Skip Navigation
Skip to contents

JMD : Journal of Movement Disorders

OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > J Mov Disord > Volume 9(1); 2016 > Article
Review Article
Mechanism of Anti-α-Synuclein Immunotherapy
Jun Sung Lee, Seung-Jae Lee
Journal of Movement Disorders 2016;9(1):14-19.
DOI: https://doi.org/10.14802/jmd.15059
Published online: January 25, 2016

Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea

Corresponding author: Seung-Jae Lee, PhD, Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea Tel: +82-2-3668-7037 Fax: +82-2-447-5683 E-mail: sjlee66@snu.ac.kr
• Received: November 25, 2015   • Accepted: November 30, 2015

Copyright © 2016 The Korean Movement Disorder Society

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 25,516 Views
  • 332 Download
  • 35 Web of Science
  • 35 Crossref
  • Immunization therapy targeting α-synuclein has emerged as a promising approach for Parkinson’s disease and perhaps for other synucleinopathies. Several antibodies have shown therapeutic effects in mouse models of synucleinopathies and have alleviated the pathological and behavioral phenotypes of these mice. The mechanisms through which the immunization therapy works were initially puzzling, especially given that α-synuclein is a typical cytosolic protein. Recent studies, however, suggested that extracellular α-synuclein is an important pathogenic entity, and hence, a target for immunotherapy. Here, we review the literature describing immunization therapy for synucleinopathies in mouse models and provide current thoughts on the potential mechanisms underlying the therapeutic effects of α-synuclein immunotherapy.
α-synuclein is a neuronal protein highly expressed in presynaptic terminals and is thought to be involved in the regulation of synaptic functions [1]. More attention has been placed on the pathogenic functions of this protein in a group of neurodegenerative diseases referred to as synucleinopathies. These diseases, which include Parkinson’s disease (PD), dementia with Lewy bodies, multiple system atrophy, and a large proportion of Alzheimer’s disease, are pathologically characterized by abnormal accumulation of α-synuclein aggregates. The link between α-synuclein and disease is particularly strong in PD, in which aggregated α-synuclein accumulates in structures known as Lewy bodies and Lewy neurites [2]. Several missense mutations [3-7] and gene multiplication mutations [8-11] in SNCA, the gene for α-synuclein, have been linked to familial forms of PD. Furthermore, SNCA is the gene that is most strongly and consistently associated with sporadic PD [12,13].
PD is clinically characterized by parkinsonian motor symptoms, including resting tremor, muscle tone rigidity, bradykinesia, and postural instability [14]. However, PD patients also manifest a variety of non-motor symptoms, such as autonomic dysfunctions, sensory abnormalities, psychiatric symptoms, sleep disorders, and dementia [15]. The majority of PD patients develop these symptoms sequentially as the disease progresses. Strikingly, an analysis of Lewy bodies revealed a progressive spreading of α-synuclein aggregates with disease progression, and the pattern in which the aggregates spread through the brain seemed to correlate with the clinical progression of the disease [16]. These findings strongly suggest that the spread of α-synuclein aggregates drives the disease progression, and therefore, stopping the spread of α-synuclein aggregates might halt the disease progression. Recent studies provide strong evidence that cell-to-cell propagation of α-synuclein aggregates is the underlying mechanism for the spreading of Lewy pathology [17].
Studies during the past twenty years testify to the importance of α-synuclein and its aggregation in the initiation and progression of PD, and probably other synucleinopathies, making this protein the most promising therapeutic target for these diseases. However, α-synuclein-targeting drugs have yet to be developed. In this review, we propose that immunotherapy for α-synuclein might be a promising approach for developing anti-synucleinopathy therapy and explain how this approach might work mechanistically.
In recent years, immunotherapy has emerged as a promising approach for targeting and clearing protein aggregate pathology in neurodegenerative diseases [18-22]. In a study performed ten years ago, which assessed the feasibility of PD immunotherapy, a transgenic mouse model for synucleinopathies was actively immunized with recombinant α-synuclein protein. The mice successfully generated antibodies against α-synuclein, and the behavioral deficits, α-synuclein deposition and neurodegeneration in the brains of these mice were significantly ameliorated [23]. Likewise, passive immunization with a monoclonal antibody with the epitope of the C-terminal part of α-synuclein decreased the accumulation of α-synuclein aggregates, as well as reduced the behavioral deficits in an α-synuclein transgenic mouse model [24]. Interestingly, administration of antibodies against α-synuclein oligomers reduced α-synuclein levels in both cell lysates and conditioned media [25].
Initially, the effects of immunization in the synucleinopathy models were puzzling and unexplainable, given the cytosolic nature of the target protein [26]; no rational explanation could be provided for how antibodies access α-synuclein proteins. In the following sections, we will discuss recent progress toward resolving this issue.
Secretion of α-synuclein from neuronal cells
α-synuclein is a typical cytosolic protein and is mostly present in the cytosolic fractions of brain homogenates and neuronal cell homogenates. However, a small portion of cellular α-synuclein is present in the lumen of vesicles [27], the identity of which is yet to be elucidated. These vesicular α-synuclein proteins were secreted from neuronal cells through unconventional exocytosis [28], which collectively refers to endoplasmic reticulum/Golgi-independent exocytosis. The precise mechanism of the exocytosis, however, is unknown. Recently, exosome-associated exocytosis [29] and exophagy (autophagosome-mediated exocytosis) [30] have been suggested as the mechanisms underlying α-synuclein secretion. However, the results of some studies contradict these proposals [31], and the amount of secreted α-synuclein that is associated with extracellular vesicles explains only a very small fraction of the total amount of α-synuclein secreted.
Although the mechanisms of exocytosis are unknown, we do know several conditions under which α-synuclein secretion is enhanced. These conditions, which include proteasome inhibition [28], lysosomal inhibition [32], autophagy inhibition [33], mitochondrial inhibition, oxidative modifications [34,35], and heat shock [29] which commonly affect cellular proteostasis (protein folding homeostasis). A large portion of secreted α-synuclein is oligomeric, whereas the cytosolic α-synuclein is mostly monomers [36]. From these results, we speculate that exocytosis of α-synuclein, and perhaps many other proteins that go through the same pathway, is part of the cellular response to the misfolding of the protein. More work needs to be done to resolve this problem.
Pathogenic actions of extracellular α-synuclein
After secretion from neuronal cells, α-synuclein can act on neighboring cells. Extracellular α-synuclein can be internalized into neuronal cells [37-39]. These proteins undergo endosomal trafficking [37-39] and are delivered to lysosomes where they are degraded [40]. If the internalized α-synuclein can survive the lysosomal degradation, which could result from lysosomal dysfunction, it can induce aggregation of endogenous α-synuclein proteins. Under certain conditions, this aggregate transmission coincides with neuronal cell death, both in cell cultures and in vivo [38,41,42]. However, neurodegeneration does not always occur with aggregate transmission [39,43].
Extracellular α-synuclein also acts on glial cells. α-synuclein released from neurons is transferred to astrocytes, where it induces pro-inflammatory responses [44]. Extracellular α-synuclein can also activate microglia and subsequently trigger inflammatory responses. In microglia, oligomeric forms of neuronreleased α-synuclein interact with toll-like receptor 2 (TLR2) and activate the TLR2 signaling pathway [36]. The interaction between TLR2 and α-synuclein seems to be highly conformation-selective; only certain types of oligomers interact with and activate TLR2, whereas monomers, fibrils, and some oligomer types do not. There seem to be other receptors for α-synuclein in microglia. One of these receptors might be β1-integrin, the activation of which is responsible for the morphological changes and migration of microglia [45].
Although extracellular α-synuclein accounts for only a minor portion of the total brain α-synuclein, its actions on neighboring neurons and glia suggest that it might be an excellent therapeutic target for PD and other synucleinopathies.
The mechanisms underlying the therapeutic effects of immunization against α-synuclein remain elusive. It has become increasingly clear that extracellular α-synuclein itself and the cellular events that lead to its generation and clearance are promising therapeutic targets for PD. Here, we discuss two mechanisms through which immunotherapy might work.
