Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502242

ABSTRACT

Alpha-synucleinopathies are featured by fibrillar inclusions in brain cells. Although α-synuclein fibrils display structural diversity, the origin of this diversity is not fully understood. We used molecular dynamics simulations to design synthetic peptides, based on the NAC 71-82 amino acid fragment of α-synuclein, that govern protofilament contacts and generation of twisted fibrillar polymorphs. Four peptides with structures based on either single or double fragments and capped or non-capped ends were selected for further analysis. We determined the fibrillar yield and the structures from these peptides found in the solution after fibrillisation using protein concentration determination assay and circular dichroism spectroscopy. In addition, we characterised secondary structures formed by individual fibrillar complexes using laser-tweezers Raman spectroscopy. Results suggest less mature fibrils, based on the lower relative ß-sheet content for double- than single-fragment peptide fibrils. We confirmed this structural difference by TEM analysis which revealed, in addition to short protofibrils, more elongated, twisted and rod-like fibril structures in non-capped and capped double-fragment peptide systems, respectively. Finally, time-correlated single-photon counting demonstrated a difference in the Thioflavin T fluorescence lifetime profiles upon fibril binding. It could be proposed that this difference originated from morphological differences in the fibril samples. Altogether, these results highlight the potential of using peptide models for the generation of fibrils that share morphological features relevant for disease, e.g., twisted and rod-like polymorphs.


Subject(s)
Amino Acids/chemistry , Amyloid/chemistry , Molecular Dynamics Simulation , Peptide Fragments/chemistry , alpha-Synuclein/chemistry , Humans , Protein Conformation , Protein Conformation, beta-Strand , Protein Structure, Secondary
2.
Int J Mol Sci ; 21(5)2020 Feb 27.
Article in English | MEDLINE | ID: mdl-32120928

ABSTRACT

Although Lewy bodies and Lewy neurites are hallmarks of Parkinson's disease (PD) and dementia with Lewy bodies (DLB), misfolded α-synuclein oligomers are nowadays believed to be key for the development of these diseases. Attempts to target soluble misfolded species of the full-length protein have been limited so far, probably due to the fast aggregation kinetics and burial of aggregation prone segments in final cross-ß-sheet fibrils. A previous characterisation study of fibrils prepared from a capped peptide of the non-amyloid ß-component (NAC) 71-82 amino acid stretch of α-synuclein demonstrated an increased aggregation propensity resulting in a cross-ß-structure that is also found in prion proteins. From this, it was suggested that capped NAC 71-82 peptide oligomers would provide interesting motifs with a capacity to regulate disease development. Here, we demonstrated, from a series of circular dichroism spectroscopic measurements and molecular dynamics simulations, the molecular-environment-sensitive behaviour of the capped NAC 71-82 peptide in a solution phase and the formation of ß-sheet oligomeric structures in the supernatant of a fibrillisation mixture. These results highlighted the use of the capped NAC 71-82 peptide as a motif in the preparation of oligomeric ß-sheet structures that potentially could be used in therapeutic strategies in the fight against progressive neurodegenerative disorders, such as PD and DLB.


Subject(s)
Amyloid beta-Peptides/chemistry , alpha-Synuclein/chemistry , Benzothiazoles/chemistry , Circular Dichroism , Fluorescence , Molecular Dynamics Simulation , Peptides/chemistry , Protein Conformation, beta-Strand , Protein Folding , Solubility , alpha-Synuclein/metabolism
3.
Sci Rep ; 9(1): 15949, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31685848

ABSTRACT

The 71-82 fragment of the non-amyloid-ß component (NAC) region of the Parkinson's disease (PD) and dementia with Lewy bodies (DLB) related protein α-Synuclein, has been reported to be important during protein misfolding. Although reports have demonstrated the importance of this fragment for the aggregation properties of the full-length protein, its exact role in pre-fibrillar oligomerisation, fibrillar growth and morphology has not yet been fully elucidated. Here, we provide evidence that fibrils prepared from an acetylated and methyl amidated peptide of the NAC 71-82 amino acid stretch of α-Synuclein are amyloid and contain, in addition to the cross-ß structure detected in the full-length protein fibrils, a cross-ß structure previously observed in prion proteins. These results shed light on the aggregation propensity of the NAC 71-82 amino acid stretch of the full-length protein but also the roles of the N- and C-terminal domains of α-Synuclein in balancing this aggregation propensity. The results also suggest that early aggregated forms of the capped NAC 71-82 peptide generated structures were stabilised by an anti-parallel and twisted ß-sheet motif. Due to its expected toxicity, this ß-sheet motif may be a promising molecular target for the development of therapeutic strategies for PD and DLB.


