Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Type of study
Language
Publication year range
1.
Biochim Biophys Acta Gen Subj ; 1862(12): 2835-2845, 2018 12.
Article in English | MEDLINE | ID: mdl-30251672

ABSTRACT

BACKGROUND: Salsolinol (SALSO), a product from the reaction of dopamine (DA) with acetaldehyde, is found increased in dopaminergic neurons of Parkinson's disease (PD) patients. The administration of SALSO in rats causes myenteric neurodegeneration followed by the formation of deposits of the protein α-synuclein (aS), whose aggregation is intimately associated to PD. METHODS: NMR, isothermal titration calorimetry and MS were used to evaluate the interaction of SALSO with aS. The toxicity of SALSO and in vitro-produced aS-SALSO species was evaluated on mesencephalic primary neurons from mice. RESULTS: SALSO, under oxidative conditions, stabilizes the monomeric state besides a minor population of oligomers of aS, resulting in a strong inhibition of the fibrillation process. SALSO does not promote any chemical modification of the protein. Instead, the interaction of SALSO with aS seems to occur via hydrophobic effect, likely mediated by the NAC (non-amyloid component) domain of the protein. aS-SALSO species were found to be innocuous on primary neurons, while SALSO alone induces apoptosis via caspase-3 activation. Importantly, exogenous aS monomer was capable of protecting neurons against SALSO toxicity irrespective whether the protein was co-administered with SALSO or added until 2 h after SALSO, as evidenced by DAPI and cleaved-caspase 3 assays. Similar protective action of aS was found by pre-incubating neurons with aS before the administration of SALSO. CONCLUSIONS: Interaction of SALSO with aS leads to the formation of fibril-incompetent and innocuous adducts. SALSO toxicity is attenuated by aS monomer. SIGNIFICANCE: aS could exhibit a protective role against the neurotoxic effects of SALSO in dopaminergic neuron.


Subject(s)
Dopaminergic Neurons/drug effects , Isoquinolines/toxicity , Synapses/metabolism , alpha-Synuclein/physiology , Animals , Apoptosis/drug effects , Calorimetry , Caspase 3/metabolism , Cells, Cultured , Chromatography, Gel , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Humans , Mass Spectrometry , Mice , Oxidation-Reduction , Rats , Spectrometry, Fluorescence , alpha-Synuclein/metabolism
2.
Mol Neurobiol ; 47(2): 525-36, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22923367

ABSTRACT

The toxicity of α-synuclein in the neuropathology of Parkinson's disease which includes its hallmark aggregation has been studied scrupulously in the last decade. Although little is known regarding the normal functions of α-synuclein, its association with membrane phospholipids suggests its potential role in signaling pathways. Following extensive evidences for its nuclear localization, we and others recently demonstrated DNA binding activity of α-synuclein that modulates its conformation as well as aggregation properties. Furthermore, we also underscored the similarities among various amyloidogenic proteins involved in neurodegenerative diseases including amyloid beta peptides and tau. Our more recent studies show that α-synuclein is glycated and glycosylated both in vitro and in neurons, significantly affecting its folding, oligomeric, and DNA binding properties. Glycated α-synuclein causes increased genome damage both via its direct interaction with DNA and by increased generation of reactive oxygen species as glycation byproduct. In this review, we discuss the mechanisms of glycation and other posttranslational modifications of α-synuclein, including phosphorylation and nitration, and their role in neuronal death in Parkinson's disease.


Subject(s)
Glycation End Products, Advanced/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/physiology , Animals , Cell Death/physiology , Glycosylation , Humans , Parkinson Disease/pathology , Phosphorylation/physiology , Protein Binding/physiology , Protein Folding , alpha-Synuclein/adverse effects , alpha-Synuclein/toxicity
3.
J Biol Chem ; 287(44): 37206-18, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22918834

