101
|
Dharmadana D, Reynolds NP, Conn CE, Valéry C. Molecular interactions of amyloid nanofibrils with biological aggregation modifiers: implications for cytotoxicity mechanisms and biomaterial design. Interface Focus 2017; 7:20160160. [PMID: 28630679 PMCID: PMC5474041 DOI: 10.1098/rsfs.2016.0160] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Amyloid nanofibrils are ubiquitous biological protein fibrous aggregates, with a wide range of either toxic or beneficial activities that are relevant to human disease and normal biology. Protein amyloid fibrillization occurs via nucleated polymerization, through non-covalent interactions. As such, protein nanofibril formation is based on a complex interplay between kinetic and thermodynamic factors. The process entails metastable oligomeric species and a highly thermodynamically favoured end state. The kinetics, and the reaction pathway itself, can be influenced by third party moieties, either molecules or surfaces. Specifically, in the biological context, different classes of biomolecules are known to act as catalysts, inhibitors or modifiers of the generic protein fibrillization process. The biological aggregation modifiers reviewed here include lipid membranes of varying composition, glycosaminoglycans and metal ions, with a final word on xenobiotic compounds. The corresponding molecular interactions are critically analysed and placed in the context of the mechanisms of cytotoxicity of the amyloids involved in diverse pathologies and the non-toxicity of functional amyloids (at least towards their biological host). Finally, the utilization of this knowledge towards the design of bio-inspired and biocompatible nanomaterials is explored.
Collapse
Affiliation(s)
- Durga Dharmadana
- School of Health and Biomedical Sciences, Discipline of Pharmaceutical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3001, Australia
| | - Nicholas P. Reynolds
- ARC Training Centre for Biodevices, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia
| | - Charlotte E. Conn
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3001, Australia
| | - Céline Valéry
- School of Health and Biomedical Sciences, Discipline of Pharmaceutical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| |
Collapse
|
102
|
Wei G, Su Z, Reynolds NP, Arosio P, Hamley IW, Gazit E, Mezzenga R. Self-assembling peptide and protein amyloids: from structure to tailored function in nanotechnology. Chem Soc Rev 2017; 46:4661-4708. [PMID: 28530745 PMCID: PMC6364806 DOI: 10.1039/c6cs00542j] [Citation(s) in RCA: 545] [Impact Index Per Article: 77.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled peptide and protein amyloid nanostructures have traditionally been considered only as pathological aggregates implicated in human neurodegenerative diseases. In more recent times, these nanostructures have found interesting applications as advanced materials in biomedicine, tissue engineering, renewable energy, environmental science, nanotechnology and material science, to name only a few fields. In all these applications, the final function depends on: (i) the specific mechanisms of protein aggregation, (ii) the hierarchical structure of the protein and peptide amyloids from the atomistic to mesoscopic length scales and (iii) the physical properties of the amyloids in the context of their surrounding environment (biological or artificial). In this review, we will discuss recent progress made in the field of functional and artificial amyloids and highlight connections between protein/peptide folding, unfolding and aggregation mechanisms, with the resulting amyloid structure and functionality. We also highlight current advances in the design and synthesis of amyloid-based biological and functional materials and identify new potential fields in which amyloid-based structures promise new breakthroughs.
Collapse
Affiliation(s)
- Gang Wei
- Faculty of Production Engineering, University of Bremen, Bremen,
Germany
| | - Zhiqiang Su
- State Key Laboratory of Chemical Resource Engineering, Beijing
University of Chemical Technology, China
| | - Nicholas P. Reynolds
- ARC Training Centre for Biodevices, Swinburne University of
Technology, Melbourne, Australia
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, ETH-Zurich,
Switzerland
| | | | - Ehud Gazit
- Faculty of Life Sciences, Tel Aviv University, Israel
| | - Raffaele Mezzenga
- Department of Health Science and Technology, ETH-Zurich,
Switzerland
| |
Collapse
|
103
|
Membrane-Bound Alpha Synuclein Clusters Induce Impaired Lipid Diffusion and Increased Lipid Packing. Biophys J 2017; 111:2440-2449. [PMID: 27926845 DOI: 10.1016/j.bpj.2016.10.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 10/09/2016] [Accepted: 10/13/2016] [Indexed: 12/21/2022] Open
Abstract
The aggregation of membrane-bound α-synuclein (αS) into oligomers and/or amyloid fibrils has been suggested to cause membrane damage in in vitro model phospholipid membrane systems and in vivo. In this study, we investigate how αS interactions that precede the formation of well-defined aggregates influence physical membrane properties. Using three truncated variants of αS with different aggregation propensities and comparable phospholipid membrane binding affinities we show, using fluorescence recovery after photobleaching (FRAP) and fluorescence anisotropy measurements, that formation of αS clusters on supported lipid bilayers (SLBs) impairs lateral lipid diffusion and increases lipid packing beneath the αS clusters. Formation of protein clusters starts immediately after monomer addition. The magnitudes of the changes in effective lipid diffusion and lipid order increase with the protein cluster size. Our results show that the combination of inter-αS and αS-membrane interactions can drive the formation of more ordered lipid domains. Considering the functional involvement of membrane micro-domains in biological membranes, αS-induced domain formation may be relevant for alternative disease mechanisms.
Collapse
|
104
|
Shrivastava AN, Aperia A, Melki R, Triller A. Physico-Pathologic Mechanisms Involved in Neurodegeneration: Misfolded Protein-Plasma Membrane Interactions. Neuron 2017; 95:33-50. [DOI: 10.1016/j.neuron.2017.05.026] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 05/12/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
|
105
|
Flagmeier P, De S, Wirthensohn DC, Lee SF, Vincke C, Muyldermans S, Knowles TPJ, Gandhi S, Dobson CM, Klenerman D. Ultrasensitive Measurement of Ca 2+
Influx into Lipid Vesicles Induced by Protein Aggregates. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201700966] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Patrick Flagmeier
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Suman De
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - David C. Wirthensohn
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Steven F. Lee
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels Belgium
| | - Tuomas P. J. Knowles
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Sonia Gandhi
- Department of Molecular Neuroscience; Institute of Neurology; University College London; Queen Square London WC1N 3BG UK
| | - Christopher M. Dobson
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - David Klenerman
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| |
Collapse
|
106
|
Flagmeier P, De S, Wirthensohn DC, Lee SF, Vincke C, Muyldermans S, Knowles TPJ, Gandhi S, Dobson CM, Klenerman D. Ultrasensitive Measurement of Ca 2+ Influx into Lipid Vesicles Induced by Protein Aggregates. Angew Chem Int Ed Engl 2017; 56:7750-7754. [PMID: 28474754 PMCID: PMC5615231 DOI: 10.1002/anie.201700966] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/23/2017] [Indexed: 12/15/2022]
Abstract
To quantify and characterize the potentially toxic protein aggregates associated with neurodegenerative diseases, a high‐throughput assay based on measuring the extent of aggregate‐induced Ca2+ entry into individual lipid vesicles has been developed. This approach was implemented by tethering vesicles containing a Ca2+ sensitive fluorescent dye to a passivated surface and measuring changes in the fluorescence as a result of membrane disruption using total internal reflection microscopy. Picomolar concentrations of Aβ42 oligomers could be observed to induce Ca2+ influx, which could be inhibited by the addition of a naturally occurring chaperone and a nanobody designed to bind to the Aβ peptide. We show that the assay can be used to study aggregates from other proteins, such as α‐synuclein, and to probe the effects of complex biofluids, such as cerebrospinal fluid, and thus has wide applicability.
Collapse
Affiliation(s)
- Patrick Flagmeier
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Suman De
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - David C Wirthensohn
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Steven F Lee
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tuomas P J Knowles
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Sonia Gandhi
- Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| |
Collapse
|
107
|
Chaudhary H, Subramaniam V, Claessens MMAE. Direct Visualization of Model Membrane Remodeling by α-Synuclein Fibrillization. Chemphyschem 2017; 18:1620-1626. [PMID: 28370874 PMCID: PMC5485007 DOI: 10.1002/cphc.201700050] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Indexed: 12/03/2022]
Abstract
The interaction of α‐synuclein (αS) with membranes is thought to be critical in the etiology of Parkinson's disease. Besides oligomeric αS aggregates that possibly form membrane pores, the aggregation of αS into amyloid fibrils has been reported to disrupt membranes. The mechanism by which aggregation affects the integrity of membranes is, however, unknown. Here, we show that whereas mature αS fibrils only weakly adhere to POPC/POPG giant unilamellar vesicles (GUVs), fibrillization of αS on the membrane results in large‐scale membrane remodeling. Fibrils that grow on the vesicle surface stiffen the membrane and make the initially spherical membrane become polyhedral. Additionally, membrane‐attached fibrils extract lipids. The lipid extraction and membrane remodeling of growing fibrils can consume the complete bilayer surface and results in loss of vesicle content. These observations suggest that there are several mechanisms by which growing fibrils can disrupt membrane function.
Collapse
Affiliation(s)
- Himanshu Chaudhary
- Nanobiophysics, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7500AE, Enschede, The Netherlands
| | - Vinod Subramaniam
- Vrije Universiteit Amsterdam, De Boelelaan 1104, 1081HV, Amsterdam, The Netherlands
| | - Mireille M A E Claessens
- Nanobiophysics, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7500AE, Enschede, The Netherlands
| |
Collapse
|
108
|
Unraveling amyloid formation paths of Parkinson's disease protein α-synuclein triggered by anionic vesicles. Q Rev Biophys 2017; 50:e3. [DOI: 10.1017/s0033583517000026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractAmyloid formation of the synaptic brain proteinα-synuclein (αS) is related to degeneration of dopaminergic neurons in Parkinson's disease patients.αS is thought to function in vesicle transport and fusion and it binds strongly to negatively charged vesiclesin vitro. Here we combined circular dichroism, fluorescence and imaging methodsin vitroto characterize the interaction ofαS with negatively charged vesicles of DOPS (1,2-dioleoyl-sn-glycero-3-phospho-L-serine, sodium salt) and DOPG (1,2-dioleoyl-sn-glycero-3-phospho-(1′-rac-glycerol), sodium salt) and the consequences of such interactions onαS amyloid formation. We found that lipid head-group chemistry modulatesαS interactions and also affects amyloid fiber formation. During the course of the experiments, we made the unexpected discovery that pre-formedαS oligomers, typically present in a small amount in theαS starting material, acted as templates for linear growth of anomalous amyloid fibers in the presence of vesicles. At the same time, the remainingαS monomers were restricted from vesicle-mediated nucleation of amyloid fibers. Although not a dominant process in bulk experiments, this hiddenαS aggregation pathway may be of importancein vivo.