Clearance of extracellular α-synuclein
Extracellular α-synuclein aggregates can be internalized to neurons and glia. Among the cell types that can internalize α-synuclein aggregates, microglia exhibit the most rapid clearance of these proteins [46]. More recently, it was shown that antibody assisted in the clearance of extracellular α-synuclein, resulting in reduced neuronal and glial accumulation of α-synuclein [47]. This antibody treatment also ameliorated neurodegeneration and behavioral deficits in a mouse model of synucleinopathy. That study showed that microglia became better scavengers for extracellular α-synuclein aggregates in the presence of specific antibodies against α-synuclein. Antibody-α-synuclein immune complexes entered microglia through the Fcγ receptors, which led to efficient delivery of these immune complexes to lysosomes, hence resulting in fast degradation. By clearing extracellular α-synuclein, antibody treatment significantly reduced the extent of cell-to-cell transmission of the protein in mouse models, suggesting that this approach is effective in slowing the disease progression. Furthermore, clinical advantages might be expected if extracellular α-synuclein is selectively targeted by immunotherapy with the intraneuronal α-synuclein being left intact.
Physical blocking of extracellular α-synuclein
In addition to the accelerated clearance of extracellular α-synuclein, antibodies could directly block cell-to-cell transmission of the protein. Tran et al. [48] recently showed that several antibodies against α-synuclein physically blocked intercellular transmission of α-synuclein aggregates in cell cultures and animal models. Therefore, from the results thus far, we propose a working model in which antibodies capture extracellular α-synuclein aggregates and physically interfere with the transfer of the protein to neighboring cells, thus enhancing the efficiency of its uptake into microglia for clearance (Figure 1).
Studies so far have been encouraging in terms of targeting α-synuclein for PD immunotherapy. However, we are facing a number of potential issues or problems in developing immunotherapy for PD. First, immunotherapy should not interfere with the physiological function of α-synuclein. Generation of conformation-specific antibodies for the “pathogenic” forms of α-synuclein would allow us to overcome this possible problem. Second, delivery of antibodies to the brain parenchyma might be a problem. Engineering the antibodies so they can penetrate the blood-brain barrier might be a necessary step. Finally, the efficacy of immunotherapy needs to be validated in non-human primate models. For passive immunization, the antibodies need to be “humanized”, and thus, the toxicity and efficacy of the humanized antibodies have to be tested in non-human primates.

Conflicts of Interest

The authors have no financial conflicts of interest.

This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI14C0093).
Figure 1.
Proposed mechanisms of anti-α-synuclein immunotherapy targeting extracellular α-synuclein. A: Pathogenic roles of extracellular α-synuclein. Cellular α-synuclein is released from neuronal cells into the extracellular space. Extracellular α-synuclein aggregates can be taken up by neighboring neurons, where aggregation of the protein is transmitted. In addition, neuron-released α-synuclein can induce microglial activation and migration by stimulating TLR2 and β1-integrin, respectively. These direct (neuron-to-neuron) and indirect (microglia-mediated) functions of extracellular α-synuclein may account for the neurodegeneration observed in synucleinopathies. B: Mechanisms of anti-α-synuclein immunotherapy. Administration of antibodies targeting α-synuclein may have a neuroprotective effect by 1) blocking the direct transfer of extracellular α-synuclein into neurons and 2) facilitating Fcγ receptor-mediated internalization of extracellular α-synuclein into microglia for subsequent lysosomal degradation. Accelerated clearance would prevent the pathogenic actions of extracellular α-synuclein on neurons and microglia. TLR2: toll-like receptor 2.
