1
|
Sechi GP, Sechi MM. Small Molecules, α-Synuclein Pathology, and the Search for Effective Treatments in Parkinson's Disease. Int J Mol Sci 2024; 25:11198. [PMID: 39456980 PMCID: PMC11508228 DOI: 10.3390/ijms252011198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Parkinson's disease (PD) is a progressive age-related neurodegenerative disorder affecting millions of people worldwide. Essentially, it is characterised by selective degeneration of dopamine neurons of the nigro-striatal pathway and intraneuronal aggregation of misfolded α-synuclein with formation of Lewy bodies and Lewy neurites. Moreover, specific small molecules of intermediary metabolism may have a definite pathophysiological role in PD. These include dopamine, levodopa, reduced glutathione, glutathione disulfide/oxidised glutathione, and the micronutrients thiamine and ß-Hydroxybutyrate. Recent research indicates that these small molecules can interact with α-synuclein and regulate its folding and potential aggregation. In this review, we discuss the current knowledge on interactions between α-synuclein and both the small molecules of intermediary metabolism in the brain relevant to PD, and many other natural and synthetic small molecules that regulate α-synuclein aggregation. Additionally, we analyse some of the relevant molecular mechanisms potentially involved. A better understanding of these interactions may have relevance for the development of rational future therapies. In particular, our observations suggest that the micronutrients ß-Hydroxybutyrate and thiamine might have a synergistic therapeutic role in halting or reversing the progression of PD and other neuronal α-synuclein disorders.
Collapse
Affiliation(s)
- Gian Pietro Sechi
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | | |
Collapse
|
2
|
Imaura R, Kawata Y, Matsuo K. Salt-Induced Hydrophobic C-Terminal Region of α-Synuclein Triggers Its Fibrillation under the Mimic Physiologic Condition. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:20537-20549. [PMID: 39285698 DOI: 10.1021/acs.langmuir.4c02178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
α-Synuclein (αS) causes Parkinson's disease due to the structural alteration into amyloid fibrils that form after the interaction with synaptic membranes in neurons. To understand the alternation mechanism, the effect of salt (NaCl) on the interaction of αS with synaptic mimic membrane was characterized at the molecular level because salt triggered the amyloid fibril formation. The membrane-bound conformation (or the initial conformation before fibrillation) showed that NaCl decreased the number of helical structures and Tyr residues interacting with the membrane surface compared to when NaCl was absent, implying an increase in solvent-exposed regions. The N-terminal region of αS interacted with the membrane, forming the helical structures regardless of NaCl, while the C-terminal region formed a random structure with weak membrane interaction, but NaCl inhibited the interaction of its hydrophobic area, suggesting that salt promoted amyloid fibril formations by exposing the hydrophobic C-terminal region, which can intermolecularly interact with free αS.
Collapse
Affiliation(s)
- Ryota Imaura
- Graduate School of Advanced Science and Engineering, Hiroshima University, Hiroshima 739-8511, Japan
| | - Yasushi Kawata
- Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Koichi Matsuo
- Graduate School of Advanced Science and Engineering, Hiroshima University, Hiroshima 739-8511, Japan
- Research Institute for Synchrotron Radiation Science, Hiroshima University, Hiroshima 739-0046, Japan
- International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM2), Hiroshima University, Hiroshima 739-0046, Japan
| |
Collapse
|
3
|
Mazzotta GM, Conte C. Alpha Synuclein Toxicity and Non-Motor Parkinson's. Cells 2024; 13:1265. [PMID: 39120295 PMCID: PMC11311369 DOI: 10.3390/cells13151265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Parkinson's disease (PD) is a common multisystem neurodegenerative disorder affecting 1% of the population over the age of 60 years. The main neuropathological features of PD are the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the presence of alpha synuclein (αSyn)-rich Lewy bodies both manifesting with classical motor signs. αSyn has emerged as a key protein in PD pathology as it can spread through synaptic networks to reach several anatomical regions of the body contributing to the appearance of non-motor symptoms (NMS) considered prevalent among individuals prior to PD diagnosis and persisting throughout the patient's life. NMS mainly includes loss of taste and smell, constipation, psychiatric disorders, dementia, impaired rapid eye movement (REM) sleep, urogenital dysfunction, and cardiovascular impairment. This review summarizes the more recent findings on the impact of αSyn deposits on several prodromal NMS and emphasizes the importance of early detection of αSyn toxic species in biofluids and peripheral biopsies as prospective biomarkers in PD.
Collapse
Affiliation(s)
| | - Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| |
Collapse
|
4
|
Maraldo A, Rnjak-Kovacina J, Marquis C. Tyrosine - a structural glue for hierarchical protein assembly. Trends Biochem Sci 2024; 49:633-648. [PMID: 38653686 DOI: 10.1016/j.tibs.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024]
Abstract
Protein self-assembly, guided by the interplay of sequence- and environment-dependent liquid-liquid phase separation (LLPS), constitutes a fundamental process in the assembly of numerous intrinsically disordered proteins. Heuristic examination of these proteins has underscored the role of tyrosine residues, evident in their conservation and pivotal involvement in initiating LLPS and subsequent liquid-solid phase transitions (LSPT). The development of tyrosine-templated constructs, designed to mimic their natural counterparts, emerges as a promising strategy for creating adaptive, self-assembling systems with diverse applications. This review explores the central role of tyrosine in orchestrating protein self-assembly, delving into key interactions and examining its potential in innovative applications, including responsive biomaterials and bioengineering.
Collapse
Affiliation(s)
- Anton Maraldo
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia.
| | - Christopher Marquis
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
5
|
Wagner WJ, Gross ML. Using mass spectrometry-based methods to understand amyloid formation and inhibition of alpha-synuclein and amyloid beta. MASS SPECTROMETRY REVIEWS 2024; 43:782-825. [PMID: 36224716 PMCID: PMC10090239 DOI: 10.1002/mas.21814] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Amyloid fibrils, insoluble β-sheets structures that arise from protein misfolding, are associated with several neurodegenerative disorders. Many small molecules have been investigated to prevent amyloid fibrils from forming; however, there are currently no therapeutics to combat these diseases. Mass spectrometry (MS) is proving to be effective for studying the high order structure (HOS) of aggregating proteins and for determining structural changes accompanying protein-inhibitor interactions. When combined with native MS (nMS), gas-phase ion mobility, protein footprinting, and chemical cross-linking, MS can afford regional and sometimes amino acid spatial resolution of the aggregating protein. The spatial resolution is greater than typical low-resolution spectroscopic, calorimetric, and the traditional ThT fluorescence methods used in amyloid research today. High-resolution approaches can struggle when investigating protein aggregation, as the proteins exist as complex oligomeric mixtures of many sizes and several conformations or polymorphs. Thus, MS is positioned to complement both high- and low-resolution approaches to studying amyloid fibril formation and protein-inhibitor interactions. This review covers basics in MS paired with ion mobility, continuous hydrogen-deuterium exchange (continuous HDX), pulsed hydrogen-deuterium exchange (pulsed HDX), fast photochemical oxidation of proteins (FPOP) and other irreversible labeling methods, and chemical cross-linking. We then review the applications of these approaches to studying amyloid-prone proteins with a focus on amyloid beta and alpha-synuclein. Another focus is the determination of protein-inhibitor interactions. The expectation is that MS will bring new insights to amyloid formation and thereby play an important role to prevent their formation.
Collapse
Affiliation(s)
- Wesley J Wagner
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
6
|
Sulatsky MI, Stepanenko OV, Stepanenko OV, Povarova OI, Kuznetsova IM, Turoverov KK, Sulatskaya AI. Broken but not beaten: Challenge of reducing the amyloids pathogenicity by degradation. J Adv Res 2024:S2090-1232(24)00161-9. [PMID: 38642804 DOI: 10.1016/j.jare.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND The accumulation of ordered protein aggregates, amyloid fibrils, accompanies various neurodegenerative diseases (such as Parkinson's, Huntington's, Alzheimer's, etc.) and causes a wide range of systemic and local amyloidoses (such as insulin, hemodialysis amyloidosis, etc.). Such pathologies are usually diagnosed when the disease is already irreversible and a large amount of amyloid plaques have accumulated. In recent years, new drugs aimed at reducing amyloid levels have been actively developed. However, although clinical trials have demonstrated a reduction in amyloid plaque size with these drugs, their effect on disease progression has been controversial and associated with significant side effects, the reasons of which are not fully understood. AIM OF REVIEW The purpose of this review is to summarize extensive array of data on the effect of exogenous and endogenous factors (physico-mechanical effects, chemical effects of low molecular weight compounds, macromolecules and their complexes) on the structure and pathogenicity of mature amyloids for proposing future directions of the development of effective and safe anti-amyloid therapeutics. KEY SCIENTIFIC CONCEPTS OF REVIEW Our analysis show that destruction of amyloids is in most cases incomplete and degradation products often retain the properties of amyloids (including high and sometimes higher than fibrils, cytotoxicity), accelerate amyloidogenesis and promote the propagation of amyloids between cells. Probably, the appearance of protein aggregates, polymorphic in structure and properties (such as amorphous aggregates, fibril fragments, amyloid oligomers, etc.), formed because of uncontrolled degradation of amyloids, may be one of the reasons for the ambiguous effectiveness and serious side effects of the anti-amyloid drugs. This means that all medications that are supposed to be used both for degradation and slow down the fibrillogenesis must first be tested on mature fibrils: the mechanism of drug action and cytotoxic, seeding, and infectious activity of the degradation products must be analyzed.
Collapse
Affiliation(s)
- Maksim I Sulatsky
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Olga V Stepanenko
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Olesya V Stepanenko
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Olga I Povarova
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Irina M Kuznetsova
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Konstantin K Turoverov
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Anna I Sulatskaya
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia.
| |
Collapse
|
7
|
D'Incecco P, Dallavalle S, Musso L, Rosi V, Sindaco M, Pellegrino L. Formation of di-Tyrosine in pasteurized milk during shelf storage. Food Chem 2024; 435:137566. [PMID: 37778263 DOI: 10.1016/j.foodchem.2023.137566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
Formation of the protein crosslink di-Tyrosine was studied in PET-bottled pasteurized milk exposed to fluorescent light in a commercial display cabinet. An HPLC method with fluorescence detection was developed and intra-laboratory validated using pure di-Tyrosine synthesized on purpose. Di-Tyrosine was detected after 1-day lightening and increased up to 7 days, reaching around 250 and 320 µg/g protein in whole and partly skimmed milk, respectively. Afterward, a progressive decrease occurred. By transmission electron microscopy with specific immune gold labelling, presence of di-Tyrosine was observed for the first time on the surface of casein micelles of lightened milk. The crosslink formation, however, did not bring to protein aggregation phenomena detectable by laser light scattering measurements. Exposure to light also induced degradation of riboflavin and decrease of yellowness index. Di-Tyrosine proved to be a suitable indicator to evaluate the progress of protein oxidation in pasteurized milk during storage on the market.
Collapse
Affiliation(s)
- Paolo D'Incecco
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Via Celoria 2, Milan, Italy.
| | - Sabrina Dallavalle
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Via Celoria 2, Milan, Italy
| | - Loana Musso
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Via Celoria 2, Milan, Italy
| | - Veronica Rosi
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Via Celoria 2, Milan, Italy
| | - Marta Sindaco
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Via Celoria 2, Milan, Italy
| | - Luisa Pellegrino
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Via Celoria 2, Milan, Italy
| |
Collapse
|
8
|
Han J. Copper trafficking systems in cells: insights into coordination chemistry and toxicity. Dalton Trans 2023; 52:15277-15296. [PMID: 37702384 DOI: 10.1039/d3dt02166a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Transition metal ions, such as copper, are indispensable components in the biological system. Copper ions which primarily exist in two major oxidation states Cu(I) and Cu(II) play crucial roles in various cellular processes including antioxidant defense, biosynthesis of neurotransmitters, and energy metabolism, owing to their inherent redox activity. The disturbance in copper homeostasis can contribute to the development of copper metabolism disorders, cancer, and neurodegenerative diseases, highlighting the significance of understanding the copper trafficking system in cellular environments. This review aims to offer a comprehensive overview of copper homeostatic machinery, with an emphasis on the coordination chemistry of copper transporters and trafficking proteins. While copper chaperones and the corresponding metalloenzymes are thoroughly discussed, we also explore the potential existence of low-molecular-mass metal complexes within cellular systems. Furthermore, we summarize the toxicity mechanisms originating from copper deficiency or accumulation, which include the dysregulation of oxidative stress, signaling pathways, signal transduction, and amyloidosis. This perspective review delves into the current knowledge regarding the intricate aspects of the copper trafficking system, providing valuable insights into potential treatment strategies from the standpoint of bioinorganic chemistry.
Collapse
Affiliation(s)
- Jiyeon Han
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea.
| |
Collapse
|
9
|
Ortigosa-Pascual L, Leiding T, Linse S, Pálmadóttir T. Photo-Induced Cross-Linking of Unmodified α-Synuclein Oligomers. ACS Chem Neurosci 2023; 14:3192-3205. [PMID: 37621159 PMCID: PMC10485903 DOI: 10.1021/acschemneuro.3c00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Photo-induced cross-linking of unmodified proteins (PICUP) has been used in the past to study size distributions of protein assemblies. PICUP may, for example, overcome the significant experimental challenges related to the transient nature, heterogeneity, and low concentration of amyloid protein oligomers relative to monomeric and fibrillar species. In the current study, a reaction chamber was designed, produced, and used for PICUP reaction optimization in terms of reaction conditions and lighting time from ms to s. These efforts make the method more reproducible and accessible and enable the use of shorter reaction times compared to previous studies. We applied the optimized method to an α-synuclein aggregation time course to monitor the relative concentration and size distribution of oligomers over time. The data are compared to the time evolution of the fibril mass concentration, as monitored by thioflavin T fluorescence. At all time points, the smaller the oligomer, the higher its concentration observed after PICUP. Moreover, the total oligomer concentration is highest at short aggregation times, and the decline over time follows the disappearance of monomers. We can therefore conclude that these oligomers form from monomers.
Collapse
Affiliation(s)
- Lei Ortigosa-Pascual
- Department of Biochemistry and Structural
Biology, Lund University, 221 00 Lund, Sweden
| | - Thom Leiding
- Department of Biochemistry and Structural
Biology, Lund University, 221 00 Lund, Sweden
| | - Sara Linse
- Department of Biochemistry and Structural
Biology, Lund University, 221 00 Lund, Sweden
| | - Tinna Pálmadóttir
- Department of Biochemistry and Structural
Biology, Lund University, 221 00 Lund, Sweden
| |
Collapse
|
10
|
Howell-Bray T, Byrne L. The effect of prions on cellular metabolism: The metabolic impact of the [RNQ +] prion and potential role of native Rnq1p. RESEARCH SQUARE 2023:rs.3.rs-2511186. [PMID: 36909567 PMCID: PMC10002837 DOI: 10.21203/rs.3.rs-2511186/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Within the field of amyloid and prion disease there is a need for a more comprehensive understanding of the fundamentals of disease biology. In order to facilitate the progression treatment and underpin comprehension of toxicity, fundamental understanding of the disruption to normal cellular biochemistry and trafficking is needed. Here, by removing the complex biochemistry of the brain, we have utilised known prion forming strains of Saccharomyces cerevisiae carrying different conformational variants of the Rnq1p to obtain Liquid Chromatography-Mass Spectrometry (LC-MS) metabolic profiles and identify key perturbations of prion presence. These studies reveal that prion containing [RNQ+] cells display a significant reduction in amino acid biosynthesis and distinct perturbations in sphingolipid metabolism, with significant downregulation in metabolites within these pathways. Moreover, that native Rnq1p appears to downregulate ubiquinone biosynthesis pathways within cells, suggesting that Rnq1p may play a lipid/mevalonate-based cytoprotective role as a regulator of ubiquinone production. These findings contribute to the understanding of how prion proteins interact in vivo in both their prion and non-prion confirmations and indicate potential targets for the mitigation of these effects. We demonstrate specific sphingolipid centred metabolic disruptions due to prion presence and give insight into a potential cytoprotective role of the native Rnq1 protein. This provides evidence of metabolic similarities between yeast and mammalian cells as a consequence of prion presence and establishes the application of metabolomics as a tool to investigate prion/amyloid-based phenomena.
Collapse
|
11
|
Maina MB, Al-Hilaly YK, Serpell LC. Dityrosine cross-linking and its potential roles in Alzheimer's disease. Front Neurosci 2023; 17:1132670. [PMID: 37034163 PMCID: PMC10075315 DOI: 10.3389/fnins.2023.1132670] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/01/2023] [Indexed: 04/11/2023] Open
Abstract
Oxidative stress is a significant source of damage that accumulates during aging and contributes to Alzheimer's disease (AD) pathogenesis. Oxidation of proteins can give rise to covalent links between adjacent tyrosines known as dityrosine (DiY) cross-linking, amongst other modifications, and this observation suggests that DiY could serve as a biomarker of accumulated oxidative stress over the lifespan. Many studies have focused on understanding the contribution of DiY to AD pathogenesis and have revealed that DiY crosslinks can be found in both Aβ and tau deposits - the two key proteins involved in the formation of amyloid plaques and tau tangles, respectively. However, there is no consensus yet in the field on the impact of DiY on Aβ and tau function, aggregation, and toxicity. Here we review the current understanding of the role of DiY on Aβ and tau gathered over the last 20 years since the first observation, and discuss the effect of this modification for Aβ and tau aggregation, and its potential as a biomarker for AD.
Collapse
Affiliation(s)
- Mahmoud B. Maina
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, United Kingdom
- Biomedical Science Research and Training Centre, College of Medical Sciences, Yobe State University, Damaturu, Nigeria
| | - Youssra K. Al-Hilaly
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, United Kingdom
- Department of Chemistry, College of Science, Mustansiriyah University, Baghdad, Iraq
| | - Louise C. Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, United Kingdom
- *Correspondence: Louise C. Serpell,
| |
Collapse
|
12
|
Docherty J, Leheste JR, Mancini J, Yao S. Preliminary Effects of Osteopathic Manipulative Medicine on Reactive Oxygen Species in Parkinson’s Disease: A Randomized Controlled Pilot Study. Cureus 2022; 14:e31504. [DOI: 10.7759/cureus.31504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2022] [Indexed: 11/16/2022] Open
|
13
|
Maina MB, Al-Hilaly YK, Oakley S, Burra G, Khanom T, Biasetti L, Mengham K, Marshall K, Harrington CR, Wischik CM, Serpell LC. Dityrosine Cross-links are Present in Alzheimer's Disease-derived Tau Oligomers and Paired Helical Filaments (PHF) which Promotes the Stability of the PHF-core Tau (297-391) In Vitro. J Mol Biol 2022; 434:167785. [PMID: 35961386 DOI: 10.1016/j.jmb.2022.167785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022]
Abstract
A characteristic hallmark of Alzheimer's Disease (AD) is the pathological aggregation and deposition of tau into paired helical filaments (PHF) in neurofibrillary tangles (NFTs). Oxidative stress is an early event during AD pathogenesis and is associated with tau-mediated AD pathology. Oxidative environments can result in the formation of covalent dityrosine crosslinks that can increase protein stability and insolubility. Dityrosine cross-linking has been shown in Aβ plaques in AD and α-synuclein aggregates in Lewy bodies in ex vivo tissue sections, and this modification may increase the insolubility of these aggregates and their resistance to degradation. Using the PHF-core tau fragment (residues 297 - 391) as a model, we have previously demonstrated that dityrosine formation traps tau assemblies to reduce further elongation. However, it is unknown whether dityrosine crosslinks are found in tau deposits in vivo in AD and its relevance to disease mechanism is unclear. Here, using transmission electron microscope (TEM) double immunogold-labelling, we reveal that neurofibrillary NFTs in AD are heavily decorated with dityrosine crosslinks alongside tau. Single immunogold-labelling TEM and fluorescence spectroscopy revealed the presence of dityrosine on AD brain-derived tau oligomers and fibrils. Using the tau (297-391) PHF-core fragment as a model, we further showed that prefibrillar tau species are more amenable to dityrosine crosslinking than tau fibrils. Dityrosine formation results in heat and SDS stability of oxidised prefibrillar and fibrillar tau assemblies. This finding has implications for understanding the mechanism governing the insolubility and toxicity of tau assemblies in vivo.
Collapse
Affiliation(s)
- Mahmoud B Maina
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK; Biomedical Science Research and Training Centre, Yobe State University, Nigeria. https://twitter.com/mahmoudbukar
| | - Youssra K Al-Hilaly
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK; Chemistry Department, College of Sciences, Mustansiriyah University, Baghdad, Iraq
| | - Sebastian Oakley
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Gunasekhar Burra
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK; Analytical Development Biologics, Biopharmaceutical Development, Syngene International Limited, Biocon Park, Bommasandra Jigani Link Road, Bangalore 560009, India
| | - Tahmida Khanom
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Luca Biasetti
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Kurtis Mengham
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Karen Marshall
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Charles R Harrington
- Institute of Medical Sciences, University of Aberdeen, UK; TauRx Therapeutics Ltd, Aberdeen, UK
| | - Claude M Wischik
- Institute of Medical Sciences, University of Aberdeen, UK; TauRx Therapeutics Ltd, Aberdeen, UK
| | - Louise C Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK.
| |
Collapse
|
14
|
Abbas M, Law JO, Grellscheid SN, Huck WTS, Spruijt E. Peptide-Based Coacervate-Core Vesicles with Semipermeable Membranes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202913. [PMID: 35796384 DOI: 10.1002/adma.202202913] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Coacervates droplets have long been considered as potential protocells to mimic living cells. However, these droplets lack a membrane and are prone to coalescence, limiting their ability to survive, interact, and organize into higher-order assemblies. This work shows that tyrosine-rich peptide conjugates can undergo liquid-liquid phase separation in a well-defined pH window and transform into stable membrane-enclosed protocells by enzymatic oxidation and cross-linking at the liquid-liquid interface. The oxidation of the tyrosine-rich peptides into dityrosine creates a semipermeable, flexible membrane around the coacervates with tunable thickness, which displays strong intrinsic fluorescence, and stabilizes the coacervate protocells against coalescence. The membranes have an effective molecular weight cut-off of 2.5 kDa, as determined from the partitioning of small dyes and labeled peptides, RNA, and polymers into the membrane-enclosed coacervate protocells. Flicker spectroscopy reveals a membrane bending rigidity of only 0.1kB T, which is substantially lower than phospholipid bilayers despite a larger membrane thickness. Finally, it is shown that enzymes can be stably encapsulated inside the protocells and be supplied with substrates from outside, which opens the way for these membrane-bound compartments to be used as molecularly crowded artificial cells capable of communication or as a vehicle for drug delivery.
Collapse
Affiliation(s)
- Manzar Abbas
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, 6525 AJ, The Netherlands
| | - Jack O Law
- Computational Biology Unit, University of Bergen, Bergen, 5020, Norway
| | - Sushma N Grellscheid
- Computational Biology Unit, University of Bergen, Bergen, 5020, Norway
- Department of Biosciences, Durham University, Durham, DH1 3LE, UK
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, 6525 AJ, The Netherlands
| | - Evan Spruijt
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, 6525 AJ, The Netherlands
| |
Collapse
|
15
|
Radomska K, Wolszczak M. Spontaneous and Ionizing Radiation-Induced Aggregation of Human Serum Albumin: Dityrosine as a Fluorescent Probe. Int J Mol Sci 2022; 23:ijms23158090. [PMID: 35897662 PMCID: PMC9331647 DOI: 10.3390/ijms23158090] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/04/2023] Open
Abstract
The use of spectroscopic techniques has shown that human serum albumin (HSA) undergoes reversible self-aggregation through protein−protein interactions. It ensures the subsequent overlapping of electron clouds along with the stiffening of the conformation of the interpenetrating network of amino acids of adjacent HSA molecules. The HSA oxidation process related to the transfer of one electron was investigated by pulse radiolysis and photochemical methods. It has been shown that the irradiation of HSA solutions under oxidative stress conditions results in the formation of stable protein aggregates. The HSA aggregates induced by ionizing radiation are characterized by specific fluorescence compared to the emission of non-irradiated solutions. We assume that HSA dimers are mainly responsible for the new emission. Dityrosine produced by the intermolecular recombination of protein tyrosine radicals as a result of radiolysis of an aqueous solution of the protein is the main cause of HSA aggregation by cross-linking. Analysis of the oxidation process of HSA confirmed that the reaction of mild oxidants (Br2•−, N3•, SO4•−) with albumin leads to the formation of covalent bonds between tyrosine residues. In the case of •OH radicals and partly, Cl2•−, species other than DT are formed. The light emission of this species is similar to the emission of self-associated HSA.
Collapse
|
16
|
Sahin C, Østerlund EC, Österlund N, Costeira-Paulo J, Pedersen JN, Christiansen G, Nielsen J, Grønnemose AL, Amstrup SK, Tiwari MK, Rao RSP, Bjerrum MJ, Ilag LL, Davies MJ, Marklund EG, Pedersen JS, Landreh M, Møller IM, Jørgensen TJD, Otzen DE. Structural Basis for Dityrosine-Mediated Inhibition of α-Synuclein Fibrillization. J Am Chem Soc 2022; 144:11949-11954. [PMID: 35749730 PMCID: PMC9284551 DOI: 10.1021/jacs.2c03607] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
α-Synuclein
(α-Syn) is an intrinsically disordered
protein which self-assembles into highly organized β-sheet structures
that accumulate in plaques in brains of Parkinson’s disease
patients. Oxidative stress influences α-Syn structure and self-assembly;
however, the basis for this remains unclear. Here we characterize
the chemical and physical effects of mild oxidation on monomeric α-Syn
and its aggregation. Using a combination of biophysical methods, small-angle
X-ray scattering, and native ion mobility mass spectrometry, we find
that oxidation leads to formation of intramolecular dityrosine cross-linkages
and a compaction of the α-Syn monomer by a factor of √2.
Oxidation-induced compaction is shown to inhibit ordered self-assembly
and amyloid formation by steric hindrance, suggesting an important
role of mild oxidation in preventing amyloid formation.
Collapse
Affiliation(s)
- Cagla Sahin
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, DK-8000 Aarhus C, Denmark
| | - Eva Christina Østerlund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Nicklas Österlund
- Department of Biochemistry and Biophysics, Stockholm University, SE-114 18 Stockholm, Sweden
| | - Joana Costeira-Paulo
- Department of Chemistry - BMC, BMC - Uppsala University, Box 576, SE-751 23 Uppsala, Sweden
| | - Jannik Nedergaard Pedersen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Gunna Christiansen
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Fredrik Bajers Vej 3b, DK-9220 Aalborg Ø, Denmark
| | - Janni Nielsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Anne Louise Grønnemose
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark.,Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Søren Kirk Amstrup
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, DK-8000 Aarhus C, Denmark
| | - Manish K Tiwari
- Department Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen Ø, Denmark
| | - R Shyama Prasad Rao
- Biostatistics and Bioinformatics Division, Yenepoya Research Center, Yenepoya University, Mangaluru-575018, Karnataka, India
| | - Morten Jannik Bjerrum
- Department Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen Ø, Denmark
| | - Leopold L Ilag
- Department of Materials and Environmental Chemistry, Stockholm University, SE-114 18 Stockholm, Sweden
| | - Michael J Davies
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Erik G Marklund
- Department of Chemistry - BMC, BMC - Uppsala University, Box 576, SE-751 23 Uppsala, Sweden
| | - Jan Skov Pedersen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark.,Department of Chemistry, Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Michael Landreh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, SE-171 65 Solna, Sweden
| | - Ian Max Møller
- Department of Molecular Biology and Genetics, Aarhus University, Forsøgsvej 1, DK-4200 Slagelse, Denmark
| | - Thomas J D Jørgensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Daniel Erik Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, DK-8000 Aarhus C, Denmark
| |
Collapse
|
17
|
Robustelli P, Ibanez-de-Opakua A, Campbell-Bezat C, Giordanetto F, Becker S, Zweckstetter M, Pan AC, Shaw DE. Molecular Basis of Small-Molecule Binding to α-Synuclein. J Am Chem Soc 2022; 144:2501-2510. [PMID: 35130691 PMCID: PMC8855421 DOI: 10.1021/jacs.1c07591] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
Intrinsically disordered
proteins (IDPs) are implicated in many
human diseases. They have generally not been amenable to conventional
structure-based drug design, however, because their intrinsic conformational
variability has precluded an atomic-level understanding of their binding
to small molecules. Here we present long-time-scale, atomic-level
molecular dynamics (MD) simulations of monomeric α-synuclein
(an IDP whose aggregation is associated with Parkinson’s disease)
binding the small-molecule drug fasudil in which the observed protein–ligand
interactions were found to be in good agreement with previously reported
NMR chemical shift data. In our simulations, fasudil, when bound,
favored certain charge–charge and π-stacking interactions
near the C terminus of α-synuclein but tended not to form these
interactions simultaneously, rather breaking one of these interactions
and forming another nearby (a mechanism we term dynamic shuttling). Further simulations with small molecules chosen to modify these
interactions yielded binding affinities and key structural features
of binding consistent with subsequent NMR experiments, suggesting
the potential for MD-based strategies to facilitate the rational design
of small molecules that bind with disordered proteins.
Collapse
Affiliation(s)
- Paul Robustelli
- D. E. Shaw Research, New York, New York 10036, United States.,Department of Chemistry, Dartmouth College, Hanover, New Hampshire 03755, United States
| | | | | | | | - Stefan Becker
- Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen, Germany.,Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37073 Göttingen, Germany
| | - Albert C Pan
- D. E. Shaw Research, New York, New York 10036, United States
| | - David E Shaw
- D. E. Shaw Research, New York, New York 10036, United States.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, United States
| |
Collapse
|
18
|
Lang Y, Zhang H, Yu H, Li Y, Liu X, Li M. Long non-coding RNA myocardial infarction-associated transcript promotes 1-Methyl-4-phenylpyridinium ion-induced neuronal inflammation and oxidative stress in Parkinson's disease through regulating microRNA-221-3p/ transforming growth factor /nuclear factor E2-related factor 2 axis. Bioengineered 2021; 13:930-940. [PMID: 34967706 PMCID: PMC8805986 DOI: 10.1080/21655979.2021.2015527] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
This study attempted to evaluate the role of long non-coding RNA myocardial infarction-associated transcript (LncRNA MIAT) in Parkinson’s disease (PD). The mouse model was established through intraperitoneal injection with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and in vitro model was induced by administrating cell with 1-Methyl-4-phenylpyridinium ion (MPP+). Rotarod test was conducted to evaluate the motor coordination of PD mice. In order to investigate the roles of LncRNA MIAT in neuronal inflammation and oxidative stress, MIAT shRNA (shMIAT) was transfected into MPP+-treated cells, and cell viability, cell apoptosis and oxidative stress response were evaluated. To evaluate the interactions between LncRNA MIAT and microRNA-221-3p (miR-221-3p)/TGF-β1/Nrf2, miR-221-3p mimic, miR-221-3p inhibitor, NC-inhibitor and transforming growth factor-β1 shRNA (shTGF-β1) were subsequently transfected into MPP+-treated cells. Dual-luciferase reporter gene assays were performed to determine the interaction of miR-221-3p with MIAT or TGFB receptor 1 (TGFBR1). The expressions of LncRNA MIAT, miR-221-3p, TGFBR1, transforming growth factor (TGF-β1) and nuclear factor E2-related factor 2 (Nrf2) were measured by quantitative reverse-transcription polymerase chain reaction (RT-qPCR) and immunoblotting. As a result, LncRNA MIAT was abundantly expressed in PD mice and cells, while downregulation of LncRNA MIAT promoted the survival of neurons, inhibited apoptosis and oxidative stress in neurons. LncRNA MIAT bound to miR-221-3p, and there was a negative correlation between miR-221-3p and LncRNA MIAT expression. In addition, miR-221-3p targeted TGFBR1 and suppressed TGF-β1 expression but increased Nrf2 expression. LncRNA MIAT promoted MPP+-induced neuronal injury in PD via regulating TGF-β1/Nrf2 axis through binding with miR-221-3p.
Collapse
Affiliation(s)
- Yue Lang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Hui Zhang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Haojia Yu
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Yu Li
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Xiao Liu
- Graduate School, Dalian Medical University, Dalian City, Liaoning Province, China
| | - Minjie Li
- Graduate School, Dalian Medical University, Dalian City, Liaoning Province, China
| |
Collapse
|
19
|
Fuentes-Lemus E, Hägglund P, López-Alarcón C, Davies MJ. Oxidative Crosslinking of Peptides and Proteins: Mechanisms of Formation, Detection, Characterization and Quantification. Molecules 2021; 27:15. [PMID: 35011250 PMCID: PMC8746199 DOI: 10.3390/molecules27010015] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 12/14/2022] Open
Abstract
Covalent crosslinks within or between proteins play a key role in determining the structure and function of proteins. Some of these are formed intentionally by either enzymatic or molecular reactions and are critical to normal physiological function. Others are generated as a consequence of exposure to oxidants (radicals, excited states or two-electron species) and other endogenous or external stimuli, or as a result of the actions of a number of enzymes (e.g., oxidases and peroxidases). Increasing evidence indicates that the accumulation of unwanted crosslinks, as is seen in ageing and multiple pathologies, has adverse effects on biological function. In this article, we review the spectrum of crosslinks, both reducible and non-reducible, currently known to be formed on proteins; the mechanisms of their formation; and experimental approaches to the detection, identification and characterization of these species.
Collapse
Affiliation(s)
- Eduardo Fuentes-Lemus
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark; (E.F.-L.); (P.H.)
| | - Per Hägglund
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark; (E.F.-L.); (P.H.)
| | - Camilo López-Alarcón
- Departamento de Química Física, Facultad de Química y de Farmacia, Pontificia Universidad Catolica de Chile, Santiago 7820436, Chile;
| | - Michael J. Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark; (E.F.-L.); (P.H.)
| |
Collapse
|
20
|
Di-Tyrosine Crosslinking and NOX4 Expression as Oxidative Pathological Markers in the Lungs of Patients with Idiopathic Pulmonary Fibrosis. Antioxidants (Basel) 2021; 10:antiox10111833. [PMID: 34829703 PMCID: PMC8615037 DOI: 10.3390/antiox10111833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 12/27/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a noninflammatory progressive lung disease. Oxidative damage is a hallmark of IPF, but the sources and consequences of oxidant generation in the lungs are unclear. In this study, we addressed the link between the H2O2-generating enzyme NADPH oxidase 4 (NOX4) and di-tyrosine (DT), an oxidative post-translational modification in IPF lungs. We performed immunohistochemical staining for DT and NOX4 in pulmonary tissue from patients with IPF and controls using validated antibodies. In the healthy lung, DT showed little or no staining and NOX4 was mostly present in normal vascular endothelium. On the other hand, both markers were detected in several cell types in the IPF patients, including vascular smooth muscle cells and epithelium (bronchial cells and epithelial cells type II). The link between NOX4 and DT was addressed in human fibroblasts deficient for NOX4 activity (mutation in the CYBA gene). Induction of NOX4 by Transforming growth factor beta 1 (TGFβ1) in fibroblasts led to moderate DT staining after the addition of a heme-containing peroxidase in control cells but not in the fibroblasts deficient for NOX4 activity. Our data indicate that DT is a histological marker of IPF and that NOX4 can generate a sufficient amount of H2O2 for DT formation in vitro.
Collapse
|
21
|
Shan FY, Fung KM, Zieneldien T, Kim J, Cao C, Huang JH. Examining the Toxicity of α-Synuclein in Neurodegenerative Disorders. Life (Basel) 2021; 11:life11111126. [PMID: 34833002 PMCID: PMC8621244 DOI: 10.3390/life11111126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Neurodegenerative disorders are complex disorders that display a variety of clinical manifestations. The second-most common neurodegenerative disorder is Parkinson’s disease, and the leading pathological protein of the disorder is considered to be α-synuclein. Nonetheless, α-synuclein accumulation also seems to result in multiple system atrophy and dementia with Lewy bodies. In order to obtain a more proficient understanding in the pathological progression of these synucleinopathies, it is crucial to observe the post-translational modifications of α-synuclein and the conformations of α-synuclein, as well as its role in the dysfunction of cellular pathways. Abstract α-synuclein is considered the main pathological protein in a variety of neurodegenerative disorders, such as Parkinson’s disease, multiple system atrophy, and dementia with Lewy bodies. As of now, numerous studies have been aimed at examining the post-translational modifications of α-synuclein to determine their effects on α-synuclein aggregation, propagation, and oligomerization, as well as the potential cellular pathway dysfunctions caused by α-synuclein, to determine the role of the protein in disease progression. Furthermore, α-synuclein also appears to contribute to the fibrilization of tau and amyloid beta, which are crucial proteins in Alzheimer’s disease, advocating for α-synuclein’s preeminent role in neurodegeneration. Due to this, investigating the mechanisms of toxicity of α-synuclein in neurodegeneration may lead to a more proficient understanding of the timeline progression in neurodegenerative synucleinopathies and could thereby lead to the development of potent targeted therapies.
Collapse
Affiliation(s)
- Frank Y. Shan
- Department of Anatomic Pathology, Baylor Scott & White Medical Center, College of Medicine, Texas A&M University, Temple, TX 76508, USA
- Correspondence: (F.Y.S.); (T.Z.)
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Medical Center, University of Oklahoma, Norman, OK 73019, USA;
| | - Tarek Zieneldien
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
- Correspondence: (F.Y.S.); (T.Z.)
| | - Janice Kim
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
| | - Jason H. Huang
- Department of Neurosurgery, Baylor Scott & White Medical Center, College of Medicine, Texas A&M University, Temple, TX 76508, USA;
| |
Collapse
|
22
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
23
|
New insights into the mechanisms of age-related protein-protein crosslinking in the human lens. Exp Eye Res 2021; 209:108679. [PMID: 34147508 DOI: 10.1016/j.exer.2021.108679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/31/2022]
Abstract
Although protein crosslinking is often linked with aging as well as some age-related diseases, very few molecular details are available on the nature of the amino acids involved, or mechanisms that are responsible for crosslinking. Recent research has shown that several amino acids are able to generate reactive intermediates that ultimately lead to covalent crosslinking through multiple non-enzymatic mechanisms. This information has been derived from proteomic investigations on aged human lenses and the mechanisms of crosslinking, in each case, have been elucidated using model peptides. Residues involved in spontaneous protein-protein crosslinking include aspartic acid, asparagine, cysteine, lysine, phosphoserine, phosphothreonine, glutamic acid and glutamine. It has become clear, therefore, that several amino acids can act as potential sites for crosslinking in the long-lived proteins that are present in aged individuals. Moreover, the lens has been an invaluable model tissue and source of crosslinked proteins from which to determine crosslinking mechanisms that may lead to crosslinking in other human tissues.
Collapse
|
24
|
Cheng W, Zhou L, Hu K, Kong D, Huang W, Xu C, Li H, Li J. Enzyme-Initiated Assembly of an Extracellular-Like Two-Dimensional Nanonetwork as a Method to Detect Procancerous Activity. ACS Sens 2021; 6:1815-1822. [PMID: 33909412 DOI: 10.1021/acssensors.0c02651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Extracellular matrix (ECM) enzymes such as lysyl oxidase (LOX) provide a new possibility to contain the invasive progress of cancer. Unlike conventional enzymes, the activity of ECM enzymes is not simply the conversion of the substrate to the product; the amount of enzymes such as matrix metalloproteinases in the ECM changes the structural integrity and morphology of the ECM. These are all important aspects that must be monitored in a spatiotemporally coupled fashion to fully understand their procancerous effect. To achieve this goal, a new molecular probe is developed, which, unlike antibodies or aptamers, can interact with the target enzyme in a more interactive way: the probe can withdraw the metal ion cofactor of the enzyme and modulate its catalytic ability. This can lead to self-propagated cross-linking of the probes to form a network not dissimilar to the collagen and elastin network of the ECM, formed through LOX activity. Thus, the biosensing process itself is a biomimetic of what may occur in vivo in the ECM, and three distinct types of signal readouts can be simultaneously recorded on the sensing surface to provide a fuller picture of ECM enzyme activity, not achievable with traditional designs. Using this method, a parallel between the detected signal and the progress of colorectal cancer can be observed. These results may point to prospective application of this method in evaluating ECM-related tumor invasiveness in the future.
Collapse
Affiliation(s)
- Wenting Cheng
- Department of Clinical Laboratory, Nanjing Gaochun People’s Hospital, Nanjing 211300, China
| | - Lei Zhou
- School of Biological Science and Technology, University of Jinan, No. 106 Jiwei Road, Jinan, Shandong 250022, China
| | - Kai Hu
- Department of Ophthalmology, The Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210004, China
| | - Dehua Kong
- Department of Clinical Laboratory, Nanjing Gaochun People’s Hospital, Nanjing 211300, China
| | - Wei Huang
- Department of Clinical Laboratory, Nanjing Gaochun People’s Hospital, Nanjing 211300, China
| | - Chuanjun Xu
- Department of Laboratory Medicine, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Hao Li
- Department of Ophthalmology, The Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210004, China
| | - Jinlong Li
- Department of Laboratory Medicine, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| |
Collapse
|
25
|
NMR unveils an N-terminal interaction interface on acetylated-α-synuclein monomers for recruitment to fibrils. Proc Natl Acad Sci U S A 2021; 118:2017452118. [PMID: 33903234 DOI: 10.1073/pnas.2017452118] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Amyloid fibril formation of α-synuclein (αS) is associated with multiple neurodegenerative diseases, including Parkinson's disease (PD). Growing evidence suggests that progression of PD is linked to cell-to-cell propagation of αS fibrils, which leads to seeding of endogenous intrinsically disordered monomer via templated elongation and secondary nucleation. A molecular understanding of the seeding mechanism and driving interactions is crucial to inhibit progression of amyloid formation. Here, using relaxation-based solution NMR experiments designed to probe large complexes, we probe weak interactions of intrinsically disordered acetylated-αS (Ac-αS) monomers with seeding-competent Ac-αS fibrils and seeding-incompetent off-pathway oligomers to identify Ac-αS monomer residues at the binding interface. Under conditions that favor fibril elongation, we determine that the first 11 N-terminal residues on the monomer form a common binding site for both fibrils and off-pathway oligomers. Additionally, the presence of off-pathway oligomers within a fibril seeding environment suppresses seeded amyloid formation, as observed through thioflavin-T fluorescence experiments. This highlights that off-pathway αS oligomers can act as an auto-inhibitor against αS fibril elongation. Based on these data taken together with previous results, we propose a model in which Ac-αS monomer recruitment to the fibril is driven by interactions between the intrinsically disordered monomer N terminus and the intrinsically disordered flanking regions (IDR) on the fibril surface. We suggest that this monomer recruitment may play a role in the elongation of amyloid fibrils and highlight the potential of the IDRs of the fibril as important therapeutic targets against seeded amyloid formation.
Collapse
|
26
|
Maina MB, Al-Hilaly YK, Burra G, Rickard JE, Harrington CR, Wischik CM, Serpell LC. Oxidative Stress Conditions Result in Trapping of PHF-Core Tau (297-391) Intermediates. Cells 2021; 10:cells10030703. [PMID: 33809978 PMCID: PMC8005035 DOI: 10.3390/cells10030703] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 01/23/2023] Open
Abstract
The self-assembly of tau into paired helical filaments (PHFs) in neurofibrillary tangles (NFTs) is a significant event in Alzheimer's disease (AD) pathogenesis. Numerous post-translational modifications enhance or inhibit tau assembly into NFTs. Oxidative stress, which accompanies AD, induces multiple post-translational modifications in proteins, including the formation of dityrosine (DiY) cross-links. Previous studies have revealed that metal-catalysed oxidation (MCO) using Cu2+ and H2O2 leads to the formation of DiY cross-links in two misfolding proteins, Aβ and α-synuclein, associated with AD and Parkinson's disease respectively. The effect of MCO on tau remains unknown. Here, we examined the effect of MCO and ultra-violet oxidation to study the influence of DiY cross-linking on the self-assembly of the PHF-core tau fragment. We report that DiY cross-linking facilitates tau assembly into tau oligomers that fail to bind thioflavin S, lack β-sheet structure and prevents their elongation into filaments. At a higher concentration, Cu2+ (without H2O2) also facilitates the formation of these tau oligomers. The DiY cross-linked tau oligomers do not cause cell death. Our findings suggest that DiY cross-linking of pre-assembled tau promotes the formation of soluble tau oligomers that show no acute impact on cell viability.
Collapse
Affiliation(s)
- Mahmoud B. Maina
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK; (M.B.M.); (Y.K.A.-H.); (G.B.)
- College of Medical Sciences, Yobe State University, Damaturu P.M.B. 1144, Nigeria
| | - Youssra K. Al-Hilaly
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK; (M.B.M.); (Y.K.A.-H.); (G.B.)
- Chemistry Department, College of Sciences, Mustansiriyah University, Baghdad, Iraq
| | - Gunasekhar Burra
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK; (M.B.M.); (Y.K.A.-H.); (G.B.)
- Analytical Research and Development, Pharma Division, Biological E. Limited, Genome Valley, IKP-Shameerpet, Hyderabad 500 078, Telangana, India
| | - Janet E. Rickard
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (J.E.R.); (C.R.H.); (C.M.W.)
| | - Charles R. Harrington
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (J.E.R.); (C.R.H.); (C.M.W.)
- TauRx Therapeutics Ltd., Aberdeen AB24 5RP, UK
| | - Claude M. Wischik
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (J.E.R.); (C.R.H.); (C.M.W.)
- TauRx Therapeutics Ltd., Aberdeen AB24 5RP, UK
| | - Louise C. Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK; (M.B.M.); (Y.K.A.-H.); (G.B.)
- Correspondence:
| |
Collapse
|
27
|
Frataxins Emerge as New Players of the Intracellular Antioxidant Machinery. Antioxidants (Basel) 2021; 10:antiox10020315. [PMID: 33672495 PMCID: PMC7923443 DOI: 10.3390/antiox10020315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/30/2022] Open
Abstract
Frataxin is a mitochondrial protein which deficiency causes Friedreich's ataxia, a cardio-neurodegenerative disease. The lack of frataxin induces the dysregulation of mitochondrial iron homeostasis and oxidative stress, which finally causes the neuronal death. The mechanism through which frataxin regulates the oxidative stress balance is rather complex and poorly understood. While the absence of human (Hfra) and yeast (Yfh1) frataxins turn out cells sensitive to oxidative stress, this does not occur when the frataxin gene is knocked-out in E. coli. To better understand the biological roles of Hfra and Yfh1 as endogenous antioxidants, we have studied their ability to inhibit the formation of reactive oxygen species (ROS) from Cu2+- and Fe3+-catalyzed degradation of ascorbic acid. Both proteins drastically reduce the formation of ROS, and during this process they are not oxidized. In addition, we have also demonstrated that merely the presence of Yfh1 or Hfra is enough to protect a highly oxidation-prone protein such as α-synuclein. This unspecific intervention (without a direct binding) suggests that frataxins could act as a shield to prevent the oxidation of a broad set of intracellular proteins, and reinforces that idea that frataxin can be used to prevent neurological pathologies linked to an enhanced oxidative stress.
Collapse
|
28
|
Folkes LK, Bartesaghi S, Trujillo M, Wardman P, Radi R. The effects of nitric oxide or oxygen on the stable products formed from the tyrosine phenoxyl radical. Free Radic Res 2021; 55:141-153. [PMID: 33399021 DOI: 10.1080/10715762.2020.1870684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tyrosine is a critical component of many proteins and can be the subject of oxidative posttranslational modifications. Furthermore, the oxidation of tyrosine residues to phenoxyl radicals, sometimes quite stable, is essential for some enzymatic functions. The lifetime and fate of tyrosine phenoxyl radicals in biological systems are largely driven by the availability and proximity of oxidants and reductants. Tyrosine phenoxyl radicals have extremely low reactivity with molecular oxygen whereas reactions with nitric oxide are diffusion controlled. This is in contrast to equivalent reactions with tryptophanyl and cysteinyl radicals where reactions with oxygen are much faster. Despite, the quite disparate apparent reactivity of tyrosine phenoxyl radicals with oxygen and nitric oxide being known, the products of the reactions are not well established. Changes in the levels from expected basal concentrations of stable products resulting from tyrosine phenoxyl radicals, for example naturally occurring 3,3'-dityrosine, 3-nitrotyrosine, and 3-hydroxytyrosine, can be indicative of oxidative and/or nitrosative stress. Using the radiolytic generation of specific oxidizing radicals to form tyrosine phenoxyl radicals in an aqueous solution at a known rate, we have compared the products in the absence and presence of nitric oxide or oxygen. Possible reactions of the phenoxyl radicals with oxygen remain unclear although we show evidence for a small decrease in the yield of dityrosine and loss of tyrosine in the presence of 20% oxygen. Low concentrations of nitric oxide in anoxic conditions react with tyrosine phenoxyl radicals, by what is most probably through the formation of an unstable intermediate, regenerating tyrosine and forming nitrite.
Collapse
Affiliation(s)
- Lisa K Folkes
- MRC Oxford Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, UK
| | - Silvina Bartesaghi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| | - Peter Wardman
- MRC Oxford Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, UK
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
29
|
Gatin A, Billault I, Duchambon P, Van der Rest G, Sicard-Roselli C. Oxidative radicals (HO • or N 3•) induce several di-tyrosine bridge isomers at the protein scale. Free Radic Biol Med 2021; 162:461-470. [PMID: 33217505 DOI: 10.1016/j.freeradbiomed.2020.10.324] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/27/2020] [Accepted: 10/31/2020] [Indexed: 11/17/2022]
Abstract
Among protein oxidative damages, di-tyrosine bridges formation has been evidenced in many neuropathological diseases. Combining oxidative radical production by gamma radiolysis with very performant chromatographic separation coupled to mass spectrometry detection, we brought into light new insights of tyrosine dimerization. Hydroxyl and azide radical tyrosine oxidation leading to di-tyrosine bridges formation was studied for different biological compounds: a full-length protein (Δ25-centrin 2), a five amino acid peptide (KTSLY) and free tyrosine. We highlighted that both radicals generate high proportion of dimers even for low doses. Surprisingly, no less than five different di-tyrosine isomers were evidenced for the protein and the peptide. For tyrosine alone, at least four distinct dimers were evidenced. These results raise some questions about their respective role in vivo and hence their relative toxicity. Also, as di-tyrosine is often used as a biomarker, a better knowledge of the type of dimer detected in vivo is now required.
Collapse
Affiliation(s)
- Anouchka Gatin
- Université Paris-Saclay, CNRS, Institut de Chimie Physique UMR 8000, 91405, Orsay Cedex, France
| | - Isabelle Billault
- Université Paris-Saclay, CNRS, Institut de Chimie Physique UMR 8000, 91405, Orsay Cedex, France
| | - Patricia Duchambon
- CNRS UMR9187, INSERM U1196, Institut Curie, Université Paris Saclay, 91405, Orsay Cedex, France
| | - Guillaume Van der Rest
- Université Paris-Saclay, CNRS, Institut de Chimie Physique UMR 8000, 91405, Orsay Cedex, France
| | - Cécile Sicard-Roselli
- Université Paris-Saclay, CNRS, Institut de Chimie Physique UMR 8000, 91405, Orsay Cedex, France.
| |
Collapse
|
30
|
Megadalton-sized Dityrosine Aggregates of α-Synuclein Retain High Degrees of Structural Disorder and Internal Dynamics. J Mol Biol 2020; 432:166689. [PMID: 33211011 PMCID: PMC7779668 DOI: 10.1016/j.jmb.2020.10.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
Despite their large size, αSyn dityrosine aggregates are dynamic and disordered. αSyn dityrosine aggregates specifically form in complex environments. αSyn dityrosine aggregates retain residual membrane binding activity. Dityrosine aggregates inhibit amyloid formation of monomeric αSyn. αSyn dityrosine aggregates are not cytotoxic.
Heterogeneous aggregates of the human protein α-synuclein (αSyn) are abundantly found in Lewy body inclusions of Parkinson’s disease patients. While structural information on classical αSyn amyloid fibrils is available, little is known about the conformational properties of disease-relevant, non-canonical aggregates. Here, we analyze the structural and dynamic properties of megadalton-sized dityrosine adducts of αSyn that form in the presence of reactive oxygen species and cytochrome c, a proapoptotic peroxidase that is released from mitochondria during sustained oxidative stress. In contrast to canonical cross-β amyloids, these aggregates retain high degrees of internal dynamics, which enables their characterization by solution-state NMR spectroscopy. We find that intermolecular dityrosine crosslinks restrict αSyn motions only locally whereas large segments of concatenated molecules remain flexible and disordered. Indistinguishable aggregates form in crowded in vitro solutions and in complex environments of mammalian cell lysates, where relative amounts of free reactive oxygen species, rather than cytochrome c, are rate limiting. We further establish that dityrosine adducts inhibit classical amyloid formation by maintaining αSyn in its monomeric form and that they are non-cytotoxic despite retaining basic membrane-binding properties. Our results suggest that oxidative αSyn aggregation scavenges cytochrome c’s activity into the formation of amorphous, high molecular-weight structures that may contribute to the structural diversity of Lewy body deposits.
Collapse
|
31
|
Pignataro MF, Herrera MG, Dodero VI. Evaluation of Peptide/Protein Self-Assembly and Aggregation by Spectroscopic Methods. Molecules 2020; 25:E4854. [PMID: 33096797 PMCID: PMC7587993 DOI: 10.3390/molecules25204854] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023] Open
Abstract
The self-assembly of proteins is an essential process for a variety of cellular functions including cell respiration, mobility and division. On the other hand, protein or peptide misfolding and aggregation is related to the development of Parkinson's disease and Alzheimer's disease, among other aggregopathies. As a consequence, significant research efforts are directed towards the understanding of this process. In this review, we are focused on the use of UV-Visible Absorption Spectroscopy, Fluorescence Spectroscopy and Circular Dichroism to evaluate the self-organization of proteins and peptides in solution. These spectroscopic techniques are commonly available in most chemistry and biochemistry research laboratories, and together they are a powerful approach for initial as well as routine evaluation of protein and peptide self-assembly and aggregation under different environmental stimulus. Furthermore, these spectroscopic techniques are even suitable for studying complex systems like those in the food industry or pharmaceutical formulations, providing an overall idea of the folding, self-assembly, and aggregation processes, which is challenging to obtain with high-resolution methods. Here, we compiled and discussed selected examples, together with our results and those that helped us better to understand the process of protein and peptide aggregation. We put particular emphasis on the basic description of the methods as well as on the experimental considerations needed to obtain meaningful information, to help those who are just getting into this exciting area of research. Moreover, this review is particularly useful to those out of the field who would like to improve reproducibility in their cellular and biomedical experiments, especially while working with peptide and protein systems as an external stimulus. Our final aim is to show the power of these low-resolution techniques to improve our understanding of the self-assembly of peptides and proteins and translate this fundamental knowledge in biomedical research or food applications.