Collapse
|
109
|
Abstract
Amyloids are highly ordered protein aggregates that are associated with both disease (including PrP prion, Alzheimer's, and Parkinson's) and biological function. The amyloid structure is composed of the cross-β-sheet entity, which is an almost indefinitely repeating two-layered intermolecular β-sheet motif. The three-dimensional (3D) structure is unique among protein folds because it folds only upon intermolecular contacts (for a folding to occur, only short sequences of amino acid residues are required), and the structure repeats itself at the atomic level (i.e., every 4.7 Å). As a consequence of this structure, among others, it can grow by recruiting corresponding amyloid peptide/protein and thus has the capacity to be an infectious protein (i.e., a prion). Furthermore, its repetitiveness can translate what would be a nonspecific activity as monomer into a potent one through cooperativity. Because of these and other properties, the activities of amyloids are manifold and include peptide storage, template assistance, loss of function, gain of function, generation of toxicity, membrane binding, infectivity, and more. This review summarizes the structural nature of the cross-β-sheet motif on the basis of a few high-resolution structural studies of amyloids in the context of potential biological activities.
Collapse
Affiliation(s)
- Roland Riek
- Laboratory of Physical Chemistry, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
110
|
Eichmann C, Kumari P, Riek R. High-density lipoprotein-like particle formation of Synuclein variants. FEBS Lett 2017; 591:304-311. [PMID: 28027392 DOI: 10.1002/1873-3468.12543] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 11/11/2022]
Abstract
α-Synuclein (α-Syn) is an intrinsically disordered protein in solution whose fibrillar aggregates are the hallmark of Parkinson's disease (PD). Although the specific function of α-Syn is still unclear, its high structural plasticity is key for the interactions of α-Syn with biological membranes. Recently, it has been observed that α-Syn is able to form high-density lipoprotein-like (HDL-like) particles that are reminiscent of self-assembling phospholipid bilayer nanodiscs. Here, we extended our preparation method for the production of α-Syn lipoprotein particles to the β- and γ-Syn variants, and the PD-related familial α-Syn mutants. We show that all human Syns can form stable and homogeneous populations of HDL-like particles with distinct morphologies. Our results characterize the impact of the individual Syns on the formation capacity of these particles and indicate that Syn HDL-like particles are neither causing toxicity nor a toxicity-related loss of α-Syn in PD.
Collapse
Affiliation(s)
- Cédric Eichmann
- In-Cell NMR Laboratory, Department of NMR-supported Structural Biology, Leibniz Institute of Molecular Pharmacology (FMP Berlin), Berlin, Germany
| | - Pratibha Kumari
- Laboratory of Physical Chemistry, Swiss Federal Institute of Technology, Zürich, Switzerland
| | - Roland Riek
- Laboratory of Physical Chemistry, Swiss Federal Institute of Technology, Zürich, Switzerland
| |
Collapse
|
111
|
Endosulfine-alpha inhibits membrane-induced α-synuclein aggregation and protects against α-synuclein neurotoxicity. Acta Neuropathol Commun 2017; 5:3. [PMID: 28069058 PMCID: PMC5223451 DOI: 10.1186/s40478-016-0403-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 12/10/2016] [Indexed: 12/21/2022] Open
Abstract
Neuropathological and genetic findings suggest that the presynaptic protein α-synuclein (aSyn) is involved in the pathogenesis of synucleinopathy disorders, including Parkinson’s disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy. Evidence suggests that the self-assembly of aSyn conformers bound to phospholipid membranes in an aggregation-prone state plays a key role in aSyn neurotoxicity. Accordingly, we hypothesized that protein binding partners of lipid-associated aSyn could inhibit the formation of toxic aSyn oligomers at membrane surfaces. To address this hypothesis, we characterized the protein endosulfine-alpha (ENSA), previously shown to interact selectively with membrane-bound aSyn, in terms of its effects on the membrane-induced aggregation and neurotoxicity of two familial aSyn mutants, A30P and G51D. We found that wild-type ENSA, but not the non-aSyn-binding S109E variant, interfered with membrane-induced aSyn self-assembly, aSyn-mediated vesicle disruption and aSyn neurotoxicity. Immunoblotting analyses revealed that ENSA was down-regulated in the brains of synucleinopathy patients versus non-diseased individuals. Collectively, these results suggest that ENSA can alleviate neurotoxic effects of membrane-bound aSyn via an apparent chaperone-like activity at the membrane surface, and a decrease in ENSA expression may contribute to aSyn neuropathology in synucleinopathy disorders. More generally, our findings suggest that promoting interactions between lipid-bound, amyloidogenic proteins and their binding partners is a viable strategy to alleviate cytotoxicity in a range of protein misfolding disorders.
Collapse
|
112
|
van Diggelen F, Tepper AWJW, Apetri MM, Otzen DE. α-Synuclein Oligomers: A Study in Diversity. Isr J Chem 2016. [DOI: 10.1002/ijch.201600116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Femke van Diggelen
- Crossbeta Biosciences; Padualaan 8 3584CH Utrecht The Netherlands
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| | | | | | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| |
Collapse
|
113
|
Baumann A, Jorge-Finnigan A, Jung-Kc K, Sauter A, Horvath I, Morozova-Roche LA, Martinez A. Tyrosine Hydroxylase Binding to Phospholipid Membranes Prompts Its Amyloid Aggregation and Compromises Bilayer Integrity. Sci Rep 2016; 6:39488. [PMID: 28004763 PMCID: PMC5177901 DOI: 10.1038/srep39488] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 11/21/2016] [Indexed: 12/14/2022] Open
Abstract
Tyrosine hydroxylase (TH), a rate-limiting enzyme in the synthesis of catecholamine neurotransmitters and hormones, binds to negatively charged phospholipid membranes. Binding to both large and giant unilamellar vesicles causes membrane permeabilization, as observed by efflux and influx of fluorescence dyes. Whereas the initial protein-membrane interaction involves the N-terminal tail that constitutes an extension of the regulatory ACT-domain, prolonged membrane binding induces misfolding and self-oligomerization of TH over time as shown by circular dichroism and Thioflavin T fluorescence. The gradual amyloid-like aggregation likely occurs through cross-β interactions involving aggregation-prone motives in the catalytic domains, consistent with the formation of chain and ring-like protofilaments observed by atomic force microscopy in monolayer-bound TH. PC12 cells treated with the neurotoxin 6-hydroxydopamine displayed increased TH levels in the mitochondrial fraction, while incubation of isolated mitochondria with TH led to a decrease in the mitochondrial membrane potential. Furthermore, cell-substrate impedance and viability assays showed that supplementing the culture media with TH compromises cell viability over time. Our results revealed that the disruptive effect of TH on cell membranes may be a cytotoxic and pathogenic factor if the regulation and intracellular stability of TH is compromised.
Collapse
Affiliation(s)
- Anne Baumann
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway.,Division of Psychiatry, Haukeland University Hospital, 5021 Bergen, Norway
| | - Ana Jorge-Finnigan
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway.,K.G. Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, 5009 Bergen, Norway
| | - Kunwar Jung-Kc
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Alexander Sauter
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway.,Department of Clinical Dentistry, University of Bergen, 5009 Bergen, Norway
| | - Istvan Horvath
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, Sweden
| | | | - Aurora Martinez
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway.,K.G. Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, 5009 Bergen, Norway
| |
Collapse
|
114
|
Riek R, Eisenberg DS. The activities of amyloids from a structural perspective. Nature 2016; 539:227-235. [PMID: 27830791 DOI: 10.1038/nature20416] [Citation(s) in RCA: 339] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/14/2016] [Indexed: 12/16/2022]
Abstract
The aggregation of proteins into structures known as amyloids is observed in many neurodegenerative diseases, including Alzheimer's disease. Amyloids are composed of pairs of tightly interacting, many stranded and repetitive intermolecular β-sheets, which form the cross-β-sheet structure. This structure enables amyloids to grow by recruitment of the same protein and its repetition can transform a weak biological activity into a potent one through cooperativity and avidity. Amyloids therefore have the potential to self-replicate and can adapt to the environment, yielding cell-to-cell transmissibility, prion infectivity and toxicity.
Collapse
Affiliation(s)
- Roland Riek
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zürich, Switzerland
| | - David S Eisenberg
- UCLA-DOE Institute, Los Angeles, California 90095-1570, USA.,Howard Hughes Medical Institute, Los Angeles, California 90095-1570, USA
| |
Collapse
|
115
|
Hecel A, De Ricco R, Valensin D. Influence of membrane environments and copper ions on the structural features of amyloidogenic proteins correlated to neurodegeneration. Coord Chem Rev 2016. [DOI: 10.1016/j.ccr.2016.06.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
116
|
Eichmann C, Campioni S, Kowal J, Maslennikov I, Gerez J, Liu X, Verasdonck J, Nespovitaya N, Choe S, Meier BH, Picotti P, Rizo J, Stahlberg H, Riek R. Preparation and Characterization of Stable α-Synuclein Lipoprotein Particles. J Biol Chem 2016; 291:8516-27. [PMID: 26846854 DOI: 10.1074/jbc.m115.707968] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Indexed: 01/11/2023] Open
Abstract
Multiple neurodegenerative diseases are caused by the aggregation of the human α-Synuclein (α-Syn) protein. α-Syn possesses high structural plasticity and the capability of interacting with membranes. Both features are not only essential for its physiological function but also play a role in the aggregation process. Recently it has been proposed that α-Syn is able to form lipid-protein particles reminiscent of high-density lipoproteins. Here, we present a method to obtain a stable and homogeneous population of nanometer-sized particles composed of α-Syn and anionic phospholipids. These particles are called α-Syn lipoprotein (nano)particles to indicate their relationship to high-density lipoproteins formed by human apolipoproteins in vivo and of in vitro self-assembling phospholipid bilayer nanodiscs. Structural investigations of the α-Syn lipoprotein particles by circular dichroism (CD) and magic angle solid-state nuclear magnetic resonance (MAS SS-NMR) spectroscopy establish that α-Syn adopts a helical secondary structure within these particles. Based on cryo-electron microscopy (cryo-EM) and dynamic light scattering (DLS) α-Syn lipoprotein particles have a defined size with a diameter of ∼23 nm. Chemical cross-linking in combination with solution-state NMR and multiangle static light scattering (MALS) of α-Syn particles reveal a high-order protein-lipid entity composed of ∼8-10 α-Syn molecules. The close resemblance in size between cross-linked in vitro-derived α-Syn lipoprotein particles and a cross-linked species of endogenous α-Syn from SH-SY5Y human neuroblastoma cells indicates a potential functional relevance of α-Syn lipoprotein nanoparticles.
Collapse
Affiliation(s)
| | | | - Julia Kowal
- Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | | | - Juan Gerez
- Institute of Biochemistry, Swiss Federal Institute of Technology, ETH-Hönggerberg, CH-8093 Zürich, Switzerland
| | - Xiaoxia Liu
- Department of Biophysics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | | | | | - Senyon Choe
- Structural Biology Laboratory, The Salk Institute, La Jolla, California 92037 and
| | | | - Paola Picotti
- Institute of Biochemistry, Swiss Federal Institute of Technology, ETH-Hönggerberg, CH-8093 Zürich, Switzerland
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | - Henning Stahlberg
- Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Roland Riek
- From the Laboratory of Physical Chemistry and Structural Biology Laboratory, The Salk Institute, La Jolla, California 92037 and
| |
Collapse
|
117
|
Ghio S, Kamp F, Cauchi R, Giese A, Vassallo N. Interaction of α-synuclein with biomembranes in Parkinson's disease--role of cardiolipin. Prog Lipid Res 2015; 61:73-82. [PMID: 26703192 DOI: 10.1016/j.plipres.2015.10.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/14/2015] [Accepted: 10/30/2015] [Indexed: 12/15/2022]
Abstract
One of the key molecular events underlying the pathogenesis of Parkinson's disease (PD) is the aberrant misfolding and aggregation of the α-synuclein (αS) protein into higher-order oligomers that play a key role in neuronal dysfunction and degeneration. A wealth of experimental data supports the hypothesis that the neurotoxicity of αS oligomers is intrinsically linked with their ability to interact with, and disrupt, biological membranes; especially those membranes having negatively-charged surfaces and/or lipid packing defects. Consequences of αS-lipid interaction include increased membrane tension, permeation by pore formation, membrane lysis and/or leakage due to the extraction of lipids from the bilayer. Moreover, we assert that the interaction of αS with a liquid-disordering phospholipid uniquely enriched in mitochondrial membranes, namely cardiolipin (1,3-diphosphatidyl-sn-glycerol, CL), helps target the αS oligomeric complexes intracellularly to mitochondria. Binding mediated by CL may thus represent an important pathomechanism by which cytosolic αS could physically associate with mitochondrial membranes and disrupt their integrity. Impaired mitochondrial function culminates in a cellular bioenergetic crisis and apoptotic death. To conclude, we advocate the accelerated discovery of new drugs targeting this pathway in order to restore mitochondrial function in PD.
Collapse
Affiliation(s)
- Stephanie Ghio
- Dept. of Physiology and Biochemistry, University of Malta, Msida, Malta
| | - Frits Kamp
- Biomedical Center (BMC), Biochemistry, Ludwig-Maximilians-University & DZNE, 81377 Munich, Germany
| | - Ruben Cauchi
- Dept. of Physiology and Biochemistry, University of Malta, Msida, Malta
| | - Armin Giese
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Neville Vassallo
- Dept. of Physiology and Biochemistry, University of Malta, Msida, Malta.
| |
Collapse
|
118
|
Vidic J, Richard CA, Péchoux C, Da Costa B, Bertho N, Mazerat S, Delmas B, Chevalier C. Amyloid Assemblies of Influenza A Virus PB1-F2 Protein Damage Membrane and Induce Cytotoxicity. J Biol Chem 2015; 291:739-51. [PMID: 26601953 DOI: 10.1074/jbc.m115.652917] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Indexed: 01/10/2023] Open
Abstract
PB1-F2 is a small accessory protein encoded by an alternative open reading frame in PB1 segments of most influenza A virus. PB1-F2 is involved in virulence by inducing mitochondria-mediated immune cells apoptosis, increasing inflammation, and enhancing predisposition to secondary bacterial infections. Using biophysical approaches we characterized membrane disruptive activity of the full-length PB1-F2 (90 amino acids), its N-terminal domain (52 amino acids), expressed by currently circulating H1N1 viruses, and its C-terminal domain (38 amino acids). Both full-length and N-terminal domain of PB1-F2 are soluble at pH values ≤6, whereas the C-terminal fragment was found soluble only at pH ≤ 3. All three peptides are intrinsically disordered. At pH ≥ 7, the C-terminal part of PB1-F2 spontaneously switches to amyloid oligomers, whereas full-length and the N-terminal domain of PB1-F2 aggregate to amorphous structures. When incubated with anionic liposomes at pH 5, full-length and the C-terminal part of PB1-F2 assemble into amyloid structures and disrupt membrane at nanomolar concentrations. PB1-F2 and its C-terminal exhibit no significant antimicrobial activity. When added in the culture medium of mammalian cells, PB1-F2 amorphous aggregates show no cytotoxicity, whereas PB1-F2 pre-assembled into amyloid oligomers or fragmented nanoscaled fibrils was highly cytotoxic. Furthermore, the formation of PB1-F2 amyloid oligomers in infected cells was directly reflected by membrane disruption and cell death as observed in U937 and A549 cells. Altogether our results demonstrate that membrane-lytic activity of PB1-F2 is closely linked to supramolecular organization of the protein.
Collapse
Affiliation(s)
- Jasmina Vidic
- From the Unité de Virologie et Immunologie Moléculaires, INRA, UR892, Domaine de Vilvert, 78350 Jouy en Josas,
| | - Charles-Adrien Richard
- From the Unité de Virologie et Immunologie Moléculaires, INRA, UR892, Domaine de Vilvert, 78350 Jouy en Josas
| | - Christine Péchoux
- the Génétique Animale et Biologie Intégrative, INRA, UMR1313, Domaine de Vilvert, 78350 Jouy en Josas, and
| | - Bruno Da Costa
- From the Unité de Virologie et Immunologie Moléculaires, INRA, UR892, Domaine de Vilvert, 78350 Jouy en Josas
| | - Nicolas Bertho
- From the Unité de Virologie et Immunologie Moléculaires, INRA, UR892, Domaine de Vilvert, 78350 Jouy en Josas
| | - Sandra Mazerat
- the Institut de Chimie Moléculaire et des Matériaux d'Orsay, Université Paris-Sud, CNRS, UMR 8182, 91400 Orsay, France
| | - Bernard Delmas
- From the Unité de Virologie et Immunologie Moléculaires, INRA, UR892, Domaine de Vilvert, 78350 Jouy en Josas
| | - Christophe Chevalier
- From the Unité de Virologie et Immunologie Moléculaires, INRA, UR892, Domaine de Vilvert, 78350 Jouy en Josas
| |
Collapse
|
119
|
Chiang YL, Chang YC, Chiang IC, Mak HM, Hwang IS, Shih YL. Atomic Force Microscopy Characterization of Protein Fibrils Formed by the Amyloidogenic Region of the Bacterial Protein MinE on Mica and a Supported Lipid Bilayer. PLoS One 2015; 10:e0142506. [PMID: 26562523 PMCID: PMC4642933 DOI: 10.1371/journal.pone.0142506] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/22/2015] [Indexed: 11/18/2022] Open
Abstract
Amyloid fibrils play a crucial role in many human diseases and are found to function in a range of physiological processes from bacteria to human. They have also been gaining importance in nanotechnology applications. Understanding the mechanisms behind amyloid formation can help develop strategies towards the prevention of fibrillation processes or create new technological applications. It is thus essential to observe the structures of amyloids and their self-assembly processes at the nanometer-scale resolution under physiological conditions. In this work, we used highly force-sensitive frequency-modulation atomic force microscopy (FM-AFM) to characterize the fibril structures formed by the N-terminal domain of a bacterial division protein MinE in solution. The approach enables us to investigate the fibril morphology and protofibril organization over time progression and in response to changes in ionic strength, molecular crowding, and upon association with different substrate surfaces. In addition to comparison of the fibril structure and behavior of MinE1-31 under varying conditions, the study also broadens our understanding of the versatile behavior of amyloid-substrate surface interactions.
Collapse
Affiliation(s)
- Ya-Ling Chiang
- Institute of Physics, Academia Sinica, Taipei, Taiwan
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yuan-Chih Chang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - I-Chen Chiang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Huey-Ming Mak
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Ing-Shouh Hwang
- Institute of Physics, Academia Sinica, Taipei, Taiwan
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
- * E-mail: (ISH), (YLS)
| | - Yu-Ling Shih
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
- * E-mail: (ISH), (YLS)
| |
Collapse
|
120
|
Brudek T, Winge K, Rasmussen NB, Bahl JMC, Tanassi J, Agander TK, Hyde TM, Pakkenberg B. Altered α-synuclein, parkin, and synphilin isoform levels in multiple system atrophy brains. J Neurochem 2015; 136:172-85. [DOI: 10.1111/jnc.13392] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 09/29/2015] [Indexed: 01/16/2023]
Affiliation(s)
- Tomasz Brudek
- Research Laboratory for Stereology and Neuroscience; Bispebjerg-Frederiksberg Hospital; University Hospital of Copenhagen; Copenhagen NV Denmark
- Bispebjerg Movement Disorders Biobank; Bispebjerg-Frederiksberg Hospital; University Hospital of Copenhagen; Copenhagen N Denmark
| | - Kristian Winge
- Department of Neurology; Bispebjerg-Frederiksberg Hospital; University Hospital of Copenhagen; Copenhagen NV Denmark
- Bispebjerg Movement Disorders Biobank; Bispebjerg-Frederiksberg Hospital; University Hospital of Copenhagen; Copenhagen N Denmark
| | - Nadja Bredo Rasmussen
- Research Laboratory for Stereology and Neuroscience; Bispebjerg-Frederiksberg Hospital; University Hospital of Copenhagen; Copenhagen NV Denmark
| | | | - Julia Tanassi
- Department of Autoimmunology and Biomarkers; Statens Serum Institut; Copenhagen S Denmark
| | | | - Thomas M. Hyde
- Lieber Institute for Brain Development; Johns Hopkins Medical Campus; Baltimore Maryland USA
- Department of Psychiatry and Behavioral Sciences; Johns Hopkins University School of Medicine; Baltimore Maryland USA
- Department of Neurology; Johns Hopkins University School of Medicine; Baltimore Maryland USA
| | - Bente Pakkenberg
- Research Laboratory for Stereology and Neuroscience; Bispebjerg-Frederiksberg Hospital; University Hospital of Copenhagen; Copenhagen NV Denmark
| |
Collapse
|
121
|
Is membrane homeostasis the missing link between inflammation and neurodegenerative diseases? Cell Mol Life Sci 2015; 72:4795-805. [PMID: 26403788 PMCID: PMC5005413 DOI: 10.1007/s00018-015-2038-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/16/2015] [Accepted: 09/03/2015] [Indexed: 12/14/2022]
Abstract
Systemic inflammation and infections are associated with neurodegenerative diseases. Unfortunately, the molecular bases of this link are still largely undiscovered. We, therefore, review how inflammatory processes can imbalance membrane homeostasis and theorize how this may have an effect on the aggregation behavior of the proteins implicated in such diseases. Specifically, we describe the processes that generate such imbalances at the molecular level, and try to understand how they affect protein folding and localization. Overall, current knowledge suggests that microglia pro-inflammatory mediators can generate membrane damage, which may have an impact in terms of triggering or accelerating disease manifestation.