jmd-15059f1.gif
  • 1. Lashuel HA, Overk CR, Oueslati A, Masliah E. The many faces of α-synuclein: from structure and toxicity to therapeutic target. Nat Rev Neurosci 2013;14:38–48.ArticlePubMedPMCPDF
  • 2. Forno LS. Neuropathology of Parkinson’s disease. J Neuropathol Exp Neurol 1996;55:259–272.ArticlePubMedPDF
  • 3. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997;276:2045–2047.ArticlePubMed
  • 4. Krüger R, Kuhn W, Müller T, Woitalla D, Graeber M, Kösel S, et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat Genet 1998;18:106–108.ArticlePubMedPDF
  • 5. Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, Ampuero I, et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol 2004;55:164–173.ArticlePubMed
  • 6. Appel-Cresswell S, Vilarino-Guell C, Encarnacion M, Sherman H, Yu I, Shah B, et al. Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson’s disease. Mov Disord 2013;28:811–813.ArticlePubMed
  • 7. Lesage S, Anheim M, Letournel F, Bousset L, Honoré A, Rozas N, et al. G51D α-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann Neurol 2013;73:459–471.ArticlePubMed
  • 8. Ibáñez P, Bonnet AM, Débarges B, Lohmann E, Tison F, Pollak P, et al. Causal relation between alpha-synuclein gene duplication and familial Parkinson’s disease. Lancet 2004;364:1169–1171.ArticlePubMed
  • 9. Ross OA, Braithwaite AT, Skipper LM, Kachergus J, Hulihan MM, Middleton FA, et al. Genomic investigation of alpha-synuclein multiplication and parkinsonism. Ann Neurol 2008;63:743–750.ArticlePubMedPMC
  • 10. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science 2003;302:841.ArticlePubMed
  • 11. Chartier-Harlin MC, Kachergus J, Roumier C, Mouroux V, Douay X, Lincoln S, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet 2004;364:1167–1169.ArticlePubMed
  • 12. Satake W, Nakabayashi Y, Mizuta I, Hirota Y, Ito C, Kubo M, et al. Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson’s disease. Nat Genet 2009;41:1303–1307.ArticlePubMed
  • 13. Simón-Sánchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat Genet 2009;41:1308–1312.ArticlePubMedPMC
  • 14. Fahn S, Sulzer D. Neurodegeneration and neuroprotection in Parkinson disease. NeuroRx 2004;1:139–154.ArticlePubMedPMC
  • 15. Ferrer I. Neuropathology and neurochemistry of nonmotor symptoms in Parkinson’s disease. Parkinsons Dis 2011;2011:708404.ArticlePubMedPMCPDF
  • 16. Braak H, Del Tredici K. Invited Article: Nervous system pathology in sporadic Parkinson disease. Neurology 2008;70:1916–1925.ArticlePubMed
  • 17. Lee HJ, Bae EJ, Lee SJ. Extracellular α--synuclein-a novel and crucial factor in Lewy body diseases. Nat Rev Neurol 2014;10:92–98.ArticlePubMedPDF
  • 18. Atwal JK, Chen Y, Chiu C, Mortensen DL, Meilandt WJ, Liu Y, et al. A therapeutic antibody targeting BACE1 inhibits amyloid-β production in vivo. Sci Transl Med 2011;3:84ra43.Article
  • 19. Delrieu J, Ousset PJ, Caillaud C, Vellas B. ‘Clinical trials in Alzheimer’s disease’: immunotherapy approaches. J Neurochem 2012;120 Suppl 1:186–193.ArticlePubMed
  • 20. Gros-Louis F, Soucy G, Larivière R, Julien JP. Intracerebroventricular infusion of monoclonal antibody or its derived Fab fragment against misfolded forms of SOD1 mutant delays mortality in a mouse model of ALS. J Neurochem 2010;113:1188–1199.ArticlePubMed
  • 21. Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, Logroscino G, Santamato A, et al. Immunotherapy for Alzheimer’s disease: from anti-β-amyloid to tau-based immunization strategies. Immunotherapy 2012;4:213–238.ArticlePubMed
  • 22. Lemere CA, Masliah E. Can Alzheimer disease be prevented by amyloid-beta immunotherapy? Nat Rev Neurol 2010;6:108–119.ArticlePubMedPMCPDF
  • 23. Masliah E, Rockenstein E, Adame A, Alford M, Crews L, Hashimoto M, et al. Effects of alpha-synuclein immunization in a mouse model of Parkinson’s disease. Neuron 2005;46:857–868.ArticlePubMed
  • 24. Masliah E, Rockenstein E, Mante M, Crews L, Spencer B, Adame A, et al. Passive immunization reduces behavioral and neuropathological deficits in an alpha-synuclein transgenic model of Lewy body disease. PLoS One 2011;6:e19338.ArticlePubMedPMC
  • 25. Näsström T, Gonçalves S, Sahlin C, Nordström E, Screpanti Sundquist V, Lannfelt L, et al. Antibodies against alpha-synuclein reduce oligomerization in living cells. PLoS One 2011;6:e27230.ArticlePubMedPMC
  • 26. Iwai A, Masliah E, Yoshimoto M, Ge N, Flanagan L, de Silva HA, et al. The precursor protein of non-A beta component of Alzheimer’s disease amyloid is a presynaptic protein of the central nervous system. Neuron 1995;14:467–475.ArticlePubMed
  • 27. Lee HJ, Patel S, Lee SJ. Intravesicular localization and exocytosis of alpha-synuclein and its aggregates. J Neurosci 2005;25:6016–6024.ArticlePubMedPMC
  • 28. Jang A, Lee HJ, Suk JE, Jung JW, Kim KP, Lee SJ. Non-classical exocytosis of alpha-synuclein is sensitive to folding states and promoted under stress conditions. J Neurochem 2010;113:1263–1274.PubMed
  • 29. Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis SD, Ntzouni M, Margaritis LH, et al. Cell-produced alphasynuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J Neurosci 2010;30:6838–6851.ArticlePubMedPMC
  • 30. Ejlerskov P, Rasmussen I, Nielsen TT, Bergström AL, Tohyama Y, Jensen PH, et al. Tubulin polymerization-promoting protein (TPPP/p25α) promotes unconventional secretion of α-synuclein through exophagy by impairing autophagosome-lysosome fusion. J Biol Chem 2013;288:17313–17335.ArticlePubMedPMC
  • 31. Grathwohl SA, Steiner JA, Britschgi M, Brundin P. Mind the gut: secretion of α-synuclein by enteric neurons. J Neurochem 2013;125:487–490.ArticlePubMed
  • 32. Bae EJ, Yang NY, Song M, Lee CS, Lee JS, Jung BC, et al. Glucocerebrosidase depletion enhances cell-to-cell transmission of α-synuclein. Nat Commun 2014;5:4755.ArticlePubMedPMCPDF
  • 33. Lee HJ, Cho ED, Lee KW, Kim JH, Cho SG, Lee SJ. Autophagic failure promotes the exocytosis and intercellular transfer of α-synuclein. Exp Mol Med 2013;45:e22. ArticlePubMedPMCPDF
  • 34. Bae EJ, Ho DH, Park E, Jung JW, Cho K, Hong JH, et al. Lipid peroxidation product 4-hydroxy-2-nonenal promotes seeding-capable oligomer formation and cell-to-cell transfer of α-synuclein. Antioxid Redox Signal 2013;18:770–783.ArticlePubMedPMC
  • 35. Lee HJ, Baek SM, Ho DH, Suk JE, Cho ED, Lee SJ. Dopamine promotes formation and secretion of non-fibrillar alpha-synuclein oligomers. Exp Mol Med 2011;43:216–222.ArticlePubMedPMC
  • 36. Kim C, Ho DH, Suk JE, You S, Michael S, Kang J, et al. Neuron-released oligomeric α-synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nat Commun 2013;4:1562.ArticlePubMedPMCPDF
  • 37. Danzer KM, Ruf WP, Putcha P, Joyner D, Hashimoto T, Glabe C, et al. Heat-shock protein 70 modulates toxic extracellular α-synuclein oligomers and rescues trans-synaptic toxicity. FASEB J 2011;25:326–336.ArticlePubMedPMC
  • 38. Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci USA 2009;106:13010–13015.ArticlePubMedPMC
  • 39. Hansen C, Angot E, Bergström AL, Steiner JA, Pieri L, Paul G, et al. α-Synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J Clin Invest 2011;121:715–725.ArticlePubMedPMC
  • 40. Lee HJ, Suk JE, Bae EJ, Lee JH, Paik SR, Lee SJ. Assembly-dependent endocytosis and clearance of extracellular alpha-synuclein. Int J Biochem Cell Biol 2008;40:1835–1849.ArticlePubMed
  • 41. Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Trojanowski JQ, et al. Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 2012;338:949–953.ArticlePubMedPMC
  • 42. Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, et al. Exogenous α-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron 2011;72:57–71.ArticlePubMedPMC
  • 43. Masuda-Suzukake M, Nonaka T, Hosokawa M, Oikawa T, Arai T, Akiyama H, et al. Prion-like spreading of pathological α-synuclein in brain. Brain 2013;136(Pt 4):1128–1138.ArticlePubMedPMCPDF
  • 44. Lee HJ, Suk JE, Patrick C, Bae EJ, Cho JH, Rho S, et al. Direct transfer of alpha-synuclein from neuron to astroglia causes inflammatory responses in synucleinopathies. J Biol Chem 2010;285:9262–9272.ArticlePubMedPMC
  • 45. Kim C, Cho ED, Kim HK, You S, Lee HJ, Hwang D, et al. β1-integrin-dependent migration of microglia in response to neuron-released α-synuclein. Exp Mol Med 2014;46:e91.ArticlePubMedPMCPDF
  • 46. Lee HJ, Suk JE, Bae EJ, Lee SJ. Clearance and deposition of extracellular alpha-synuclein aggregates in microglia. Biochem Biophys Res Commun 2008;372:423–428.ArticlePubMed
  • 47. Bae EJ, Lee HJ, Rockenstein E, Ho DH, Park EB, Yang NY, et al. Antibody-aided clearance of extracellular α-synuclein prevents cell-to-cell aggregate transmission. J Neurosci 2012;32:13454–13469.ArticlePubMedPMC
  • 48. Tran HT, Chung CH, Iba M, Zhang B, Trojanowski JQ, Luk KC, et al. Α-synuclein immunotherapy blocks uptake and templated propagation of misfolded α-synuclein and neurodegeneration. Cell Rep 2014;7:2054–2065.ArticlePubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Aggregation-Induced Emission Luminogens: A New Possibility for Efficient Visualization of RNA in Plants
      Zheng-Chao Yang, Li-Xiang Zhao, Yu-Qi Sang, Xin Huang, Xuan-Chen Lin, Zhi-Ming Yu
      Plants.2024; 13(5): 743.     CrossRef
    • Immunisation with UB-312 in the Thy1SNCA mouse prevents motor performance deficits and oligomeric α-synuclein accumulation in the brain and gut
      Jacqui T. Nimmo, Harry Smith, Chang Yi Wang, Jessica L. Teeling, James A. R. Nicoll, Ajay Verma, Jean-Cosme Dodart, Zhi Liu, Feng Lin, Roxana O. Carare
      Acta Neuropathologica.2022; 143(1): 55.     CrossRef
    • Autophagy-Related Pathways in Vesicular Unconventional Protein Secretion
      Shin Hye Noh, Ye Jin Kim, Min Goo Lee
      Frontiers in Cell and Developmental Biology.2022;[Epub]     CrossRef
    • Targeting Macroautophagy as a Therapeutic Opportunity to Treat Parkinson’s Disease
      Irene Sanchez-Mirasierra, Saurav Ghimire, Sergio Hernandez-Diaz, Sandra-Fausia Soukup
      Frontiers in Cell and Developmental Biology.2022;[Epub]     CrossRef
    • A quantitative systems pharmacology model for simulating OFF-Time in augmentation trials for Parkinson’s disease: application to preladenant
      Rachel Rose, Emma Mitchell, Piet Van Der Graaf, Daisuke Takaichi, Jun Hosogi, Hugo Geerts
      Journal of Pharmacokinetics and Pharmacodynamics.2022; 49(6): 593.     CrossRef
    • Neuroimmune crosstalk and evolving pharmacotherapies in neurodegenerative diseases
      Falguni Baidya, Mariya Bohra, Aishika Datta, Deepaneeta Sarmah, Birva Shah, Priya Jagtap, Swapnil Raut, Ankan Sarkar, Upasna Singh, Kiran Kalia, Anupom Borah, Xin Wang, Kunjan R. Dave, Dileep R. Yavagal, Pallab Bhattacharya
      Immunology.2021; 162(2): 160.     CrossRef
    • Electrogastrography for diagnosis of early-stage Parkinson's disease
      Nobuyuki Araki, Yoshitaka Yamanaka, Anupama Poudel, Yoshikatsu Fujinuma, Akira Katagiri, Satoshi Kuwabara, Masato Asahina
      Parkinsonism & Related Disorders.2021; 86: 61.     CrossRef
    • Natural Alkaloid Compounds as Inhibitors for Alpha-Synuclein Seeded Fibril Formation and Toxicity
      Simona S. Ghanem, Hend S. Fayed, Qi Zhu, Jia-Hong Lu, Nishant N. Vaikath, Janarthanan Ponraj, Said Mansour, Omar M. A. El-Agnaf
      Molecules.2021; 26(12): 3736.     CrossRef
    • Tat-p27 Ameliorates Neuronal Damage Reducing α-Synuclein and Inflammatory Responses in Motor Neurons After Spinal Cord Ischemia
      Woosuk Kim, Hyun Jung Kwon, Hyo Young Jung, Kyu Ri Hahn, Seung Myung Moon, Yeo Sung Yoon, In Koo Hwang, Soo Young Choi, Dae Won Kim
      Neurochemical Research.2021; 46(12): 3123.     CrossRef
    • Immunotherapies for Parkinson’s Disease: Progression of Clinical Development
      Jet Shee Teng, Yin Yin Ooi, Soi Moi Chye, Anna Pick Kiong Ling, Rhun Yian Koh
      CNS & Neurological Disorders - Drug Targets.2021; 20(9): 802.     CrossRef
    • Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection
      Sheila K. Pirooznia, Liana S. Rosenthal, Valina L. Dawson, Ted M. Dawson, Eric Barker
      Pharmacological Reviews.2021; 73(4): 1204.     CrossRef
    • Anti-inflammatory and Neuroprotective Agents in Clinical Trials for CNS Disease and Injury: Where Do We Go From Here?
      Khalil Mallah, Christine Couch, Davis M. Borucki, Amer Toutonji, Mohammed Alshareef, Stephen Tomlinson
      Frontiers in Immunology.2020;[Epub]     CrossRef
    • Targeting α-synuclein by PD03 AFFITOPE® and Anle138b rescues neurodegenerative pathology in a model of multiple system atrophy: clinical relevance
      Miguel Lemos, Serena Venezia, Violetta Refolo, Antonio Heras-Garvin, Sabine Schmidhuber, Armin Giese, Andrei Leonov, Sergey Ryazanov, Christian Griesinger, Gergana Galabova, Guenther Staffler, Gregor Karl Wenning, Nadia Stefanova
      Translational Neurodegeneration.2020;[Epub]     CrossRef
    • Insights Into Peptide Inhibition of Alpha-Synuclein Aggregation
      James H. Torpey, Richard M. Meade, Ravina Mistry, Jody M. Mason, Jillian Madine
      Frontiers in Neuroscience.2020;[Epub]     CrossRef
    • Novel antibodies detect additional α-synuclein pathology in synucleinopathies: potential development for immunotherapy
      Jacqui T. Nimmo, Ajay Verma, Jean-Cosme Dodart, Chang Yi Wang, Jimmy Savistchenko, Ronald Melki, Roxana O. Carare, James A. R. Nicoll
      Alzheimer's Research & Therapy.2020;[Epub]     CrossRef
    • Cellular and Molecular Aspects of Parkinson Treatment: Future Therapeutic Perspectives
      Khosro Jamebozorgi, Eskandar Taghizadeh, Daryoush Rostami, Hosein Pormasoumi, George E. Barreto, Seyed Mohammad Gheibi Hayat, Amirhossein Sahebkar
      Molecular Neurobiology.2019; 56(7): 4799.     CrossRef
    • The Associations between Immunological Reactivity to the Haptenation of Unconjugated Bisphenol A to Albumin and Protein Disulfide Isomerase with Alpha-Synuclein Antibodies
      Datis Kharrazian, Martha Herbert, Aristo Vojdani
      Toxics.2019; 7(2): 26.     CrossRef
    • Effects of single and combined immunotherapy approach targeting amyloid β protein and α‐synuclein in a dementia with Lewy bodies–like model
      Markus Mandler, Edward Rockenstein, Cassia Overk, Michael Mante, Jazmin Florio, Anthony Adame, Changyoun Kim, Radmila Santic, Achim Schneeberger, Frank Mattner, Sabine Schmidhuber, Gergana Galabova, Brian Spencer, Eliezer Masliah, Robert A. Rissman
      Alzheimer's & Dementia.2019; 15(9): 1133.     CrossRef
    • Translational therapies for multiple system atrophy: Bottlenecks and future directions
      Nadia Stefanova
      Autonomic Neuroscience.2018; 211: 7.     CrossRef
    • Integrin CD11b mediates α-synuclein-induced activation of NADPH oxidase through a Rho-dependent pathway
      Liyan Hou, Xiuqi Bao, Caixia Zang, Hanyu Yang, Fuqiang Sun, Yuning Che, Xuefei Wu, Shao Li, Dan Zhang, Qingshan Wang
      Redox Biology.2018; 14: 600.     CrossRef
    • Serum titers of autoantibodies against α-synuclein and tau in child- and adulthood
      Isabell Kuhn, Tobias Rogosch, Theresa I. Schindler, Björn Tackenberg, Michael Zemlin, Rolf F. Maier, Richard Dodel, Yannick Kronimus
      Journal of Neuroimmunology.2018; 315: 33.     CrossRef
    • Beneficial Effects of Flavonoids Against Parkinson's Disease
      Un Ju Jung, Sang Ryong Kim
      Journal of Medicinal Food.2018; 21(5): 421.     CrossRef
    • Unconventional protein secretion – new insights into the pathogenesis and therapeutic targets of human diseases
      Jiyoon Kim, Heon Yung Gee, Min Goo Lee
      Journal of Cell Science.2018;[Epub]     CrossRef