Collapse
Affiliation(s)
- María Florencia Pignataro
- Department of Physiology and Molecular and Cellular Biology, Institute of Biosciences, Biotechnology and Translational Biology (iB3), Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires C1428EG, Argentina;
- Institute of Biological Chemistry and Physical Chemistry, Dr. Alejandro Paladini, University of Buenos Aires-CONICET, Buenos Aires C1113AAD, Argentina
| | - María Georgina Herrera
- Department of Physiology and Molecular and Cellular Biology, Institute of Biosciences, Biotechnology and Translational Biology (iB3), Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires C1428EG, Argentina;
- Institute of Biological Chemistry and Physical Chemistry, Dr. Alejandro Paladini, University of Buenos Aires-CONICET, Buenos Aires C1113AAD, Argentina
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| | - Verónica Isabel Dodero
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| |
Collapse
|
32
|
Calvo JS, Mulpuri NV, Dao A, Qazi NK, Meloni G. Membrane insertion exacerbates the α-Synuclein-Cu(II) dopamine oxidase activity: Metallothionein-3 targets and silences all α-synuclein-Cu(II) complexes. Free Radic Biol Med 2020; 158:149-161. [PMID: 32712192 PMCID: PMC7484060 DOI: 10.1016/j.freeradbiomed.2020.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 10/23/2022]
Abstract
Copper binding to α-synuclein (α-Syn), the major component of intracellular Lewy body inclusions in substantia nigra dopaminergic neurons, potentiate its toxic redox-reactivity and plays a detrimental role in the etiology of Parkinson disease (PD). Soluble α-synuclein-Cu(II) complexes possess dopamine oxidase activity and catalyze ROS production in the presence of biological reducing agents via Cu(II)/Cu(I) redox cycling. These metal-centered redox reactivities harmfully promote the oxidation and oligomerization of α-Syn. While this chemistry has been investigated on recombinantly expressed soluble α-Syn, in vivo, α-Syn is acetylated at its N-terminus and is present in equilibrium between soluble and membrane-bound forms. This post-translational modification and membrane-binding alter the Cu(II) coordination environment and binding modes and are expected to affect the α-Syn-Cu(II) reactivity. In this work, we first investigated the reactivity of acetylated and membrane-bound complexes, and subsequently addressed whether the brain metalloprotein Zn7-metallothionein-3 (Zn7MT-3) possesses a multifaceted-role in targeting these aberrant copper interactions and consequent reactivity. Through biochemical characterization of the reactivity of the non-acetylated/N-terminally acetylated soluble or membrane-bound α-Syn-Cu(II) complexes towards dopamine, oxygen, and ascorbate, we reveal that membrane insertion dramatically exacerbates the catechol oxidase-like reactivity of α-Syn-Cu(II) as a result of a change in the Cu(II) coordination environment, thereby potentiating its toxicity. Moreover, we show that Zn7MT-3 can efficiently target all α-Syn-Cu(II) complexes through Cu(II) removal, preventing their deleterious redox activities. We demonstrate that the Cu(II) reduction by the thiolate ligands of Zn7MT-3 and the formation of Cu(I)4Zn4MT-3 featuring an unusual redox-inert Cu(I)4-thiolate cluster is the molecular mechanism responsible for the protective effect exerted by MT-3 towards α-Syn-Cu(II). This work provides the molecular basis for new therapeutic interventions to control the deleterious bioinorganic chemistry of α-Syn-Cu(II).
Collapse
Affiliation(s)
- Jenifer S Calvo
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Neha V Mulpuri
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Alex Dao
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Nabeeha K Qazi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Gabriele Meloni
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
33
|
Metal- and UV- Catalyzed Oxidation Results in Trapped Amyloid-β Intermediates Revealing that Self-Assembly Is Required for Aβ-Induced Cytotoxicity. iScience 2020; 23:101537. [PMID: 33083713 PMCID: PMC7516296 DOI: 10.1016/j.isci.2020.101537] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/05/2020] [Accepted: 09/03/2020] [Indexed: 02/08/2023] Open
Abstract
Dityrosine (DiY), via the cross-linking of tyrosine residues, is a marker of protein oxidation, which increases with aging. Amyloid-β (Aβ) forms DiY in vitro and DiY-cross-linked Aβ is found in the brains of patients with Alzheimer disease. Metal- or UV- catalyzed oxidation of Aβ42 results in an increase in DiY cross-links. Using DiY as a marker of oxidation, we compare the self-assembly propensity and DiY cross-link formation for a non-assembly competent variant of Aβ42 (vAβ) with wild-type Aβ42. Oxidation results in the formation of trapped wild-type Aβ assemblies with increased DiY cross-links that are unable to elongate further. Assembly-incompetent vAβ and trapped Aβ assemblies are non-toxic to neuroblastoma cells at all stages of self-assembly, in contrast to oligomeric, non-cross-linked Aβ. These findings point to a mechanism of toxicity that necessitates dynamic self-assembly whereby trapped Aβ assemblies and assembly-incompetent variant Aβ are unable to result in cell death. Metal- (Cu2+ H202) or UV- catalyzedoxidation results in dityrosine (DiY) formation Oxidation results in DiY cross-link formation in Aβ and halts further assembly Non-assembling Aβ (trapped Aβ or variant Αβ monomer) is not cytotoxic
Collapse
|
34
|
Curry AM, Fernàndez RD, Pagani TD, Abeyawardhane DL, Trahan ML, Lucas HR. Mapping of Photochemically-Derived Dityrosine across Fe-Bound N-Acetylated α-Synuclein. Life (Basel) 2020; 10:life10080124. [PMID: 32726960 PMCID: PMC7459884 DOI: 10.3390/life10080124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 01/27/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurological disease and belongs to a group of neurodegenerative disorders called synucleinopathies in which pathological aggregates of N-terminally acetylated α-synuclein (NAcα-Syn) accumulate in various regions of the brain. In PD, these NAcα-Syn aggregates have been found to contain covalent dityrosine crosslinks, which can occur either intermolecularly or intramolecularly. Cerebral metal imbalance is also a hallmark of PD, warranting investigations into the effects of brain biometals on NAcα-Syn. NAcα-Syn is an intrinsically disordered protein, and metal-mediated conformational modifications of this structurally dynamic protein have been demonstrated to influence its propensity for dityrosine formation. In this study, a library of tyrosine-to-phenylalanine (Y-to-F) NAcα-Syn constructs were designed in order to elucidate the nature and the precise residues involved in dityrosine crosslinking of Fe-bound NAcα-Syn. The structural capacity of each mutant to form dityrosine crosslinks was assessed using Photo-Induced Cross-Linking of Unmodified Proteins (PICUP), demonstrating that coordination of either FeIII or FeII to NAcα-Syn inhibits dityrosine crosslinking among the C-terminal residues. We further demonstrate that Y39 is the main contributor to dityrosine formation of Fe-bound NAcα-Syn, while Y125 is the main residue involved in dityrosine crosslinks in unmetalated NAcα-Syn. Our results confirm that iron coordination has a global effect on NAcα-Syn structure and reactivity.
Collapse
|
35
|
Reversible Oxidative Modifications in Myoglobin and Functional Implications. Antioxidants (Basel) 2020; 9:antiox9060549. [PMID: 32599765 PMCID: PMC7346209 DOI: 10.3390/antiox9060549] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/14/2020] [Accepted: 06/18/2020] [Indexed: 12/22/2022] Open
Abstract
Myoglobin (Mb), an oxygen-binding heme protein highly expressed in heart and skeletal muscle, has been shown to undergo oxidative modifications on both an inter- and intramolecular level when exposed to hydrogen peroxide (H2O2) in vitro. Here, we show that exposure to H2O2 increases the peroxidase activity of Mb. Reaction of Mb with H2O2 causes covalent binding of heme to the Mb protein (Mb-X), corresponding to an increase in peroxidase activity when ascorbic acid is the reducing co-substrate. Treatment of H2O2-reacted Mb with ascorbic acid reverses the Mb-X crosslink. Reaction with H2O2 causes Mb to form dimers, trimers, and larger molecular weight Mb aggregates, and treatment with ascorbic acid regenerates Mb monomers. Reaction of Mb with H2O2 causes formation of dityrosine crosslinks, though the labile nature of the crosslinks broken by treatment with ascorbic acid suggests that the reversible aggregation of Mb is mediated by crosslinks other than dityrosine. Disappearance of a peptide containing a tryptophan residue when Mb is treated with H2O2 and the peptide’s reappearance after subsequent treatment with ascorbic acid suggest that tryptophan side chains might participate in the labile crosslinking. Taken together, these data suggest that while exposure to H2O2 causes Mb-X formation, increases Mb peroxidase activity, and causes Mb aggregation, these oxidative modifications are reversible by treatment with ascorbic acid. A caveat is that future studies should demonstrate that these and other in vitro findings regarding properties of Mb have relevance in the intracellular milieu, especially in regard to actual concentrations of metMb, H2O2, and ascorbate that would be found in vivo.
Collapse
|
36
|
Scutellarin inhibits the uninduced and metal-induced aggregation of α-Synuclein and disaggregates preformed fibrils: implications for Parkinson's disease. Biochem J 2020; 477:645-670. [DOI: 10.1042/bcj20190705] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/08/2020] [Accepted: 01/15/2020] [Indexed: 01/07/2023]
Abstract
The aggregation of the protein alpha synuclein (α-Syn), a known contributor in Parkinson's disease (PD) pathogenesis is triggered by transition metal ions through occupational exposure and disrupted metal ion homeostasis. Naturally occurring small molecules such as polyphenols have emerged as promising inhibitors of α-Syn fibrillation and toxicity and could be potential therapeutic agents against PD. Here, using an array of biophysical tools combined with cellular assays, we demonstrate that the novel polyphenolic compound scutellarin efficiently inhibits the uninduced and metal-induced fibrillation of α-Syn by acting at the nucleation stage and stabilizes a partially folded intermediate of α-Syn to form SDS-resistant, higher-order oligomers (∼680 kDa) and also disaggregates preformed fibrils of α-Syn into similar type of higher-order oligomers. ANS binding assay, fluorescence lifetime measurements and cell-toxicity experiments reveal scutellarin-generated oligomers as compact, low hydrophobicity structures with modulated surface properties and significantly reduced cytotoxicity than the fibrillation intermediates of α-Syn control. Fluorescence spectroscopy and isothermal titration calorimetry establish the binding between scutellarin and α-Syn to be non-covalent in nature and of moderate affinity (Ka ∼ 105 M−1). Molecular docking approaches suggest binding of scutellarin to the residues present in the NAC region and C-terminus of monomeric α-Syn and the C-terminal residues of fibrillar α-Syn, demonstrating inhibition of fibrillation upon binding to these residues and possible stabilization of the autoinhibitory conformation of α-Syn. These findings reveal interesting insights into the mechanism of scutellarin action and establish it as an efficient modulator of uninduced as well as metal-induced α-Syn fibrillation and toxicity.
Collapse
|
37
|
Ozyurt VH, Otles S. Investigation of the effect of sodium nitrite on protein oxidation markers in food protein suspensions. J Food Biochem 2020; 44:e13152. [PMID: 31950521 DOI: 10.1111/jfbc.13152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/08/2019] [Accepted: 01/02/2020] [Indexed: 11/29/2022]
Abstract
The aim of this study is to investigate the effect of sodium nitrite (NaNO2 ) on protein oxidation and the use of 3-nitrotyrosine (3NT) as a protein oxidation marker in suspensions of the food protein. Food proteins, namely bovine serum albumin, casein, and myofibrillar protein, were suspended in 100 mM sodium phosphate buffer and nitrated with 25 µM iron (III) chloride, 2.5 mM hydrogen peroxide, and 150 mg/kg NaNO2 at 37°C for a period of 24 hr. The food protein suspensions were analyzed at different sampling periods for the loss of tryptophan (TRY) residues as well as the formation of Schiff bases (SBs), protein carbonyls, 3NT, and dityrosine (DT). It was found that NaNO2 has pro-oxidant activity in NaNO2 -added food protein suspensions due to the increased amounts of SBs, protein carbonyls, 3NT, and DT as well as decreased TRY fluorescence. Positive correlations between the 3NT and other protein oxidation markers except for TRY fluorescence were found in NaNO2 -added food protein suspensions. In conclusion, these findings on the detection of 3NT indicate that it might be a useful tool as a new protein oxidation biomarker in food samples. PRACTICAL APPLICATIONS: Sodium nitrite (NaNO2 ) supports protein oxidation in different food protein suspension. 3-nitrotyrosine (3NT) was found in food protein suspensions and can be a potential biomarker for protein nitration in food sample due to potential relationship between 3NT and other oxidation markers. The results showed that this study has formed novel insight into interaction between NaNO2 and food proteins and indicate that it might affect the food quality and its nutritional value. Moreover, the relationship between protein oxidation/nitration and food quality as well as the comprehension of the scientific and technological meaning of these phenomena has been hindered because of the lack of knowledge about the basic chemistry behind the protein oxidation and nitration pathways. For the clarification of these phenomena, further studies are still needed.
Collapse
Affiliation(s)
- Vasfiye Hazal Ozyurt
- Faculty of Engineering, Department of Food Engineering, Near East University, Lefkosa, Turkey.,Faculty of Engineering, Department of Food Engineering, Ege University, Izmir, Turkey
| | - Semih Otles
- Faculty of Engineering, Department of Food Engineering, Ege University, Izmir, Turkey
| |
Collapse
|
38
|
Figueroa JD, Zárate AM, Fuentes-Lemus E, Davies MJ, López-Alarcón C. Formation and characterization of crosslinks, including Tyr–Trp species, on one electron oxidation of free Tyr and Trp residues by carbonate radical anion. RSC Adv 2020; 10:25786-25800. [PMID: 35518626 PMCID: PMC9055361 DOI: 10.1039/d0ra04051g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/28/2020] [Indexed: 01/04/2023] Open
Abstract
Dityrosine and ditryptophan bonds have been implied in protein crosslinking. This is associated with oxidative stress conditions including those involved in neurodegenerative pathologies and age-related processes. Formation of dityrosine and ditryptophan derives from radical–radical reactions involving Tyr˙ and Trp˙ radicals. However, cross reactions of Tyr˙ and Trp˙ leading to Tyr–Trp crosslinks and their biological consequences have been less explored. In the present work we hypothesized that exposure of free Tyr and Trp to a high concentration of carbonate anion radicals (CO3˙−), under anaerobic conditions, would result in the formation of Tyr–Trp species, as well as dityrosine and ditryptophan crosslinks. Here we report a simple experimental procedure, employing CO3˙− generated photochemically by illumination of a Co(iii) complex at 254 nm, that produces micromolar concentrations of Tyr–Trp crosslinks. Analysis by mass spectrometry of solutions containing only the individual amino acids, and the Co(iii) complex, provided evidence for the formation of o,o′-dityrosine and isodityrosine from Tyr, and three ditryptophan dimers from Trp. When mixtures of Tyr and Trp were illuminated in an identical manner, Tyr–Trp crosslinks were detected together with dityrosine and ditryptophan dimers. These results indicate that there is a balance between the formation of these three classes of crosslinks, which is dependent on the Tyr and Trp concentrations. The methods reported here allow the generation of significant yields of isolated Tyr–Trp adducts and their characterization. This technology should facilitate the detection, and examination of the biological consequences of Tyr–Trp crosslink formation in complex systems in future investigations. Exposure of free Tyr and Trp to a high concentration of carbonate anion radicals (CO3˙−), under anaerobic conditions, result in the formation of Tyr–Trp species, as well as dityrosine and ditryptophan crosslinks.![]()
Collapse
Affiliation(s)
- Juan David Figueroa
- Pontificia Universidad Católica de Chile, Facultad de Química y de Farmacia
- Departamento de Química Física
- Santiago
- Chile
| | - Ana María Zárate
- Pontificia Universidad Católica de Chile, Facultad de Química y de Farmacia
- Departamento de Química Física
- Santiago
- Chile
| | - Eduardo Fuentes-Lemus
- Pontificia Universidad Católica de Chile, Facultad de Química y de Farmacia
- Departamento de Química Física
- Santiago
- Chile
| | - Michael J. Davies
- University of Copenhagen
- Department of Biomedical Sciences
- Copenhagen
- Denmark
| | - Camilo López-Alarcón
- Pontificia Universidad Católica de Chile, Facultad de Química y de Farmacia
- Departamento de Química Física
- Santiago
- Chile
| |
Collapse
|
39
|
Hawkins CL, Davies MJ. Detection, identification, and quantification of oxidative protein modifications. J Biol Chem 2019; 294:19683-19708. [PMID: 31672919 PMCID: PMC6926449 DOI: 10.1074/jbc.rev119.006217] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Exposure of biological molecules to oxidants is inevitable and therefore commonplace. Oxidative stress in cells arises from both external agents and endogenous processes that generate reactive species, either purposely (e.g. during pathogen killing or enzymatic reactions) or accidentally (e.g. exposure to radiation, pollutants, drugs, or chemicals). As proteins are highly abundant and react rapidly with many oxidants, they are highly susceptible to, and major targets of, oxidative damage. This can result in changes to protein structure, function, and turnover and to loss or (occasional) gain of activity. Accumulation of oxidatively-modified proteins, due to either increased generation or decreased removal, has been associated with both aging and multiple diseases. Different oxidants generate a broad, and sometimes characteristic, spectrum of post-translational modifications. The kinetics (rates) of damage formation also vary dramatically. There is a pressing need for reliable and robust methods that can detect, identify, and quantify the products formed on amino acids, peptides, and proteins, especially in complex systems. This review summarizes several advances in our understanding of this complex chemistry and highlights methods that are available to detect oxidative modifications-at the amino acid, peptide, or protein level-and their nature, quantity, and position within a peptide sequence. Although considerable progress has been made in the development and application of new techniques, it is clear that further development is required to fully assess the relative importance of protein oxidation and to determine whether an oxidation is a cause, or merely a consequence, of injurious processes.
Collapse
Affiliation(s)
- Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
40
|
Abeyawardhane DL, Curry AM, Forney AK, Roberts JW, Lucas HR. Biometals as conformational modulators of α-synuclein photochemical crosslinking. J Biol Inorg Chem 2019; 24:1261-1268. [PMID: 31728738 PMCID: PMC7334974 DOI: 10.1007/s00775-019-01738-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 10/26/2019] [Indexed: 11/28/2022]
Abstract
Metal dyshomeostasis has long been linked to Parkinson's disease (PD), and the amyloidogenic protein α-synuclein (αS) is universally recognized as a key player in PD pathology. Structural consequences upon coordination of copper and iron to αS have gained attention due to significant dyshomeostasis of both metals in the PD brain. Protein-metal association can navigate protein folding in distinctive pathways based on the identity of the bio-metal in question. In this work, we employed photo-chemical crosslinking of unmodified proteins (PICUP) to evaluate these potential metal ion-induced structural alterations in the folding dynamics of N-terminally acetylated αS (NAcαS) following metal coordination. Through fluorescence analysis and immunoblotting analyses following photoirradiation, we discovered that coordination of iron obstructs copper-promoted crosslinking. The absence of intra-molecular crosslinking upon iron association further supports its C-terminal coordination site and suggests a potential role for iron in mitigating nearby post-translational modification of tyrosine residues. Decreased fluorescence emission upon synergistic coordination of both copper and iron highlighted that although copper acts as a conformational promotor of NAcαS crosslinking, iron inhibits analogous conformational changes within the protein. The metal coordination preferences of NAcαS suggest that both competitive binding sites as well as dual metal coordination contribute to the changes in folding dynamics, unveiling unique structural orientations for NAcαS that have a direct and measureable influence on photoinitiated dityrosine crosslinks. Moreover, our findings have physiological implications in that iron overload, as is associated with PD-insulted brain tissue, may serve as a conformational block of copper-promoted protein oxidation.
Collapse
Affiliation(s)
| | - Alyson M Curry
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Ashley K Forney
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Joel W Roberts
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Heather R Lucas
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, 23284, USA.
| |
Collapse
|
41
|
Tian Y, Stanyon HF, Barritt JD, Mayet U, Patel P, Karamani E, Fusco G, Viles JH. Copper2+ Binding to α-Synuclein. Histidine50 Can Form a Ternary Complex with Cu2+ at the N-Terminus but Not a Macrochelate. Inorg Chem 2019; 58:15580-15589. [DOI: 10.1021/acs.inorgchem.9b02644] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Yao Tian
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Helen F. Stanyon
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Joseph D Barritt
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Uroosa Mayet
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Pelak Patel
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Elena Karamani
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Giuliana Fusco
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB1 1EW, United Kingdom
| | - John H. Viles
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| |
Collapse
|
42
|
Fricano A, Librizzi F, Rao E, Alfano C, Vetri V. Blue autofluorescence in protein aggregates “lighted on” by UV induced oxidation. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:140258. [DOI: 10.1016/j.bbapap.2019.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/18/2019] [Accepted: 07/26/2019] [Indexed: 11/27/2022]
|
43
|
Iron and other metals in the pathogenesis of Parkinson's disease: Toxic effects and possible detoxification. J Inorg Biochem 2019; 199:110717. [DOI: 10.1016/j.jinorgbio.2019.110717] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/24/2022]
|
44
|
Metal binding to the amyloid-β peptides in the presence of biomembranes: potential mechanisms of cell toxicity. J Biol Inorg Chem 2019; 24:1189-1196. [PMID: 31562546 DOI: 10.1007/s00775-019-01723-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/10/2019] [Indexed: 12/18/2022]
Abstract
The amyloid-β (Aβ) peptides are key molecules in Alzheimer's disease (AD) pathology. They interact with cellular membranes, and can bind metal ions outside the membrane. Certain oligomeric Aβ aggregates are known to induce membrane perturbations and the structure of these oligomers-and their membrane-perturbing effects-can be modulated by metal ion binding. If the bound metal ions are redox active, as e.g., Cu and Fe ions are, they will generate harmful reactive oxygen species (ROS) just outside the membrane surface. Thus, the membrane damage incurred by toxic Aβ oligomers is likely aggravated when redox-active metal ions are present. The combined interactions between Aβ oligomers, metal ions, and biomembranes may be responsible for at least some of the neuronal death in AD patients.
Collapse
|
45
|
Mukherjee S, Fang M, Kok WM, Kapp EA, Thombare VJ, Huguet R, Hutton CA, Reid GE, Roberts BR. Establishing Signature Fragments for Identification and Sequencing of Dityrosine Cross-Linked Peptides Using Ultraviolet Photodissociation Mass Spectrometry. Anal Chem 2019; 91:12129-12133. [DOI: 10.1021/acs.analchem.9b02986] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Soumya Mukherjee
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Mengxuan Fang
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, 3010, Australia
| | - W. Mei Kok
- University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland 4072, Australia
| | - Eugene A. Kapp
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Varsha J. Thombare
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, 3010, Australia
| | - Romain Huguet
- Thermo Fisher Scientific, San Jose, California 95134, United States
| | - Craig A. Hutton
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, 3010, Australia
| | - Gavin E. Reid
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, 3010, Australia
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Blaine R. Roberts
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
46
|
Sarafian TA, Yacoub A, Kunz A, Aranki B, Serobyan G, Cohn W, Whitelegge JP, Watson JB. Enhanced mitochondrial inhibition by 3,4-dihydroxyphenyl-acetaldehyde (DOPAL)-oligomerized α-synuclein. J Neurosci Res 2019; 97:1689-1705. [PMID: 31420910 DOI: 10.1002/jnr.24513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/24/2019] [Accepted: 07/23/2019] [Indexed: 12/29/2022]
Abstract
Oligomeric forms of α-synuclein are believed to cause mitochondrial injury, which may contribute to neurotoxicity in Parkinson's disease (PD). Here oligomers of α-synuclein were prepared using the dopamine metabolite, DOPAL (3,4-dihydroxyphenyl-acetaldehyde), in the presence of guanidinium hydrochloride. Electron microscopy, mass spectrometry, and Western blotting studies revealed enhanced and stable oligomerization with DOPAL compared with dopamine or CuCl2 /H2 O2 . Using isolated mouse brain mitochondria, DOPAL-oligomerized α-synuclein (DOS) significantly inhibited oxygen consumption rates compared with untreated, control-fibrillated, and dopamine-fibrillated synuclein, or with monomeric α-synuclein. Inhibition was greater in the presence of malate plus pyruvate than with succinate, suggesting the involvement of mitochondrial complex I. Mitochondrial membrane potential studies using fluorescent probes, JC-1, and Safranin O also detected enhanced inhibition by DOS compared with the other aggregated forms of α-synuclein. Testing a small customized chemical library, four compounds were identified that rescued membrane potential from DOS injury. While diverse in chemical structure and mechanism, each compound has been reported to interact with mitochondrial complex I. Western blotting studies revealed that none of the four compounds disrupted the oligomeric banding pattern of DOS, suggesting their protection involved direct mitochondrial interaction. The remaining set of chemicals also did not disrupt oligomeric banding, attesting to the high structural stability of this α-synuclein proteoform. DOPAL and α-synuclein are both found in dopaminergic neurons, where their levels are elevated in PD and in animal models exposed to chemical toxicants, including agricultural pesticides. The current study provides further evidence of α-synuclein-induced mitochondrial injury and a likely role in PD neuropathology.
Collapse
Affiliation(s)
- Theodore A Sarafian
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Amneh Yacoub
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Anastasia Kunz
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Burkan Aranki
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Grigor Serobyan
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Whitaker Cohn
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Julian P Whitelegge
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Joseph B Watson
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| |
Collapse
|
47
|
Martínez-Orozco H, Mariño L, Uceda AB, Ortega-Castro J, Vilanova B, Frau J, Adrover M. Nitration and Glycation Diminish the α-Synuclein Role in the Formation and Scavenging of Cu 2+-Catalyzed Reactive Oxygen Species. ACS Chem Neurosci 2019; 10:2919-2930. [PMID: 30973706 DOI: 10.1021/acschemneuro.9b00142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Human α-synuclein is a small monomeric protein (140 residues) essential to maintain the function of the dopaminergic neurons and the neuronal redox balance. However, it holds a dark side since it is able to clump inside the neurons forming insoluble aggregates known as Lewy bodies, which are considered the hallmark of Parkinson's disease. Sporadic mutations and nonenzymatic post-translational modifications are well-known to stimulate the formation of Lewy bodies. Yet, the effect of nonenzymatic post-translational modifications on the function of α-synuclein has been studied less intense. Therefore, here we study how nitration and glycation mediated by methylglyoxal affect the redox features of α-synuclein. Both diminish the ability of α-synuclein to chelate Cu2+, except when Nε-(carboxyethyl)lysine or Nε-(carboxymethyl)lysine (two advanced glycation end products highly prevalent in vivo) are formed. This results in a lower capacity to prevent the Cu-catalyzed ascorbic acid degradation and to delay the formation of H2O2. However, only methylglyoxal was able to abolish the ability of α-synuclein to inhibit the free radical release. Both nitration and glycation enhanced the α-synuclein availability to be damaged by O2•-, although glycation made α-synuclein less reactive toward HO•. Our data represent the first report describing how nonenzymatic post-translational modifications might affect the redox function of α-synuclein, thus contributing to a better understanding of its pathological implications.
Collapse
Affiliation(s)
- Humberto Martínez-Orozco
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Laura Mariño
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Ana Belén Uceda
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Joaquín Ortega-Castro
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Bartolomé Vilanova
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Juan Frau
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Miquel Adrover
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| |
Collapse
|
48
|
Cracco L, Xiao X, Nemani SK, Lavrich J, Cali I, Ghetti B, Notari S, Surewicz WK, Gambetti P. Gerstmann-Sträussler-Scheinker disease revisited: accumulation of covalently-linked multimers of internal prion protein fragments. Acta Neuropathol Commun 2019; 7:85. [PMID: 31142381 PMCID: PMC6540574 DOI: 10.1186/s40478-019-0734-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 05/09/2019] [Indexed: 12/01/2022] Open
Abstract
Despite their phenotypic heterogeneity, most human prion diseases belong to two broadly defined groups: Creutzfeldt-Jakob disease (CJD) and Gerstmann-Sträussler-Scheinker disease (GSS). While the structural characteristics of the disease-related proteinase K-resistant prion protein (resPrPD) associated with the CJD group are fairly well established, many features of GSS-associated resPrPD are unclear. Electrophoretic profiles of resPrPD associated with GSS variants typically show 6-8 kDa bands corresponding to the internal PrP fragments as well as a variable number of higher molecular weight bands, the molecular nature of which has not been investigated. Here we have performed systematic studies of purified resPrPD species extracted from GSS cases with the A117V (GSSA117V) and F198S (GSSF198S) PrP gene mutations. The combined analysis based on epitope mapping, deglycosylation treatment and direct amino acid sequencing by mass spectrometry provided a conclusive evidence that high molecular weight resPrPD species seen in electrophoretic profiles represent covalently-linked multimers of the internal ~ 7 and ~ 8 kDa fragments. This finding reveals a mechanism of resPrPD aggregate formation that has not been previously established in prion diseases.
Collapse
Affiliation(s)
- Laura Cracco
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Xiangzhu Xiao
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Satish K Nemani
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Jody Lavrich
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Ignazio Cali
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, OH, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Silvio Notari
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Witold K Surewicz
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Pierluigi Gambetti
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
49
|
Switching on Endogenous Metal Binding Proteins in Parkinson's Disease. Cells 2019; 8:cells8020179. [PMID: 30791479 PMCID: PMC6406413 DOI: 10.3390/cells8020179] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/26/2019] [Accepted: 02/05/2019] [Indexed: 12/28/2022] Open
Abstract
The formation of cytotoxic intracellular protein aggregates is a pathological signature of multiple neurodegenerative diseases. The principle aggregating protein in Parkinson’s disease (PD) and atypical Parkinson’s diseases is α-synuclein (α-syn), which occurs in neural cytoplasmic inclusions. Several factors have been found to trigger α-syn aggregation, including raised calcium, iron, and copper. Transcriptional inducers have been explored to upregulate expression of endogenous metal-binding proteins as a potential neuroprotective strategy. The vitamin-D analogue, calcipotriol, induced increased expression of the neuronal vitamin D-dependent calcium-binding protein, calbindin-D28k, and this significantly decreased the occurrence of α-syn aggregates in cells with transiently raised intracellular free Ca, thereby increasing viability. More recently, the induction of endogenous expression of the Zn and Cu binding protein, metallothionein, by the glucocorticoid analogue, dexamethasone, gave a specific reduction in Cu-dependent α-syn aggregates. Fe accumulation has long been associated with PD. Intracellularly, Fe is regulated by interactions between the Fe storage protein ferritin and Fe transporters, such as poly(C)-binding protein 1. Analysis of the transcriptional regulation of Fe binding proteins may reveal potential inducers that could modulate Fe homoeostasis in disease. The current review highlights recent studies that suggest that transcriptional inducers may have potential as novel mechanism-based drugs against metal overload in PD.
Collapse
|
50
|
Living in Promiscuity: The Multiple Partners of Alpha-Synuclein at the Synapse in Physiology and Pathology. Int J Mol Sci 2019; 20:ijms20010141. [PMID: 30609739 PMCID: PMC6337145 DOI: 10.3390/ijms20010141] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 12/24/2018] [Accepted: 12/26/2018] [Indexed: 12/18/2022] Open
Abstract
Alpha-synuclein (α-syn) is a small protein that, in neurons, localizes predominantly to presynaptic terminals. Due to elevated conformational plasticity, which can be affected by environmental factors, in addition to undergoing disorder-to-order transition upon interaction with different interactants, α-syn is counted among the intrinsically disordered proteins (IDPs) family. As with many other IDPs, α-syn is considered a hub protein. This function is particularly relevant at synaptic sites, where α-syn is abundant and interacts with many partners, such as monoamine transporters, cytoskeletal components, lipid membranes, chaperones and synaptic vesicles (SV)-associated proteins. These protein–protein and protein–lipid membrane interactions are crucial for synaptic functional homeostasis, and alterations in α-syn can cause disruption of this complex network, and thus a failure of the synaptic machinery. Alterations of the synaptic environment or post-translational modification of α-syn can induce its misfolding, resulting in the formation of oligomers or fibrillary aggregates. These α-syn species are thought to play a pathological role in neurodegenerative disorders with α-syn deposits such as Parkinson’s disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), which are referred to as synucleinopathies. Here, we aim at revising the complex and promiscuous role of α-syn at synaptic terminals in order to decipher whether α-syn molecular interactants may influence its conformational state, contributing to its aggregation, or whether they are just affected by it.
Collapse
|