Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1494 word(s) 1494 2021-06-21 08:50:19

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Picconi, B. Alpha-Synuclein. Encyclopedia. Available online: https://encyclopedia.pub/entry/11429 (accessed on 28 March 2024).
Picconi B. Alpha-Synuclein. Encyclopedia. Available at: https://encyclopedia.pub/entry/11429. Accessed March 28, 2024.
Picconi, Barbara. "Alpha-Synuclein" Encyclopedia, https://encyclopedia.pub/entry/11429 (accessed March 28, 2024).
Picconi, B. (2021, June 29). Alpha-Synuclein. In Encyclopedia. https://encyclopedia.pub/entry/11429
Picconi, Barbara. "Alpha-Synuclein." Encyclopedia. Web. 29 June, 2021.
Alpha-Synuclein
Edit

Alpha-Synuclein (α-syn) has strong connection with Parkinson’s disease. Parkinson’s disease (PD) is considered the most common disorder of synucleinopathy, which is characterised by intracellular inclusions of aggregated and misfolded α-syn protein in various brain regions, and the loss of dopaminergic neurons. During the early prodromal phase of PD, synaptic alterations happen before cell death, which is linked to the synaptic accumulation of toxic α-syn specifically in the presynaptic terminals, affecting neurotransmitter release. The oligomers and protofibrils of α-syn are the most toxic species, and their overexpression impairs the distribution and activation of synaptic proteins, such as the SNARE complex, preventing neurotransmitter exocytosis and neuronal synaptic communication. In the last few years, the role of the immune system in PD has been increasingly considered. Microglial and astrocyte activation, the gene expression of proinflammatory factors, and the infiltration of immune cells from the periphery to the central nervous system (CNS) represent the main features of the inflammatory response. One of the actors of these processes is α-syn accumulation.

synaptopathy α-synuclein dopamine neuroinflammation immune system

1. Introduction

The reduction of striatal dopaminergic neurons triggers motor symptoms that include bradykinesia, uncontrollable tremor at rest, postural impairment, and rigidity, which together characterise Parkinson’s disease (PD) as a movement disorder [1][2]. Neurodegeneration in the Substantia Nigra pars compacta (SNpc) leads to a marked decrease of dopamine (DA) levels in the synaptic terminals of the dorsal striatum and the consequent loss of nigrostriatal pathway, which allows PD to be described as a synaptopathy [3][4]. Synaptopathy is linked to α-synuclein (α-syn), a small, soluble protein encoded by the SNCA gene on human chromosome 4 [5][6], which is physiologically mainly localised in the presynaptic nerve terminals [7], the mitochondrial-associated membrane (MAM) [8], in which its overexpression increases the extent of contact sites and downregulates MAM activity [9][10][11][12], and in the nucleus [13][14][15][16].
α-syn accumulation compromises the fusion and clustering activity of the synaptic vesicles [17] and then influences neurotransmitter release, inducing the death of nigrostriatal neurons [18]. The transmission of α-syn pathology crosses different brain regions [19], though the impacts of extracellular α-syn on synaptic activity remains largely unknown [20][21]. Pacheco and colleagues [22] showed that extracellular α-syn oligomers facilitate the perforation of the neuronal plasma membrane, increasing its conductance and the influx of both calcium (Ca2+) and glucose, explaining in part the synaptotoxicity observed in PD. α-syn occurs in a dynamic balance between the monomeric and oligomeric forms, which are not easily prone to form fibrils under physiological conditions. Identifying the most toxic species, between fibrils and oligomers, has been difficult. There is evidence that the formation of fibrils mediates α-syn toxicity [23]. On the contrary, oligomeric forms are considered the most toxic species at the synapses [24], where they impair long term potentiation (LTP) and increase basal synaptic transmission through a mechanism dependent on N-Methyl-D-Aspartate (NMDA) receptor activation [25][26][27][28].
PD has a multifactorial aetiology. Indeed, the possible causes depend both on genetic and environmental factors that engage several biological mechanisms and processes, such as the cited α-syn misfolding, mitochondrial dysfunction, oxidative stress, synaptic plasticity, and neuroinflammation. Neuroinflammation, in recent years, has assumed a central role in the pathophysiology of PD and other neurodegenerative diseases. Evidence from post-mortem brains of PD patients, as well as in in vitro and in in vivo models, has highlighted the inflammatory contribution to the disease’s neuropathology [29][30][31][32][33]. In 1988, McGeer and collaborators disclosed the presence of HLA-DR+ microglia (macrophages) in the SN of idiopathic PD patients through immunohistochemical staining [34]. This finding proved the existence of the reactive microglia state around dead and dying dopaminergic neurons. Moreover, these macrophages exhibited phagocytic activity, as demonstrated by the presence of melanin detritus inside them [34]. This study, like others in the recent literature, suggested the involvement of immune system alteration in PD. Moreover, GWAS (genome-wide associations studies) analyses have unveiled the contribution of both innate and adaptive immune responses [35][36][37][38]. Furthermore, data support that oligomeric and fibrillary α-syn forms can activate microglial cells [39][40], suggesting a clear role for this protein in the central inflammation in PD affecting neuronal homeostasis through the modulation of microglia function, which could be either protective or detrimental in PD.

