Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Acta Neuropathol Commun ; 12(1): 91, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38858742

ABSTRACT

Synucleinopathies are a group of neurodegenerative disorders characterized by the presence of misfolded α-Synuclein (αSyn) in the brain. These conditions manifest with diverse clinical and pathophysiological characteristics. This disease diversity is hypothesized to be driven by αSyn strains with differing biophysical properties, potentially influencing prion-type propagation and consequentially the progression of illness. Previously, we investigated this hypothesis by injecting brain lysate (seeds) from deceased individuals with various synucleinopathies or human recombinant αSyn preformed fibrils (PFFs) into transgenic mice overexpressing either wild type or A53T human αSyn. In the studies herein, we expanded on these experiments, utilizing a panel of antibodies specific for the major carboxyl-terminally truncated forms of αSyn (αSynΔC). These modified forms of αSyn are found enriched in human disease brains to inform on potential strain-specific proteolytic patterns. With monoclonal antibodies specific for human αSyn cleaved at residues 103, 114, 122, 125, and 129, we demonstrate that multiple system atrophy (MSA) seeds and PFFs induce differing neuroanatomical spread of αSyn pathology associated with host specific profiles. Overall, αSyn cleaved at residue 103 was most widely present in the induced pathological inclusions. Furthermore, αSynΔC-positive inclusions were present in astrocytes, but more frequently in activated microglia, with patterns dependent on host and inoculum. These findings support the hypothesis that synucleinopathy heterogeneity might stem from αSyn strains with unique biochemical properties that include proteolytic processing, which could result in dominant strain properties.


Subject(s)
Brain , Disease Models, Animal , Mice, Transgenic , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/immunology , Animals , Humans , Mice , Brain/pathology , Brain/metabolism , Synucleinopathies/pathology , Synucleinopathies/metabolism , Synucleinopathies/immunology , Antibodies, Monoclonal , Multiple System Atrophy/pathology , Multiple System Atrophy/immunology , Multiple System Atrophy/metabolism , Prions/immunology , Prions/metabolism , Female
3.
Int J Mol Sci ; 24(18)2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37761979

ABSTRACT

Tau proteins within the adult central nervous system (CNS) are found to be abnormally aggregated into heterogeneous filaments in neurodegenerative diseases, termed tauopathies. These tau inclusions are pathological hallmarks of Alzheimer's disease (AD), Pick's disease (PiD), corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP). The neuropathological hallmarks of these diseases burden several cell types within the CNS, and have also been shown to be abundantly phosphorylated. The mechanism(s) by which tau aggregates in the CNS is not fully known, but it is hypothesized that hyperphosphorylated tau may precede and further promote filament formation, leading to the production of these pathological inclusions. In the studies herein, we generated and thoroughly characterized two novel conformation-dependent tau monoclonal antibodies that bind to residues Pro218-Glu222, but are sensitive to denaturing conditions and highly modulated by adjacent downstream phosphorylation sites. These epitopes are present in the neuropathological hallmarks of several tauopathies, including AD, PiD, CBD, and PSP. These novel antibodies will further enable investigation of tau-dependent pathological inclusion formation and enhance our understanding of the phosphorylation signatures within tauopathies with the possibility of new biomarker developments.


Subject(s)
Alzheimer Disease , Pick Disease of the Brain , Tauopathies , Adult , Humans , Phosphorylation , Antibodies, Monoclonal , Central Nervous System
4.
Acta Neuropathol Commun ; 11(1): 119, 2023 07 23.
Article in English | MEDLINE | ID: mdl-37482615

ABSTRACT

Post-translational modifications to the carboxyl (C) terminus domain of α-synuclein can play an important role in promoting the pathologic aggregation of α-synuclein. Various cleavages that diminish this highly charged, proline-rich region can result in exposure of hydrophobic, aggregation-prone regions, thereby accelerating the aggregation kinetics of α-synuclein into misfolded, pathologic forms. C-terminally truncated forms of α-synuclein are abundant in human diseased brains compared to controls, suggesting a role in disease pathogenesis. Factors that alter the homeostatic proteolytic processing of α-synuclein may ultimately tip the balance towards a progressive disease state. Apolipoprotein E (APOE) has been implicated in the acceleration of cognitive impairment in patients with Lewy body diseases. The APOE4 isoform has been found to cause dysregulation in the endosomal-lysosomal pathway, which could result in altered α-synuclein degradation as a potential mechanism for promoting its pathologic misfolding. Herein, we investigate the spatiotemporal accumulation of C-terminally truncated α-synuclein in a seeded and progressive mouse model of synucleinopathy. Furthermore, we study how this process is influenced in the context of mice that are altered to express either the human APOE3 or APOE4 isoforms. We found that specific C-terminal truncation of α-synuclein occurs at early stages of pathogenesis. We also found that proteolytic processing of this domain differs across various brain regions and is influenced by the presence of different human APOE isoforms. Our data demonstrate an early pathogenic role for C-terminally truncated α-synuclein, and highlight the influence of APOE isoforms in modulating its impact.


Subject(s)
Apolipoprotein E4 , alpha-Synuclein , Animals , Humans , Mice , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Apolipoproteins E/metabolism , Carrier Proteins , Mice, Transgenic , Protein Isoforms/genetics , Protein Isoforms/metabolism
5.
J Biol Chem ; 299(7): 104912, 2023 07.
Article in English | MEDLINE | ID: mdl-37307916

ABSTRACT

α-synuclein (αS) is an abundant, neuronal protein that assembles into fibrillar pathological inclusions in a spectrum of neurodegenerative diseases that include Lewy body diseases (LBD) and Multiple System Atrophy (MSA). The cellular and regional distributions of pathological inclusions vary widely between different synucleinopathies contributing to the spectrum of clinical presentations. Extensive cleavage within the carboxy (C)-terminal region of αS is associated with inclusion formation, although the events leading to these modifications and the implications for pathobiology are of ongoing study. αS preformed fibrils can induce prion-like spread of αS pathology in both in vitro and animal models of disease. Using C truncation-specific antibodies, we demonstrated here that prion-like cellular uptake and processing of αS preformed fibrils resulted in two major cleavages at residues 103 and 114. A third cleavage product (122 αS) accumulated upon application of lysosomal protease inhibitors. In vitro, both 1-103 and 1-114 αS polymerized rapidly and extensively in isolation and in the presence of full-length αS. 1-103 αS also demonstrated more extensive aggregation when expressed in cultured cells. Furthermore, we used novel antibodies to αS cleaved at residue Glu114, to assess x-114 αS pathology in postmortem brain tissue from patients with LBD and MSA, as well as three different transgenic αS mouse models of prion-like induction. The distribution of x-114 αS pathology was distinct from that of overall αS pathology. These studies reveal the cellular formation and behavior of αS C-truncated at residues 114 and 103 as well as the disease dependent distribution of x-114 αS pathology.


Subject(s)
Lewy Body Disease , Multiple System Atrophy , alpha-Synuclein , Animals , Mice , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Mice, Transgenic , Multiple System Atrophy/metabolism , Multiple System Atrophy/pathology , Prions/chemistry , Prions/metabolism , Humans , Lysosomes/enzymology , Protease Inhibitors , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Autopsy , Glutamic Acid/metabolism
7.
Acta Neuropathol Commun ; 10(1): 94, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35765058

ABSTRACT

Tau is a predominantly neuronal, soluble and natively unfolded protein that can bind and stabilize microtubules in the central nervous system. Tau has been extensively studied over several decades, especially in the context of neurodegenerative diseases where it can aberrantly aggregate to form a spectrum of pathological inclusions. The presence of tau inclusions in the form of neurofibrillary tangles, neuropil threads and dystrophic neurites within senile plaques are essential and defining features of Alzheimer's disease. The current dogma favors the notion that tau is predominantly an axonal protein, and that in Alzheimer's disease there is a redistribution of tau towards the neuronal soma that is associated with the formation of pathological inclusions such as neurofibrillary tangles and neuropil threads. Using novel as well as previously established highly specific tau antibodies, we demonstrate that contrary to this overwhelmingly accepted fact, as asserted in numerous articles and reviews, in adult human brain, tau is more abundant in cortical gray matter that is enriched in neuronal soma and dendrites compared to white matter that is predominantly rich in neuronal axons. Additionally, in Alzheimer's disease tau pathology is significantly more abundant in the brain cortical gray matter of affected brain regions compared to the adjacent white matter regions. These findings have important implications for the biological function of tau as well as the mechanisms involved in the progressive spread of tau associated with the insidious nature of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Adult , Alzheimer Disease/pathology , Brain/pathology , Humans , Neurofibrillary Tangles/pathology , Neurons/metabolism , tau Proteins/metabolism
8.
Commun Biol ; 5(1): 446, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35550593

ABSTRACT

Pathological tau inclusions are neuropathologic hallmarks of many neurodegenerative diseases. We generated and characterized a transgenic mouse model expressing pathogenic human tau with S320F and P301S aggregating mutations (SPAM) at transgene levels below endogenous mouse tau protein levels. This mouse model develops a predictable temporal progression of tau pathology in the brain with biochemical and ultrastructural properties akin to authentic tau inclusions. Surprisingly, pathogenic human tau extensively recruited endogenous mouse tau into insoluble aggregates. Despite the early onset and rapid progressive nature of tau pathology, major neuroinflammatory and transcriptional changes were only detectable at later time points. Moreover, tau SPAM mice are the first model to develop loss of enteric neurons due to tau accumulation resulting in a lethal phenotype. With moderate transgene expression, rapidly progressing tau pathology, and a highly predictable lethal phenotype, the tau SPAM model reveals new associations of tau neurotoxicity in the brain and intestinal tract.


Subject(s)
Brain , tau Proteins , Animals , Brain/metabolism , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Mutation , Neurons/metabolism , tau Proteins/genetics , tau Proteins/metabolism
9.
Acta Neuropathol ; 143(6): 663-685, 2022 06.
Article in English | MEDLINE | ID: mdl-35488930

ABSTRACT

α-synuclein (αSyn) is an intrinsically disordered protein which can undergo structural transformations, resulting in the formation of stable, insoluble fibrils. αSyn amyloid-type nucleation can be induced by misfolded 'seeds' serving as a conformational template, tantamount to the prion-like mechanism. Accumulation of αSyn inclusions is a key feature of dementia with Lewy bodies (DLB) and multiple system atrophy (MSA), and are found as additional pathology in Alzheimer's disease (AD) such as AD with amygdala predominant Lewy bodies (AD/ALB). While these disorders accumulate the same pathological protein, they exhibit heterogeneity in clinical and histological features; however, the mechanism(s) underlying this variability remains elusive. Accruing data from human autopsy studies, animal inoculation modeling, and in vitro characterization experiments, have lent credence to the hypothesis that conformational polymorphism of the αSyn amyloid-type fibril structure results in distinct "strains" with categorical infectivity traits. Herein, we directly compare the seeding abilities and outcome of human brain lysates from these diseases, as well as recombinant preformed human αSyn fibrils by the intracerebral inoculation of transgenic mice overexpressing either human wild-type αSyn or human αSyn with the familial A53T mutation. Our study has revealed that the initiating inoculum heavily dictates the phenotypic and pathological course of disease. Interestingly, we have also established relevant host-dependent distinctions between propagation profiles, including burden and spread of inclusion pathology throughout the neuroaxis, as well as severity of neurological symptoms. These findings provide compelling evidence supporting the hypothesis that diverse prion-type conformers may explain the variability seen in synucleinopathies.


Subject(s)
Alzheimer Disease , Multiple System Atrophy , Prions , Synucleinopathies , Alzheimer Disease/pathology , Amyloid , Animals , Humans , Mice , Mice, Transgenic , Multiple System Atrophy/pathology , Prions/genetics , Prions/metabolism , Synucleinopathies/genetics , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
10.
Mol Neurodegener ; 16(1): 63, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34503546

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Dementia/metabolism , Disease Models, Animal , Protein Aggregation, Pathological , alpha-Synuclein/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Crosses, Genetic , Dementia/pathology , Gliosis/metabolism , Gliosis/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Injections , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Neuroinflammatory Diseases , Parkinson Disease/metabolism , Parkinson Disease/pathology , Prions/chemistry , Protein Aggregates , Recombinant Proteins/metabolism , alpha-Synuclein/toxicity
11.
Acta Neuropathol Commun ; 8(1): 88, 2020 06 22.
Article in English | MEDLINE | ID: mdl-32571418

ABSTRACT

Tau protein abnormally aggregates in tauopathies, a diverse group of neurologic diseases that includes Alzheimer's disease (AD). In early stages of disease, tau becomes hyperphosphorylated and mislocalized, which can contribute to its aggregation and toxicity. We demonstrate that tau phosphorylation at Ser208 (pSer208) promotes microtubule dysfunction and tau aggregation in cultured cells. Comparative assessment of the epitopes recognized by antibodies AT8, CP13, and 7F2 demonstrates that CP13 and 7F2 are specific for tau phosphorylation at Ser202 and Thr205, respectively, independently of the phosphorylation state of adjacent phosphorylation sites. Supporting the involvement of pSer208 in tau pathology, a novel monoclonal antibody 3G12 specific for tau phosphorylation at Ser208 revealed strong reactivity of tau inclusions in the brains of PS19 and rTg4510 transgenic mouse models of tauopathy. 3G12 also labelled neurofibrillary tangles in brains of patients with AD but revealed differential staining compared to CP13 and 7F2 for other types of tau pathologies such as in neuropil threads and neuritic plaques in AD, tufted astrocytes in progressive supranuclear palsy and astrocytic plaques in corticobasal degeneration. These results support the hypothesis that tau phosphorylation at Ser208 strongly contributes to unique types of tau aggregation and may be a reliable marker for the presence of mature neurofibrillary tangles.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Transgenic , Phosphorylation , Serine/metabolism
12.
Neurosci Lett ; 732: 135017, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32371157

ABSTRACT

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.


Subject(s)
Synucleinopathies/metabolism , alpha-Synuclein/metabolism , Animals , Central Nervous System/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Transgenic , Neurons , Parkinson Disease/metabolism , Phosphorylation , Prions , Protein Processing, Post-Translational , Survival Analysis , Synucleinopathies/pathology
13.
FEBS Lett ; 594(8): 1271-1283, 2020 04.
Article in English | MEDLINE | ID: mdl-31912891

ABSTRACT

α-synuclein (αsyn) forms pathologic inclusions in several neurodegenerative diseases termed synucleinopathies. The inclusions are comprised of αsyn fibrils harboring prion-like properties. Prion-like activity of αsyn has been studied by intracerebral injection of fibrils into mice, where the presence of a species barrier requires the use of mouse αsyn. Post-translational modifications to αsyn such as carboxy (C)-terminal truncation occur in synucleinopathies, and their implications for prion-like aggregation and seeding are under investigation. Herein, C-truncated forms of αsyn found in human disease are recapitulated in mouse αsyn to study their seeding activity in vitro, in HEK293T cells, in neuronal-glial culture, and in nontransgenic mice. The results show that C-truncation of mouse αsyn accelerates aggregation of αsyn but alters prion-like seeding of inclusion formation.


Subject(s)
Neurons/metabolism , alpha-Synuclein/metabolism , Amyloid/metabolism , Animals , Cells, Cultured , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Injections , Mice, Inbred Strains , Neuroglia/cytology , Neuroglia/metabolism , Neurons/pathology , Prions/metabolism , alpha-Synuclein/administration & dosage , alpha-Synuclein/genetics
14.
Acta Neuropathol Commun ; 7(1): 142, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477175

ABSTRACT

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.