Collapse
|
122
|
Anunciado D, Rai DK, Qian S, Urban V, O'Neill H. Small-angle neutron scattering reveals the assembly of alpha-synuclein in lipid membranes. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1881-1889. [PMID: 26321599 DOI: 10.1016/j.bbapap.2015.08.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 08/12/2015] [Accepted: 08/25/2015] [Indexed: 12/15/2022]
Abstract
The aggregation of α-synuclein (asyn), an intrinsically disordered protein (IDP), is a hallmark in Parkinson's disease (PD). We investigated the conformational changes that asyn undergoes in the presence of membrane and membrane mimetics using small-angle neutron scattering (SANS). In solution, asyn is monomeric and unfolded assuming an ensemble of conformers spanning extended and compact conformations. Using the contrast variation technique and SANS, the protein scattering signal in the membrane-protein complexes is selectively highlighted in order to monitor its conformational changes in this environment. We showed that in the presence of phospholipid membranes asyn transitions from a monodisperse state to aggregated structures with sizes ranging from 200 to 900Å coexisting with the monomeric species. Detailed SANS data analysis revealed that asyn aggregates have a hierarchical organization in which clusters of smaller asyn aggregates assemble to form the larger structures. This study provides new insight into the mechanism of asyn aggregation. We propose an aggregation mechanism in which stable asyn aggregates seed the aggregation process and hence the hierarchical assembly of structures. Our findings demonstrate that membrane-induced conformational changes in asyn lead to its heterogeneous aggregation which could be physiologically relevant in its function or in the diseased state.
Collapse
Affiliation(s)
- Divina Anunciado
- Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, United States
| | - Durgesh K Rai
- Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, United States
| | - Shuo Qian
- Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, United States
| | - Volker Urban
- Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, United States
| | - Hugh O'Neill
- Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, United States
| |
Collapse
|
123
|
Smith RAS, Nabok A, Blakeman BJF, Xue WF, Abell B, Smith DP. Analysis of Toxic Amyloid Fibril Interactions at Natively Derived Membranes by Ellipsometry. PLoS One 2015; 10:e0132309. [PMID: 26172440 PMCID: PMC4501548 DOI: 10.1371/journal.pone.0132309] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/11/2015] [Indexed: 12/26/2022] Open
Abstract
There is an ongoing debate regarding the culprits of cytotoxicity associated with amyloid disorders. Although small pre-fibrillar amyloid oligomers have been implicated as the primary toxic species, the fibrillar amyloid material itself can also induce cytotoxicity. To investigate membrane disruption and cytotoxic effects associated with intact and fragmented fibrils, the novel in situ spectroscopic technique of Total Internal Reflection Ellipsometry (TIRE) was used. Fibril lipid interactions were monitored using natively derived whole cell membranes as a model of the in vivo environment. We show that fragmented fibrils have an increased ability to disrupt these natively derived membranes by causing a loss of material from the deposited surface when compared with unfragmented fibrils. This effect was corroborated by observations of membrane disruption in live cells, and by dye release assay using synthetic liposomes. Through these studies we demonstrate the use of TIRE for the analysis of protein-lipid interactions on natively derived lipid surfaces, and provide an explanation on how amyloid fibrils can cause a toxic gain of function, while entangled amyloid plaques exert minimal biological activity.
Collapse
Affiliation(s)
- Rachel A. S. Smith
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - Aleksey Nabok
- Materials and Engineering Research Institute, Sheffield Hallam University, Sheffield, United Kingdom
| | - Ben J. F. Blakeman
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Wei-Feng Xue
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Benjamin Abell
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - David P. Smith
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
124
|
Fibril growth and seeding capacity play key roles in α-synuclein-mediated apoptotic cell death. Cell Death Differ 2015; 22:2107-22. [PMID: 26138444 DOI: 10.1038/cdd.2015.79] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 04/30/2015] [Accepted: 05/06/2015] [Indexed: 02/08/2023] Open
Abstract
The role of extracellular α-synuclein (α-syn) in the initiation and the spreading of neurodegeneration in Parkinson's disease (PD) has been studied extensively over the past 10 years. However, the nature of the α-syn toxic species and the molecular mechanisms by which they may contribute to neuronal cell loss remain controversial. In this study, we show that fully characterized recombinant monomeric, fibrillar or stabilized forms of oligomeric α-syn do not trigger significant cell death when added individually to neuroblastoma cell lines. However, a mixture of preformed fibrils (PFFs) with monomeric α-syn becomes toxic under conditions that promote their growth and amyloid formation. In hippocampal primary neurons and ex vivo hippocampal slice cultures, α-syn PFFs are capable of inducing a moderate toxicity over time that is greatly exacerbated upon promoting fibril growth by addition of monomeric α-syn. The causal relationship between α-syn aggregation and cellular toxicity was further investigated by assessing the effect of inhibiting fibrillization on α-syn-induced cell death. Remarkably, our data show that blocking fibril growth by treatment with known pharmacological inhibitor of α-syn fibrillization (Tolcapone) or replacing monomeric α-syn by monomeric β-synuclein in α-syn mixture composition prevent α-syn-induced toxicity in both neuroblastoma cell lines and hippocampal primary neurons. We demonstrate that exogenously added α-syn fibrils bind to the plasma membrane and serve as nucleation sites for the formation of α-syn fibrils and promote the accumulation and internalization of these aggregates that in turn activate both the extrinsic and intrinsic apoptotic cell death pathways in our cellular models. Our results support the hypothesis that ongoing aggregation and fibrillization of extracellular α-syn play central roles in α-syn extracellular toxicity, and suggest that inhibiting fibril growth and seeding capacity constitute a viable strategy for protecting against α-syn-induced toxicity and slowing the progression of neurodegeneration in PD and other synucleinopathies.
Collapse
|
125
|
Verma M, Vats A, Taneja V. Toxic species in amyloid disorders: Oligomers or mature fibrils. Ann Indian Acad Neurol 2015; 18:138-45. [PMID: 26019408 PMCID: PMC4445186 DOI: 10.4103/0972-2327.144284] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 09/05/2014] [Accepted: 09/21/2014] [Indexed: 11/04/2022] Open
Abstract
Protein aggregation is the hallmark of several neurodegenerative disorders. These protein aggregation (fibrillization) disorders are also known as amyloid disorders. The mechanism of protein aggregation involves conformation switch of the native protein, oligomer formation leading to protofibrils and finally mature fibrils. Mature fibrils have long been considered as the cause of disease pathogenesis; however, recent evidences suggest oligomeric intermediates formed during fibrillization to be toxic. In this review, we have tried to address the ongoing debate for these toxic amyloid species. We did an extensive literature search and collated information from Pubmed (http://www.ncbi.nlm.nih.gov) and Google search using various permutations and combinations of the following keywords: Neurodegeneration, amyloid disorders, protein aggregation, fibrils, oligomers, toxicity, Alzheimer's Disease, Parkinson's Disease. We describe different instances showing the toxicity of mature fibrils as well as oligomers in Alzheimer's Disease and Parkinson's Disease. Distinct structural framework and morphology of amyloid oligomers suggests difference in toxic effect between oligomers and fibrils. We highlight the difference in structure and proposed toxicity pathways for fibrils and oligomers. We also highlight the evidences indicating that intermediary oligomeric species can act as potential diagnostic biomarker. Since the formation of these toxic species follow a common structural switch among various amyloid disorders, the protein aggregation events can be targeted for developing broad-range therapeutics. The therapeutic trials based on the understanding of different protein conformers (monomers, oligomers, protofibrils and fibrils) in amyloid cascade are also described.
Collapse
Affiliation(s)
- Meenakshi Verma
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research-Institute of Genomics and Integrated Biology, Sir Ganga Ram Hospital, New Delhi, India
| | - Abhishek Vats
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India ; Department of Biotechnology, Jamia Hamdard University, New Delhi, India
| | - Vibha Taneja
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
126
|
Reynolds NP, Charnley M, Bongiovanni MN, Hartley PG, Gras SL. Biomimetic Topography and Chemistry Control Cell Attachment to Amyloid Fibrils. Biomacromolecules 2015; 16:1556-65. [DOI: 10.1021/acs.biomac.5b00114] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Nicholas P. Reynolds
- Manufacturing
Flagship, CSIRO, Bayview Avenue, Clayton, Victoria 3169, Australia
| | | | - Marie N. Bongiovanni
- Department
of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Patrick G. Hartley
- Energy
Flagship, CSIRO, Private Bag 10, Bayview Avenue, Clayton, Victoria 3169, Australia
| | | |
Collapse
|
127
|
Ysselstein D, Joshi M, Mishra V, Griggs AM, Asiago JM, McCabe GP, Stanciu LA, Post CB, Rochet JC. Effects of impaired membrane interactions on α-synuclein aggregation and neurotoxicity. Neurobiol Dis 2015; 79:150-63. [PMID: 25931201 DOI: 10.1016/j.nbd.2015.04.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/20/2015] [Accepted: 04/21/2015] [Indexed: 01/08/2023] Open
Abstract
The post-mortem brains of individuals with Parkinson's disease (PD) and other synucleinopathy disorders are characterized by the presence of aggregated forms of the presynaptic protein α-synuclein (aSyn). Understanding the molecular mechanism of aSyn aggregation is essential for the development of neuroprotective strategies to treat these diseases. In this study, we examined how interactions between aSyn and phospholipid vesicles influence the protein's aggregation and toxicity to dopaminergic neurons. Two-dimensional NMR data revealed that two familial aSyn mutants, A30P and G51D, populated an exposed, membrane-bound conformer in which the central hydrophobic region was dissociated from the bilayer to a greater extent than in the case of wild-type aSyn. A30P and G51D had a greater propensity to undergo membrane-induced aggregation and elicited greater toxicity to primary dopaminergic neurons compared to the wild-type protein. In contrast, the non-familial aSyn mutant A29E exhibited a weak propensity to aggregate in the presence of phospholipid vesicles or to elicit neurotoxicity, despite adopting a relatively exposed membrane-bound conformation. Our findings suggest that the aggregation of exposed, membrane-bound aSyn conformers plays a key role in the protein's neurotoxicity in PD and other synucleinopathy disorders.
Collapse
Affiliation(s)
- Daniel Ysselstein
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Mehul Joshi
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Vartika Mishra
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Amy M Griggs
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Josephat M Asiago
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - George P McCabe
- Department of Statistics, Purdue University, West Lafayette, IN, USA
| | - Lia A Stanciu
- Schools of Materials Engineering and Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Carol Beth Post
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
128
|
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: 142] [Impact Index Per Article: 15.8] [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
|
129
|
Hähl H, Möller I, Kiesel I, Campioni S, Riek R, Verdes D, Seeger S. α-Synuclein insertion into supported lipid bilayers as seen by in situ X-ray reflectivity. ACS Chem Neurosci 2015; 6:374-9. [PMID: 25523270 DOI: 10.1021/cn5002683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Large aggregates of misfolded α-synuclein inside neuronal cells are the hallmarks of Parkinson's disease. The protein's natural function and its supposed toxicity, however, are believed to be closely related to its interaction with cell and vesicle membranes. Upon this interaction, the protein folds into an α-helical structure and intercalates into the membrane. In this study, we focus on the changes in the lipid bilayer caused by this intrusion. In situ X-ray reflectivity was applied to determine the vertical density structure of the bilayer before and after exposure to α-synuclein. It was found that the α-synuclein insertion, wild type and E57K variant, caused a reduction in bilayer thickness. This effect may be one factor in the membrane pore formation ability of α-synuclein.
Collapse
Affiliation(s)
- Hendrik Hähl
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Isabelle Möller
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Irena Kiesel
- Fakultät Physik/DELTA, Technische Universität Dortmund, 44221 Dortmund, Germany
| | - Silvia Campioni
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Str. 10, 8093 Zurich, Switzerland
| | - Roland Riek
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Str. 10, 8093 Zurich, Switzerland
| | - Dorinel Verdes
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Stefan Seeger
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
130
|
Tsigelny IF, Sharikov Y, Kouznetsova VL, Greenberg JP, Wrasidlo W, Overk C, Gonzalez T, Trejo M, Spencer B, Kosberg K, Masliah E. Molecular determinants of α-synuclein mutants' oligomerization and membrane interactions. ACS Chem Neurosci 2015; 6:403-16. [PMID: 25561023 DOI: 10.1021/cn500332w] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is associated with the formation of toxic α-synuclein oligomers that can penetrate the cell membrane. Familial forms of PD are caused by the point mutations A53T, A30P, E46K, and H50Q. Artificial point mutations E35K and E57K also increase oligomerization and pore formation. We generated structural conformations of α-synuclein and the above-mentioned mutants using molecular dynamics. We elucidated four main regions in these conformers contacting the membrane and found that the region including residues 39-45 (Zone2) may have maximum membrane penetration. E57K mutant had the highest rate of interaction with the membrane, followed by A53T, E46K, and E35K mutants and wild type (wt) α-synuclein. The mutant A30P had the smallest percentage of conformers that contact the membrane by Zone 2 than all other mutants and wt α-synuclein. These results were confirmed experimentally in vitro. We identified the key amino acids that can interact with the membrane (Y38, E62, and N65 (first hydrophilic layer); E104, E105, and D115 (second hydrophilic layer), and V15 and V26 (central hydrophobic layer)) and the residues that are involved in the interprotein contacts (L38, V48, V49, Q62, and T64). Understanding the molecular interactions of α-synuclein mutants is important for the design of compounds blocking the formation of toxic oligomers.
Collapse
Affiliation(s)
- Igor F. Tsigelny
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Yuriy Sharikov
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Valentina L. Kouznetsova
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Jerry P. Greenberg
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Wolf Wrasidlo
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Cassia Overk
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Tania Gonzalez
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Margarita Trejo
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Brian Spencer
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Kori Kosberg
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| | - Eliezer Masliah
- San Diego Supercomputer Center, ‡Moores Cancer Center, §Department of Neurosciences, and ∥Department of
Pathology, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
131
|
Iyer A, Petersen NO, Claessens MMAE, Subramaniam V. Amyloids of alpha-synuclein affect the structure and dynamics of supported lipid bilayers. Biophys J 2015; 106:2585-94. [PMID: 24940776 DOI: 10.1016/j.bpj.2014.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/14/2014] [Accepted: 05/01/2014] [Indexed: 02/06/2023] Open
Abstract
Interactions of monomeric alpha-synuclein (αS) with lipid membranes have been suggested to play an important role in initiating aggregation of αS. We have systematically analyzed the distribution and self-assembly of monomeric αS on supported lipid bilayers. We observe that at protein/lipid ratios higher than 1:10, αS forms micrometer-sized clusters, leading to observable membrane defects and decrease in lateral diffusion of both lipids and proteins. An αS deletion mutant lacking amino-acid residues 71-82 binds to membranes, but does not observably affect membrane integrity. Although this deletion mutant cannot form amyloid, significant amyloid formation is observed in the wild-type αS clusters. These results suggest that the process of amyloid formation, rather than binding of αS on membranes, is crucial in compromising membrane integrity.
Collapse
Affiliation(s)
- Aditya Iyer
- Nanoscale Biophysics Group, FOM Institute AMOLF, Amsterdam, The Netherlands; Nanobiophysics Group, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Nils O Petersen
- Nanobiophysics Group, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mireille M A E Claessens
- Nanobiophysics Group, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Vinod Subramaniam
- Nanoscale Biophysics Group, FOM Institute AMOLF, Amsterdam, The Netherlands; Nanobiophysics Group, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
132
|
Ardah MT, Paleologou KE, Lv G, Menon SA, Abul Khair SB, Lu JH, Safieh-Garabedian B, Al-Hayani AA, Eliezer D, Li M, El-Agnaf OMA. Ginsenoside Rb1 inhibits fibrillation and toxicity of alpha-synuclein and disaggregates preformed fibrils. Neurobiol Dis 2015; 74:89-101. [PMID: 25449909 PMCID: PMC4882765 DOI: 10.1016/j.nbd.2014.11.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/04/2014] [Accepted: 11/07/2014] [Indexed: 01/10/2023] Open
Abstract
Compelling evidence indicates that α-synuclein (α-syn) aggregation plays a central role in the pathogenesis of Parkinson's disease (PD) and other synucleinopathies. Identification of compounds that inhibit or reverse the aggregation process may thus represent a viable therapeutic strategy against PD and related disorders. Ginseng is a well-known medicinal plant that has been used in East Asia for more than two thousand years to treat several conditions. It is now understood that the pharmacological properties of ginseng can be attributed to its biologically active components, the ginsenosides, which in turn have been shown to have neuroprotective properties. We therefore sought to determine for the first time, the potential of the most frequently used and studied ginsenosides, namely Rg1, Rg3 and Rb1, as anti-amyloidogenic agents. The effect of Rg1, Rg3 and Rb1 on α-syn aggregation and toxicity was determined by an array of biophysical, biochemical and cell-culture-based techniques. Among the screened ginsenosides, only Rb1 was shown to be a potent inhibitor of α-syn fibrillation and toxicity. Additionally, Rb1 exhibited a strong ability to disaggregate preformed fibrils and to inhibit the seeded polymerization of α-syn. Interestingly, Rb1 was found to stabilize soluble non-toxic oligomers with no β-sheet content, that were susceptible to proteinase K digestion, and the binding of Rb1 to those oligomers may represent a potential mechanism of action. Thus, Rb1 could represent the starting point for designing new molecules that could be utilized as drugs for the treatment of PD and related disorders.
Collapse
Affiliation(s)
- Mustafa T Ardah
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Katerina E Paleologou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Guohua Lv
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, USA
| | - Sindhu A Menon
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Salema B Abul Khair
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Jia-Hong Lu
- State Key Lab of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | | | - Abdulmonem A Al-Hayani
- Department of Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - David Eliezer
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, USA
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Omar M A El-Agnaf
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates; Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
133
|
Chaudhary H, Stefanovic AND, Subramaniam V, Claessens MMAE. Membrane interactions and fibrillization of α-synuclein play an essential role in membrane disruption. FEBS Lett 2015; 588:4457-63. [PMID: 25448986 DOI: 10.1016/j.febslet.2014.10.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/13/2014] [Accepted: 10/15/2014] [Indexed: 12/22/2022]
Abstract
We studied α-synuclein (αS) aggregation in giant vesicles, and observed dramatic membrane disintegration, as well as lipid incorporation into micrometer-sized suprafibrillar aggregates. In the presence of dye-filled vesicles, dye leakage and fibrillization happen concurrently. However, growing fibrils do not impair the integrity of phospholipid vesicles that have a low affinity for αS. Seeding αS aggregation accelerates dye leakage, indicating that oligomeric species are not required to explain the observed effect. The evolving picture suggests that fibrils that appear in solution bind membranes and recruit membrane-bound monomers, resulting in lipid extraction, membrane destabilization and the formation of lipid-containing suprafibrillar aggregates.
Collapse
Affiliation(s)
- Himanshu Chaudhary
- Nanobiophysics Group, MESA+ Institute for Nanotechnology, Department of Science and Technology, University Twente, 7500 AE Enschede, The Netherlands
| | | | | | | |
Collapse
|
134
|
Möller I, Seeger S. Solid supported lipid bilayers from artificial and natural lipid mixtures – long-term stable, homogeneous and reproducible. J Mater Chem B 2015; 3:6046-6056. [DOI: 10.1039/c5tb00437c] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We show the assembly of reproducible, long-term stable, homogeneous solid supported lipid bilayers under flow conditions by the vesicle deposition method from various artificial and natural lipid mixtures.
Collapse
Affiliation(s)
- Isabelle Möller
- Department of Chemistry
- University of Zurich
- 8057 Zurich
- Switzerland
| | - Stefan Seeger
- Department of Chemistry
- University of Zurich
- 8057 Zurich
- Switzerland
| |
Collapse
|
135
|
Garten M, Prévost C, Cadart C, Gautier R, Bousset L, Melki R, Bassereau P, Vanni S. Methyl-branched lipids promote the membrane adsorption of α-synuclein by enhancing shallow lipid-packing defects. Phys Chem Chem Phys 2015; 17:15589-97. [DOI: 10.1039/c5cp00244c] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Reconstitution experiments on Giant Unilamellar Vesicles and Molecular Dynamics Simulations indicate that alpha-synuclein binds to neutral flat membranes in the presence of methyl-branched lipids.
Collapse
Affiliation(s)
| | | | | | - Romain Gautier
- Institut de Pharmacologie Moléculaire et Cellulaire
- Université de Nice Sophia-Antipolis and Centre National de la Recherche Scientifique
- UMR 7275
- 06560 Valbonne
- France
| | - Luc Bousset
- CNRS
- Paris Saclay Institute of Neuroscience
- Gif-sur-Yvette
- France
| | - Ronald Melki
- CNRS
- Paris Saclay Institute of Neuroscience
- Gif-sur-Yvette
- France
| | | | - Stefano Vanni
- Institut de Pharmacologie Moléculaire et Cellulaire
- Université de Nice Sophia-Antipolis and Centre National de la Recherche Scientifique
- UMR 7275
- 06560 Valbonne
- France
| |
Collapse
|
136
|
Shan Z, Cai S, Zhang T, Kuang L, Wang Q, Xiu H, Wen J, Gu H, Xu K. Effects of sevoflurane on leucine-rich repeat kinase 2-associated Drosophila model of Parkinson's disease. Mol Med Rep 2014; 11:2062-70. [PMID: 25406035 DOI: 10.3892/mmr.2014.2966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 09/24/2014] [Indexed: 11/05/2022] Open
Abstract
Patients with Parkinson's disease (PD) often require surgery, and therefore may receive inhalation anesthesia. However, it is currently unknown whether inhalation anesthetics affect the prognosis of the disease. Leucine‑rich repeat kinase 2 (LRRK2) genetic mutations are the most common cause of familial PD, contributing to ~39% of all cases in certain populations. The aim of the present study was to determine the effects of inhaled anesthetics on PD, by observing the influence of sevoflurane on a LRRK2‑associated Drosophila model of PD. PD transgenic Drosophila overexpressing LRRK2 were generated by crossing flies expressing an LRRK2 upstream activation sequence, with tyrosine hydroxylase (TH)‑Gal4 flies. Western blot analysis successfully verified that the transgenic Drosophila overexpressed LRRK2. Three days prior to eclosion, three genotypes of Drosophila were divided into four groups, and were exposed to air, 1, 2, or 3% sevoflurane, for 5 hours. Twenty‑four hours after the exposure, the electrophysiological activities of the projection neurons (PN) in the brains of the Drosophila were recorded using a patch clamp. The locomotor activities were tested on days 5, 10, 15, 20, 25, 30, 35 and 40 following eclosion. The frequency of miniature excitatory synaptic currents (mEPSCs) obtained from the PNs of the TH‑wild type LRRK2 (TH‑WT) Drosophila brain, following exposure to air (1.60±0.05 Hz), was lower as compared with the wild type LRRK2 (WT) (2.51±0.07 Hz) and W1118 (2.41±0.10 Hz) Drosophila. After exposure to 1, 2 and 3% sevoflurane, the frequency of mEPSCs in the brains of the TH‑WT group decreased to 0.82±0.04 Hz, 0.63±0.16 Hz and 0.55±0.04 Hz, respectively. The percentage decrease of the frequency of mEPSCs, from exposure to air to 1% sevoflurane, of the TH‑WT group (48.32%±3.08%) was significantly higher, as compared with the WT (39.17%±1.42%) and W1118 (35.10%±2.66%) groups, and there was no statistical difference between the WT and W1118 groups. The transgenic TH‑WT Drosophila presented an early decrease in locomotor ability, as compared with the WT and W1118 groups. Following a 5 hour exposure to sevoflurane, the percentage decrease of the climbing abilities of the TH‑WT group, from exposure to air to 1% sevoflurane, were significantly lower, as compared with the WT and W1118 groups. In conclusion, sevoflurane had negative effects on the control W1118 flies, and also severely aggravated the prognosis of PD in the LRRK2‑associated Drosophila model, through synaptic cholinergic deficits and impairment on locomotor abilities.
Collapse
Affiliation(s)
- Zhiming Shan
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Song Cai
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Tao Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Liting Kuang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Qi Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Huanhuan Xiu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jing Wen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Huaiyu Gu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Kangqing Xu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
137
|
Terakawa MS, Yagi H, Adachi M, Lee YH, Goto Y. Small liposomes accelerate the fibrillation of amyloid β (1-40). J Biol Chem 2014; 290:815-26. [PMID: 25406316 DOI: 10.1074/jbc.m114.592527] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The deposition of amyloid β (Aβ) peptides is a pathological hallmark of Alzheimer disease. Aβ peptides were previously considered to interact specifically with ganglioside-containing membranes. Several studies have suggested that Aβ peptides also bind to phosphatidylcholine membranes, which lead to deformation of membranes and fibrillation of Aβ. Moreover, the role of membrane curvature, one type of deformation produced by binding of proteins to a membrane, in the binding and fibrillation of Aβ remains unclear. To clearly understand the relationship between the binding, consequent membrane deformation, and fibrillation of Aβ, we examined the amyloid fibrillation of Aβ-(1-40) in the presence of liposomes of various sizes. Membrane curvature increased with a decrease in the size of the liposomes. We used liposomes made of 1,2-dioleoyl-sn-glycero-3-phosphocholine to eliminate electrostatic effects. The results obtained showed that liposomes of smaller sizes (≤50 nm) significantly accelerated the nucleation step, thereby shortening the lag time of fibrillation. On the other hand, liposomes of larger sizes decreased the amount of fibrils but did not notably affect the lag time. The morphologies of fibrils, which were monitored by total internal reflection fluorescence microscopy, atomic force microscopy, and transmission electron microscopy, revealed that the length of Aβ-(1-40) fibrils became shorter and the amount of amorphous aggregates became larger as liposomes increased in size. These results suggest that the curvature of membranes coupled with an increase in water-accessible hydrophobic regions is important for binding and concentrating Aβ monomers, leading to amyloid nucleation. Furthermore, amyloid fibrillation on membranes may compete with non-productive binding to produce amorphous aggregates.
Collapse
Affiliation(s)
- Mayu S Terakawa
- From the Institute for Protein Research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan
| | - Hisashi Yagi
- From the Institute for Protein Research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan
| | - Masayuki Adachi
- From the Institute for Protein Research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan
| | - Young-Ho Lee
- From the Institute for Protein Research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan
| | - Yuji Goto
- From the Institute for Protein Research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan
| |
Collapse
|
138
|
van Maarschalkerweerd A, Vetri V, Langkilde AE, Foderà V, Vestergaard B. Protein/lipid coaggregates are formed during α-synuclein-induced disruption of lipid bilayers. Biomacromolecules 2014; 15:3643-54. [PMID: 25210839 DOI: 10.1021/bm500937p] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Amyloid formation is associated with neurodegenerative diseases such as Parkinson's disease (PD). Significant α-synuclein (αSN) deposition in lipid-rich Lewy bodies is a hallmark of PD. Nonetheless, an unraveling of the connection between neurodegeneration and amyloid fibrils, including the molecular mechanisms behind potential amyloid-mediated toxic effects, is still missing. Interaction between amyloid aggregates and the lipid cell membrane is expected to play a key role in the disease progress. Here, we present experimental data based on hybrid analysis of two-photon-microscopy, solution small-angle X-ray scattering and circular dichroism data. Data show in real time changes in liposome morphology and stability upon protein addition and reveal that membrane disruption mediated by amyloidogenic αSN is associated with dehydration of anionic lipid membranes and stimulation of protein secondary structure. As a result of membrane fragmentation, soluble αSN:-lipid coaggregates are formed, hence, suggesting a novel molecular mechanism behind PD amyloid cytotoxicity.
Collapse
Affiliation(s)
- Andreas van Maarschalkerweerd
- Department of Drug Design and Pharmacology, University of Copenhagen , Universitetsparken 2, 2100 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
139
|
Reducing C-terminal-truncated alpha-synuclein by immunotherapy attenuates neurodegeneration and propagation in Parkinson's disease-like models. J Neurosci 2014; 34:9441-54. [PMID: 25009275 DOI: 10.1523/jneurosci.5314-13.2014] [Citation(s) in RCA: 234] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are common neurodegenerative disorders of the aging population, characterized by progressive and abnormal accumulation of α-synuclein (α-syn). Recent studies have shown that C-terminus (CT) truncation and propagation of α-syn play a role in the pathogenesis of PD/DLB. Therefore, we explored the effect of passive immunization against the CT of α-syn in the mThy1-α-syn transgenic (tg) mouse model, which resembles the striato-nigral and motor deficits of PD. Mice were immunized with the new monoclonal antibodies 1H7, 5C1, or 5D12, all directed against the CT of α-syn. CT α-syn antibodies attenuated synaptic and axonal pathology, reduced the accumulation of CT-truncated α-syn (CT-α-syn) in axons, rescued the loss of tyrosine hydroxylase fibers in striatum, and improved motor and memory deficits. Among them, 1H7 and 5C1 were most effective at decreasing levels of CT-α-syn and higher-molecular-weight aggregates. Furthermore, in vitro studies showed that preincubation of recombinant α-syn with 1H7 and 5C1 prevented CT cleavage of α-syn. In a cell-based system, CT antibodies reduced cell-to-cell propagation of full-length α-syn, but not of the CT-α-syn that lacked the 118-126 aa recognition site needed for antibody binding. Furthermore, the results obtained after lentiviral expression of α-syn suggest that antibodies might be blocking the extracellular truncation of α-syn by calpain-1. Together, these results demonstrate that antibodies against the CT of α-syn reduce levels of CT-truncated fragments of the protein and its propagation, thus ameliorating PD-like pathology and improving behavioral and motor functions in a mouse model of this disease.
Collapse
|
140
|
Goodchild SC, Sheynis T, Thompson R, Tipping KW, Xue WF, Ranson NA, Beales PA, Hewitt EW, Radford SE. β2-Microglobulin amyloid fibril-induced membrane disruption is enhanced by endosomal lipids and acidic pH. PLoS One 2014; 9:e104492. [PMID: 25100247 PMCID: PMC4123989 DOI: 10.1371/journal.pone.0104492] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 07/11/2014] [Indexed: 12/28/2022] Open
Abstract
Although the molecular mechanisms underlying the pathology of amyloidoses are not well understood, the interaction between amyloid proteins and cell membranes is thought to play a role in several amyloid diseases. Amyloid fibrils of β2-microglobulin (β2m), associated with dialysis-related amyloidosis (DRA), have been shown to cause disruption of anionic lipid bilayers in vitro. However, the effect of lipid composition and the chemical environment in which β2m-lipid interactions occur have not been investigated previously. Here we examine membrane damage resulting from the interaction of β2m monomers and fibrils with lipid bilayers. Using dye release, tryptophan fluorescence quenching and fluorescence confocal microscopy assays we investigate the effect of anionic lipid composition and pH on the susceptibility of liposomes to fibril-induced membrane damage. We show that β2m fibril-induced membrane disruption is modulated by anionic lipid composition and is enhanced by acidic pH. Most strikingly, the greatest degree of membrane disruption is observed for liposomes containing bis(monoacylglycero)phosphate (BMP) at acidic pH, conditions likely to reflect those encountered in the endocytic pathway. The results suggest that the interaction between β2m fibrils and membranes of endosomal origin may play a role in the molecular mechanism of β2m amyloid-associated osteoarticular tissue destruction in DRA.
Collapse
Affiliation(s)
- Sophia C. Goodchild
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Tania Sheynis
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Rebecca Thompson
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Kevin W. Tipping
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Wei-Feng Xue
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Neil A. Ranson
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Paul A. Beales
- Astbury Centre for Structural Molecular Biology and School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - Eric W. Hewitt
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Sheena E. Radford
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
141
|
Nübling GS, Levin J, Bader B, Lorenzl S, Hillmer A, Högen T, Kamp F, Giese A. Modelling Ser129 phosphorylation inhibits membrane binding of pore-forming alpha-synuclein oligomers. PLoS One 2014; 9:e98906. [PMID: 24911099 PMCID: PMC4049638 DOI: 10.1371/journal.pone.0098906] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 05/08/2014] [Indexed: 12/16/2022] Open
Abstract
Background In several neurodegenerative diseases, hyperphosphorylation at position Ser129 is found in fibrillar deposits of alpha-synuclein (asyn), implying a pathophysiological role of asyn phosphorylation in neurodegeneration. However, recent animal models applying asyn phosphorylation mimics demonstrated a protective effect of phosphorylation. Since metal-ion induced asyn oligomers were identified as a potential neurotoxic aggregate species with membrane pore-forming abilities, the current study was undertaken to determine effects of asyn phosphorylation on oligomer membrane binding. Methods We investigated the influence of S129 phosphorylation on interactions of metal-ion induced asyn oligomers with small unilamellar lipid vesicles (SUV) composed of POPC and DPPC applying the phosphorylation mimic asyn129E. Confocal single-particle fluorescence techniques were used to monitor membrane binding at the single-particle level. Results Binding of asyn129E monomers to gel-state membranes (DPPC-SUV) is slightly reduced compared to wild-type asyn, while no interactions with membranes in the liquid-crystalline state (POPC-SUV) are seen for both asyn and asyn129E. Conversely, metal-ion induced oligomer formation is markedly increased in asyn129E. Surprisingly, membrane binding to POPC-SUV is nearly absent in Fe3+ induced asyn129E oligomers and markedly reduced in Al3+ induced oligomers. Conclusion The protective effect of pseudophosphorylation seen in animal models may be due to impeded oligomer membrane binding. Phosphorylation at Ser129 may thus have a protective effect against neurotoxic asyn oligomers by preventing oligomer membrane binding and disruption of the cellular electrophysiological equilibrium. Importantly, these findings put a new complexion on experimental pharmaceutical interventions against POLO-2 kinase.
Collapse
Affiliation(s)
- Georg Sebastian Nübling
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
- Department of Palliative Medicine, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
- * E-mail:
| | - Johannes Levin
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
| | - Benedikt Bader
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Lorenzl
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
- Department of Palliative Medicine, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
- Endowed Professorship for Interdisciplinary Research in Palliative Care, Institute of Nursing Science and –Practice, Paracelsus Medical University, Salzburg, Austria
| | - Andreas Hillmer
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
| | - Tobias Högen
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
| | - Frits Kamp
- Adolf-Butenandt-Institute, Munich, Germany
| | - Armin Giese
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
142
|
Probing the interplay between amyloidogenic proteins and membranes using lipid monolayers and bilayers. Adv Colloid Interface Sci 2014; 207:81-92. [PMID: 24200086 DOI: 10.1016/j.cis.2013.10.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/11/2013] [Accepted: 10/13/2013] [Indexed: 11/21/2022]
Abstract
Many degenerative diseases such as Alzheimer's and Parkinson's involve proteins that have a tendency to misfold and aggregate eventually forming amyloid fibers. This review describes the use of monolayers, bilayers, supported membranes, and vesicles as model systems that have helped elucidate the mechanisms and consequences of the interactions between amyloidogenic proteins and membranes. These are twofold: membranes favor the formation of amyloid structures and these induce damage in those membranes. We describe studies that show how interfaces, especially charged ones, favor amyloidogenic protein aggregation by several means. First, surfaces increase the effective protein concentration reducing a three-dimensional system to a two-dimensional one. Second, charged surfaces allow electrostatic interactions with the protein. Anionic lipids as well as rafts, rich in cholesterol and gangliosides, prove to play an especially important role. Finally, these amphipathic systems also offer a hydrophobic environment favoring conformational changes, oligomerization, and eventual formation of mature fibers. In addition, we examine several models for membrane permeabilization: protein pores, leakage induced by extraction of lipids, chaotic pores, and membrane tension, presenting illustrative examples of experimental evidence in support of these models. The picture that emerges from recent work is one where more than one mechanism is in play. Which mechanism prevails depends on the protein, its aggregation state, and the lipid environment in which the interactions occur.
Collapse
|
143
|
Rockenstein E, Nuber S, Overk CR, Ubhi K, Mante M, Patrick C, Adame A, Trejo-Morales M, Gerez J, Picotti P, Jensen PH, Campioni S, Riek R, Winkler J, Gage FH, Winner B, Masliah E. Accumulation of oligomer-prone α-synuclein exacerbates synaptic and neuronal degeneration in vivo. ACTA ACUST UNITED AC 2014; 137:1496-513. [PMID: 24662516 DOI: 10.1093/brain/awu057] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In Parkinson's disease and dementia with Lewy bodies, α-synuclein aggregates to form oligomers and fibrils; however, the precise nature of the toxic α-synuclein species remains unclear. A number of synthetic α-synuclein mutations were recently created (E57K and E35K) that produce species of α-synuclein that preferentially form oligomers and increase α-synuclein-mediated toxicity. We have shown that acute lentiviral expression of α-synuclein E57K leads to the degeneration of dopaminergic neurons; however, the effects of chronic expression of oligomer-prone α-synuclein in synapses throughout the brain have not been investigated. Such a study could provide insight into the possible mechanism(s) through which accumulation of α-synuclein oligomers in the synapse leads to neurodegeneration. For this purpose, we compared the patterns of neurodegeneration and synaptic damage between a newly generated mThy-1 α-synuclein E57K transgenic mouse model that is prone to forming oligomers and the mThy-1 α-synuclein wild-type mouse model (Line 61), which accumulates various forms of α-synuclein. Three lines of α-synuclein E57K (Lines 9, 16 and 54) were generated and compared with the wild-type. The α-synuclein E57K Lines 9 and 16 were higher expressings of α-synuclein, similar to α-synuclein wild-type Line 61, and Line 54 was a low expressing of α-synuclein compared to Line 61. By immunoblot analysis, the higher-expressing α-synuclein E57K transgenic mice showed abundant oligomeric, but not fibrillar, α-synuclein whereas lower-expressing mice accumulated monomeric α-synuclein. Monomers, oligomers, and fibrils were present in α-synuclein wild-type Line 61. Immunohistochemical and ultrastructural analyses demonstrated that α-synuclein accumulated in the synapses but not in the neuronal cells bodies, which was different from the α-synuclein wild-type Line 61, which accumulates α-synuclein in the soma. Compared to non-transgenic and lower-expressing mice, the higher-expressing α-synuclein E57K mice displayed synaptic and dendritic loss, reduced levels of synapsin 1 and synaptic vesicles, and behavioural deficits. Similar alterations, but to a lesser extent, were seen in the α-synuclein wild-type mice. Moreover, although the oligomer-prone α-synuclein mice displayed neurodegeneration in the frontal cortex and hippocampus, the α-synuclein wild-type only displayed neuronal loss in the hippocampus. These results support the hypothesis that accumulating oligomeric α-synuclein may mediate early synaptic pathology in Parkinson's disease and dementia with Lewy bodies by disrupting synaptic vesicles. This oligomer-prone model might be useful for evaluating therapies directed at oligomer reduction.