Subject(s)
Amino Acids/chemistry , Amyloid/chemistry , Peptides/chemistry , Prion Proteins/chemistry , Protein Structure, Secondary , alpha-Synuclein/chemistry , Acetylation , Amyloid/ultrastructure , Methylation , Molecular Dynamics Simulation , Protein Conformation , Spectroscopy, Fourier Transform Infrared , alpha-Synuclein/metabolism
4.
Amyloid ; 20(4): 233-44, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24053224

ABSTRACT

Aggregated α-synuclein is the major component of Lewy bodies, protein inclusions observed in the brain in neurodegenerative disorders such as Parkinson's disease and dementia with Lewy bodies. Experimental evidence indicates that α-synuclein potentially can be transferred between cells and act as a seed to accelerate the aggregation process. Here, we investigated in vitro and in vivo seeding effects of α-synuclein oligomers induced by the reactive aldehyde 4-oxo-2-nonenal (ONE). As measured by a Thioflavin-T based fibrillization assay, there was an earlier onset of aggregation when α-synuclein oligomers were added to monomeric α-synuclein. In contrast, exogenously added α-synuclein oligomers did not induce aggregation in a cell model. However, cells overexpressing α-synuclein that were treated with the oligomers displayed reduced α-synuclein levels, indicating that internalized oligomers either decreased the expression or accelerated the degradation of transfected α-synuclein. Also in vivo there were no clear seeding effects, as intracerebral injections of α-synuclein oligomers into the neocortex of α-synuclein transgenic mice did not induce formation of proteinase K resistant α-synuclein pathology. Taken together, we could observe a seeding effect of the ONE-induced α-synuclein oligomers in a fibrillization assay, but neither in a cell nor in a mouse model.


Subject(s)
alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Aldehydes/chemistry , Animals , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Humans , Mice , Mice, Transgenic , Microscopy, Atomic Force , Parkinson Disease/metabolism , Parkinson Disease/pathology
5.
J Neurochem ; 126(1): 131-44, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23363402

ABSTRACT

Inclusions of intraneuronal alpha-synuclein (α-synuclein) can be detected in brains of patients with Parkinson's disease and dementia with Lewy bodies. The aggregation of α-synuclein is a central feature of the disease pathogenesis. Among the different α-synuclein species, large oligomers/protofibrils have particular neurotoxic properties and should therefore be suitable as both therapeutic and diagnostic targets. Two monoclonal antibodies, mAb38F and mAb38E2, with high affinity and strong selectivity for large α-synuclein oligomers were generated. These antibodies, which do not bind amyloid-beta or tau, recognize Lewy body pathology in brains from patients with Parkinson's disease and dementia with Lewy bodies and detect pathology earlier in α-synuclein transgenic mice than linear epitope antibodies. An oligomer-selective sandwich ELISA, based on mAb38F, was set up to analyze brain extracts of the transgenic mice. The overall levels of α-synuclein oligomers/protofibrils were found to increase with age in these mice, although the levels displayed a large interindividual variation. Upon subcellular fractionation, higher levels of α-synuclein oligomers/protofibrils could be detected in the endoplasmic reticulum around the age when behavioral disturbances develop. In summary, our novel oligomer-selective α-synuclein antibodies recognize relevant pathology and should be important tools to further explore the pathogenic mechanisms in Lewy body disorders. Moreover, they could be potential candidates both for immunotherapy and as reagents in an assay to assess a potential disease biomarker.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain/pathology , Lewy Body Disease/genetics , Lewy Body Disease/pathology , alpha-Synuclein/genetics , alpha-Synuclein/immunology , Animals , Blotting, Western , DNA, Complementary/genetics , Enzyme-Linked Immunosorbent Assay , Epitopes , Formates/chemistry , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Mutation/genetics , Mutation/physiology , Subcellular Fractions/metabolism
6.
J Neurosci ; 32(34): 11750-62, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22915117

ABSTRACT

Parkinson's disease (PD) is the most common representative of a group of disorders known as synucleinopathies, in which misfolding and aggregation of α-synuclein (a-syn) in various brain regions is the major pathological hallmark. Indeed, the motor symptoms in PD are caused by a heterogeneous degeneration of brain neurons not only in substantia nigra pars compacta but also in other extrastriatal areas of the brain. In addition to the well known motor dysfunction in PD patients, cognitive deficits and memory impairment are also an important part of the disorder, probably due to disruption of synaptic transmission and plasticity in extrastriatal areas, including the hippocampus. Here, we investigated the impact of a-syn aggregation on AMPA and NMDA receptor-mediated rat hippocampal (CA3-CA1) synaptic transmission and long-term potentiation (LTP), the neurophysiological basis for learning and memory. Our data show that prolonged exposure to a-syn oligomers, but not monomers or fibrils, increases basal synaptic transmission through NMDA receptor activation, triggering enhanced contribution of calcium-permeable AMPA receptors. Slices treated with a-syn oligomers were unable to respond with further potentiation to theta-burst stimulation, leading to impaired LTP. Prior delivery of a low-frequency train reinstated the ability to express LTP, implying that exposure to a-syn oligomers drives the increase of glutamatergic synaptic transmission, preventing further potentiation by physiological stimuli. Our novel findings provide mechanistic insight on how a-syn oligomers may trigger neuronal dysfunction and toxicity in PD and other synucleinopathies.


