many neurodegenerative diseases termed synucleinopathies, neuropathologically defined by inclusions containing aggregated α-synuclein (αS). αS gene (SNCA) mutations can directly cause autosomal dominant PD. In vitro studies demonstrated that SNCA missense mutations may either enhance or diminish αS aggregation but cross-seeding of mutant and wild-type αS proteins appear to reduce aggregation efficiency. Here, we extended these studies by assessing the effects of seeded αS aggregation in αS transgenic mice through intracerebral or peripheral injection of various mutant αS fibrils. We observed modestly decreased time to paralysis in mice transgenic for human A53T αS (line M83) intramuscularly injected with H50Q, G51D or A53E αS fibrils relative to wild-type αS fibrils. Conversely, E46K αS fibril seeding was significantly delayed and less efficient in the same experimental paradigm. However, the amount and distribution of αS inclusions in the central nervous system were similar for all αS fibril muscle injected mice that developed paralysis. Mice transgenic for human αS (line M20) injected in the hippocampus with wild-type, H50Q, G51D or A53E αS fibrils displayed induction of αS inclusion pathology that increased and spread over time. By comparison, induction of αS aggregation following the intrahippocampal injection of E46K αS fibrils in M20 mice was much less efficient. These findings show that H50Q, G51D or A53E can efficiently cross-seed and induce αS pathology in vivo. In contrast, E46K αS fibrils are intrinsically inefficient at seeding αS inclusion pathology. Consistent with previous in vitro studies, E46K αS polymers are likely distinct aggregated conformers that may represent a unique prion-like strain of αS.


Assuntos
Doença de Parkinson/genética , alfa-Sinucleína/metabolismo , Animais , Sistema Nervoso Central/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Humanos , Corpos de Inclusão/metabolismo , Camundongos , Camundongos Transgênicos , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/fisiologia
10.
PLoS One ; 12(9): e0184731, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28910367

RESUMO

Synucleinopathies are a spectrum of neurodegenerative diseases characterized by the intracellular deposition of the protein α-synuclein leading to multiple outcomes, including dementia and Parkinsonism. Recent findings support the notion that across the spectrum of synucleinopathies there exist diverse but specific biochemical modifications and/or structural conformations of α-synuclein, which would give rise to protein strain specific prion-like intercellular transmission, a proposed model that could explain synucleinopathies disease progression. Herein, we characterized a panel of antibodies with epitopes within both the C- and N- termini of α-synuclein. A comprehensive analysis of human pathological tissue and mouse models of synucleinopathy with these antibodies support the notion that α-synuclein exists in distinct modified forms and/or structural variants. Furthermore, these well-characterized and specific tools allow the investigation of biochemical changes associated with α-synuclein inclusion formation. We have identified several antibodies of interest with diverse staining and epitope properties that will prove useful in future investigations of strain specific disease progression and the development of targeted immunotherapeutic approaches to synucleinopathies.


Assuntos
Anticorpos/análise , Doença por Corpos de Lewy/metabolismo , alfa-Sinucleína/química , Animais , Modelos Animais de Doenças , Epitopos/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , alfa-Sinucleína/genética , alfa-Sinucleína/imunologia
11.
Mol Ther Oncolytics ; 4: 31-40, 2017 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-28345022

RESUMO

Allogeneic stem cell transplant-derived T cells have the potential to seek and eliminate sites of residual cancer that escaped primary therapy. Oncolytic myxoma virus (MYXV) exhibits potent anti-cancer efficacy against human cancers like multiple myeloma (MM) and can arm transplant-derived T cells to become more effective cancer killers in vitro and in an immunodeficient xenotransplant murine model. Here, we tested ex vivo MYXV virotherapy against residual murine MM in immunocompetent mice using an allogeneic mouse-mouse model. In contrast to all human MM cell lines previously tested, the murine MM cell line tested here was highly resistant to direct MYXV infection and oncolysis in vitro. Despite this in vitro resistance, we found that ex vivo MYXV-armed allogeneic bone marrow (BM) transplantation dramatically ablated pre-seeded residual MM in vivo. Unexpectedly, we show that both neutrophils and activated T cells from the donor function as virus-armed carrier cells, and MYXV-preloaded cells enhanced MM killing. Our results demonstrate a novel therapeutic paradigm for residual cancer, in which multiple classes of allotransplant leukocytes can be armed by MYXV ex vivo to enhance the graft-versus-tumor effects.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA