Development of Small Molecules Targeting α-Synuclein Aggregation: A Promising Strategy to Treat Parkinson’s Disease
Abstract
1. Introduction
2. Historical Overview
3. Symptomatology
4. Risk Factors and Genetics of Parkinson’s Disease
5. Molecular Mechanism Implicated in PD Development
6. Alpha-Synuclein
7. New Therapies: Modulating α-Synuclein Aggregation
7.1. Polyphenolic Scaffolds
7.2. Repositioned Compounds
7.3. Compounds Developed by Rational Design
7.4. Compounds Derived from High-Throughput Screenings
7.5. Structure-Based Strategies for Drug Discovery
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Sabate, R.; Ventura, S. Cross-beta-sheet supersecondary structure in amyloid folds: Techniques for detection and characterization. Methods Mol. Biol. 2013, 932, 237–257. [Google Scholar] [CrossRef]
- Sunde, M.; Serpell, L.C.; Bartlam, M.; Fraser, P.E.; Pepys, M.B.; Blake, C.C. Common core structure of amyloid fibrils by synchrotron X-ray diffraction. J. Mol. Biol. 1997, 273, 729–739. [Google Scholar] [CrossRef] [PubMed]
- Ke, P.C.; Zhou, R.; Serpell, L.C.; Riek, R.; Knowles, T.P.J.; Lashuel, H.A.; Gazit, E.; Hamley, I.W.; Davis, T.P.; Fandrich, M.; et al. Half a century of amyloids: Past, present and future. Chem. Soc. Rev. 2020, 49, 5473–5509. [Google Scholar] [CrossRef] [PubMed]
- Santos, J.; Ventura, S. Functional Amyloids Germinate in Plants. Trends Plant Sci. 2021, 26, 7–10. [Google Scholar] [CrossRef]
- Claessen, D.; Rink, R.; de Jong, W.; Siebring, J.; de Vreugd, P.; Boersma, F.G.; Dijkhuizen, L.; Wosten, H.A. A novel class of secreted hydrophobic proteins is involved in aerial hyphae formation in Streptomyces coelicolor by forming amyloid-like fibrils. Genes Dev. 2003, 17, 1714–1726. [Google Scholar] [CrossRef]
- Chapman, M.R.; Robinson, L.S.; Pinkner, J.S.; Roth, R.; Heuser, J.; Hammar, M.; Normark, S.; Hultgren, S.J. Role of Escherichia coli curli operons in directing amyloid fiber formation. Science 2002, 295, 851–855. [Google Scholar] [CrossRef] [PubMed]
- Fowler, D.M.; Koulov, A.V.; Alory-Jost, C.; Marks, M.S.; Balch, W.E.; Kelly, J.W. Functional amyloid formation within mammalian tissue. PLoS Biol. 2006, 4, e6. [Google Scholar] [CrossRef] [PubMed]
- Otzen, D.; Riek, R. Functional Amyloids. Cold Spring Harb. Perspect. Biol. 2019, 11, a033860. [Google Scholar] [CrossRef]
- Garcia-Pardo, J.; Bartolome-Nafria, A.; Chaves-Sanjuan, A.; Gil-Garcia, M.; Visentin, C.; Bolognesi, M.; Ricagno, S.; Ventura, S. Cryo-EM structure of hnRNPDL-2 fibrils, a functional amyloid associated with limb-girdle muscular dystrophy D3. Nat. Commun. 2023, 14, 239. [Google Scholar] [CrossRef]
- Berson, J.F.; Harper, D.C.; Tenza, D.; Raposo, G.; Marks, M.S. Pmel17 initiates premelanosome morphogenesis within multivesicular bodies. Mol. Biol. Cell 2001, 12, 3451–3464. [Google Scholar] [CrossRef]
- Hervas, R.; Rau, M.J.; Park, Y.; Zhang, W.; Murzin, A.G.; Fitzpatrick, J.A.J.; Scheres, S.H.W.; Si, K. Cryo-EM structure of a neuronal functional amyloid implicated in memory persistence in Drosophila. Science 2020, 367, 1230–1234. [Google Scholar] [CrossRef] [PubMed]
- Maji, S.K.; Perrin, M.H.; Sawaya, M.R.; Jessberger, S.; Vadodaria, K.; Rissman, R.A.; Singru, P.S.; Nilsson, K.P.; Simon, R.; Schubert, D.; et al. Functional amyloids as natural storage of peptide hormones in pituitary secretory granules. Science 2009, 325, 328–332. [Google Scholar] [CrossRef]
- Chiti, F.; Dobson, C.M. Protein misfolding, functional amyloid, and human disease. Annu. Rev. Biochem. 2006, 75, 333–366. [Google Scholar] [CrossRef] [PubMed]
- Nussbaum, R.L.; Ellis, C.E. Alzheimer’s disease and Parkinson’s disease. New Engl. J. Med. 2003, 348, 1356–1364. [Google Scholar] [CrossRef] [PubMed]
- Dexter, D.T.; Jenner, P. Parkinson disease: From pathology to molecular disease mechanisms. Free Radic. Biol. Med. 2013, 62, 132–144. [Google Scholar] [CrossRef] [PubMed]
- Pringsheim, T.; Jette, N.; Frolkis, A.; Steeves, T.D. The prevalence of Parkinson’s disease: A systematic review and meta-analysis. Mov. Disord. Off. J. Mov. Disord. Soc. 2014, 29, 1583–1590. [Google Scholar] [CrossRef]
- Goetz, C.G. The history of Parkinson’s disease: Early clinical descriptions and neurological therapies. Cold Spring Harbor. Perspect. Med. 2011, 1, a008862. [Google Scholar] [CrossRef]
- Duncan, G.W.; Khoo, T.K.; Yarnall, A.J.; O’Brien, J.T.; Coleman, S.Y.; Brooks, D.J.; Barker, R.A.; Burn, D.J. Health-related quality of life in early Parkinson’s disease: The impact of nonmotor symptoms. Mov. Disord. Off. J. Mov. Disord. Soc. 2014, 29, 195–202. [Google Scholar] [CrossRef]
- Martinez-Martin, P.; Rodriguez-Blazquez, C.; Kurtis, M.M.; Chaudhuri, K.R.; Group, N.V. The impact of non-motor symptoms on health-related quality of life of patients with Parkinson’s disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2011, 26, 399–406. [Google Scholar] [CrossRef]
- Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
- Spillantini, 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. [Google Scholar] [CrossRef]
- Cotzias, G.C. L-Dopa for Parkinsonism. New Engl. J. Med. 1968, 278, 630. [Google Scholar] [CrossRef] [PubMed]
- Hornykiewicz, O. L-DOPA: From a biologically inactive amino acid to a successful therapeutic agent. Amino Acids 2002, 23, 65–70. [Google Scholar] [CrossRef]
- Baba, M.; Nakajo, S.; Tu, P.H.; Tomita, T.; Nakaya, K.; Lee, V.M.; Trojanowski, J.Q.; Iwatsubo, T. Aggregation of alpha-synuclein in Lewy bodies of sporadic Parkinson’s disease and dementia with Lewy bodies. Am. J. Pathol. 1998, 152, 879–884. [Google Scholar]
- Wakabayashi, K.; Hayashi, S.; Kakita, A.; Yamada, M.; Toyoshima, Y.; Yoshimoto, M.; Takahashi, H. Accumulation of alpha-synuclein/NACP is a cytopathological feature common to Lewy body disease and multiple system atrophy. Acta Neuropathol. 1998, 96, 445–452. [Google Scholar] [CrossRef]
- Spillantini, M.G.; Crowther, R.A.; Jakes, R.; Cairns, N.J.; Lantos, P.L.; Goedert, M. Filamentous alpha-synuclein inclusions link multiple system atrophy with Parkinson’s disease and dementia with Lewy bodies. Neurosci. Lett. 1998, 251, 205–208. [Google Scholar] [CrossRef]
- Goedert, M.; Spillantini, M.G. Lewy body diseases and multiple system atrophy as alpha-synucleinopathies. Mol. Psychiatry 1998, 3, 462–465. [Google Scholar] [CrossRef] [PubMed]
- Spillantini, M.G.; Crowther, R.A.; Jakes, R.; Hasegawa, M.; Goedert, M. Alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with lewy bodies. Proc. Natl. Acad. Sci. USA 1998, 95, 6469–6473. [Google Scholar] [CrossRef] [PubMed]
- Parkinson, J. An essay on the shaking palsy. 1817. J. Neuropsychiatry Clin. Neurosci. 2002, 14, 223–236, discussion 222. [Google Scholar] [CrossRef]
- Charcot, J.-M. Leçons sur le maladies du système nerveus. In Bureaux du Progrès Medical; Oeuvres Complètes (Tome 1); Delahaye, A., Lecronsnier, E., Eds.; Bureaux du Progrès Médical: Paris, France, 1872; pp. 155–188. [Google Scholar]
- Blocq, C.; Marinescu, G. Sur un cas de tremblement parkinsonien hémiplégique symptomatique d’une tumeur du pédoncle cérébral. C. R. Cos. Biol. 1893, 5, 105–111. [Google Scholar]
- Brissaud, E. Leçons sur les Maladies Nerveuses; Masson & Associates, Inc.: Escondido, CA, USA, 1899; Volume 2. [Google Scholar]
- Trétiakoff, C.D. Contribution à L’étude de L’anatomie Pathologique du Locus Niger de Soemmering Avec Quelques Deductions Relatives A la Pathogenie des Troubles du Tonus Musculaire et de la Maladie de Parkinson; Université de Paris: Paris, France, 1919. [Google Scholar]
- Lewy, F. Zur pathologischen anatomie der paralysis agitans. Dtsch. Z. Nervenheilkd. 1912, 50, 50–55. [Google Scholar]
- Postuma, R.B.; Berg, D. Advances in markers of prodromal Parkinson disease. Nat. Rev. Neurol. 2016, 12, 622–634. [Google Scholar] [CrossRef]
- Carlsson, A.; Lindqvist, M.; Magnusson, T. 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists. Nature 1957, 180, 1200. [Google Scholar] [CrossRef] [PubMed]
- Ehringer, H.; Hornykiewicz, O. Distribution of noradrenaline and dopamine (3-hydroxytyramine) in the human brain and their behavior in diseases of the extrapyramidal system. Klin. Wochenschr. 1960, 38, 1236–1239. [Google Scholar] [CrossRef]
- Polymeropoulos, M.H.; Lavedan, C.; Leroy, E.; Ide, S.E.; Dehejia, A.; Dutra, A.; Pike, B.; Root, H.; Rubenstein, J.; Boyer, R.; et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997, 276, 2045–2047. [Google Scholar] [CrossRef] [PubMed]
- Kruger, R.; Kuhn, W.; Muller, T.; Woitalla, D.; Graeber, M.; Kosel, S.; Przuntek, H.; Epplen, J.T.; Schols, L.; Riess, O. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat. Genet. 1998, 18, 106–108. [Google Scholar] [CrossRef] [PubMed]
- Athanassiadou, A.; Voutsinas, G.; Psiouri, L.; Leroy, E.; Polymeropoulos, M.H.; Ilias, A.; Maniatis, G.M.; Papapetropoulos, T. Genetic analysis of families with Parkinson disease that carry the Ala53Thr mutation in the gene encoding alpha-synuclein. Am. J. Hum. Genet. 1999, 65, 555–558. [Google Scholar] [CrossRef]
- Zarranz, J.J.; Alegre, J.; Gomez-Esteban, J.C.; Lezcano, E.; Ros, R.; Ampuero, I.; Vidal, L.; Hoenicka, J.; Rodriguez, O.; Atares, B.; et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann. Neurol. 2004, 55, 164–173. [Google Scholar] [CrossRef] [PubMed]
- Chartier-Harlin, M.C.; Kachergus, J.; Roumier, C.; Mouroux, V.; Douay, X.; Lincoln, S.; Levecque, C.; Larvor, L.; Andrieux, J.; Hulihan, M.; et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet 2004, 364, 1167–1169. [Google Scholar] [CrossRef]
- Appel-Cresswell, S.; Vilarino-Guell, C.; Encarnacion, M.; Sherman, H.; Yu, I.; Shah, B.; Weir, D.; Thompson, C.; Szu-Tu, C.; Trinh, J.; et al. Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson’s disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2013, 28, 811–813. [Google Scholar] [CrossRef]
- Kiely, A.P.; Asi, Y.T.; Kara, E.; Limousin, P.; Ling, H.; Lewis, P.; Proukakis, C.; Quinn, N.; Lees, A.J.; Hardy, J.; et al. alpha-Synucleinopathy associated with G51D SNCA mutation: A link between Parkinson’s disease and multiple system atrophy? Acta Neuropathol. 2013, 125, 753–769. [Google Scholar] [CrossRef] [PubMed]
- Singleton, A.B.; Farrer, M.J.; Bonifati, V. The genetics of Parkinson’s disease: Progress and therapeutic implications. Mov. Disord. Off. J. Mov. Disord. Soc. 2013, 28, 14–23. [Google Scholar] [CrossRef] [PubMed]
- Kitada, T.; Asakawa, S.; Hattori, N.; Matsumine, H.; Yamamura, Y.; Minoshima, S.; Yokochi, M.; Mizuno, Y.; Shimizu, N. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998, 392, 605–608. [Google Scholar] [CrossRef]
- Samaranch, L.; Lorenzo-Betancor, O.; Arbelo, J.M.; Ferrer, I.; Lorenzo, E.; Irigoyen, J.; Pastor, M.A.; Marrero, C.; Isla, C.; Herrera-Henriquez, J.; et al. PINK1-linked parkinsonism is associated with Lewy body pathology. Brain A J. Neurol. 2010, 133, 1128–1142. [Google Scholar] [CrossRef]
- Taipa, R.; Pereira, C.; Reis, I.; Alonso, I.; Bastos-Lima, A.; Melo-Pires, M.; Magalhaes, M. DJ-1 linked parkinsonism (PARK7) is associated with Lewy body pathology. Brain A J. Neurol. 2016, 139, 1680–1687. [Google Scholar] [CrossRef] [PubMed]
- Zimprich, A.; Biskup, S.; Leitner, P.; Lichtner, P.; Farrer, M.; Lincoln, S.; Kachergus, J.; Hulihan, M.; Uitti, R.J.; Calne, D.B.; et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 2004, 44, 601–607. [Google Scholar] [CrossRef]
- Schober, A. Classic toxin-induced animal models of Parkinson’s disease: 6-OHDA and MPTP. Cell Tissue Res. 2004, 318, 215–224. [Google Scholar] [CrossRef]
- Ko, W.K.D.; Bezard, E. Experimental animal models of Parkinson’s disease: A transition from assessing symptomatology to alpha-synuclein targeted disease modification. Exp. Neurol. 2017, 298, 172–179. [Google Scholar] [CrossRef]
- Koprich, J.B.; Kalia, L.V.; Brotchie, J.M. Animal models of alpha-synucleinopathy for Parkinson disease drug development. Nat. Rev. Neurosci. 2017, 18, 515–529. [Google Scholar] [CrossRef]
- Lazaro, D.F.; Pavlou, M.A.S.; Outeiro, T.F. Cellular models as tools for the study of the role of alpha-synuclein in Parkinson’s disease. Exp. Neurol. 2017, 298, 162–171. [Google Scholar] [CrossRef]
- Marmion, D.J.; Kordower, J.H. alpha-Synuclein nonhuman primate models of Parkinson’s disease. J. Neural Transm. 2018, 125, 385–400. [Google Scholar] [CrossRef] [PubMed]
- Trigo-Damas, I.; Del Rey, N.L.; Blesa, J. Novel models for Parkinson’s disease and their impact on future drug discovery. Expert Opin. Drug Discov. 2018, 13, 229–239. [Google Scholar] [CrossRef] [PubMed]
- Dehay, B.; Bourdenx, M.; Gorry, P.; Przedborski, S.; Vila, M.; Hunot, S.; Singleton, A.; Olanow, C.W.; Merchant, K.M.; Bezard, E.; et al. Targeting alpha-synuclein for treatment of Parkinson’s disease: Mechanistic and therapeutic considerations. Lancet Neurol. 2015, 14, 855–866. [Google Scholar] [CrossRef]
- Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480.e1412. [Google Scholar] [CrossRef] [PubMed]
- Coune, P.G.; Schneider, B.L.; Aebischer, P. Parkinson’s disease: Gene therapies. Cold Spring Harb. Perspect. Med. 2012, 2, a009431. [Google Scholar] [CrossRef]
- Tolosa, E.; Wenning, G.; Poewe, W. The diagnosis of Parkinson’s disease. Lancet Neurol. 2006, 5, 75–86. [Google Scholar] [CrossRef]
- Postuma, R.B.; Berg, D.; Stern, M.; Poewe, W.; Olanow, C.W.; Oertel, W.; Obeso, J.; Marek, K.; Litvan, I.; Lang, A.E.; et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2015, 30, 1591–1601. [Google Scholar] [CrossRef]
- Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Prim. 2017, 3, 17013. [Google Scholar] [CrossRef]
- Marder, K.S.; Tang, M.X.; Mejia-Santana, H.; Rosado, L.; Louis, E.D.; Comella, C.L.; Colcher, A.; Siderowf, A.D.; Jennings, D.; Nance, M.A.; et al. Predictors of parkin mutations in early-onset Parkinson disease: The consortium on risk for early-onset Parkinson disease study. Arch. Neurol. 2010, 67, 731–738. [Google Scholar] [CrossRef]
- Pont-Sunyer, C.; Hotter, A.; Gaig, C.; Seppi, K.; Compta, Y.; Katzenschlager, R.; Mas, N.; Hofeneder, D.; Brucke, T.; Bayes, A.; et al. The onset of nonmotor symptoms in Parkinson’s disease (the ONSET PD study). Mov. Disord. Off. J. Mov. Disord. Soc. 2015, 30, 229–237. [Google Scholar] [CrossRef]
- Schrag, A.; Sauerbier, A.; Chaudhuri, K.R. New clinical trials for nonmotor manifestations of Parkinson’s disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2015, 30, 1490–1504. [Google Scholar] [CrossRef]
- Hinnell, C.; Hurt, C.S.; Landau, S.; Brown, R.G.; Samuel, M.; Group, P.-P.S. Nonmotor versus motor symptoms: How much do they matter to health status in Parkinson’s disease? Mov. Disord. Off. J. Mov. Disord. Soc. 2012, 27, 236–241. [Google Scholar] [CrossRef]
- Storch, A.; Schneider, C.B.; Wolz, M.; Sturwald, Y.; Nebe, A.; Odin, P.; Mahler, A.; Fuchs, G.; Jost, W.H.; Chaudhuri, K.R.; et al. Nonmotor fluctuations in Parkinson disease: Severity and correlation with motor complications. Neurology 2013, 80, 800–809. [Google Scholar] [CrossRef] [PubMed]
- Weerkamp, N.J.; Tissingh, G.; Poels, P.J.; Zuidema, S.U.; Munneke, M.; Koopmans, R.T.; Bloem, B.R. Nonmotor symptoms in nursing home residents with Parkinson’s disease: Prevalence and effect on quality of life. J. Am. Geriatr. Soc. 2013, 61, 1714–1721. [Google Scholar] [CrossRef]
- Kalia, L.V.; Brotchie, J.M.; Fox, S.H. Novel nondopaminergic targets for motor features of Parkinson’s disease: Review of recent trials. Mov. Disord. Off. J. Mov. Disord. Soc. 2013, 28, 131–144. [Google Scholar] [CrossRef] [PubMed]
- Karachi, C.; Grabli, D.; Bernard, F.A.; Tande, D.; Wattiez, N.; Belaid, H.; Bardinet, E.; Prigent, A.; Nothacker, H.P.; Hunot, S.; et al. Cholinergic mesencephalic neurons are involved in gait and postural disorders in Parkinson disease. J. Clin. Investig. 2010, 120, 2745–2754. [Google Scholar] [CrossRef]
- Hirsch, E.C.; Graybiel, A.M.; Duyckaerts, C.; Javoy-Agid, F. Neuronal loss in the pedunculopontine tegmental nucleus in Parkinson disease and in progressive supranuclear palsy. Proc. Natl. Acad. Sci. USA 1987, 84, 5976–5980. [Google Scholar] [CrossRef]
- Seppi, K.; Weintraub, D.; Coelho, M.; Perez-Lloret, S.; Fox, S.H.; Katzenschlager, R.; Hametner, E.M.; Poewe, W.; Rascol, O.; Goetz, C.G.; et al. The Movement Disorder Society Evidence-Based Medicine Review Update: Treatments for the non-motor symptoms of Parkinson’s disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2011, 26 (Suppl. S3), S42–S80. [Google Scholar] [CrossRef] [PubMed]
- Connolly, B.; Fox, S.H. Treatment of cognitive, psychiatric, and affective disorders associated with Parkinson’s disease. Neurotherapeutics 2014, 11, 78–91. [Google Scholar] [CrossRef]
- Mahlknecht, P.; Seppi, K.; Poewe, W. The Concept of Prodromal Parkinson’s Disease. J. Park. Dis. 2015, 5, 681–697. [Google Scholar] [CrossRef]
- Salat, D.; Noyce, A.J.; Schrag, A.; Tolosa, E. Challenges of modifying disease progression in prediagnostic Parkinson’s disease. Lancet Neurol. 2016, 15, 637–648. [Google Scholar] [CrossRef] [PubMed]
- O’Sullivan, S.S.; Williams, D.R.; Gallagher, D.A.; Massey, L.A.; Silveira-Moriyama, L.; Lees, A.J. Nonmotor symptoms as presenting complaints in Parkinson’s disease: A clinicopathological study. Mov. Disord. Off. J. Mov. Disord. Soc. 2008, 23, 101–106. [Google Scholar] [CrossRef]
- Hely, M.A.; Reid, W.G.; Adena, M.A.; Halliday, G.M.; Morris, J.G. The Sydney multicenter study of Parkinson’s disease: The inevitability of dementia at 20 years. Mov. Disord. Off. J. Mov. Disord. Soc. 2008, 23, 837–844. [Google Scholar] [CrossRef]
- Hely, M.A.; Morris, J.G.; Reid, W.G.; Trafficante, R. Sydney Multicenter Study of Parkinson’s disease: Non-L-dopa-responsive problems dominate at 15 years. Mov. Disord. Off. J. Mov. Disord. Soc. 2005, 20, 190–199. [Google Scholar] [CrossRef] [PubMed]
- Noyce, A.J.; Bestwick, J.P.; Silveira-Moriyama, L.; Hawkes, C.H.; Giovannoni, G.; Lees, A.J.; Schrag, A. Meta-analysis of early nonmotor features and risk factors for Parkinson disease. Ann. Neurol. 2012, 72, 893–901. [Google Scholar] [CrossRef] [PubMed]
- Papadimitriou, D.; Antonelou, R.; Miligkos, M.; Maniati, M.; Papagiannakis, N.; Bostantjopoulou, S.; Leonardos, A.; Koros, C.; Simitsi, A.; Papageorgiou, S.G.; et al. Motor and Nonmotor Features of Carriers of the p.A53T Alpha-Synuclein Mutation: A Longitudinal Study. Mov. Disord. Off. J. Mov. Disord. Soc. 2016, 31, 1226–1230. [Google Scholar] [CrossRef]
- Koros, C.; Simitsi, A.; Stefanis, L. Genetics of Parkinson’s Disease: Genotype-Phenotype Correlations. Int. Rev. Neurobiol. 2017, 132, 197–231. [Google Scholar] [CrossRef]
- Somme, J.H.; Gomez-Esteban, J.C.; Molano, A.; Tijero, B.; Lezcano, E.; Zarranz, J.J. Initial neuropsychological impairments in patients with the E46K mutation of the alpha-synuclein gene (PARK 1). J. Neurol. Sci. 2011, 310, 86–89. [Google Scholar] [CrossRef]
- Ghosh, D.; Sahay, S.; Ranjan, P.; Salot, S.; Mohite, G.M.; Singh, P.K.; Dwivedi, S.; Carvalho, E.; Banerjee, R.; Kumar, A.; et al. The newly discovered Parkinson’s disease associated Finnish mutation (A53E) attenuates alpha-synuclein aggregation and membrane binding. Biochemistry 2014, 53, 6419–6421. [Google Scholar] [CrossRef]
- Kiely, A.P.; Ling, H.; Asi, Y.T.; Kara, E.; Proukakis, C.; Schapira, A.H.; Morris, H.R.; Roberts, H.C.; Lubbe, S.; Limousin, P.; et al. Distinct clinical and neuropathological features of G51D SNCA mutation cases compared with SNCA duplication and H50Q mutation. Mol. Neurodegener. 2015, 10, 41. [Google Scholar] [CrossRef]
- Lesage, S.; Anheim, M.; Letournel, F.; Bousset, L.; Honore, A.; Rozas, N.; Pieri, L.; Madiona, K.; Durr, A.; Melki, R.; et al. G51D alpha-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann. Neurol. 2013, 73, 459–471. [Google Scholar] [CrossRef] [PubMed]
- Tokutake, T.; Ishikawa, A.; Yoshimura, N.; Miyashita, A.; Kuwano, R.; Nishizawa, M.; Ikeuchi, T. Clinical and neuroimaging features of patient with early-onset Parkinson’s disease with dementia carrying SNCA p. G51D mutation. Park. Relat. Disord. 2014, 20, 262–264. [Google Scholar] [CrossRef] [PubMed]
- Martikainen, M.H.; Paivarinta, M.; Hietala, M.; Kaasinen, V. Clinical and imaging findings in Parkinson disease associated with the A53E SNCA mutation. Neurol. Genet. 2015, 1, e27. [Google Scholar] [CrossRef]
- Pasanen, P.; Myllykangas, L.; Siitonen, M.; Raunio, A.; Kaakkola, S.; Lyytinen, J.; Tienari, P.J.; Poyhonen, M.; Paetau, A. Novel alpha-synuclein mutation A53E associated with atypical multiple system atrophy and Parkinson’s disease-type pathology. Neurobiol. Aging 2014, 35, 2180.e1–2180.e5. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.T.; Mahul-Mellier, A.L.; Hegde, R.N.; Riviere, G.; Moons, R.; Ibanez de Opakua, A.; Magalhaes, P.; Rostami, I.; Donzelli, S.; Sobott, F.; et al. A NAC domain mutation (E83Q) unlocks the pathogenicity of human alpha-synuclein and recapitulates its pathological diversity. Sci. Adv. 2022, 8, eabn0044. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Koros, C.; Strohaker, T.; Schulte, C.; Bozi, M.; Varvaresos, S.; Ibanez de Opakua, A.; Simitsi, A.M.; Bougea, A.; Voumvourakis, K.; et al. A Novel SNCA A30G Mutation Causes Familial Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2021, 36, 1624–1633. [Google Scholar] [CrossRef]
- Lee, S.; Imai, Y.; Gehrke, S.; Liu, S.; Lu, B. The synaptic function of LRRK2. Biochem. Soc. Trans. 2012, 40, 1047–1051. [Google Scholar] [CrossRef]
- Rassu, M.; Del Giudice, M.G.; Sanna, S.; Taymans, J.M.; Morari, M.; Brugnoli, A.; Frassineti, M.; Masala, A.; Esposito, S.; Galioto, M.; et al. Role of LRRK2 in the regulation of dopamine receptor trafficking. PLoS ONE 2017, 12, e0179082. [Google Scholar] [CrossRef]
- Paisan-Ruiz, C.; Jain, S.; Evans, E.W.; Gilks, W.P.; Simon, J.; van der Brug, M.; Lopez de Munain, A.; Aparicio, S.; Gil, A.M.; Khan, N.; et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron 2004, 44, 595–600. [Google Scholar] [CrossRef]
- Zabetian, C.P.; Samii, A.; Mosley, A.D.; Roberts, J.W.; Leis, B.C.; Yearout, D.; Raskind, W.H.; Griffith, A. A clinic-based study of the LRRK2 gene in Parkinson disease yields new mutations. Neurology 2005, 65, 741–744. [Google Scholar] [CrossRef]
- Bose, A.; Beal, M.F. Mitochondrial dysfunction in Parkinson’s disease. J. Neurochem. 2016, 139 (Suppl. S1), 216–231. [Google Scholar] [CrossRef] [PubMed]
- Cookson, M.R. Cellular effects of LRRK2 mutations. Biochem. Soc. Trans. 2012, 40, 1070–1073. [Google Scholar] [CrossRef] [PubMed]
- Lill, C.M. Genetics of Parkinson’s disease. Mol. Cell. Probes 2016, 30, 386–396. [Google Scholar] [CrossRef]
- Thaler, A.; Gurevich, T.; Bar Shira, A.; Gana Weisz, M.; Ash, E.; Shiner, T.; Orr-Urtreger, A.; Giladi, N.; Mirelman, A. A “dose” effect of mutations in the GBA gene on Parkinson’s disease phenotype. Park. Relat. Disord. 2017, 36, 47–51. [Google Scholar] [CrossRef] [PubMed]
- Montfort, M.; Chabas, A.; Vilageliu, L.; Grinberg, D. Functional analysis of 13 GBA mutant alleles identified in Gaucher disease patients: Pathogenic changes and “modifier” polymorphisms. Hum. Mutat. 2004, 23, 567–575. [Google Scholar] [CrossRef] [PubMed]
- Sidransky, E.; Lopez, G. The link between the GBA gene and parkinsonism. Lancet Neurol. 2012, 11, 986–998. [Google Scholar] [CrossRef]
- Rocha, E.M.; Smith, G.A.; Park, E.; Cao, H.; Brown, E.; Hallett, P.; Isacson, O. Progressive decline of glucocerebrosidase in aging and Parkinson’s disease. Ann. Clin. Transl. Neurol. 2015, 2, 433–438. [Google Scholar] [CrossRef] [PubMed]
- Sidransky, E.; Nalls, M.A.; Aasly, J.O.; Aharon-Peretz, J.; Annesi, G.; Barbosa, E.R.; Bar-Shira, A.; Berg, D.; Bras, J.; Brice, A.; et al. Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease. New Engl. J. Med. 2009, 361, 1651–1661. [Google Scholar] [CrossRef] [PubMed]
- Mazzulli, J.R.; Xu, Y.H.; Sun, Y.; Knight, A.L.; McLean, P.J.; Caldwell, G.A.; Sidransky, E.; Grabowski, G.A.; Krainc, D. Gaucher disease glucocerebrosidase and alpha-synuclein form a bidirectional pathogenic loop in synucleinopathies. Cell 2011, 146, 37–52. [Google Scholar] [CrossRef]
- Sardi, S.P.; Clarke, J.; Viel, C.; Chan, M.; Tamsett, T.J.; Treleaven, C.M.; Bu, J.; Sweet, L.; Passini, M.A.; Dodge, J.C.; et al. Augmenting CNS glucocerebrosidase activity as a therapeutic strategy for parkinsonism and other Gaucher-related synucleinopathies. Proc. Natl. Acad. Sci. USA 2013, 110, 3537–3542. [Google Scholar] [CrossRef]
- Sardi, S.P.; Cheng, S.H.; Shihabuddin, L.S. Gaucher-related synucleinopathies: The examination of sporadic neurodegeneration from a rare (disease) angle. Prog. Neurobiol. 2015, 125, 47–62. [Google Scholar] [CrossRef]
- Riley, B.E.; Lougheed, J.C.; Callaway, K.; Velasquez, M.; Brecht, E.; Nguyen, L.; Shaler, T.; Walker, D.; Yang, Y.; Regnstrom, K.; et al. Structure and function of Parkin E3 ubiquitin ligase reveals aspects of RING and HECT ligases. Nat. Commun. 2013, 4, 1982. [Google Scholar] [CrossRef]
- Smith, W.W.; Pei, Z.; Jiang, H.; Moore, D.J.; Liang, Y.; West, A.B.; Dawson, V.L.; Dawson, T.M.; Ross, C.A. Leucine-rich repeat kinase 2 (LRRK2) interacts with parkin, and mutant LRRK2 induces neuronal degeneration. Proc. Natl. Acad. Sci. USA 2005, 102, 18676–18681. [Google Scholar] [CrossRef]
- van der Merwe, C.; Jalali Sefid Dashti, Z.; Christoffels, A.; Loos, B.; Bardien, S. Evidence for a common biological pathway linking three Parkinson’s disease-causing genes: Parkin, PINK1 and DJ-1. Eur. J. Neurosci. 2015, 41, 1113–1125. [Google Scholar] [CrossRef]
- Bonifati, V.; Rizzu, P.; van Baren, M.J.; Schaap, O.; Breedveld, G.J.; Krieger, E.; Dekker, M.C.; Squitieri, F.; Ibanez, P.; Joosse, M.; et al. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 2003, 299, 256–259. [Google Scholar] [CrossRef]
- Lesage, S.; Drouet, V.; Majounie, E.; Deramecourt, V.; Jacoupy, M.; Nicolas, A.; Cormier-Dequaire, F.; Hassoun, S.M.; Pujol, C.; Ciura, S.; et al. Loss of VPS13C Function in Autosomal-Recessive Parkinsonism Causes Mitochondrial Dysfunction and Increases PINK1/Parkin-Dependent Mitophagy. Am. J. Hum. Genet. 2016, 98, 500–513. [Google Scholar] [CrossRef]
- Pickrell, A.M.; Youle, R.J. The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron 2015, 85, 257–273. [Google Scholar] [CrossRef]
- Di Nottia, M.; Masciullo, M.; Verrigni, D.; Petrillo, S.; Modoni, A.; Rizzo, V.; Di Giuda, D.; Rizza, T.; Niceta, M.; Torraco, A.; et al. DJ-1 modulates mitochondrial response to oxidative stress: Clues from a novel diagnosis of PARK7. Clin. Genet. 2017, 92, 18–25. [Google Scholar] [CrossRef]
- Guzman, J.N.; Sanchez-Padilla, J.; Wokosin, D.; Kondapalli, J.; Ilijic, E.; Schumacker, P.T.; Surmeier, D.J. Oxidant stress evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. Nature 2010, 468, 696–700. [Google Scholar] [CrossRef]
- McCoy, M.K.; Cookson, M.R. Mitochondrial quality control and dynamics in Parkinson’s disease. Antioxid. Redox Signal. 2012, 16, 869–882. [Google Scholar] [CrossRef]
- Valente, E.M.; Abou-Sleiman, P.M.; Caputo, V.; Muqit, M.M.; Harvey, K.; Gispert, S.; Ali, Z.; Del Turco, D.; Bentivoglio, A.R.; Healy, D.G.; et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science 2004, 304, 1158–1160. [Google Scholar] [CrossRef]
- Abou-Sleiman, P.M.; Muqit, M.M.; McDonald, N.Q.; Yang, Y.X.; Gandhi, S.; Healy, D.G.; Harvey, K.; Harvey, R.J.; Deas, E.; Bhatia, K.; et al. A heterozygous effect for PINK1 mutations in Parkinson’s disease? Ann. Neurol. 2006, 60, 414–419. [Google Scholar] [CrossRef]
- Kim, C.; Lee, S.J. Controlling the mass action of alpha-synuclein in Parkinson’s disease. J. Neurochem. 2008, 107, 303–316. [Google Scholar] [CrossRef]
- Melki, R. Role of Different Alpha-Synuclein Strains in Synucleinopathies, Similarities with other Neurodegenerative Diseases. J. Park. Dis. 2015, 5, 217–227. [Google Scholar] [CrossRef]
- Kaushik, S.; Cuervo, A.M. Proteostasis and aging. Nat. Med. 2015, 21, 1406–1415. [Google Scholar] [CrossRef]
- Xilouri, M.; Brekk, O.R.; Stefanis, L. alpha-Synuclein and protein degradation systems: A reciprocal relationship. Mol. Neurobiol. 2013, 47, 537–551. [Google Scholar] [CrossRef]
- Braak, H.; Del Tredici, K.; Rub, U.; de Vos, R.A.; Jansen Steur, E.N.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef]
- Angot, E.; Steiner, J.A.; Hansen, C.; Li, J.Y.; Brundin, P. Are synucleinopathies prion-like disorders? Lancet Neurol. 2010, 9, 1128–1138. [Google Scholar] [CrossRef]
- Brundin, P.; Melki, R.; Kopito, R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat. Rev. Mol. Cell Biol. 2010, 11, 301–307. [Google Scholar] [CrossRef]
- George, S.; Rey, N.L.; Reichenbach, N.; Steiner, J.A.; Brundin, P. alpha-Synuclein: The long distance runner. Brain Pathol. 2013, 23, 350–357. [Google Scholar] [CrossRef]
- Brundin, P.; Li, J.Y.; Holton, J.L.; Lindvall, O.; Revesz, T. Research in motion: The enigma of Parkinson’s disease pathology spread. Nat. Rev. Neurosci. 2008, 9, 741–745. [Google Scholar] [CrossRef]
- Xilouri, M.; Vogiatzi, T.; Vekrellis, K.; Park, D.; Stefanis, L. Abberant alpha-synuclein confers toxicity to neurons in part through inhibition of chaperone-mediated autophagy. PLoS ONE 2009, 4, e5515. [Google Scholar] [CrossRef]
- Volpicelli-Daley, L.A.; Abdelmotilib, H.; Liu, Z.; Stoyka, L.; Daher, J.P.; Milnerwood, A.J.; Unni, V.K.; Hirst, W.D.; Yue, Z.; Zhao, H.T.; et al. G2019S-LRRK2 Expression Augments alpha-Synuclein Sequestration into Inclusions in Neurons. J. Neurosci. Off. J. Soc. Neurosci. 2016, 36, 7415–7427. [Google Scholar] [CrossRef]
- Fernandes, H.J.; Hartfield, E.M.; Christian, H.C.; Emmanoulidou, E.; Zheng, Y.; Booth, H.; Bogetofte, H.; Lang, C.; Ryan, B.J.; Sardi, S.P.; et al. ER Stress and Autophagic Perturbations Lead to Elevated Extracellular alpha-Synuclein in GBA-N370S Parkinson’s iPSC-Derived Dopamine Neurons. Stem Cell Rep. 2016, 6, 342–356. [Google Scholar] [CrossRef]
- Tang, F.L.; Erion, J.R.; Tian, Y.; Liu, W.; Yin, D.M.; Ye, J.; Tang, B.; Mei, L.; Xiong, W.C. VPS35 in Dopamine Neurons Is Required for Endosome-to-Golgi Retrieval of Lamp2a, a Receptor of Chaperone-Mediated Autophagy That Is Critical for alpha-Synuclein Degradation and Prevention of Pathogenesis of Parkinson’s Disease. J. Neurosci. Off. J. Soc. Neurosci. 2015, 35, 10613–10628. [Google Scholar] [CrossRef]
- Steele, J.W.; Ju, S.; Lachenmayer, M.L.; Liken, J.; Stock, A.; Kim, S.H.; Delgado, L.M.; Alfaro, I.E.; Bernales, S.; Verdile, G.; et al. Latrepirdine stimulates autophagy and reduces accumulation of alpha-synuclein in cells and in mouse brain. Mol. Psychiatry 2013, 18, 882–888. [Google Scholar] [CrossRef]
- Sarkar, S.; Davies, J.E.; Huang, Z.; Tunnacliffe, A.; Rubinsztein, D.C. Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and alpha-synuclein. J. Biol. Chem. 2007, 282, 5641–5652. [Google Scholar] [CrossRef]
- Emmanouilidou, E.; Stefanis, L.; Vekrellis, K. Cell-produced alpha-synuclein oligomers are targeted to, and impair, the 26S proteasome. Neurobiol. Aging 2010, 31, 953–968. [Google Scholar] [CrossRef]
- Zheng, B.; Liao, Z.; Locascio, J.J.; Lesniak, K.A.; Roderick, S.S.; Watt, M.L.; Eklund, A.C.; Zhang-James, Y.; Kim, P.D.; Hauser, M.A.; et al. PGC-1alpha, a potential therapeutic target for early intervention in Parkinson’s disease. Sci. Transl. Med. 2010, 2, 52ra73. [Google Scholar] [CrossRef]
- Devi, L.; Raghavendran, V.; Prabhu, B.M.; Avadhani, N.G.; Anandatheerthavarada, H.K. Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J. Biol. Chem. 2008, 283, 9089–9100. [Google Scholar] [CrossRef]
- Eschbach, J.; von Einem, B.; Muller, K.; Bayer, H.; Scheffold, A.; Morrison, B.E.; Rudolph, K.L.; Thal, D.R.; Witting, A.; Weydt, P.; et al. Mutual exacerbation of peroxisome proliferator-activated receptor gamma coactivator 1alpha deregulation and alpha-synuclein oligomerization. Ann. Neurol. 2015, 77, 15–32. [Google Scholar] [CrossRef]
- Dias, V.; Junn, E.; Mouradian, M.M. The role of oxidative stress in Parkinson’s disease. J. Park. Dis. 2013, 3, 461–491. [Google Scholar] [CrossRef]
- Bolam, J.P.; Pissadaki, E.K. Living on the edge with too many mouths to feed: Why dopamine neurons die. Mov. Disord. Off. J. Mov. Disord. Soc. 2012, 27, 1478–1483. [Google Scholar] [CrossRef]
- Pissadaki, E.K.; Bolam, J.P. The energy cost of action potential propagation in dopamine neurons: Clues to susceptibility in Parkinson’s disease. Front. Comput. Neurosci. 2013, 7, 13. [Google Scholar] [CrossRef]
- Surmeier, D.J.; Guzman, J.N.; Sanchez-Padilla, J.; Schumacker, P.T. The role of calcium and mitochondrial oxidant stress in the loss of substantia nigra pars compacta dopaminergic neurons in Parkinson’s disease. Neuroscience 2011, 198, 221–231. [Google Scholar] [CrossRef]
- Surmeier, D.J.; Schumacker, P.T.; Guzman, J.D.; Ilijic, E.; Yang, B.; Zampese, E. Calcium and Parkinson’s disease. Biochem. Biophys. Res. Commun. 2017, 483, 1013–1019. [Google Scholar] [CrossRef]
- Moehle, M.S.; West, A.B. M1 and M2 immune activation in Parkinson’s Disease: Foe and ally? Neuroscience 2015, 302, 59–73. [Google Scholar] [CrossRef]
- Ransohoff, R.M. How neuroinflammation contributes to neurodegeneration. Science 2016, 353, 777–783. [Google Scholar] [CrossRef]
- Hirsch, E.C.; Hunot, S. Neuroinflammation in Parkinson’s disease: A target for neuroprotection? Lancet Neurol. 2009, 8, 382–397. [Google Scholar] [CrossRef]
- Gao, H.M.; Kotzbauer, P.T.; Uryu, K.; Leight, S.; Trojanowski, J.Q.; Lee, V.M. Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. J. Neurosci. Off. J. Soc. Neurosci. 2008, 28, 7687–7698. [Google Scholar] [CrossRef]
- Jao, C.C.; Hegde, B.G.; Chen, J.; Haworth, I.S.; Langen, R. Structure of membrane-bound alpha-synuclein from site-directed spin labeling and computational refinement. Proc. Natl. Acad. Sci. USA 2008, 105, 19666–19671. [Google Scholar] [CrossRef] [PubMed]
- Bartels, T.; Choi, J.G.; Selkoe, D.J. alpha-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation. Nature 2011, 477, 107–110. [Google Scholar] [CrossRef] [PubMed]
- Bendor, J.T.; Logan, T.P.; Edwards, R.H. The function of alpha-synuclein. Neuron 2013, 79, 1044–1066. [Google Scholar] [CrossRef] [PubMed]