Subject(s)
Long-Term Potentiation/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , alpha-Synuclein/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Biophysics , Biotinylation , Cell Line, Tumor , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Extracellular Fluid/metabolism , Hippocampus/cytology , Humans , Insulin/pharmacology , L-Lactate Dehydrogenase/metabolism , Long-Term Potentiation/physiology , Male , Neuroblastoma/pathology , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Valine/analogs & derivatives , Valine/pharmacology , alpha-Synuclein/biosynthesis , alpha-Synuclein/chemistry
7.
PLoS One ; 6(10): e27230, 2011.
Article in English | MEDLINE | ID: mdl-22073131

ABSTRACT

Recent research implicates soluble aggregated forms of α-synuclein as neurotoxic species with a central role in the pathogenesis of Parkinson's disease and related disorders. The pathway by which α-synuclein aggregates is believed to follow a step-wise pattern, in which dimers and smaller oligomers are initially formed. Here, we used H4 neuroglioma cells expressing α-synuclein fused to hemi:GFP constructs to study the effects of α-synuclein monoclonal antibodies on the early stages of aggregation, as quantified by Bimolecular Fluorescence Complementation assay. Widefield and confocal microscopy revealed that cells treated for 48 h with monoclonal antibodies internalized antibodies to various degrees. C-terminal and oligomer-selective α-synuclein antibodies reduced the extent of α-synuclein dimerization/oligomerization, as indicated by decreased GFP fluorescence signal. Furthermore, ELISA measurements on lysates and conditioned media from antibody treated cells displayed lower α-synuclein levels compared to untreated cells, suggesting increased protein turnover. Taken together, our results propose that extracellular administration of monoclonal antibodies can modify or inhibit early steps in the aggregation process of α-synuclein, thus providing further support for passive immunization against diseases with α-synuclein pathology.


Subject(s)
Antibodies, Monoclonal/pharmacology , Glioma/metabolism , Green Fluorescent Proteins/metabolism , Protein Multimerization/drug effects , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Animals , Blotting, Western , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Glioma/genetics , Glioma/immunology , Green Fluorescent Proteins/genetics , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Protein Multimerization/immunology , Tumor Cells, Cultured , alpha-Synuclein/immunology
8.
Biochem Biophys Res Commun ; 412(1): 32-8, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21798243

ABSTRACT

Deposition of fibrillar α-synuclein as Lewy bodies is the neuropathological hallmark of Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Apart from α-synuclein, these intraneuronal inclusions contain over 250 different proteins. The actin binding protein gelsolin, has previously been suggested to be part of the Lewy body, but its potential role in α-synuclein aggregation remains unknown. Here, we studied the association between gelsolin and α-synuclein in brain tissue from PD and DLB patients as well as in a cell model for α-synuclein aggregation. Moreover, the potential effect of gelsolin on α-synuclein fibrillization was also investigated. Our data demonstrate that gelsolin co-occured with α-synuclein in Lewy bodies from affected human brain as well as with Lewy body-like inclusions in α-synuclein over expressing cells. Furthermore, in the presence of calcium chloride, gelsolin was found to enhance the aggregation rate of α-synuclein in vitro. Moreover, no apparent structural differences could be observed between fibrils formed in the presence or absence of gelsolin. Further studies on gelsolin and other Lewy body associated proteins are warranted to learn more about their potential role in the α-synuclein aggregation process.


Subject(s)
Brain/metabolism , Gelsolin/metabolism , Lewy Bodies/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Aged , Calcium/metabolism , Cell Line , Humans , Male
9.
Free Radic Biol Med ; 50(3): 428-37, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21130160

ABSTRACT

Oxidative stress has been implicated in the etiology of neurodegenerative disorders with α-synuclein pathology. Lipid peroxidation products such as 4-oxo-2-nonenal (ONE) and 4-hydroxy-2-nonenal (HNE) can covalently modify and structurally alter proteins. Herein, we have characterized ONE- or HNE-induced α-synuclein oligomers. Our results demonstrate that both oligomers are rich in ß-sheet structure and have a molecular weight of about 2000 kDa. Atomic force microscopy analysis revealed that ONE-induced α-synuclein oligomers were relatively amorphous, with a diameter of 40-80 nm and a height of 4-8 nm. In contrast, the HNE-induced α-synuclein oligomers had a protofibril-like morphology with a width of 100-200 nm and a height of 2-4 nm. Furthermore, neither oligomer type polymerized into amyloid-like fibrils despite prolonged incubation. Although more SDS and urea stable, because of a higher degree of cross-linking, ONE-induced α-synuclein oligomers were less compact and more sensitive to proteinase K treatment. Finally, both ONE- and HNE-induced α-synuclein oligomers were cytotoxic when added exogenously to a neuroblastoma cell line, but HNE-induced α-synuclein oligomers were taken up by the cells to a significantly higher degree. Despite nearly identical chemical structures, ONE and HNE induce the formation of off-pathway α-synuclein oligomers with distinct biochemical, morphological, and functional properties.


Subject(s)
Aldehydes/pharmacology , Lipid Peroxidation , alpha-Synuclein/metabolism , Aldehydes/metabolism , Cell Survival/drug effects , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/ultrastructure , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Tumor Cells, Cultured , alpha-Synuclein/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...