Structure-Based Discovery of Small-Molecule Inhibitors of the Autocatalytic Proliferation of α-Synuclein AggregatesClick to copy article linkArticle link copied!
- Sean ChiaSean ChiaCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Sean Chia
- Z. Faidon BrotzakisZ. Faidon BrotzakisCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Z. Faidon Brotzakis
- Robert I. HorneRobert I. HorneCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Robert I. Horne
- Andrea PossentiAndrea PossentiCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Andrea Possenti
- Benedetta ManniniBenedetta ManniniCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Benedetta Mannini
- Rodrigo CataldiRodrigo CataldiCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Rodrigo Cataldi
- Magdalena NowinskaMagdalena NowinskaCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Magdalena Nowinska
- Roxine StaatsRoxine StaatsCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Roxine Staats
- Sara LinseSara LinseDepartment of Biochemistry & Structural Biology, Center for Molecular Protein Science, Lund University, 221 00Lund, SwedenMore by Sara Linse
- Tuomas P. J. KnowlesTuomas P. J. KnowlesCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.Department of Physics, Cavendish Laboratory, CambridgeCB3 0HE, U.K.More by Tuomas P. J. Knowles
- Johnny HabchiJohnny HabchiCentre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Johnny Habchi
- Michele Vendruscolo*Michele Vendruscolo*Email: [email protected]Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.More by Michele Vendruscolo
Abstract
The presence of amyloid fibrils of α-synuclein is closely associated with Parkinson’s disease and related synucleinopathies. It is still very challenging, however, to systematically discover small molecules that prevent the formation of these aberrant aggregates. Here, we describe a structure-based approach to identify small molecules that specifically inhibit the surface-catalyzed secondary nucleation step in the aggregation of α-synuclein by binding to the surface of the amyloid fibrils. The resulting small molecules are screened using a range of kinetic and thermodynamic assays for their ability to bind α-synuclein fibrils and prevent the further generation of α-synuclein oligomers. This study demonstrates that the combination of structure-based and kinetic-based drug discovery methods can lead to the identification of small molecules that selectively inhibit the autocatalytic proliferation of α-synuclein aggregates.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Materials and Methods
Computational Docking
Preparation of Compounds and Chemicals
Preparation of α-Synuclein
Preparation of α-Synuclein Fibril Seeds
Kinetic Assays
Transmission Electron Microscopy (TEM)
Determination of the Elongation Rate Constant
Determination of the Fibril Amplification Rate Constant
Determination of the Oligomer Flux
Fluorescence Polarization
Mass Spectrometry
Cell Cultures
Colocalization Assay
Results
Framework to Identify Compounds That Bind α-Synuclein Fibrils
Figure 1
Figure 1. Combined structure-based and kinetic-based approach to identify small molecules that bind α-synuclein fibrils and inhibit its aggregation. In the first step, computational docking is performed on a large library of small molecules. The top candidates are then clustered to identify a subset of chemically diverse compounds that exhibit high predicted binding scores for α-synuclein fibrils. Subsequently, these compounds are experimentally validated through a kinetic assay for their ability to inhibit the secondary nucleation aggregation of α-synuclein by binding to the surface of fibrils. Further rate constant analysis and fibril-binding experiments allow for the positive compounds to be characterized based on both their inhibition of the kinetic assay, as well as their binding affinity toward α-synuclein fibrils.
In Silico Docking of Compounds Predicted to Bind α-Synuclein Fibrils
Identification of Compounds That Inhibit α-Synuclein Secondary Nucleation
Figure 2
Figure 2. Five compounds selected from the docking library inhibit the aggregation of α-synuclein. (A) Kinetic profiles of a 10 μM solution of α-synuclein in the presence of 25 nM seeds at pH 4.8 and 37 °C, in the presence of 1% DMSO alone (beige), in the presence of 10 molar equivalents of compounds A–E (represented in different colors), or in the presence of 10 molar equivalents of other compounds in the docking library that did not affect significantly α-synuclein aggregation (black). (B) Relative t1/2 of the aggregation of α-synuclein in the presence of compounds A–E as shown in (A), normalized to the DMSO control. (C) Chemical structures of compounds A–E. Throughout, error bars represent mean ± SEM of two replicates.
Figure 3
Figure 3. Computational docking of compounds to α-synuclein fibrils. (A–E) Binding poses of compounds A–E to the selected binding pocket in α-synuclein fibrils (centered between residues His50 and Glu57), determined either through FRED or AutoDock Vina. (F) Representation of possible binding pockets in the fibril structure (PDB: 6cu7, cyan) identified by Fpocket, with pockets in the fibril core (blue spheres), and at the fibril surface (red spheres). Key binding site residues His50 and Glu57 are shown in licorice representation.
Kinetic Analysis of α-Synuclein Aggregation in the Presence of the Inhibitors
Figure 4
Figure 4. Compounds identified by docking specifically inhibit the proliferation of α-synuclein aggregates by secondary nucleation. (A) Kinetic profiles of a 10 μM solution of α-synuclein in the presence of 25 nM seeds at pH 4.8, 37 °C, in the presence of either 1% DMSO alone (purple) or increasing molar equivalents of compound C (represented in different colors). (B) Relative rate of fibril amplification of α-synuclein in the presence of compounds A–E as shown in (A) and Figure S3, normalized to the DMSO control. (C) Kinetic profiles of a 10 μM solution of α-synuclein in the presence of 5 μM seeds at pH 4.8, 37 °C, in the presence of either 1% DMSO alone (purple) or increasing molar equivalents of compound C (represented in different colors). Dotted lines indicate the vmax of the reaction which is used to extract the elongation rate of the aggregation process. (D) Relative rate of fibril elongation of α-synuclein in the presence of compounds A–E as shown in (C) and Figure S5,6, normalized to the DMSO control. Throughout, error bars represent mean ± SEM of three replicates.
Compound C Inhibits α-Synuclein Oligomer Formation
Figure 5
Figure 5. Compound C inhibits the reactive flux toward α-synuclein oligomers and displays binding affinity and specificity toward α-synuclein fibrils. (A) Time dependence of the reactive flux toward α-synuclein oligomers either in the presence of 1% DMSO alone (purple) or in the presence of increasing molar equivalents of compound C (represented in different colors), normalized to the DMSO control. (B) Change in fluorescence polarization (in mP units) of 10 μM compound C with increasing concentrations of either α-synuclein fibrils (purple) or Aβ42 fibrils (red). The solid lines are fits to the points using a one-step binding curve, estimating a Kd of 4 μM for compound C toward α-synuclein fibrils. (C) Total ion current (TIC) of 10 μM compound C bound and unbound to 10 μM α-synuclein fibrils detected by mass spectrometry (see the Materials and Methods section). (D) Representative images indicating either the fluorescence of the red channel (amyloid-specific dye pFTAA) or the green channel (compound C) following incubation in the absence (top) or presence (bottom) of 100 nM α-synuclein fibrils. Throughout, error bars represent mean ± SEM of two replicates.
Compound C Binds α-Synuclein Fibrils
Discussion
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.molpharmaceut.2c00548.
Predicted binding affinity of compounds via computational docking; TEM images of α-synuclein fibrils formed in the presence of molecules; kinetic profiles of α-synuclein aggregation in the presence of molecules; time dependence of reactive flux toward α-synuclein oligomers in the presence of molecules; fluorescence emission spectra of molecule C; colocalization of molecules with α-synuclein fibrils in the presence of neuroblastoma cells; list of all molecules from docking studies experimentally validated; and supplementary methods, supplementary figures, and supplementary table (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Kd | apparent dissociation constant |
SAR | structure–activity relationship |
TEM | transmission electron microscopy |
References
This article references 50 other publications.
- 1Poewe, W.; Seppi, K.; Tanner, C. M.; Halliday, G. M.; Brundin, P.; Volkmann, J.; Schrag, A. E.; Lang, A. E. Parkinson Disease. Nat. Rev. Dis. Primers 2017, 3, 17013, DOI: 10.1038/nrdp.2017.13Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cvgtFChsA%253D%253D&md5=b624b37fee08b8b663ea88b83bc47865Parkinson diseasePoewe Werner; Seppi Klaus; Tanner Caroline M; Tanner Caroline M; Halliday Glenda M; Halliday Glenda M; Brundin Patrik; Volkmann Jens; Schrag Anette-Eleonore; Lang Anthony ENature reviews. Disease primers (2017), 3 (), 17013 ISSN:.Parkinson disease is the second-most common neurodegenerative disorder that affects 2-3% of the population ≥65 years of age. Neuronal loss in the substantia nigra, which causes striatal dopamine deficiency, and intracellular inclusions containing aggregates of α-synuclein are the neuropathological hallmarks of Parkinson disease. Multiple other cell types throughout the central and peripheral autonomic nervous system are also involved, probably from early disease onwards. Although clinical diagnosis relies on the presence of bradykinesia and other cardinal motor features, Parkinson disease is associated with many non-motor symptoms that add to overall disability. The underlying molecular pathogenesis involves multiple pathways and mechanisms: α-synuclein proteostasis, mitochondrial function, oxidative stress, calcium homeostasis, axonal transport and neuroinflammation. Recent research into diagnostic biomarkers has taken advantage of neuroimaging in which several modalities, including PET, single-photon emission CT (SPECT) and novel MRI techniques, have been shown to aid early and differential diagnosis. Treatment of Parkinson disease is anchored on pharmacological substitution of striatal dopamine, in addition to non-dopaminergic approaches to address both motor and non-motor symptoms and deep brain stimulation for those developing intractable L-DOPA-related motor complications. Experimental therapies have tried to restore striatal dopamine by gene-based and cell-based approaches, and most recently, aggregation and cellular transport of α-synuclein have become therapeutic targets. One of the greatest current challenges is to identify markers for prodromal disease stages, which would allow novel disease-modifying therapies to be started earlier.
- 2Nichols, E.; Szoeke, C. E. I.; Vollset, S. E.; Abbasi, N.; Abd-Allah, F.; Abdela, J.; Aichour, M. T. E.; Akinyemi, R. O.; Alahdab, F.; Asgedom, S. W.; Awasthi, A.; Barker-Collo, S. L.; Baune, B. T.; Béjot, Y.; Belachew, A. B.; Bennett, D. A.; Biadgo, B.; Bijani, A.; Bin Sayeed, M. S.; Brayne, C.; Carpenter, D. O.; Carvalho, F.; Catalá-López, F.; Cerin, E.; Choi, J. Y. J.; Dang, A. K.; Degefa, M. G.; Djalalinia, S.; Dubey, M.; Duken, E. E.; Edvardsson, D.; Endres, M.; Eskandarieh, S.; Faro, A.; Farzadfar, F.; Fereshtehnejad, S. M.; Fernandes, E.; Filip, I.; Fischer, F.; Gebre, A. K.; Geremew, D.; Ghasemi-Kasman, M.; Gnedovskaya, E. V.; Gupta, R.; Hachinski, V.; Hagos, T. B.; Hamidi, S.; Hankey, G. J.; Haro, J. M.; Hay, S. I.; Irvani, S. S. N.; Jha, R. P.; Jonas, J. B.; Kalani, R.; Karch, A.; Kasaeian, A.; Khader, Y. S.; Khalil, I. A.; Khan, E. A.; Khanna, T.; Khoja, T. A. M.; Khubchandani, J.; Kisa, A.; Kissimova-Skarbek, K.; Kivimäki, M.; Koyanagi, A.; Krohn, K. J.; Logroscino, G.; Lorkowski, S.; Majdan, M.; Malekzadeh, R.; März, W.; Massano, J.; Mengistu, G.; Meretoja, A.; Mohammadi, M.; Mohammadi-Khanaposhtani, M.; Mokdad, A. H.; Mondello, S.; Moradi, G.; Nagel, G.; Naghavi, M.; Naik, G.; Nguyen, L. H.; Nguyen, T. H.; Nirayo, Y. L.; Nixon, M. R.; Ofori-Asenso, R.; Ogbo, F. A.; Olagunju, A. T.; Owolabi, M. O.; Panda-Jonas, S.; Passos, V. M. d. A.; Pereira, D. M.; Pinilla-Monsalve, G. D.; Piradov, M. A.; Pond, C. D.; Poustchi, H.; Qorbani, M.; Radfar, A.; Reiner, R. C.; Robinson, S. R.; Roshandel, G.; Rostami, A.; Russ, T. C.; Sachdev, P. S.; Safari, H.; Safiri, S.; Sahathevan, R.; Salimi, Y.; Satpathy, M.; Sawhney, M.; Saylan, M.; Sepanlou, S. G.; Shafieesabet, A.; Shaikh, M. A.; Sahraian, M. A.; Shigematsu, M.; Shiri, R.; Shiue, I.; Silva, J. P.; Smith, M.; Sobhani, S.; Stein, D. J.; Tabarés-Seisdedos, R.; Tovani-Palone, M. R.; Tran, B. X.; Tran, T. T.; Tsegay, A. T.; Ullah, I.; Venketasubramanian, N.; Vlassov, V.; Wang, Y. P.; Weiss, J.; Westerman, R.; Wijeratne, T.; Wyper, G. M. A.; Yano, Y.; Yimer, E. M.; Yonemoto, N.; Yousefifard, M.; Zaidi, Z.; Zare, Z.; Vos, T.; Feigin, V. L.; Murray, C. J. L. Global, Regional, and National Burden of Alzheimer’s Disease and Other Dementias, 1990–2016: A Systematic Analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 88– 106, DOI: 10.1016/S1474-4422(18)30403-4Google ScholarThere is no corresponding record for this reference.
- 3Balestrino, R.; Schapira, A. H. V. Parkinson Disease. Eur. J. Neurol. 2020, 27, 27– 42, DOI: 10.1111/ene.14108Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MnpslGguw%253D%253D&md5=2a9d346119d47bb04883b69786b0308cParkinson diseaseBalestrino R; Schapira A H VEuropean journal of neurology (2020), 27 (1), 27-42 ISSN:.Parkinson disease (PD) is the most common neurodegenerative movement disorder. In Europe, prevalence and incidence rates for PD are estimated at approximately 108-257/100 000 and 11-19/100 000 per year, respectively. Risk factors include age, male gender and some environmental factors. The aetiology of the disease in most patients is unknown, but different genetic causes have been identified. Although familial forms of PD account for only 5%-15% of cases, studies on these families provided interesting insight on the genetics and the pathogenesis of the disease allowing the identification of genes implicated in its pathogenesis and offering critical insights into the mechanisms of disease. The cardinal motor symptoms of PD are tremor, rigidity, bradykinesia/akinesia and postural instability, but the clinical picture includes other motor and non-motor symptoms. Its diagnosis is principally clinical, although specific investigations can help the differential diagnosis from other forms of parkinsonism. Pathologically, PD is characterized by the loss of dopaminergic neurons in the pars compacta of the substantia nigra and by accumulation of misfolded α-synuclein, which is found in intra-cytoplasmic inclusions called Lewy bodies. Currently available treatments offer good control of motor symptoms but do not modify the evolution of the disease. This article is intended to provide a comprehensive, general and practical review of PD for the general neurologist.
- 4Pringsheim, T.; Jette, N.; Frolkis, A.; Steeves, T. D. L. The Prevalence of Parkinson’s Disease: A Systematic Review and Meta-Analysis. Mov. Disord. 2014, 29, 1583– 1590, DOI: 10.1002/mds.25945Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cfns1yiuw%253D%253D&md5=6e281ab13ef57a2c8c7c91d44cced519The prevalence of Parkinson's disease: a systematic review and meta-analysisPringsheim Tamara; Jette Nathalie; Frolkis Alexandra; Steeves Thomas D LMovement disorders : official journal of the Movement Disorder Society (2014), 29 (13), 1583-90 ISSN:.Parkinson's Disease (PD) is a common neurodegenerative disorder. We sought to synthesize studies on the prevalence of PD to obtain an overall view of how the prevalence of this disease varies by age, by sex, and by geographic location. We searched MEDLINE and EMBASE for epidemiological studies of PD from 1985 to 2010. Data were analyzed by age group, geographic location, and sex. Geographic location was stratified by the following groups: 1) Asia, 2) Africa, 3) South America, and 4) Europe/North America/Australia. Meta-regression was used to determine whether a significant difference was present between groups. Forty-seven studies were included in the analysis. Meta-analysis of the worldwide data showed a rising prevalence of PD with age (all per 100,000): 41 in 40 to 49 years; 107 in 50 to 59 years; 173 in 55 to 64 years; 428 in 60 to 69 years; 425 in 65 to 74 years; 1087 in 70 to 79 years; and 1903 in older than age 80. A significant difference was seen in prevalence by geographic location only for individuals 70 to 79 years old, with a prevalence of 1,601 in individuals from North America, Europe, and Australia, compared with 646 in individuals from Asia (P < 0.05). A significant difference in prevalence by sex was found only for individuals 50 to 59 years old, with a prevalence of 41 in females and 134 in males (P < 0.05). PD prevalence increases steadily with age. Some differences in prevalence by geographic location and sex can be detected.
- 5Spillantini, M. G.; Schmidt, M. L.; Lee, V. M.; Trojanowski, J. Q.; Jakes, R.; Goedert, M. Alpha-Synuclein in Lewy Bodies. Nature 1997, 388, 839– 840, DOI: 10.1038/42166Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXlslWru7o%253D&md5=ef5d22aaeb6f08828ea33c50e3f7c06dα-Synuclein in Lewy bodiesSpillantini, Maria Grazia; Schmidt, Marie Luise; Lee, Virginia M.-Y.; Trojanowski, John Q.; Jakes, Ross; Goedert, MichelNature (London) (1997), 388 (6645), 839-840CODEN: NATUAS; ISSN:0028-0836. (Macmillan Magazines)The authors report strong staining of Lewy bodies in brain in idiopathic Parkinson's disease and dementia with Lewy bodies with antibodies for α-synuclein. They conclude that Parkinson's disease and dementia with Lewy bodies may both be α-synuclein diseases.
- 6Goedert, M.; Spillantini, M. G.; Del Tredici, K.; Braak, H. 100 Years of Lewy Pathology. Nat. Rev. Neurol. 2013, 9, 13– 24, DOI: 10.1038/nrneurol.2012.242Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXntlWr&md5=947a357746ed61eeab6d2891bf10b013100 years of Lewy pathologyGoedert, Michel; Spillantini, Maria Grazia; Del Tredici, Kelly; Braak, HeikoNature Reviews Neurology (2013), 9 (1), 13-24CODEN: NRNACP; ISSN:1759-4758. (Nature Publishing Group)A review. In 1817, James Parkinson described the symptoms of the shaking palsy, a disease that was subsequently defined in greater detail, and named after Parkinson, by Jean-Martin Charcot. Parkinson expected that the publication of his monograph would lead to a rapid elucidation of the anatomical substrate of the shaking palsy; in the event, this process took almost a century. In 1912, Fritz Heinrich Lewy identified the protein aggregates that define Parkinson disease (PD) in some brain regions outside the substantia nigra. In 1919, Konstantin Nikolaevich Tretiakoff found similar aggregates in the substantia nigra and named them after Lewy. In the 1990s, α-synuclein was identified as the main constituent of the Lewy pathol., and its aggregation was shown to be central to PD, dementia with Lewy bodies, and multiple system atrophy. In 2003, a staging scheme for idiopathic PD was introduced, according to which α-synuclein pathol. originates in the dorsal motor nucleus of the vagal nerve and progresses from there to other brain regions, including the substantia nigra. In this article, we review the relevance of Lewy's discovery 100 years ago for the current understanding of PD and related disorders.
- 7Fusco, G.; Chen, S. W.; Williamson, P. T. F.; Cascella, R.; Perni, M.; Jarvis, J. A.; Cecchi, C.; Vendruscolo, M.; Chiti, F.; Cremades, N.; Ying, L. Structural Basis of Membrane Disruption and Cellular Toxicity by α-Synuclein Oligomers. Science 2017, 358, 1440– 1443, DOI: 10.1126/science.aan6160Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvFGmtrzN&md5=5e54a267d090e6f747aeca57de8b19b8Structural basis of membrane disruption and cellular toxicity by α-synuclein oligomersFusco, Giuliana; Chen, Serene W.; Williamson, Philip T. F.; Cascella, Roberta; Perni, Michele; Jarvis, James A.; Cecchi, Cristina; Vendruscolo, Michele; Chiti, Fabrizio; Cremades, Nunilo; Ying, Liming; Dobson, Christopher M.; De Simone, AlfonsoScience (Washington, DC, United States) (2017), 358 (6369), 1440-1443CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Oligomeric species populated during the aggregation process of α-synuclein have been linked to neuronal impairment in Parkinson's disease and related neurodegenerative disorders. By using soln. and solid-state NMR techniques in conjunction with other structural methods, we identified the fundamental characteristics that enable toxic α-synuclein oligomers to perturb biol. membranes and disrupt cellular function; these include a highly lipophilic element that promotes strong membrane interactions and a structured region that inserts into lipid bilayers and disrupts their integrity. In support of these conclusions, mutations that target the region that promotes strong membrane interactions by α-synuclein oligomers suppressed their toxicity in neuroblastoma cells and primary cortical neurons.
- 8Lashuel, H. A.; Overk, C. R.; Oueslati, A.; Masliah, E. The Many Faces of α-Synuclein: From Structure and Toxicity to Therapeutic Target. Nat. Rev. Neurosci. 2013, 14, 38– 48, DOI: 10.1038/nrn3406Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVGls7jN&md5=930fda9ece4cfa715d3fa58d496e7a13The many faces of α-synuclein: from structure and toxicity to therapeutic targetLashuel, Hilal A.; Overk, Cassia R.; Oueslati, Abid; Masliah, EliezerNature Reviews Neuroscience (2013), 14 (1), 38-48CODEN: NRNAAN; ISSN:1471-003X. (Nature Publishing Group)A review. Disorders characterized by α-synuclein (α-syn) accumulation, Lewy body formation and parkinsonism (and in some cases dementia) are collectively known as Lewy body diseases. The mol. mechanism (or mechanisms) through which α-syn abnormally accumulates and contributes to neurodegeneration in these disorders remains unknown. Here, we provide an overview of current knowledge and prevailing hypotheses regarding the conformational, oligomerization and aggregation states of α-syn and their role in regulating α-syn function in health and disease. Understanding the nature of the various α-syn structures, how they are formed and their relative contributions to α-syn-mediated toxicity may inform future studies aiming to develop therapeutic prevention and intervention.
- 9Haass, C.; Selkoe, D. J. Soluble Protein Oligomers in Neurodegeneration: Lessons from the Alzheimer’s Amyloid β-Peptide. Nat. Rev. Mol. Cell Biol. 2007, 8, 101– 112, DOI: 10.1038/nrm2101Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXotFKmtw%253D%253D&md5=e8634ff5c1491580f623c90ded91b4beSoluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid β-peptideHaass, Christian; Selkoe, Dennis J.Nature Reviews Molecular Cell Biology (2007), 8 (2), 101-112CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. The distinct protein aggregates that are found in Alzheimer's, Parkinson's, Huntington's and prion diseases seem to cause these disorders. Small intermediates - sol. oligomers - in the aggregation process can confer synaptic dysfunction, whereas large, insol. deposits might function as reservoirs of the bioactive oligomers. These emerging concepts are exemplified by Alzheimer's disease, in which amyloid β-protein oligomers adversely affect synaptic structure and plasticity. Findings in other neurodegenerative diseases indicate that a broadly similar process of neuronal dysfunction is induced by diffusible oligomers of misfolded proteins.
- 10Prots, I.; Grosch, J.; Brazdis, R. M.; Simmnacher, K.; Veber, V.; Havlicek, S.; Hannappel, C.; Krach, F.; Krumbiegel, M.; Schütz, O.; Reis, A.; Wrasidlo, W.; Galasko, D. R.; Groemer, T. W.; Masliah, E.; Schlötzer-Schrehardt, U.; Xiang, W.; Winkler, J.; Winner, B. α-Synuclein Oligomers Induce Early Axonal Dysfunction in Human IPSC-Based Models of Synucleinopathies. Proc. Natl. Acad. Sci. U.S.A. 2018, 115, 7813– 7818, DOI: 10.1073/pnas.1713129115Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFCmtrjL&md5=583d4ea493a22aafa3aeb0af58f0755bα-Synuclein oligomers induce early axonal dysfunction in human iPSC-based models of synucleinopathiesProts, Iryna; Grosch, Janina; Brazdis, Razvan-Marius; Simmnacher, Katrin; Veber, Vanesa; Havlicek, Steven; Hannappel, Christian; Krach, Florian; Krumbiegel, Mandy; Schuetz, Oliver; Reis, Andre; Wrasidlo, Wolfgang; Galasko, Douglas R.; Groemer, Teja W.; Masliah, Eliezer; Schloetzer-Schrehardt, Ursula; Xiang, Wei; Winkler, Juergen; Winner, BeateProceedings of the National Academy of Sciences of the United States of America (2018), 115 (30), 7813-7818CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)α-Synuclein (α-Syn) aggregation, proceeding from oligomers to fibrils, is one central hallmark of neurodegeneration in synucleinopathies. α-Syn oligomers are toxic by triggering neurodegenerative processes in in vitro and in vivo models. However, the precise contribution of α-Syn oligomers to neurite pathol. in human neurons and the underlying mechanisms remain unclear. Here, we demonstrate the formation of oligomeric α-Syn intermediates and reduced axonal mitochondrial transport in human neurons derived from induced pluripotent stem cells (iPSC) from a Parkinson's disease patient carrying an α-Syn gene duplication. We further show that increased levels of α-Syn oligomers disrupt axonal integrity in human neurons. We apply an α-Syn oligomerization model by expressing α-Syn oligomer-forming mutants (E46K and E57K) and wild-type α-Syn in human iPSC-derived neurons. Pronounced α-Syn oligomerization led to impaired anterograde axonal transport of mitochondria, which can be restored by the inhibition of α-Syn oligomer formation. Furthermore, α-Syn oligomers were assocd. with a subcellular relocation of transport-regulating proteins Miro1, KLC1, and Tau as well as reduced ATP levels, underlying axonal transport deficits. Consequently, reduced axonal d. and structural synaptic degeneration were obsd. in human neurons in the presence of high levels of α-Syn oligomers. Together, increased dosage of α-Syn resulting in α-Syn oligomerization causes axonal transport disruption and energy deficits, leading to synapse loss in human neurons. This study identifies α-Syn oligomers as the crit. species triggering early axonal dysfunction in synucleinopathies.
- 11Ludtmann, M. H. R.; Angelova, P. R.; Horrocks, M. H.; Choi, M. L.; Rodrigues, M.; Baev, A. Y.; Berezhnov, A. V.; Yao, Z.; Little, D.; Banushi, B.; Al-Menhali, A. S.; Ranasinghe, R. T.; Whiten, D. R.; Yapom, R.; Dolt, K. S.; Devine, M. J.; Gissen, P.; Kunath, T.; Jaganjac, M.; Pavlov, E. V.; Klenerman, D.; Abramov, A. Y.; Gandhi, S. α-Synuclein Oligomers Interact with ATP Synthase and Open the Permeability Transition Pore in Parkinson’s Disease. Nat. Commun. 2018, 9, 2293 DOI: 10.1038/s41467-018-04422-2Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1Mbms1Kjsw%253D%253D&md5=7f4729f901fbb58e29c180ab98999bbaα-synuclein oligomers interact with ATP synthase and open the permeability transition pore in Parkinson's diseaseLudtmann Marthe H R; Angelova Plamena R; Abramov Andrey Y; Ludtmann Marthe H R; Horrocks Mathew H; Rodrigues Margarida; Ranasinghe Rohan T; Whiten Daniel R; Klenerman David; Horrocks Mathew H; Horrocks Mathew H; Choi Minee L; Yao Zhi; Gandhi Sonia; Choi Minee L; Yao Zhi; Gandhi Sonia; Baev Artyom Y; Berezhnov Alexey V; Little Daniel; Banushi Blerida; Devine Michael J; Gissen Paul; Al-Menhali Afnan Saleh; Jaganjac Morana; Yapom Ratsuda; Dolt Karamjit Singh; Kunath Tilo; Devine Michael J; Pavlov Evgeny V; Klenerman DavidNature communications (2018), 9 (1), 2293 ISSN:.Protein aggregation causes α-synuclein to switch from its physiological role to a pathological toxic gain of function. Under physiological conditions, monomeric α-synuclein improves ATP synthase efficiency. Here, we report that aggregation of monomers generates beta sheet-rich oligomers that localise to the mitochondria in close proximity to several mitochondrial proteins including ATP synthase. Oligomeric α-synuclein impairs complex I-dependent respiration. Oligomers induce selective oxidation of the ATP synthase beta subunit and mitochondrial lipid peroxidation. These oxidation events increase the probability of permeability transition pore (PTP) opening, triggering mitochondrial swelling, and ultimately cell death. Notably, inhibition of oligomer-induced oxidation prevents the pathological induction of PTP. Inducible pluripotent stem cells (iPSC)-derived neurons bearing SNCA triplication, generate α-synuclein aggregates that interact with the ATP synthase and induce PTP opening, leading to neuronal death. This study shows how the transition of α-synuclein from its monomeric to oligomeric structure alters its functional consequences in Parkinson's disease.
- 12Hughes, C. D.; Choi, M. L.; Ryten, M.; Hopkins, L.; Drews, A.; Botía, J. A.; Iljina, M.; Rodrigues, M.; Gagliano, S. A.; Gandhi, S.; Klenerman, D.; Bryant, C. Picomolar Concentrations of Oligomeric Alpha - Synuclein Sensitizes TLR4 to Play an Initiating Role in Parkinson’ s Disease Pathogenesis. Acta Neuropathol. 2019, 137, 103– 120, DOI: 10.1007/s00401-018-1907-yGoogle Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslCjtrjJ&md5=d54754b630183e9c7fbea6a8ff763c49Picomolar concentrations of oligomeric alpha-synuclein sensitizes TLR4 to play an initiating role in Parkinson's disease pathogenesisHughes, Craig D.; Choi, Minee L.; Ryten, Mina; Hopkins, Lee; Drews, Anna; Botia, Juan A.; Iljina, Maria; Rodrigues, Magarida; Gagliano, Sarah A.; Gandhi, Sonia; Bryant, Clare; Klenerman, DavidActa Neuropathologica (2019), 137 (1), 103-120CODEN: ANPTAL; ISSN:0001-6322. (Springer)Despite the wealth of genomic and transcriptomic data in Parkinson's disease (PD), the initial mol. events are unknown. Using LD score regression anal., we show significant enrichment in PD heritability within regulatory sites for LPS-activated monocytes and that TLR4 expression is highest within human substantia nigra, the most affected brain region, suggesting a role for TLR4 inflammatory responses. We then performed extended incubation of cells with physiol. concns. of small alpha-synuclein oligomers observing the development of a TLR4-dependent sensitized inflammatory response with time, including TNF-α prodn. ROS and cell death in primary neuronal cultures were significantly reduced by TLR4 antagonists revealing that an indirect inflammatory mechanism involving cytokines produced by glial cells makes a major contribution to neuronal death. Prolonged exposure to low levels of alpha-synuclein oligomers sensitizes TLR4 responsiveness in astrocytes and microglial, explaining how they become pro-inflammatory, and may be an early causative event in PD.
- 13Li, B.; Ge, P.; Murray, K. A.; Sheth, P.; Zhang, M.; Nair, G.; Sawaya, M. R.; Shin, W. S.; Boyer, D. R.; Ye, S.; Eisenberg, D. S.; Zhou, Z. H.; Jiang, L. Cryo-EM of Full-Length α-Synuclein Reveals Fibril Polymorphs with a Common Structural Kernel. Nat. Commun. 2018, 9, 3609 DOI: 10.1038/s41467-018-05971-2Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3c3mtlCmsw%253D%253D&md5=e3cd60ca2cf31d76e4d33fccd8bfa683Cryo-EM of full-length α-synuclein reveals fibril polymorphs with a common structural kernelLi Binsen; Sheth Phorum; Nair Gayatri; Shin Woo Shik; Jiang Lin; Ge Peng; Ye Shulin; Zhou Z Hong; Murray Kevin A; Zhang Meng; Sawaya Michael R; Boyer David R; Eisenberg David S; Zhou Z HongNature communications (2018), 9 (1), 3609 ISSN:.α-Synuclein (aSyn) fibrillar polymorphs have distinct in vitro and in vivo seeding activities, contributing differently to synucleinopathies. Despite numerous prior attempts, how polymorphic aSyn fibrils differ in atomic structure remains elusive. Here, we present fibril polymorphs from the full-length recombinant human aSyn and their seeding capacity and cytotoxicity in vitro. By cryo-electron microscopy helical reconstruction, we determine the structures of the two predominant species, a rod and a twister, both at 3.7 ÅA resolution. Our atomic models reveal that both polymorphs share a kernel structure of a bent β-arch, but differ in their inter-protofilament interfaces. Thus, different packing of the same kernel structure gives rise to distinct fibril polymorphs. Analyses of disease-related familial mutations suggest their potential contribution to the pathogenesis of synucleinopathies by altering population distribution of the fibril polymorphs. Drug design targeting amyloid fibrils in neurodegenerative diseases should consider the formation and distribution of concurrent fibril polymorphs.
- 14Guerrero-Ferreira, R.; Taylor, N. M. I.; Mona, D.; Ringler, P.; Lauer, M. E.; Riek, R.; Britschgi, M.; Stahlberg, H. Cryo-EM Structure of Alpha-Synuclein Fibrils. eLife 2018, 7, e36402 DOI: 10.7554/eLife.36402Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlyrtL3F&md5=c1f72f2655276d3eb3ec4e030ff2cdcaCryo-EM structure of alpha-synuclein fibrilsGuerrero-Ferreira, Ricardo; Taylor, Nicholas M. I.; Mona, Daniel; Ringler, Philippe; Lauer, Matthias E.; Riek, Roland; Britschgi, Markus; Stahlberg, HenningeLife (2018), 7 (), e36402/1-e36402/18CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Parkinson's disease is a progressive neuropathol. disorder that belongs to the class of synucleinopathies, in which the protein alpha-synuclein is found at abnormally high concns. in affected neurons. Its hallmark are intracellular inclusions called Lewy bodies and Lewy neurites. We here report the structure of cytotoxic alpha-synuclein fibrils (residues 1-121), detd. by cryo-electron microscopy at a resoln. of 3.4 Å . Two protofilaments form a polar fibril composed of staggered b-strands. The backbone of residues 38 to 95, including the fibril core and the non-amyloid component region, are well resolved in the EM map. Residues 50-57, contg. three of the mutation sites assocd. with familial synucleinopathies, form the interface between the two protofilaments and contribute to fibril stability. A hydrophobic cleft at one end of the fibril may have implications for fibril elongation, and invites for the design of mols. for diagnosis and treatment of synucleinopathies.
- 15Li, Y.; Zhao, C.; Luo, F.; Liu, Z.; Gui, X.; Luo, Z.; Zhang, X.; Li, D.; Liu, C.; Li, X. Amyloid Fibril Structure of α-Synuclein Determined by Cryo-Electron Microscopy. Cell Res. 2018, 28, 897– 903, DOI: 10.1038/s41422-018-0075-xGoogle Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVahtbvL&md5=053ffd5d6d3beca7c9a3fa322d243b83Amyloid fibril structure of α-synuclein determined by cryo-electron microscopyLi, Yaowang; Zhao, Chunyu; Luo, Feng; Liu, Zhenying; Gui, Xinrui; Luo, Zhipu; Zhang, Xiang; Li, Dan; Liu, Cong; Li, XuemingCell Research (2018), 28 (9), 897-903CODEN: CREEB6; ISSN:1001-0602. (Nature Research)α-Synuclein (α-syn) amyloid fibrils are the major component of Lewy bodies, which are the pathol. hallmark of Parkinson's disease (PD) and other synucleinopathies. High-resoln. structure of α-syn fibril is important for understanding its assembly and pathol. mechanism. Here, we detd. a fibril structure of full-length α-syn (1-140) at the resoln. of 3.07 Å by cryo-electron microscopy (cryo-EM). The fibrils are cytotoxic, and transmissible to induce endogenous α-syn aggregation in primary neurons. Based on the reconstructed cryo-EM d. map, we were able to unambiguously build the fibril structure comprising residues 37-99. The α-syn amyloid fibril structure shows two protofilaments intertwining along an approx. 21 screw axis into a left-handed helix. Each protofilament features a Greek key-like topol. Remarkably, five out of the six early-onset PD familial mutations are located at the dimer interface of the fibril (H50Q, G51D, and A53T/E) or involved in the stabilization of the protofilament (E46K). Furthermore, these PD mutations lead to the formation of fibrils with polymorphic structures distinct from that of the wild-type. Our study provides mol. insight into the fibrillar assembly of α-syn at the at. level and sheds light on the mol. pathogenesis caused by familial PD mutations of α-syn.
- 16Rodriguez, J. A.; Ivanova, M. I.; Sawaya, M. R.; Cascio, D.; Reyes, F. E.; Shi, D.; Sangwan, S.; Guenther, E. L.; Johnson, L. M.; Zhang, M.; Jiang, L.; Arbing, M. A.; Nannenga, B. L.; Hattne, J.; Whitelegge, J.; Brewster, A. S.; Messerschmidt, M.; Boutet, S.; Sauter, N. K.; Gonen, T.; Eisenberg, D. S. Structure of the Toxic Core of α-Synuclein from Invisible Crystals. Nature 2015, 525, 486– 490, DOI: 10.1038/nature15368Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVOls7zN&md5=a6c64c087d6e5b10e4190bfdf2267665Structure of the toxic core of α-synuclein from invisible crystalsRodriguez, Jose A.; Ivanova, Magdalena I.; Sawaya, Michael R.; Cascio, Duilio; Reyes, Francis E.; Shi, Dan; Sangwan, Smriti; Guenther, Elizabeth L.; Johnson, Lisa M.; Zhang, Meng; Jiang, Lin; Arbing, Mark A.; Nannenga, Brent L.; Hattne, Johan; Whitelegge, Julian; Brewster, Aaron S.; Messerschmidt, Marc; Boutet, Sebastien; Sauter, Nicholas K.; Gonen, Tamir; Eisenberg, David S.Nature (London, United Kingdom) (2015), 525 (7570), 486-490CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)α-Synuclein is the main component of Lewy bodies, the neuron-assocd. aggregates seen in Parkinson disease and other neurodegenerative pathologies. An 11-residue segment, which the authors term NACore, appears to be responsible for amyloid formation and cytotoxicity of human α-synuclein. Here, the authors describe crystals of NACore that have dimensions smaller than the wavelength of visible light and thus are invisible by optical microscopy. As the crystals are thousands of times too small for structure detn. by synchrotron x-ray diffraction, the authors used micro-electron diffraction to det. the structure at at. resoln. The 1.4-Å-resoln. structure demonstrated that this method can det. previously unknown protein structures and here yielded, to the authors' knowledge, the highest resoln. achieved by any cryo-electron microscopy method to date. The structure exhibited protofibrils built of pairs of face-to-face β-sheets. X-ray fiber diffraction patterns showed the similarity of NACore to toxic fibrils of full-length α-synuclein. The NACore structure, together with that of a 2nd segment, inspired a model for most of the ordered portion of the toxic, full-length α-synuclein fibril, presenting opportunities for the design of inhibitors of α-synuclein fibrils.
- 17Schweighauser, M.; Shi, Y.; Tarutani, A.; Kametani, F.; Murzin, A. G.; Ghetti, B.; Matsubara, T.; Tomita, T.; Ando, T.; Hasegawa, K.; Murayama, S.; Yoshida, M.; Hasegawa, M.; Scheres, S. H. W.; Goedert, M. Structures of α-Synuclein Filaments from Multiple System Atrophy. Nature 2020, 585, 464– 469, DOI: 10.1038/s41586-020-2317-6Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVantbjK&md5=2227413593f48413b7129ff9d8128e6eStructures of α-synuclein filaments from multiple system atrophySchweighauser, Manuel; Shi, Yang; Tarutani, Airi; Kametani, Fuyuki; Murzin, Alexey G.; Ghetti, Bernardino; Matsubara, Tomoyasu; Tomita, Taisuke; Ando, Takashi; Hasegawa, Kazuko; Murayama, Shigeo; Yoshida, Mari; Hasegawa, Masato; Scheres, Sjors H. W.; Goedert, MichelNature (London, United Kingdom) (2020), 585 (7825), 464-469CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Synucleinopathies, which include multiple system atrophy (MSA), Parkinson's disease, Parkinson's disease with dementia and dementia with Lewy bodies (DLB), are human neurodegenerative diseases1. Existing treatments are at best symptomatic. These diseases are characterized by the presence of, and believed to be caused by the formation of, filamentous inclusions of α-synuclein in brain cells2,3. However, the structures of α-synuclein filaments from the human brain are unknown. Here, using cryo-electron microscopy, we show that α-synuclein inclusions from the brains of individuals with MSA are made of two types of filament, each of which consists of two different protofilaments. In each type of filament, non-proteinaceous mols. are present at the interface of the two protofilaments. Using two-dimensional class averaging, we show that α-synuclein filaments from the brains of individuals with MSA differ from those of individuals with DLB, which suggests that distinct conformers or strains characterize specific synucleinopathies. As is the case with tau assemblies4-9, the structures of α-synuclein filaments extd. from the brains of individuals with MSA differ from those formed in vitro using recombinant proteins, which has implications for understanding the mechanisms of aggregate propagation and neurodegeneration in the human brain. These findings have diagnostic and potential therapeutic relevance, esp. because of the unmet clin. need to be able to image filamentous α-synuclein inclusions in the human brain.
- 18Sangwan, S.; Sahay, S.; Murray, K. A.; Morgan, S.; Guenther, E. L.; Jiang, L.; Williams, C. K.; Vinters, H. V.; Goedert, M.; Eisenberg, D. S. Inhibition of Synucleinopathic Seeding by Rationally Designed Inhibitors. eLife 2020, 9, e46775 DOI: 10.7554/eLife.46775Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlarsLrM&md5=1161e8cdb0482e8bee3cd6253a3cd4f5Inhibition of synucleinopathic seeding by rationally designed inhibitorsSangwan, Smriti; Sahay, Shruti; Murray, Kevin A.; Morgan, Sophie; Guenther, Elizabeth L.; Jiang, Lin; Williams, Christopher K.; Vinters, Harry V.; Goedert, Michel; Eisenberg, David S.eLife (2020), 9 (), e46775CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Seeding, in the context of amyloid disease, is the sequential transfer of pathogenic protein aggregates from cell-to-cell within affected tissues. The structure of pathogenic seeds provides the mol. basis and enables rapid conversion of sol. protein into fibrils. To date, there are no inhibitors that specifically target seeding of Parkinson's disease (PD)-assocd. asynuclein (a-syn) fibrils, in part, due to lack of information of the structural properties of pathol. seeds. Here we design small peptidic inhibitors based on the at. structure of the core of a-syn fibrils. The inhibitors prevent a-syn aggregation in vitro and in cell culture models with binding affinities of 0.5 mM to a-syn fibril seeds. The inhibitors also show efficacy in preventing seeding by human patient-derived a-syn fibrils. Our results suggest that pathogenic seeds of a-syn contain steric zippers and suggest a therapeutic approach targeted at the spread and progression that may be applicable for PD and related synucleinopathies.
- 19Priss, A.; Afitska, K.; Galkin, M.; Yushchenko, D. A.; Shvadchak, V. V. Rationally Designed Protein-Based Inhibitor of α-Synuclein Fibrillization in Cells. J. Med. Chem. 2021, 64, 6827– 6837, DOI: 10.1021/acs.jmedchem.1c00086Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVCiu7%252FM&md5=42e14747550775b61e5c84d161b58efaRationally Designed Protein-Based Inhibitor of α-Synuclein Fibrillization in CellsPriss, Anastasiia; Afitska, Kseniia; Galkin, Maksym; Yushchenko, Dmytro A.; Shvadchak, Volodymyr V.Journal of Medicinal Chemistry (2021), 64 (10), 6827-6837CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Misfolding of the neuronal protein α-synuclein (αSyn) into amyloid fibrils is involved in the development of Parkinson's disease (PD), and inhibition of this process is considered to be a promising therapeutic approach. In this work, we engineered protein inhibitors that bind to fibrils with higher affinity than the monomeric αSyn. They were developed based on the recent structural data of the αSyn fibrils and were shown to prevent fibril elongation upon binding to fibril ends. These inhibitors are highly selective to the misfolded αSyn, nontoxic, and active in cytosol in small concns. The best-performing inhibitor shows IC50 ~ 10 nM in a cell-based assay, which corresponds to the ~ 1:60 molar ratio to αSyn. It can suppress the formation of αSyn aggregates in cells that can be potentially used to slow down the spreading of the pathol. aggregates from cell to cell during the course of the PD.
- 20Gao, L.; Wang, W.; Wang, X.; Yang, F.; Xie, L.; Shen, J.; Brimble, M. A.; Xiao, Q.; Yao, S. Q. Fluorescent Probes for Bioimaging of Potential Biomarkers in Parkinson’s Disease. Chem. Soc. Rev. 2021, 50, 1219– 1250, DOI: 10.1039/d0cs00115eGoogle Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFSgtb%252FE&md5=ab063111c2d1f8f66453b1bff9c5becdFluorescent probes for bioimaging of potential biomarkers in Parkinson's diseaseGao, Liqian; Wang, Wei; Wang, Xuan; Yang, Fen; Xie, Liuxing; Shen, Jun; Brimble, Margaret A.; Xiao, Qicai; Yao, Shao Q.Chemical Society Reviews (2021), 50 (2), 1219-1250CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Parkinson's disease (PD), as the second most common neurodegenerative disease, is caused by complex pathol. processes and currently remains very difficult to treat. PD brings great distress to patients and imposes a heavy economic burden on society. The no. of PD patients is growing as the aging population increases worldwide. Therefore, it is crucial to develop new tools for aiding the early diagnosis and treatment of PD. The significant pathol. features involved in PD include the abnormal accumulation of α-synuclein, metal ion dyshomeostasis, oxidative stress, mitochondrial dysfunction and neurotransmitter deficiencies. In recent years, fluorescent probes have emerged as a powerful bioimaging tool with potential to help understand the pathol. processes of PD via the detection and monitoring of pathol. features. In this review, we comprehensively summarize the design and working mechanisms of fluorescent probes along with their applications in the detection of various PD biomarkers. We also discuss the current limitations of fluorescent probes and provide perspectives on how these limitations can be overcome to develop better fluorescent probes suitable for application in clin. trials in the future. We hope that this review provides valuable information and guidance for the development of new fluorescent probes that can be used clin. in the early diagnosis of PD and contributes to the development of efficient PD drugs in the future.
- 21Ferrie, J. J.; Lengyel-Zhand, Z.; Janssen, B.; Lougee, M. G.; Giannakoulias, S.; Hsieh, C.-J.; Pagar, V. V.; Weng, C.-C.; Xu, H.; Graham, T. J. A.; Lee, V. M.-Y.; Mach, R. H.; Petersson, E. J. Identification of a Nanomolar Affinity α-Synuclein Fibril Imaging Probe by Ultra-High Throughput in Silico Screening. Chem. Sci. 2020, 11, 12746– 12754, DOI: 10.1039/d0sc02159hGoogle Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsl2ktb%252FJ&md5=9a34fe33b5d896d3bf60cbe264d74452Identification of a nanomolar affinity alpha-synuclein fibril imaging probe by ultra-high throughput in silico screeningFerrie, John J.; Lengyel-Zhand, Zsofia; Janssen, Bieneke; Lougee, Marshall G.; Giannakoulias, Sam; Hsieh, Chia-Ju; Pagar, Vinayak Vishnu; Weng, Chi-Chang; Xu, Hong; Graham, Thomas J. A.; Lee, Virginia M.-Y.; Mach, Robert H.; Petersson, E. JamesChemical Science (2020), 11 (47), 12746-12754CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Small mols. that bind with high affinity and specificity to fibrils of the α-synuclein (αS) protein have the potential to serve as positron emission tomog. (PET) imaging probes to aid in the diagnosis of Parkinson's disease and related synucleinopathies. To identify such mols., we employed an ultra-high throughput in silico screening strategy using idealized pseudo-ligands termed exemplars to identify compds. for exptl. binding studies. For the top hit from this screen, we used photo-crosslinking to confirm its binding site and studied the structure-activity relationship of its analogs to develop multiple mols. with nanomolar affinity for αS fibrils and moderate specificity for α;S over Aβfibrils. Lastly, we demonstrated the potential of the lead analog as an imaging probe by measuring binding to αS-enriched homogenates from mouse brain tissue using a radiolabeled analog of the identified mols. This study demonstrates the validity of our powerful new approach to the discovery of PET probes for challenging mol. targets.
- 22Lengyel-Zhand, Z.; Ferrie, J. J.; Janssen, B.; Hsieh, C. J.; Graham, T.; Xu, K. Y.; Haney, C. M.; Lee, V. M. Y.; Trojanowski, J. Q.; Petersson, E. J.; Mach, R. H. Synthesis and Characterization of High Affinity Fluorogenic α-Synuclein Probes. Chem. Commun. 2020, 56, 3567– 3570, DOI: 10.1039/c9cc09849fGoogle Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjsF2ntrw%253D&md5=a2e24885326f473f4ede5c722d027692Synthesis and characterization of high affinity fluorogenic α-synuclein probesLengyel-Zhand, Zsofia; Ferrie, John J.; Janssen, Bieneke; Hsieh, Chia-Ju; Graham, Thomas; Xu, Kui-ying; Haney, Conor M.; Lee, Virginia M.-Y.; Trojanowski, John Q.; Petersson, E. James; Mach, Robert H.Chemical Communications (Cambridge, United Kingdom) (2020), 56 (24), 3567-3570CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)Fluorescent small mols. are powerful tools for imaging α-synuclein pathol. in vitro and in vivo. In this work, we explore benzofuranone as a potential scaffold for the design of fluorescent α-synuclein probes. These compds. have high affinity for α-synuclein, show fluorescent turn-on upon binding to fibrils, and display different binding to Lewy bodies, Lewy neurites and glial cytoplasmic inclusion pathologies in post-mortem brain tissue. These studies not only reveal the potential of benzofuranone compds. as α-synuclein specific fluorescent probes, but also have implications for the ways in which α-synucleinopathies are conformationally different and display distinct small mol. binding sites.
- 23Hsieh, C. J.; Ferrie, J. J.; Xu, K.; Lee, I.; Graham, T. J. A.; Tu, Z.; Yu, J.; Dhavale, D.; Kotzbauer, P.; Petersson, E. J.; Mach, R. H. Alpha Synuclein Fibrils Contain Multiple Binding Sites for Small Molecules. ACS Chem. Neurosci. 2018, 9, 2521– 2527, DOI: 10.1021/acschemneuro.8b00177Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXptlajurs%253D&md5=f16af627b180f8b1cac79ff08bebc643Alpha Synuclein Fibrils Contain Multiple Binding Sites for Small MoleculesHsieh, Chia-Ju; Ferrie, John J.; Xu, Kuiying; Lee, Iljung; Graham, Thomas J. A.; Tu, Zhude; Yu, Jennifer; Dhavale, Dhruva; Kotzbauer, Paul; Petersson, E. James; Mach, Robert H.ACS Chemical Neuroscience (2018), 9 (11), 2521-2527CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)The fibrillary aggregation of the α-synuclein (Asyn) is a hallmark of Parkinson's disease, and the identification of small mol. binding sites on fibrils is essential to the development of diagnostic imaging probes. Here, a series of mol. modeling, photoaffinity labeling, mass spectrometry,, and radioligand binding studies were conducted on Asyn fibrils. The results of these studies revealed the presence of 3 different binding sites within fibrillar Asyn capable of binding small mols. with moderate to high affinity. A knowledge of the amino acid residues in these binding sites will be important in the design of high affinity probes capable of imaging fibrillary species of Asyn.
- 24Eberling, J. L.; Dave, K. D.; Frasier, M. A. α-Synuclein Imaging: A Critical Need for Parkinson’s Disease Research. J. Parkinson’s Dis. 2013, 3, 565– 567, DOI: 10.3233/JPD-130247Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvV2iur%252FO&md5=74cb27856e9d5c7b593b0c357ef7138aα-synuclein Imaging: A Critical Need for Parkinson's Disease ResearchEberling, Jamie L.; Dave, Kuldip D.; Frasier, Mark A.Journal of Parkinson's Disease (2013), 3 (4), 565-567CODEN: JPDOAP; ISSN:1877-7171. (IOS Press)A review. The development of an α-synuclein imaging agent could be transformative for Parkinson's disease research and drug development. The ability to image α-synuclein in the brain would enable tracking of the degree and location of pathol. over time and monitoring of therapies aimed at reducing α-synuclein levels. The Michael J. Fox Foundation has assembled a consortium of researchers to develop an α-synuclein radiotracer for use in positron emission tomog. (PET) imaging studies. While this poses a no. of challenges they should not be insurmountable and lessons learned from the development of tau radiotracers should provide valuable insights.
- 25Kulenkampff, K.; Wolf Perez, A. M.; Sormanni, P.; Habchi, J.; Vendruscolo, M. Quantifying Misfolded Protein Oligomers as Drug Targets and Biomarkers in Alzheimer and Parkinson Diseases. Nat. Rev. Chem. 2021, 5, 277– 294, DOI: 10.1038/s41570-021-00254-9Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXosFers7s%253D&md5=d7a63b934405847347c68bd061ab34e9Quantifying misfolded protein oligomers as drug targets and biomarkers in Alzheimer and Parkinson diseasesKulenkampff, Klara; Wolf Perez, Adriana-M.; Sormanni, Pietro; Habchi, Johnny; Vendruscolo, MicheleNature Reviews Chemistry (2021), 5 (4), 277-294CODEN: NRCAF7; ISSN:2397-3358. (Nature Portfolio)A review. Protein misfolding and aggregation are characteristic of a wide range of neurodegenerative disorders, including Alzheimer and Parkinson diseases. A hallmark of these diseases is the aggregation of otherwise sol. and functional proteins into amyloid aggregates. Although for many decades such amyloid deposits have been thought to be responsible for disease progression, it is now increasingly recognized that the misfolded protein oligomers formed during aggregation are, instead, the main agents causing pathol. processes. These oligomers are transient and heterogeneous, which makes it difficult to detect and quantify them, generating confusion about their exact role in disease. The lack of suitable methods to address these challenges has hampered efforts to investigate the mol. mechanisms of oligomer toxicity and to develop oligomer-based diagnostic and therapeutic tools to combat protein misfolding diseases. In this Review, we describe methods to quantify misfolded protein oligomers, with particular emphasis on diagnostic applications as disease biomarkers and on therapeutic applications as target biomarkers. The development of these methods is ongoing, and we discuss the challenges that remain to be addressed to establish measurement tools capable of overcoming existing limitations and to meet present needs.
- 26Fayyad, M.; Salim, S.; Majbour, N.; Erskine, D.; Stoops, E.; Mollenhauer, B.; El-Agnaf, O. M. A. Parkinson’s Disease Biomarkers Based on α-Synuclein. J. Neurochem. 2019, 150, 626– 636, DOI: 10.1111/jnc.14809Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsVCmt7nF&md5=28fbc154ad78042e2a331e234f525d0bParkinson's disease biomarkers based on α-synucleinFayyad, Muneera; Salim, Safa; Majbour, Nour; Erskine, Daniel; Stoops, Erik; Mollenhauer, Brit; El-Agnaf, Omar M. A.Journal of Neurochemistry (2019), 150 (5), 626-636CODEN: JONRA9; ISSN:0022-3042. (Wiley-Blackwell)A review. Parkinson's disease is the second most common neurodegenerative disorder after Alzheimer's disease and is estd. to affect approx. 1-4% of individuals aged over 60 years old. Although considerable efforts have been invested into developing disease-modifying therapies for Parkinson's disease, such efforts have been confounded by the difficulty in accurately diagnosing Parkinson's disease during life to enable accurate patient stratification for clin. trialling of candidate therapeutics. Therefore, the search for effective biomarkers that can be accurately evaluated during life with non-invasive means is a pressing issue in the field. Since the discovery of α-synuclein (α-syn) as a protein linked to a familial form of Parkinson's disease, later identified as the major protein component of the neuropathol. hallmark of idiopathic Parkinson's disease, considerable interest has focused on this protein and its distinct conformers. We describe here the progress that has been made in the area of Parkinson's disease biomarker discovery with a focus on α-synuclein. In particular, we highlight the novel assays that have been employed and the increasing complexity in evaluating α-synuclein with regard to the considerable diversity of conformers that exist in the biofluids and peripheral tissues under disease conditions.
- 27Brown, J. W. P.; Buell, A. K.; Michaels, T. C. T.; Meisl, G.; Carozza, J.; Flagmeier, P.; Vendruscolo, M.; Knowles, T. P. J.; Dobson, C. M.; Galvagnion, C. β-Synuclein Suppresses Both the Initiation and Amplification Steps of α-Synuclein Aggregation via Competitive Binding to Surfaces. Sci. Rep. 2016, 6, 36010 DOI: 10.1038/srep36010Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVSisbjJ&md5=aa6a38bfdb70917406e08a40448bfc01β-Synuclein suppresses both the initiation and amplification steps of α-synuclein aggregation via competitive binding to surfacesBrown, James W. P.; Buell, Alexander K.; Michaels, Thomas C. T.; Meisl, Georg; Carozza, Jacqueline; Flagmeier, Patrick; Vendruscolo, Michele; Knowles, Tuomas P. J.; Dobson, Christopher M.; Galvagnion, CelineScientific Reports (2016), 6 (), 36010CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)α-Synuclein is an intrinsically disordered protein that is assocd. with the pathogenesis of Parkinson's disease through the processes involved in the formation of amyloid fibrils. α and β-synuclein are homologous proteins found at comparable levels in presynaptic terminals but β-synuclein has a greatly reduced propensity to aggregate and indeed has been found to inhibit α-synuclein aggregation. In this paper, we describe how sequence differences between α- and β-synuclein affect individual microscopic processes in amyloid formation. In particular, we show that β-synuclein strongly suppresses both lipid-induced aggregation and secondary nucleation of α-synuclein by competing for binding sites at the surfaces of lipid vesicles and fibrils, resp. These results suggest that β-synuclein can act as a natural inhibitor of α-synuclein aggregation by reducing both the initiation of its self-assembly and the proliferation of its aggregates.
- 28Staats, R.; Michaels, T. C. T.; Flagmeier, P.; Chia, S.; Horne, R. I.; Habchi, J.; Linse, S.; Knowles, T. P. J.; Dobson, C. M.; Vendruscolo, M. Screening of Small Molecules Using the Inhibition of Oligomer Formation in α-Synuclein Aggregation as a Selection Parameter. Commun. Chem. 2020, 3, 191, DOI: 10.1038/s42004-020-00412-yGoogle Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVerur3E&md5=3dedbd30c621b0c6a777f3e6ad7c503eScreening of small molecules using the inhibition of oligomer formation in α-synuclein aggregation as a selection parameterStaats, Roxine; Michaels, Thomas C. T.; Flagmeier, Patrick; Chia, Sean; Horne, Robert I.; Habchi, Johnny; Linse, Sara; Knowles, Tuomas P. J.; Dobson, Christopher M.; Vendruscolo, MicheleCommunications Chemistry (2020), 3 (1), 191CODEN: CCOHCT; ISSN:2399-3669. (Nature Research)The aggregation of α-synuclein is a central event in Parkinsons disease and related synucleinopathies. Since pharmacol. targeting this process, however, has not yet resulted in approved disease-modifying treatments, there is an unmet need of developing novel methods of drug discovery. In this context, the use of chem. kinetics has recently enabled accurate quantifications of the microscopic steps leading to the proliferation of protein misfolded oligomers. As these species are highly neurotoxic, effective therapeutic strategies may be aimed at reducing their nos. Here, we exploit this quant. approach to develop a screening strategy that uses the reactive flux toward α-synuclein oligomers as a selection parameter. Using this approach, we evaluate the efficacy of a library of flavone derivs., identifying apigenin as a compd. that simultaneously delays and reduces the formation of α-synuclein oligomers. These results demonstrate a compd. selection strategy based on the inhibition of the formation of α-synuclein oligomers, which may be key in identifying small mols. in drug discovery pipelines for diseases assocd. with α-synuclein aggregation.
- 29Perni, M.; Flagmeier, P.; Limbocker, R.; Cascella, R.; Aprile, F. A.; Galvagnion, C.; Heller, G. T.; Meisl, G.; Chen, S. W.; Kumita, J. R.; Challa, P. K.; Kirkegaard, J. B.; Cohen, S. I. A.; Mannini, B.; Barbut, D.; Nollen, E. A. A.; Cecchi, C.; Cremades, N.; Knowles, T. P. J.; Chiti, F.; Zasloff, M.; Vendruscolo, M.; Dobson, C. M. Multistep Inhibition of α-Synuclein Aggregation and Toxicity in Vitro and in Vivo by Trodusquemine. ACS Chem. Biol. 2018, 13, 2308– 2319, DOI: 10.1021/acschembio.8b00466Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1SqtLfN&md5=7bcab9b174a7dd4958233f8d8457834eMultistep Inhibition of α-Synuclein Aggregation and Toxicity in Vitro and in Vivo by TrodusqueminePerni, Michele; Flagmeier, Patrick; Limbocker, Ryan; Cascella, Roberta; Aprile, Francesco A.; Galvagnion, Celine; Heller, Gabriella T.; Meisl, Georg; Chen, Serene W.; Kumita, Janet R.; Challa, Pavan K.; Kirkegaard, Julius B.; Cohen, Samuel I. A.; Mannini, Benedetta; Barbut, Denise; Nollen, Ellen A. A.; Cecchi, Cristina; Cremades, Nunilo; Knowles, Tuomas P. J.; Chiti, Fabrizio; Zasloff, Michael; Vendruscolo, Michele; Dobson, Christopher M.ACS Chemical Biology (2018), 13 (8), 2308-2319CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The aggregation of α-synuclein, an intrinsically disordered protein that is highly abundant in neurons, is closely assocd. with the onset and progression of Parkinson's disease. We have shown previously that the aminosterol squalamine can inhibit the lipid induced initiation process in the aggregation of α-synuclein, and we report here that the related compd. trodusquemine is capable of inhibiting not only this process but also the fibril-dependent secondary pathways in the aggregation reaction. We further demonstrate that trodusquemine can effectively suppress the toxicity of α-synuclein oligomers in neuronal cells, and that its administration, even after the initial growth phase, leads to a dramatic redn. in the no. of α-synuclein inclusions in a Caenorhabditis elegans model of Parkinson's disease, eliminates the related muscle paralysis, and increases lifespan. On the basis of these findings, we show that trodusquemine is able to inhibit multiple events in the aggregation process of α-synuclein and hence to provide important information about the link between such events and neurodegeneration, as it is initiated and progresses. Particularly in the light of the previously reported ability of trodusquemine to cross the blood-brain barrier and to promote tissue regeneration, the present results suggest that this compd. has the potential to be an important therapeutic candidate for Parkinson's disease and related disorders.
- 30Pujols, J.; Peña-Díaz, S.; Pallarès, I.; Ventura, S. Chemical Chaperones as Novel Drugs for Parkinson’s Disease. Trends Mol. Med. 2020, 26, 408– 421, DOI: 10.1016/j.molmed.2020.01.005Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlGrt7w%253D&md5=e56194e5e4f70b2fec1fff492fd62919Chemical Chaperones as Novel Drugs for Parkinson's DiseasePujols, Jordi; Pena-Diaz, Samuel; Pallares, Irantzu; Ventura, SalvadorTrends in Molecular Medicine (2020), 26 (4), 408-421CODEN: TMMRCY; ISSN:1471-4914. (Elsevier Ltd.)A review. Parkinson's disease (PD) is characterized by progressive loss of dopaminergic neurons and the accumulation of deposits of α-synuclein (α-syn) in the brain. The pivotal role of α-syn aggregation in PD makes it an attractive target for potential disease-modifying therapies. However, the disordered nature of the protein, its multistep aggregation mechanism, and the lack of structural information on intermediate species complicate the discovery of modulators of α-syn amyloid deposition. Despite these difficulties, small mols. have been shown to block the misfolding and aggregation of α-syn, and can even disentangle mature α-syn amyloid fibrils. In this review we provide an updated overview of these leading small compds. and discuss how these chem. chaperones hold great promise to alter the course of PD progression.
- 31Gaspar, R.; Meisl, G.; Buell, A. K.; Young, L.; Kaminski, C. F.; Knowles, T. P. J.; Sparr, E.; Linse, S. Secondary Nucleation of Monomers on Fibril Surface Dominates α-Synuclein Aggregation and Provides Autocatalytic Amyloid Amplification. Q. Rev. Biophys. 2017, 50, e6 DOI: 10.1017/S0033583516000172Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXkvVaqtrw%253D&md5=1b0069435bf692abc35d3b62a6cbac51Secondary nucleation of monomers on fibril surface dominates α-synuclein aggregation and provides autocatalytic amyloid amplificationGaspar, Ricardo; Meisl, Georg; Buell, Alexander K.; Young, Laurence; Kaminski, Clemens F.; Knowles, Tuomas P. J.; Sparr, Emma; Linse, SaraQuarterly Reviews of Biophysics (2017), 50 (), e6/1-e6/12CODEN: QURBAW; ISSN:0033-5835. (Cambridge University Press)Parkinson's disease (PD) is characterized by proteinaceous aggregates named Lewy Bodies and Lewy Neurites contg. α-synuclein fibrils. The underlying aggregation mechanism of this protein is dominated by a secondary process at mildly acidic pH, as in endosomes and other organelles. This effect manifests as a strong acceleration of the aggregation in the presence of seeds and a weak dependence of the aggregation rate on monomer concn. The mol. mechanism underlying this process could be nucleation of monomers on fibril surfaces or fibril fragmentation. Here, we aim to distinguish between these mechanisms. The nature of the secondary processes was investigated using differential sedimentation anal., trap and seed expts., quartz crystal microbalance expts. and super-resoln. microscopy. The results identify secondary nucleation of monomers on the fibril surface as the dominant secondary process leading to rapid generation of new aggregates, while no significant contribution from fragmentation was found. The newly generated oligomeric species quickly elongate to further serve as templates for secondary nucleation and this may have important implications in the spreading of PD.
- 32Michaels, T. C. T.; Lazell, H. W.; Arosio, P.; Knowles, T. P. J. Dynamics of Protein Aggregation and Oligomer Formation Governed by Secondary Nucleation. J. Chem. Phys. 2015, 143, 054901 DOI: 10.1063/1.4927655Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht12gtL7P&md5=43386e2cdf09735afe1c274bca781845Dynamics of protein aggregation and oligomer formation governed by secondary nucleationMichaels, Thomas C. T.; Lazell, Hamish W.; Arosio, Paolo; Knowles, Tuomas P. J.Journal of Chemical Physics (2015), 143 (5), 054901/1-054901/15CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The formation of aggregates in many protein systems can be significantly accelerated by secondary nucleation, a process where existing assemblies catalyze the nucleation of new species. In particular, secondary nucleation has emerged as a central process controlling the proliferation of many filamentous protein structures, including mol. species related to diseases such as sickle cell anemia and a range of neurodegenerative conditions. Increasing evidence suggests that the phys. size of protein filaments plays a key role in detg. their potential for deleterious interactions with living cells, with smaller aggregates of misfolded proteins, oligomers, being particularly toxic. It is thus crucial to progress towards an understanding of the factors that control the sizes of protein aggregates. However, the influence of secondary nucleation on the time evolution of aggregate size distributions has been challenging to quantify. This difficulty originates in large part from the fact that secondary nucleation couples the dynamics of species distant in size space. Here, we approach this problem by presenting an anal. treatment of the master equation describing the growth kinetics of linear protein structures proliferating through secondary nucleation and provide closed-form expressions for the temporal evolution of the resulting aggregate size distribution. We show how the availability of anal. solns. for the full filament distribution allows us to identify the key phys. parameters that control the sizes of growing protein filaments. Furthermore, we use these results to probe the dynamics of the populations of small oligomeric species as they are formed through secondary nucleation and discuss the implications of our work for understanding the factors that promote or curtail the prodn. of these species with a potentially high deleterious biol. activity. (c) 2015 American Institute of Physics.
- 33Chia, S.; Habchi, J.; Michaels, T. C. T.; Cohen, S. I. A.; Linse, S.; Dobson, C. M.; Knowles, T. P. J.; Vendruscolo, M. SAR by Kinetics for Drug Discovery in Protein Misfolding Diseases. Proc. Natl. Acad. Sci. U.S.A. 2018, 115, 10245– 10250, DOI: 10.1073/pnas.1807884115Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFOntLnP&md5=13ac0b5763f8a9779de80af1c9700978SAR by kinetics for drug discovery in protein misfolding diseasesChia, Sean; Habchi, Johnny; Michaels, Thomas C. T.; Cohen, Samuel I. A.; Linse, Sara; Dobson, Christopher M.; Knowles, Tuomas P. J.; Vendruscolo, MicheleProceedings of the National Academy of Sciences of the United States of America (2018), 115 (41), 10245-10250CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)To develop effective therapeutic strategies for protein misfolding diseases, a promising route is to identify compds. that inhibit the formation of protein oligomers. To achieve this goal, the authors report a structure-activity relation (SAR) approach based on chem. kinetics to est. quant. how small mols. modify the reactive flux toward oligomers. The authors use this est. to derive chem. rules in the case of the amyloid beta peptide (Aβ), which the authors then exploit to optimize starting compds. to curtail Aβ oligomer formation. The authors demonstrate this approach by converting an inactive rhodanine compd. into an effective inhibitor of Aβ oligomer formation by generating chem. derivs. in a systematic manner. These results provide an initial demonstration of the potential of drug discovery strategies based on targeting directly the prodn. of protein oligomers.
- 34Le Guilloux, V.; Schmidtke, P.; Tuffery, P. Fpocket: An Open Source Platform for Ligand Pocket Detection. BMC Bioinf. 2009, 10, S6 DOI: 10.1186/1471-2105-10-168Google ScholarThere is no corresponding record for this reference.
- 35Buell, A. K.; Galvagnion, C.; Gaspar, R.; Sparr, E.; Vendruscolo, M.; Knowles, T. P. J.; Linse, S.; Dobson, C. M. Solution Conditions Determine the Relative Importance of Nucleation and Growth Processes in α-Synuclein Aggregation. Proc. Natl. Acad. Sci. U.S.A. 2014, 111, 7671– 7676, DOI: 10.1073/pnas.1315346111Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnsl2msL4%253D&md5=f08c789180029c972001b80b401cc78eSolution conditions determine the relative importance of nucleation and growth processes in α-synuclein aggregationBuell, Alexander K.; Galvagnion, Celine; Gaspar, Ricardo; Sparr, Emma; Vendruscolo, Michele; Knowles, Tuomas P. J.; Linse, Sara; Dobson, Christopher M.Proceedings of the National Academy of Sciences of the United States of America (2014), 111 (21), 7671-7676CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The formation of amyloid fibrils by the intrinsically disordered protein α-synuclein is a hallmark of Parkinson's disease (PD). To characterize the microscopic steps in the mechanism of aggregation of this protein, the authors used in vitro aggregation assays in the presence of preformed seed fibrils to det. the mol. rate const. of fibril elongation under a range of different conditions. The authors showed that α-synuclein amyloid fibrils grew by monomer and not oligomer addn. and were subject to higher-order assembly processes that decreased their capacity to grow. The authors also found that at neutral pH under quiescent conditions homogeneous primary nucleation and secondary processes, such as fragmentation and surface-assisted nucleation, which can lead to proliferation of the total no. of aggregates, were undetectable. At pH values of <6, however, the rate of secondary nucleation increased dramatically, leading to a completely different balance between the nucleation and growth of aggregates. Thus, at mildly acidic pH values, such as those, e.g., that are present in some intracellular locations, including endosomes and lysosomes, multiplication of aggregates was much faster than at normal physiol. pH values, largely as a consequence of much more rapid secondary nucleation. These findings provide new insights into possible mechanisms of α-synuclein aggregation and aggregate spreading in the context of PD.
- 36Sterling, T.; Irwin, J. J. ZINC 15 - Ligand Discovery for Everyone. J. Chem. Inf. Model. 2015, 55, 2324– 2337, DOI: 10.1021/acs.jcim.5b00559Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhs1OhurbF&md5=e767c26c1d0aff7432c403c59a5d2b8bZINC 15 - Ligand Discovery for EveryoneSterling, Teague; Irwin, John J.Journal of Chemical Information and Modeling (2015), 55 (11), 2324-2337CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Many questions about the biol. activity and availability of small mols. remain inaccessible to investigators who could most benefit from their answers. To narrow the gap between chemoinformatics and biol., we have developed a suite of ligand annotation, purchasability, target, and biol. assocn. tools, incorporated into ZINC and meant for investigators who are not computer specialists. The new version contains over 120 million purchasable "drug-like" compds. - effectively all org. mols. that are for sale - a quarter of which are available for immediate delivery. ZINC connects purchasable compds. to high-value ones such as metabolites, drugs, natural products, and annotated compds. from the literature. Compds. may be accessed by the genes for which they are annotated as well as the major and minor target classes to which those genes belong. It offers new anal. tools that are easy for nonspecialists yet with few limitations for experts. ZINC retains its original 3D roots - all mols. are available in biol. relevant, ready-to-dock formats. ZINC is freely available at http://zinc15.docking.org.
- 37Wager, T. T.; Hou, X.; Verhoest, P. R.; Villalobos, A. Moving beyond Rules: The Development of a Central Nervous System Multiparameter Optimization (CNS MPO) Approach to Enable Alignment of Druglike Properties. ACS Chem. Neurosci. 2010, 1, 435– 449, DOI: 10.1021/cn100008cGoogle Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjvVejtr8%253D&md5=e94998f65a420be6ee4dec4a987cb983Moving beyond Rules: The Development of a Central Nervous System Multiparameter Optimization (CNS MPO) Approach To Enable Alignment of Druglike PropertiesWager, Travis T.; Hou, Xinjun; Verhoest, Patrick R.; Villalobos, AnabellaACS Chemical Neuroscience (2010), 1 (6), 435-449CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)The interplay among commonly used physicochem. properties in drug design was examd. and utilized to create a prospective design tool focused on the alignment of key druglike attributes. Using a set of six physicochem. parameters ((a) lipophilicity, calcd. partition coeff. (ClogP); (b) calcd. distribution coeff. at pH = 7.4 (ClogD); (c) mol. wt. (MW); (d) topol. polar surface area (TPSA); (e) no. of hydrogen bond donors (HBD); (f) most basic center (pKa)), a druglikeness central nervous system multiparameter optimization (CNS MPO) algorithm was built and applied to a set of marketed CNS drugs (N = 119) and Pfizer CNS candidates (N = 108), as well as to a large diversity set of Pfizer proprietary compds. (N = 11 303). The novel CNS MPO algorithm showed that 74% of marketed CNS drugs displayed a high CNS MPO score (MPO desirability score ≥ 4, using a scale of 0-6), in comparison to 60% of the Pfizer CNS candidates. This anal. suggests that this algorithm could potentially be used to identify compds. with a higher probability of successfully testing hypotheses in the clinic. In addn., a relationship between an increasing CNS MPO score and alignment of key in vitro attributes of drug discovery (favorable permeability, P-glycoprotein (P-gp) efflux, metabolic stability, and safety) was seen in the marketed CNS drug set, the Pfizer candidate set, and the Pfizer proprietary diversity set. The CNS MPO scoring function offers advantages over hard cutoffs or utilization of single parameters to optimize structure-activity relationships (SAR) by expanding medicinal chem. design space through a holistic assessment approach. Based on six physicochem. properties commonly used by medicinal chemists, the CNS MPO function may be used prospectively at the design stage to accelerate the identification of compds. with increased probability of success.
- 38Trott, O.; Olson, A. J. AutoDock Vina: Improving the Speed and Accuracy of Docking with a New Scoring Function, Efficient Optimization, and Multithreading. J. Comput. Chem. 2009, 31, 455– 461, DOI: 10.1002/jcc.21334Google ScholarThere is no corresponding record for this reference.
- 39Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: A New Approach for Rapid, Accurate Docking and Scoring. 1. Method and Assessment of Docking Accuracy. J. Med. Chem. 2004, 47, 1739– 1749, DOI: 10.1021/jm0306430Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit74%253D&md5=8cc2f0022318b12dd972e9c493375bf9Glide: A new approach for rapid, accurate docking and scoring. 1. method and assessment of docking accuracyFriesner, Richard A.; Banks, Jay L.; Murphy, Robert B.; Halgren, Thomas A.; Klicic, Jasna J.; Mainz, Daniel T.; Repasky, Matthew P.; Knoll, Eric H.; Shelley, Mee; Perry, Jason K.; Shaw, David E.; Francis, Perry; Shenkin, Peter S.Journal of Medicinal Chemistry (2004), 47 (7), 1739-1749CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Unlike other methods for docking ligands to the rigid 3D structure of a known protein receptor, Glide approximates a complete systematic search of the conformational, orientational, and positional space of the docked ligand. In this search, an initial rough positioning and scoring phase that dramatically narrows the search space is followed by torsionally flexible energy optimization on an OPLS-AA nonbonded potential grid for a few hundred surviving candidate poses. The very best candidates are further refined via a Monte Carlo sampling of pose conformation; in some cases, this is crucial to obtaining an accurate docked pose. Selection of the best docked pose uses a model energy function that combines empirical and force-field-based terms. Docking accuracy is assessed by redocking ligands from 282 cocrystd. PDB complexes starting from conformationally optimized ligand geometries that bear no memory of the correctly docked pose. Errors in geometry for the top-ranked pose are less than 1 Å in nearly half of the cases and are greater than 2 Å in only about one-third of them. Comparisons to published data on rms deviations show that Glide is nearly twice as accurate as GOLD and more than twice as accurate as FlexX for ligands having up to 20 rotatable bonds. Glide is also found to be more accurate than the recently described Surflex method.
- 40Butina, D. Unsupervised Data Base Clustering Based on Daylight’s Fingerprint and Tanimoto Similarity: A Fast and Automated Way to Cluster Small and Large Data Sets. J. Chem. Inf. Comput. Sci. 1999, 39, 747– 750, DOI: 10.1021/ci9803381Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXktVWhtLw%253D&md5=e11c02e1e777f88e208ec5fdc5405c47Unsupervised data base clustering based on Daylight's fingerprint and Tanimoto similarity: a fast and automated way to cluster small and large data setsButina, DarkoJournal of Chemical Information and Computer Sciences (1999), 39 (4), 747-750CODEN: JCISD8; ISSN:0095-2338. (American Chemical Society)Jarvis-Patrick's (J-P) algorithm is one of the most commonly used clustering algorithms within the global pharmaceutical industry. The implementation of the J-P under Daylight software, using Daylight's fingerprints and the Tanimoto similarity index, can deal with sets of 100 k mols. in a matter of a few hours. However, the J-P clustering algorithm has several assocd. problems which make it difficult to cluster large data sets in a consistent and timely manner. The clusters produced are greatly dependent on the choice of the two parameters needed to run J-P clustering, such that this method tends to produce clusters which are either very large and heterogeneous or homogeneous but too small. In any case, J-P always requires time-consuming manual tuning. An algorithm which will identify dense clusters where similarity within each cluster reflects the Tanimoto value used for the clustering and, more importantly, where the cluster centroid will be at least similar, at the given Tanimoto value, to every other mol. within the cluster in a consistent and automated manner, is presented. The similarity term used throughout the paper reflects the overall similarity between two given mols., as defined by Daylight's fingerprints and the Tanimoto similarity index.
- 41Flagmeier, P.; Meisl, G.; Vendruscolo, M.; Knowles, T. P. J.; Dobson, C. M.; Buell, A. K.; Galvagnion, C. Mutations Associated with Familial Parkinson’s Disease Alter the Initiation and Amplification Steps of α-Synuclein Amyloid Formation. Proc. Natl. Acad. Sci. U.S.A. 2016, 113, 10328– 10333, DOI: 10.1073/pnas.1604645113Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVWks7jL&md5=70a504e389bfb0f58ae555b9d5726eceMutations associated with familial Parkinson's disease alter the initiation and amplification steps of α-synuclein aggregationFlagmeier, Patrick; Meisl, Georg; Vendruscolo, Michele; Knowles, Tuomas P. J.; Dobson, Christopher M.; Buell, Alexander K.; Galvagnion, CelineProceedings of the National Academy of Sciences of the United States of America (2016), 113 (37), 10328-10333CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Parkinson's disease is a highly debilitating neurodegenerative condition whose pathol. hallmark is the presence in nerve cells of proteinaceous deposits, known as Lewy bodies, composed primarily of amyloid fibrils of α-synuclein. Several missense mutations in the gene encoding α-synuclein have been assocd. with familial variants of Parkinson's disease and have been shown to affect the kinetics of the aggregation of the protein. Using a combination of exptl. and theor. approaches, we present a systematic in vitro study of the influence of disease-assocd. single-point mutations on the individual processes involved in α-synuclein aggregation into amyloid fibrils. We find that lipid-induced fibril prodn. and surface catalyzed fibril amplification are the processes most strongly affected by these mutations and show that familial mutations can induce dramatic changes in the crucial processes thought to be assocd. with the initiation and spreading of the aggregation of α-synuclein.
- 42Galvagnion, C.; Buell, A. K.; Meisl, G.; Michaels, T. C. T.; Vendruscolo, M.; Knowles, T. P. J.; Dobson, C. M. Lipid Vesicles Trigger α-Synuclein Aggregation by Stimulating Primary Nucleation. Nat. Chem. Biol. 2015, 11, 229– 234, DOI: 10.1038/nchembio.1750Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFCntro%253D&md5=1ceed4bdf36d5cf4663395ab4443b720Lipid vesicles trigger α-synuclein aggregation by stimulating primary nucleationGalvagnion, Celine; Buell, Alexander K.; Meisl, Georg; Michaels, Thomas C. T.; Vendruscolo, Michele; Knowles, Tuomas P. J.; Dobson, Christopher M.Nature Chemical Biology (2015), 11 (3), 229-234CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)α-Synuclein (α-syn) is a 140-residue intrinsically disordered protein that is involved in neuronal and synaptic vesicle plasticity, but its aggregation to form amyloid fibrils is the hallmark of Parkinson's disease (PD). The interaction between α-syn and lipid surfaces is believed to be a key feature for mediation of its normal function, but under other circumstances it is able to modulate amyloid fibril formation. Using a combination of exptl. and theor. approaches, we identify the mechanism through which facile aggregation of α-syn is induced under conditions where it binds a lipid bilayer, and we show that the rate of primary nucleation can be enhanced by three orders of magnitude or more under such conditions. These results reveal the key role that membrane interactions can have in triggering conversion of α-syn from its sol. state to the aggregated state that is assocd. with neurodegeneration and to its assocd. disease states.
- 43Michaels, T. C. T.; Cohen, S. I. A.; Vendruscolo, M.; Dobson, C. M.; Knowles, T. P. J. Hamiltonian Dynamics of Protein Filament Formation. Phys. Rev. Lett. 2016, 116, 038101, DOI: 10.1103/PhysRevLett.116.038101Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtF2ksr3J&md5=fb3129c50d3e9aaf4d5dea257fa941f2Hamiltonian dynamics of protein filament formationMichaels, Thomas C. T.; Cohen, Samuel I. A.; Vendruscolo, Michele; Dobson, Christopher M.; Knowles, Tuomas P. J.Physical Review Letters (2016), 116 (3), 038101/1-038101/6CODEN: PRLTAO; ISSN:0031-9007. (American Physical Society)We establish the Hamiltonian structure of the rate equations describing the formation of protein filaments. We then show that this formalism provides a unified view of the behavior of a range of biol. self-assembling systems as diverse as actin. prions. and amyloidogenic polypeptides. We further demonstrate that the time-translation symmetry of the resulting Hamiltonian leads to previously unsuggested conservation laws that connect the no. and mass concns. of fibrils and allow linear growth phenomena to be equated with autocatalytic growth processes. We finally show how these results reveal simple rate laws that provide the basis for interpreting exptl. data in terms of specific mechanisms controlling the proliferation of fibrils.
- 44Palacio-Rodríguez, K.; Lans, I.; Cavasotto, C. N.; Cossio, P. Exponential Consensus Ranking Improves the Outcome in Docking and Receptor Ensemble Docking. Sci. Rep. 2019, 9, 5142 DOI: 10.1038/s41598-019-41594-3Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cbosVCksA%253D%253D&md5=949544789593b1c55faa32bfcce3afc2Exponential consensus ranking improves the outcome in docking and receptor ensemble dockingPalacio-Rodriguez Karen; Lans Isaias; Cossio Pilar; Cavasotto Claudio N; Cavasotto Claudio N; Cavasotto Claudio N; Cossio PilarScientific reports (2019), 9 (1), 5142 ISSN:.Consensus-scoring methods are commonly used with molecular docking in virtual screening campaigns to filter potential ligands for a protein target. Traditional consensus methods combine results from different docking programs by averaging the score or rank of each molecule obtained from individual programs. Unfortunately, these methods fail if one of the docking programs has poor performance, which is likely to occur due to training-set dependencies and scoring-function parameterization. In this work, we introduce a novel consensus method that overcomes these limitations. We combine the results from individual docking programs using a sum of exponential distributions as a function of the molecule rank for each program. We test the method over several benchmark systems using individual and ensembles of target structures from diverse protein families with challenging decoy/ligand datasets. The results demonstrate that the novel method outperforms the best traditional consensus strategies over a wide range of systems. Moreover, because the novel method is based on the rank rather than the score, it is independent of the score units, scales and offsets, which can hinder the combination of results from different structures or programs. Our method is simple and robust, providing a theoretical basis not only for molecular docking but also for any consensus strategy in general.
- 45Wilhelm, B. G.; Mandad, S.; Truckenbrodt, S.; Kröhnert, K.; Schäfer, C.; Rammner, B.; Koo, S. J.; Claßen, Ga.; Krauss, M.; Haucke, V.; Urlaub, H.; Rizzoli, S. O. Composition of Isolated Synaptic Boutons Reveals the Amounts of Vesicle Trafficking Proteins. Science 2014, 344, 1023– 1028, DOI: 10.1126/science.1252884Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXosFSmurc%253D&md5=0e85f1ad06f7cd9cfca66cea6a3b59adComposition of isolated synaptic boutons reveals the amounts of vesicle trafficking proteinsWilhelm, Benjamin G.; Mandad, Sunit; Truckenbrodt, Sven; Kroehnert, Katharina; Schaefer, Christina; Rammner, Burkhard; Koo, Seong Joo; Classen, Gala A.; Krauss, Michael; Haucke, Volker; Urlaub, Henning; Rizzoli, Silvio O.Science (Washington, DC, United States) (2014), 344 (6187), 1023-1028CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Synaptic vesicle recycling has long served as a model for the general mechanisms of cellular trafficking. We used an integrative approach, combining quant. immunoblotting and mass spectrometry to det. protein nos.; electron microscopy to measure organelle nos., sizes, and positions; and super-resoln. fluorescence microscopy to localize the proteins. Using these data, we generated a three-dimensional model of an "av." synapse, displaying 300,000 proteins in at. detail. The copy nos. of proteins involved in the same step of synaptic vesicle recycling correlated closely. In contrast, copy nos. varied over more than three orders of magnitude between steps, from about 150 copies for the endosomal fusion proteins to more than 20,000 for the exocytotic ones.
- 46Yang, Y.; Shi, Y.; Schweighauser, M.; Zhang, X.; Kotecha, A.; Murzin, A. G.; Garringer, H. J.; Cullinane, P. W.; Saito, Y.; Foroud, T.; Warner, T. T.; Hasegawa, K.; Vidal, R.; Murayama, S.; Revesz, T.; Ghetti, B.; Hasegawa, M.; Lashley, T.; Scheres, S. H. W.; Goedert, M. Structures of α-synuclein filaments from human brains with Lewy pathology, Nature 2022. 610 791 795 DOI: 10.1101/2022.07.12.499706610 .Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xis1Onu7%252FN&md5=00b50c7577f4c53e7250a09bb96bfda8Structures of alpha synuclein filaments from human brains with Lewy pathologyYang, Yang; Shi, Yang; Schweighauser, Manuel; Zhang, Xianjun; Kotecha, Abhay; Murzin, Alexey G.; Garringer, Holly J.; Cullinane, Patrick W.; Saito, Yuko; Foroud, Tatiana; Warner, Thomas T.; Hasegawa, Kazuko; Vidal, Ruben; Murayama, Shigeo; Revesz, Tamas; Ghetti, Bernardino; Hasegawa, Masato; Lashley, Tammaryn; Scheres, Sjors H. W.; Goedert, MichelNature (London, United Kingdom) (2022), 610 (7933), 791-795CODEN: NATUAS; ISSN:1476-4687. (Nature Portfolio)Parkinson disease (PD) is the most common movement disorder, with resting tremor, rigidity, bradykinesia and postural instability being major symptoms. Neuropathol. it is characterized by the presence of abundant filamentous inclusions of α-synuclein in the form of Lewy bodies and Lewy neurites in some brain cells, including dopaminergic nerve cells of the substantia nigra. PD is increasingly recognized as a multisystem disorder, with cognitive decline being one of its most common non-motor symptoms. Many patients with PD develop dementia more than 10 years after diagnosis. PD dementia (PDD) is clin. and neuropathol. similar to dementia with Lewy bodies (DLB), which is diagnosed when cognitive impairment precedes parkinsonian motor signs or begins within one year from their onset. In PDD, cognitive impairment develops in the setting of well-established PD. Besides PD and DLB, multiple system atrophy (MSA) is the third major synucleinopathy. It is characterized by the presence of abundant filamentous α-synuclein inclusions in brain cells, esp. oligodendrocytes (Papp-Lantos bodies). We previously reported the electron cryo-microscopy structures of two types of α-synuclein filament extd. from the brains of individuals with MSA6. Each filament type is made of two different protofilaments. Here we report that the cryo-electron microscopy structures of α-synuclein filaments from the brains of individuals with PD, PDD and DLB are made of a single protofilament (Lewy fold) that is markedly different from the protofilaments of MSA. These findings establish the existence of distinct mol. conformers of assembled α-synuclein in neurodegenerative disease.
- 47Cohen, S. I. A.; Arosio, P.; Presto, J.; Kurudenkandy, F. R.; Biverstål, H.; Dolfe, L.; Dunning, C.; Yang, X.; Frohm, B.; Vendruscolo, M.; Johansson, J.; Dobson, C. M.; Fisahn, A.; Knowles, T. P. J.; Linse, S. A Molecular Chaperone Breaks the Catalytic Cycle That Generates Toxic Aβ Oligomers. Nat. Struct. Mol. Biol. 2015, 22, 207– 213, DOI: 10.1038/nsmb.2971Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXivVSitLY%253D&md5=03c46f97e7f2361193667db1d4b94f07A molecular chaperone breaks the catalytic cycle that generates toxic Aβ oligomersCohen, Samuel I. A.; Arosio, Paolo; Presto, Jenny; Kurudenkandy, Firoz Roshan; Biverstal, Henrik; Dolfe, Lisa; Dunning, Christopher; Yang, Xiaoting; Frohm, Birgitta; Vendruscolo, Michele; Johansson, Jan; Dobson, Christopher M.; Fisahn, Andre; Knowles, Tuomas P. J.; Linse, SaraNature Structural & Molecular Biology (2015), 22 (3), 207-213CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Alzheimer's disease is an increasingly prevalent neurodegenerative disorder whose pathogenesis has been assocd. with aggregation of the amyloid-β peptide (Aβ42). Recent studies have revealed that once Aβ42 fibrils are generated, their surfaces effectively catalyze the formation of neurotoxic oligomers. Here we show that a mol. chaperone, a human Brichos domain, can specifically inhibit this catalytic cycle and limit human Aβ42 toxicity. We demonstrate in vitro that Brichos achieves this inhibition by binding to the surfaces of fibrils, thereby redirecting the aggregation reaction to a pathway that involves minimal formation of toxic oligomeric intermediates. We verify that this mechanism occurs in living mouse brain tissue by cytotoxicity and electrophysiol. expts. These results reveal that mol. chaperones can help maintain protein homeostasis by selectively suppressing crit. microscopic steps within the complex reaction pathways responsible for the toxic effects of protein misfolding and aggregation.
- 48Perni, M.; Galvagnion, C.; Maltsev, A.; Meisl, G.; Müller, M. B. D.; Challa, P. K.; Kirkegaard, J. B.; Cohen, S. I. A.; Cascella, R.; Chen, S. W.; Limboker, R.; Sormanni, P.; Heller, G. T.; Francesco, A.; Cremades, N.; Cecchi, C.; Chiti, F.; Ellen, A. A.; Knowles, T. P. J.; Vendruscolo, M.; Bax, A.; Zasloff, M.; Dobson, C. M.; Perni, M.; Galvagnion, C.; Maltsev, A.; Meisl, G.; Müller, M. B. D.; Challa, P. K.; Julius, B.; Flagmeier, P.; Cohen, S. I. A.; Chen, S. W.; Limbocker, R.; Sormanni, P.; Heller, G. T.; Aprile, F. A.; Cremades, N.; Cecchi, C.; Chiti, F.; Nollen, E. A. A.; Tuomas, P. J.; Vendruscolo, M.; Bax, A.; Dobson, C. M.; Perni, M.; Galvagnion, C.; Maltsev, A.; Meisl, G.; Müller, M. B. D.; Challa, P. K. A Natural Product Inhibits the Initiation of α-Synuclein Aggregation and Suppresses Its Toxicity. Proc. Natl. Acad. Sci. U.S.A. 2017, 114, E1009– E1017, DOI: 10.1073/pnas.1610586114Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFarsLY%253D&md5=116485fd417a51fac86fb5d78c604020A natural product inhibits the initiation of α-synuclein aggregation and suppresses its toxicityPerni, Michele; Galvagnion, Celine; Maltsev, Alexander; Meisl, Georg; Muller, Martin B. D.; Challa, Pavan K.; Kirkegaard, Julius B.; Flagmeier, Patrick; Cohen, Samuel I. A.; Cascella, Roberta; Chen, Serene W.; Limboker, Ryan; Sormanni, Pietro; Heller, Gabriella T.; Aprile, Francesco A.; Cremades, Nunilo; Cecchi, Cristina; Chiti, Fabrizio; Nollen, Ellen A. A.; Knowles, Tuomas P. J.; Vendruscolo, Michele; Bax, Adriaan; Zasloff, Michael; Dobson, Christopher M.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (6), E1009-E1017CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The self-assembly of α-synuclein is closely assocd. with Parkinson's disease and related syndromes. The authors show that squalamine, a natural product with known anticancer and antiviral activity, dramatically affects α-synuclein aggregation in vitro and in vivo. The authors elucidate the mechanism of action of squalamine by investigating its interaction with lipid vesicles, which are known to stimulate nucleation, and find that this compd. displaces α-synuclein from the surfaces of such vesicles, thereby blocking the first steps in its aggregation process. The authors also show that squalamine almost completely suppresses the toxicity of α-synuclein oligomers in human neuroblastoma cells by inhibiting their interactions with lipid membranes. The authors further examine the effects of squalamine in a Caenorhabditis elegans strain overexpressing α-synuclein, observing a dramatic redn. of α-synuclein aggregation and an almost complete elimination of muscle paralysis. These findings suggest that squalamine could be a means of therapeutic intervention in Parkinson's disease and related conditions.
- 49Agerschou, E. D.; Flagmeier, P.; Saridaki, T.; Galvagnion, C.; Komnig, D.; Heid, L.; Prasad, V.; Shaykhalishahi, H.; Willbold, D.; Dobson, C. M.; Voigt, A.; Falkenburger, B.; Hoyer, W.; Buell, A. K. An Engineered Monomer Binding-Protein for α-Synuclein Efficiently Inhibits the Proliferation of Amyloid Fibrils. eLife 2019, 8, e46112 DOI: 10.7554/eLife.46112Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlaltLzE&md5=5d8e3c3ee85ea1ec460bdcd61c05ca83An engineered monomer binding-protein for α-synuclein efficiently inhibits the proliferation of amyloid fibrilsAgerschou, Emil Dandanell; Flagmeier, Patrick; Saridaki, Theodora; Galvagnion, Celine; Komnig, Daniel; Heid, Laetitia; Prasad, Vibha; Shaykhalishahi, Hamed; Willbold, Dieter; Dobson, Christopher M.; Voigt, Aaron; Falkenburger, Bjoern; Hoyer, Wolfgang; Buell, Alexander K.eLife (2019), 8 (), e46112/1-e46112/31CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Removing or preventing the formation of α-synuclein aggregates is a plausible strategy against Parkinson's disease. To this end, we have engineered the β-wrapin AS69 to bind monomeric α-synuclein with high affinity. In cultured cells, AS69 reduced the self-interaction of α-synuclein and formation of visible α-synuclein aggregates. In flies, AS69 reduced α-synuclein aggregates and the locomotor deficit resulting from α-synuclein expression in neuronal cells. In biophys. expts. in vitro, AS69 highly sub-stoichiometrically inhibited both primary and autocatalytic secondary nucleation processes, even in the presence of a large excess of monomer. We present evidence that the AS69-α-synuclein complex, rather than the free AS69, is the inhibitory species responsible for sub-stoichiometric inhibition of secondary nucleation. These results represent a new paradigm that high affinity monomer binders can lead to strongly substoichiometric inhibition of nucleation processes.
- 50Linse, S.; Scheidt, T.; Bernfur, K.; Vendruscolo, M.; Dobson, C. M.; Cohen, S. I. A.; Sileikis, E.; Lundqvist, M.; Qian, F.; O’Malley, T.; Bussiere, T.; Weinreb, P. H.; Xu, C. K.; Meisl, G.; Devenish, S. R. A.; Knowles, T. P. J.; Hansson, O. Kinetic Fingerprints Differentiate the Mechanisms of Action of Anti-Aβ Antibodies. Nat. Struct. Mol. Biol. 2020, 27, 1125– 1133, DOI: 10.1038/s41594-020-0505-6Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFegurjF&md5=2adc8febabbb832a3c8db63019db83e6Kinetic fingerprints differentiate the mechanisms of action of anti-Aβ antibodiesLinse, Sara; Scheidt, Tom; Bernfur, Katja; Vendruscolo, Michele; Dobson, Christopher M.; Cohen, Samuel I. A.; Sileikis, Eimantas; Lundqvist, Martin; Qian, Fang; O'Malley, Tiernan; Bussiere, Thierry; Weinreb, Paul H.; Xu, Catherine K.; Meisl, Georg; Devenish, Sean R. A.; Knowles, Tuomas P. J.; Hansson, OskarNature Structural & Molecular Biology (2020), 27 (12), 1125-1133CODEN: NSMBCU; ISSN:1545-9993. (Nature Research)The amyloid cascade hypothesis, according to which the self-assembly of amyloid-β peptide (Aβ) is a causative process in Alzheimer's disease, has driven many therapeutic efforts for the past 20 years. Failures of clin. trials investigating Aβ-targeted therapies have been interpreted as evidence against this hypothesis, irresp. of the characteristics and mechanisms of action of the therapeutic agents, which are highly challenging to assess. Here, we combine kinetic analyses with quant. binding measurements to address the mechanism of action of four clin. stage anti-Aβ antibodies, aducanumab, gantenerumab, bapineuzumab and solanezumab. We quantify the influence of these antibodies on the aggregation kinetics and on the prodn. of oligomeric aggregates and link these effects to the affinity and stoichiometry of each antibody for monomeric and fibrillar forms of Aβ. Our results reveal that, uniquely among these four antibodies, aducanumab dramatically reduces the flux of Aβ oligomers.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 20 publications.
- Susanta Das, Kenneth M. Merz, Jr.. Molecular Gas-Phase Conformational Ensembles. Journal of Chemical Information and Modeling 2024, 64
(3)
, 749-760. https://doi.org/10.1021/acs.jcim.3c01309
- Robert I. Horne, Jared Wilson-Godber, Alicia González Díaz, Z. Faidon Brotzakis, Srijit Seal, Rebecca C. Gregory, Andrea Possenti, Sean Chia, Michele Vendruscolo. Using Generative Modeling to Endow with Potency Initially Inert Compounds with Good Bioavailability and Low Toxicity. Journal of Chemical Information and Modeling 2024, 64
(3)
, 590-596. https://doi.org/10.1021/acs.jcim.3c01777
- Sarah E. Sandler, Robert I. Horne, Sara Rocchetti, Robert Novak, Nai-Shu Hsu, Marta Castellana Cruz, Z. Faidon Brotzakis, Rebecca C. Gregory, Sean Chia, Gonçalo J. L. Bernardes, Ulrich F. Keyser, Michele Vendruscolo. Multiplexed Digital Characterization of Misfolded Protein Oligomers via Solid-State Nanopores. Journal of the American Chemical Society 2023, 145
(47)
, 25776-25788. https://doi.org/10.1021/jacs.3c09335
- Robert I. Horne, Michael A. Metrick II, Wing Man, Dillon J. Rinauro, Z. Faidon Brotzakis, Sean Chia, Georg Meisl, Michele Vendruscolo. Secondary Processes Dominate the Quiescent, Spontaneous Aggregation of α-Synuclein at Physiological pH with Sodium Salts. ACS Chemical Neuroscience 2023, 14
(17)
, 3125-3131. https://doi.org/10.1021/acschemneuro.3c00282
- Robert I. Horne, Mhd Hussein Murtada, Donghui Huo, Z. Faidon Brotzakis, Rebecca C. Gregory, Andrea Possenti, Sean Chia, Michele Vendruscolo. Exploration and Exploitation Approaches Based on Generative Machine Learning to Identify Potent Small Molecule Inhibitors of α-Synuclein Secondary Nucleation. Journal of Chemical Theory and Computation 2023, 19
(14)
, 4701-4710. https://doi.org/10.1021/acs.jctc.2c01303
- Robert I. Horne, Sarah E. Sandler, Michele Vendruscolo, Ulrich F. Keyser. Detection of protein oligomers with nanopores . Nature Reviews Chemistry 2025, 72 https://doi.org/10.1038/s41570-025-00694-7
- Samuel Bismut, Matthias M. Schneider, Masashi Miyasaki, Yuqing Feng, Ellis J. Wilde, M. Dylan Gunawardena, Tuomas P.J. Knowles, Gabriele S Kaminski Schierle, Laura Susan Itzhaki, Janet Reiko Kumita. Using a stable protein scaffold to display peptides that bind to alpha-synuclein fibrils. 2025https://doi.org/10.1101/2025.02.04.636406
- Samuel T. Dada, Zenon Toprakcioglu, Mariana P. Cali, Alexander Röntgen, Maarten C. Hardenberg, Owen M. Morris, Lena K. Mrugalla, Tuomas P. J. Knowles, Michele Vendruscolo. Pharmacological inhibition of α-synuclein aggregation within liquid condensates. Nature Communications 2024, 15
(1)
https://doi.org/10.1038/s41467-024-47585-x
- Priscilla Chinchilla, Baifan Wang, Joseph H. Lubin, Xue Yang, Jonathan Roth, Sagar D. Khare, Jean Baum. Synergistic Multi-Pronged Interactions Mediate the Effective Inhibition of Alpha-Synuclein Aggregation by the Chaperone HtrA1. 2024https://doi.org/10.1101/2024.11.25.624572
- Sneha Menon, Jagannath Mondal. Simulating the anti-aggregative effect of fasudil in early dimerisation process of α-synuclein. Biophysical Chemistry 2024, 314 , 107319. https://doi.org/10.1016/j.bpc.2024.107319
- Eva D. Ruiz-Ortega, Anna Wilkaniec, Agata Adamczyk. Liquid–liquid phase separation and conformational strains of α-Synuclein: implications for Parkinson’s disease pathogenesis. Frontiers in Molecular Neuroscience 2024, 17 https://doi.org/10.3389/fnmol.2024.1494218
- Gian Pietro Sechi, M. Margherita Sechi. Small Molecules, α-Synuclein Pathology, and the Search for Effective Treatments in Parkinson’s Disease. International Journal of Molecular Sciences 2024, 25
(20)
, 11198. https://doi.org/10.3390/ijms252011198
- Robert I. Horne, Ewa A. Andrzejewska, Parvez Alam, Z. Faidon Brotzakis, Ankit Srivastava, Alice Aubert, Magdalena Nowinska, Rebecca C. Gregory, Roxine Staats, Andrea Possenti, Sean Chia, Pietro Sormanni, Bernardino Ghetti, Byron Caughey, Tuomas P. J. Knowles, Michele Vendruscolo. Discovery of potent inhibitors of α-synuclein aggregation using structure-based iterative learning. Nature Chemical Biology 2024, 20
(5)
, 634-645. https://doi.org/10.1038/s41589-024-01580-x
- Timothy S. Chisholm, Christopher A. Hunter. A closer look at amyloid ligands, and what they tell us about protein aggregates. Chemical Society Reviews 2024, 53
(3)
, 1354-1374. https://doi.org/10.1039/D3CS00518F
- Maksym Galkin, Anastasiia Priss, Yevhenii Kyriukha, Volodymyr Shvadchak. Navigating α‐Synuclein Aggregation Inhibition: Methods, Mechanisms, and Molecular Targets. The Chemical Record 2024, 24
(2)
https://doi.org/10.1002/tcr.202300282
- Milica Marković, Jelica Milošević, Weirong Wang, Yanguang Cao. Passive Immunotherapies Targeting Amyloid-β in Alzheimer’s Disease: A Quantitative Systems Pharmacology Perspective. Molecular Pharmacology 2024, 105
(1)
, 1-13. https://doi.org/10.1124/molpharm.123.000726
- Keyur N. Patel, Dhruvil Chavda, Moutusi Manna. Molecular Docking of Intrinsically Disordered Proteins: Challenges and Strategies. 2024, 165-201. https://doi.org/10.1007/978-1-0716-3985-6_11
- Roxine Staats, Z. Faidon Brotzakis, Sean Chia, Robert I. Horne, Michele Vendruscolo. Optimization of a small molecule inhibitor of secondary nucleation in α-synuclein aggregation. Frontiers in Molecular Biosciences 2023, 10 https://doi.org/10.3389/fmolb.2023.1155753
- Michele Vendruscolo. Thermodynamic and kinetic approaches for drug discovery to target protein misfolding and aggregation. Expert Opinion on Drug Discovery 2023, 18
(8)
, 881-891. https://doi.org/10.1080/17460441.2023.2221024
- Natalia Siwecka, Kamil Saramowicz, Grzegorz Galita, Wioletta Rozpędek-Kamińska, Ireneusz Majsterek. Inhibition of Protein Aggregation and Endoplasmic Reticulum Stress as a Targeted Therapy for α-Synucleinopathy. Pharmaceutics 2023, 15
(8)
, 2051. https://doi.org/10.3390/pharmaceutics15082051
- Samuel Peña-Díaz, Javier García-Pardo, Salvador Ventura. Development of Small Molecules Targeting α-Synuclein Aggregation: A Promising Strategy to Treat Parkinson’s Disease. Pharmaceutics 2023, 15
(3)
, 839. https://doi.org/10.3390/pharmaceutics15030839
- Kwangho Nam, Magnus Wolf-Watz. Protein dynamics: The future is bright and complicated!. Structural Dynamics 2023, 10
(1)
, 014301. https://doi.org/10.1063/4.0000179
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Combined structure-based and kinetic-based approach to identify small molecules that bind α-synuclein fibrils and inhibit its aggregation. In the first step, computational docking is performed on a large library of small molecules. The top candidates are then clustered to identify a subset of chemically diverse compounds that exhibit high predicted binding scores for α-synuclein fibrils. Subsequently, these compounds are experimentally validated through a kinetic assay for their ability to inhibit the secondary nucleation aggregation of α-synuclein by binding to the surface of fibrils. Further rate constant analysis and fibril-binding experiments allow for the positive compounds to be characterized based on both their inhibition of the kinetic assay, as well as their binding affinity toward α-synuclein fibrils.
Figure 2
Figure 2. Five compounds selected from the docking library inhibit the aggregation of α-synuclein. (A) Kinetic profiles of a 10 μM solution of α-synuclein in the presence of 25 nM seeds at pH 4.8 and 37 °C, in the presence of 1% DMSO alone (beige), in the presence of 10 molar equivalents of compounds A–E (represented in different colors), or in the presence of 10 molar equivalents of other compounds in the docking library that did not affect significantly α-synuclein aggregation (black). (B) Relative t1/2 of the aggregation of α-synuclein in the presence of compounds A–E as shown in (A), normalized to the DMSO control. (C) Chemical structures of compounds A–E. Throughout, error bars represent mean ± SEM of two replicates.
Figure 3
Figure 3. Computational docking of compounds to α-synuclein fibrils. (A–E) Binding poses of compounds A–E to the selected binding pocket in α-synuclein fibrils (centered between residues His50 and Glu57), determined either through FRED or AutoDock Vina. (F) Representation of possible binding pockets in the fibril structure (PDB: 6cu7, cyan) identified by Fpocket, with pockets in the fibril core (blue spheres), and at the fibril surface (red spheres). Key binding site residues His50 and Glu57 are shown in licorice representation.
Figure 4
Figure 4. Compounds identified by docking specifically inhibit the proliferation of α-synuclein aggregates by secondary nucleation. (A) Kinetic profiles of a 10 μM solution of α-synuclein in the presence of 25 nM seeds at pH 4.8, 37 °C, in the presence of either 1% DMSO alone (purple) or increasing molar equivalents of compound C (represented in different colors). (B) Relative rate of fibril amplification of α-synuclein in the presence of compounds A–E as shown in (A) and Figure S3, normalized to the DMSO control. (C) Kinetic profiles of a 10 μM solution of α-synuclein in the presence of 5 μM seeds at pH 4.8, 37 °C, in the presence of either 1% DMSO alone (purple) or increasing molar equivalents of compound C (represented in different colors). Dotted lines indicate the vmax of the reaction which is used to extract the elongation rate of the aggregation process. (D) Relative rate of fibril elongation of α-synuclein in the presence of compounds A–E as shown in (C) and Figure S5,6, normalized to the DMSO control. Throughout, error bars represent mean ± SEM of three replicates.
Figure 5
Figure 5. Compound C inhibits the reactive flux toward α-synuclein oligomers and displays binding affinity and specificity toward α-synuclein fibrils. (A) Time dependence of the reactive flux toward α-synuclein oligomers either in the presence of 1% DMSO alone (purple) or in the presence of increasing molar equivalents of compound C (represented in different colors), normalized to the DMSO control. (B) Change in fluorescence polarization (in mP units) of 10 μM compound C with increasing concentrations of either α-synuclein fibrils (purple) or Aβ42 fibrils (red). The solid lines are fits to the points using a one-step binding curve, estimating a Kd of 4 μM for compound C toward α-synuclein fibrils. (C) Total ion current (TIC) of 10 μM compound C bound and unbound to 10 μM α-synuclein fibrils detected by mass spectrometry (see the Materials and Methods section). (D) Representative images indicating either the fluorescence of the red channel (amyloid-specific dye pFTAA) or the green channel (compound C) following incubation in the absence (top) or presence (bottom) of 100 nM α-synuclein fibrils. Throughout, error bars represent mean ± SEM of two replicates.
References
This article references 50 other publications.
- 1Poewe, W.; Seppi, K.; Tanner, C. M.; Halliday, G. M.; Brundin, P.; Volkmann, J.; Schrag, A. E.; Lang, A. E. Parkinson Disease. Nat. Rev. Dis. Primers 2017, 3, 17013, DOI: 10.1038/nrdp.2017.131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cvgtFChsA%253D%253D&md5=b624b37fee08b8b663ea88b83bc47865Parkinson diseasePoewe Werner; Seppi Klaus; Tanner Caroline M; Tanner Caroline M; Halliday Glenda M; Halliday Glenda M; Brundin Patrik; Volkmann Jens; Schrag Anette-Eleonore; Lang Anthony ENature reviews. Disease primers (2017), 3 (), 17013 ISSN:.Parkinson disease is the second-most common neurodegenerative disorder that affects 2-3% of the population ≥65 years of age. Neuronal loss in the substantia nigra, which causes striatal dopamine deficiency, and intracellular inclusions containing aggregates of α-synuclein are the neuropathological hallmarks of Parkinson disease. Multiple other cell types throughout the central and peripheral autonomic nervous system are also involved, probably from early disease onwards. Although clinical diagnosis relies on the presence of bradykinesia and other cardinal motor features, Parkinson disease is associated with many non-motor symptoms that add to overall disability. The underlying molecular pathogenesis involves multiple pathways and mechanisms: α-synuclein proteostasis, mitochondrial function, oxidative stress, calcium homeostasis, axonal transport and neuroinflammation. Recent research into diagnostic biomarkers has taken advantage of neuroimaging in which several modalities, including PET, single-photon emission CT (SPECT) and novel MRI techniques, have been shown to aid early and differential diagnosis. Treatment of Parkinson disease is anchored on pharmacological substitution of striatal dopamine, in addition to non-dopaminergic approaches to address both motor and non-motor symptoms and deep brain stimulation for those developing intractable L-DOPA-related motor complications. Experimental therapies have tried to restore striatal dopamine by gene-based and cell-based approaches, and most recently, aggregation and cellular transport of α-synuclein have become therapeutic targets. One of the greatest current challenges is to identify markers for prodromal disease stages, which would allow novel disease-modifying therapies to be started earlier.
- 2Nichols, E.; Szoeke, C. E. I.; Vollset, S. E.; Abbasi, N.; Abd-Allah, F.; Abdela, J.; Aichour, M. T. E.; Akinyemi, R. O.; Alahdab, F.; Asgedom, S. W.; Awasthi, A.; Barker-Collo, S. L.; Baune, B. T.; Béjot, Y.; Belachew, A. B.; Bennett, D. A.; Biadgo, B.; Bijani, A.; Bin Sayeed, M. S.; Brayne, C.; Carpenter, D. O.; Carvalho, F.; Catalá-López, F.; Cerin, E.; Choi, J. Y. J.; Dang, A. K.; Degefa, M. G.; Djalalinia, S.; Dubey, M.; Duken, E. E.; Edvardsson, D.; Endres, M.; Eskandarieh, S.; Faro, A.; Farzadfar, F.; Fereshtehnejad, S. M.; Fernandes, E.; Filip, I.; Fischer, F.; Gebre, A. K.; Geremew, D.; Ghasemi-Kasman, M.; Gnedovskaya, E. V.; Gupta, R.; Hachinski, V.; Hagos, T. B.; Hamidi, S.; Hankey, G. J.; Haro, J. M.; Hay, S. I.; Irvani, S. S. N.; Jha, R. P.; Jonas, J. B.; Kalani, R.; Karch, A.; Kasaeian, A.; Khader, Y. S.; Khalil, I. A.; Khan, E. A.; Khanna, T.; Khoja, T. A. M.; Khubchandani, J.; Kisa, A.; Kissimova-Skarbek, K.; Kivimäki, M.; Koyanagi, A.; Krohn, K. J.; Logroscino, G.; Lorkowski, S.; Majdan, M.; Malekzadeh, R.; März, W.; Massano, J.; Mengistu, G.; Meretoja, A.; Mohammadi, M.; Mohammadi-Khanaposhtani, M.; Mokdad, A. H.; Mondello, S.; Moradi, G.; Nagel, G.; Naghavi, M.; Naik, G.; Nguyen, L. H.; Nguyen, T. H.; Nirayo, Y. L.; Nixon, M. R.; Ofori-Asenso, R.; Ogbo, F. A.; Olagunju, A. T.; Owolabi, M. O.; Panda-Jonas, S.; Passos, V. M. d. A.; Pereira, D. M.; Pinilla-Monsalve, G. D.; Piradov, M. A.; Pond, C. D.; Poustchi, H.; Qorbani, M.; Radfar, A.; Reiner, R. C.; Robinson, S. R.; Roshandel, G.; Rostami, A.; Russ, T. C.; Sachdev, P. S.; Safari, H.; Safiri, S.; Sahathevan, R.; Salimi, Y.; Satpathy, M.; Sawhney, M.; Saylan, M.; Sepanlou, S. G.; Shafieesabet, A.; Shaikh, M. A.; Sahraian, M. A.; Shigematsu, M.; Shiri, R.; Shiue, I.; Silva, J. P.; Smith, M.; Sobhani, S.; Stein, D. J.; Tabarés-Seisdedos, R.; Tovani-Palone, M. R.; Tran, B. X.; Tran, T. T.; Tsegay, A. T.; Ullah, I.; Venketasubramanian, N.; Vlassov, V.; Wang, Y. P.; Weiss, J.; Westerman, R.; Wijeratne, T.; Wyper, G. M. A.; Yano, Y.; Yimer, E. M.; Yonemoto, N.; Yousefifard, M.; Zaidi, Z.; Zare, Z.; Vos, T.; Feigin, V. L.; Murray, C. J. L. Global, Regional, and National Burden of Alzheimer’s Disease and Other Dementias, 1990–2016: A Systematic Analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 88– 106, DOI: 10.1016/S1474-4422(18)30403-4There is no corresponding record for this reference.
- 3Balestrino, R.; Schapira, A. H. V. Parkinson Disease. Eur. J. Neurol. 2020, 27, 27– 42, DOI: 10.1111/ene.141083https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MnpslGguw%253D%253D&md5=2a9d346119d47bb04883b69786b0308cParkinson diseaseBalestrino R; Schapira A H VEuropean journal of neurology (2020), 27 (1), 27-42 ISSN:.Parkinson disease (PD) is the most common neurodegenerative movement disorder. In Europe, prevalence and incidence rates for PD are estimated at approximately 108-257/100 000 and 11-19/100 000 per year, respectively. Risk factors include age, male gender and some environmental factors. The aetiology of the disease in most patients is unknown, but different genetic causes have been identified. Although familial forms of PD account for only 5%-15% of cases, studies on these families provided interesting insight on the genetics and the pathogenesis of the disease allowing the identification of genes implicated in its pathogenesis and offering critical insights into the mechanisms of disease. The cardinal motor symptoms of PD are tremor, rigidity, bradykinesia/akinesia and postural instability, but the clinical picture includes other motor and non-motor symptoms. Its diagnosis is principally clinical, although specific investigations can help the differential diagnosis from other forms of parkinsonism. Pathologically, PD is characterized by the loss of dopaminergic neurons in the pars compacta of the substantia nigra and by accumulation of misfolded α-synuclein, which is found in intra-cytoplasmic inclusions called Lewy bodies. Currently available treatments offer good control of motor symptoms but do not modify the evolution of the disease. This article is intended to provide a comprehensive, general and practical review of PD for the general neurologist.
- 4Pringsheim, T.; Jette, N.; Frolkis, A.; Steeves, T. D. L. The Prevalence of Parkinson’s Disease: A Systematic Review and Meta-Analysis. Mov. Disord. 2014, 29, 1583– 1590, DOI: 10.1002/mds.259454https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cfns1yiuw%253D%253D&md5=6e281ab13ef57a2c8c7c91d44cced519The prevalence of Parkinson's disease: a systematic review and meta-analysisPringsheim Tamara; Jette Nathalie; Frolkis Alexandra; Steeves Thomas D LMovement disorders : official journal of the Movement Disorder Society (2014), 29 (13), 1583-90 ISSN:.Parkinson's Disease (PD) is a common neurodegenerative disorder. We sought to synthesize studies on the prevalence of PD to obtain an overall view of how the prevalence of this disease varies by age, by sex, and by geographic location. We searched MEDLINE and EMBASE for epidemiological studies of PD from 1985 to 2010. Data were analyzed by age group, geographic location, and sex. Geographic location was stratified by the following groups: 1) Asia, 2) Africa, 3) South America, and 4) Europe/North America/Australia. Meta-regression was used to determine whether a significant difference was present between groups. Forty-seven studies were included in the analysis. Meta-analysis of the worldwide data showed a rising prevalence of PD with age (all per 100,000): 41 in 40 to 49 years; 107 in 50 to 59 years; 173 in 55 to 64 years; 428 in 60 to 69 years; 425 in 65 to 74 years; 1087 in 70 to 79 years; and 1903 in older than age 80. A significant difference was seen in prevalence by geographic location only for individuals 70 to 79 years old, with a prevalence of 1,601 in individuals from North America, Europe, and Australia, compared with 646 in individuals from Asia (P < 0.05). A significant difference in prevalence by sex was found only for individuals 50 to 59 years old, with a prevalence of 41 in females and 134 in males (P < 0.05). PD prevalence increases steadily with age. Some differences in prevalence by geographic location and sex can be detected.
- 5Spillantini, M. G.; Schmidt, M. L.; Lee, V. M.; Trojanowski, J. Q.; Jakes, R.; Goedert, M. Alpha-Synuclein in Lewy Bodies. Nature 1997, 388, 839– 840, DOI: 10.1038/421665https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXlslWru7o%253D&md5=ef5d22aaeb6f08828ea33c50e3f7c06dα-Synuclein in Lewy bodiesSpillantini, Maria Grazia; Schmidt, Marie Luise; Lee, Virginia M.-Y.; Trojanowski, John Q.; Jakes, Ross; Goedert, MichelNature (London) (1997), 388 (6645), 839-840CODEN: NATUAS; ISSN:0028-0836. (Macmillan Magazines)The authors report strong staining of Lewy bodies in brain in idiopathic Parkinson's disease and dementia with Lewy bodies with antibodies for α-synuclein. They conclude that Parkinson's disease and dementia with Lewy bodies may both be α-synuclein diseases.
- 6Goedert, M.; Spillantini, M. G.; Del Tredici, K.; Braak, H. 100 Years of Lewy Pathology. Nat. Rev. Neurol. 2013, 9, 13– 24, DOI: 10.1038/nrneurol.2012.2426https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXntlWr&md5=947a357746ed61eeab6d2891bf10b013100 years of Lewy pathologyGoedert, Michel; Spillantini, Maria Grazia; Del Tredici, Kelly; Braak, HeikoNature Reviews Neurology (2013), 9 (1), 13-24CODEN: NRNACP; ISSN:1759-4758. (Nature Publishing Group)A review. In 1817, James Parkinson described the symptoms of the shaking palsy, a disease that was subsequently defined in greater detail, and named after Parkinson, by Jean-Martin Charcot. Parkinson expected that the publication of his monograph would lead to a rapid elucidation of the anatomical substrate of the shaking palsy; in the event, this process took almost a century. In 1912, Fritz Heinrich Lewy identified the protein aggregates that define Parkinson disease (PD) in some brain regions outside the substantia nigra. In 1919, Konstantin Nikolaevich Tretiakoff found similar aggregates in the substantia nigra and named them after Lewy. In the 1990s, α-synuclein was identified as the main constituent of the Lewy pathol., and its aggregation was shown to be central to PD, dementia with Lewy bodies, and multiple system atrophy. In 2003, a staging scheme for idiopathic PD was introduced, according to which α-synuclein pathol. originates in the dorsal motor nucleus of the vagal nerve and progresses from there to other brain regions, including the substantia nigra. In this article, we review the relevance of Lewy's discovery 100 years ago for the current understanding of PD and related disorders.
- 7Fusco, G.; Chen, S. W.; Williamson, P. T. F.; Cascella, R.; Perni, M.; Jarvis, J. A.; Cecchi, C.; Vendruscolo, M.; Chiti, F.; Cremades, N.; Ying, L. Structural Basis of Membrane Disruption and Cellular Toxicity by α-Synuclein Oligomers. Science 2017, 358, 1440– 1443, DOI: 10.1126/science.aan61607https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvFGmtrzN&md5=5e54a267d090e6f747aeca57de8b19b8Structural basis of membrane disruption and cellular toxicity by α-synuclein oligomersFusco, Giuliana; Chen, Serene W.; Williamson, Philip T. F.; Cascella, Roberta; Perni, Michele; Jarvis, James A.; Cecchi, Cristina; Vendruscolo, Michele; Chiti, Fabrizio; Cremades, Nunilo; Ying, Liming; Dobson, Christopher M.; De Simone, AlfonsoScience (Washington, DC, United States) (2017), 358 (6369), 1440-1443CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Oligomeric species populated during the aggregation process of α-synuclein have been linked to neuronal impairment in Parkinson's disease and related neurodegenerative disorders. By using soln. and solid-state NMR techniques in conjunction with other structural methods, we identified the fundamental characteristics that enable toxic α-synuclein oligomers to perturb biol. membranes and disrupt cellular function; these include a highly lipophilic element that promotes strong membrane interactions and a structured region that inserts into lipid bilayers and disrupts their integrity. In support of these conclusions, mutations that target the region that promotes strong membrane interactions by α-synuclein oligomers suppressed their toxicity in neuroblastoma cells and primary cortical neurons.
- 8Lashuel, H. A.; Overk, C. R.; Oueslati, A.; Masliah, E. The Many Faces of α-Synuclein: From Structure and Toxicity to Therapeutic Target. Nat. Rev. Neurosci. 2013, 14, 38– 48, DOI: 10.1038/nrn34068https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVGls7jN&md5=930fda9ece4cfa715d3fa58d496e7a13The many faces of α-synuclein: from structure and toxicity to therapeutic targetLashuel, Hilal A.; Overk, Cassia R.; Oueslati, Abid; Masliah, EliezerNature Reviews Neuroscience (2013), 14 (1), 38-48CODEN: NRNAAN; ISSN:1471-003X. (Nature Publishing Group)A review. Disorders characterized by α-synuclein (α-syn) accumulation, Lewy body formation and parkinsonism (and in some cases dementia) are collectively known as Lewy body diseases. The mol. mechanism (or mechanisms) through which α-syn abnormally accumulates and contributes to neurodegeneration in these disorders remains unknown. Here, we provide an overview of current knowledge and prevailing hypotheses regarding the conformational, oligomerization and aggregation states of α-syn and their role in regulating α-syn function in health and disease. Understanding the nature of the various α-syn structures, how they are formed and their relative contributions to α-syn-mediated toxicity may inform future studies aiming to develop therapeutic prevention and intervention.
- 9Haass, C.; Selkoe, D. J. Soluble Protein Oligomers in Neurodegeneration: Lessons from the Alzheimer’s Amyloid β-Peptide. Nat. Rev. Mol. Cell Biol. 2007, 8, 101– 112, DOI: 10.1038/nrm21019https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXotFKmtw%253D%253D&md5=e8634ff5c1491580f623c90ded91b4beSoluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid β-peptideHaass, Christian; Selkoe, Dennis J.Nature Reviews Molecular Cell Biology (2007), 8 (2), 101-112CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. The distinct protein aggregates that are found in Alzheimer's, Parkinson's, Huntington's and prion diseases seem to cause these disorders. Small intermediates - sol. oligomers - in the aggregation process can confer synaptic dysfunction, whereas large, insol. deposits might function as reservoirs of the bioactive oligomers. These emerging concepts are exemplified by Alzheimer's disease, in which amyloid β-protein oligomers adversely affect synaptic structure and plasticity. Findings in other neurodegenerative diseases indicate that a broadly similar process of neuronal dysfunction is induced by diffusible oligomers of misfolded proteins.
- 10Prots, I.; Grosch, J.; Brazdis, R. M.; Simmnacher, K.; Veber, V.; Havlicek, S.; Hannappel, C.; Krach, F.; Krumbiegel, M.; Schütz, O.; Reis, A.; Wrasidlo, W.; Galasko, D. R.; Groemer, T. W.; Masliah, E.; Schlötzer-Schrehardt, U.; Xiang, W.; Winkler, J.; Winner, B. α-Synuclein Oligomers Induce Early Axonal Dysfunction in Human IPSC-Based Models of Synucleinopathies. Proc. Natl. Acad. Sci. U.S.A. 2018, 115, 7813– 7818, DOI: 10.1073/pnas.171312911510https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFCmtrjL&md5=583d4ea493a22aafa3aeb0af58f0755bα-Synuclein oligomers induce early axonal dysfunction in human iPSC-based models of synucleinopathiesProts, Iryna; Grosch, Janina; Brazdis, Razvan-Marius; Simmnacher, Katrin; Veber, Vanesa; Havlicek, Steven; Hannappel, Christian; Krach, Florian; Krumbiegel, Mandy; Schuetz, Oliver; Reis, Andre; Wrasidlo, Wolfgang; Galasko, Douglas R.; Groemer, Teja W.; Masliah, Eliezer; Schloetzer-Schrehardt, Ursula; Xiang, Wei; Winkler, Juergen; Winner, BeateProceedings of the National Academy of Sciences of the United States of America (2018), 115 (30), 7813-7818CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)α-Synuclein (α-Syn) aggregation, proceeding from oligomers to fibrils, is one central hallmark of neurodegeneration in synucleinopathies. α-Syn oligomers are toxic by triggering neurodegenerative processes in in vitro and in vivo models. However, the precise contribution of α-Syn oligomers to neurite pathol. in human neurons and the underlying mechanisms remain unclear. Here, we demonstrate the formation of oligomeric α-Syn intermediates and reduced axonal mitochondrial transport in human neurons derived from induced pluripotent stem cells (iPSC) from a Parkinson's disease patient carrying an α-Syn gene duplication. We further show that increased levels of α-Syn oligomers disrupt axonal integrity in human neurons. We apply an α-Syn oligomerization model by expressing α-Syn oligomer-forming mutants (E46K and E57K) and wild-type α-Syn in human iPSC-derived neurons. Pronounced α-Syn oligomerization led to impaired anterograde axonal transport of mitochondria, which can be restored by the inhibition of α-Syn oligomer formation. Furthermore, α-Syn oligomers were assocd. with a subcellular relocation of transport-regulating proteins Miro1, KLC1, and Tau as well as reduced ATP levels, underlying axonal transport deficits. Consequently, reduced axonal d. and structural synaptic degeneration were obsd. in human neurons in the presence of high levels of α-Syn oligomers. Together, increased dosage of α-Syn resulting in α-Syn oligomerization causes axonal transport disruption and energy deficits, leading to synapse loss in human neurons. This study identifies α-Syn oligomers as the crit. species triggering early axonal dysfunction in synucleinopathies.
- 11Ludtmann, M. H. R.; Angelova, P. R.; Horrocks, M. H.; Choi, M. L.; Rodrigues, M.; Baev, A. Y.; Berezhnov, A. V.; Yao, Z.; Little, D.; Banushi, B.; Al-Menhali, A. S.; Ranasinghe, R. T.; Whiten, D. R.; Yapom, R.; Dolt, K. S.; Devine, M. J.; Gissen, P.; Kunath, T.; Jaganjac, M.; Pavlov, E. V.; Klenerman, D.; Abramov, A. Y.; Gandhi, S. α-Synuclein Oligomers Interact with ATP Synthase and Open the Permeability Transition Pore in Parkinson’s Disease. Nat. Commun. 2018, 9, 2293 DOI: 10.1038/s41467-018-04422-211https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1Mbms1Kjsw%253D%253D&md5=7f4729f901fbb58e29c180ab98999bbaα-synuclein oligomers interact with ATP synthase and open the permeability transition pore in Parkinson's diseaseLudtmann Marthe H R; Angelova Plamena R; Abramov Andrey Y; Ludtmann Marthe H R; Horrocks Mathew H; Rodrigues Margarida; Ranasinghe Rohan T; Whiten Daniel R; Klenerman David; Horrocks Mathew H; Horrocks Mathew H; Choi Minee L; Yao Zhi; Gandhi Sonia; Choi Minee L; Yao Zhi; Gandhi Sonia; Baev Artyom Y; Berezhnov Alexey V; Little Daniel; Banushi Blerida; Devine Michael J; Gissen Paul; Al-Menhali Afnan Saleh; Jaganjac Morana; Yapom Ratsuda; Dolt Karamjit Singh; Kunath Tilo; Devine Michael J; Pavlov Evgeny V; Klenerman DavidNature communications (2018), 9 (1), 2293 ISSN:.Protein aggregation causes α-synuclein to switch from its physiological role to a pathological toxic gain of function. Under physiological conditions, monomeric α-synuclein improves ATP synthase efficiency. Here, we report that aggregation of monomers generates beta sheet-rich oligomers that localise to the mitochondria in close proximity to several mitochondrial proteins including ATP synthase. Oligomeric α-synuclein impairs complex I-dependent respiration. Oligomers induce selective oxidation of the ATP synthase beta subunit and mitochondrial lipid peroxidation. These oxidation events increase the probability of permeability transition pore (PTP) opening, triggering mitochondrial swelling, and ultimately cell death. Notably, inhibition of oligomer-induced oxidation prevents the pathological induction of PTP. Inducible pluripotent stem cells (iPSC)-derived neurons bearing SNCA triplication, generate α-synuclein aggregates that interact with the ATP synthase and induce PTP opening, leading to neuronal death. This study shows how the transition of α-synuclein from its monomeric to oligomeric structure alters its functional consequences in Parkinson's disease.
- 12Hughes, C. D.; Choi, M. L.; Ryten, M.; Hopkins, L.; Drews, A.; Botía, J. A.; Iljina, M.; Rodrigues, M.; Gagliano, S. A.; Gandhi, S.; Klenerman, D.; Bryant, C. Picomolar Concentrations of Oligomeric Alpha - Synuclein Sensitizes TLR4 to Play an Initiating Role in Parkinson’ s Disease Pathogenesis. Acta Neuropathol. 2019, 137, 103– 120, DOI: 10.1007/s00401-018-1907-y12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslCjtrjJ&md5=d54754b630183e9c7fbea6a8ff763c49Picomolar concentrations of oligomeric alpha-synuclein sensitizes TLR4 to play an initiating role in Parkinson's disease pathogenesisHughes, Craig D.; Choi, Minee L.; Ryten, Mina; Hopkins, Lee; Drews, Anna; Botia, Juan A.; Iljina, Maria; Rodrigues, Magarida; Gagliano, Sarah A.; Gandhi, Sonia; Bryant, Clare; Klenerman, DavidActa Neuropathologica (2019), 137 (1), 103-120CODEN: ANPTAL; ISSN:0001-6322. (Springer)Despite the wealth of genomic and transcriptomic data in Parkinson's disease (PD), the initial mol. events are unknown. Using LD score regression anal., we show significant enrichment in PD heritability within regulatory sites for LPS-activated monocytes and that TLR4 expression is highest within human substantia nigra, the most affected brain region, suggesting a role for TLR4 inflammatory responses. We then performed extended incubation of cells with physiol. concns. of small alpha-synuclein oligomers observing the development of a TLR4-dependent sensitized inflammatory response with time, including TNF-α prodn. ROS and cell death in primary neuronal cultures were significantly reduced by TLR4 antagonists revealing that an indirect inflammatory mechanism involving cytokines produced by glial cells makes a major contribution to neuronal death. Prolonged exposure to low levels of alpha-synuclein oligomers sensitizes TLR4 responsiveness in astrocytes and microglial, explaining how they become pro-inflammatory, and may be an early causative event in PD.
- 13Li, B.; Ge, P.; Murray, K. A.; Sheth, P.; Zhang, M.; Nair, G.; Sawaya, M. R.; Shin, W. S.; Boyer, D. R.; Ye, S.; Eisenberg, D. S.; Zhou, Z. H.; Jiang, L. Cryo-EM of Full-Length α-Synuclein Reveals Fibril Polymorphs with a Common Structural Kernel. Nat. Commun. 2018, 9, 3609 DOI: 10.1038/s41467-018-05971-213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3c3mtlCmsw%253D%253D&md5=e3cd60ca2cf31d76e4d33fccd8bfa683Cryo-EM of full-length α-synuclein reveals fibril polymorphs with a common structural kernelLi Binsen; Sheth Phorum; Nair Gayatri; Shin Woo Shik; Jiang Lin; Ge Peng; Ye Shulin; Zhou Z Hong; Murray Kevin A; Zhang Meng; Sawaya Michael R; Boyer David R; Eisenberg David S; Zhou Z HongNature communications (2018), 9 (1), 3609 ISSN:.α-Synuclein (aSyn) fibrillar polymorphs have distinct in vitro and in vivo seeding activities, contributing differently to synucleinopathies. Despite numerous prior attempts, how polymorphic aSyn fibrils differ in atomic structure remains elusive. Here, we present fibril polymorphs from the full-length recombinant human aSyn and their seeding capacity and cytotoxicity in vitro. By cryo-electron microscopy helical reconstruction, we determine the structures of the two predominant species, a rod and a twister, both at 3.7 ÅA resolution. Our atomic models reveal that both polymorphs share a kernel structure of a bent β-arch, but differ in their inter-protofilament interfaces. Thus, different packing of the same kernel structure gives rise to distinct fibril polymorphs. Analyses of disease-related familial mutations suggest their potential contribution to the pathogenesis of synucleinopathies by altering population distribution of the fibril polymorphs. Drug design targeting amyloid fibrils in neurodegenerative diseases should consider the formation and distribution of concurrent fibril polymorphs.
- 14Guerrero-Ferreira, R.; Taylor, N. M. I.; Mona, D.; Ringler, P.; Lauer, M. E.; Riek, R.; Britschgi, M.; Stahlberg, H. Cryo-EM Structure of Alpha-Synuclein Fibrils. eLife 2018, 7, e36402 DOI: 10.7554/eLife.3640214https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlyrtL3F&md5=c1f72f2655276d3eb3ec4e030ff2cdcaCryo-EM structure of alpha-synuclein fibrilsGuerrero-Ferreira, Ricardo; Taylor, Nicholas M. I.; Mona, Daniel; Ringler, Philippe; Lauer, Matthias E.; Riek, Roland; Britschgi, Markus; Stahlberg, HenningeLife (2018), 7 (), e36402/1-e36402/18CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Parkinson's disease is a progressive neuropathol. disorder that belongs to the class of synucleinopathies, in which the protein alpha-synuclein is found at abnormally high concns. in affected neurons. Its hallmark are intracellular inclusions called Lewy bodies and Lewy neurites. We here report the structure of cytotoxic alpha-synuclein fibrils (residues 1-121), detd. by cryo-electron microscopy at a resoln. of 3.4 Å . Two protofilaments form a polar fibril composed of staggered b-strands. The backbone of residues 38 to 95, including the fibril core and the non-amyloid component region, are well resolved in the EM map. Residues 50-57, contg. three of the mutation sites assocd. with familial synucleinopathies, form the interface between the two protofilaments and contribute to fibril stability. A hydrophobic cleft at one end of the fibril may have implications for fibril elongation, and invites for the design of mols. for diagnosis and treatment of synucleinopathies.
- 15Li, Y.; Zhao, C.; Luo, F.; Liu, Z.; Gui, X.; Luo, Z.; Zhang, X.; Li, D.; Liu, C.; Li, X. Amyloid Fibril Structure of α-Synuclein Determined by Cryo-Electron Microscopy. Cell Res. 2018, 28, 897– 903, DOI: 10.1038/s41422-018-0075-x15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVahtbvL&md5=053ffd5d6d3beca7c9a3fa322d243b83Amyloid fibril structure of α-synuclein determined by cryo-electron microscopyLi, Yaowang; Zhao, Chunyu; Luo, Feng; Liu, Zhenying; Gui, Xinrui; Luo, Zhipu; Zhang, Xiang; Li, Dan; Liu, Cong; Li, XuemingCell Research (2018), 28 (9), 897-903CODEN: CREEB6; ISSN:1001-0602. (Nature Research)α-Synuclein (α-syn) amyloid fibrils are the major component of Lewy bodies, which are the pathol. hallmark of Parkinson's disease (PD) and other synucleinopathies. High-resoln. structure of α-syn fibril is important for understanding its assembly and pathol. mechanism. Here, we detd. a fibril structure of full-length α-syn (1-140) at the resoln. of 3.07 Å by cryo-electron microscopy (cryo-EM). The fibrils are cytotoxic, and transmissible to induce endogenous α-syn aggregation in primary neurons. Based on the reconstructed cryo-EM d. map, we were able to unambiguously build the fibril structure comprising residues 37-99. The α-syn amyloid fibril structure shows two protofilaments intertwining along an approx. 21 screw axis into a left-handed helix. Each protofilament features a Greek key-like topol. Remarkably, five out of the six early-onset PD familial mutations are located at the dimer interface of the fibril (H50Q, G51D, and A53T/E) or involved in the stabilization of the protofilament (E46K). Furthermore, these PD mutations lead to the formation of fibrils with polymorphic structures distinct from that of the wild-type. Our study provides mol. insight into the fibrillar assembly of α-syn at the at. level and sheds light on the mol. pathogenesis caused by familial PD mutations of α-syn.
- 16Rodriguez, J. A.; Ivanova, M. I.; Sawaya, M. R.; Cascio, D.; Reyes, F. E.; Shi, D.; Sangwan, S.; Guenther, E. L.; Johnson, L. M.; Zhang, M.; Jiang, L.; Arbing, M. A.; Nannenga, B. L.; Hattne, J.; Whitelegge, J.; Brewster, A. S.; Messerschmidt, M.; Boutet, S.; Sauter, N. K.; Gonen, T.; Eisenberg, D. S. Structure of the Toxic Core of α-Synuclein from Invisible Crystals. Nature 2015, 525, 486– 490, DOI: 10.1038/nature1536816https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVOls7zN&md5=a6c64c087d6e5b10e4190bfdf2267665Structure of the toxic core of α-synuclein from invisible crystalsRodriguez, Jose A.; Ivanova, Magdalena I.; Sawaya, Michael R.; Cascio, Duilio; Reyes, Francis E.; Shi, Dan; Sangwan, Smriti; Guenther, Elizabeth L.; Johnson, Lisa M.; Zhang, Meng; Jiang, Lin; Arbing, Mark A.; Nannenga, Brent L.; Hattne, Johan; Whitelegge, Julian; Brewster, Aaron S.; Messerschmidt, Marc; Boutet, Sebastien; Sauter, Nicholas K.; Gonen, Tamir; Eisenberg, David S.Nature (London, United Kingdom) (2015), 525 (7570), 486-490CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)α-Synuclein is the main component of Lewy bodies, the neuron-assocd. aggregates seen in Parkinson disease and other neurodegenerative pathologies. An 11-residue segment, which the authors term NACore, appears to be responsible for amyloid formation and cytotoxicity of human α-synuclein. Here, the authors describe crystals of NACore that have dimensions smaller than the wavelength of visible light and thus are invisible by optical microscopy. As the crystals are thousands of times too small for structure detn. by synchrotron x-ray diffraction, the authors used micro-electron diffraction to det. the structure at at. resoln. The 1.4-Å-resoln. structure demonstrated that this method can det. previously unknown protein structures and here yielded, to the authors' knowledge, the highest resoln. achieved by any cryo-electron microscopy method to date. The structure exhibited protofibrils built of pairs of face-to-face β-sheets. X-ray fiber diffraction patterns showed the similarity of NACore to toxic fibrils of full-length α-synuclein. The NACore structure, together with that of a 2nd segment, inspired a model for most of the ordered portion of the toxic, full-length α-synuclein fibril, presenting opportunities for the design of inhibitors of α-synuclein fibrils.
- 17Schweighauser, M.; Shi, Y.; Tarutani, A.; Kametani, F.; Murzin, A. G.; Ghetti, B.; Matsubara, T.; Tomita, T.; Ando, T.; Hasegawa, K.; Murayama, S.; Yoshida, M.; Hasegawa, M.; Scheres, S. H. W.; Goedert, M. Structures of α-Synuclein Filaments from Multiple System Atrophy. Nature 2020, 585, 464– 469, DOI: 10.1038/s41586-020-2317-617https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVantbjK&md5=2227413593f48413b7129ff9d8128e6eStructures of α-synuclein filaments from multiple system atrophySchweighauser, Manuel; Shi, Yang; Tarutani, Airi; Kametani, Fuyuki; Murzin, Alexey G.; Ghetti, Bernardino; Matsubara, Tomoyasu; Tomita, Taisuke; Ando, Takashi; Hasegawa, Kazuko; Murayama, Shigeo; Yoshida, Mari; Hasegawa, Masato; Scheres, Sjors H. W.; Goedert, MichelNature (London, United Kingdom) (2020), 585 (7825), 464-469CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Synucleinopathies, which include multiple system atrophy (MSA), Parkinson's disease, Parkinson's disease with dementia and dementia with Lewy bodies (DLB), are human neurodegenerative diseases1. Existing treatments are at best symptomatic. These diseases are characterized by the presence of, and believed to be caused by the formation of, filamentous inclusions of α-synuclein in brain cells2,3. However, the structures of α-synuclein filaments from the human brain are unknown. Here, using cryo-electron microscopy, we show that α-synuclein inclusions from the brains of individuals with MSA are made of two types of filament, each of which consists of two different protofilaments. In each type of filament, non-proteinaceous mols. are present at the interface of the two protofilaments. Using two-dimensional class averaging, we show that α-synuclein filaments from the brains of individuals with MSA differ from those of individuals with DLB, which suggests that distinct conformers or strains characterize specific synucleinopathies. As is the case with tau assemblies4-9, the structures of α-synuclein filaments extd. from the brains of individuals with MSA differ from those formed in vitro using recombinant proteins, which has implications for understanding the mechanisms of aggregate propagation and neurodegeneration in the human brain. These findings have diagnostic and potential therapeutic relevance, esp. because of the unmet clin. need to be able to image filamentous α-synuclein inclusions in the human brain.
- 18Sangwan, S.; Sahay, S.; Murray, K. A.; Morgan, S.; Guenther, E. L.; Jiang, L.; Williams, C. K.; Vinters, H. V.; Goedert, M.; Eisenberg, D. S. Inhibition of Synucleinopathic Seeding by Rationally Designed Inhibitors. eLife 2020, 9, e46775 DOI: 10.7554/eLife.4677518https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlarsLrM&md5=1161e8cdb0482e8bee3cd6253a3cd4f5Inhibition of synucleinopathic seeding by rationally designed inhibitorsSangwan, Smriti; Sahay, Shruti; Murray, Kevin A.; Morgan, Sophie; Guenther, Elizabeth L.; Jiang, Lin; Williams, Christopher K.; Vinters, Harry V.; Goedert, Michel; Eisenberg, David S.eLife (2020), 9 (), e46775CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Seeding, in the context of amyloid disease, is the sequential transfer of pathogenic protein aggregates from cell-to-cell within affected tissues. The structure of pathogenic seeds provides the mol. basis and enables rapid conversion of sol. protein into fibrils. To date, there are no inhibitors that specifically target seeding of Parkinson's disease (PD)-assocd. asynuclein (a-syn) fibrils, in part, due to lack of information of the structural properties of pathol. seeds. Here we design small peptidic inhibitors based on the at. structure of the core of a-syn fibrils. The inhibitors prevent a-syn aggregation in vitro and in cell culture models with binding affinities of 0.5 mM to a-syn fibril seeds. The inhibitors also show efficacy in preventing seeding by human patient-derived a-syn fibrils. Our results suggest that pathogenic seeds of a-syn contain steric zippers and suggest a therapeutic approach targeted at the spread and progression that may be applicable for PD and related synucleinopathies.
- 19Priss, A.; Afitska, K.; Galkin, M.; Yushchenko, D. A.; Shvadchak, V. V. Rationally Designed Protein-Based Inhibitor of α-Synuclein Fibrillization in Cells. J. Med. Chem. 2021, 64, 6827– 6837, DOI: 10.1021/acs.jmedchem.1c0008619https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVCiu7%252FM&md5=42e14747550775b61e5c84d161b58efaRationally Designed Protein-Based Inhibitor of α-Synuclein Fibrillization in CellsPriss, Anastasiia; Afitska, Kseniia; Galkin, Maksym; Yushchenko, Dmytro A.; Shvadchak, Volodymyr V.Journal of Medicinal Chemistry (2021), 64 (10), 6827-6837CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Misfolding of the neuronal protein α-synuclein (αSyn) into amyloid fibrils is involved in the development of Parkinson's disease (PD), and inhibition of this process is considered to be a promising therapeutic approach. In this work, we engineered protein inhibitors that bind to fibrils with higher affinity than the monomeric αSyn. They were developed based on the recent structural data of the αSyn fibrils and were shown to prevent fibril elongation upon binding to fibril ends. These inhibitors are highly selective to the misfolded αSyn, nontoxic, and active in cytosol in small concns. The best-performing inhibitor shows IC50 ~ 10 nM in a cell-based assay, which corresponds to the ~ 1:60 molar ratio to αSyn. It can suppress the formation of αSyn aggregates in cells that can be potentially used to slow down the spreading of the pathol. aggregates from cell to cell during the course of the PD.
- 20Gao, L.; Wang, W.; Wang, X.; Yang, F.; Xie, L.; Shen, J.; Brimble, M. A.; Xiao, Q.; Yao, S. Q. Fluorescent Probes for Bioimaging of Potential Biomarkers in Parkinson’s Disease. Chem. Soc. Rev. 2021, 50, 1219– 1250, DOI: 10.1039/d0cs00115e20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFSgtb%252FE&md5=ab063111c2d1f8f66453b1bff9c5becdFluorescent probes for bioimaging of potential biomarkers in Parkinson's diseaseGao, Liqian; Wang, Wei; Wang, Xuan; Yang, Fen; Xie, Liuxing; Shen, Jun; Brimble, Margaret A.; Xiao, Qicai; Yao, Shao Q.Chemical Society Reviews (2021), 50 (2), 1219-1250CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Parkinson's disease (PD), as the second most common neurodegenerative disease, is caused by complex pathol. processes and currently remains very difficult to treat. PD brings great distress to patients and imposes a heavy economic burden on society. The no. of PD patients is growing as the aging population increases worldwide. Therefore, it is crucial to develop new tools for aiding the early diagnosis and treatment of PD. The significant pathol. features involved in PD include the abnormal accumulation of α-synuclein, metal ion dyshomeostasis, oxidative stress, mitochondrial dysfunction and neurotransmitter deficiencies. In recent years, fluorescent probes have emerged as a powerful bioimaging tool with potential to help understand the pathol. processes of PD via the detection and monitoring of pathol. features. In this review, we comprehensively summarize the design and working mechanisms of fluorescent probes along with their applications in the detection of various PD biomarkers. We also discuss the current limitations of fluorescent probes and provide perspectives on how these limitations can be overcome to develop better fluorescent probes suitable for application in clin. trials in the future. We hope that this review provides valuable information and guidance for the development of new fluorescent probes that can be used clin. in the early diagnosis of PD and contributes to the development of efficient PD drugs in the future.
- 21Ferrie, J. J.; Lengyel-Zhand, Z.; Janssen, B.; Lougee, M. G.; Giannakoulias, S.; Hsieh, C.-J.; Pagar, V. V.; Weng, C.-C.; Xu, H.; Graham, T. J. A.; Lee, V. M.-Y.; Mach, R. H.; Petersson, E. J. Identification of a Nanomolar Affinity α-Synuclein Fibril Imaging Probe by Ultra-High Throughput in Silico Screening. Chem. Sci. 2020, 11, 12746– 12754, DOI: 10.1039/d0sc02159h21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsl2ktb%252FJ&md5=9a34fe33b5d896d3bf60cbe264d74452Identification of a nanomolar affinity alpha-synuclein fibril imaging probe by ultra-high throughput in silico screeningFerrie, John J.; Lengyel-Zhand, Zsofia; Janssen, Bieneke; Lougee, Marshall G.; Giannakoulias, Sam; Hsieh, Chia-Ju; Pagar, Vinayak Vishnu; Weng, Chi-Chang; Xu, Hong; Graham, Thomas J. A.; Lee, Virginia M.-Y.; Mach, Robert H.; Petersson, E. JamesChemical Science (2020), 11 (47), 12746-12754CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Small mols. that bind with high affinity and specificity to fibrils of the α-synuclein (αS) protein have the potential to serve as positron emission tomog. (PET) imaging probes to aid in the diagnosis of Parkinson's disease and related synucleinopathies. To identify such mols., we employed an ultra-high throughput in silico screening strategy using idealized pseudo-ligands termed exemplars to identify compds. for exptl. binding studies. For the top hit from this screen, we used photo-crosslinking to confirm its binding site and studied the structure-activity relationship of its analogs to develop multiple mols. with nanomolar affinity for αS fibrils and moderate specificity for α;S over Aβfibrils. Lastly, we demonstrated the potential of the lead analog as an imaging probe by measuring binding to αS-enriched homogenates from mouse brain tissue using a radiolabeled analog of the identified mols. This study demonstrates the validity of our powerful new approach to the discovery of PET probes for challenging mol. targets.
- 22Lengyel-Zhand, Z.; Ferrie, J. J.; Janssen, B.; Hsieh, C. J.; Graham, T.; Xu, K. Y.; Haney, C. M.; Lee, V. M. Y.; Trojanowski, J. Q.; Petersson, E. J.; Mach, R. H. Synthesis and Characterization of High Affinity Fluorogenic α-Synuclein Probes. Chem. Commun. 2020, 56, 3567– 3570, DOI: 10.1039/c9cc09849f22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjsF2ntrw%253D&md5=a2e24885326f473f4ede5c722d027692Synthesis and characterization of high affinity fluorogenic α-synuclein probesLengyel-Zhand, Zsofia; Ferrie, John J.; Janssen, Bieneke; Hsieh, Chia-Ju; Graham, Thomas; Xu, Kui-ying; Haney, Conor M.; Lee, Virginia M.-Y.; Trojanowski, John Q.; Petersson, E. James; Mach, Robert H.Chemical Communications (Cambridge, United Kingdom) (2020), 56 (24), 3567-3570CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)Fluorescent small mols. are powerful tools for imaging α-synuclein pathol. in vitro and in vivo. In this work, we explore benzofuranone as a potential scaffold for the design of fluorescent α-synuclein probes. These compds. have high affinity for α-synuclein, show fluorescent turn-on upon binding to fibrils, and display different binding to Lewy bodies, Lewy neurites and glial cytoplasmic inclusion pathologies in post-mortem brain tissue. These studies not only reveal the potential of benzofuranone compds. as α-synuclein specific fluorescent probes, but also have implications for the ways in which α-synucleinopathies are conformationally different and display distinct small mol. binding sites.
- 23Hsieh, C. J.; Ferrie, J. J.; Xu, K.; Lee, I.; Graham, T. J. A.; Tu, Z.; Yu, J.; Dhavale, D.; Kotzbauer, P.; Petersson, E. J.; Mach, R. H. Alpha Synuclein Fibrils Contain Multiple Binding Sites for Small Molecules. ACS Chem. Neurosci. 2018, 9, 2521– 2527, DOI: 10.1021/acschemneuro.8b0017723https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXptlajurs%253D&md5=f16af627b180f8b1cac79ff08bebc643Alpha Synuclein Fibrils Contain Multiple Binding Sites for Small MoleculesHsieh, Chia-Ju; Ferrie, John J.; Xu, Kuiying; Lee, Iljung; Graham, Thomas J. A.; Tu, Zhude; Yu, Jennifer; Dhavale, Dhruva; Kotzbauer, Paul; Petersson, E. James; Mach, Robert H.ACS Chemical Neuroscience (2018), 9 (11), 2521-2527CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)The fibrillary aggregation of the α-synuclein (Asyn) is a hallmark of Parkinson's disease, and the identification of small mol. binding sites on fibrils is essential to the development of diagnostic imaging probes. Here, a series of mol. modeling, photoaffinity labeling, mass spectrometry,, and radioligand binding studies were conducted on Asyn fibrils. The results of these studies revealed the presence of 3 different binding sites within fibrillar Asyn capable of binding small mols. with moderate to high affinity. A knowledge of the amino acid residues in these binding sites will be important in the design of high affinity probes capable of imaging fibrillary species of Asyn.
- 24Eberling, J. L.; Dave, K. D.; Frasier, M. A. α-Synuclein Imaging: A Critical Need for Parkinson’s Disease Research. J. Parkinson’s Dis. 2013, 3, 565– 567, DOI: 10.3233/JPD-13024724https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvV2iur%252FO&md5=74cb27856e9d5c7b593b0c357ef7138aα-synuclein Imaging: A Critical Need for Parkinson's Disease ResearchEberling, Jamie L.; Dave, Kuldip D.; Frasier, Mark A.Journal of Parkinson's Disease (2013), 3 (4), 565-567CODEN: JPDOAP; ISSN:1877-7171. (IOS Press)A review. The development of an α-synuclein imaging agent could be transformative for Parkinson's disease research and drug development. The ability to image α-synuclein in the brain would enable tracking of the degree and location of pathol. over time and monitoring of therapies aimed at reducing α-synuclein levels. The Michael J. Fox Foundation has assembled a consortium of researchers to develop an α-synuclein radiotracer for use in positron emission tomog. (PET) imaging studies. While this poses a no. of challenges they should not be insurmountable and lessons learned from the development of tau radiotracers should provide valuable insights.
- 25Kulenkampff, K.; Wolf Perez, A. M.; Sormanni, P.; Habchi, J.; Vendruscolo, M. Quantifying Misfolded Protein Oligomers as Drug Targets and Biomarkers in Alzheimer and Parkinson Diseases. Nat. Rev. Chem. 2021, 5, 277– 294, DOI: 10.1038/s41570-021-00254-925https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXosFers7s%253D&md5=d7a63b934405847347c68bd061ab34e9Quantifying misfolded protein oligomers as drug targets and biomarkers in Alzheimer and Parkinson diseasesKulenkampff, Klara; Wolf Perez, Adriana-M.; Sormanni, Pietro; Habchi, Johnny; Vendruscolo, MicheleNature Reviews Chemistry (2021), 5 (4), 277-294CODEN: NRCAF7; ISSN:2397-3358. (Nature Portfolio)A review. Protein misfolding and aggregation are characteristic of a wide range of neurodegenerative disorders, including Alzheimer and Parkinson diseases. A hallmark of these diseases is the aggregation of otherwise sol. and functional proteins into amyloid aggregates. Although for many decades such amyloid deposits have been thought to be responsible for disease progression, it is now increasingly recognized that the misfolded protein oligomers formed during aggregation are, instead, the main agents causing pathol. processes. These oligomers are transient and heterogeneous, which makes it difficult to detect and quantify them, generating confusion about their exact role in disease. The lack of suitable methods to address these challenges has hampered efforts to investigate the mol. mechanisms of oligomer toxicity and to develop oligomer-based diagnostic and therapeutic tools to combat protein misfolding diseases. In this Review, we describe methods to quantify misfolded protein oligomers, with particular emphasis on diagnostic applications as disease biomarkers and on therapeutic applications as target biomarkers. The development of these methods is ongoing, and we discuss the challenges that remain to be addressed to establish measurement tools capable of overcoming existing limitations and to meet present needs.
- 26Fayyad, M.; Salim, S.; Majbour, N.; Erskine, D.; Stoops, E.; Mollenhauer, B.; El-Agnaf, O. M. A. Parkinson’s Disease Biomarkers Based on α-Synuclein. J. Neurochem. 2019, 150, 626– 636, DOI: 10.1111/jnc.1480926https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsVCmt7nF&md5=28fbc154ad78042e2a331e234f525d0bParkinson's disease biomarkers based on α-synucleinFayyad, Muneera; Salim, Safa; Majbour, Nour; Erskine, Daniel; Stoops, Erik; Mollenhauer, Brit; El-Agnaf, Omar M. A.Journal of Neurochemistry (2019), 150 (5), 626-636CODEN: JONRA9; ISSN:0022-3042. (Wiley-Blackwell)A review. Parkinson's disease is the second most common neurodegenerative disorder after Alzheimer's disease and is estd. to affect approx. 1-4% of individuals aged over 60 years old. Although considerable efforts have been invested into developing disease-modifying therapies for Parkinson's disease, such efforts have been confounded by the difficulty in accurately diagnosing Parkinson's disease during life to enable accurate patient stratification for clin. trialling of candidate therapeutics. Therefore, the search for effective biomarkers that can be accurately evaluated during life with non-invasive means is a pressing issue in the field. Since the discovery of α-synuclein (α-syn) as a protein linked to a familial form of Parkinson's disease, later identified as the major protein component of the neuropathol. hallmark of idiopathic Parkinson's disease, considerable interest has focused on this protein and its distinct conformers. We describe here the progress that has been made in the area of Parkinson's disease biomarker discovery with a focus on α-synuclein. In particular, we highlight the novel assays that have been employed and the increasing complexity in evaluating α-synuclein with regard to the considerable diversity of conformers that exist in the biofluids and peripheral tissues under disease conditions.
- 27Brown, J. W. P.; Buell, A. K.; Michaels, T. C. T.; Meisl, G.; Carozza, J.; Flagmeier, P.; Vendruscolo, M.; Knowles, T. P. J.; Dobson, C. M.; Galvagnion, C. β-Synuclein Suppresses Both the Initiation and Amplification Steps of α-Synuclein Aggregation via Competitive Binding to Surfaces. Sci. Rep. 2016, 6, 36010 DOI: 10.1038/srep3601027https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVSisbjJ&md5=aa6a38bfdb70917406e08a40448bfc01β-Synuclein suppresses both the initiation and amplification steps of α-synuclein aggregation via competitive binding to surfacesBrown, James W. P.; Buell, Alexander K.; Michaels, Thomas C. T.; Meisl, Georg; Carozza, Jacqueline; Flagmeier, Patrick; Vendruscolo, Michele; Knowles, Tuomas P. J.; Dobson, Christopher M.; Galvagnion, CelineScientific Reports (2016), 6 (), 36010CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)α-Synuclein is an intrinsically disordered protein that is assocd. with the pathogenesis of Parkinson's disease through the processes involved in the formation of amyloid fibrils. α and β-synuclein are homologous proteins found at comparable levels in presynaptic terminals but β-synuclein has a greatly reduced propensity to aggregate and indeed has been found to inhibit α-synuclein aggregation. In this paper, we describe how sequence differences between α- and β-synuclein affect individual microscopic processes in amyloid formation. In particular, we show that β-synuclein strongly suppresses both lipid-induced aggregation and secondary nucleation of α-synuclein by competing for binding sites at the surfaces of lipid vesicles and fibrils, resp. These results suggest that β-synuclein can act as a natural inhibitor of α-synuclein aggregation by reducing both the initiation of its self-assembly and the proliferation of its aggregates.
- 28Staats, R.; Michaels, T. C. T.; Flagmeier, P.; Chia, S.; Horne, R. I.; Habchi, J.; Linse, S.; Knowles, T. P. J.; Dobson, C. M.; Vendruscolo, M. Screening of Small Molecules Using the Inhibition of Oligomer Formation in α-Synuclein Aggregation as a Selection Parameter. Commun. Chem. 2020, 3, 191, DOI: 10.1038/s42004-020-00412-y28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVerur3E&md5=3dedbd30c621b0c6a777f3e6ad7c503eScreening of small molecules using the inhibition of oligomer formation in α-synuclein aggregation as a selection parameterStaats, Roxine; Michaels, Thomas C. T.; Flagmeier, Patrick; Chia, Sean; Horne, Robert I.; Habchi, Johnny; Linse, Sara; Knowles, Tuomas P. J.; Dobson, Christopher M.; Vendruscolo, MicheleCommunications Chemistry (2020), 3 (1), 191CODEN: CCOHCT; ISSN:2399-3669. (Nature Research)The aggregation of α-synuclein is a central event in Parkinsons disease and related synucleinopathies. Since pharmacol. targeting this process, however, has not yet resulted in approved disease-modifying treatments, there is an unmet need of developing novel methods of drug discovery. In this context, the use of chem. kinetics has recently enabled accurate quantifications of the microscopic steps leading to the proliferation of protein misfolded oligomers. As these species are highly neurotoxic, effective therapeutic strategies may be aimed at reducing their nos. Here, we exploit this quant. approach to develop a screening strategy that uses the reactive flux toward α-synuclein oligomers as a selection parameter. Using this approach, we evaluate the efficacy of a library of flavone derivs., identifying apigenin as a compd. that simultaneously delays and reduces the formation of α-synuclein oligomers. These results demonstrate a compd. selection strategy based on the inhibition of the formation of α-synuclein oligomers, which may be key in identifying small mols. in drug discovery pipelines for diseases assocd. with α-synuclein aggregation.
- 29Perni, M.; Flagmeier, P.; Limbocker, R.; Cascella, R.; Aprile, F. A.; Galvagnion, C.; Heller, G. T.; Meisl, G.; Chen, S. W.; Kumita, J. R.; Challa, P. K.; Kirkegaard, J. B.; Cohen, S. I. A.; Mannini, B.; Barbut, D.; Nollen, E. A. A.; Cecchi, C.; Cremades, N.; Knowles, T. P. J.; Chiti, F.; Zasloff, M.; Vendruscolo, M.; Dobson, C. M. Multistep Inhibition of α-Synuclein Aggregation and Toxicity in Vitro and in Vivo by Trodusquemine. ACS Chem. Biol. 2018, 13, 2308– 2319, DOI: 10.1021/acschembio.8b0046629https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1SqtLfN&md5=7bcab9b174a7dd4958233f8d8457834eMultistep Inhibition of α-Synuclein Aggregation and Toxicity in Vitro and in Vivo by TrodusqueminePerni, Michele; Flagmeier, Patrick; Limbocker, Ryan; Cascella, Roberta; Aprile, Francesco A.; Galvagnion, Celine; Heller, Gabriella T.; Meisl, Georg; Chen, Serene W.; Kumita, Janet R.; Challa, Pavan K.; Kirkegaard, Julius B.; Cohen, Samuel I. A.; Mannini, Benedetta; Barbut, Denise; Nollen, Ellen A. A.; Cecchi, Cristina; Cremades, Nunilo; Knowles, Tuomas P. J.; Chiti, Fabrizio; Zasloff, Michael; Vendruscolo, Michele; Dobson, Christopher M.ACS Chemical Biology (2018), 13 (8), 2308-2319CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The aggregation of α-synuclein, an intrinsically disordered protein that is highly abundant in neurons, is closely assocd. with the onset and progression of Parkinson's disease. We have shown previously that the aminosterol squalamine can inhibit the lipid induced initiation process in the aggregation of α-synuclein, and we report here that the related compd. trodusquemine is capable of inhibiting not only this process but also the fibril-dependent secondary pathways in the aggregation reaction. We further demonstrate that trodusquemine can effectively suppress the toxicity of α-synuclein oligomers in neuronal cells, and that its administration, even after the initial growth phase, leads to a dramatic redn. in the no. of α-synuclein inclusions in a Caenorhabditis elegans model of Parkinson's disease, eliminates the related muscle paralysis, and increases lifespan. On the basis of these findings, we show that trodusquemine is able to inhibit multiple events in the aggregation process of α-synuclein and hence to provide important information about the link between such events and neurodegeneration, as it is initiated and progresses. Particularly in the light of the previously reported ability of trodusquemine to cross the blood-brain barrier and to promote tissue regeneration, the present results suggest that this compd. has the potential to be an important therapeutic candidate for Parkinson's disease and related disorders.
- 30Pujols, J.; Peña-Díaz, S.; Pallarès, I.; Ventura, S. Chemical Chaperones as Novel Drugs for Parkinson’s Disease. Trends Mol. Med. 2020, 26, 408– 421, DOI: 10.1016/j.molmed.2020.01.00530https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlGrt7w%253D&md5=e56194e5e4f70b2fec1fff492fd62919Chemical Chaperones as Novel Drugs for Parkinson's DiseasePujols, Jordi; Pena-Diaz, Samuel; Pallares, Irantzu; Ventura, SalvadorTrends in Molecular Medicine (2020), 26 (4), 408-421CODEN: TMMRCY; ISSN:1471-4914. (Elsevier Ltd.)A review. Parkinson's disease (PD) is characterized by progressive loss of dopaminergic neurons and the accumulation of deposits of α-synuclein (α-syn) in the brain. The pivotal role of α-syn aggregation in PD makes it an attractive target for potential disease-modifying therapies. However, the disordered nature of the protein, its multistep aggregation mechanism, and the lack of structural information on intermediate species complicate the discovery of modulators of α-syn amyloid deposition. Despite these difficulties, small mols. have been shown to block the misfolding and aggregation of α-syn, and can even disentangle mature α-syn amyloid fibrils. In this review we provide an updated overview of these leading small compds. and discuss how these chem. chaperones hold great promise to alter the course of PD progression.
- 31Gaspar, R.; Meisl, G.; Buell, A. K.; Young, L.; Kaminski, C. F.; Knowles, T. P. J.; Sparr, E.; Linse, S. Secondary Nucleation of Monomers on Fibril Surface Dominates α-Synuclein Aggregation and Provides Autocatalytic Amyloid Amplification. Q. Rev. Biophys. 2017, 50, e6 DOI: 10.1017/S003358351600017231https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXkvVaqtrw%253D&md5=1b0069435bf692abc35d3b62a6cbac51Secondary nucleation of monomers on fibril surface dominates α-synuclein aggregation and provides autocatalytic amyloid amplificationGaspar, Ricardo; Meisl, Georg; Buell, Alexander K.; Young, Laurence; Kaminski, Clemens F.; Knowles, Tuomas P. J.; Sparr, Emma; Linse, SaraQuarterly Reviews of Biophysics (2017), 50 (), e6/1-e6/12CODEN: QURBAW; ISSN:0033-5835. (Cambridge University Press)Parkinson's disease (PD) is characterized by proteinaceous aggregates named Lewy Bodies and Lewy Neurites contg. α-synuclein fibrils. The underlying aggregation mechanism of this protein is dominated by a secondary process at mildly acidic pH, as in endosomes and other organelles. This effect manifests as a strong acceleration of the aggregation in the presence of seeds and a weak dependence of the aggregation rate on monomer concn. The mol. mechanism underlying this process could be nucleation of monomers on fibril surfaces or fibril fragmentation. Here, we aim to distinguish between these mechanisms. The nature of the secondary processes was investigated using differential sedimentation anal., trap and seed expts., quartz crystal microbalance expts. and super-resoln. microscopy. The results identify secondary nucleation of monomers on the fibril surface as the dominant secondary process leading to rapid generation of new aggregates, while no significant contribution from fragmentation was found. The newly generated oligomeric species quickly elongate to further serve as templates for secondary nucleation and this may have important implications in the spreading of PD.
- 32Michaels, T. C. T.; Lazell, H. W.; Arosio, P.; Knowles, T. P. J. Dynamics of Protein Aggregation and Oligomer Formation Governed by Secondary Nucleation. J. Chem. Phys. 2015, 143, 054901 DOI: 10.1063/1.492765532https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht12gtL7P&md5=43386e2cdf09735afe1c274bca781845Dynamics of protein aggregation and oligomer formation governed by secondary nucleationMichaels, Thomas C. T.; Lazell, Hamish W.; Arosio, Paolo; Knowles, Tuomas P. J.Journal of Chemical Physics (2015), 143 (5), 054901/1-054901/15CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The formation of aggregates in many protein systems can be significantly accelerated by secondary nucleation, a process where existing assemblies catalyze the nucleation of new species. In particular, secondary nucleation has emerged as a central process controlling the proliferation of many filamentous protein structures, including mol. species related to diseases such as sickle cell anemia and a range of neurodegenerative conditions. Increasing evidence suggests that the phys. size of protein filaments plays a key role in detg. their potential for deleterious interactions with living cells, with smaller aggregates of misfolded proteins, oligomers, being particularly toxic. It is thus crucial to progress towards an understanding of the factors that control the sizes of protein aggregates. However, the influence of secondary nucleation on the time evolution of aggregate size distributions has been challenging to quantify. This difficulty originates in large part from the fact that secondary nucleation couples the dynamics of species distant in size space. Here, we approach this problem by presenting an anal. treatment of the master equation describing the growth kinetics of linear protein structures proliferating through secondary nucleation and provide closed-form expressions for the temporal evolution of the resulting aggregate size distribution. We show how the availability of anal. solns. for the full filament distribution allows us to identify the key phys. parameters that control the sizes of growing protein filaments. Furthermore, we use these results to probe the dynamics of the populations of small oligomeric species as they are formed through secondary nucleation and discuss the implications of our work for understanding the factors that promote or curtail the prodn. of these species with a potentially high deleterious biol. activity. (c) 2015 American Institute of Physics.
- 33Chia, S.; Habchi, J.; Michaels, T. C. T.; Cohen, S. I. A.; Linse, S.; Dobson, C. M.; Knowles, T. P. J.; Vendruscolo, M. SAR by Kinetics for Drug Discovery in Protein Misfolding Diseases. Proc. Natl. Acad. Sci. U.S.A. 2018, 115, 10245– 10250, DOI: 10.1073/pnas.180788411533https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFOntLnP&md5=13ac0b5763f8a9779de80af1c9700978SAR by kinetics for drug discovery in protein misfolding diseasesChia, Sean; Habchi, Johnny; Michaels, Thomas C. T.; Cohen, Samuel I. A.; Linse, Sara; Dobson, Christopher M.; Knowles, Tuomas P. J.; Vendruscolo, MicheleProceedings of the National Academy of Sciences of the United States of America (2018), 115 (41), 10245-10250CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)To develop effective therapeutic strategies for protein misfolding diseases, a promising route is to identify compds. that inhibit the formation of protein oligomers. To achieve this goal, the authors report a structure-activity relation (SAR) approach based on chem. kinetics to est. quant. how small mols. modify the reactive flux toward oligomers. The authors use this est. to derive chem. rules in the case of the amyloid beta peptide (Aβ), which the authors then exploit to optimize starting compds. to curtail Aβ oligomer formation. The authors demonstrate this approach by converting an inactive rhodanine compd. into an effective inhibitor of Aβ oligomer formation by generating chem. derivs. in a systematic manner. These results provide an initial demonstration of the potential of drug discovery strategies based on targeting directly the prodn. of protein oligomers.
- 34Le Guilloux, V.; Schmidtke, P.; Tuffery, P. Fpocket: An Open Source Platform for Ligand Pocket Detection. BMC Bioinf. 2009, 10, S6 DOI: 10.1186/1471-2105-10-168There is no corresponding record for this reference.
- 35Buell, A. K.; Galvagnion, C.; Gaspar, R.; Sparr, E.; Vendruscolo, M.; Knowles, T. P. J.; Linse, S.; Dobson, C. M. Solution Conditions Determine the Relative Importance of Nucleation and Growth Processes in α-Synuclein Aggregation. Proc. Natl. Acad. Sci. U.S.A. 2014, 111, 7671– 7676, DOI: 10.1073/pnas.131534611135https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnsl2msL4%253D&md5=f08c789180029c972001b80b401cc78eSolution conditions determine the relative importance of nucleation and growth processes in α-synuclein aggregationBuell, Alexander K.; Galvagnion, Celine; Gaspar, Ricardo; Sparr, Emma; Vendruscolo, Michele; Knowles, Tuomas P. J.; Linse, Sara; Dobson, Christopher M.Proceedings of the National Academy of Sciences of the United States of America (2014), 111 (21), 7671-7676CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The formation of amyloid fibrils by the intrinsically disordered protein α-synuclein is a hallmark of Parkinson's disease (PD). To characterize the microscopic steps in the mechanism of aggregation of this protein, the authors used in vitro aggregation assays in the presence of preformed seed fibrils to det. the mol. rate const. of fibril elongation under a range of different conditions. The authors showed that α-synuclein amyloid fibrils grew by monomer and not oligomer addn. and were subject to higher-order assembly processes that decreased their capacity to grow. The authors also found that at neutral pH under quiescent conditions homogeneous primary nucleation and secondary processes, such as fragmentation and surface-assisted nucleation, which can lead to proliferation of the total no. of aggregates, were undetectable. At pH values of <6, however, the rate of secondary nucleation increased dramatically, leading to a completely different balance between the nucleation and growth of aggregates. Thus, at mildly acidic pH values, such as those, e.g., that are present in some intracellular locations, including endosomes and lysosomes, multiplication of aggregates was much faster than at normal physiol. pH values, largely as a consequence of much more rapid secondary nucleation. These findings provide new insights into possible mechanisms of α-synuclein aggregation and aggregate spreading in the context of PD.
- 36Sterling, T.; Irwin, J. J. ZINC 15 - Ligand Discovery for Everyone. J. Chem. Inf. Model. 2015, 55, 2324– 2337, DOI: 10.1021/acs.jcim.5b0055936https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhs1OhurbF&md5=e767c26c1d0aff7432c403c59a5d2b8bZINC 15 - Ligand Discovery for EveryoneSterling, Teague; Irwin, John J.Journal of Chemical Information and Modeling (2015), 55 (11), 2324-2337CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Many questions about the biol. activity and availability of small mols. remain inaccessible to investigators who could most benefit from their answers. To narrow the gap between chemoinformatics and biol., we have developed a suite of ligand annotation, purchasability, target, and biol. assocn. tools, incorporated into ZINC and meant for investigators who are not computer specialists. The new version contains over 120 million purchasable "drug-like" compds. - effectively all org. mols. that are for sale - a quarter of which are available for immediate delivery. ZINC connects purchasable compds. to high-value ones such as metabolites, drugs, natural products, and annotated compds. from the literature. Compds. may be accessed by the genes for which they are annotated as well as the major and minor target classes to which those genes belong. It offers new anal. tools that are easy for nonspecialists yet with few limitations for experts. ZINC retains its original 3D roots - all mols. are available in biol. relevant, ready-to-dock formats. ZINC is freely available at http://zinc15.docking.org.
- 37Wager, T. T.; Hou, X.; Verhoest, P. R.; Villalobos, A. Moving beyond Rules: The Development of a Central Nervous System Multiparameter Optimization (CNS MPO) Approach to Enable Alignment of Druglike Properties. ACS Chem. Neurosci. 2010, 1, 435– 449, DOI: 10.1021/cn100008c37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjvVejtr8%253D&md5=e94998f65a420be6ee4dec4a987cb983Moving beyond Rules: The Development of a Central Nervous System Multiparameter Optimization (CNS MPO) Approach To Enable Alignment of Druglike PropertiesWager, Travis T.; Hou, Xinjun; Verhoest, Patrick R.; Villalobos, AnabellaACS Chemical Neuroscience (2010), 1 (6), 435-449CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)The interplay among commonly used physicochem. properties in drug design was examd. and utilized to create a prospective design tool focused on the alignment of key druglike attributes. Using a set of six physicochem. parameters ((a) lipophilicity, calcd. partition coeff. (ClogP); (b) calcd. distribution coeff. at pH = 7.4 (ClogD); (c) mol. wt. (MW); (d) topol. polar surface area (TPSA); (e) no. of hydrogen bond donors (HBD); (f) most basic center (pKa)), a druglikeness central nervous system multiparameter optimization (CNS MPO) algorithm was built and applied to a set of marketed CNS drugs (N = 119) and Pfizer CNS candidates (N = 108), as well as to a large diversity set of Pfizer proprietary compds. (N = 11 303). The novel CNS MPO algorithm showed that 74% of marketed CNS drugs displayed a high CNS MPO score (MPO desirability score ≥ 4, using a scale of 0-6), in comparison to 60% of the Pfizer CNS candidates. This anal. suggests that this algorithm could potentially be used to identify compds. with a higher probability of successfully testing hypotheses in the clinic. In addn., a relationship between an increasing CNS MPO score and alignment of key in vitro attributes of drug discovery (favorable permeability, P-glycoprotein (P-gp) efflux, metabolic stability, and safety) was seen in the marketed CNS drug set, the Pfizer candidate set, and the Pfizer proprietary diversity set. The CNS MPO scoring function offers advantages over hard cutoffs or utilization of single parameters to optimize structure-activity relationships (SAR) by expanding medicinal chem. design space through a holistic assessment approach. Based on six physicochem. properties commonly used by medicinal chemists, the CNS MPO function may be used prospectively at the design stage to accelerate the identification of compds. with increased probability of success.
- 38Trott, O.; Olson, A. J. AutoDock Vina: Improving the Speed and Accuracy of Docking with a New Scoring Function, Efficient Optimization, and Multithreading. J. Comput. Chem. 2009, 31, 455– 461, DOI: 10.1002/jcc.21334There is no corresponding record for this reference.
- 39Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: A New Approach for Rapid, Accurate Docking and Scoring. 1. Method and Assessment of Docking Accuracy. J. Med. Chem. 2004, 47, 1739– 1749, DOI: 10.1021/jm030643039https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit74%253D&md5=8cc2f0022318b12dd972e9c493375bf9Glide: A new approach for rapid, accurate docking and scoring. 1. method and assessment of docking accuracyFriesner, Richard A.; Banks, Jay L.; Murphy, Robert B.; Halgren, Thomas A.; Klicic, Jasna J.; Mainz, Daniel T.; Repasky, Matthew P.; Knoll, Eric H.; Shelley, Mee; Perry, Jason K.; Shaw, David E.; Francis, Perry; Shenkin, Peter S.Journal of Medicinal Chemistry (2004), 47 (7), 1739-1749CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Unlike other methods for docking ligands to the rigid 3D structure of a known protein receptor, Glide approximates a complete systematic search of the conformational, orientational, and positional space of the docked ligand. In this search, an initial rough positioning and scoring phase that dramatically narrows the search space is followed by torsionally flexible energy optimization on an OPLS-AA nonbonded potential grid for a few hundred surviving candidate poses. The very best candidates are further refined via a Monte Carlo sampling of pose conformation; in some cases, this is crucial to obtaining an accurate docked pose. Selection of the best docked pose uses a model energy function that combines empirical and force-field-based terms. Docking accuracy is assessed by redocking ligands from 282 cocrystd. PDB complexes starting from conformationally optimized ligand geometries that bear no memory of the correctly docked pose. Errors in geometry for the top-ranked pose are less than 1 Å in nearly half of the cases and are greater than 2 Å in only about one-third of them. Comparisons to published data on rms deviations show that Glide is nearly twice as accurate as GOLD and more than twice as accurate as FlexX for ligands having up to 20 rotatable bonds. Glide is also found to be more accurate than the recently described Surflex method.
- 40Butina, D. Unsupervised Data Base Clustering Based on Daylight’s Fingerprint and Tanimoto Similarity: A Fast and Automated Way to Cluster Small and Large Data Sets. J. Chem. Inf. Comput. Sci. 1999, 39, 747– 750, DOI: 10.1021/ci980338140https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXktVWhtLw%253D&md5=e11c02e1e777f88e208ec5fdc5405c47Unsupervised data base clustering based on Daylight's fingerprint and Tanimoto similarity: a fast and automated way to cluster small and large data setsButina, DarkoJournal of Chemical Information and Computer Sciences (1999), 39 (4), 747-750CODEN: JCISD8; ISSN:0095-2338. (American Chemical Society)Jarvis-Patrick's (J-P) algorithm is one of the most commonly used clustering algorithms within the global pharmaceutical industry. The implementation of the J-P under Daylight software, using Daylight's fingerprints and the Tanimoto similarity index, can deal with sets of 100 k mols. in a matter of a few hours. However, the J-P clustering algorithm has several assocd. problems which make it difficult to cluster large data sets in a consistent and timely manner. The clusters produced are greatly dependent on the choice of the two parameters needed to run J-P clustering, such that this method tends to produce clusters which are either very large and heterogeneous or homogeneous but too small. In any case, J-P always requires time-consuming manual tuning. An algorithm which will identify dense clusters where similarity within each cluster reflects the Tanimoto value used for the clustering and, more importantly, where the cluster centroid will be at least similar, at the given Tanimoto value, to every other mol. within the cluster in a consistent and automated manner, is presented. The similarity term used throughout the paper reflects the overall similarity between two given mols., as defined by Daylight's fingerprints and the Tanimoto similarity index.
- 41Flagmeier, P.; Meisl, G.; Vendruscolo, M.; Knowles, T. P. J.; Dobson, C. M.; Buell, A. K.; Galvagnion, C. Mutations Associated with Familial Parkinson’s Disease Alter the Initiation and Amplification Steps of α-Synuclein Amyloid Formation. Proc. Natl. Acad. Sci. U.S.A. 2016, 113, 10328– 10333, DOI: 10.1073/pnas.160464511341https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVWks7jL&md5=70a504e389bfb0f58ae555b9d5726eceMutations associated with familial Parkinson's disease alter the initiation and amplification steps of α-synuclein aggregationFlagmeier, Patrick; Meisl, Georg; Vendruscolo, Michele; Knowles, Tuomas P. J.; Dobson, Christopher M.; Buell, Alexander K.; Galvagnion, CelineProceedings of the National Academy of Sciences of the United States of America (2016), 113 (37), 10328-10333CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Parkinson's disease is a highly debilitating neurodegenerative condition whose pathol. hallmark is the presence in nerve cells of proteinaceous deposits, known as Lewy bodies, composed primarily of amyloid fibrils of α-synuclein. Several missense mutations in the gene encoding α-synuclein have been assocd. with familial variants of Parkinson's disease and have been shown to affect the kinetics of the aggregation of the protein. Using a combination of exptl. and theor. approaches, we present a systematic in vitro study of the influence of disease-assocd. single-point mutations on the individual processes involved in α-synuclein aggregation into amyloid fibrils. We find that lipid-induced fibril prodn. and surface catalyzed fibril amplification are the processes most strongly affected by these mutations and show that familial mutations can induce dramatic changes in the crucial processes thought to be assocd. with the initiation and spreading of the aggregation of α-synuclein.
- 42Galvagnion, C.; Buell, A. K.; Meisl, G.; Michaels, T. C. T.; Vendruscolo, M.; Knowles, T. P. J.; Dobson, C. M. Lipid Vesicles Trigger α-Synuclein Aggregation by Stimulating Primary Nucleation. Nat. Chem. Biol. 2015, 11, 229– 234, DOI: 10.1038/nchembio.175042https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFCntro%253D&md5=1ceed4bdf36d5cf4663395ab4443b720Lipid vesicles trigger α-synuclein aggregation by stimulating primary nucleationGalvagnion, Celine; Buell, Alexander K.; Meisl, Georg; Michaels, Thomas C. T.; Vendruscolo, Michele; Knowles, Tuomas P. J.; Dobson, Christopher M.Nature Chemical Biology (2015), 11 (3), 229-234CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)α-Synuclein (α-syn) is a 140-residue intrinsically disordered protein that is involved in neuronal and synaptic vesicle plasticity, but its aggregation to form amyloid fibrils is the hallmark of Parkinson's disease (PD). The interaction between α-syn and lipid surfaces is believed to be a key feature for mediation of its normal function, but under other circumstances it is able to modulate amyloid fibril formation. Using a combination of exptl. and theor. approaches, we identify the mechanism through which facile aggregation of α-syn is induced under conditions where it binds a lipid bilayer, and we show that the rate of primary nucleation can be enhanced by three orders of magnitude or more under such conditions. These results reveal the key role that membrane interactions can have in triggering conversion of α-syn from its sol. state to the aggregated state that is assocd. with neurodegeneration and to its assocd. disease states.
- 43Michaels, T. C. T.; Cohen, S. I. A.; Vendruscolo, M.; Dobson, C. M.; Knowles, T. P. J. Hamiltonian Dynamics of Protein Filament Formation. Phys. Rev. Lett. 2016, 116, 038101, DOI: 10.1103/PhysRevLett.116.03810143https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtF2ksr3J&md5=fb3129c50d3e9aaf4d5dea257fa941f2Hamiltonian dynamics of protein filament formationMichaels, Thomas C. T.; Cohen, Samuel I. A.; Vendruscolo, Michele; Dobson, Christopher M.; Knowles, Tuomas P. J.Physical Review Letters (2016), 116 (3), 038101/1-038101/6CODEN: PRLTAO; ISSN:0031-9007. (American Physical Society)We establish the Hamiltonian structure of the rate equations describing the formation of protein filaments. We then show that this formalism provides a unified view of the behavior of a range of biol. self-assembling systems as diverse as actin. prions. and amyloidogenic polypeptides. We further demonstrate that the time-translation symmetry of the resulting Hamiltonian leads to previously unsuggested conservation laws that connect the no. and mass concns. of fibrils and allow linear growth phenomena to be equated with autocatalytic growth processes. We finally show how these results reveal simple rate laws that provide the basis for interpreting exptl. data in terms of specific mechanisms controlling the proliferation of fibrils.
- 44Palacio-Rodríguez, K.; Lans, I.; Cavasotto, C. N.; Cossio, P. Exponential Consensus Ranking Improves the Outcome in Docking and Receptor Ensemble Docking. Sci. Rep. 2019, 9, 5142 DOI: 10.1038/s41598-019-41594-344https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cbosVCksA%253D%253D&md5=949544789593b1c55faa32bfcce3afc2Exponential consensus ranking improves the outcome in docking and receptor ensemble dockingPalacio-Rodriguez Karen; Lans Isaias; Cossio Pilar; Cavasotto Claudio N; Cavasotto Claudio N; Cavasotto Claudio N; Cossio PilarScientific reports (2019), 9 (1), 5142 ISSN:.Consensus-scoring methods are commonly used with molecular docking in virtual screening campaigns to filter potential ligands for a protein target. Traditional consensus methods combine results from different docking programs by averaging the score or rank of each molecule obtained from individual programs. Unfortunately, these methods fail if one of the docking programs has poor performance, which is likely to occur due to training-set dependencies and scoring-function parameterization. In this work, we introduce a novel consensus method that overcomes these limitations. We combine the results from individual docking programs using a sum of exponential distributions as a function of the molecule rank for each program. We test the method over several benchmark systems using individual and ensembles of target structures from diverse protein families with challenging decoy/ligand datasets. The results demonstrate that the novel method outperforms the best traditional consensus strategies over a wide range of systems. Moreover, because the novel method is based on the rank rather than the score, it is independent of the score units, scales and offsets, which can hinder the combination of results from different structures or programs. Our method is simple and robust, providing a theoretical basis not only for molecular docking but also for any consensus strategy in general.
- 45Wilhelm, B. G.; Mandad, S.; Truckenbrodt, S.; Kröhnert, K.; Schäfer, C.; Rammner, B.; Koo, S. J.; Claßen, Ga.; Krauss, M.; Haucke, V.; Urlaub, H.; Rizzoli, S. O. Composition of Isolated Synaptic Boutons Reveals the Amounts of Vesicle Trafficking Proteins. Science 2014, 344, 1023– 1028, DOI: 10.1126/science.125288445https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXosFSmurc%253D&md5=0e85f1ad06f7cd9cfca66cea6a3b59adComposition of isolated synaptic boutons reveals the amounts of vesicle trafficking proteinsWilhelm, Benjamin G.; Mandad, Sunit; Truckenbrodt, Sven; Kroehnert, Katharina; Schaefer, Christina; Rammner, Burkhard; Koo, Seong Joo; Classen, Gala A.; Krauss, Michael; Haucke, Volker; Urlaub, Henning; Rizzoli, Silvio O.Science (Washington, DC, United States) (2014), 344 (6187), 1023-1028CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Synaptic vesicle recycling has long served as a model for the general mechanisms of cellular trafficking. We used an integrative approach, combining quant. immunoblotting and mass spectrometry to det. protein nos.; electron microscopy to measure organelle nos., sizes, and positions; and super-resoln. fluorescence microscopy to localize the proteins. Using these data, we generated a three-dimensional model of an "av." synapse, displaying 300,000 proteins in at. detail. The copy nos. of proteins involved in the same step of synaptic vesicle recycling correlated closely. In contrast, copy nos. varied over more than three orders of magnitude between steps, from about 150 copies for the endosomal fusion proteins to more than 20,000 for the exocytotic ones.
- 46Yang, Y.; Shi, Y.; Schweighauser, M.; Zhang, X.; Kotecha, A.; Murzin, A. G.; Garringer, H. J.; Cullinane, P. W.; Saito, Y.; Foroud, T.; Warner, T. T.; Hasegawa, K.; Vidal, R.; Murayama, S.; Revesz, T.; Ghetti, B.; Hasegawa, M.; Lashley, T.; Scheres, S. H. W.; Goedert, M. Structures of α-synuclein filaments from human brains with Lewy pathology, Nature 2022. 610 791 795 DOI: 10.1101/2022.07.12.499706610 .46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xis1Onu7%252FN&md5=00b50c7577f4c53e7250a09bb96bfda8Structures of alpha synuclein filaments from human brains with Lewy pathologyYang, Yang; Shi, Yang; Schweighauser, Manuel; Zhang, Xianjun; Kotecha, Abhay; Murzin, Alexey G.; Garringer, Holly J.; Cullinane, Patrick W.; Saito, Yuko; Foroud, Tatiana; Warner, Thomas T.; Hasegawa, Kazuko; Vidal, Ruben; Murayama, Shigeo; Revesz, Tamas; Ghetti, Bernardino; Hasegawa, Masato; Lashley, Tammaryn; Scheres, Sjors H. W.; Goedert, MichelNature (London, United Kingdom) (2022), 610 (7933), 791-795CODEN: NATUAS; ISSN:1476-4687. (Nature Portfolio)Parkinson disease (PD) is the most common movement disorder, with resting tremor, rigidity, bradykinesia and postural instability being major symptoms. Neuropathol. it is characterized by the presence of abundant filamentous inclusions of α-synuclein in the form of Lewy bodies and Lewy neurites in some brain cells, including dopaminergic nerve cells of the substantia nigra. PD is increasingly recognized as a multisystem disorder, with cognitive decline being one of its most common non-motor symptoms. Many patients with PD develop dementia more than 10 years after diagnosis. PD dementia (PDD) is clin. and neuropathol. similar to dementia with Lewy bodies (DLB), which is diagnosed when cognitive impairment precedes parkinsonian motor signs or begins within one year from their onset. In PDD, cognitive impairment develops in the setting of well-established PD. Besides PD and DLB, multiple system atrophy (MSA) is the third major synucleinopathy. It is characterized by the presence of abundant filamentous α-synuclein inclusions in brain cells, esp. oligodendrocytes (Papp-Lantos bodies). We previously reported the electron cryo-microscopy structures of two types of α-synuclein filament extd. from the brains of individuals with MSA6. Each filament type is made of two different protofilaments. Here we report that the cryo-electron microscopy structures of α-synuclein filaments from the brains of individuals with PD, PDD and DLB are made of a single protofilament (Lewy fold) that is markedly different from the protofilaments of MSA. These findings establish the existence of distinct mol. conformers of assembled α-synuclein in neurodegenerative disease.
- 47Cohen, S. I. A.; Arosio, P.; Presto, J.; Kurudenkandy, F. R.; Biverstål, H.; Dolfe, L.; Dunning, C.; Yang, X.; Frohm, B.; Vendruscolo, M.; Johansson, J.; Dobson, C. M.; Fisahn, A.; Knowles, T. P. J.; Linse, S. A Molecular Chaperone Breaks the Catalytic Cycle That Generates Toxic Aβ Oligomers. Nat. Struct. Mol. Biol. 2015, 22, 207– 213, DOI: 10.1038/nsmb.297147https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXivVSitLY%253D&md5=03c46f97e7f2361193667db1d4b94f07A molecular chaperone breaks the catalytic cycle that generates toxic Aβ oligomersCohen, Samuel I. A.; Arosio, Paolo; Presto, Jenny; Kurudenkandy, Firoz Roshan; Biverstal, Henrik; Dolfe, Lisa; Dunning, Christopher; Yang, Xiaoting; Frohm, Birgitta; Vendruscolo, Michele; Johansson, Jan; Dobson, Christopher M.; Fisahn, Andre; Knowles, Tuomas P. J.; Linse, SaraNature Structural & Molecular Biology (2015), 22 (3), 207-213CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Alzheimer's disease is an increasingly prevalent neurodegenerative disorder whose pathogenesis has been assocd. with aggregation of the amyloid-β peptide (Aβ42). Recent studies have revealed that once Aβ42 fibrils are generated, their surfaces effectively catalyze the formation of neurotoxic oligomers. Here we show that a mol. chaperone, a human Brichos domain, can specifically inhibit this catalytic cycle and limit human Aβ42 toxicity. We demonstrate in vitro that Brichos achieves this inhibition by binding to the surfaces of fibrils, thereby redirecting the aggregation reaction to a pathway that involves minimal formation of toxic oligomeric intermediates. We verify that this mechanism occurs in living mouse brain tissue by cytotoxicity and electrophysiol. expts. These results reveal that mol. chaperones can help maintain protein homeostasis by selectively suppressing crit. microscopic steps within the complex reaction pathways responsible for the toxic effects of protein misfolding and aggregation.
- 48Perni, M.; Galvagnion, C.; Maltsev, A.; Meisl, G.; Müller, M. B. D.; Challa, P. K.; Kirkegaard, J. B.; Cohen, S. I. A.; Cascella, R.; Chen, S. W.; Limboker, R.; Sormanni, P.; Heller, G. T.; Francesco, A.; Cremades, N.; Cecchi, C.; Chiti, F.; Ellen, A. A.; Knowles, T. P. J.; Vendruscolo, M.; Bax, A.; Zasloff, M.; Dobson, C. M.; Perni, M.; Galvagnion, C.; Maltsev, A.; Meisl, G.; Müller, M. B. D.; Challa, P. K.; Julius, B.; Flagmeier, P.; Cohen, S. I. A.; Chen, S. W.; Limbocker, R.; Sormanni, P.; Heller, G. T.; Aprile, F. A.; Cremades, N.; Cecchi, C.; Chiti, F.; Nollen, E. A. A.; Tuomas, P. J.; Vendruscolo, M.; Bax, A.; Dobson, C. M.; Perni, M.; Galvagnion, C.; Maltsev, A.; Meisl, G.; Müller, M. B. D.; Challa, P. K. A Natural Product Inhibits the Initiation of α-Synuclein Aggregation and Suppresses Its Toxicity. Proc. Natl. Acad. Sci. U.S.A. 2017, 114, E1009– E1017, DOI: 10.1073/pnas.161058611448https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFarsLY%253D&md5=116485fd417a51fac86fb5d78c604020A natural product inhibits the initiation of α-synuclein aggregation and suppresses its toxicityPerni, Michele; Galvagnion, Celine; Maltsev, Alexander; Meisl, Georg; Muller, Martin B. D.; Challa, Pavan K.; Kirkegaard, Julius B.; Flagmeier, Patrick; Cohen, Samuel I. A.; Cascella, Roberta; Chen, Serene W.; Limboker, Ryan; Sormanni, Pietro; Heller, Gabriella T.; Aprile, Francesco A.; Cremades, Nunilo; Cecchi, Cristina; Chiti, Fabrizio; Nollen, Ellen A. A.; Knowles, Tuomas P. J.; Vendruscolo, Michele; Bax, Adriaan; Zasloff, Michael; Dobson, Christopher M.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (6), E1009-E1017CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The self-assembly of α-synuclein is closely assocd. with Parkinson's disease and related syndromes. The authors show that squalamine, a natural product with known anticancer and antiviral activity, dramatically affects α-synuclein aggregation in vitro and in vivo. The authors elucidate the mechanism of action of squalamine by investigating its interaction with lipid vesicles, which are known to stimulate nucleation, and find that this compd. displaces α-synuclein from the surfaces of such vesicles, thereby blocking the first steps in its aggregation process. The authors also show that squalamine almost completely suppresses the toxicity of α-synuclein oligomers in human neuroblastoma cells by inhibiting their interactions with lipid membranes. The authors further examine the effects of squalamine in a Caenorhabditis elegans strain overexpressing α-synuclein, observing a dramatic redn. of α-synuclein aggregation and an almost complete elimination of muscle paralysis. These findings suggest that squalamine could be a means of therapeutic intervention in Parkinson's disease and related conditions.
- 49Agerschou, E. D.; Flagmeier, P.; Saridaki, T.; Galvagnion, C.; Komnig, D.; Heid, L.; Prasad, V.; Shaykhalishahi, H.; Willbold, D.; Dobson, C. M.; Voigt, A.; Falkenburger, B.; Hoyer, W.; Buell, A. K. An Engineered Monomer Binding-Protein for α-Synuclein Efficiently Inhibits the Proliferation of Amyloid Fibrils. eLife 2019, 8, e46112 DOI: 10.7554/eLife.4611249https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlaltLzE&md5=5d8e3c3ee85ea1ec460bdcd61c05ca83An engineered monomer binding-protein for α-synuclein efficiently inhibits the proliferation of amyloid fibrilsAgerschou, Emil Dandanell; Flagmeier, Patrick; Saridaki, Theodora; Galvagnion, Celine; Komnig, Daniel; Heid, Laetitia; Prasad, Vibha; Shaykhalishahi, Hamed; Willbold, Dieter; Dobson, Christopher M.; Voigt, Aaron; Falkenburger, Bjoern; Hoyer, Wolfgang; Buell, Alexander K.eLife (2019), 8 (), e46112/1-e46112/31CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Removing or preventing the formation of α-synuclein aggregates is a plausible strategy against Parkinson's disease. To this end, we have engineered the β-wrapin AS69 to bind monomeric α-synuclein with high affinity. In cultured cells, AS69 reduced the self-interaction of α-synuclein and formation of visible α-synuclein aggregates. In flies, AS69 reduced α-synuclein aggregates and the locomotor deficit resulting from α-synuclein expression in neuronal cells. In biophys. expts. in vitro, AS69 highly sub-stoichiometrically inhibited both primary and autocatalytic secondary nucleation processes, even in the presence of a large excess of monomer. We present evidence that the AS69-α-synuclein complex, rather than the free AS69, is the inhibitory species responsible for sub-stoichiometric inhibition of secondary nucleation. These results represent a new paradigm that high affinity monomer binders can lead to strongly substoichiometric inhibition of nucleation processes.
- 50Linse, S.; Scheidt, T.; Bernfur, K.; Vendruscolo, M.; Dobson, C. M.; Cohen, S. I. A.; Sileikis, E.; Lundqvist, M.; Qian, F.; O’Malley, T.; Bussiere, T.; Weinreb, P. H.; Xu, C. K.; Meisl, G.; Devenish, S. R. A.; Knowles, T. P. J.; Hansson, O. Kinetic Fingerprints Differentiate the Mechanisms of Action of Anti-Aβ Antibodies. Nat. Struct. Mol. Biol. 2020, 27, 1125– 1133, DOI: 10.1038/s41594-020-0505-650https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFegurjF&md5=2adc8febabbb832a3c8db63019db83e6Kinetic fingerprints differentiate the mechanisms of action of anti-Aβ antibodiesLinse, Sara; Scheidt, Tom; Bernfur, Katja; Vendruscolo, Michele; Dobson, Christopher M.; Cohen, Samuel I. A.; Sileikis, Eimantas; Lundqvist, Martin; Qian, Fang; O'Malley, Tiernan; Bussiere, Thierry; Weinreb, Paul H.; Xu, Catherine K.; Meisl, Georg; Devenish, Sean R. A.; Knowles, Tuomas P. J.; Hansson, OskarNature Structural & Molecular Biology (2020), 27 (12), 1125-1133CODEN: NSMBCU; ISSN:1545-9993. (Nature Research)The amyloid cascade hypothesis, according to which the self-assembly of amyloid-β peptide (Aβ) is a causative process in Alzheimer's disease, has driven many therapeutic efforts for the past 20 years. Failures of clin. trials investigating Aβ-targeted therapies have been interpreted as evidence against this hypothesis, irresp. of the characteristics and mechanisms of action of the therapeutic agents, which are highly challenging to assess. Here, we combine kinetic analyses with quant. binding measurements to address the mechanism of action of four clin. stage anti-Aβ antibodies, aducanumab, gantenerumab, bapineuzumab and solanezumab. We quantify the influence of these antibodies on the aggregation kinetics and on the prodn. of oligomeric aggregates and link these effects to the affinity and stoichiometry of each antibody for monomeric and fibrillar forms of Aβ. Our results reveal that, uniquely among these four antibodies, aducanumab dramatically reduces the flux of Aβ oligomers.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.molpharmaceut.2c00548.
Predicted binding affinity of compounds via computational docking; TEM images of α-synuclein fibrils formed in the presence of molecules; kinetic profiles of α-synuclein aggregation in the presence of molecules; time dependence of reactive flux toward α-synuclein oligomers in the presence of molecules; fluorescence emission spectra of molecule C; colocalization of molecules with α-synuclein fibrils in the presence of neuroblastoma cells; list of all molecules from docking studies experimentally validated; and supplementary methods, supplementary figures, and supplementary table (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.