    • Region-Specific Effects of Immunotherapy With Antibodies Targeting α-synuclein in a Transgenic Model of Synucleinopathy
      Martin Kallab, Marcos Herrera-Vaquero, Malin Johannesson, Fredrik Eriksson, Jessica Sigvardson, Werner Poewe, Gregor K. Wenning, Eva Nordström, Nadia Stefanova
      Frontiers in Neuroscience.2018;[Epub]     CrossRef
    • Gene therapy for neurological disorders: progress and prospects
      Benjamin E. Deverman, Bernard M. Ravina, Krystof S. Bankiewicz, Steven M. Paul, Dinah W. Y. Sah
      Nature Reviews Drug Discovery.2018; 17(9): 641.     CrossRef
    • Combined Active Humoral and Cellular Immunization Approaches for the Treatment of Synucleinopathies
      Edward Rockenstein, Gary Ostroff, Fusun Dikengil, Florentina Rus, Michael Mante, Jazmin Florio, Anthony Adame, Ivy Trinh, Changyoun Kim, Cassia Overk, Eliezer Masliah, Robert A. Rissman
      The Journal of Neuroscience.2018; 38(4): 1000.     CrossRef
    • Holocranohistochemistry enables the visualization of α-synuclein expression in the murine olfactory system and discovery of its systemic anti-microbial effects
      Julianna J. Tomlinson, Bojan Shutinoski, Li Dong, Fanyi Meng, Dina Elleithy, Nathalie A. Lengacher, Angela P. Nguyen, Greg O. Cron, Qiubo Jiang, Erik D. Roberson, Robert L. Nussbaum, Nour K. Majbour, Omar M. El-Agnaf, Steffany A. Bennett, Diane C. Lagace,
      Journal of Neural Transmission.2017; 124(6): 721.     CrossRef
    • Structure, Distribution, and Genetic Profile of α-Synuclein and Their Potential Clinical Application in Parkinson’s Disease
      Xiaoli Si, Jiali Pu, Baorong Zhang
      Journal of Movement Disorders.2017; 10(2): 69.     CrossRef
    • Membrane Trafficking Illuminates a Path to Parkinson’s Disease
      Takafumi Hasegawa, Naoto Sugeno, Akio Kikuchi, Toru Baba, Masashi Aoki
      The Tohoku Journal of Experimental Medicine.2017; 242(1): 63.     CrossRef
    • Decelerated neurodegeneration after intravitreal injection of α-synuclein antibodies in a glaucoma animal model
      J. Teister, F. Anders, S. Beck, S. Funke, H. von Pein, V. Prokosch, N. Pfeiffer, F. Grus
      Scientific Reports.2017;[Epub]     CrossRef
    • Impact of aging immune system on neurodegeneration and potential immunotherapies
      Zhanfeng Liang, Yang Zhao, Linhui Ruan, Linnan Zhu, Kunlin Jin, Qichuan Zhuge, Dong-Ming Su, Yong Zhao
      Progress in Neurobiology.2017; 157: 2.     CrossRef
    • A novel panel of α-synuclein antibodies reveal distinctive staining profiles in synucleinopathies
      Jess-Karan S. Dhillon, Cara Riffe, Brenda D. Moore, Yong Ran, Paramita Chakrabarty, Todd E. Golde, Benoit I. Giasson, Stephan N. Witt
      PLOS ONE.2017; 12(9): e0184731.     CrossRef
    • Vaccination strategies in tauopathies and synucleinopathies
      Anne K. Braczynski, Jörg B. Schulz, Jan‐Philipp Bach
      Journal of Neurochemistry.2017; 143(5): 467.     CrossRef
    • Missions of <italic>Journal of Movement Disorders</italic>
      Yun Joong Kim
      Journal of Movement Disorders.2016; 9(1): 1.     CrossRef
    • How strong is the evidence that Parkinson's disease is a prion disorder?
      Patrik Brundin, Jiyan Ma, Jeffrey H. Kordower
      Current Opinion in Neurology.2016; 29(4): 459.     CrossRef

    Comments on this article

    Add a comment
    Figure

    JMD : Journal of Movement Disorders