Subject(s)
Amygdala/metabolism , Amygdala/pathology , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Disease Progression , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Mice, Transgenic , Middle Aged , Neurons/metabolism , Neurons/pathology
15.
Neurosci Lett ; 692: 187-192, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30423399

ABSTRACT

Alzheimer's disease and other tauopathies are characterized by the brain accumulation of hyperphosphorylated aggregated tau protein forming pathological inclusions. Although elevated tau phosphorylated at many amino acid residues is a hallmark of pathological tau, some evidence suggest that tau phosphorylation at unique sites, especially within its microtubule-binding domain, might inhibit aggregation. In this study, the effects of phosphorylation of two unique residues within this domain, serine 305 (S305) and serine 320 (S320), were examined in the context of established aggregation and seeding models. It was found that the S305E phosphomimetic significantly inhibited both tau seeding and tau aggregation in this model, while S320E did not. To further explore S305 phosphorylation in vivo, a monoclonal antibody (2G2) specific for tau phosphorylated at S305 was generated and characterized. Consistent with inhibition of tau aggregation, phosphorylation of S305 was not detected in pathological tau inclusions in Alzheimer's disease brain tissue. This study indicates that phosphorylation of unique tau residues can be inhibitory to aggregate formation, and has important implications for potential kinase therapies. Additionally, it creates new tools for observing these changes in vivo.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Protein Aggregation, Pathological/metabolism , Serine/metabolism , tau Proteins/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Cells, Cultured , HEK293 Cells , Humans , Mice , Phosphorylation , tau Proteins/immunology
16.
J Biol Chem ; 293(49): 18914-18932, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30327435

ABSTRACT

α-Synuclein (αsyn) aggregates into toxic fibrils in multiple neurodegenerative diseases where these fibrils form characteristic pathological inclusions such as Lewy bodies (LBs). The mechanisms initiating αsyn aggregation into fibrils are unclear, but ubiquitous post-translational modifications of αsyn present in LBs may play a role. Specific C-terminally (C)-truncated forms of αsyn are present within human pathological inclusions and form under physiological conditions likely in lysosome-associated pathways, but the roles for these C-truncated forms of αsyn in inclusion formation and disease are not well understood. Herein, we characterized the in vitro aggregation properties, amyloid fibril structures, and ability to induce full-length (FL) αsyn aggregation through prion-like mechanisms for eight of the most common physiological C-truncated forms of αsyn (1-115, 1-119, 1-122, 1-124, 1-125, 1-129, 1-133, and 1-135). In vitro, C-truncated αsyn aggregated more readily than FL αsyn and formed fibrils with unique morphologies. The presence of C-truncated αsyn potentiated aggregation of FL αsyn in vitro through co-polymerization. Specific C-truncated forms of αsyn in cells also exacerbated seeded aggregation of αsyn. Furthermore, in primary neuronal cultures, co-polymers of C-truncated and FL αsyn were potent prion-like seeds, but polymers composed solely of the C-truncated protein were not. These experiments indicated that specific physiological C-truncated forms of αsyn have distinct aggregation properties, including the ability to modulate the prion-like aggregation and seeding activity of FL αsyn. Proteolytic formation of these C-truncated species may have an important role in both the initiation of αsyn pathological inclusions and further progression of disease with strain-like properties.


Subject(s)
Amyloid/metabolism , Peptide Fragments/metabolism , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/metabolism , Amyloid/immunology , Animals , Antibodies, Monoclonal/immunology , HEK293 Cells , Humans , Mice, Inbred BALB C , Peptide Fragments/immunology , Protein Multimerization , Proteolysis , alpha-Synuclein/immunology
17.
Neurosci Lett ; 661: 114-120, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-28964772

ABSTRACT

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.


Subject(s)
Intermediate Filaments/metabolism , Neurons/metabolism , Prions/metabolism , alpha-Synuclein/metabolism , Animals , Brain/metabolism , Inclusion Bodies/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurodegenerative Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...