- Sun, J.; Wang, L.; Bao, H.; Premi, S.; Das, U.; Chapman, E.R.; Roy, S. Functional cooperation of alpha-synuclein and VAMP2 in synaptic vesicle recycling. Proc. Natl. Acad. Sci. USA 2019, 116, 11113–11115. [Google Scholar] [CrossRef]
- Burre, J.; Sharma, M.; Tsetsenis, T.; Buchman, V.; Etherton, M.R.; Sudhof, T.C. Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science 2010, 329, 1663–1667. [Google Scholar] [CrossRef]
- Burre, J.; Sharma, M.; Sudhof, T.C. alpha-Synuclein assembles into higher-order multimers upon membrane binding to promote SNARE complex formation. Proc. Natl. Acad. Sci. USA 2014, 111, E4274–E4283. [Google Scholar] [CrossRef]
- Hallacli, E.; Kayatekin, C.; Nazeen, S.; Wang, X.H.; Sheinkopf, Z.; Sathyakumar, S.; Sarkar, S.; Jiang, X.; Dong, X.; Di Maio, R.; et al. The Parkinson’s disease protein alpha-synuclein is a modulator of processing bodies and mRNA stability. Cell 2022, 185, 2035–2056.e33. [Google Scholar] [CrossRef]
- Schaser, A.J.; Osterberg, V.R.; Dent, S.E.; Stackhouse, T.L.; Wakeham, C.M.; Boutros, S.W.; Weston, L.J.; Owen, N.; Weissman, T.A.; Luna, E.; et al. Alpha-synuclein is a DNA binding protein that modulates DNA repair with implications for Lewy body disorders. Sci. Rep. 2019, 9, 10919. [Google Scholar] [CrossRef]
- Fusco, G.; De Simone, A.; Gopinath, T.; Vostrikov, V.; Vendruscolo, M.; Dobson, C.M.; Veglia, G. Direct observation of the three regions in alpha-synuclein that determine its membrane-bound behaviour. Nat. Commun. 2014, 5, 3827. [Google Scholar] [CrossRef]
- Ulmer, T.S.; Bax, A.; Cole, N.B.; Nussbaum, R.L. Structure and dynamics of micelle-bound human alpha-synuclein. J. Biol. Chem. 2005, 280, 9595–9603. [Google Scholar] [CrossRef]
- Zhu, M.; Li, J.; Fink, A.L. The association of alpha-synuclein with membranes affects bilayer structure, stability, and fibril formation. J. Biol. Chem. 2003, 278, 40186–40197. [Google Scholar] [CrossRef] [PubMed]
- Galvagnion, C.; Brown, J.W.; Ouberai, M.M.; Flagmeier, P.; Vendruscolo, M.; Buell, A.K.; Sparr, E.; Dobson, C.M. Chemical properties of lipids strongly affect the kinetics of the membrane-induced aggregation of alpha-synuclein. Proc. Natl. Acad. Sci. USA 2016, 113, 7065–7070. [Google Scholar] [CrossRef] [PubMed]
- Kiechle, M.; Grozdanov, V.; Danzer, K.M. The Role of Lipids in the Initiation of alpha-Synuclein Misfolding. Front. Cell Dev. Biol. 2020, 8, 562241. [Google Scholar] [CrossRef]
- O’Leary, E.I.; Lee, J.C. Interplay between alpha-synuclein amyloid formation and membrane structure. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 483–491. [Google Scholar] [CrossRef] [PubMed]
- Doherty, C.P.A.; Ulamec, S.M.; Maya-Martinez, R.; Good, S.C.; Makepeace, J.; Khan, G.N.; van Oosten-Hawle, P.; Radford, S.E.; Brockwell, D.J. A short motif in the N-terminal region of alpha-synuclein is critical for both aggregation and function. Nat. Struct. Mol. Biol. 2020, 27, 249–259. [Google Scholar] [CrossRef]
- McGlinchey, R.P.; Ni, X.; Shadish, J.A.; Jiang, J.; Lee, J.C. The N terminus of alpha-synuclein dictates fibril formation. Proc. Natl. Acad. Sci. USA 2021, 118, e2023487118. [Google Scholar] [CrossRef]
- Buratti, F.A.; Boeffinger, N.; Garro, H.A.; Flores, J.S.; Hita, F.J.; Goncalves, P.D.C.; Copello, F.D.R.; Lizarraga, L.; Rossetti, G.; Carloni, P.; et al. Aromaticity at position 39 in alpha-synuclein: A modulator of amyloid fibril assembly and membrane-bound conformations. Protein Sci. 2022, 31, e4360. [Google Scholar] [CrossRef]
- Ueda, K.; Fukushima, H.; Masliah, E.; Xia, Y.; Iwai, A.; Yoshimoto, M.; Otero, D.A.; Kondo, J.; Ihara, Y.; Saitoh, T. Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc. Natl. Acad. Sci. USA 1993, 90, 11282–11286. [Google Scholar] [CrossRef]
- Rodriguez, 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.; et al. Structure of the toxic core of alpha-synuclein from invisible crystals. Nature 2015, 525, 486–490. [Google Scholar] [CrossRef]
- Guerrero-Ferreira, R.; Taylor, N.M.; Mona, D.; Ringler, P.; Lauer, M.E.; Riek, R.; Britschgi, M.; Stahlberg, H. Cryo-EM structure of alpha-synuclein fibrils. eLife 2018, 7. [Google Scholar] [CrossRef]
- Chakraborty, R.; Chattopadhyay, K. Cryo-Electron Microscopy Uncovers Key Residues within the Core of Alpha-Synuclein Fibrils. ACS Chem. Neurosci. 2019, 10, 1135–1136. [Google Scholar] [CrossRef] [PubMed]
- Izawa, Y.; Tateno, H.; Kameda, H.; Hirakawa, K.; Hato, K.; Yagi, H.; Hongo, K.; Mizobata, T.; Kawata, Y. Role of C-terminal negative charges and tyrosine residues in fibril formation of alpha-synuclein. Brain Behav. 2012, 2, 595–605. [Google Scholar] [CrossRef]
- Tuttle, M.D.; Comellas, G.; Nieuwkoop, A.J.; Covell, D.J.; Berthold, D.A.; Kloepper, K.D.; Courtney, J.M.; Kim, J.K.; Barclay, A.M.; Kendall, A.; et al. Solid-state NMR structure of a pathogenic fibril of full-length human alpha-synuclein. Nat. Struct. Mol. Biol. 2016, 23, 409–415. [Google Scholar] [CrossRef]
- Gillam, J.E.; MacPhee, C.E. Modelling amyloid fibril formation kinetics: Mechanisms of nucleation and growth. J. Phys. Condens. Matter Inst. Phys. J. 2013, 25, 373101. [Google Scholar] [CrossRef]
- Winner, B.; Jappelli, R.; Maji, S.K.; Desplats, P.A.; Boyer, L.; Aigner, S.; Hetzer, C.; Loher, T.; Vilar, M.; Campioni, S.; et al. In vivo demonstration that alpha-synuclein oligomers are toxic. Proc. Natl. Acad. Sci. USA 2011, 108, 4194–4199. [Google Scholar] [CrossRef]
- Invernizzi, G.; Papaleo, E.; Sabate, R.; Ventura, S. Protein aggregation: Mechanisms and functional consequences. Int. J. Biochem. Cell Biol. 2012, 44, 1541–1554. [Google Scholar] [CrossRef] [PubMed]
- Marvian, A.T.; Koss, D.J.; Aliakbari, F.; Morshedi, D.; Outeiro, T.F. In vitro models of synucleinopathies: Informing on molecular mechanisms and protective strategies. J. Neurochem. 2019, 150, 535–565. [Google Scholar] [CrossRef]
- Rutherford, N.J.; Moore, B.D.; Golde, T.E.; Giasson, B.I. Divergent effects of the H50Q and G51D SNCA mutations on the aggregation of alpha-synuclein. J. Neurochem. 2014, 131, 859–867. [Google Scholar] [CrossRef] [PubMed]
- Uversky, V.N.; Li, J.; Fink, A.L. Evidence for a partially folded intermediate in alpha-synuclein fibril formation. J. Biol. Chem. 2001, 276, 10737–10744. [Google Scholar] [CrossRef]
- Gaspar, R.; Lund, M.; Sparr, E.; Linse, S. Anomalous Salt Dependence Reveals an Interplay of Attractive and Repulsive Electrostatic Interactions in α-synuclein Fibril Formation. QRB Discov. 2020, 1, e2. [Google Scholar] [CrossRef]
- Ziaunys, M.; Sakalauskas, A.; Mikalauskaite, K.; Smirnovas, V. Polymorphism of Alpha-Synuclein Amyloid Fibrils Depends on Ionic Strength and Protein Concentration. Int. J. Mol. Sci. 2021, 22, 12382. [Google Scholar] [CrossRef]
- de Oliveira, G.A.P.; Silva, J.L. Alpha-synuclein stepwise aggregation reveals features of an early onset mutation in Parkinson’s disease. Commun. Biol. 2019, 2, 374. [Google Scholar] [CrossRef] [PubMed]
- Buell, A.K.; Galvagnion, C.; Gaspar, R.; Sparr, E.; Vendruscolo, M.; Knowles, T.P.; Linse, S.; Dobson, C.M. Solution conditions determine the relative importance of nucleation and growth processes in alpha-synuclein aggregation. Proc. Natl. Acad. Sci. USA 2014, 111, 7671–7676. [Google Scholar] [CrossRef]
- Bousset, L.; Pieri, L.; Ruiz-Arlandis, G.; Gath, J.; Jensen, P.H.; Habenstein, B.; Madiona, K.; Olieric, V.; Bockmann, A.; Meier, B.H.; et al. Structural and functional characterization of two alpha-synuclein strains. Nat. Commun. 2013, 4, 2575. [Google Scholar] [CrossRef]
- Pena-Diaz, S.; Pujols, J.; Vasili, E.; Pinheiro, F.; Santos, J.; Manglano-Artunedo, Z.; Outeiro, T.F.; Ventura, S. The small aromatic compound SynuClean-D inhibits the aggregation and seeded polymerization of multiple alpha-synuclein strains. J. Biol. Chem. 2022, 298, 101902. [Google Scholar] [CrossRef] [PubMed]
- Aguzzi, A.; Heikenwalder, M.; Polymenidou, M. Insights into prion strains and neurotoxicity. Nat. Rev. Mol. Cell Biol. 2007, 8, 552–561. [Google Scholar] [CrossRef] [PubMed]
- Behbahanipour, M.; Garcia-Pardo, J.; Ventura, S. Decoding the role of coiled-coil motifs in human prion-like proteins. Prion 2021, 15, 143–154. [Google Scholar] [CrossRef]
- Peelaerts, W.; Bousset, L.; Van der Perren, A.; Moskalyuk, A.; Pulizzi, R.; Giugliano, M.; Van den Haute, C.; Melki, R.; Baekelandt, V. alpha-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature 2015, 522, 340–344. [Google Scholar] [CrossRef]
- Yamasaki, T.R.; Holmes, B.B.; Furman, J.L.; Dhavale, D.D.; Su, B.W.; Song, E.S.; Cairns, N.J.; Kotzbauer, P.T.; Diamond, M.I. Parkinson’s disease and multiple system atrophy have distinct alpha-synuclein seed characteristics. J. Biol. Chem. 2019, 294, 1045–1058. [Google Scholar] [CrossRef]
- Guerrero-Ferreira, R.; Taylor, N.M.; Arteni, A.A.; Kumari, P.; Mona, D.; Ringler, P.; Britschgi, M.; Lauer, M.E.; Makky, A.; Verasdonck, J.; et al. Two new polymorphic structures of human full-length alpha-synuclein fibrils solved by cryo-electron microscopy. eLife 2019, 8, e48907. [Google Scholar] [CrossRef]
- Boyer, D.R.; Li, B.; Sun, C.; Fan, W.; Sawaya, M.R.; Jiang, L.; Eisenberg, D.S. Structures of fibrils formed by alpha-synuclein hereditary disease mutant H50Q reveal new polymorphs. Nat. Struct. Mol. Biol. 2019, 26, 1044–1052. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Ge, P.; Murray, K.A.; Sheth, P.; Zhang, M.; Nair, G.; Sawaya, M.R.; Shin, W.S.; Boyer, D.R.; Ye, S.; et al. Cryo-EM of full-length alpha-synuclein reveals fibril polymorphs with a common structural kernel. Nat. Commun. 2018, 9, 3609. [Google Scholar] [CrossRef] [PubMed]
- Schweighauser, M.; Shi, Y.; Tarutani, A.; Kametani, F.; Murzin, A.G.; Ghetti, B.; Matsubara, T.; Tomita, T.; Ando, T.; Hasegawa, K.; et al. Structures of alpha-synuclein filaments from multiple system atrophy. Nature 2020, 585, 464–469. [Google Scholar] [CrossRef] [PubMed]
- Shahnawaz, M.; Mukherjee, A.; Pritzkow, S.; Mendez, N.; Rabadia, P.; Liu, X.; Hu, B.; Schmeichel, A.; Singer, W.; Wu, G.; et al. Discriminating alpha-synuclein strains in Parkinson’s disease and multiple system atrophy. Nature 2020, 578, 273–277. [Google Scholar] [CrossRef] [PubMed]
- Lau, A.; So, R.W.L.; Lau, H.H.C.; Sang, J.C.; Ruiz-Riquelme, A.; Fleck, S.C.; Stuart, E.; Menon, S.; Visanji, N.P.; Meisl, G.; et al. alpha-Synuclein strains target distinct brain regions and cell types. Nat. Neurosci. 2020, 23, 21–31. [Google Scholar] [CrossRef] [PubMed]
- Ray, S.; Singh, N.; Kumar, R.; Patel, K.; Pandey, S.; Datta, D.; Mahato, J.; Panigrahi, R.; Navalkar, A.; Mehra, S.; et al. alpha-Synuclein aggregation nucleates through liquid-liquid phase separation. Nat. Chem. 2020, 12, 705–716. [Google Scholar] [CrossRef]
- Hardenberg, M.C.; Sinnige, T.; Casford, S.; Dada, S.T.; Poudel, C.; Robinson, E.A.; Fuxreiter, M.; Kaminksi, C.F.; Kaminski Schierle, G.S.; Nollen, E.A.A.; et al. Observation of an alpha-synuclein liquid droplet state and its maturation into Lewy body-like assemblies. J. Mol. Cell Biol. 2021, 13, 282–294. [Google Scholar] [CrossRef]
- Takamuku, M.; Sugishita, T.; Tamaki, H.; Dong, L.; So, M.; Fujiwara, T.; Matsuki, Y. Evolution of alpha-synuclein conformation ensemble toward amyloid fibril via liquid-liquid phase separation (LLPS) as investigated by dynamic nuclear polarization-enhanced solid-state MAS NMR. Neurochem. Int. 2022, 157, 105345. [Google Scholar] [CrossRef]
- Brangwynne, C.P.; Eckmann, C.R.; Courson, D.S.; Rybarska, A.; Hoege, C.; Gharakhani, J.; Julicher, F.; Hyman, A.A. Germline P granules are liquid droplets that localize by controlled dissolution/condensation. Science 2009, 324, 1729–1732. [Google Scholar] [CrossRef]
- Li, X.H.; Chavali, P.L.; Pancsa, R.; Chavali, S.; Babu, M.M. Function and Regulation of Phase-Separated Biological Condensates. Biochemistry 2018, 57, 2452–2461. [Google Scholar] [CrossRef]
- Molliex, A.; Temirov, J.; Lee, J.; Coughlin, M.; Kanagaraj, A.P.; Kim, H.J.; Mittag, T.; Taylor, J.P. Phase separation by low complexity domains promotes stress granule assembly and drives pathological fibrillization. Cell 2015, 163, 123–133. [Google Scholar] [CrossRef] [PubMed]
- Berry, J.; Weber, S.C.; Vaidya, N.; Haataja, M.; Brangwynne, C.P. RNA transcription modulates phase transition-driven nuclear body assembly. Proc. Natl. Acad. Sci. USA 2015, 112, E5237–E5245. [Google Scholar] [CrossRef] [PubMed]
- Li, P.; Banjade, S.; Cheng, H.C.; Kim, S.; Chen, B.; Guo, L.; Llaguno, M.; Hollingsworth, J.V.; King, D.S.; Banani, S.F.; et al. Phase transitions in the assembly of multivalent signalling proteins. Nature 2012, 483, 336–340. [Google Scholar] [CrossRef] [PubMed]
- Woodruff, J.B.; Ferreira Gomes, B.; Widlund, P.O.; Mahamid, J.; Honigmann, A.; Hyman, A.A. The Centrosome Is a Selective Condensate that Nucleates Microtubules by Concentrating Tubulin. Cell 2017, 169, 1066–1077.e1010. [Google Scholar] [CrossRef] [PubMed]
- Sawner, A.S.; Ray, S.; Yadav, P.; Mukherjee, S.; Panigrahi, R.; Poudyal, M.; Patel, K.; Ghosh, D.; Kummerant, E.; Kumar, A.; et al. Modulating alpha-Synuclein Liquid-Liquid Phase Separation. Biochemistry 2021, 60, 3676–3696. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.; Xu, B.; Liu, Y. Calcium promotes alpha-synuclein liquid-liquid phase separation to accelerate amyloid aggregation. Biochem. Biophys. Res. Commun. 2022, 603, 13–20. [Google Scholar] [CrossRef]
- Xu, B.; Huang, S.; Liu, Y.; Wan, C.; Gu, Y.; Wang, D.; Yu, H. Manganese promotes alpha-synuclein amyloid aggregation through the induction of protein phase transition. J. Biol. Chem. 2022, 298, 101469. [Google Scholar] [CrossRef]
- Oliva, R.; Mukherjee, S.K.; Ostermeier, L.; Pazurek, L.A.; Kriegler, S.; Bader, V.; Prumbaum, D.; Raunser, S.; Winklhofer, K.F.; Tatzelt, J.; et al. Remodeling of the Fibrillation Pathway of alpha-Synuclein by Interaction with Antimicrobial Peptide LL-III. Chemistry 2021, 27, 11845–11851. [Google Scholar] [CrossRef]
- Agarwal, A.; Arora, L.; Rai, S.K.; Avni, A.; Mukhopadhyay, S. Spatiotemporal modulations in heterotypic condensates of prion and alpha-synuclein control phase transitions and amyloid conversion. Nat. Commun. 2022, 13, 1154. [Google Scholar] [CrossRef]
- Siegert, A.; Rankovic, M.; Favretto, F.; Ukmar-Godec, T.; Strohaker, T.; Becker, S.; Zweckstetter, M. Interplay between tau and alpha-synuclein liquid-liquid phase separation. Protein Sci. 2021, 30, 1326–1336. [Google Scholar] [CrossRef]
- Fusco, G.; Chen, S.W.; Williamson, P.T.F.; Cascella, R.; Perni, M.; Jarvis, J.A.; Cecchi, C.; Vendruscolo, M.; Chiti, F.; Cremades, N.; et al. Structural basis of membrane disruption and cellular toxicity by alpha-synuclein oligomers. Science 2017, 358, 1440–1443. [Google Scholar] [CrossRef] [PubMed]
- Cremades, N.; Cohen, S.I.; Deas, E.; Abramov, A.Y.; Chen, A.Y.; Orte, A.; Sandal, M.; Clarke, R.W.; Dunne, P.; Aprile, F.A.; et al. Direct observation of the interconversion of normal and toxic forms of alpha-synuclein. Cell 2012, 149, 1048–1059. [Google Scholar] [CrossRef] [PubMed]
- Campioni, S.; Mannini, B.; Zampagni, M.; Pensalfini, A.; Parrini, C.; Evangelisti, E.; Relini, A.; Stefani, M.; Dobson, C.M.; Cecchi, C.; et al. A causative link between the structure of aberrant protein oligomers and their toxicity. Nat. Chem. Biol. 2010, 6, 140–147. [Google Scholar] [CrossRef]
- Cascella, R.; Chen, S.W.; Bigi, A.; Camino, J.D.; Xu, C.K.; Dobson, C.M.; Chiti, F.; Cremades, N.; Cecchi, C. The release of toxic oligomers from alpha-synuclein fibrils induces dysfunction in neuronal cells. Nat. Commun. 2021, 12, 1814. [Google Scholar] [CrossRef] [PubMed]
- Gadhe, L.; Sakunthala, A.; Mukherjee, S.; Gahlot, N.; Bera, R.; Sawner, A.S.; Kadu, P.; Maji, S.K. Intermediates of alpha-synuclein aggregation: Implications in Parkinson’s disease pathogenesis. Biophys. Chem. 2022, 281, 106736. [Google Scholar] [CrossRef]
- Cascella, R.; Bigi, A.; Cremades, N.; Cecchi, C. Effects of oligomer toxicity, fibril toxicity and fibril spreading in synucleinopathies. Cell. Mol. Life Sci. 2022, 79, 174. [Google Scholar] [CrossRef]
- Hansen, C.; Angot, E.; Bergstrom, A.L.; Steiner, J.A.; Pieri, L.; Paul, G.; Outeiro, T.F.; Melki, R.; Kallunki, P.; Fog, K.; et al. alpha-Synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J. Clin. Investig. 2011, 121, 715–725. [Google Scholar] [CrossRef]
- Rietdijk, C.D.; Perez-Pardo, P.; Garssen, J.; van Wezel, R.J.; Kraneveld, A.D. Exploring Braak’s Hypothesis of Parkinson’s Disease. Front. Neurol. 2017, 8, 37. [Google Scholar] [CrossRef]
- Alba, R.; Bosch, A.; Chillon, M. Gutless adenovirus: Last-generation adenovirus for gene therapy. Gene Ther. 2005, 12 (Suppl. S1), S18–S27. [Google Scholar] [CrossRef]
- Froelich, S.; Tai, A.; Wang, P. Lentiviral vectors for immune cells targeting. Immunopharmacol. Immunotoxicol. 2010, 32, 208–218. [Google Scholar] [CrossRef]
- Kotterman, M.A.; Schaffer, D.V. Engineering adeno-associated viruses for clinical gene therapy. Nat. Rev. Genet 2014, 15, 445–451. [Google Scholar] [CrossRef] [PubMed]
- LeWitt, P.A.; Rezai, A.R.; Leehey, M.A.; Ojemann, S.G.; Flaherty, A.W.; Eskandar, E.N.; Kostyk, S.K.; Thomas, K.; Sarkar, A.; Siddiqui, M.S.; et al. AAV2-GAD gene therapy for advanced Parkinson’s disease: A double-blind, sham-surgery controlled, randomised trial. Lancet Neurol. 2011, 10, 309–319. [Google Scholar] [CrossRef] [PubMed]
- Muramatsu, S.; Wang, L.; Ikeguchi, K.; Fujimoto, K.; Nakano, I.; Ozawa, K. Recombinant adeno-associated viral vectors bring gene therapy for Parkinson’s disease closer to reality. J. Neurol. 2002, 249 (Suppl. S2), II36–II40. [Google Scholar] [CrossRef]
- Jarraya, B.; Boulet, S.; Ralph, G.S.; Jan, C.; Bonvento, G.; Azzouz, M.; Miskin, J.E.; Shin, M.; Delzescaux, T.; Drouot, X.; et al. Dopamine gene therapy for Parkinson’s disease in a nonhuman primate without associated dyskinesia. Sci. Transl. Med. 2009, 1, 2ra4. [Google Scholar] [CrossRef]
- Marks, W.J., Jr.; Bartus, R.T.; Siffert, J.; Davis, C.S.; Lozano, A.; Boulis, N.; Vitek, J.; Stacy, M.; Turner, D.; Verhagen, L.; et al. Gene delivery of AAV2-neurturin for Parkinson’s disease: A double-blind, randomised, controlled trial. Lancet Neurol. 2010, 9, 1164–1172. [Google Scholar] [CrossRef] [PubMed]
- Bjorklund, A.; Kirik, D.; Rosenblad, C.; Georgievska, B.; Lundberg, C.; Mandel, R.J. Towards a neuroprotective gene therapy for Parkinson’s disease: Use of adenovirus, AAV and lentivirus vectors for gene transfer of GDNF to the nigrostriatal system in the rat Parkinson model. Brain Res. 2000, 886, 82–98. [Google Scholar] [CrossRef]
- Tian, Y.Y.; Tang, C.J.; Wang, J.N.; Feng, Y.; Chen, X.W.; Wang, L.; Qiao, X.; Sun, S.G. Favorable effects of VEGF gene transfer on a rat model of Parkinson disease using adeno-associated viral vectors. Neurosci. Lett. 2007, 421, 239–244. [Google Scholar] [CrossRef]
- Oh, S.M.; Chang, M.Y.; Song, J.J.; Rhee, Y.H.; Joe, E.H.; Lee, H.S.; Yi, S.H.; Lee, S.H. Combined Nurr1 and Foxa2 roles in the therapy of Parkinson’s disease. EMBO Mol. Med. 2015, 7, 510–525. [Google Scholar] [CrossRef]
- Rahman, M.U.; Bilal, M.; Shah, J.A.; Kaushik, A.; Teissedre, P.L.; Kujawska, M. CRISPR-Cas9-Based Technology and Its Relevance to Gene Editing in Parkinson’s Disease. Pharmaceutics 2022, 14, 1252. [Google Scholar] [CrossRef]
- Lindvall, O.; Brundin, P.; Widner, H.; Rehncrona, S.; Gustavii, B.; Frackowiak, R.; Leenders, K.L.; Sawle, G.; Rothwell, J.C.; Marsden, C.D.; et al. Grafts of fetal dopamine neurons survive and improve motor function in Parkinson’s disease. Science 1990, 247, 574–577. [Google Scholar] [CrossRef]
- Piccini, P.; Brooks, D.J.; Bjorklund, A.; Gunn, R.N.; Grasby, P.M.; Rimoldi, O.; Brundin, P.; Hagell, P.; Rehncrona, S.; Widner, H.; et al. Dopamine release from nigral transplants visualized in vivo in a Parkinson’s patient. Nat. Neurosci. 1999, 2, 1137–1140. [Google Scholar] [CrossRef] [PubMed]
- Barker, R.A.; Drouin-Ouellet, J.; Parmar, M. Cell-based therapies for Parkinson disease-past insights and future potential. Nat. Rev. Neurol. 2015, 11, 492–503. [Google Scholar] [CrossRef] [PubMed]
- Petit, G.H.; Olsson, T.T.; Brundin, P. The future of cell therapies and brain repair: Parkinson’s disease leads the way. Neuropathol. Appl. Neurobiol. 2014, 40, 60–70. [Google Scholar] [CrossRef] [PubMed]
- Hauser, R.A. alpha-Synuclein in Parkinson’s disease: Getting to the core of the matter. Lancet Neurol. 2015, 14, 785–786. [Google Scholar] [CrossRef]
- Kingwell, K. Zeroing in on neurodegenerative alpha-synuclein. Nat. Rev. Drug Discov. 2017, 16, 371–373. [Google Scholar] [CrossRef] [PubMed]
- Mikitsh, J.L.; Chacko, A.M. Pathways for small molecule delivery to the central nervous system across the blood-brain barrier. Perspect. Medicin. Chem. 2014, 6, 11–24. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Zhu, M.; Manning-Bog, A.B.; Di Monte, D.A.; Fink, A.L. Dopamine and L-dopa disaggregate amyloid fibrils: Implications for Parkinson’s and Alzheimer’s disease. FASEB J. 2004, 18, 962–964. [Google Scholar] [CrossRef] [PubMed]
- Li, H.T.; Lin, D.H.; Luo, X.Y.; Zhang, F.; Ji, L.N.; Du, H.N.; Song, G.Q.; Hu, J.; Zhou, J.W.; Hu, H.Y. Inhibition of alpha-synuclein fibrillization by dopamine analogs via reaction with the amino groups of alpha-synuclein. Implication for dopaminergic neurodegeneration. FEBS J. 2005, 272, 3661–3672. [Google Scholar] [CrossRef] [PubMed]
- Leong, S.L.; Pham, C.L.; Galatis, D.; Fodero-Tavoletti, M.T.; Perez, K.; Hill, A.F.; Masters, C.L.; Ali, F.E.; Barnham, K.J.; Cappai, R. Formation of dopamine-mediated alpha-synuclein-soluble oligomers requires methionine oxidation. Free Radic. Biol. Med. 2009, 46, 1328–1337. [Google Scholar] [CrossRef]
- Uversky, V.N.; Yamin, G.; Souillac, P.O.; Goers, J.; Glaser, C.B.; Fink, A.L. Methionine oxidation inhibits fibrillation of human alpha-synuclein in vitro. FEBS Lett. 2002, 517, 239–244. [Google Scholar] [CrossRef]
- Norris, E.H.; Giasson, B.I.; Hodara, R.; Xu, S.; Trojanowski, J.Q.; Ischiropoulos, H.; Lee, V.M. Reversible inhibition of alpha-synuclein fibrillization by dopaminochrome-mediated conformational alterations. J. Biol. Chem. 2005, 280, 21212–21219. [Google Scholar] [CrossRef] [PubMed]
- Latawiec, D.; Herrera, F.; Bek, A.; Losasso, V.; Candotti, M.; Benetti, F.; Carlino, E.; Kranjc, A.; Lazzarino, M.; Gustincich, S.; et al. Modulation of alpha-synuclein aggregation by dopamine analogs. PLoS ONE 2010, 5, e9234. [Google Scholar] [CrossRef] [PubMed]
- Herrera, F.E.; Chesi, A.; Paleologou, K.E.; Schmid, A.; Munoz, A.; Vendruscolo, M.; Gustincich, S.; Lashuel, H.A.; Carloni, P. Inhibition of alpha-synuclein fibrillization by dopamine is mediated by interactions with five C-terminal residues and with E83 in the NAC region. PLoS ONE 2008, 3, e3394. [Google Scholar] [CrossRef] [PubMed]
- Mazzulli, J.R.; Armakola, M.; Dumoulin, M.; Parastatidis, I.; Ischiropoulos, H. Cellular oligomerization of alpha-synuclein is determined by the interaction of oxidized catechols with a C-terminal sequence. J. Biol. Chem. 2007, 282, 31621–31630. [Google Scholar] [CrossRef] [PubMed]
- Dibenedetto, D.; Rossetti, G.; Caliandro, R.; Carloni, P. A molecular dynamics simulation-based interpretation of nuclear magnetic resonance multidimensional heteronuclear spectra of alpha-synuclein.dopamine adducts. Biochemistry 2013, 52, 6672–6683. [Google Scholar] [CrossRef]
- Mor, D.E.; Tsika, E.; Mazzulli, J.R.; Gould, N.S.; Kim, H.; Daniels, M.J.; Doshi, S.; Gupta, P.; Grossman, J.L.; Tan, V.X.; et al. Dopamine induces soluble alpha-synuclein oligomers and nigrostriatal degeneration. Nat. Neurosci. 2017, 20, 1560–1568. [Google Scholar] [CrossRef]
- Mor, D.E.; Daniels, M.J.; Ischiropoulos, H. The usual suspects, dopamine and alpha-synuclein, conspire to cause neurodegeneration. Mov. Disord. Off. J. Mov. Disord. Soc. 2019, 34, 167–179. [Google Scholar] [CrossRef]
- Ono, K.; Yamada, M. Antioxidant compounds have potent anti-fibrillogenic and fibril-destabilizing effects for alpha-synuclein fibrils in vitro. J. Neurochem. 2006, 97, 105–115. [Google Scholar] [CrossRef] [PubMed]
- Masuda, M.; Suzuki, N.; Taniguchi, S.; Oikawa, T.; Nonaka, T.; Iwatsubo, T.; Hisanaga, S.; Goedert, M.; Hasegawa, M. Small molecule inhibitors of alpha-synuclein filament assembly. Biochemistry 2006, 45, 6085–6094. [Google Scholar] [CrossRef]
- Dhouafli, Z.; Cuanalo-Contreras, K.; Hayouni, E.A.; Mays, C.E.; Soto, C.; Moreno-Gonzalez, I. Inhibition of protein misfolding and aggregation by natural phenolic compounds. Cell. Mol. Life Sci. 2018, 75, 3521–3538. [Google Scholar] [CrossRef]
- Hornedo-Ortega, R.; Alvarez-Fernandez, M.A.; Cerezo, A.B.; Richard, T.; Troncoso, A.M.A.; Garcia-Parrilla, M.A.C. Protocatechuic Acid: Inhibition of Fibril Formation, Destabilization of Preformed Fibrils of Amyloid-beta and alpha-Synuclein, and Neuroprotection. J. Agric. Food Chem. 2016, 64, 7722–7732. [Google Scholar] [CrossRef] [PubMed]
- Pandey, N.; Strider, J.; Nolan, W.C.; Yan, S.X.; Galvin, J.E. Curcumin inhibits aggregation of alpha-synuclein. Acta Neuropathol. 2008, 115, 479–489. [Google Scholar] [CrossRef] [PubMed]
- Zhu, M.; Rajamani, S.; Kaylor, J.; Han, S.; Zhou, F.; Fink, A.L. The flavonoid baicalein inhibits fibrillation of alpha-synuclein and disaggregates existing fibrils. J. Biol. Chem. 2004, 279, 26846–26857. [Google Scholar] [CrossRef] [PubMed]
- Fazili, N.A.; Naeem, A. Anti-fibrillation potency of caffeic acid against an antidepressant induced fibrillogenesis of human alpha-synuclein: Implications for Parkinson’s disease. Biochimie 2015, 108, 178–185. [Google Scholar] [CrossRef] [PubMed]
- Ahmad, B.; Lapidus, L.J. Curcumin prevents aggregation in alpha-synuclein by increasing reconfiguration rate. J. Biol. Chem. 2012, 287, 9193–9199. [Google Scholar] [CrossRef]
- Ehrnhoefer, D.E.; Bieschke, J.; Boeddrich, A.; Herbst, M.; Masino, L.; Lurz, R.; Engemann, S.; Pastore, A.; Wanker, E.E. EGCG redirects amyloidogenic polypeptides into unstructured, off-pathway oligomers. Nat. Struct. Mol. Biol. 2008, 15, 558–566. [Google Scholar] [CrossRef]
- Lorenzen, N.; Nielsen, S.B.; Yoshimura, Y.; Vad, B.S.; Andersen, C.B.; Betzer, C.; Kaspersen, J.D.; Christiansen, G.; Pedersen, J.S.; Jensen, P.H.; et al. How epigallocatechin gallate can inhibit alpha-synuclein oligomer toxicity in vitro. J. Biol. Chem. 2014, 289, 21299–21310. [Google Scholar] [CrossRef]
- Bieschke, J.; Russ, J.; Friedrich, R.P.; Ehrnhoefer, D.E.; Wobst, H.; Neugebauer, K.; Wanker, E.E. EGCG remodels mature alpha-synuclein and amyloid-beta fibrils and reduces cellular toxicity. Proc. Natl. Acad. Sci. USA 2010, 107, 7710–7715. [Google Scholar] [CrossRef]
- Singh, P.K.; Kotia, V.; Ghosh, D.; Mohite, G.M.; Kumar, A.; Maji, S.K. Curcumin modulates alpha-synuclein aggregation and toxicity. ACS Chem. Neurosci. 2013, 4, 393–407. [Google Scholar] [CrossRef]
- Xu, B.; Chen, J.; Liu, Y. Curcumin Interacts with alpha-Synuclein Condensates To Inhibit Amyloid Aggregation under Phase Separation. ACS Omega 2022, 7, 30281–30290. [Google Scholar] [CrossRef]
- Xu, B.; Mo, X.; Chen, J.; Yu, H.; Liu, Y. Myricetin Inhibits alpha-Synuclein Amyloid Aggregation by Delaying the Liquid-to-Solid Phase Transition. Chembiochem 2022, 23, e202200216. [Google Scholar] [CrossRef] [PubMed]
- Ardah, M.T.; Paleologou, K.E.; Lv, G.; Abul Khair, S.B.; Kazim, A.S.; Minhas, S.T.; Al-Tel, T.H.; Al-Hayani, A.A.; Haque, M.E.; Eliezer, D.; et al. Structure activity relationship of phenolic acid inhibitors of alpha-synuclein fibril formation and toxicity. Front. Aging Neurosci. 2014, 6, 197. [Google Scholar] [CrossRef] [PubMed]
- Kujawska, M.; Jodynis-Liebert, J. Polyphenols in Parkinson’s Disease: A Systematic Review of In Vivo Studies. Nutrients 2018, 10, 642. [Google Scholar] [CrossRef] [PubMed]
- Hu, Q.; Uversky, V.N.; Huang, M.; Kang, H.; Xu, F.; Liu, X.; Lian, L.; Liang, Q.; Jiang, H.; Liu, A.; et al. Baicalein inhibits alpha-synuclein oligomer formation and prevents progression of alpha-synuclein accumulation in a rotenone mouse model of Parkinson’s disease. Biochim. Biophys. Acta 2016, 1862, 1883–1890. [Google Scholar] [CrossRef]
- Sharma, N.; Nehru, B. Curcumin affords neuroprotection and inhibits alpha-synuclein aggregation in lipopolysaccharide-induced Parkinson’s disease model. Inflammopharmacology 2018, 26, 349–360. [Google Scholar] [CrossRef]
- Ogawa, N.; Mizukawa, K.; Hirose, Y.; Kajita, S.; Ohara, S.; Watanabe, Y. MPTP-induced parkinsonian model in mice: Biochemistry, pharmacology and behavior. Eur. Neurol. 1987, 26 (Suppl. S1), 16–23. [Google Scholar] [CrossRef]
- Jeon, B.S.; Jackson-Lewis, V.; Burke, R.E. 6-Hydroxydopamine lesion of the rat substantia nigra: Time course and morphology of cell death. Neurodegeneration 1995, 4, 131–137. [Google Scholar] [CrossRef]
- Pan-Montojo, F.; Anichtchik, O.; Dening, Y.; Knels, L.; Pursche, S.; Jung, R.; Jackson, S.; Gille, G.; Spillantini, M.G.; Reichmann, H.; et al. Progression of Parkinson’s disease pathology is reproduced by intragastric administration of rotenone in mice. PLoS ONE 2010, 5, e8762. [Google Scholar] [CrossRef]
- Vajragupta, O.; Boonchoong, P.; Watanabe, H.; Tohda, M.; Kummasud, N.; Sumanont, Y. Manganese complexes of curcumin and its derivatives: Evaluation for the radical scavenging ability and neuroprotective activity. Free Radic. Biol. Med. 2003, 35, 1632–1644. [Google Scholar] [CrossRef]
- Rajeswari, A.; Sabesan, M. Inhibition of monoamine oxidase-B by the polyphenolic compound, curcumin and its metabolite tetrahydrocurcumin, in a model of Parkinson’s disease induced by MPTP neurodegeneration in mice. Inflammopharmacology 2008, 16, 96–99. [Google Scholar] [CrossRef]
- Nagarajan, S.; Chellappan, D.R.; Chinnaswamy, P.; Thulasingam, S. Ferulic acid pretreatment mitigates MPTP-induced motor impairment and histopathological alterations in C57BL/6 mice. Pharm. Biol. 2015, 53, 1591–1601. [Google Scholar] [CrossRef]
- Lv, C.; Hong, T.; Yang, Z.; Zhang, Y.; Wang, L.; Dong, M.; Zhao, J.; Mu, J.; Meng, Y. Effect of Quercetin in the 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-Induced Mouse Model of Parkinson’s Disease. Evid. Based Complement. Alternat. Med. 2012, 2012, 928643. [Google Scholar] [CrossRef]
- Yu, X.; He, G.R.; Sun, L.; Lan, X.; Shi, L.L.; Xuan, Z.H.; Du, G.H. Assessment of the treatment effect of baicalein on a model of Parkinsonian tremor and elucidation of the mechanism. Life Sci. 2012, 91, 5–13. [Google Scholar] [CrossRef]
- Li, S.; Pu, X.P. Neuroprotective effect of kaempferol against a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse model of Parkinson’s disease. Biol. Pharm. Bull. 2011, 34, 1291–1296. [Google Scholar] [CrossRef]
- Lee, E.; Park, H.R.; Ji, S.T.; Lee, Y.; Lee, J. Baicalein attenuates astroglial activation in the 1-methyl-4-phenyl-1,2,3,4-tetrahydropyridine-induced Parkinson’s disease model by downregulating the activations of nuclear factor-kappaB, ERK, and JNK. J. Neurosci. Res. 2014, 92, 130–139. [Google Scholar] [CrossRef]
- Chen, M.; Wang, T.; Yue, F.; Li, X.; Wang, P.; Li, Y.; Chan, P.; Yu, S. Tea polyphenols alleviate motor impairments, dopaminergic neuronal injury, and cerebral alpha-synuclein aggregation in MPTP-intoxicated parkinsonian monkeys. Neuroscience 2015, 286, 383–392. [Google Scholar] [CrossRef] [PubMed]
- Spinelli, K.J.; Osterberg, V.R.; Meshul, C.K.; Soumyanath, A.; Unni, V.K. Curcumin Treatment Improves Motor Behavior in alpha-Synuclein Transgenic Mice. PLoS ONE 2015, 10, e0128510. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.; Kumar, R.; Kumari, M.; Kumari, R.; Saha, S.; Bhavesh, N.S.; Maiti, T.K. Ellagic Acid Inhibits alpha-Synuclein Aggregation at Multiple Stages and Reduces Its Cytotoxicity. ACS Chem. Neurosci. 2021, 12, 1919–1930. [Google Scholar] [CrossRef] [PubMed]
- Ardah, M.T.; Eid, N.; Kitada, T.; Haque, M.E. Ellagic Acid Prevents alpha-Synuclein Aggregation and Protects SH-SY5Y Cells from Aggregated alpha-Synuclein-Induced Toxicity via Suppression of Apoptosis and Activation of Autophagy. Int. J. Mol. Sci. 2021, 22, 13398. [Google Scholar] [CrossRef]
- Chen, M.; Liu, A.; Ouyang, Y.; Huang, Y.; Chao, X.; Pi, R. Fasudil and its analogs: A new powerful weapon in the long war against central nervous system disorders? Expert Opin. Investig. Drugs 2013, 22, 537–550. [Google Scholar] [CrossRef]
- He, Q.; Li, Y.H.; Guo, S.S.; Wang, Y.; Lin, W.; Zhang, Q.; Wang, J.; Ma, C.G.; Xiao, B.G. Inhibition of Rho-kinase by Fasudil protects dopamine neurons and attenuates inflammatory response in an intranasal lipopolysaccharide-mediated Parkinson’s model. Eur. J. Neurosci. 2016, 43, 41–52. [Google Scholar] [CrossRef]
- Tatenhorst, L.; Eckermann, K.; Dambeck, V.; Fonseca-Ornelas, L.; Walle, H.; Lopes da Fonseca, T.; Koch, J.C.; Becker, S.; Tonges, L.; Bahr, M.; et al. Fasudil attenuates aggregation of alpha-synuclein in models of Parkinson’s disease. Acta Neuropathol. Commun. 2016, 4, 39. [Google Scholar] [CrossRef] [PubMed]
- Robustelli, P.; Ibanez-de-Opakua, A.; Campbell-Bezat, C.; Giordanetto, F.; Becker, S.; Zweckstetter, M.; Pan, A.C.; Shaw, D.E. Molecular Basis of Small-Molecule Binding to alpha-Synuclein. J. Am. Chem. Soc. 2022, 144, 2501–2510. [Google Scholar] [CrossRef] [PubMed]
- Giasson, B.I.; Duda, J.E.; Quinn, S.M.; Zhang, B.; Trojanowski, J.Q.; Lee, V.M. Neuronal alpha-synucleinopathy with severe movement disorder in mice expressing A53T human alpha-synuclein. Neuron 2002, 34, 521–533. [Google Scholar] [CrossRef] [PubMed]
- Koprich, J.B.; Johnston, T.H.; Reyes, M.G.; Sun, X.; Brotchie, J.M. Expression of human A53T alpha-synuclein in the rat substantia nigra using a novel AAV1/2 vector produces a rapidly evolving pathology with protein aggregation, dystrophic neurite architecture and nigrostriatal degeneration with potential to model the pathology of Parkinson’s disease. Mol. Neurodegener. 2010, 5, 43. [Google Scholar] [CrossRef]
- Yang, Y.J.; Bu, L.L.; Shen, C.; Ge, J.J.; He, S.J.; Yu, H.L.; Tang, Y.L.; Jue, Z.; Sun, Y.M.; Yu, W.B.; et al. Fasudil Promotes alpha-Synuclein Clearance in an AAV-Mediated alpha-Synuclein Rat Model of Parkinson’s Disease by Autophagy Activation. J. Park. Dis. 2020, 10, 969–979. [Google Scholar] [CrossRef]
- Wood, J.G.; Mirra, S.S.; Pollock, N.J.; Binder, L.I. Neurofibrillary tangles of Alzheimer disease share antigenic determinants with the axonal microtubule-associated protein tau (tau). Proc. Natl. Acad. Sci. USA 1986, 83, 4040–4043. [Google Scholar] [CrossRef]
- Delacourte, A.; Defossez, A. Alzheimer’s disease: Tau proteins, the promoting factors of microtubule assembly, are major components of paired helical filaments. J. Neurol. Sci. 1986, 76, 173–186. [Google Scholar] [CrossRef]
- Wischik, C.M.; Edwards, P.C.; Lai, R.Y.; Roth, M.; Harrington, C.R. Selective inhibition of Alzheimer disease-like tau aggregation by phenothiazines. Proc. Natl. Acad. Sci. USA 1996, 93, 11213–11218. [Google Scholar] [CrossRef]
- Wischik, C.M.; Staff, R.T.; Wischik, D.J.; Bentham, P.; Murray, A.D.; Storey, J.M.; Kook, K.A.; Harrington, C.R. Tau aggregation inhibitor therapy: An exploratory phase 2 study in mild or moderate Alzheimer’s disease. J. Alzheimers Dis. 2015, 44, 705–720. [Google Scholar] [CrossRef]
- Gauthier, S.; Feldman, H.H.; Schneider, L.S.; Wilcock, G.K.; Frisoni, G.B.; Hardlund, J.H.; Moebius, H.J.; Bentham, P.; Kook, K.A.; Wischik, D.J.; et al. Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer’s disease: A randomised, controlled, double-blind, parallel-arm, phase 3 trial. Lancet 2016, 388, 2873–2884. [Google Scholar] [CrossRef] [PubMed]
- Wilcock, G.K.; Gauthier, S.; Frisoni, G.B.; Jia, J.; Hardlund, J.H.; Moebius, H.J.; Bentham, P.; Kook, K.A.; Schelter, B.O.; Wischik, D.J.; et al. Potential of Low Dose Leuco-Methylthioninium Bis(Hydromethanesulphonate) (LMTM) Monotherapy for Treatment of Mild Alzheimer’s Disease: Cohort Analysis as Modified Primary Outcome in a Phase III Clinical Trial. J. Alzheimers Dis. 2018, 61, 435–457. [Google Scholar] [CrossRef] [PubMed]
- Schwab, K.; Frahm, S.; Horsley, D.; Rickard, J.E.; Melis, V.; Goatman, E.A.; Magbagbeolu, M.; Douglas, M.; Leith, M.G.; Baddeley, T.C.; et al. A Protein Aggregation Inhibitor, Leuco-Methylthioninium Bis(Hydromethanesulfonate), Decreases alpha-Synuclein Inclusions in a Transgenic Mouse Model of Synucleinopathy. Front. Mol. Neurosci. 2017, 10, 447. [Google Scholar] [CrossRef] [PubMed]
- Frahm, S.; Melis, V.; Horsley, D.; Rickard, J.E.; Riedel, G.; Fadda, P.; Scherma, M.; Harrington, C.R.; Wischik, C.M.; Theuring, F.; et al. Alpha-Synuclein transgenic mice, h-alpha-SynL62, display alpha-Syn aggregation and a dopaminergic phenotype reminiscent of Parkinson’s disease. Behav. Brain Res. 2018, 339, 153–168. [Google Scholar] [CrossRef] [PubMed]
- Brycki, B.; Koenig, H.; Pospieszny, T. Quaternary Alkylammonium Conjugates of Steroids: Synthesis, Molecular Structure, and Biological Studies. Molecules 2015, 20, 20887–20900. [Google Scholar] [CrossRef] [PubMed]
- Moore, K.S.; Wehrli, S.; Roder, H.; Rogers, M.; Forrest, J.N., Jr.; McCrimmon, D.; Zasloff, M. Squalamine: An aminosterol antibiotic from the shark. Proc. Natl. Acad. Sci. USA 1993, 90, 1354–1358. [Google Scholar] [CrossRef] [PubMed]
- Khelaifia, S.; Drancourt, M. Susceptibility of archaea to antimicrobial agents: Applications to clinical microbiology. Clin. Microbiol. Infect. 2012, 18, 841–848. [Google Scholar] [CrossRef]
- Cushnie, T.P.; Cushnie, B.; Lamb, A.J. Alkaloids: An overview of their antibacterial, antibiotic-enhancing and antivirulence activities. Int. J. Antimicrob. Agents 2014, 44, 377–386. [Google Scholar] [CrossRef]
- Schlottmann, P.G.; Alezzandrini, A.A.; Zas, M.; Rodriguez, F.J.; Luna, J.D.; Wu, L. New Treatment Modalities for Neovascular Age-Related Macular Degeneration. Asia Pac. J. Ophthalmol. 2017, 6, 514–519. [Google Scholar] [CrossRef]
- Perni, M.; Galvagnion, C.; Maltsev, A.; Meisl, G.; Muller, M.B.; Challa, P.K.; Kirkegaard, J.B.; Flagmeier, P.; Cohen, S.I.; Cascella, R.; et al. A natural product inhibits the initiation of alpha-synuclein aggregation and suppresses its toxicity. Proc. Natl. Acad. Sci. USA 2017, 114, E1009–E1017. [Google Scholar] [CrossRef]
- Yeung, T.; Gilbert, G.E.; Shi, J.; Silvius, J.; Kapus, A.; Grinstein, S. Membrane phosphatidylserine regulates surface charge and protein localization. Science 2008, 319, 210–213. [Google Scholar] [CrossRef]
- Sumioka, A.; Yan, D.; Tomita, S. TARP phosphorylation regulates synaptic AMPA receptors through lipid bilayers. Neuron 2010, 66, 755–767. [Google Scholar] [CrossRef] [PubMed]
- Alexander, R.T.; Jaumouille, V.; Yeung, T.; Furuya, W.; Peltekova, I.; Boucher, A.; Zasloff, M.; Orlowski, J.; Grinstein, S. Membrane surface charge dictates the structure and function of the epithelial Na+/H+ exchanger. EMBO J. 2011, 30, 679–691. [Google Scholar] [CrossRef] [PubMed]
- Galvagnion, C.; Buell, A.K.; Meisl, G.; Michaels, T.C.; Vendruscolo, M.; Knowles, T.P.; Dobson, C.M. Lipid vesicles trigger alpha-synuclein aggregation by stimulating primary nucleation. Nat. Chem. Biol. 2015, 11, 229–234. [Google Scholar] [CrossRef]
- Limbocker, R.; Staats, R.; Chia, S.; Ruggeri, F.S.; Mannini, B.; Xu, C.K.; Perni, M.; Cascella, R.; Bigi, A.; Sasser, L.R.; et al. Squalamine and Its Derivatives Modulate the Aggregation of Amyloid-beta and alpha-Synuclein and Suppress the Toxicity of Their Oligomers. Front. Neurosci. 2021, 15, 680026. [Google Scholar] [CrossRef] [PubMed]
- Smith, A.M.; Maguire-Nguyen, K.K.; Rando, T.A.; Zasloff, M.A.; Strange, K.B.; Yin, V.P. The protein tyrosine phosphatase 1B inhibitor MSI-1436 stimulates regeneration of heart and multiple other tissues. NPJ Regen. Med. 2017, 2, 4. [Google Scholar] [CrossRef] [PubMed]
- Perni, M.; Flagmeier, P.; Limbocker, R.; Cascella, R.; Aprile, F.A.; Galvagnion, C.; Heller, G.T.; Meisl, G.; Chen, S.W.; Kumita, J.R.; et al. Multistep Inhibition of alpha-Synuclein Aggregation and Toxicity in vitro and in vivo by Trodusquemine. ACS Chem. Biol. 2018, 13, 2308–2319. [Google Scholar] [CrossRef] [PubMed]
- Limbocker, R.; Mannini, B.; Ruggeri, F.S.; Cascella, R.; Xu, C.K.; Perni, M.; Chia, S.; Chen, S.W.; Habchi, J.; Bigi, A.; et al. Trodusquemine displaces protein misfolded oligomers from cell membranes and abrogates their cytotoxicity through a generic mechanism. Commun. Biol. 2020, 3, 435. [Google Scholar] [CrossRef] [PubMed]
- Flagmeier, P.; Meisl, G.; Vendruscolo, M.; Knowles, T.P.; Dobson, C.M.; Buell, A.K.; Galvagnion, C. Mutations associated with familial Parkinson’s disease alter the initiation and amplification steps of alpha-synuclein aggregation. Proc. Natl. Acad. Sci. USA 2016, 113, 10328–10333. [Google Scholar] [CrossRef]
- Brown, J.W.; Buell, A.K.; Michaels, T.C.; Meisl, G.; Carozza, J.; Flagmeier, P.; Vendruscolo, M.; Knowles, T.P.; Dobson, C.M.; Galvagnion, C. Beta-Synuclein suppresses both the initiation and amplification steps of alpha-synuclein aggregation via competitive binding to surfaces. Sci. Rep. 2016, 6, 36010. [Google Scholar] [CrossRef]
- Chen, S.W.; Drakulic, S.; Deas, E.; Ouberai, M.; Aprile, F.A.; Arranz, R.; Ness, S.; Roodveldt, C.; Guilliams, T.; De-Genst, E.J.; et al. Structural characterization of toxic oligomers that are kinetically trapped during alpha-synuclein fibril formation. Proc. Natl. Acad. Sci. USA 2015, 112, E1994–E2003. [Google Scholar] [CrossRef] [PubMed]
- Evangelisti, E.; Cascella, R.; Becatti, M.; Marrazza, G.; Dobson, C.M.; Chiti, F.; Stefani, M.; Cecchi, C. Binding affinity of amyloid oligomers to cellular membranes is a generic indicator of cellular dysfunction in protein misfolding diseases. Sci. Rep. 2016, 6, 32721. [Google Scholar] [CrossRef] [PubMed]
- Prabhudesai, S.; Sinha, S.; Attar, A.; Kotagiri, A.; Fitzmaurice, A.G.; Lakshmanan, R.; Ivanova, M.I.; Loo, J.A.; Klarner, F.G.; Schrader, T.; et al. A novel “molecular tweezer” inhibitor of alpha-synuclein neurotoxicity in vitro and in vivo. Neurotherapeutics 2012, 9, 464–476. [Google Scholar] [CrossRef] [PubMed]
- Acharya, S.; Safaie, B.M.; Wongkongkathep, P.; Ivanova, M.I.; Attar, A.; Klarner, F.G.; Schrader, T.; Loo, J.A.; Bitan, G.; Lapidus, L.J. Molecular basis for preventing alpha-synuclein aggregation by a molecular tweezer. J. Biol. Chem. 2014, 289, 10727–10737. [Google Scholar] [CrossRef] [PubMed]
- Rink, E.; Wullimann, M.F. The teleostean (zebrafish) dopaminergic system ascending to the subpallium (striatum) is located in the basal diencephalon (posterior tuberculum). Brain Res. 2001, 889, 316–330. [Google Scholar] [CrossRef]
- Chesselet, M.F.; Richter, F.; Zhu, C.; Magen, I.; Watson, M.B.; Subramaniam, S.R. A progressive mouse model of Parkinson’s disease: The Thy1-aSyn (“Line 61”) mice. Neurotherapeutics 2012, 9, 297–314. [Google Scholar] [CrossRef]
- Richter, F.; Subramaniam, S.R.; Magen, I.; Lee, P.; Hayes, J.; Attar, A.; Zhu, C.; Franich, N.R.; Bove, N.; De La Rosa, K.; et al. A Molecular Tweezer Ameliorates Motor Deficits in Mice Overexpressing alpha-Synuclein. Neurotherapeutics 2017, 14, 1107–1119. [Google Scholar] [CrossRef]
- Janezic, S.; Threlfell, S.; Dodson, P.D.; Dowie, M.J.; Taylor, T.N.; Potgieter, D.; Parkkinen, L.; Senior, S.L.; Anwar, S.; Ryan, B.; et al. Deficits in dopaminergic transmission precede neuron loss and dysfunction in a new Parkinson model. Proc. Natl. Acad. Sci. USA 2013, 110, E4016–E4025. [Google Scholar] [CrossRef]
- Refolo, V.; Bez, F.; Polissidis, A.; Kuzdas-Wood, D.; Sturm, E.; Kamaratou, M.; Poewe, W.; Stefanis, L.; Angela Cenci, M.; Romero-Ramos, M.; et al. Progressive striatonigral degeneration in a transgenic mouse model of multiple system atrophy: Translational implications for interventional therapies. Acta Neuropathol. Commun. 2018, 6, 2. [Google Scholar] [CrossRef]
- Bengoa-Vergniory, N.; Faggiani, E.; Ramos-Gonzalez, P.; Kirkiz, E.; Connor-Robson, N.; Brown, L.V.; Siddique, I.; Li, Z.; Vingill, S.; Cioroch, M.; et al. CLR01 protects dopaminergic neurons in vitro and in mouse models of Parkinson’s disease. Nat. Commun. 2020, 11, 4885. [Google Scholar] [CrossRef]
- Herrera-Vaquero, M.; Bouquio, D.; Kallab, M.; Biggs, K.; Nair, G.; Ochoa, J.; Heras-Garvin, A.; Heid, C.; Hadrovic, I.; Poewe, W.; et al. The molecular tweezer CLR01 reduces aggregated, pathologic, and seeding-competent alpha-synuclein in experimental multiple system atrophy. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 165513. [Google Scholar] [CrossRef] [PubMed]
- Sinha, S.; Lopes, D.H.; Du, Z.; Pang, E.S.; Shanmugam, A.; Lomakin, A.; Talbiersky, P.; Tennstaedt, A.; McDaniel, K.; Bakshi, R.; et al. Lysine-specific molecular tweezers are broad-spectrum inhibitors of assembly and toxicity of amyloid proteins. J. Am. Chem. Soc. 2011, 133, 16958–16969. [Google Scholar] [CrossRef]
- Ferreira, N.; Pereira-Henriques, A.; Attar, A.; Klarner, F.G.; Schrader, T.; Bitan, G.; Gales, L.; Saraiva, M.J.; Almeida, M.R. Molecular tweezers targeting transthyretin amyloidosis. Neurotherapeutics 2014, 11, 450–461. [Google Scholar] [CrossRef]
- Tsigelny, I.F.; Sharikov, Y.; Miller, M.A.; Masliah, E. Mechanism of alpha-synuclein oligomerization and membrane interaction: Theoretical approach to unstructured proteins studies. Nanomedicine 2008, 4, 350–357. [Google Scholar] [CrossRef] [PubMed]
- Tsigelny, I.F.; Sharikov, Y.; Wrasidlo, W.; Gonzalez, T.; Desplats, P.A.; Crews, L.; Spencer, B.; Masliah, E. Role of alpha-synuclein penetration into the membrane in the mechanisms of oligomer pore formation. FEBS J. 2012, 279, 1000–1013. [Google Scholar] [CrossRef]
- Wrasidlo, W.; Tsigelny, I.F.; Price, D.L.; Dutta, G.; Rockenstein, E.; Schwarz, T.C.; Ledolter, K.; Bonhaus, D.; Paulino, A.; Eleuteri, S.; et al. A de novo compound targeting alpha-synuclein improves deficits in models of Parkinson’s disease. Brain A J. Neurol. 2016, 139, 3217–3236. [Google Scholar] [CrossRef] [PubMed]
- Price, D.L.; Koike, M.A.; Khan, A.; Wrasidlo, W.; Rockenstein, E.; Masliah, E.; Bonhaus, D. The small molecule alpha-synuclein misfolding inhibitor, NPT200-11, produces multiple benefits in an animal model of Parkinson’s disease. Sci. Rep. 2018, 8, 16165. [Google Scholar] [CrossRef]
- Khan, A.; Johnson, R.; Wittmer, C.; Maile, M.; Tatsukawa, K.; Wong, J.L.; Gill, M.B.; Stocking, E.M.; Natala, S.R.; Paulino, A.D.; et al. NPT520-34 improves neuropathology and motor deficits in a transgenic mouse model of Parkinson’s disease. Brain A J. Neurol. 2021, 144, 3692–3709. [Google Scholar] [CrossRef]
- Maculins, T.; Garcia-Pardo, J.; Skenderovic, A.; Gebel, J.; Putyrski, M.; Vorobyov, A.; Busse, P.; Varga, G.; Kuzikov, M.; Zaliani, A.; et al. Discovery of Protein-Protein Interaction Inhibitors by Integrating Protein Engineering and Chemical Screening Platforms. Cell Chem. Biol. 2020, 27, 1441–1451.e1447. [Google Scholar] [CrossRef]
- Wagner, J.; Ryazanov, S.; Leonov, A.; Levin, J.; Shi, S.; Schmidt, F.; Prix, C.; Pan-Montojo, F.; Bertsch, U.; Mitteregger-Kretzschmar, G.; et al. Anle138b: A novel oligomer modulator for disease-modifying therapy of neurodegenerative diseases such as prion and Parkinson’s disease. Acta Neuropathol. 2013, 125, 795–813. [Google Scholar] [CrossRef]
- Kurnik, M.; Sahin, C.; Andersen, C.B.; Lorenzen, N.; Giehm, L.; Mohammad-Beigi, H.; Jessen, C.M.; Pedersen, J.S.; Christiansen, G.; Petersen, S.V.; et al. Potent alpha-Synuclein Aggregation Inhibitors, Identified by High-Throughput Screening, Mainly Target the Monomeric State. Cell Chem. Biol. 2018, 25, 1389–1402.e1389. [Google Scholar] [CrossRef]
- Hideshima, M.; Kimura, Y.; Aguirre, C.; Kakuda, K.; Takeuchi, T.; Choong, C.J.; Doi, J.; Nabekura, K.; Yamaguchi, K.; Nakajima, K.; et al. Two-step screening method to identify alpha-synuclein aggregation inhibitors for Parkinson’s disease. Sci. Rep. 2022, 12, 351. [Google Scholar] [CrossRef]
- Staats, R.; Michaels, T.C.T.; Flagmeier, P.; Chia, S.A.; 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 alpha-synuclein aggregation as a selection parameter. Commun. Chem. 2020, 3, 191. [Google Scholar] [CrossRef] [PubMed]
- Antonschmidt, L.; Matthes, D.; Dervisoglu, R.; Frieg, B.; Dienemann, C.; Leonov, A.; Nimerovsky, E.; Sant, V.; Ryazanov, S.; Giese, A.; et al. The clinical drug candidate anle138b binds in a cavity of lipidic alpha-synuclein fibrils. Nat. Commun. 2022, 13, 5385. [Google Scholar] [CrossRef]
- Levin, J.; Schmidt, F.; Boehm, C.; Prix, C.; Botzel, K.; Ryazanov, S.; Leonov, A.; Griesinger, C.; Giese, A. The oligomer modulator anle138b inhibits disease progression in a Parkinson mouse model even with treatment started after disease onset. Acta Neuropathol. 2014, 127, 779–780. [Google Scholar] [CrossRef]
- Heras-Garvin, A.; Weckbecker, D.; Ryazanov, S.; Leonov, A.; Griesinger, C.; Giese, A.; Wenning, G.K.; Stefanova, N. Anle138b modulates alpha-synuclein oligomerization and prevents motor decline and neurodegeneration in a mouse model of multiple system atrophy. Mov. Disord. Off. J. Mov. Disord. Soc. 2019, 34, 255–263. [Google Scholar] [CrossRef]
- Fellner, L.; Kuzdas-Wood, D.; Levin, J.; Ryazanov, S.; Leonov, A.; Griesinger, C.; Giese, A.; Wenning, G.K.; Stefanova, N. Anle138b Partly Ameliorates Motor Deficits Despite Failure of Neuroprotection in a Model of Advanced Multiple System Atrophy. Front. Neurosci. 2016, 10, 99. [Google Scholar] [CrossRef] [PubMed]
- Levin, J.; Sing, N.; Melbourne, S.; Morgan, A.; Mariner, C.; Spillantini, M.G.; Wegrzynowicz, M.; Dalley, J.W.; Langer, S.; Ryazanov, S.; et al. Safety, tolerability and pharmacokinetics of the oligomer modulator anle138b with exposure levels sufficient for therapeutic efficacy in a murine Parkinson model: A randomised, double-blind, placebo-controlled phase 1a trial. EBioMedicine 2022, 80, 104021. [Google Scholar] [CrossRef]
- Pujols, J.; Pena-Diaz, S.; Conde-Gimenez, M.; Pinheiro, F.; Navarro, S.; Sancho, J.; Ventura, S. High-Throughput Screening Methodology to Identify Alpha-Synuclein Aggregation Inhibitors. Int. J. Mol. Sci. 2017, 18, 478. [Google Scholar] [CrossRef] [PubMed]
- Pujols, J.; Pena-Diaz, S.; Lazaro, D.F.; Peccati, F.; Pinheiro, F.; Gonzalez, D.; Carija, A.; Navarro, S.; Conde-Gimenez, M.; Garcia, J.; et al. Small molecule inhibits alpha-synuclein aggregation, disrupts amyloid fibrils, and prevents degeneration of dopaminergic neurons. Proc. Natl. Acad. Sci. USA 2018, 115, 10481–10486. [Google Scholar] [CrossRef]
- Pena-Diaz, S.; Pujols, J.; Conde-Gimenez, M.; Carija, A.; Dalfo, E.; Garcia, J.; Navarro, S.; Pinheiro, F.; Santos, J.; Salvatella, X.; et al. ZPD-2, a Small Compound That Inhibits alpha-Synuclein Amyloid Aggregation and Its Seeded Polymerization. Front. Mol. Neurosci. 2019, 12, 306. [Google Scholar] [CrossRef] [PubMed]
- Pena-Diaz, S.; Pujols, J.; Pinheiro, F.; Santos, J.; Pallares, I.; Navarro, S.; Conde-Gimenez, M.; Garcia, J.; Salvatella, X.; Dalfo, E.; et al. Inhibition of alpha-Synuclein Aggregation and Mature Fibril Disassembling With a Minimalistic Compound, ZPDm. Front. Bioeng. Biotechnol. 2020, 8, 588947. [Google Scholar] [CrossRef] [PubMed]
- van Ham, T.J.; Thijssen, K.L.; Breitling, R.; Hofstra, R.M.; Plasterk, R.H.; Nollen, E.A.C. elegans model identifies genetic modifiers of alpha-synuclein inclusion formation during aging. PLoS Genet. 2008, 4, e1000027. [Google Scholar] [CrossRef]
- Hamamichi, S.; Rivas, R.N.; Knight, A.L.; Cao, S.; Caldwell, K.A.; Caldwell, G.A. Hypothesis-based RNAi screening identifies neuroprotective genes in a Parkinson’s disease model. Proc. Natl. Acad. Sci. USA 2008, 105, 728–733. [Google Scholar] [CrossRef]
- Harrington, A.J.; Yacoubian, T.A.; Slone, S.R.; Caldwell, K.A.; Caldwell, G.A. Functional analysis of VPS41-mediated neuroprotection in Caenorhabditis elegans and mammalian models of Parkinson’s disease. J. Neurosci. Off. J. Soc. Neurosci. 2012, 32, 2142–2153. [Google Scholar] [CrossRef]
- Pena, D.S.; Ventura, S. One ring is sufficient to inhibit alpha-synuclein aggregation. Neural Regen. Res. 2022, 17, 508–511. [Google Scholar] [CrossRef]
- Mahia, A.; Pena-Diaz, S.; Navarro, S.; Jose Galano-Frutos, J.; Pallares, I.; Pujols, J.; Diaz-de-Villegas, M.D.; Galvez, J.A.; Ventura, S.; Sancho, J. Design, synthesis and structure-activity evaluation of novel 2-pyridone-based inhibitors of alpha-synuclein aggregation with potentially improved BBB permeability. Bioorg. Chem. 2021, 117, 105472. [Google Scholar] [CrossRef] [PubMed]
- De Luca, L.; Vittorio, S.; Pena-Diaz, S.; Pitasi, G.; Fornt-Sune, M.; Bucolo, F.; Ventura, S.; Gitto, R. Ligand-Based Discovery of a Small Molecule as Inhibitor of alpha-Synuclein Amyloid Formation. Int. J. Mol. Sci. 2022, 23, 14844. [Google Scholar] [CrossRef]
- Gitto, R.; Vittorio, S.; Bucolo, F.; Pena-Diaz, S.; Siracusa, R.; Cuzzocrea, S.; Ventura, S.; Di Paola, R.; De Luca, L. Discovery of Neuroprotective Agents Based on a 5-(4-Pyridinyl)-1,2,4-triazole Scaffold. ACS Chem. Neurosci. 2022, 13, 581–586. [Google Scholar] [CrossRef]
- Braun, A.R.; Liao, E.E.; Horvath, M.; Kalra, P.; Acosta, K.; Young, M.C.; Kochen, N.N.; Lo, C.H.; Brown, R.; Evans, M.D.; et al. Potent inhibitors of toxic alpha-synuclein identified via cellular time-resolved FRET biosensors. NPJ Park. Dis. 2021, 7, 52. [Google Scholar] [CrossRef]
- Wang, Q.; Yao, S.; Yang, Z.X.; Zhou, C.; Zhang, Y.; Zhang, Y.; Zhang, L.; Li, J.T.; Xu, Z.J.; Zhu, W.L.; et al. Pharmacological characterization of the small molecule 03A10 as an inhibitor of alpha-synuclein aggregation for Parkinson’s disease treatment. Acta Pharmacol. Sin. 2023. [Google Scholar] [CrossRef] [PubMed]
- Priss, A.; Afitska, K.; Galkin, M.; Yushchenko, D.A.; Shvadchak, V.V. Rationally Designed Protein-Based Inhibitor of alpha-Synuclein Fibrillization in Cells. J. Med. Chem. 2021, 64, 6827–6837. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Hu, J.; Zhang, G.; Qin, L.; Wen, H.; Tang, Y. Pharmacophore modeling and 3D-QSAR study for the design of novel alpha-synuclein aggregation inhibitors. J. Mol. Model. 2021, 27, 260. [Google Scholar] [CrossRef]
- Sahihi, M.; Gaci, F.; Navizet, I. Identification of new alpha-synuclein fibrillogenesis inhibitor using in silico structure-based virtual screening. J. Mol. Graph. Model. 2021, 108, 108010. [Google Scholar] [CrossRef]
- Chia, S.; Faidon Brotzakis, Z.; Horne, R.I.; Possenti, A.; Mannini, B.; Cataldi, R.; Nowinska, M.; Staats, R.; Linse, S.; Knowles, T.P.J.; et al. Structure-Based Discovery of Small-Molecule Inhibitors of the Autocatalytic Proliferation of alpha-Synuclein Aggregates. Mol. Pharm. 2023, 20, 183–193. [Google Scholar] [CrossRef] [PubMed]
Gene | Role in PD |
---|---|
SNCA (α-Synuclein) | Protein aggregation Prion-like transmission Synaptic function and dopamine transmission |
GBA (Glucocerebrosidase) | Lysosome mediated autophagy pathway |
LRRK2 | Neurite structure Protein and membrane trafficking Lysosome-mediated autophagy pathway Synaptic function and dopamine transmission |
MAPT (Tau) | Protein aggregation Neurite structure |
VPS35 | Protein and membrane trafficking Lysosome-mediated autophagy pathway |
DNAJC13 (REM-8) | Protein and membrane trafficking Lysosome-mediated autophagy pathway |
GAK | Protein and membrane trafficking |
RAB7L1 | Protein and membrane trafficking |
RAB39B | Protein and membrane trafficking |
Parkin | Ubiquitin-mediated proteasome Mitochondrial dysfunction and mitophagy |
FBX07 | Ubiquitin-mediated proteasome |
SCA3 (Ataxin-3) | Ubiquitin-mediated proteasome |
PINK1 | Mitochondrial dysfunction and mitophagy |
DJ-1 | Mitochondrial dysfunction and mitophagy |
CHCHD2 | Mitochondrial dysfunction and mitophagy |
POLG1 | Mitochondrial dysfunction and mitophagy |
SREVF1 | Mitochondrial dysfunction and mitophagy |
ATP12A2 | Lysosome-mediated autophagy pathway |
SCARB2 (LIMP-2) | Lysosome-mediated autophagy pathway |
SYNJ1 (Synaptojanin 1) | Synaptic function and dopamine transmission |
GCH1 | Synaptic function and dopamine transmission |
STX1B (Syntaxin-1B) | Synaptic function and dopamine transmission |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Peña-Díaz, S.; García-Pardo, J.; Ventura, S. Development of Small Molecules Targeting α-Synuclein Aggregation: A Promising Strategy to Treat Parkinson’s Disease. Pharmaceutics 2023, 15, 839. https://doi.org/10.3390/pharmaceutics15030839
Peña-Díaz S, García-Pardo J, Ventura S. 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
Chicago/Turabian StylePeña-Díaz, Samuel, Javier García-Pardo, and Salvador Ventura. 2023. "Development of Small Molecules Targeting α-Synuclein Aggregation: A Promising Strategy to Treat Parkinson’s Disease" Pharmaceutics 15, no. 3: 839. https://doi.org/10.3390/pharmaceutics15030839
APA StylePeña-Díaz, S., García-Pardo, J., & Ventura, S. (2023). Development of Small Molecules Targeting α-Synuclein Aggregation: A Promising Strategy to Treat Parkinson’s Disease. Pharmaceutics, 15(3), 839. https://doi.org/10.3390/pharmaceutics15030839