Collapse
Affiliation(s)
- Edward Rockenstein
- 1 Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Lara C, Reynolds NP, Berryman JT, Xu A, Zhang A, Mezzenga R. ILQINS Hexapeptide, Identified in Lysozyme Left-Handed Helical Ribbons and Nanotubes, Forms Right-Handed Helical Ribbons and Crystals. J Am Chem Soc 2014; 136:4732-9. [DOI: 10.1021/ja500445z] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Cecile Lara
- Food & Soft Materials, Department of Health Science & Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23, 8092 Zürich, Switzerland
| | - Nicholas P. Reynolds
- Materials
Science and Engineering, CSIRO, Private Bag 10, Bayview Avenue, Clayton, Vic 3169, Australia
| | - Joshua T. Berryman
- Faculty
of Science Technology and Communication, University of Luxembourg, 162a Avenue de la Faïencerie, L-1511 Luxembourg
| | - Anqiu Xu
- Department
of Polymer Materials, Shanghai University, Nanchen Street 333, Shanghai 200444, China
| | - Afang Zhang
- Department
of Polymer Materials, Shanghai University, Nanchen Street 333, Shanghai 200444, China
| | - Raffaele Mezzenga
- Food & Soft Materials, Department of Health Science & Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23, 8092 Zürich, Switzerland
| |
Collapse
|
145
|
Asi YT, Simpson JE, Heath PR, Wharton SB, Lees AJ, Revesz T, Houlden H, Holton JL. Alpha-synuclein mRNA expression in oligodendrocytes in MSA. Glia 2014; 62:964-70. [PMID: 24590631 PMCID: PMC4238782 DOI: 10.1002/glia.22653] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 01/25/2014] [Accepted: 02/10/2014] [Indexed: 01/14/2023]
Abstract
Multiple system atrophy (MSA) is a progressive neurodegenerative disease presenting clinically with parkinsonian, cerebellar, and autonomic features. α-Synuclein (αsyn), encoded by the gene SNCA, is the main constituent of glial cytoplasmic inclusion (GCI) found in oligodendrocytes in MSA, but the methods of its accumulation have not been established. The aim of this study is to investigate alterations in regional and cellular SNCA mRNA expression in MSA as a possible substrate for GCI formation. Quantitative reverse transcription polymerase chain reaction (qPCR) was performed on postmortem brain samples from 15 MSA, 5 IPD, and 5 control cases to investigate regional expression in the frontal and occipital regions, dorsal putamen, pontine base, and cerebellum. For cellular expression analysis, neurons and oligodendrocytes were isolated by laser-capture microdissection from five MSA and five control cases. SNCA mRNA expression was not significantly different between the MSA, IPD and control cases in all regions (multilevel model, P = 0.14). After adjusting for group effect, the highest expression was found in the occipital cortex while the lowest was in the putamen (multilevel model, P < 0.0001). At the cellular level, MSA oligodendrocytes expressed more SNCA than control oligodendrocytes and expression in MSA neurons was slightly lower than that in controls, however, these results did not reach statistical significance. We have demonstrated regional variations in SNCA expression, which is higher in cortical than subcortical regions. This study is the first to demonstrate SNCA mRNA expression by oligodendrocytes in human postmortem tissue using qPCR and, although not statistically significant, could suggest that this may be increased in MSA compared to controls.
Collapse
Affiliation(s)
- Yasmine T Asi
- Queen Square Brain Bank, Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Reynolds NP, Charnley M, Mezzenga R, Hartley PG. Engineered lysozyme amyloid fibril networks support cellular growth and spreading. Biomacromolecules 2014; 15:599-608. [PMID: 24432698 DOI: 10.1021/bm401646x] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Fibrous networks assembled from synthetic peptides are promising candidates for biomimetic cell culture platforms and implantable biomaterials. The ability of the materials to reproduce physiological cell-matrix interactions is essential. However, the synthetic complexity of such systems limits their applications, thus alternative materials are desirable. Here, we design lysozyme derived amyloid fibril networks with controllable topographies, and perform a comprehensive study of the response of cultured fibroblast and epithelial cells. At high surface coverage a favorable increase in spreading and the generation of focal adhesions was observed, due to a combination of biomimetic chemistry and morphology. Their ease of synthesis, makes the nanoscale fibrils presented here ideal materials for future clinical applications whereby large volumes of biomimetic biomaterials are required. Furthermore, the surface chemistry of the fibrils is sufficient for the promotion of focal adhesions with cultured cells, eliminating the need for complex protocols for fibril decoration with bioactive moieties.
Collapse
Affiliation(s)
- Nicholas P Reynolds
- CSIRO, Materials Science and Engineering, Private Bag 10, Bayview Avenue, Clayton, Victoria 3169, Australia
| | | | | | | |
Collapse
|
147
|
Biophysical groundwork as a hinge to unravel the biology of α-synuclein aggregation and toxicity. Q Rev Biophys 2014; 47:1-48. [PMID: 24443929 DOI: 10.1017/s0033583513000097] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Alpha-synuclein (aS) and its aggregation properties are central in the development and spread of Parkinson's disease. Point mutations and multiplications of the SNCA gene encoding aS cause autosomal dominant forms of the disorder. Moreover, protein inclusions found in the surviving neurons of parkinsonian brains consist mainly of a fibrillar form of aS. Aggregates of aS, which form a transient, complex and heterogeneous ensemble, participate in a wide variety of toxic mechanisms that may be amplified by aS spreading among neighbouring neurons. Recently, significant effort has been directed into the study of the aS aggregation process and the impact of aS aggregates on neuron survival. In this review, we present state-of-the-art biophysical studies on the aS aggregation process in vitro and in cellular models. We comprehensively review the new insights generated by the recent biophysical investigations, which could provide a solid basis from which to design future biomedical studies. The diverse cellular models of aS toxicity and their potential use in the biophysical investigation are also discussed.
Collapse
|
148
|
Lee SJC, Lee JW, Choi TS, Jin KS, Lee S, Ban C, Kim HI. Probing Conformational Change of Intrinsically Disordered α-Synuclein to Helical Structures by Distinctive Regional Interactions with Lipid Membranes. Anal Chem 2014; 86:1909-16. [DOI: 10.1021/ac404132g] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Shin Jung C. Lee
- Department of Chemistry, ‡Pohang Accelerator
Laboratory, §Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang, North Gyeongsang 790-784, South Korea
| | - Jong Wha Lee
- Department of Chemistry, ‡Pohang Accelerator
Laboratory, §Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang, North Gyeongsang 790-784, South Korea
| | - Tae Su Choi
- Department of Chemistry, ‡Pohang Accelerator
Laboratory, §Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang, North Gyeongsang 790-784, South Korea
| | - Kyeong Sik Jin
- Department of Chemistry, ‡Pohang Accelerator
Laboratory, §Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang, North Gyeongsang 790-784, South Korea
| | - Seonghwan Lee
- Department of Chemistry, ‡Pohang Accelerator
Laboratory, §Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang, North Gyeongsang 790-784, South Korea
| | - Changill Ban
- Department of Chemistry, ‡Pohang Accelerator
Laboratory, §Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang, North Gyeongsang 790-784, South Korea
| | - Hugh I. Kim
- Department of Chemistry, ‡Pohang Accelerator
Laboratory, §Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang, North Gyeongsang 790-784, South Korea
| |
Collapse
|
149
|
Fecchio C, De Franceschi G, Relini A, Greggio E, Dalla Serra M, Bubacco L, Polverino de Laureto P. α-Synuclein oligomers induced by docosahexaenoic acid affect membrane integrity. PLoS One 2013; 8:e82732. [PMID: 24312431 PMCID: PMC3843715 DOI: 10.1371/journal.pone.0082732] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/04/2013] [Indexed: 11/18/2022] Open
Abstract
A key feature of Parkinson disease is the aggregation of α-synuclein and its intracellular deposition in fibrillar form. Increasing evidence suggests that the pathogenicity of α-synuclein is correlated with the activity of oligomers formed in the early stages of its aggregation process. Oligomers toxicity seems to be associated with both their ability to bind and affect the integrity of lipid membranes. Previously, we demonstrated that α-synuclein forms oligomeric species in the presence of docosahexaenoic acid and that these species are toxic to cells. Here we studied how interaction of these oligomers with membranes results in cell toxicity, using cellular membrane-mimetic and cell model systems. We found that α-synuclein oligomers are able to interact with large and small unilamellar negatively charged vesicles acquiring an increased amount of α-helical structure, which induces small molecules release. We explored the possibility that oligomers effects on membranes could be due to pore formation, to a detergent-like effect or to fibril growth on the membrane. Our biophysical and cellular findings are consistent with a model where α-synuclein oligomers are embedded into the lipid bilayer causing transient alteration of membrane permeability.
Collapse
Affiliation(s)
- Chiara Fecchio
- CRIBI, Biotechnology Centre, Department of Pharmaceutical Sciences, University of Padova, Padova, Italy
| | - Giorgia De Franceschi
- CRIBI, Biotechnology Centre, Department of Pharmaceutical Sciences, University of Padova, Padova, Italy
| | | | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
| | - Mauro Dalla Serra
- Institute of Biophysics, National Research Council of Italy and Bruno Kessler Foundation, Trento, Italy
| | - Luigi Bubacco
- Department of Biology, University of Padova, Padova, Italy
| | | |
Collapse
|
150
|
The interplay between lipids and dopamine on α-synuclein oligomerization and membrane binding. Biosci Rep 2013; 33:BSR20130092. [PMID: 24066973 PMCID: PMC3804888 DOI: 10.1042/bsr20130092] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The deposition of α-syn (α-synuclein) as amyloid fibrils and the selective loss of DA (dopamine) containing neurons in the substantia nigra are two key features of PD (Parkinson's disease). α-syn is a natively unfolded protein and adopts an α-helical conformation upon binding to lipid membrane. Oligomeric species of α-syn have been proposed to be the pathogenic species associated with PD because they can bind lipid membranes and disrupt membrane integrity. DA is readily oxidized to generate reactive intermediates and ROS (reactive oxygen species) and in the presence of DA, α-syn form of SDS-resistant soluble oligomers. It is postulated that the formation of the α-syn:DA oligomers involves the cross-linking of DA-melanin with α-syn, via covalent linkage, hydrogen and hydrophobic interactions. We investigate the effect of lipids on DA-induced α-syn oligomerization and studied the ability of α-syn:DA oligomers to interact with lipids vesicles. Our results show that the interaction of α-syn with lipids inhibits the formation of DA-induced α-syn oligomers. Moreover, the α-syn:DA oligomer cannot interact with lipid vesicles or cause membrane permeability. Thus, the formation of α-syn:DA oligomers may alter the actions of α-syn which require membrane association, leading to disruption of its normal cellular function.
Collapse
|