ABSTRACT

The accumulation of amyloid fibrils is a feature of amyloid diseases, where cell toxicity is due to soluble oligomeric species that precede fibril formation or are formed by fibril fragmentation, but the mechanism(s) of fragmentation is still unclear. Neutrophil-derived elastase and histones were found in amyloid deposits from patients with different systemic amyloidoses. Neutrophil extracellular traps (NETs) are key players in a death mechanism in which neutrophils release DNA traps decorated with proteins such as elastase and histones to entangle pathogens. Here, we asked whether NETs are triggered by amyloid fibrils, reasoning that because proteases are present in NETs, protease digestion of amyloid may generate soluble, cytotoxic species. We show that amyloid fibrils from three different sources (α-synuclein, Sup35, and transthyretin) induced NADPH oxidase-dependent NETs in vitro from human neutrophils. Surprisingly, NET-associated elastase digested amyloid fibrils into short species that were cytotoxic for BHK-21 and HepG2 cells. In tissue sections from patients with primary amyloidosis, we also observed the co-localization of NETs with amyloid deposits as well as with oligomers, which are probably derived from elastase-induced fibril degradation (amyloidolysis). These data reveal that release of NETs, so far described to be elicited by pathogens, can also be triggered by amyloid fibrils. Moreover, the involvement of NETs in amyloidoses might be crucial for the production of toxic species derived from fibril fragmentation.


Subject(s)
Amyloid/physiology , Chromatin/metabolism , Neutrophils/pathology , Acetophenones/pharmacology , Amyloid/chemistry , Amyloid/genetics , Amyloid Neuropathies, Familial/enzymology , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/pathology , Amyloidosis/enzymology , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Biomarkers/metabolism , Cell Nucleus/metabolism , Cell Survival/drug effects , Chromatin/enzymology , Cricetinae , Extracellular Space/enzymology , Extracellular Space/metabolism , Hep G2 Cells , Humans , Immunoglobulin Light-chain Amyloidosis , Lung/enzymology , Lung/metabolism , Lung/pathology , Mutation, Missense , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neutrophils/enzymology , Neutrophils/metabolism , Onium Compounds/pharmacology , Pancreatic Elastase , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Prealbumin/chemistry , Prealbumin/genetics , Prealbumin/physiology , Protein Structure, Quaternary , Proteolysis , Reactive Oxygen Species/metabolism , Skin/enzymology , Skin/metabolism , Skin/pathology , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/physiology
4.
Free Radic Biol Med ; 53(4): 1004-15, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22771470

ABSTRACT

Alpha-synuclein (αS), a 140 amino acid presynaptic protein, is the major component of the fibrillar aggregates (Lewy bodies) observed in dopaminergic neurons of patients affected by Parkinson's disease. It is currently believed that noncovalent oligomeric forms of αS, arising as intermediates in its aggregation, may constitute the major neurotoxic species. However, attempts to isolate and characterize such oligomers in vitro, and even more so in living cells, have been hampered by their transient nature, low concentration, polymorphism, and inherent instability. In this work, we describe the preparation and characterization of low molecular weight covalently bound oligomeric species of αS obtained by crosslinking via tyrosyl radicals generated by blue-light photosensitization of the metal coordination complex ruthenium (II) tris-bipyridine in the presence of ammonium persulfate. Numerous analytical techniques were used to characterize the αS oligomers: biochemical (anion-exchange chromatography, SDS-PAGE, and Western blotting); spectroscopic (optical: UV/Vis absorption, steady state, dynamic fluorescence, and dynamic light scattering); mass spectrometry; and electrochemical. Light-controlled protein oligomerization was mediated by formation of Tyr-Tyr (dityrosine) dimers through -C-C- bonds acting as covalent bridges, with a predominant involvement of residue Y39. The diverse oligomeric species exhibited a direct effect on the in vitro aggregation behavior of wild-type monomeric αS, decreasing the total yield of amyloid fibrils in aggregation assays monitored by thioflavin T (ThioT) fluorescence and light scattering, and by atomic force microscopy (AFM). Compared to the unmodified monomer, the photoinduced covalent oligomeric species demonstrated increased toxic effects on differentiated neuronal-like SH-SY5Y cells. The results highlight the importance of protein modification induced by oxidative stress in the initial molecular events leading to Parkinson's disease.


Subject(s)
Amyloid/chemistry , Free Radicals/chemistry , Tyrosine/chemistry , alpha-Synuclein/chemistry , Ammonium Sulfate/chemistry , Amyloid/chemical synthesis , Amyloid/physiology , Cell Line , Cell Survival , Cross-Linking Reagents/chemistry , Humans , Kinetics , Organometallic Compounds/chemistry , Oxidative Stress , Photochemical Processes , Photosensitizing Agents/chemistry , Protein Stability , alpha-Synuclein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL