51
|
Hadi F, Akrami H, Totonchi M, Barzegar A, Nabavi SM, Shahpasand K. α-synuclein abnormalities trigger focal tau pathology, spreading to various brain areas in Parkinson disease. J Neurochem 2021; 157:727-751. [PMID: 33264426 DOI: 10.1111/jnc.15257] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/28/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022]
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disorder, whose prevalence is 2~3% in the population over 65. α-Synuclein aggregation is the major pathological hallmark of PD. However, recent studies have demonstrated enhancing evidence of tau pathology in PD. Despite extensive considerations, thus far, the actual spreading mechanism of neurodegeneration has remained elusive in a PD brain. This study aimed to further investigate the development of α-synuclein and tau pathology. We employed various PD models, including cultured neurons treated with either 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or with recombinant α-synuclein. Also, we studied dopaminergic neurons of cytokine Interferon-β knock-out. Moreover, we examined rats treated with 6-hydroxydopamine, Rhesus monkeys administrated with MPTP neurotoxin, and finally, human post-mortem brains. We found the α-synuclein phosphorylation triggers tau pathogenicity. Also, we observed more widespread phosphorylated tau than α-synuclein with prion-like nature in various brain areas. We optionally removed P-tau or P-α-synuclein from cytokine interferon-β knock out with respective monoclonal antibodies. We found that tau immunotherapy suppressed neurodegeneration more than α-synuclein elimination. Our findings indicate that the pathogenic tau could be one of the leading causes of comprehensive neurodegeneration triggered by PD. Thus, we can propose an efficient therapeutic target to fight the devastating disorder.
Collapse
Affiliation(s)
- Fatemeh Hadi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Hassan Akrami
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Mehdi Totonchi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | | | - Seyed Massood Nabavi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| |
Collapse
|
52
|
Ramalingam N, Dettmer U. Temperature is a key determinant of alpha- and beta-synuclein membrane interactions in neurons. J Biol Chem 2021; 296:100271. [PMID: 33428933 PMCID: PMC7949061 DOI: 10.1016/j.jbc.2021.100271] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 02/02/2023] Open
Abstract
Aggregation of α-synuclein (αS) leads to the hallmark neuropathology of Parkinson's disease (PD) and related synucleinopathies. αS has been described to exist in both cytosolic and membrane-associated forms, the relative abundance of which has remained unsettled. To study αS under the most relevant conditions by a quantitative method, we cultured and matured rodent primary cortical neurons for >17 days and determined αS cytosol:membrane distribution via centrifugation-free sequential extractions based on the weak ionic detergent digitonin. We noticed that at lower temperatures (4 °C or room temperature), αS was largely membrane-associated. At 37 °C, however, αS solubility was markedly increased. In contrast, the extraction of control proteins (GAPDH, cytosolic; calnexin, membrane) was not affected by temperature. When we compared the relative distribution of the synuclein homologs αS and β-synuclein (βS) under various conditions that differed in temperature and digitonin concentration (200-1200 μg/ml), we consistently found αS to be more membrane-associated than βS. Both proteins, however, exhibited temperature-dependent membrane binding. Under the most relevant conditions (37 °C and 800 μg/ml digitonin, i.e., the lowest digitonin concentration that extracted cytosolic GAPDH to near completion), cytosolic distribution was 49.8% ± 9.0% for αS and 63.6% ± 6.6% for βS. PD-linked αS A30P was found to be largely cytosolic, confirming previous studies that had used different methods. Our work highlights the dynamic nature of cellular synuclein behavior and has important implications for protein-biochemical and cell-biological studies of αS proteostasis, such as testing the effects of genetic and pharmacological manipulations.
Collapse
Affiliation(s)
- Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
53
|
Cawood EE, Karamanos TK, Wilson AJ, Radford SE. Visualizing and trapping transient oligomers in amyloid assembly pathways. Biophys Chem 2021; 268:106505. [PMID: 33220582 PMCID: PMC8188297 DOI: 10.1016/j.bpc.2020.106505] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/31/2022]
Abstract
Oligomers which form during amyloid fibril assembly are considered to be key contributors towards amyloid disease. However, understanding how such intermediates form, their structure, and mechanisms of toxicity presents significant challenges due to their transient and heterogeneous nature. Here, we discuss two different strategies for addressing these challenges: use of (1) methods capable of detecting lowly-populated species within complex mixtures, such as NMR, single particle methods (including fluorescence and force spectroscopy), and mass spectrometry; and (2) chemical and biological tools to bias the amyloid energy landscape towards specific oligomeric states. While the former methods are well suited to following the kinetics of amyloid assembly and obtaining low-resolution structural information, the latter are capable of producing oligomer samples for high-resolution structural studies and inferring structure-toxicity relationships. Together, these different approaches should enable a clearer picture to be gained of the nature and role of oligomeric intermediates in amyloid formation and disease.
Collapse
Affiliation(s)
- Emma E Cawood
- Astbury Centre for Structural Molecular Biology, School of Chemistry, University of Leeds, LS2 9JT, UK; Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, UK
| | - Theodoros K Karamanos
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, UK; Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew J Wilson
- Astbury Centre for Structural Molecular Biology, School of Chemistry, University of Leeds, LS2 9JT, UK.
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, UK.
| |
Collapse
|
54
|
Musteikytė G, Jayaram AK, Xu CK, Vendruscolo M, Krainer G, Knowles TPJ. Interactions of α-synuclein oligomers with lipid membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183536. [PMID: 33373595 DOI: 10.1016/j.bbamem.2020.183536] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/24/2022]
Abstract
Parkinson's disease is an increasingly prevalent and currently incurable neurodegenerative disorder. At the molecular level, this disease is characterized by the formation of aberrant intracellular protein deposits known as Lewy bodies. Oligomeric forms of the protein α-synuclein (αS), which are believed to be both intermediates and by-products of Lewy body formation, are considered to be the main pathogenic species. Interactions of such oligomers with lipid membranes are increasingly emerging as a major molecular pathway underpinning their toxicity. Here we review recent progress in our understanding of the interactions of αS oligomers with lipid membranes. We highlight key structural and biophysical features of αS oligomers, the effects of these features on αS oligomer membrane binding properties, and resultant implications for understanding the etiology of Parkinson's disease. We discuss mechanistic modes of αS oligomer-lipid membrane interactions and the effects of environmental factors to such modes. Finally, we provide an overview of the current understanding of the main molecular determinants of αS oligomer toxicity in vivo.
Collapse
Affiliation(s)
- Greta Musteikytė
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Akhila K Jayaram
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom; Cavendish Laboratory, University of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| | - Catherine K Xu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Georg Krainer
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom; Cavendish Laboratory, University of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, United Kingdom.
| |
Collapse
|
55
|
Sun Y, Pham AN, Hider RC, Zheng H, Waite TD. Effectiveness of the Iron Chelator CN128 in Mitigating the Formation of Dopamine Oxidation Products Associated with the Progression of Parkinson's Disease. ACS Chem Neurosci 2020; 11:3646-3657. [PMID: 33143428 DOI: 10.1021/acschemneuro.0c00557] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The occurrence and progression of Parkinson's disease (PD) has been associated with the observation of elevated iron concentrations in the substantia nigra pars compacta (SNpc). While the reasons for the impact of elevated iron concentrations remain unclear, one hypothesis is that the presence of labile iron induces the oxidation of dopamine (DA) to toxic quinones such as aminochrome (DAC) and reactive oxygen species (ROS). As such, one of the proposed therapeutic strategies has been the use of iron chelators such as deferiprone (DFP) (which is recognized to have limitations related to its rapid degradation in the liver) to reduce the concentration of labile iron. In this study, a detailed investigation regarding the novel iron chelator, CN128, was conducted and a kinetic model developed to elucidate the fundamental behavior of this chelator. The results in this work reveal that CN128 is effective in alleviating the toxicity induced by iron and DA to neurons when DA is present at moderate concentrations. When all the iron is chelated by CN128, the formation of DAC and the oxidation of DA can be reduced to levels identical to that in the absence of iron. The production of H2O2 is lower than that generated via the autoxidation of the same amount of DA. However, when severe leakage of DA occurs, the application of CN128 is insufficient to alleviate the associated toxicity. This is attibuted to the less important role of iron in the production of toxic intermediates at high concentrations of DA. CN128 is superior to DFP with regard to the reduction in formation of DAC and elevation in DA concentration. In summary, the results of this study suggest that prodromal application of the chelator CN128 could be effective in preventing the onset and slowing the early stage development of PD symptoms associated with oxidants and toxic intermediates resulting from the iron-mediated oxidation of the neurotransmitter dopamine with CN128 likely to be superior to DFP in view of its greater in vivo availability and less problematic side effects.
Collapse
Affiliation(s)
- Yingying Sun
- Water Research Centre and School of Civil and Environmental Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - A. Ninh Pham
- Water Research Centre and School of Civil and Environmental Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Robert C. Hider
- Institute of Pharmaceutical Science, King’s College, London, WC2R 2LS, United Kingdom
| | - Haolin Zheng
- Water Research Centre and School of Civil and Environmental Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - T. David Waite
- Water Research Centre and School of Civil and Environmental Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
56
|
Farotti L, Paolini Paoletti F, Simoni S, Parnetti L. Unraveling Pathophysiological Mechanisms of Parkinson's Disease: Contribution of CSF Biomarkers. Biomark Insights 2020; 15:1177271920964077. [PMID: 33110345 PMCID: PMC7555566 DOI: 10.1177/1177271920964077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 01/08/2023] Open
Abstract
Diagnosis of Parkinson's disease (PD) relies on clinical history and physical examination, but misdiagnosis is common in early stages. Identification of biomarkers for PD may allow for early and more precise diagnosis and provide information about prognosis. Developments in analytical chemistry allow for the detection of a large number of molecules in cerebrospinal fluid (CSF), which are known to be associated with the pathogenesis of PD. Given the pathophysiology of PD, CSF α-synuclein species have the strongest rationale for use, also providing encouraging preliminary results in terms of early diagnosis. In the field of classical Alzheimer's disease (AD) biomarkers, low CSF Aβ42 levels have shown a robust prognostic value in terms of development of cognitive impairment. Other CSF biomarkers including lysosomal enzymes, neurofilament light chain, markers of neuroinflammation and oxidative stress, although promising, have not proved to be reliable for diagnostic and prognostic purposes yet. Overall, the implementation of CSF biomarkers may give a substantial contribution to the optimal use of disease-modifying drugs.
Collapse
Affiliation(s)
- Lucia Farotti
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | | | - Simone Simoni
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
57
|
Hill I, Palombo M, Santin M, Branzoli F, Philippe AC, Wassermann D, Aigrot MS, Stankoff B, Baron-Van Evercooren A, Felfli M, Langui D, Zhang H, Lehericy S, Petiet A, Alexander DC, Ciccarelli O, Drobnjak I. Machine learning based white matter models with permeability: An experimental study in cuprizone treated in-vivo mouse model of axonal demyelination. Neuroimage 2020; 224:117425. [PMID: 33035669 DOI: 10.1016/j.neuroimage.2020.117425] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 01/14/2023] Open
Abstract
The intra-axonal water exchange time (τi), a parameter associated with axonal permeability, could be an important biomarker for understanding and treating demyelinating pathologies such as Multiple Sclerosis. Diffusion-Weighted MRI (DW-MRI) is sensitive to changes in permeability; however, the parameter has so far remained elusive due to the lack of general biophysical models that incorporate it. Machine learning based computational models can potentially be used to estimate such parameters. Recently, for the first time, a theoretical framework using a random forest (RF) regressor suggests that this is a promising new approach for permeability estimation. In this study, we adopt such an approach and for the first time experimentally investigate it for demyelinating pathologies through direct comparison with histology. We construct a computational model using Monte Carlo simulations and an RF regressor in order to learn a mapping between features derived from DW-MRI signals and ground truth microstructure parameters. We test our model in simulations, and find strong correlations between the predicted and ground truth parameters (intra-axonal volume fraction f: R2 =0.99, τi: R2 =0.84, intrinsic diffusivity d: R2 =0.99). We then apply the model in-vivo, on a controlled cuprizone (CPZ) mouse model of demyelination, comparing the results from two cohorts of mice, CPZ (N=8) and healthy age-matched wild-type (WT, N=8). We find that the RF model estimates sensible microstructure parameters for both groups, matching values found in literature. Furthermore, we perform histology for both groups using electron microscopy (EM), measuring the thickness of the myelin sheath as a surrogate for exchange time. Histology results show that our RF model estimates are very strongly correlated with the EM measurements (ρ = 0.98 for f, ρ = 0.82 for τi). Finally, we find a statistically significant decrease in τi in all three regions of the corpus callosum (splenium/genu/body) of the CPZ cohort (<τi>=310ms/330ms/350ms) compared to the WT group (<τi>=370ms/370ms/380ms). This is in line with our expectations that τi is lower in regions where the myelin sheath is damaged, as axonal membranes become more permeable. Overall, these results demonstrate, for the first time experimentally and in vivo, that a computational model learned from simulations can reliably estimate microstructure parameters, including the axonal permeability .
Collapse
Affiliation(s)
- Ioana Hill
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK
| | - Marco Palombo
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK.
| | - Mathieu Santin
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France; Institut du Cerveau et de la Moelle épinière, ICM, Centre de NeuroImagerie de Recherche, CENIR, Paris, France
| | - Francesca Branzoli
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France; Institut du Cerveau et de la Moelle épinière, ICM, Centre de NeuroImagerie de Recherche, CENIR, Paris, France
| | - Anne-Charlotte Philippe
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France
| | - Demian Wassermann
- Université Côte d'Azur, Inria, Sophia-Antipolis, France; Parietal, CEA, Inria, Saclay, Île-de-France
| | - Marie-Stephane Aigrot
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France
| | - Bruno Stankoff
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France; AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Anne Baron-Van Evercooren
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France
| | - Mehdi Felfli
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France
| | - Dominique Langui
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France
| | - Hui Zhang
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK
| | - Stephane Lehericy
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France; Institut du Cerveau et de la Moelle épinière, ICM, Centre de NeuroImagerie de Recherche, CENIR, Paris, France
| | - Alexandra Petiet
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm 1127, CNRS UMR 7225, F-75013, Paris, France; Institut du Cerveau et de la Moelle épinière, ICM, Centre de NeuroImagerie de Recherche, CENIR, Paris, France
| | - Daniel C Alexander
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK
| | - Olga Ciccarelli
- Dept. of Neuroinflammation, University College London, Queen Square Institute of Neurology, University College London, London, UK
| | - Ivana Drobnjak
- Centre for Medical Image Computing and Dept of Computer Science, University College London, London, UK
| |
Collapse
|
58
|
Kumar ST, Jagannath S, Francois C, Vanderstichele H, Stoops E, Lashuel HA. How specific are the conformation-specific α-synuclein antibodies? Characterization and validation of 16 α-synuclein conformation-specific antibodies using well-characterized preparations of α-synuclein monomers, fibrils and oligomers with distinct structures and morphology. Neurobiol Dis 2020; 146:105086. [PMID: 32971232 DOI: 10.1016/j.nbd.2020.105086] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/30/2020] [Accepted: 09/17/2020] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence suggests that alpha-synuclein (α-syn) oligomers are obligate intermediates in the pathway involved in α-syn fibrillization and Lewy body (LB) formation, and may also accumulate within LBs in Parkinson's disease (PD) and other synucleinopathies. Therefore, the development of tools and methods to detect and quantify α-syn oligomers has become increasingly crucial for mechanistic studies to understand their role in PD, and to develop new diagnostic methods and therapies for PD and other synucleinopathies. The majority of these tools and methods rely primarily on the use of aggregation state-specific or conformation-specific antibodies. Given the impact of the data and knowledge generated using these antibodies on shaping the foundation and directions of α-syn and PD research, it is crucial that these antibodies are thoroughly characterized, and their specificity or ability to capture diverse α-syn species is tested and validated. Herein, we describe an antibody characterization and validation pipeline that allows a systematic investigation of the specificity of α-syn antibodies using well-defined and well-characterized preparations of various α-syn species, including monomers, fibrils, and different oligomer preparations that are characterized by distinct morphological, chemical and secondary structure properties. This pipeline was used to characterize 18 α-syn antibodies, 16 of which have been reported as conformation- or oligomer-specific antibodies, using an array of techniques, including immunoblot analysis (slot blot and Western blot), a digital ELISA assay using single molecule array technology and surface plasmon resonance. Our results show that i) none of the antibodies tested are specific for one particular type of α-syn species, including monomers, oligomers or fibrils; ii) all antibodies that were reported to be oligomer-specific also recognized fibrillar α-syn; and iii) a few antibodies showed high specificity for oligomers and fibrils but did not bind to monomers. These findings suggest that the great majority of α-syn aggregate-specific antibodies do not differentiate between oligomers and fibrils, thus highlighting the importance of exercising caution when interpreting results obtained using these antibodies. Our results also underscore the critical importance of the characterization and validation of antibodies before their use in mechanistic studies and as diagnostic tools or therapeutic agents. This will not only improve the quality and reproducibility of research and reduce costs but will also reduce the number of therapeutic antibody failures in the clinic.
Collapse
Affiliation(s)
- Senthil T Kumar
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, EPFL, Switzerland
| | - Somanath Jagannath
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, EPFL, Switzerland
| | | | - Hugo Vanderstichele
- ADx NeuroSciences, Technologiepark 94, Ghent, Belgium; Biomarkable, Gent, Belgium
| | - Erik Stoops
- ADx NeuroSciences, Technologiepark 94, Ghent, Belgium
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, EPFL, Switzerland.
| |
Collapse
|
59
|
Investigating the effect of sugar-terminated nanoparticles on amyloid fibrillogenesis of β-lactoglobulin. Int J Biol Macromol 2020; 165:291-307. [PMID: 32961178 DOI: 10.1016/j.ijbiomac.2020.09.104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 08/19/2020] [Accepted: 09/14/2020] [Indexed: 11/22/2022]
Abstract
In vivo tissue deposition of fibrillar protein aggregates is the cause of several degenerative diseases. Evidence suggests that interfering with the pathology-associated amyloid fibrillogenesis by inhibitory molecules is envisaged as the primary therapeutic strategy. Amyloid fibril formation of proteins has been demonstrated to be influenced by nanoparticles/nanomaterials. As compared with their molecular form counterpart, this work examined the effect of sucrose-terminated nanoparticles on the in vitro amyloid fibrillogenesis and structural properties of β-lactoglobulin at pH 2.0 and 80 °C. ThT binding and electron microscopy results demonstrated that sucrose-terminated nanoparticles were able to suppress β-lactoglobulin fibrillogenesis in a concentration-dependent fashion. Importantly, sucrose-terminated nanoparticles showed better β-lactoglobulin fibril-inhibiting ability than sucrose molecules. ANS fluorescence and right-angle light scattering results showed reduced solvent exposure and decreased aggregation, respectively, in the β-lactoglobulin samples upon treatment with sucrose-terminated nanoparticles. Moreover, fluorescence quenching analyses revealed that the static quenching mechanism and formation of a non-fluorescent fluorophore-nanoparticle complex are involved in the nanoparticle-β-lactoglobulin interaction. We believe that the results from this study may suggest that the nanoparticle form of biocompatible sugar-related osmolytes may serve as effective inhibiting/suppressing agents toward protein fibrillogenesis.
Collapse
|
60
|
Landeck N, Strathearn KE, Ysselstein D, Buck K, Dutta S, Banerjee S, Lv Z, Hulleman JD, Hindupur J, Lin LK, Padalkar S, Stanciu LA, Lyubchenko YL, Kirik D, Rochet JC. Two C-terminal sequence variations determine differential neurotoxicity between human and mouse α-synuclein. Mol Neurodegener 2020; 15:49. [PMID: 32900375 PMCID: PMC7487555 DOI: 10.1186/s13024-020-00380-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND α-Synuclein (aSyn) aggregation is thought to play a central role in neurodegenerative disorders termed synucleinopathies, including Parkinson's disease (PD). Mouse aSyn contains a threonine residue at position 53 that mimics the human familial PD substitution A53T, yet in contrast to A53T patients, mice show no evidence of aSyn neuropathology even after aging. Here, we studied the neurotoxicity of human A53T, mouse aSyn, and various human-mouse chimeras in cellular and in vivo models, as well as their biochemical properties relevant to aSyn pathobiology. METHODS Primary midbrain cultures transduced with aSyn-encoding adenoviruses were analyzed immunocytochemically to determine relative dopaminergic neuron viability. Brain sections prepared from rats injected intranigrally with aSyn-encoding adeno-associated viruses were analyzed immunohistochemically to determine nigral dopaminergic neuron viability and striatal dopaminergic terminal density. Recombinant aSyn variants were characterized in terms of fibrillization rates by measuring thioflavin T fluorescence, fibril morphologies via electron microscopy and atomic force microscopy, and protein-lipid interactions by monitoring membrane-induced aSyn aggregation and aSyn-mediated vesicle disruption. Statistical tests consisted of ANOVA followed by Tukey's multiple comparisons post hoc test and the Kruskal-Wallis test followed by a Dunn's multiple comparisons test or a two-tailed Mann-Whitney test. RESULTS Mouse aSyn was less neurotoxic than human aSyn A53T in cell culture and in rat midbrain, and data obtained for the chimeric variants indicated that the human-to-mouse substitutions D121G and N122S were at least partially responsible for this decrease in neurotoxicity. Human aSyn A53T and a chimeric variant with the human residues D and N at positions 121 and 122 (respectively) showed a greater propensity to undergo membrane-induced aggregation and to elicit vesicle disruption. Differences in neurotoxicity among the human, mouse, and chimeric aSyn variants correlated weakly with differences in fibrillization rate or fibril morphology. CONCLUSIONS Mouse aSyn is less neurotoxic than the human A53T variant as a result of inhibitory effects of two C-terminal amino acid substitutions on membrane-induced aSyn aggregation and aSyn-mediated vesicle permeabilization. Our findings highlight the importance of membrane-induced self-assembly in aSyn neurotoxicity and suggest that inhibiting this process by targeting the C-terminal domain could slow neurodegeneration in PD and other synucleinopathy disorders.
Collapse
Affiliation(s)
- Natalie Landeck
- Brain Repair and Imaging in Neural Systems, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Katherine E. Strathearn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN USA
- Present address: Fujifilm Irvine Scientific, Santa Ana, CA USA
| | - Daniel Ysselstein
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN USA
- Present address: Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Kerstin Buck
- Brain Repair and Imaging in Neural Systems, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Present address: AbbVie Deutschland GmbH & Co KG, Ludwigshafen, Germany
| | - Sayan Dutta
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN USA
| | - Siddhartha Banerjee
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE USA
| | - Zhengjian Lv
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE USA
- Present address: Bruker Nanosurfaces Division, Goleta, Santa Barbara, CA USA
| | - John D. Hulleman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN USA
- Present address: Departments of Ophthalmology and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Jagadish Hindupur
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN USA
- Present address: Liveon Biolabs Pvt. Ltd., Tumakuru, Karnataka India
| | - Li-Kai Lin
- School of Materials Engineering, Purdue University, West Lafayette, IN USA
| | - Sonal Padalkar
- School of Materials Engineering, Purdue University, West Lafayette, IN USA
- Present address: Department of Mechanical Engineering, Iowa State University, Ames, IA USA
| | - Lia A. Stanciu
- School of Materials Engineering, Purdue University, West Lafayette, IN USA
| | - Yuri L. Lyubchenko
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE USA
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN USA
| |
Collapse
|
61
|
Parres-Gold J, Chieng A, Su SW, Wang Y. Real-Time Characterization of Cell Membrane Disruption by α-Synuclein Oligomers in Live SH-SY5Y Neuroblastoma Cells. ACS Chem Neurosci 2020; 11:2528-2534. [PMID: 32786327 PMCID: PMC7484231 DOI: 10.1021/acschemneuro.0c00309] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aggregation of the natively unfolded protein α-synuclein (α-Syn) has been widely correlated to the neuronal death associated with Parkinson's disease. Mutations and protein overaccumulation can promote the aggregation of α-Syn into oligomers and fibrils. Recent work has suggested that α-Syn oligomers can permeabilize the neuronal membrane, promoting calcium influx and cell death. However, the mechanism of this permeabilization is still uncertain and has yet to be characterized in live cells. This work uses scanning ion conductance microscopy (SICM) to image, in real time and without using chemical probes, the topographies of live SH-SY5Y neuroblastoma cells after exposure to α-Syn oligomers. Substantial morphological changes were observed, with micrometer-scale hills and troughs observed at lower α-Syn concentrations (1.00 μM) and large, transient pores observed at higher α-Syn concentrations (6.0 μM). These findings suggest that α-Syn oligomers may permeabilize the neuronal membrane by destabilizing the lipid bilayer and opening transient pores.
Collapse
Affiliation(s)
- Jacob Parres-Gold
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, California 90032, United States
| | - Andy Chieng
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, California 90032, United States
| | - Stephanie Wong Su
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, California 90032, United States
| | - Yixian Wang
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, California 90032, United States
| |
Collapse
|
62
|
Proteotoxicity and Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21165646. [PMID: 32781742 PMCID: PMC7460676 DOI: 10.3390/ijms21165646] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/01/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases are a major burden for our society, affecting millions of people worldwide. A main goal of past and current research is to enhance our understanding of the mechanisms underlying proteotoxicity, a common theme among these incurable and debilitating conditions. Cell proteome alteration is considered to be one of the main driving forces that triggers neurodegeneration, and unraveling the biological complexity behind the affected molecular pathways constitutes a daunting challenge. This review summarizes the current state on key processes that lead to cellular proteotoxicity in Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, providing a comprehensive landscape of recent literature. A foundational understanding of how proteotoxicity affects disease etiology and progression may provide essential insight towards potential targets amenable of therapeutic intervention.
Collapse
|
63
|
Lee EY, Srinivasan Y, de Anda J, Nicastro LK, Tükel Ç, Wong GCL. Functional Reciprocity of Amyloids and Antimicrobial Peptides: Rethinking the Role of Supramolecular Assembly in Host Defense, Immune Activation, and Inflammation. Front Immunol 2020; 11:1629. [PMID: 32849553 PMCID: PMC7412598 DOI: 10.3389/fimmu.2020.01629] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
Pathological self-assembly is a concept that is classically associated with amyloids, such as amyloid-β (Aβ) in Alzheimer's disease and α-synuclein in Parkinson's disease. In prokaryotic organisms, amyloids are assembled extracellularly in a similar fashion to human amyloids. Pathogenicity of amyloids is attributed to their ability to transform into several distinct structural states that reflect their downstream biological consequences. While the oligomeric forms of amyloids are thought to be responsible for their cytotoxicity via membrane permeation, their fibrillar conformations are known to interact with the innate immune system to induce inflammation. Furthermore, both eukaryotic and prokaryotic amyloids can self-assemble into molecular chaperones to bind nucleic acids, enabling amplification of Toll-like receptor (TLR) signaling. Recent work has shown that antimicrobial peptides (AMPs) follow a strikingly similar paradigm. Previously, AMPs were thought of as peptides with the primary function of permeating microbial membranes. Consistent with this, many AMPs are facially amphiphilic and can facilitate membrane remodeling processes such as pore formation and fusion. We show that various AMPs and chemokines can also chaperone and organize immune ligands into amyloid-like ordered supramolecular structures that are geometrically optimized for binding to TLRs, thereby amplifying immune signaling. The ability of amphiphilic AMPs to self-assemble cooperatively into superhelical protofibrils that form structural scaffolds for the ordered presentation of immune ligands like DNA and dsRNA is central to inflammation. It is interesting to explore the notion that the assembly of AMP protofibrils may be analogous to that of amyloid aggregates. Coming full circle, recent work has suggested that Aβ and other amyloids also have AMP-like antimicrobial functions. The emerging perspective is one in which assembly affords a more finely calibrated system of recognition and response: the detection of single immune ligands, immune ligands bound to AMPs, and immune ligands spatially organized to varying degrees by AMPs, result in different immunologic outcomes. In this framework, not all ordered structures generated during multi-stepped AMP (or amyloid) assembly are pathological in origin. Supramolecular structures formed during this process serve as signatures to the innate immune system to orchestrate immune amplification in a proportional, situation-dependent manner.
Collapse
Affiliation(s)
- Ernest Y Lee
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States.,UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yashes Srinivasan
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jaime de Anda
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lauren K Nicastro
- Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Çagla Tükel
- Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Gerard C L Wong
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, United States.,California Nano Systems Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
64
|
Terry-Kantor E, Tripathi A, Imberdis T, LaVoie ZM, Ho GPH, Selkoe D, Fanning S, Ramalingam N, Dettmer U. Rapid Alpha-Synuclein Toxicity in a Neural Cell Model and Its Rescue by a Stearoyl-CoA Desaturase Inhibitor. Int J Mol Sci 2020; 21:E5193. [PMID: 32707907 PMCID: PMC7432784 DOI: 10.3390/ijms21155193] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 01/28/2023] Open
Abstract
Genetic and biochemical evidence attributes neuronal loss in Parkinson's disease (PD) and related brain diseases to dyshomeostasis of the 14 kDa protein α-synuclein (αS). There is no consensus on how αS exerts toxicity. Explanations range from disturbed vesicle biology to proteotoxicity caused by fibrillar aggregates. To probe these mechanisms further, robust cellular toxicity models are needed, but their availability is limited. We previously reported that a shift from dynamic multimers to monomers is an early event in αS dyshomeostasis, as caused by familial PD (fPD)-linked mutants such as E46K. Excess monomers accumulate in round, lipid-rich inclusions. Engineered αS '3K' (E35K+E46K+E61K) amplifies E46K, causing a PD-like, L-DOPA-responsive motor phenotype in transgenic mice. Here, we present a cellular model of αS neurotoxicity after transducing human neuroblastoma cells to express yellow fluorescent protein (YFP)-tagged αS 3K in a doxycycline-dependent manner. αS-3K::YFP induction causes pronounced growth defects that accord with cell death. We tested candidate compounds for their ability to restore growth, and stearoyl-CoA desaturase (SCD) inhibitors emerged as a molecule class with growth-restoring capacity, but the therapeutic window varied among compounds. The SCD inhibitor MF-438 fully restored growth while exerting no apparent cytotoxicity. Our αS bioassay will be useful for elucidating compound mechanisms, for pharmacokinetic studies, and for compound/genetic screens.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (E.T.-K.); (A.T.); (T.I.); (Z.M.L.); (G.P.H.H.); (D.S.); (S.F.)
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (E.T.-K.); (A.T.); (T.I.); (Z.M.L.); (G.P.H.H.); (D.S.); (S.F.)
| |
Collapse
|
65
|
Single-vesicle imaging reveals lipid-selective and stepwise membrane disruption by monomeric α-synuclein. Proc Natl Acad Sci U S A 2020; 117:14178-14186. [PMID: 32513706 PMCID: PMC7322013 DOI: 10.1073/pnas.1914670117] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neurodegenerative diseases are increasing among the world's population, but there are no cures. These disorders all involve proteins that assemble into amyloid fibers which results in brain cell death. Evidence suggests that association of these proteins with lipid membranes is crucial for both functional and pathological roles. In Parkinson's disease, the involved protein, α-synuclein, is thought to function in trafficking of lipid vesicles in the brain. In search of mechanistic origins, increasing focus is put on identifying neurotoxic reactions induced by membrane interactions. To contribute new clues to this question, we here employed a new surface-sensitive scattering microscopy technique. With this approach, we discovered that α-synuclein perturbs vesicles in a stepwise and lipid-dependent fashion already at very low protein coverage. The interaction of the neuronal protein α-synuclein with lipid membranes appears crucial in the context of Parkinson’s disease, but the underlying mechanistic details, including the roles of different lipids in pathogenic protein aggregation and membrane disruption, remain elusive. Here, we used single-vesicle resolution fluorescence and label-free scattering microscopy to investigate the interaction kinetics of monomeric α-synuclein with surface-tethered vesicles composed of different negatively charged lipids. Supported by a theoretical model to account for structural changes in scattering properties of surface-tethered lipid vesicles, the data demonstrate stepwise vesicle disruption and asymmetric membrane deformation upon α-synuclein binding to phosphatidylglycerol vesicles at protein concentrations down to 10 nM (∼100 proteins per vesicle). In contrast, phosphatidylserine vesicles were only marginally affected. These insights into structural consequences of α-synuclein interaction with lipid vesicles highlight the contrasting roles of different anionic lipids, which may be of mechanistic relevance for both normal protein function (e.g., synaptic vesicle binding) and dysfunction (e.g., mitochondrial membrane interaction).
Collapse
|
66
|
Advances in modelling alpha-synuclein-induced Parkinson’s diseases in rodents: Virus-based models versus inoculation of exogenous preformed toxic species. J Neurosci Methods 2020; 338:108685. [DOI: 10.1016/j.jneumeth.2020.108685] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 11/22/2022]
|
67
|
Sanderson JB, De S, Jiang H, Rovere M, Jin M, Zaccagnini L, Hays Watson A, De Boni L, Lagomarsino VN, Young-Pearse TL, Liu X, Pochapsky TC, Hyman BT, Dickson DW, Klenerman D, Selkoe DJ, Bartels T. Analysis of α-synuclein species enriched from cerebral cortex of humans with sporadic dementia with Lewy bodies. Brain Commun 2020; 2:fcaa010. [PMID: 32280944 PMCID: PMC7130446 DOI: 10.1093/braincomms/fcaa010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/23/2019] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Since researchers identified α-synuclein as the principal component of Lewy bodies and Lewy neurites, studies have suggested that it plays a causative role in the pathogenesis of dementia with Lewy bodies and other 'synucleinopathies'. While α-synuclein dyshomeostasis likely contributes to the neurodegeneration associated with the synucleinopathies, few direct biochemical analyses of α-synuclein from diseased human brain tissue currently exist. In this study, we analysed sequential protein extracts from a substantial number of patients with neuropathological diagnoses of dementia with Lewy bodies and corresponding controls, detecting a shift of cytosolic and membrane-bound physiological α-synuclein to highly aggregated forms. We then fractionated aqueous extracts (cytosol) from cerebral cortex using non-denaturing methods to search for soluble, disease-associated high molecular weight species potentially associated with toxicity. We applied these fractions and corresponding insoluble fractions containing Lewy-type aggregates to several reporter assays to determine their bioactivity and cytotoxicity. Ultimately, high molecular weight cytosolic fractions enhances phospholipid membrane permeability, while insoluble, Lewy-associated fractions induced morphological changes in the neurites of human stem cell-derived neurons. While the concentrations of soluble, high molecular weight α-synuclein were only slightly elevated in brains of dementia with Lewy bodies patients compared to healthy, age-matched controls, these observations suggest that a small subset of soluble α-synuclein aggregates in the brain may drive early pathogenic effects, while Lewy body-associated α-synuclein can drive neurotoxicity.
Collapse
Affiliation(s)
- John B Sanderson
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Suman De
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute, Department of Chemistry, University of Cambridge, Cambridge CB2 0AH, UK
| | - Haiyang Jiang
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Matteo Rovere
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ming Jin
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ludovica Zaccagnini
- UK Dementia Research Institute, Department of Neurology, University College London, London WC1E 6BT, UK
| | - Aurelia Hays Watson
- UK Dementia Research Institute, Department of Neurology, University College London, London WC1E 6BT, UK
| | - Laura De Boni
- UK Dementia Research Institute, Department of Neurology, University College London, London WC1E 6BT, UK
| | - Valentina N Lagomarsino
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xinyue Liu
- Department of Chemistry, Rosenstiel Institute for Basic Biomedical Research, Brandeis University, Waltham, MA 02453, USA
| | - Thomas C Pochapsky
- Department of Chemistry, Rosenstiel Institute for Basic Biomedical Research, Brandeis University, Waltham, MA 02453, USA
| | - Bradley T Hyman
- Massachusetts General Hospital, Harvard Medical School, Department of Neurology, Massachusetts Institute for Neurodegenerative Disease, Boston, MA 02129, USA
| | - Dennis W Dickson
- Neuropathology Laboratory, Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL 32224, USA
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute, Department of Chemistry, University of Cambridge, Cambridge CB2 0AH, UK
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tim Bartels
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- UK Dementia Research Institute, Department of Neurology, University College London, London WC1E 6BT, UK
| |
Collapse
|
68
|
Brás IC, Xylaki M, Outeiro TF. Mechanisms of alpha-synuclein toxicity: An update and outlook. PROGRESS IN BRAIN RESEARCH 2019; 252:91-129. [PMID: 32247376 DOI: 10.1016/bs.pbr.2019.10.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Alpha-synuclein (aSyn) was identified as the main component of inclusions that define synucleinopathies more than 20 years ago. Since then, aSyn has been extensively studied in an attempt to unravel its roles in both physiology and pathology. Today, studying the mechanisms of aSyn toxicity remains in the limelight, leading to the identification of novel pathways involved in pathogenesis. In this chapter, we address the molecular mechanisms involved in synucleinopathies, from aSyn misfolding and aggregation to the various cellular effects and pathologies associated. In particular, we review our current understanding of the mechanisms involved in the spreading of aSyn between different cells, from the periphery to the brain, and back. Finally, we also review recent studies on the contribution of inflammation and the gut microbiota to pathology in synucleinopathies. Despite significant advances in our understanding of the molecular mechanisms involved, we still lack an integrated understanding of the pathways leading to neurodegeneration in PD and other synucleinopathies, compromising our ability to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Inês Caldeira Brás
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Mary Xylaki
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany; Max Planck Institute for Experimental Medicine, Göttingen, Germany; Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
69
|
Bellomo G, Paciotti S, Gatticchi L, Parnetti L. The Vicious Cycle Between
α
‐Synuclein Aggregation and Autophagic‐Lysosomal Dysfunction. Mov Disord 2019; 35:34-44. [DOI: 10.1002/mds.27895] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/31/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Giovanni Bellomo
- Magnetic Resonance Center (CERM) University of Florence Sesto Fiorentino (FI) Italy
| | - Silvia Paciotti
- Laboratory of Clinical Neurochemistry, Section of Neurology University of Perugia Perugia (PG) Italy
- Department of Experimental Medicine University of Perugia Perugia (PG) Italy
| | - Leonardo Gatticchi
- Department of Experimental Medicine University of Perugia Perugia (PG) Italy
| | - Lucilla Parnetti
- Laboratory of Clinical Neurochemistry, Section of Neurology University of Perugia Perugia (PG) Italy
| |
Collapse
|
70
|
Vincent BM, Tardiff DF, Piotrowski JS, Aron R, Lucas MC, Chung CY, Bacherman H, Chen Y, Pires M, Subramaniam R, Doshi DB, Sadlish H, Raja WK, Solís EJ, Khurana V, Le Bourdonnec B, Scannevin RH, Rhodes KJ. Inhibiting Stearoyl-CoA Desaturase Ameliorates α-Synuclein Cytotoxicity. Cell Rep 2019; 25:2742-2754.e31. [PMID: 30517862 DOI: 10.1016/j.celrep.2018.11.028] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/19/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
The lack of disease-modifying treatments for neurodegenerative disease stems in part from our rudimentary understanding of disease mechanisms and the paucity of targets for therapeutic intervention. Here we used an integrated discovery paradigm to identify a new therapeutic target for diseases caused by α-synuclein (α-syn), a small lipid-binding protein that misfolds and aggregates in Parkinson's disease and other disorders. Using unbiased phenotypic screening, we identified a series of compounds that were cytoprotective against α-syn-mediated toxicity by inhibiting the highly conserved enzyme stearoyl-CoA desaturase (SCD). Critically, reducing the levels of unsaturated membrane lipids by inhibiting SCD reduced α-syn toxicity in human induced pluripotent stem cell (iPSC) neuronal models. Taken together, these findings suggest that inhibition of fatty acid desaturation has potential as a therapeutic approach for the treatment of Parkinson's disease and other synucleinopathies.
Collapse
Affiliation(s)
- Benjamin M Vincent
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Daniel F Tardiff
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA.
| | - Jeff S Piotrowski
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Rebecca Aron
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Matthew C Lucas
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Chee Yeun Chung
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Helene Bacherman
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - YiQun Chen
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Michelle Pires
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Radha Subramaniam
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Dimple B Doshi
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Heather Sadlish
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Waseem K Raja
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Eric J Solís
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Vikram Khurana
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA; Ann Romney Center for Neurologic Disease, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Robert H Scannevin
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| | - Kenneth J Rhodes
- Yumanity Therapeutics, 790 Memorial Drive, Suite 2C, Cambridge, MA 02139, USA
| |
Collapse
|
71
|
α-Synuclein 2.0 — Moving towards Cell Type Specific Pathophysiology. Neuroscience 2019; 412:248-256. [DOI: 10.1016/j.neuroscience.2019.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/17/2022]
|
72
|
α-synuclein-lipoprotein interactions and elevated ApoE level in cerebrospinal fluid from Parkinson's disease patients. Proc Natl Acad Sci U S A 2019; 116:15226-15235. [PMID: 31270237 PMCID: PMC6660770 DOI: 10.1073/pnas.1821409116] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Two of the most important issues in Parkinson’s disease (PD) research are the identification of mechanisms underlying α-synuclein cell-to-cell transfer in the nervous system and the discovery of early diagnostic biomarkers. Both of these issues are addressed in our current manuscript. Using multiple approaches, we present that α-synuclein interacts with lipoproteins within human cerebrospinal fluid and can be taken up by cells in such a state. Moreover, using cerebrospinal fluid samples from 3 large and independent cohorts of patients, we demonstrate that apolipoprotein E is elevated in early, not yet medicated, patients with PD. Finally, using postmortem brain tissue, we provide preliminary histological evidence that apolipoprotein E is enriched in a subpopulation of dopaminergic neurons of human substantia nigra. The progressive accumulation, aggregation, and spread of α-synuclein (αSN) are common hallmarks of Parkinson’s disease (PD) pathology. Moreover, numerous proteins interact with αSN species, influencing its toxicity in the brain. In the present study, we extended analyses of αSN-interacting proteins to cerebrospinal fluid (CSF). Using coimmunoprecipitation, followed by mass spectrometry, we found that αSN colocalize with apolipoproteins on lipoprotein vesicles. We confirmed these interactions using several methods, including the enrichment of lipoproteins with a recombinant αSN, and the subsequent uptake of prepared vesicles by human dopaminergic neuronal-like cells. Further, we report an increased level of ApoE in CSF from early PD patients compared with matched controls in 3 independent cohorts. Moreover, in contrast to controls, we observed the presence of ApoE-positive neuromelanin-containing dopaminergic neurons in substantia nigra of PD patients. In conclusion, the cooccurrence of αSN on lipoprotein vesicles, and their uptake by dopaminergic neurons along with an increase of ApoE in early PD, proposes a mechanism(s) for αSN spreading in the extracellular milieu of PD.
Collapse
|
73
|
Limphaibool N, Iwanowski P, Holstad MJV, Kobylarek D, Kozubski W. Infectious Etiologies of Parkinsonism: Pathomechanisms and Clinical Implications. Front Neurol 2019; 10:652. [PMID: 31275235 PMCID: PMC6593078 DOI: 10.3389/fneur.2019.00652] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
Extensive research in recent decades has expanded our insights into the pathogenesis of Parkinson's disease (PD), though the underlying cause remains incompletely understood. Neuroinflammation have become a point of interest in the interconnecting areas of neurodegeneration and infectious diseases. The hypothesis concerning an infectious origin in PD stems from the observation of Parkinson-like symptoms in individuals infected with the influenza virus who then developed encephalitis lethargica. The implications of infectious pathogens have later been studied in neuronal pathways leading to the development of Parkinsonism and PD, through both a direct association and through synergistic effects of infectious pathogens in inducing neuroinflammation. This review explores the relationship between important infectious pathogens and Parkinsonism, including symptoms of Parkinsonism following infectious etiologies, infectious contributions to neuroinflammation and neurodegenerative processes associated with Parkinsonism, and the epidemiologic correlations between infectious pathogens and idiopathic PD.
Collapse
Affiliation(s)
| | - Piotr Iwanowski
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Dominik Kobylarek
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
74
|
Sanyal A, Dutta S, Camara A, Chandran A, Koller A, Watson BG, Sengupta R, Ysselstein D, Montenegro P, Cannon J, Rochet JC, Mattoo S. Alpha-Synuclein Is a Target of Fic-Mediated Adenylylation/AMPylation: Possible Implications for Parkinson's Disease. J Mol Biol 2019; 431:2266-2282. [PMID: 31034889 PMCID: PMC6554060 DOI: 10.1016/j.jmb.2019.04.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 01/17/2023]
Abstract
During disease, cells experience various stresses that manifest as an accumulation of misfolded proteins and eventually lead to cell death. To combat this stress, cells activate a pathway called unfolded protein response that functions to maintain endoplasmic reticulum (ER) homeostasis and determines cell fate. We recently reported a hitherto unknown mechanism of regulating ER stress via a novel post-translational modification called Fic-mediatedadenylylation/AMPylation. Specifically, we showed that the human Fic (filamentation induced by cAMP) protein, HYPE/FicD, catalyzes the addition of an adenosine monophosphate (AMP) to the ER chaperone, BiP, to alter the cell's unfolded protein response-mediated response to misfolded proteins. Here, we report that we have now identified a second target for HYPE-alpha-synuclein (αSyn), a presynaptic protein involved in Parkinson's disease. Aggregated αSyn has been shown to induce ER stress and elicit neurotoxicity in Parkinson's disease models. We show that HYPE adenylylates αSyn and reduces phenotypes associated with αSyn aggregation invitro, suggesting a possible mechanism by which cells cope with αSyn toxicity.
Collapse
Affiliation(s)
- Anwesha Sanyal
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Sayan Dutta
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Ali Camara
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Aswathy Chandran
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Antonius Koller
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Ben G Watson
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Ranjan Sengupta
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Daniel Ysselstein
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Paola Montenegro
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Jason Cannon
- School of Health Sciences, Purdue University, 915 W State St., LILYG-227, West Lafayette, IN 47907, USA
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, 915 W State St., LILYG-227, West Lafayette, IN 47907, USA
| | - Seema Mattoo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Inflammation, Immunology and Infectious Disease, Purdue University, 915 W State St., LILYG-227, West Lafayette, IN 47907, USA.
| |
Collapse
|
75
|
Khalifeh M, Barreto GE, Sahebkar A. Trehalose as a promising therapeutic candidate for the treatment of Parkinson's disease. Br J Pharmacol 2019; 176:1173-1189. [PMID: 30767205 PMCID: PMC6468260 DOI: 10.1111/bph.14623] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/13/2018] [Accepted: 01/29/2019] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a progressive movement disorder resulting primarily from loss of nigrostriatal dopaminergic neurons. PD is characterized by the accumulation of protein aggregates, and evidence suggests that aberrant protein deposition in dopaminergic neurons could be related to the dysregulation of the lysosomal autophagy pathway. The therapeutic potential of autophagy modulators has been reported in experimental models of PD. Trehalose is a natural disaccharide that has been considered as a new candidate for the treatment of neurodegenerative diseases. It has a chaperone-like activity, prevents protein misfolding or aggregation, and by promoting autophagy, contributes to the removal of accumulated proteins. In this review, we briefly summarize the role of aberrant autophagy in PD and the underlying mechanisms that lead to the development of this disease. We also discuss reports that used trehalose to counteract the neurotoxicity in PD, focusing particularly on the autophagy promoting, protein stabilization, and anti-neuroinflammatory effects of trehalose.
Collapse
Affiliation(s)
- Masoomeh Khalifeh
- Department of Medical Biotechnology, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - George E. Barreto
- Departamento de Nutrición y Bioquímica, Facultad de CienciasPontificia Universidad JaverianaBogotáColombia
- Instituto de Ciencias BiomédicasUniversidad Autónoma de ChileSantiagoChile
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research CenterMashhad University of Medical SciencesMashhadIran
- Biotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- School of PharmacyMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
76
|
Li X, Tong X, Lu W, Yu D, Diao J, Zhao Q. Label-free detection of early oligomerization of α-synuclein and its mutants A30P/E46K through solid-state nanopores. NANOSCALE 2019; 11:6480-6488. [PMID: 30892349 DOI: 10.1039/c9nr00023b] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
A30P and E46K are two mutants of α-synuclein (α-Syn) associated with familial early-onset Parkinson's disease (PD), and amyloid fibrils of α-Syn are the hallmarks of this disease. Detecting the heterogeneous system in the oligomerization stage of α-Syn is crucial for understanding the fibril formation and in vivo toxicity of α-Syn oligomers. Over the last two decades, solid-state nanopore technology has been developed into a reliable and versatile method in single-molecule studies. In this work, we study the time-dependent kinetics of early oligomerization of wild-type α-Syn, A30P, and E46K mutants through silicon nitride solid-state nanopores. By testing A30P, E46K, and wild-type α-Syn samples with different incubation times-from 3 to 15 days-we identify three typical types of oligomers formed in the oligomerization stage and confirm that A30P and E46K mutants aggregate faster than wild-type α-Syn. The results imply that the distinct aggregation pathways and kinetics featured by wild-type α-Syn and mutations may account for their distinct cytotoxicity and pathology in PD-related studies.
Collapse
Affiliation(s)
- Xiaoqing Li
- State Key Laboratory for Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, China.
| | | | | | | | | | | |
Collapse
|
77
|
Belfiore M, Cariati I, Matteucci A, Gaddini L, Macchia G, Fioravanti R, Frank C, Tancredi V, D'Arcangelo G, Diociaiuti M. Calcitonin native prefibrillar oligomers but not monomers induce membrane damage that triggers NMDA-mediated Ca 2+-influx, LTP impairment and neurotoxicity. Sci Rep 2019; 9:5144. [PMID: 30914688 PMCID: PMC6435710 DOI: 10.1038/s41598-019-41462-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/08/2019] [Indexed: 01/20/2023] Open
Abstract
Amyloid protein misfolding results in a self-assembling aggregation process, characterized by the formation of typical aggregates. The attention is focused on pre-fibrillar oligomers (PFOs), formed in the early stages and supposed to be neurotoxic. PFOs structure may change due to their instability and different experimental protocols. Consequently, it is difficult to ascertain which aggregation species are actually neurotoxic. We used salmon Calcitonin (sCT) as an amyloid model whose slow aggregation rate allowed to prepare stable samples without photochemical cross-linking. Intracellular Ca2+ rise plays a fundamental role in amyloid protein-induced neurodegerations. Two paradigms have been explored: (i) the "membrane permeabilization" due to the formation of amyloid pores or other types of membrane damage; (ii) "receptor-mediated" modulation of Ca2+ channels. In the present paper, we tested the effects of native sCT PFOs- with respect to Monomer-enriched solutions in neurons characterized by an increasing degree of differentiation, in terms of -Ca2+-influx, cellular viability, -Long-Term Potentiation impairment, Post-Synaptic Densities and synaptophysin expression. Results indicated that PFOs-, but not Monomer-enriched solutions, induced abnormal -Ca2+-influx, which could only in part be ascribed to NMDAR activation. Thus, we propose an innovative neurotoxicity mechanism for amyloid proteins where "membrane permeabilization" and "receptor-mediated" paradigms coexist.
Collapse
Affiliation(s)
- Marcello Belfiore
- National Center for Rare Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Matteucci
- National Center for Drug Research and Evalutation, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Gaddini
- National Center for Drug Research and Evalutation, Istituto Superiore di Sanità, Rome, Italy
| | | | - Raoul Fioravanti
- National Center for Rare Diseases, Istituto Superiore di Sanità, Rome, Italy.,Chemistry Department, University "Sapienza", Rome, Italy
| | - Claudio Frank
- National Center for Rare Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Marco Diociaiuti
- National Center for Rare Diseases, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
78
|
Fanni AM, Monge FA, Lin CY, Thapa A, Bhaskar K, Whitten DG, Chi EY. High Selectivity and Sensitivity of Oligomeric p-Phenylene Ethynylenes for Detecting Fibrillar and Prefibrillar Amyloid Protein Aggregates. ACS Chem Neurosci 2019; 10:1813-1825. [PMID: 30657326 DOI: 10.1021/acschemneuro.8b00719] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Misfolding and aggregation of amyloid proteins into fibrillar aggregates is a central pathogenic event in neurodegenerative disorders such as Alzheimer's (AD) and Parkinson's diseases (PD). Currently, there is a lack of reliable sensors for detecting the range of protein aggregates involved in disease etiology, particularly the prefibrillar aggregate conformations that are more neurotoxic. In this study, the fluorescent sensing of two novel oligomeric p-phenylene ethynylenes (OPEs), anionic OPE1- and cationic OPE2+, for detecting prefibrillar and fibrillar aggregates of AD-associated amyloid-β (Aβ40 and Aβ42) and PD-associated α-synuclein proteins (wildtype, and single mutants A30P, E35K, and A53T) over their monomeric counterparts, were tested. Furthermore, the performance of OPEs was evaluated and compared to thioflavin T (ThT), the most widely used fibril dye. Our results show that OPE1- and OPE2+ exhibited aggregate-specific binding inducing large fluorescence turn-on and spectral shifts based on a combination of backbone planarization, hydrophobic unquenching, and superluminescent OPE complex formation sensing modes. OPEs exhibited higher selectivity, higher binding affinity, and comparable limits of detection for Aβ40 fibrils compared to ThT. OPE2+ exhibited the largest fluorescence turn-on and highest sensitivity. Significantly, OPEs detected prefibrillar aggregates of Aβ42 and α-synuclein that ThT failed to detect. The superior sensing performance, the nonprotein specific detection, and the ability to selectively detect fibrillar and prefibrillar amyloid protein aggregates point to the potential of OPEs to overcome the limitations of existing probes and promise significant advancement in the detection of the myriad of protein aggregates involved in the early stages of AD and PD.
Collapse
|
79
|
Patel D, Xu C, Nagarajan S, Liu Z, Hemphill WO, Shi R, Uversky VN, Caldwell GA, Caldwell KA, Witt SN. Alpha-synuclein inhibits Snx3-retromer-mediated retrograde recycling of iron transporters in S. cerevisiae and C. elegans models of Parkinson's disease. Hum Mol Genet 2019; 27:1514-1532. [PMID: 29452354 DOI: 10.1093/hmg/ddy059] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/12/2018] [Indexed: 01/31/2023] Open
Abstract
We probed the role of alpha-synuclein (α-syn) in modulating sorting nexin 3 (Snx3)-retromer-mediated recycling of iron transporters in Saccharomyces cerevisiae and Caenorhabditis elegans. In yeast, the membrane-bound heterodimer Fet3/Ftr1 is the high affinity iron importer. Fet3 is a membrane-bound multicopper ferroxidase, whose ferroxidase domain is orthologous to human ceruloplasmin (Cp), that oxidizes external Fe+2 to Fe+3; the Fe+3 ions then channel through the Ftr1 permease into the cell. When the concentration of external iron is low (<1 µM), Fet3/Ftr1 is maintained on the plasma membrane by retrograde endocytic-recycling; whereas, when the concentration of external iron is high (>10 µM), Fet3/Ftr1 is endocytosed and shunted to the vacuole for degradation. We discovered that α-syn expression phenocopies the high iron condition: under the low iron condition (<1 µM), α-syn inhibits Snx3-retromer-mediated recycling of Fet3/Ftr1 and instead shunts Fet3/Ftr1 into the multivesicular body pathway to the vacuole. α-Syn inhibits recycling by blocking the association of Snx3-mCherry molecules with endocytic vesicles, possibly by interfering with the binding of Snx3 to phosphatidylinositol-3-monophosphate. In C. elegans, transgenic worms expressing α-syn exhibit an age-dependent degeneration of dopaminergic neurons that is partially rescued by the iron chelator desferoxamine. This implies that α-syn-expressing dopaminergic neurons are susceptible to changes in iron neurotoxicity with age, whereby excess iron enhances α-syn-induced neurodegeneration. In vivo genetic analysis indicates that α-syn dysregulates iron homeostasis in worm dopaminergic neurons, possibly by inhibiting SNX-3-mediated recycling of a membrane-bound ortholog of Cp (F21D5.3), the iron exporter ferroportin (FPN1.1), or both.
Collapse
Affiliation(s)
- Dhaval Patel
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Chuan Xu
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Sureshbabu Nagarajan
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Zhengchang Liu
- Department of Biological Sciences, The University of New Orleans, New Orleans, LA 70148, USA
| | - Wayne O Hemphill
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Runhua Shi
- Department of Medicine, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Stephan N Witt
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA.,Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| |
Collapse
|
80
|
Mor DE, Daniels MJ, Ischiropoulos H. The usual suspects, dopamine and alpha-synuclein, conspire to cause neurodegeneration. Mov Disord 2019; 34:167-179. [PMID: 30633814 PMCID: PMC6379109 DOI: 10.1002/mds.27607] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/15/2018] [Accepted: 12/03/2018] [Indexed: 01/07/2023] Open
Abstract
Parkinson's disease (PD) is primarily a movement disorder driven by the loss of dopamine-producing neurons in the substantia nigra (SN). Early identification of the oxidative properties of dopamine implicated it as a potential source of oxidative stress in PD, yet few studies have investigated dopamine neurotoxicity in vivo. The discovery of PD-causing mutations in α-synuclein and the presence of aggregated α-synuclein in the hallmark Lewy body pathology of PD revealed another important player. Despite extensive efforts, the precise role of α-synuclein aggregation in neurodegeneration remains unclear. We recently manipulated both dopamine levels and α-synuclein expression in aged mice and found that only the combination of these 2 factors caused progressive neurodegeneration of the SN and an associated motor deficit. Dopamine modified α-synuclein aggregation in the SN, resulting in greater abundance of α-synuclein oligomers and unique dopamine-induced oligomeric conformations. Furthermore, disruption of the dopamine-α-synuclein interaction rescued dopaminergic neurons from degeneration in transgenic Caenorhabditis elegans models. In this Perspective, we discuss these findings in the context of known α-synuclein and dopamine biology, review the evidence for α-synuclein oligomer toxicity and potential mechanisms, and discuss therapeutic implications. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Danielle E. Mor
- Lewis-Sigler Institute for Integrative Genomics and Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Malcolm J. Daniels
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Harry Ischiropoulos
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| |
Collapse
|
81
|
Transition metal complexes based aptamers as optical diagnostic tools for disease proteins and biomolecules. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2018.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
82
|
Chakraborty R, Sahoo S, Halder N, Rath H, Chattopadhyay K. Conformational-Switch Based Strategy Triggered by [18] π Heteroannulenes toward Reduction of Alpha Synuclein Oligomer Toxicity. ACS Chem Neurosci 2019; 10:573-587. [PMID: 30296047 DOI: 10.1021/acschemneuro.8b00436] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A water-soluble meso-carboxy aryl substituted [18] heteroannulene (porphyrin) and its Zn-complex have been found to be viable in targeting α-Syn aggregation at all its key microevents, namely, primary nucleation, fibril elongation, and secondary nucleation, by converting the highly heterogeneous and cytotoxic aggresome into a homogeneous population of minimally toxic off-pathway oligomers, that remained unexplored until recently. With the EC50 and dissociation constants in the low micromolar range, these heteroannulenes induce a switch in the secondary structure of toxic prefibrillar on-pathway oligomers of α-Syn, converting them into minimally toxic nonseeding off-pathway oligomers. The inhibition of the aggregation and the reduction of toxicity have been studied in vitro as well as inside neuroblastoma cells.
Collapse
Affiliation(s)
- Ritobrita Chakraborty
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Sumit Sahoo
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A/2B Raja S. C. Mullick Road, Kolkata 700032, India
| | - Nyancy Halder
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A/2B Raja S. C. Mullick Road, Kolkata 700032, India
| | - Harapriya Rath
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A/2B Raja S. C. Mullick Road, Kolkata 700032, India
| | - Krishnananda Chattopadhyay
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| |
Collapse
|
83
|
Sahoo A, Matysiak S. Computational insights into lipid assisted peptide misfolding and aggregation in neurodegeneration. Phys Chem Chem Phys 2019; 21:22679-22694. [DOI: 10.1039/c9cp02765c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
An overview of recent advances in computational investigation of peptide–lipid interactions in neurodegeneration – Alzheimer's, Parkinson's and Huntington's disease.
Collapse
Affiliation(s)
- Abhilash Sahoo
- Biophysics Program
- Institute of Physical Science and Technology
- University of Maryland
- College Park
- USA
| | - Silvina Matysiak
- Biophysics Program
- Institute of Physical Science and Technology
- University of Maryland
- College Park
- USA
| |
Collapse
|
84
|
Bacterial Amyloids: Biogenesis and Biomaterials. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1174:113-159. [DOI: 10.1007/978-981-13-9791-2_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
85
|
Aldehyde adducts inhibit 3,4-dihydroxyphenylacetaldehyde-induced α-synuclein aggregation and toxicity: Implication for Parkinson neuroprotective therapy. Eur J Pharmacol 2018; 845:65-73. [PMID: 30579934 DOI: 10.1016/j.ejphar.2018.12.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/21/2022]
Abstract
3,4-Dihydroxyphenylacetaldehyde (DOPAL), the monoamine oxidase (MAO) metabolite of dopamine, plays a role in pathogenesis of Parkinson disease, inducing α-synuclein aggregation. DOPAL generates discrete α-synuclein aggregates. Inhibiting this aggregation could provide therapy for slowing Parkinson disease progression. Primary and secondary amines form adducts with aldehydes. Rasagiline and aminoindan contain these amine groups. DOPAL-induced α-synuclein aggregates were resolved in the presence and absence of rasagiline or aminoindan using quantitative Western blotting. DOPAL levels in incubation mixtures, containing increased rasagiline or aminoindan concentrations, were determined by high pressure liquid chromatography (HPLC). Schiff base adducts between DOPAL and rasagiline or aminoindan were determined using mass spectrometry. A neuroprotective effect of rasagiline and aminoindan against DOPAL-induced toxicity was demonstrated using PC-12 cells. Rasagiline and aminoindan significantly reduced aggregation of α-synuclein of all sizes in test tube and PC-12 cells experiments. Dimethylaminoindan did not reduce aggregation. DOPAL levels in incubation mixtures were reduced with increasing rasagiline or aminoindan concentrations but not with dimethylaminoindan. Schiff base adducts between DOPAL and either rasagiline or aminoindan were demonstrated by mass spectrometry. A neuroprotective effect against DOPAL-induced toxicity in PC-12 cells was demonstrated for both rasagiline and aminoindan. Inhibiting DOPAL-induced α-synuclein aggregation through amine adducts provides a therapeutic approach for slowing Parkinson disease progression.
Collapse
|
86
|
Roy S, Bhat R. Suppression, disaggregation, and modulation of γ-Synuclein fibrillation pathway by green tea polyphenol EGCG. Protein Sci 2018; 28:382-402. [PMID: 30394624 DOI: 10.1002/pro.3549] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/31/2018] [Indexed: 12/24/2022]
Abstract
Oligomerization of γ-Synuclein is known to have implications for both neurodegeneration and cancer. Although it is known to co-exist with the fibrillar deposits of α-Synuclein (Lewy bodies), a hallmark in Parkinson's disease (PD), the effect of potential therapeutic modulators on the fibrillation pathway of γ-Syn remains unexplored. By a combined use of various biophysical tools and cytotoxicity assays we demonstrate that the flavonoid epigallocatechin-3-gallate (EGCG) significantly suppresses γ-Syn fibrillation by affecting its nucleation and binds with the unstructured, nucleus forming oligomers of γ-Syn to modulate the pathway to form α-helical containing higher-order oligomers (~158 kDa and ~ 670 kDa) that are SDS-resistant and conformationally restrained in nature. Seeding studies reveal that these oligomers although "on-pathway" in nature, are kinetically retarded and rate-limiting species that slows down fibril elongation. We observe that EGCG also disaggregates the protofibrils and mature γ-Syn fibrils into similar SDS-resistant oligomers. Steady-state and time-resolved fluorescence spectroscopy and isothermal titration calorimetry (ITC) reveal a weak non-covalent interaction between EGCG and γ-Syn with the dissociation constant in the mM range (Kd ~ 2-10 mM). Interestingly, while EGCG-generated oligomers completely rescue the breast cancer (MCF-7) cells from γ-Syn toxicity, it reduces the viability of neuroblastoma (SH-SY5Y) cells. However, the disaggregated oligomers of γ-Syn are more toxic than the disaggregated fibrils for MCF-7cells. These findings throw light on EGCG-mediated modulation of γ-Syn fibrillation and suggest that investigation on the effects of such modulators on γ-Syn fibrillation is critical in identifying effective therapeutic strategies using small molecule modulators of synucleopathies.
Collapse
Affiliation(s)
- Sneha Roy
- Biophysical Chemistry Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
| | - Rajiv Bhat
- Biophysical Chemistry Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
| |
Collapse
|
87
|
Iljina M, Dear AJ, Garcia GA, De S, Tosatto L, Flagmeier P, Whiten DR, Michaels TCT, Frenkel D, Dobson CM, Knowles TPJ, Klenerman D. Quantifying Co-Oligomer Formation by α-Synuclein. ACS NANO 2018; 12:10855-10866. [PMID: 30371053 PMCID: PMC6262461 DOI: 10.1021/acsnano.8b03575] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Small oligomers of the protein α-synuclein (αS) are highly cytotoxic species associated with Parkinson's disease (PD). In addition, αS can form co-aggregates with its mutational variants and with other proteins such as amyloid-β (Aβ) and tau, which are implicated in Alzheimer's disease. The processes of self-oligomerization and co-oligomerization of αS are, however, challenging to study quantitatively. Here, we have utilized single-molecule techniques to measure the equilibrium populations of oligomers formed in vitro by mixtures of wild-type αS with its mutational variants and with Aβ40, Aβ42, and a fragment of tau. Using a statistical mechanical model, we find that co-oligomer formation is generally more favorable than self-oligomer formation at equilibrium. Furthermore, self-oligomers more potently disrupt lipid membranes than do co-oligomers. However, this difference is sometimes outweighed by the greater formation propensity of co-oligomers when multiple proteins coexist. Our results suggest that co-oligomer formation may be important in PD and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Marija Iljina
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Alexander J. Dear
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
- Department
of Chemistry, Centre for Misfolding Diseases, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Gonzalo A. Garcia
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Suman De
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Laura Tosatto
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Patrick Flagmeier
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
- Department
of Chemistry, Centre for Misfolding Diseases, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Daniel R. Whiten
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Thomas C. T. Michaels
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Daan Frenkel
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Christopher M. Dobson
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
- Department
of Chemistry, Centre for Misfolding Diseases, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Tuomas P. J. Knowles
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
- Department
of Chemistry, Centre for Misfolding Diseases, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
- E-mail:
| | - David Klenerman
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
- UK
Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, United Kingdom
- E-mail:
| |
Collapse
|
88
|
Cheng A, Shinoda Y, Yamamoto T, Miyachi H, Fukunaga K. Development of FABP3 ligands that inhibit arachidonic acid-induced α-synuclein oligomerization. Brain Res 2018; 1707:190-197. [PMID: 30496735 DOI: 10.1016/j.brainres.2018.11.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 11/19/2018] [Accepted: 11/25/2018] [Indexed: 10/27/2022]
Abstract
In Parkinson's disease (PD), α-synuclein (αSyn) accumulation and inclusion triggers dopamine neuronal death and synapse dysfunction in vivo. We previously reported that fatty acid-binding protein 3 (FABP3) is highly expressed in the brain and accelerates αSyn oligomerization when cells are exposed to 1-Methyl-1,2,3,6-tetrahydropiridine (MPTP). Here, we demonstrate that αSyn oligomerization was markedly enhanced by co-overexpressing FABP3 in neuro-2A cells when cells were treated with arachidonic acid (AA). We developed FABP3 ligands, which bind to the fatty acid binding domain of FABP3, using an 8-Anilinonaphthalene-1-sulfonic acid (ANS) assay with a recombinant FABP3 protein. The prototype for the FABP4 ligand, BMS309403, has no affinity for FABP3. We developed more FABP3-specific ligands derived from the chemical structure of BMS309403. Like AA, ligands 1, 7, and 8 had a relatively high affinity for FAPB3 in the ANS assay. Then, we evaluated the inhibition of αSyn oligomerization in neuro-2A cells co-overexpressing FABP3 and αSyn. Importantly, AA treatments markedly enhanced αSyn oligomerization in the co-expressing cells. Ligands 1, 7, and 8 significantly reduced AA-induced αSyn oligomerization in neuro-2A cells. Taken together, our results indicate that FABP3 ligands that target FABP3 may be used as potential therapeutics that inhibit αSyn aggregation in vivo.
Collapse
Affiliation(s)
- An Cheng
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Tetsunori Yamamoto
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Hiroyuki Miyachi
- Lead Exploration Unit, Drug Discovery Initiative, The University of Tokyo, Tokyo 113-0033, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| |
Collapse
|
89
|
Brás IC, Lopes LV, Outeiro TF. Sensing α-Synuclein From the Outside via the Prion Protein: Implications for Neurodegeneration. Mov Disord 2018; 33:1675-1684. [DOI: 10.1002/mds.27478] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022] Open
Affiliation(s)
- Inês Caldeira Brás
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
| | - Luísa V. Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina; Universidade de Lisboa; Lisboa Portugal
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
- CEDOC, Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciências Médicas; Universidade Nova de Lisboa, Campo dos Mártires da Pátria; Lisboa Portugal
- Max Planck Institute for Experimental Medicine; Göttingen Germany
- Institute of Neuroscience, The Medical School; Newcastle University; Newcastle Upon Tyne UK
| |
Collapse
|
90
|
Gallea JI, Ambroggio EE, Vilcaes AA, James NG, Jameson DM, Celej MS. Amyloid oligomerization of the Parkinson's disease related protein α-synuclein impacts on its curvature-membrane sensitivity. J Neurochem 2018; 147:541-556. [PMID: 30142705 DOI: 10.1111/jnc.14573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/28/2018] [Accepted: 08/17/2018] [Indexed: 11/28/2022]
Abstract
The amyloid aggregation of the presynaptic protein α-synuclein (AS) is pathognomonic of Parkinson's disease and other neurodegenerative disorders. Physiologically, AS contributes to synaptic homeostasis by participating in vesicle maintenance, trafficking, and release. Its avidity for highly curved acidic membranes has been related to the distinct chemistry of the N-terminal amphipathic helix adopted upon binding to appropriated lipid interfaces. Pathologically, AS populate a myriad of toxic aggregates ranging from soluble oligomers to insoluble amyloid fibrils. Different gain-of-toxic function mechanisms are linked to prefibrillar oligomers which are considered as the most neurotoxic species. Here, we investigated if amyloid oligomerization could hamper AS function as a membrane curvature sensor. We used fluorescence correlation spectroscopy to quantitatively evaluate the interaction of oligomeric species, produced using a popular method based on lyophilization and rehydration, to lipid vesicles of different curvatures and compositions. We found that AS oligomerization has a profound impact on protein-lipid interaction, altering binding affinity and/or curvature sensitivity depending on membrane composition. Our work provides novel insights into how the formation of prefibrillar intermediate species could contribute to neurodegeneration due to a loss-of-function mechanism. OPEN PRACTICES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- José Ignacio Gallea
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC, CONICET and Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Ernesto E Ambroggio
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC, CONICET and Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Aldo Alejandro Vilcaes
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC, CONICET and Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Nicholas G James
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - David M Jameson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - María Soledad Celej
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC, CONICET and Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| |
Collapse
|
91
|
Impact of monomeric, oligomeric and fibrillar alpha-synuclein on astrocyte reactivity and toxicity to neurons. Biochem J 2018; 475:3153-3169. [PMID: 30185433 DOI: 10.1042/bcj20180297] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/11/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022]
Abstract
Synucleinopathies are a group of neurodegenerative disorders characterized by the presence of aggregated and fibrillar forms of alpha-synuclein (α-syn). Here, we analyze the effect of different species of α-syn, including monomeric, oligomeric and fibrillar forms of the protein, on rat astrocytes. Astrocytes treated with these distinct forms of α-syn showed an increase in long and thin processes and glial fibrillary acidic protein expression, indicating cell activation, high levels of intracellular oxidants and increased expression of cytokines. Moreover, astrocytes incubated with the different species induced hippocampal neuronal death in co-culture, and cytotoxicity was particularly enhanced by exposure to fibrillar α-syn. Further exploration of the mechanisms behind astrocyte activation and cytotoxicity revealed differences between the assessed α-syn species. Only oligomers induced mitochondrial dysfunction in astrocytes and significantly increased extracellular hydrogen peroxide production by these cells. Besides, TNF-α and IL-1β (interleukin 1β) expression presented different kinetics and levels depending on which species induced the response. Our data suggest that α-syn species (monomeric, oligomeric and fibrillar) induce astrocyte activation that can lead to neuronal death. Nevertheless, the tested α-syn species act through different preferential mechanisms and potency. All together these results help to understand the effect of α-syn species on astrocyte function and their potential impact on the pathogenesis of Parkinson's disease and related α-synucleinopathies.
Collapse
|
92
|
Iyer A, Claessens MMAE. Disruptive membrane interactions of alpha-synuclein aggregates. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1867:468-482. [PMID: 30315896 DOI: 10.1016/j.bbapap.2018.10.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/14/2018] [Accepted: 10/04/2018] [Indexed: 12/17/2022]
Abstract
Alpha synuclein (αS) is a ~14 kDa intrinsically disordered protein. Decades of research have increased our knowledge on αS yet its physiological function remains largely elusive. The conversion of monomeric αS into oligomers and amyloid fibrils is believed to play a central role of the pathology of Parkinson's disease (PD). It is becoming increasingly clear that the interactions of αS with cellular membranes are important for both αS's functional and pathogenic actions. Therefore, understanding interactions of αS with membranes seems critical to uncover functional or pathological mechanisms. This review summarizes our current knowledge of how physicochemical properties of phospholipid membranes affect the binding and aggregation of αS species and gives an overview of how post-translational modifications and point mutations in αS affect phospholipid membrane binding and protein aggregation. We discuss the disruptive effects resulting from the interaction of αS aggregate species with membranes and highlight current approaches and hypotheses that seek to understand the pathogenic and/or protective role of αS in PD.
Collapse
Affiliation(s)
- Aditya Iyer
- Membrane Enzymology Group, University of Groningen, Groningen 9747 AG, The Netherlands
| | | |
Collapse
|
93
|
Non-cell-autonomous actions of α-synuclein: Implications in glial synucleinopathies. Prog Neurobiol 2018; 169:158-171. [DOI: 10.1016/j.pneurobio.2018.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 07/24/2017] [Accepted: 06/30/2018] [Indexed: 01/11/2023]
|
94
|
Kurnik M, Sahin C, Andersen CB, Lorenzen N, Giehm L, Mohammad-Beigi H, Jessen CM, Pedersen JS, Christiansen G, Petersen SV, Staal R, Krishnamurthy G, Pitts K, Reinhart PH, Mulder FAA, Mente S, Hirst WD, Otzen DE. Potent α-Synuclein Aggregation Inhibitors, Identified by High-Throughput Screening, Mainly Target the Monomeric State. Cell Chem Biol 2018; 25:1389-1402.e9. [PMID: 30197194 DOI: 10.1016/j.chembiol.2018.08.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/12/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022]
Abstract
α-Synuclein (αSN) aggregation is central to the etiology of Parkinson's disease (PD). Large-scale screening of compounds to identify aggregation inhibitors is challenged by stochastic αSN aggregation and difficulties in detecting early-stage oligomers (αSOs). We developed a high-throughput screening assay combining SDS-stimulated αSN aggregation with FRET to reproducibly detect initial stages in αSN aggregation. We screened 746,000 compounds, leading to 58 hits that markedly inhibit αSN aggregation and reduce αSOs' membrane permeabilization activity. The most effective aggregation inhibitors were derivatives of (4-hydroxynaphthalen-1-yl)sulfonamide. They interacted strongly with the N-terminal part of monomeric αSN and reduced αSO-membrane interactions, possibly by affecting electrostatic interactions. Several compounds reduced αSO toxicity toward neuronal cell lines. The inhibitors introduced chemical modifications of αSN that were, however, not a prerequisite for inhibitory activity. We also identified several phenyl-benzoxazol compounds that promoted αSN aggregation (proaggregators). These compounds may be useful tools to modulate αSN aggregation in cellula.
Collapse
Affiliation(s)
- Martin Kurnik
- iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark
| | - Cagla Sahin
- iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus, Denmark
| | | | - Nikolai Lorenzen
- iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark
| | - Lise Giehm
- iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark
| | - Hossein Mohammad-Beigi
- iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark; Biotechnology Group, Faculty of Chemical Engineering, Tarbiat Modares University, P.O. Box 14115-143, Tehran, Iran
| | - Christian Moestrup Jessen
- iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark; Department of Chemistry, Aarhus University, Langelandsgade 140, 8000 Aarhus, Denmark
| | - Jan Skov Pedersen
- iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark; Department of Chemistry, Aarhus University, Langelandsgade 140, 8000 Aarhus, Denmark
| | | | | | | | | | - Keith Pitts
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Peter H Reinhart
- Forma Therapeutics, Inc.Institute for Applied Life Sciences, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, MA 01003-9364, USA
| | - Frans A A Mulder
- iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark; Department of Chemistry, Aarhus University, Langelandsgade 140, 8000 Aarhus, Denmark
| | - Scot Mente
- Forma Therapeutics, Inc., 500 Arsenal Street, Suite 100, Watertown, MA 02472, USA
| | | | - Daniel E Otzen
- iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus, Denmark.
| |
Collapse
|
95
|
Choi TS, Han JY, Heo CE, Lee SW, Kim HI. Electrostatic and hydrophobic interactions of lipid-associated α-synuclein: The role of a water-limited interfaces in amyloid fibrillation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1854-1862. [DOI: 10.1016/j.bbamem.2018.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 12/22/2022]
|
96
|
Pan J, Dalzini A, Khadka NK, Aryal CM, Song L. Lipid Extraction by α-Synuclein Generates Semi-Transmembrane Defects and Lipoprotein Nanoparticles. ACS OMEGA 2018; 3:9586-9597. [PMID: 30198000 PMCID: PMC6120733 DOI: 10.1021/acsomega.8b01462] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/07/2018] [Indexed: 05/17/2023]
Abstract
Modulations of synaptic membranes play an essential role in the physiological and pathological functions of the presynaptic protein α-synuclein (αSyn). Here we used solution atomic force microscopy (AFM) and electron paramagnetic resonance (EPR) spectroscopy to investigate membrane modulations caused by αSyn. We used several lipid bilayers to explore how different lipid species may regulate αSyn-membrane interactions. We found that at a protein-to-lipid ratio of ∼1/9, αSyn perturbed lipid bilayers by generating semi-transmembrane defects that only span one leaflet. In addition, αSyn coaggregates with lipid molecules to produce ∼10 nm-sized lipoprotein nanoparticles. The obtained AFM data are consistent with the apolipoprotein characteristic of αSyn. The role of anionic lipids was elucidated by comparing results from zwitterionic and anionic lipid bilayers. Specifically, our AFM measurements showed that anionic bilayers had a larger tendency of forming bilayer defects; similarly, our EPR measurements revealed that anionic bilayers exhibited more substantial changes in lipid chain mobility and bilayer polarity. We also studied the effect of cholesterol. We found that cholesterol increased the capability of αSyn in inducing bilayer defects and altering lipid chain mobility and bilayer polarity. These data can be explained by an increase in the lipid headgroup-headgroup spacing and/or specific cholesterol-αSyn interactions. Interestingly, we found an inhibitory effect of the cone-shaped phosphatidylethanolamine lipids on αSyn-induced bilayer remodeling. We explained our data by considering interlipid hydrogen-bonding that can stabilize bilayer organization and suppress lipid extraction. Our results of lipid-dependent membrane modulations are likely relevant to αSyn functioning.
Collapse
Affiliation(s)
- Jianjun Pan
- Department
of Physics, University of South Florida, Tampa, Florida 33620, United States
- E-mail: (J.P.)
| | - Annalisa Dalzini
- National
High Magnetic Field Laboratory, Florida
State University, Tallahassee, Florida 32310, United States
| | - Nawal K. Khadka
- Department
of Physics, University of South Florida, Tampa, Florida 33620, United States
| | - Chinta M. Aryal
- Department
of Physics, University of South Florida, Tampa, Florida 33620, United States
| | - Likai Song
- National
High Magnetic Field Laboratory, Florida
State University, Tallahassee, Florida 32310, United States
- E-mail: (L.S.)
| |
Collapse
|
97
|
Cox D, Raeburn C, Sui X, Hatters DM. Protein aggregation in cell biology: An aggregomics perspective of health and disease. Semin Cell Dev Biol 2018; 99:40-54. [PMID: 29753879 DOI: 10.1016/j.semcdb.2018.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/21/2018] [Accepted: 05/04/2018] [Indexed: 01/08/2023]
Abstract
Maintaining protein homeostasis (proteostasis) is essential for cellular health and is governed by a network of quality control machinery comprising over 800 genes. When proteostasis becomes imbalanced, proteins can abnormally aggregate or become mislocalized. Inappropriate protein aggregation and proteostasis imbalance are two of the central pathological features of common neurodegenerative diseases including Alzheimer, Parkinson, Huntington, and motor neuron diseases. How aggregation contributes to the pathogenic mechanisms of disease remains incompletely understood. Here, we integrate some of the key and emerging ideas as to how protein aggregation relates to imbalanced proteostasis with an emphasis on Huntington disease as our area of main expertise. We propose the term "aggregomics" be coined in reference to how aggregation of particular proteins concomitantly influences the spatial organization and protein-protein interactions of the surrounding proteome. Meta-analysis of aggregated interactomes from various published datasets reveals chaperones and RNA-binding proteins are common components across various disease contexts. We conclude with an examination of therapeutic avenues targeting proteostasis mechanisms.
Collapse
Affiliation(s)
- Dezerae Cox
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Australia; Bio21 Molecular Science and Biotechnology Institute, Australia
| | - Candice Raeburn
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Australia; Bio21 Molecular Science and Biotechnology Institute, Australia
| | - Xiaojing Sui
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Australia; Bio21 Molecular Science and Biotechnology Institute, Australia
| | - Danny M Hatters
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Australia; Bio21 Molecular Science and Biotechnology Institute, Australia.
| |
Collapse
|
98
|
Lipid vesicles affect the aggregation of 4-hydroxy-2-nonenal-modified α-synuclein oligomers. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3060-3068. [PMID: 29960040 DOI: 10.1016/j.bbadis.2018.06.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 11/23/2022]
Abstract
Parkinson's disease (PD) and other synucleinopathies are characterized by accumulation of misfolded aggregates of α-synuclein (α-syn). The normal function of α-syn is still under investigation, but it has been generally linked to synaptic plasticity, neurotransmitter release and the maintenance of the synaptic pool. α-Syn localizes at synaptic terminals where it can bind to synaptic vesicles as well as to other cellular membranes. It has become clear that these interactions have an impact on both α-syn functional role and its propensity to aggregate. In this study, we investigated the aggregation process of α-syn covalently modified with 4-hydroxy-2-nonenal (HNE). HNE is a product of lipid peroxidation and has been implicated in the pathogenesis of different neurodegenerative diseases by modifying the kinetics of soluble toxic oligomers. Although HNE-modified α-syn has been reported to assemble into stable oligomers, we found that slightly acidic conditions promoted further protein aggregation. Lipid vesicles delayed the aggregation process in a concentration-dependent manner, an effect that was observed only when they were added at the beginning of the aggregation process. Co-aggregation of lipid vesicles with HNE-modified α-syn also induced cytotoxic effects on differentiated SHSY-5Y cells. Under conditions in which the aggregation process was delayed cell viability was reduced. By exploring the behavior and potential cytotoxic effects of HNE-α-syn under acidic conditions in relation to protein-lipid interactions our study gives a framework to examine a possible pathway leading from a physiological setting to the pathological outcome of PD.
Collapse
|
99
|
Caplan IF, Maguire-Zeiss KA. Toll-Like Receptor 2 Signaling and Current Approaches for Therapeutic Modulation in Synucleinopathies. Front Pharmacol 2018; 9:417. [PMID: 29780321 PMCID: PMC5945810 DOI: 10.3389/fphar.2018.00417] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 04/10/2018] [Indexed: 12/18/2022] Open
Abstract
The innate immune response in the central nervous system (CNS) is implicated as both beneficial and detrimental to health. Integral to this process are microglia, the resident immune cells of the CNS. Microglia express a wide variety of pattern-recognition receptors, such as Toll-like receptors, that detect changes in the neural environment. The activation of microglia and the subsequent proinflammatory response has become increasingly relevant to synucleinopathies, including Parkinson's disease the second most prevalent neurodegenerative disease. Within these diseases there is evidence of the accumulation of endogenous α-synuclein that stimulates an inflammatory response from microglia via the Toll-like receptors. There have been recent developments in both new and old pharmacological agents designed to target microglia and curtail the inflammatory environment. This review will aim to delineate the process of microglia-mediated inflammation and new therapeutic avenues to manage the response.
Collapse
Affiliation(s)
- Ian F Caplan
- Biology Department, Georgetown University, Washington, DC, United States
| | - Kathleen A Maguire-Zeiss
- Biology Department, Georgetown University, Washington, DC, United States.,Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
100
|
Dong C, Hoffmann M, Li X, Wang M, Garen CR, Petersen NO, Woodside MT. Structural characteristics and membrane interactions of tandem α-synuclein oligomers. Sci Rep 2018; 8:6755. [PMID: 29712958 PMCID: PMC5928076 DOI: 10.1038/s41598-018-25133-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/16/2018] [Indexed: 01/04/2023] Open
Abstract
Pre-fibrillar oligomers of α-synuclein are thought to be pathogenic molecules leading to neurotoxicity associated with Parkinson’s disease and other neurodegenerative disorders. However, small oligomers are difficult to isolate for study. To gain better insight into the properties of small α-synuclein oligomers, we investigated engineered oligomers of specific size (dimers, tetramers, and octamers) linked head-to-tail in tandem, comparing the behavior of the oligomers to monomeric α-synuclein. All oligomeric constructs remained largely disordered in solution, as determined from dynamic light scattering and size-exclusion chromatography. Electron microscopy revealed that each construct could aggregate to form fibrils similar to those formed by monomeric α-synuclein. The interactions with large unilamellar vesicles (LUVs) composed of negatively-charged lipids differed depending on size, with smaller oligomers forming more extensive helical structure as determined by CD spectroscopy. Monitoring the influx of a fluorescence bleaching agent into vesicles showed that larger oligomers were somewhat more effective at degrading vesicular integrity and inducing membrane permeabilization.
Collapse
Affiliation(s)
- Chunhua Dong
- Department of Physics, University of Alberta, Edmonton, AB, Canada.,Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Marion Hoffmann
- Department of Physics, University of Alberta, Edmonton, AB, Canada
| | - Xi Li
- Department of Physics, University of Alberta, Edmonton, AB, Canada.,Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Meijing Wang
- Department of Physics, University of Alberta, Edmonton, AB, Canada.,National Research Council, National Institute of Nanotechnology, University of Alberta, Edmonton, AB, Canada
| | - Craig R Garen
- Department of Physics, University of Alberta, Edmonton, AB, Canada
| | - Nils O Petersen
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Michael T Woodside
- Department of Physics, University of Alberta, Edmonton, AB, Canada. .,National Research Council, National Institute of Nanotechnology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|