1
|
Terrin F, Tesoriere A, Plotegher N, Dalla Valle L. Sex and Brain: The Role of Sex Chromosomes and Hormones in Brain Development and Parkinson's Disease. Cells 2023; 12:1486. [PMID: 37296608 PMCID: PMC10252697 DOI: 10.3390/cells12111486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Sex hormones and genes on the sex chromosomes are not only key factors in the regulation of sexual differentiation and reproduction but they are also deeply involved in brain homeostasis. Their action is crucial for the development of the brain, which presents different characteristics depending on the sex of individuals. The role of these players in the brain is fundamental in the maintenance of brain function during adulthood as well, thus being important also with respect to age-related neurodegenerative diseases. In this review, we explore the role of biological sex in the development of the brain and analyze its impact on the predisposition toward and the progression of neurodegenerative diseases. In particular, we focus on Parkinson's disease, a neurodegenerative disorder that has a higher incidence in the male population. We report how sex hormones and genes encoded by the sex chromosomes could protect from the disease or alternatively predispose toward its development. We finally underline the importance of considering sex when studying brain physiology and pathology in cellular and animal models in order to better understand disease etiology and develop novel tailored therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Nicoletta Plotegher
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| | - Luisa Dalla Valle
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| |
Collapse
|
2
|
Chakrabarti S, Bisaglia M. Oxidative Stress and Neuroinflammation in Parkinson's Disease: The Role of Dopamine Oxidation Products. Antioxidants (Basel) 2023; 12:antiox12040955. [PMID: 37107329 PMCID: PMC10135711 DOI: 10.3390/antiox12040955] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative condition affecting more than 1% of people over 65 years old. It is characterized by the preferential degeneration of nigrostriatal dopaminergic neurons, which is responsible for the motor symptoms of PD patients. The pathogenesis of this multifactorial disorder is still elusive, hampering the discovery of therapeutic strategies able to suppress the disease's progression. While redox alterations, mitochondrial dysfunctions, and neuroinflammation are clearly involved in PD pathology, how these processes lead to the preferential degeneration of dopaminergic neurons is still an unanswered question. In this context, the presence of dopamine itself within this neuronal population could represent a crucial determinant. In the present review, an attempt is made to link the aforementioned pathways to the oxidation chemistry of dopamine, leading to the formation of free radical species, reactive quinones and toxic metabolites, and sustaining a pathological vicious cycle.
Collapse
Affiliation(s)
- Sasanka Chakrabarti
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala 133207, India
| | - Marco Bisaglia
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Study Center for Neurodegeneration (CESNE), 35121 Padova, Italy
| |
Collapse
|
3
|
Bacchella C, Dell'Acqua S, Nicolis S, Monzani E, Casella L. The reactivity of copper complexes with neuronal peptides promoted by catecholamines and its impact on neurodegeneration. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
4
|
|
5
|
Bisi N, Feni L, Peqini K, Pérez-Peña H, Ongeri S, Pieraccini S, Pellegrino S. α-Synuclein: An All-Inclusive Trip Around its Structure, Influencing Factors and Applied Techniques. Front Chem 2021; 9:666585. [PMID: 34307295 PMCID: PMC8292672 DOI: 10.3389/fchem.2021.666585] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/02/2021] [Indexed: 12/15/2022] Open
Abstract
Alpha-synuclein (αSyn) is a highly expressed and conserved protein, typically found in the presynaptic terminals of neurons. The misfolding and aggregation of αSyn into amyloid fibrils is a pathogenic hallmark of several neurodegenerative diseases called synucleinopathies, such as Parkinson’s disease. Since αSyn is an Intrinsically Disordered Protein, the characterization of its structure remains very challenging. Moreover, the mechanisms by which the structural conversion of monomeric αSyn into oligomers and finally into fibrils takes place is still far to be completely understood. Over the years, various studies have provided insights into the possible pathways that αSyn could follow to misfold and acquire oligomeric and fibrillar forms. In addition, it has been observed that αSyn structure can be influenced by different parameters, such as mutations in its sequence, the biological environment (e.g., lipids, endogenous small molecules and proteins), the interaction with exogenous compounds (e.g., drugs, diet components, heavy metals). Herein, we review the structural features of αSyn (wild-type and disease-mutated) that have been elucidated up to present by both experimental and computational techniques in different environmental and biological conditions. We believe that this gathering of current knowledge will further facilitate studies on αSyn, helping the planning of future experiments on the interactions of this protein with targeting molecules especially taking into consideration the environmental conditions.
Collapse
Affiliation(s)
- Nicolò Bisi
- BioCIS, CNRS, Université Paris Saclay, Châtenay-Malabry Cedex, France
| | - Lucia Feni
- DISFARM-Dipartimento di Scienze Farmaceutiche, Sezione Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milan, Italy
| | - Kaliroi Peqini
- DISFARM-Dipartimento di Scienze Farmaceutiche, Sezione Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milan, Italy
| | - Helena Pérez-Peña
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
| | - Sandrine Ongeri
- BioCIS, CNRS, Université Paris Saclay, Châtenay-Malabry Cedex, France
| | | | - Sara Pellegrino
- DISFARM-Dipartimento di Scienze Farmaceutiche, Sezione Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
6
|
Antonschmidt L, Dervişoğlu R, Sant V, Tekwani Movellan K, Mey I, Riedel D, Steinem C, Becker S, Andreas LB, Griesinger C. Insights into the molecular mechanism of amyloid filament formation: Segmental folding of α-synuclein on lipid membranes. SCIENCE ADVANCES 2021; 7:7/20/eabg2174. [PMID: 33990334 PMCID: PMC8121418 DOI: 10.1126/sciadv.abg2174] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/25/2021] [Indexed: 05/15/2023]
Abstract
Recent advances in the structural biology of disease-relevant α-synuclein fibrils have revealed a variety of structures, yet little is known about the process of fibril aggregate formation. Characterization of intermediate species that form during aggregation is crucial; however, this has proven very challenging because of their transient nature, heterogeneity, and low population. Here, we investigate the aggregation of α-synuclein bound to negatively charged phospholipid small unilamellar vesicles. Through a combination of kinetic and structural studies, we identify key time points in the aggregation process that enable targeted isolation of prefibrillar and early fibrillar intermediates. By using solid-state nuclear magnetic resonance, we show the gradual buildup of structural features in an α-synuclein fibril filament, revealing a segmental folding process. We identify distinct membrane-binding domains in α-synuclein aggregates, and the combined data are used to present a comprehensive mechanism of the folding of α-synuclein on lipid membranes.
Collapse
Affiliation(s)
- Leif Antonschmidt
- Department of NMR-Based Structural Biology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Rıza Dervişoğlu
- Department of NMR-Based Structural Biology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Vrinda Sant
- Department of NMR-Based Structural Biology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, USA
| | - Kumar Tekwani Movellan
- Department of NMR-Based Structural Biology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Ingo Mey
- Institute of Organic and Biomolecular Chemistry, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Dietmar Riedel
- Laboratory of Electron Microscopy, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Claudia Steinem
- Institute of Organic and Biomolecular Chemistry, Georg-August-Universität Göttingen, Göttingen, Germany
- Biomolecular Chemistry Group, Max-Planck Institute for Dynamics and Self-Organization, Am Fassberg 17, 37077 Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Stefan Becker
- Department of NMR-Based Structural Biology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Loren B Andreas
- Department of NMR-Based Structural Biology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany.
| | - Christian Griesinger
- Department of NMR-Based Structural Biology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
7
|
Pike AF, Varanita T, Herrebout MAC, Plug BC, Kole J, Musters RJP, Teunissen CE, Hoozemans JJM, Bubacco L, Veerhuis R. α-Synuclein evokes NLRP3 inflammasome-mediated IL-1β secretion from primary human microglia. Glia 2021; 69:1413-1428. [PMID: 33506583 PMCID: PMC8247862 DOI: 10.1002/glia.23970] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 01/03/2023]
Abstract
Synucleinopathies such as Parkinson's disease (PD) are hallmarked by α‐synuclein (α‐syn) pathology and neuroinflammation. This neuroinflammation involves activated microglia with increased secretion of interleukin‐1β (IL‐1β). The main driver of IL‐1β secretion from microglia is the NLRP3 inflammasome. A critical link between microglial NLRP3 inflammasome activation and the progression of both α‐syn pathology and dopaminergic neurodegeneration has been identified in various PD models in vivo. α‐Syn is known to activate the microglial NLRP3 inflammasome in murine models, but its relationship to this inflammasome in human microglia has not been established. In this study, IL‐1β secretion from primary mouse microglia induced by α‐syn fibrils was dependent on NLRP3 inflammasome assembly and caspase‐1 activity, as previously reported. We show that exposure of primary human microglia to α‐syn fibrils also resulted in significant IL‐1β secretion that was dependent on inflammasome assembly and involved the recruitment of caspase‐1 protein to inflammasome scaffolds as visualized with superresolution microscopy. While canonical IL‐1β secretion was clearly dependent on caspase‐1 enzymatic activity, this activity was less clearly involved for α‐syn‐induced IL‐1β secretion from human microglia. This work presents similarities between primary human and mouse microglia in the mechanisms of activation of the NLRP3 inflammasome by α‐syn, but also highlights evidence to suggest that there may be a difference in the requirement for caspase‐1 activity in IL‐1β output. The data represent a novel characterization of PD‐related NLRP3 inflammasome activation in primary human microglia and further implicate this mechanism in the pathology underlying PD.
Collapse
Affiliation(s)
- Adrianne F Pike
- Amsterdam UMC, Vrije Universiteit Amsterdam, Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | | | - Maaike A C Herrebout
- Amsterdam UMC, Vrije Universiteit Amsterdam, Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Bonnie C Plug
- Amsterdam UMC, Vrije Universiteit Amsterdam, Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Jeroen Kole
- Amsterdam UMC, Vrije Universiteit Amsterdam, Laboratory for Physiology, Institute for Cardiovascular Research, Amsterdam, the Netherlands
| | - René J P Musters
- Amsterdam UMC, Vrije Universiteit Amsterdam, Laboratory for Physiology, Institute for Cardiovascular Research, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Jeroen J M Hoozemans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Neuropathology Laboratory, Department of Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Luigi Bubacco
- Department of Biology, University of Padua, Padua, Italy
| | - Robert Veerhuis
- Amsterdam UMC, Vrije Universiteit Amsterdam, Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, the Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, the Netherlands
| |
Collapse
|
8
|
Kang C, Sun R. Molecular Dynamics Study of the Interaction between the N-terminal of α-Synuclein and a Lipid Bilayer Mimicking Synaptic Vesicles. J Phys Chem B 2020; 125:1036-1048. [DOI: 10.1021/acs.jpcb.0c08620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Christopher Kang
- Department of Chemistry, University of Hawai’i at Manoa, 2545 McCarthy
Mall, Honolulu 96822-2275, Hawaii, United States
| | - Rui Sun
- Department of Chemistry, University of Hawai’i at Manoa, 2545 McCarthy
Mall, Honolulu 96822-2275, Hawaii, United States
| |
Collapse
|
9
|
Jain K, Ghribi O, Delhommelle J. Folding Free-Energy Landscape of α-Synuclein (35-97) Via Replica Exchange Molecular Dynamics. J Chem Inf Model 2020; 61:432-443. [PMID: 33350818 DOI: 10.1021/acs.jcim.0c01278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The misfolding and aggregation of α-synuclein (α-syn) in Lewy bodies are implicated in the pathogenesis of various neurodegenerative disorders, such as Parkinson's disease and dementia. The formation of α-syn fibrils is a complex process, involving various intermediates and oligomeric forms. These intermediates establish at an early stage of aggregation and subsequently lead to fibrillation. Determining which conformations are accessible to monomeric α-syn and especially, as shown in a recent work, to the central amino acids from residue 35 to residue 97 (63 residues) is thus crucial to understand the formation of these oligomers. Here, we carry out extensive replica exchange molecular dynamics (total time-18 μs) with an all-atom model and explicit solvent to characterize the free-energy landscape of human α-syn (residue 35 to residue 97). The simulation results lead us to identify two free-energy basins. Clustering analysis for the deepest free-energy minimum reveals a compact structure, with a secondary structure predominantly α-helix, while the shallower minimum corresponds to an elongated conformation, also predominantly α-helix. Furthermore, at physiological temperature, we find that conformational rearrangements happen via helix breaks due to the presence of glycine. We also show that the most likely conformations are characterized by the α-helix structure rather than the β-hairpin structure (for residue 38 to residue 53), in contrast with prior simulation studies using coarse-grained models or an implicit solvent. For higher temperatures, we observe a shift in secondary structure with a decrease in the population of α-helix in favor of random coils, β-bend, and β-turns.
Collapse
Affiliation(s)
- Karnesh Jain
- Department of Chemistry, University of North Dakota, Grand Forks, North Dakota 58202, United States
| | - Othman Ghribi
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, North Dakota 58202, United States
| | - Jerome Delhommelle
- Department of Chemistry, University of North Dakota, Grand Forks, North Dakota 58202, United States
| |
Collapse
|
10
|
Moretti P, Mariani P, Ortore MG, Plotegher N, Bubacco L, Beltramini M, Spinozzi F. Comprehensive Structural and Thermodynamic Analysis of Prefibrillar WT α-Synuclein and Its G51D, E46K, and A53T Mutants by a Combination of Small-Angle X-ray Scattering and Variational Bayesian Weighting. J Chem Inf Model 2020; 60:5265-5281. [PMID: 32866007 PMCID: PMC8154249 DOI: 10.1021/acs.jcim.0c00807] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Indexed: 12/13/2022]
Abstract
The in solution synchrotron small-angle X-ray scattering SAXS technique has been used to investigate an intrinsically disordered protein (IDP) related to Parkinson's disease, the α-synuclein (α-syn), in prefibrillar diluted conditions. SAXS experiments have been performed as a function of temperature and concentration on the wild type (WT) and on the three pathogenic mutants G51D, E46K, and A53T. To identify the conformers that populate WT α-syn and the pathogenic mutants in prefibrillar conditions, scattering data have been analyzed by a new variational bayesian weighting method (VBWSAS) based on an ensemble of conformers, which includes unfolded monomers, trimers, and tetramers, both in helical-rich and strand-rich forms. The developed VBWSAS method uses a thermodynamic scheme to account for temperature and concentration effects and considers long-range protein-protein interactions in the framework of the random phase approximation. The global analysis of the whole set of data indicates that WT α-syn is mostly present as unfolded monomers and trimers (helical-rich trimers at low T and strand-rich trimers at high T), but not tetramers, as previously derived by several studies. On the contrary, different conformer combinations characterize mutants. In the α-syn G51D mutant, the most abundant aggregates at all the temperatures are strand-rich tetramers. Strand-rich tetramers are also the predominant forms in the A53T mutant, but their weight decreases with temperature. Only monomeric conformers, with a preference for the ones with the smallest sizes, are present in the E46K mutant. The derived conformational behavior then suggests a different availability of species prone to aggregate, depending on mutation, temperature, and concentration and accounting for the different neurotoxicity of α-syn variants. Indeed, this approach may be of pivotal importance to describe conformational and aggregational properties of other IDPs.
Collapse
Affiliation(s)
- Paolo Moretti
- Department
of Life and Environmental Sciences, Polytechnic
University of Marche, 60131 Ancona, Marche, Italy
| | - Paolo Mariani
- Department
of Life and Environmental Sciences, Polytechnic
University of Marche, 60131 Ancona, Marche, Italy
| | - Maria Grazia Ortore
- Department
of Life and Environmental Sciences, Polytechnic
University of Marche, 60131 Ancona, Marche, Italy
| | | | - Luigi Bubacco
- Department
of Biology, University of Padova, 35121 Padova, Veneto, Italy
| | - Mariano Beltramini
- Department
of Biology, University of Padova, 35121 Padova, Veneto, Italy
| | - Francesco Spinozzi
- Department
of Life and Environmental Sciences, Polytechnic
University of Marche, 60131 Ancona, Marche, Italy
| |
Collapse
|
11
|
Li N, Stewart T, Sheng L, Shi M, Cilento EM, Wu Y, Hong JS, Zhang J. Immunoregulation of microglial polarization: an unrecognized physiological function of α-synuclein. J Neuroinflammation 2020; 17:272. [PMID: 32943057 PMCID: PMC7500012 DOI: 10.1186/s12974-020-01940-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022] Open
Abstract
Background Microglial function is vital for maintaining the health of the brain, and their activation is an essential component of neurodegeneration. There is significant research on factors that provoke “reactive” or “inflammatory” phenotypes in conditions of injury or disease. One such factor, exposure to the aggregated or oligomeric forms of α-synuclein, an abundant brain protein, plays an essential role in driving microglial activation; including chemotactic migration and production of inflammatory mediators in Lewy body (LB) diseases such as Parkinson’s disease. On the other hand, it is increasingly recognized that microglia also undergo changes, dependent on the cellular environment, that promote mainly reconstructive and anti-inflammatory functions, i.e., mostly desirable functions of microglia in a physiological state. What maintains microglia in this physiological state is essentially unknown. Methods In this study, using in vitro and in vivo models, we challenged primary microglia or BV2 microglia with LPS + IFN-γ, IL-4 + IL-13, α-synuclein monomer, and α-synuclein oligomer, and examined microglia phenotype and the underlying mechanism by RT-PCR, Western blot, ELISA, IF, IHC, Co-IP. Results We described a novel physiological function of α-synuclein, in which it modulates microglia toward an anti-inflammatory phenotype by interaction with extracellular signal-regulated kinase (ERK) and recruitment of the ERK, nuclear factor kappa B (NF-κB), and peroxisome proliferator-activated receptor γ (PPARγ) pathways. Conclusions These findings suggest a previously unrecognized function of monomeric α-synuclein that likely gives new insights into the pathogenesis and potential therapies for Lewy body-related diseases and beyond, given the abundance and multiple functions of α-synuclein in brain tissue.
Collapse
Affiliation(s)
- Na Li
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China.,Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Tessandra Stewart
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Lifu Sheng
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Min Shi
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Eugene M Cilento
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Yufeng Wu
- Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Jau-Syong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Jing Zhang
- Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China. .,Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98104, USA. .,Department of Pathology, Zhejiang University First Affiliated Hospital and School of Medicine, Hangzhou, Zhejiang, 310002, China.
| |
Collapse
|
12
|
Bacchella C, Nicolis S, Dell'Acqua S, Rizzarelli E, Monzani E, Casella L. Membrane Binding Strongly Affecting the Dopamine Reactivity Induced by Copper Prion and Copper/Amyloid-β (Aβ) Peptides. A Ternary Copper/Aβ/Prion Peptide Complex Stabilized and Solubilized in Sodium Dodecyl Sulfate Micelles. Inorg Chem 2019; 59:900-912. [PMID: 31869218 DOI: 10.1021/acs.inorgchem.9b03153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The combination between dyshomeostatic levels of catecholamine neurotransmitters and redox-active metals such as copper and iron exacerbates the oxidative stress condition that typically affects neurodegenerative diseases. We report a comparative study of the oxidative reactivity of copper complexes with amyloid-β (Aβ40) and the prion peptide fragment 76-114 (PrP76-114), containing the high-affinity binding site, toward dopamine and 4-methylcatechol, in aqueous buffer and in sodium dodecyl sulfate micelles, as a model membrane environment. The competitive oxidative and covalent modifications undergone by the peptides were also evaluated. The high binding affinity of Cu/peptide to micelles and lipid membranes leads to a strong reduction (Aβ40) and quenching (PrP76-114) of the oxidative efficiency of the binary complexes and to a stabilization and redox silencing of the ternary complex CuII/Aβ40/PrP76-114, which is highly reactive in solution. The results improve our understanding of the pathological and protective effects associated with these complexes, depending on the physiological environment.
Collapse
Affiliation(s)
- Chiara Bacchella
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Stefania Nicolis
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Simone Dell'Acqua
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Enrico Rizzarelli
- Istituto di Biostrutture e Bioimmagini , Consiglio Nazionale delle Ricerche , Via P. Gaifami 18 , 95125 Catania , Italy
| | - Enrico Monzani
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| | - Luigi Casella
- Dipartimento di Chimica , Università di Pavia , Via Taramelli 12 , 27100 Pavia , Italy
| |
Collapse
|
13
|
Monzani E, Nicolis S, Dell'Acqua S, Capucciati A, Bacchella C, Zucca FA, Mosharov EV, Sulzer D, Zecca L, Casella L. Dopamin, oxidativer Stress und Protein‐Chinonmodifikationen bei Parkinson und anderen neurodegenerativen Erkrankungen. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201811122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Enrico Monzani
- Department of ChemistryUniversity of Pavia 27100 Pavia Italien
| | | | | | | | | | - Fabio A. Zucca
- Institute of Biomedical TechnologiesNational Research Council of Italy Segrate (Mailand) Italien
| | - Eugene V. Mosharov
- Department of PsychiatryColumbia University Medical CenterNew York State Psychiatric Institute New York NY USA
- Departments Neurology, PharmacologyColumbia University Medical Center New York NY USA
| | - David Sulzer
- Department of PsychiatryColumbia University Medical CenterNew York State Psychiatric Institute New York NY USA
- Departments Neurology, PharmacologyColumbia University Medical Center New York NY USA
| | - Luigi Zecca
- Institute of Biomedical TechnologiesNational Research Council of Italy Segrate (Mailand) Italien
- Department of PsychiatryColumbia University Medical CenterNew York State Psychiatric Institute New York NY USA
| | - Luigi Casella
- Department of ChemistryUniversity of Pavia 27100 Pavia Italien
| |
Collapse
|
14
|
Monzani E, Nicolis S, Dell'Acqua S, Capucciati A, Bacchella C, Zucca FA, Mosharov EV, Sulzer D, Zecca L, Casella L. Dopamine, Oxidative Stress and Protein-Quinone Modifications in Parkinson's and Other Neurodegenerative Diseases. Angew Chem Int Ed Engl 2019; 58:6512-6527. [PMID: 30536578 DOI: 10.1002/anie.201811122] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/10/2018] [Indexed: 12/19/2022]
Abstract
Dopamine (DA) is the most important catecholamine in the brain, as it is the most abundant and the precursor of other neurotransmitters. Degeneration of nigrostriatal neurons of substantia nigra pars compacta in Parkinson's disease represents the best-studied link between DA neurotransmission and neuropathology. Catecholamines are reactive molecules that are handled through complex control and transport systems. Under normal conditions, small amounts of cytosolic DA are converted to neuromelanin in a stepwise process involving melanization of peptides and proteins. However, excessive cytosolic or extraneuronal DA can give rise to nonselective protein modifications. These reactions involve DA oxidation to quinone species and depend on the presence of redox-active transition metal ions such as iron and copper. Other oxidized DA metabolites likely participate in post-translational protein modification. Thus, protein-quinone modification is a heterogeneous process involving multiple DA-derived residues that produce structural and conformational changes of proteins and can lead to aggregation and inactivation of the modified proteins.
Collapse
Affiliation(s)
- Enrico Monzani
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| | - Stefania Nicolis
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| | | | | | - Chiara Bacchella
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| | - Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate (Milano), Italy
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA.,Departments of Neurology and Pharmacology, Columbia University Medical Center, New York, NY, USA
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate (Milano), Italy.,Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Luigi Casella
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| |
Collapse
|
15
|
Bhasne K, Sebastian S, Jain N, Mukhopadhyay S. Synergistic Amyloid Switch Triggered by Early Heterotypic Oligomerization of Intrinsically Disordered α-Synuclein and Tau. J Mol Biol 2018; 430:2508-2520. [PMID: 29704492 DOI: 10.1016/j.jmb.2018.04.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/05/2018] [Accepted: 04/16/2018] [Indexed: 12/31/2022]
Abstract
Amyloidogenic intrinsically disordered proteins, α-synuclein and tau are linked to Parkinson's disease and Alzheimer's disease, respectively. A body of evidence suggests that α-synuclein and tau, both present in the presynaptic nerve terminals, co-aggregate in many neurological ailments. The molecular mechanism of α-synuclein-tau hetero-assembly is poorly understood. Here we show that amyloid formation is synergistically facilitated by heterotypic association mediated by binding-induced misfolding of both α-synuclein and tau K18. We demonstrate that the intermolecular association is largely driven by the electrostatic interaction between the negatively charged C-terminal segment of α-synuclein and the positively charged tau K18 fragment. This heterotypic association results in rapid formation of oligomers that readily mature into hetero-fibrils with a much shorter lag phase compared to the individual proteins. These findings suggested that the critical intermolecular interaction between α-synuclein and tau can promote facile amyloid formation that can potentially lead to efficient sequestration of otherwise long-lived lethal oligomeric intermediates into innocuous fibrils. We next show that a well-known familial Parkinson's disease mutant (A30P) that is known to aggregate slowly via accumulation of highly toxic oligomeric species during the long lag phase converts into amyloid fibrils significantly faster in the presence of tau K18. The early intermolecular interaction profoundly accelerates the fibrillation rate of A30P α-synuclein and impels the disease mutant to behave similar to wild-type α-synuclein in the presence of tau. Our findings suggest a mechanistic underpinning of bypassing toxicity and suggest a general strategy by which detrimental amyloidogenic precursors are efficiently sequestered into more benign amyloid fibrils.
Collapse
Affiliation(s)
- Karishma Bhasne
- Centre for Protein Science, Design and Engineering, Indian Institute of Science Education and Research (IISER) Mohali, Punjab, India; Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Punjab, India
| | - Sanjana Sebastian
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Punjab, India
| | - Neha Jain
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Punjab, India; Present address: Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Ahmedabad, Gujarat, India
| | - Samrat Mukhopadhyay
- Centre for Protein Science, Design and Engineering, Indian Institute of Science Education and Research (IISER) Mohali, Punjab, India; Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Punjab, India; Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Punjab, India.
| |
Collapse
|
16
|
Biosa A, Outeiro TF, Bubacco L, Bisaglia M. Diabetes Mellitus as a Risk Factor for Parkinson's Disease: a Molecular Point of View. Mol Neurobiol 2018; 55:8754-8763. [PMID: 29594935 DOI: 10.1007/s12035-018-1025-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/20/2018] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder characterized by elevated concentrations of glucose in the blood. The chronic hyperglycemic state accounts for most of the vascular complications associated to the disease and the prevalent mechanism proposed is related to the glycating chemistry mediated by methylglyoxal (MG), which accumulates in T2DM. In recent years, a higher risk of Parkinson's disease (PD) onset in people affected by T2DM has become evident, but the molecular mechanisms underlying the interplay between T2DM and PD are still unknown. The oxidative chemistry of dopamine and its reactivity towards the protein α-Synuclein (aS) has been associated to the pathogenesis of PD. Recently, aS has also been described to interact with MG. Interestingly, MG and the dopamine oxidation products share both structural similarity and chemical reactivity. The ability of MG to spread over the site of its production and react with aS could represent the rationale to explain the higher incidence of PD in T2DM-affected people and may open opportunities for the development of novel strategies to antagonize the raise of PD.
Collapse
Affiliation(s)
- Alice Biosa
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, Padova, Italy
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Max Planck Institute for Experimental Medicine, Goettingen, Germany
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Luigi Bubacco
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, Padova, Italy
| | - Marco Bisaglia
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|
17
|
Cardiolipin exposure on the outer mitochondrial membrane modulates α-synuclein. Nat Commun 2018; 9:817. [PMID: 29483518 PMCID: PMC5827019 DOI: 10.1038/s41467-018-03241-9] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/30/2018] [Indexed: 11/09/2022] Open
Abstract
Neuronal loss in Parkinson's disease (PD) is associated with aberrant mitochondrial function and impaired proteostasis. Identifying the mechanisms that link these pathologies is critical to furthering our understanding of PD pathogenesis. Using human pluripotent stem cells (hPSCs) that allow comparison of cells expressing mutant SNCA (encoding α-synuclein (α-syn)) with isogenic controls, or SNCA-transgenic mice, we show that SNCA-mutant neurons display fragmented mitochondria and accumulate α-syn deposits that cluster to mitochondrial membranes in response to exposure of cardiolipin on the mitochondrial surface. Whereas exposed cardiolipin specifically binds to and facilitates refolding of α-syn fibrils, prolonged cardiolipin exposure in SNCA-mutants initiates recruitment of LC3 to the mitochondria and mitophagy. Moreover, we find that co-culture of SNCA-mutant neurons with their isogenic controls results in transmission of α-syn pathology coincident with mitochondrial pathology in control neurons. Transmission of pathology is effectively blocked using an anti-α-syn monoclonal antibody (mAb), consistent with cell-to-cell seeding of α-syn.
Collapse
|
18
|
Fauerbach JA, Jovin TM. Pre-aggregation kinetics and intermediates of α-synuclein monitored by the ESIPT probe 7MFE. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 47:345-362. [PMID: 29255947 PMCID: PMC5982440 DOI: 10.1007/s00249-017-1272-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/05/2017] [Accepted: 12/08/2017] [Indexed: 01/04/2023]
Abstract
The defining feature of the extensive family of amyloid diseases is the formation of networks of entangled elongated protein fibrils and amorphous aggregates exhibiting crossed β-sheet secondary structure. The time course of amyloid conversion has been studied extensively in vitro with the proteins involved in the neurodegenerative pathology of Parkinson's disease (α-synuclein), Alzheimer's disease (Tau) and Huntington's disease (Huntingtin). Although much is known about the thermodynamics and kinetics of the transition from a soluble, intrinsically disordered monomer to the fibrillar end state, the putative oligomeric intermediates, currently considered to be the major initiators of cellular toxicity, are as yet poorly defined. We have detected and characterized amyloid precursors by monitoring AS aggregation with ESIPT (excited state intramolecular protein transfer) probes, one of which, 7MFE [7-(3-maleimido-N-propanamide)-2-(4-diethyaminophenyl)-3-hydroxychromone], is introduced here and compared with a related compound, 6MFC, used previously. A series of 140 spectra for sparsely labeled AS was acquired during the course of aggregation, and resolved into the relative contributions (spectra, intensities) of discrete molecular species including the monomeric, fibrillar, and ensemble of intermediate forms. Based on these findings, a kinetic scheme was devised to simulate progress curves as a function of key parameters. An essential feature of the model, one not previously invoked in schemes of amyloid aggregation, is the catalysis of molecular fuzziness by discrete colloidal nanoparticles arising spontaneously via monomer condensation upon exposure of AS to ≥ 37 °C.
Collapse
Affiliation(s)
- Jonathan A Fauerbach
- Miltenyi Biotec GmbH, Friedrich-Ebert Str. 42, 51429, Bergisch-Gladbach, Germany
| | - Thomas M Jovin
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
19
|
Plotegher N, Duchen MR. Crosstalk between Lysosomes and Mitochondria in Parkinson's Disease. Front Cell Dev Biol 2017; 5:110. [PMID: 29312935 PMCID: PMC5732996 DOI: 10.3389/fcell.2017.00110] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/30/2017] [Indexed: 11/15/2022] Open
Abstract
Parkinson's disease (PD) is the most common motor neurodegenerative disorder. In most cases the cause of the disease is unknown, while in about 10% of subjects, it is associated with mutations in a number of different genes. Several different mutations in 15 genes have been identified as causing familial forms of the disease, while many others have been identified as risk factors. A striking number of these genes are either involved in the regulation of mitochondrial function or of endo-lysosomal pathways. Mutations affecting one of these two pathways are often coupled with defects in the other pathway, suggesting a crosstalk between them. Moreover, PD-linked mutations in genes encoding proteins with other functions are frequently associated with defects in mitochondrial and/or autophagy/lysosomal function as a secondary effect. Even toxins that impair mitochondrial function and cause parkinsonian phenotypes, such as rotenone, also impair lysosomal function. In this review, we explore the reciprocal relationship between mitochondrial and lysosomal pathways in PD. We will discuss the impact of mitochondrial dysfunction on the lysosomal compartment and of endo-lysosomal defects on mitochondrial function, and explore the roles of both causative genes and genes that are risk factors for PD. Understanding the pathways that govern these interactions should help to define a framework to understand the roles and mechanisms of mitochondrial and lysosomal miscommunication in the pathophysiology of PD.
Collapse
Affiliation(s)
- Nicoletta Plotegher
- Department of Cell and Developmental Biology and UCL Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Michael R Duchen
- Department of Cell and Developmental Biology and UCL Consortium for Mitochondrial Research, University College London, London, United Kingdom
| |
Collapse
|
20
|
Piccirilli F, Plotegher N, Ortore MG, Tessari I, Brucale M, Spinozzi F, Beltramini M, Mariani P, Militello V, Lupi S, Perucchi A, Bubacco L. High-Pressure-Driven Reversible Dissociation of α-Synuclein Fibrils Reveals Structural Hierarchy. Biophys J 2017; 113:1685-1696. [PMID: 29045863 DOI: 10.1016/j.bpj.2017.08.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/20/2017] [Accepted: 08/04/2017] [Indexed: 01/02/2023] Open
Abstract
The analysis of the α-synuclein (aS) aggregation process, which is involved in Parkinson's disease etiopathogenesis, and of the structural feature of the resulting amyloid fibrils may shed light on the relationship between the structure of aS aggregates and their toxicity. This may be considered a paradigm of the ground work needed to tackle the molecular basis of all the protein-aggregation-related diseases. With this aim, we used chemical and physical dissociation methods to explore the structural organization of wild-type aS fibrils. High pressure (in the kbar range) and alkaline pH were used to disassemble fibrils to collect information on the hierarchic pathway by which distinct β-sheets sequentially unfold using the unique possibility offered by high-pressure Fourier transform infrared spectroscopy. The results point toward the formation of kinetic traps in the energy landscape of aS fibril disassembly and the presence of transient partially folded species during the process. Since we found that the dissociation of wild-type aS fibrils by high pressure is reversible upon pressure release, the disassembled molecules likely retain structural information that favors fibril reformation. To deconstruct the role of the different regions of aS sequence in this process, we measured the high-pressure dissociation of amyloids formed by covalent chimeric dimers of aS (syn-syn) and by the aS deletion mutant that lacks the C-terminus, i.e., aS (1-99). The results allowed us to single out the role of dimerization and that of the C-terminus in the complete maturation of fibrillar aS.
Collapse
Affiliation(s)
- Federica Piccirilli
- CNR-IOM, University of Trieste, Trieste, Italy; Physics Department, University of Trieste, Trieste, Italy
| | - Nicoletta Plotegher
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Maria Grazia Ortore
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | | | | | - Francesco Spinozzi
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | | | - Paolo Mariani
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Valeria Militello
- Department of Physics and Chemistry, University of Palermo, Palermo, Italy
| | - Stefano Lupi
- Physics Department, University of Trieste, Trieste, Italy; Dipartimento di Fisica Università di Roma "La Sapienza", Rome, Italy
| | | | - Luigi Bubacco
- Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|
21
|
Pressure effects on α-synuclein amyloid fibrils: An experimental investigation on their dissociation and reversible nature. Arch Biochem Biophys 2017. [DOI: 10.1016/j.abb.2017.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
22
|
Mitochondrial Dysfunction and Neurodegeneration in Lysosomal Storage Disorders. Trends Mol Med 2017; 23:116-134. [DOI: 10.1016/j.molmed.2016.12.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 12/22/2022]
|
23
|
DOPAL derived alpha-synuclein oligomers impair synaptic vesicles physiological function. Sci Rep 2017; 7:40699. [PMID: 28084443 PMCID: PMC5233976 DOI: 10.1038/srep40699] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/06/2016] [Indexed: 12/15/2022] Open
Abstract
Parkinson’s disease is a neurodegenerative disorder characterized by the death of dopaminergic neurons and by accumulation of alpha-synuclein (aS) aggregates in the surviving neurons. The dopamine catabolite 3,4-dihydroxyphenylacetaldehyde (DOPAL) is a highly reactive and toxic molecule that leads to aS oligomerization by covalent modifications to lysine residues. Here we show that DOPAL-induced aS oligomer formation in neurons is associated with damage of synaptic vesicles, and with alterations in the synaptic vesicles pools. To investigate the molecular mechanism that leads to synaptic impairment, we first aimed to characterize the biochemical and biophysical properties of the aS-DOPAL oligomers; heterogeneous ensembles of macromolecules able to permeabilise cholesterol-containing lipid membranes. aS-DOPAL oligomers can induce dopamine leak in an in vitro model of synaptic vesicles and in cellular models. The dopamine released, after conversion to DOPAL in the cytoplasm, could trigger a noxious cycle that further fuels the formation of aS-DOPAL oligomers, inducing neurodegeneration.
Collapse
|
24
|
Sierecki E, Giles N, Bowden Q, Polinkovsky ME, Steinbeck J, Arrioti N, Rahman D, Bhumkar A, Nicovich PR, Ross I, Parton RG, Böcking T, Gambin Y. Nanomolar oligomerization and selective co-aggregation of α-synuclein pathogenic mutants revealed by single-molecule fluorescence. Sci Rep 2016; 6:37630. [PMID: 27892477 PMCID: PMC5385372 DOI: 10.1038/srep37630] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 10/28/2016] [Indexed: 02/08/2023] Open
Abstract
Protein aggregation is a hallmark of many neurodegenerative diseases, notably Alzheimer's and Parkinson's disease. Parkinson's disease is characterized by the presence of Lewy bodies, abnormal aggregates mainly composed of α-synuclein. Moreover, cases of familial Parkinson's disease have been linked to mutations in α-synuclein. In this study, we compared the behavior of wild-type (WT) α-synuclein and five of its pathological mutants (A30P, E46K, H50Q, G51D and A53T). To this end, single-molecule fluorescence detection was coupled to cell-free protein expression to measure precisely the oligomerization of proteins without purification, denaturation or labelling steps. In these conditions, we could detect the formation of oligomeric and pre-fibrillar species at very short time scale and low micromolar concentrations. The pathogenic mutants surprisingly segregated into two classes: one group forming large aggregates and fibrils while the other tending to form mostly oligomers. Strikingly, co-expression experiments reveal that members from the different groups do not generally interact with each other, both at the fibril and monomer levels. Together, this data paints a completely different picture of α-synuclein aggregation, with two possible pathways leading to the development of fibrils.
Collapse
Affiliation(s)
- Emma Sierecki
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Nichole Giles
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Quill Bowden
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Mark E. Polinkovsky
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Janina Steinbeck
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Nicholas Arrioti
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Diya Rahman
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Akshay Bhumkar
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Philip R. Nicovich
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Ian Ross
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Till Böcking
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Yann Gambin
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| |
Collapse
|
25
|
Lysines, Achilles' heel in alpha-synuclein conversion to a deadly neuronal endotoxin. Ageing Res Rev 2016; 26:62-71. [PMID: 26690800 DOI: 10.1016/j.arr.2015.12.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 12/11/2022]
Abstract
Alpha-synuclein aggregation is associated with Parkinson's disease and other neurodegenerative disorders termed synucleinopathies. The sequence of alpha-synuclein has a remarkable amount of lysines, which may be a target for modifications by several aldehydes found at increased concentration in parkinsonian brains. The involved aldehydes are the dopamine metabolite 3,4-dihydroxyphenylacetaldehyde, the lipid peroxidation products 4-hydroxynonenal, acrolein and malondialdehyde, and advanced glycation end-products. Moreover, both relative expression levels and enzymatic activity of aldehyde dehydrogenases, which are responsible for aldehydes detoxification in cells, are altered in Parkinson's disease brains. The effects of aldehyde modifications can include: (i) a perturbation in the equilibrium of cytosolic and membrane-bound alpha-synuclein, that may alter protein function and lead to aggregation; (ii) the reduction of alpha-synuclein ubiquitination and SUMOylation, affecting its cellular localization and clearance; (iii) a decreased susceptibility to cleavage at specific sites by extracellular proteases; (iv) a reduced availability of identified lysine acetylation sites; (v) the production of toxic oligomeric alpha-synuclein-aldehyde species, able to damage lipid membranes and transmissible from unhealthy to healthy neurons. All of these observations point to a complex interaction between alpha-synuclein and aldehydes in brain, which may lead to the accumulation of dysfunctional alpha-synuclein and its oligomerization.
Collapse
|
26
|
Yu H, Han W, Ma W, Schulten K. Transient β-hairpin formation in α-synuclein monomer revealed by coarse-grained molecular dynamics simulation. J Chem Phys 2015; 143:243142. [PMID: 26723627 PMCID: PMC4684271 DOI: 10.1063/1.4936910] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/18/2015] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease, originating from the intrinsically disordered peptide α-synuclein, is a common neurodegenerative disorder that affects more than 5% of the population above age 85. It remains unclear how α-synuclein monomers undergo conformational changes leading to aggregation and formation of fibrils characteristic for the disease. In the present study, we perform molecular dynamics simulations (over 180 μs in aggregated time) using a hybrid-resolution model, Proteins with Atomic details in Coarse-grained Environment (PACE), to characterize in atomic detail structural ensembles of wild type and mutant monomeric α-synuclein in aqueous solution. The simulations reproduce structural properties of α-synuclein characterized in experiments, such as secondary structure content, long-range contacts, chemical shifts, and (3)J(HNHCα )-coupling constants. Most notably, the simulations reveal that a short fragment encompassing region 38-53, adjacent to the non-amyloid-β component region, exhibits a high probability of forming a β-hairpin; this fragment, when isolated from the remainder of α-synuclein, fluctuates frequently into its β-hairpin conformation. Two disease-prone mutations, namely, A30P and A53T, significantly accelerate the formation of a β-hairpin in the stated fragment. We conclude that the formation of a β-hairpin in region 38-53 is a key event during α-synuclein aggregation. We predict further that the G47V mutation impedes the formation of a turn in the β-hairpin and slows down β-hairpin formation, thereby retarding α-synuclein aggregation.
Collapse
Affiliation(s)
- Hang Yu
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Wei Han
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Wen Ma
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Klaus Schulten
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
27
|
Rutherford NJ, Sacino AN, Brooks M, Ceballos-Diaz C, Ladd TB, Howard JK, Golde TE, Giasson BI. Studies of lipopolysaccharide effects on the induction of α-synuclein pathology by exogenous fibrils in transgenic mice. Mol Neurodegener 2015. [PMID: 26223783 PMCID: PMC4520273 DOI: 10.1186/s13024-015-0029-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative disorder that is pathologically characterized by loss of dopaminergic neurons from the substantia nigra, the presence of aggregated α-synuclein (αS) and evidence of neuroinflammation. Experimental studies have shown that the cerebral injection of recombinant fibrillar αS, especially in αS transgenic mouse models, can induce the formation and spread of αS inclusion pathology. However, studies reporting this phenomenon did not consider the presence of lipopolysaccharide (LPS) in the injected αS, produced in E. coli, as a potential confound. The objectives of this study are to develop a method to remove the LPS contamination and investigate the differences in pathologies induced by αS containing LPS or αS highly purified of LPS. RESULTS AND CONCLUSIONS We were able to remove >99.5% of the LPS contamination from the αS preparations through the addition of a cation exchange step during purification. The αS pathology induced by injection of fibrils produced from αS containing LPS or purified of LPS, showed a similar distribution pattern; however, there was less spread into the cortex of the mice injected with αS containing higher levels of LPS. As previously reported, injection of αS fibrils could induce astrogliosis, and αS inclusions were present within astrocytes in mice injected with fibrils comprised of αS with or without cation exchange purification. Furthermore, we identified the presence of αS pathology in ependymal cells in both groups of mice, which suggests the involvement of a novel mechanism for spread in this model of αS pathology.
Collapse
Affiliation(s)
- Nicola J Rutherford
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, 1275 Center Drive, Room BMS J-483, PO Box 100159, Gainesville, FL, 32610, USA.
| | - Amanda N Sacino
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, 1275 Center Drive, Room BMS J-483, PO Box 100159, Gainesville, FL, 32610, USA.
| | - Mieu Brooks
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, 1275 Center Drive, Room BMS J-483, PO Box 100159, Gainesville, FL, 32610, USA.
| | - Carolina Ceballos-Diaz
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, 1275 Center Drive, Room BMS J-483, PO Box 100159, Gainesville, FL, 32610, USA.
| | - Thomas B Ladd
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, 1275 Center Drive, Room BMS J-483, PO Box 100159, Gainesville, FL, 32610, USA.
| | - Jasie K Howard
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, 1275 Center Drive, Room BMS J-483, PO Box 100159, Gainesville, FL, 32610, USA.
| | - Todd E Golde
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, 1275 Center Drive, Room BMS J-483, PO Box 100159, Gainesville, FL, 32610, USA.
| | - Benoit I Giasson
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, 1275 Center Drive, Room BMS J-483, PO Box 100159, Gainesville, FL, 32610, USA.
| |
Collapse
|
28
|
Roberts HL, Brown DR. Seeking a mechanism for the toxicity of oligomeric α-synuclein. Biomolecules 2015; 5:282-305. [PMID: 25816357 PMCID: PMC4496673 DOI: 10.3390/biom5020282] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 03/08/2015] [Accepted: 03/11/2015] [Indexed: 12/31/2022] Open
Abstract
In a number of neurological diseases including Parkinson’s disease (PD), α‑synuclein is aberrantly folded, forming abnormal oligomers, and amyloid fibrils within nerve cells. Strong evidence exists for the toxicity of increased production and aggregation of α-synuclein in vivo. The toxicity of α-synuclein is popularly attributed to the formation of “toxic oligomers”: a heterogenous and poorly characterized group of conformers that may share common molecular features. This review presents the available evidence on the properties of α-synuclein oligomers and the potential molecular mechanisms of their cellular disruption. Toxic α-synuclein oligomers may impact cells in a number of ways, including the disruption of membranes, mitochondrial depolarization, cytoskeleton changes, impairment of protein clearance pathways, and enhanced oxidative stress. We also examine the relationship between α-synuclein toxic oligomers and amyloid fibrils, in the light of recent studies that paint a more complex picture of α-synuclein toxicity. Finally, methods of studying and manipulating oligomers within cells are described.
Collapse
Affiliation(s)
- Hazel L Roberts
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| | - David R Brown
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| |
Collapse
|
29
|
Gallegos S, Pacheco C, Peters C, Opazo CM, Aguayo LG. Features of alpha-synuclein that could explain the progression and irreversibility of Parkinson's disease. Front Neurosci 2015; 9:59. [PMID: 25805964 PMCID: PMC4353246 DOI: 10.3389/fnins.2015.00059] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/10/2015] [Indexed: 01/21/2023] Open
Abstract
Alpha-synuclein is a presynaptic protein expressed throughout the central nervous system, and it is the main component of Lewy bodies, one of the histopathological features of Parkinson's disease (PD) which is a progressive and irreversible neurodegenerative disorder. The conformational flexibility of α-synuclein allows it to adopt different conformations, i.e., bound to membranes or form aggregates, the oligomers are believed to be the more toxic species. In this review, we will focus on two major features of α-synuclein, transmission and toxicity, that could help to understand the pathological characteristics of PD. One important feature of α-synuclein is its ability to be transmitted from neuron to neuron using mechanisms such as endocytosis, plasma membrane penetration or through exosomes, thus propagating the Lewy body pathology to different brain regions thereby contributing to the progressiveness of PD. The second feature of α-synuclein is that it confers cytotoxicity to recipient cells, principally when it is in an oligomeric state. This form causes mitochondrial dysfunction, endoplasmic reticulum stress, oxidative stress, proteasome impairment, disruption of plasma membrane and pore formation that lead to apoptosis pathway activation and consequent cell death. The complexity of α-synuclein oligomerization and formation of toxic species could be a major factor for the irreversibility of PD and could also explain the lack of successful therapies to halt the disease.
Collapse
Affiliation(s)
- Scarlet Gallegos
- Laboratory of Neurophysiology, Department of Physiology, University of Concepcion Concepcio, Chile
| | - Carla Pacheco
- Laboratory of Neurophysiology, Department of Physiology, University of Concepcion Concepcio, Chile
| | - Christian Peters
- Laboratory of Neurophysiology, Department of Physiology, University of Concepcion Concepcio, Chile
| | - Carlos M Opazo
- Oxidation Biology Laboratory, The Florey Institute of Neuroscience and Mental Health, University of Melbourne Melbourne, VIC, Australia
| | - Luis G Aguayo
- Laboratory of Neurophysiology, Department of Physiology, University of Concepcion Concepcio, Chile
| |
Collapse
|
30
|
Plotegher N, Stringari C, Jahid S, Veronesi M, Girotto S, Gratton E, Bubacco L. NADH fluorescence lifetime is an endogenous reporter of α-synuclein aggregation in live cells. FASEB J 2015; 29:2484-94. [PMID: 25713058 DOI: 10.1096/fj.14-260281] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 02/06/2015] [Indexed: 12/22/2022]
Abstract
α-Synuclein (aS) aggregation has been amply investigated for its involvement in Parkinson's disease because its amyloid fibrils are the main constituent of Lewy bodies, one of the hallmarks of the disease. aS aggregation was studied here in vitro and in cellular models to correlate aggregation products with toxicity mechanisms. Independent results published elsewhere suggested that aS overexpression and/or aggregation may impair cellular metabolism and cause mitochondrial damage. In this context, we report the characterization of changes in NADH fluorescence properties in vitro and in human embryonic kidney 293 cells upon aS aggregation. The application of the phasor approach to study NADH fluorescence lifetime and emission allowed us to identify changes that correlate with aS aggregation. In particular, the fraction of bound NADH, characterized by longer lifetimes in comparison to free NADH, is increased, and the maximum of the NADH emission is shifted toward shorter wavelengths in the presence of aggregating aS both in vitro and in cells. These data suggest that NADH binds to aggregated aS. NMR experiments in vitro substantiate such binding, which occurs during aggregation. NADH fluorescence is thus useful to detect aS aggregation and by extension the associated oxidative stress.
Collapse
Affiliation(s)
- Nicoletta Plotegher
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Chiara Stringari
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Sohail Jahid
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Marina Veronesi
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Stefania Girotto
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Enrico Gratton
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Luigi Bubacco
- *Department of Biology, University of Padua, Padua, Italy; Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA; and Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
31
|
Plotegher N, Kumar D, Tessari I, Brucale M, Munari F, Tosatto L, Belluzzi E, Greggio E, Bisaglia M, Capaldi S, Aioanei D, Mammi S, Monaco HL, Samo B, Bubacco L. The chaperone-like protein 14-3-3η interacts with human α-synuclein aggregation intermediates rerouting the amyloidogenic pathway and reducing α-synuclein cellular toxicity. Hum Mol Genet 2014; 23:5615-29. [PMID: 24895406 DOI: 10.1093/hmg/ddu275] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Familial and idiopathic Parkinson's disease (PD) is associated with the abnormal neuronal accumulation of α-synuclein (aS) leading to β-sheet-rich aggregates called Lewy Bodies (LBs). Moreover, single point mutation in aS gene and gene multiplication lead to autosomal dominant forms of PD. A connection between PD and the 14-3-3 chaperone-like proteins was recently proposed, based on the fact that some of the 14-3-3 isoforms can interact with genetic PD-associated proteins such as parkin, LRRK2 and aS and were found as components of LBs in human PD. In particular, a direct interaction between 14-3-3η and aS was reported when probed by co-immunoprecipitation from cell models, from parkinsonian brains and by surface plasmon resonance in vitro. However, the mechanisms through which 14-3-3η and aS interact in PD brains remain unclear. Herein, we show that while 14-3-3η is unable to bind monomeric aS, it interacts with aS oligomers which occur during the early stages of aS aggregation. This interaction diverts the aggregation process even when 14-3-3η is present in sub-stoichiometric amounts relative to aS. When aS level is overwhelmingly higher than that of 14-3-3η, the fibrillation process becomes a sequestration mechanism for 14-3-3η, undermining all processes governed by this protein. Using a panel of complementary techniques, we single out the stage of aggregation at which the aS/14-3-3η interaction occurs, characterize the products of the resulting processes, and show how the processes elucidated in vitro are relevant in cell models. Our findings constitute a first step in elucidating the molecular mechanism of aS/14-3-3η interaction and in understanding the critical aggregation step at which 14-3-3η has the potential to rescue aS-induced cellular toxicity.
Collapse
Affiliation(s)
| | - Dhruv Kumar
- Department of Biochemistry, University of Bologna, Bologna, Italy
| | | | - Marco Brucale
- Department of Biochemistry, University of Bologna, Bologna, Italy, ISMN-National Council of Research, Rome, Italy and
| | - Francesca Munari
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | | | | | | | | | - Stefano Capaldi
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Daniel Aioanei
- Department of Biochemistry, University of Bologna, Bologna, Italy
| | - Stefano Mammi
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Hugo L Monaco
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Bruno Samo
- Department of Biochemistry, University of Bologna, Bologna, Italy
| | | |
Collapse
|