2. α-Synuclein and Its Role in Neuroinflammation in Parkinson’s Disease

As mentioned above, α-syn represents a pathological hallmark of PD, in particular intraneuronal inclusions known as LBs and/or LNs [41]. During brain physiological activity, α-syn is specially found within the presynaptic terminal of neurons belonging to the neocortex, striatum, hippocampus, thalamus, and cerebellum [42][43][44], whose cellular function has not yet been clarified, even if its involvement in synaptic plasticity and in the release of neurotransmitters and synaptic vesicles has been recognised, as extensively discussed in the previous section of this review.
The SNCA gene encodes α-syn, and can undergo missense mutations (A53T, A30T and E46K) and multiplication (duplications and triplications), which demonstrates the key role of this protein in PD [6][45][46][47][48][49][50][51]. As previously mentioned, under such pathological conditions, α-syn can be overexpressed and acquire a misfolded conformation. These misfolding species can accumulate, because of impaired autophagy or reduced phagocytic clearance [42][52][53], and could assume aggregated forms, such as oligomers or protofibrils, that cause acute toxicity in the brains of PD patients [54][55]. α-syn aggregates to resist degradation and to prompt, as shown in in vitro experiments, the impairment of macroautophagy, reducing autophasome clearance and promoting dopaminergic neuron death [56]. Moreover, post-translational modifications (ubiquitination, nitration, and phosphorylation) facilitate the formation of these aggregate species, increasing the disease process [42][53][57][58]. However, α-syn is not only a citoplasmatic protein, but can also be found in the extracellular space [59]. Neuronal cells normally throw out α-syn in the extracellular space, which impairs biological processes, such as oxidative stress or mitochondrial and lysosomal dysfunction, amplifying its release [60][61][62][63][64][65]. Thus, extracellular α-syn, probably secreted through exosomes and exocytotic vesicles [60][61][62][63][64][65], could act as damaged-associated molecular patterns (DAMPs), triggering the immune system and neuroinflammation processes [37][66]. As previously indicated, PFF aggregates could activate endogenous α-syn, triggering inflammatory and neurodegenerative responses and, in the final instance, PD pathology [27][67][68][69][70][71]. Thus, in the context of nigrostriatal degeneration in PD, misfolded α-syn species could be considered as being strictly associated with neuroinflammatory events, acting as an antigen capable of activating immune molecules.

3. Neuroinflammation as a Therapeutic Target in Parkinson’s Disease

Neuroinflammation is considered an early event in the pathophysiology of PD, or a driving mechanism of its progression. Thus, it could be an early therapeutic target to counteract PD. Many recent studies have described the use of different agents that act on inflammatory species [54][72]. In the past few years, it has been hypothesized that there is a correlation between the chronic use of non-steroidal anti-inflammatory drugs (NSAIDs) and reduced risk of developing PD [29][73]. Different meta-analyses have presented conflicting data. In particular, some studies did not find any correlation between the use of anti-inflammatory drugs and PD risk [74][75]. Other research studies showed a beneficial outcome for the use of non-aspirin NSAIDs [76][77], or aspirin in females [78]. Growing evidence supports the importance of neuroinflammation as a therapeutic target, with the aim of limiting disease progression.
Several clinical trials have been conducted in the last few years in this field (https://clinicaltrials.gov (accessed on 10 May 2021)). Immunotherapies against α-syn have been developed to reduce extracellular α-syn levels, which are responsible for triggering neuroinflammation, and its spread. There are two types of immunotherapies: active and passive [79][80].
Active immunotherapy, or vaccination, involves the production of antibodies against α-syn through the patient’s immune system. AFFITOPE® AFF1 belongs to this category, and is characterised by administration of small fragments of α-syn. This immunotherapy has successfully reduced neurodegeneration and increased anti-inflammatory factors in two animal models [72]. AFFITOPE® AFF1 is currently being considered in a phase I clinical study (NCT02267434) [72][81].
Passive immunisation involves the administration of antibodies against α-syn, and has achieved good results in pre-clinical studies by increasing the clearance of α-syn [81]. A phase 2 clinical trial that uses PRX002 as a passive immunotherapy [72][81] is currently underway.
Immunotherapies could be a valid target for therapeutic approaches, either used in combination with current medications or even as an alternative treatment in the future.

4. Conclusions

Synaptic damage and neuronal loss are major neuropathological features of PD. Misfolded α-syn aggregates are associated with disease progression; this is known as synucleinopathy. In the non-pathological brain, α-syn is not toxic and participates in several functions associated with neurotransmission and synaptic plasticity, including synaptic vesicle recycling and neurotransmitter synthesis and release. Alterations to the conformity of α-syn lead to the beginning of pathological processes in synucleinopathies. To date, many studies have identified the functions of α-syn in the regulation of neurotransmission and synaptic plasticity, but further insights are needed to outline its pathological roles. Synaptopathy, α-syn misfolding/aggregation, and neuroinflammation processes seem to interact and contribute to PD pathogenesis. As demonstrated by many of the studies cited above, these mechanisms affect each other, creating a vicious circle in which it is difficult to establish the first pathology trigger. The involvement of α-syn pathology and an altered immune response point to potential new immunomodulatory targets to slow down the progression of the disease, and/or improve its outcome.

References

  1. Obeso, J.A.; Stamelou, M.; Goetz, C.G.; Poewe, W.; Lang, A.E.; Weintraub, D.; Burn, D.; Halliday, G.M.; Bezard, E.; Przedborski, S.; et al. Past, present, and future of Parkinson’s disease: A special essay on the 200th anniversary of the shaking palsy. Mov. Disord. 2017, 32, 1264–1310.
  2. Surmeier, D.J. Determinants of dopaminergic neuron loss in Parkinson’s disease. FEBS J. 2018, 285, 3657–3668.
  3. Cheng, F.; Vivacqua, G.; Yu, S. The role of alpha-synuclein in neurotransmission and synaptic plasticity. J. Chem. Neuroanat. 2011, 42, 242–248.
  4. Picconi, B.; Hernandez, L.F.; Obeso, J.A.; Calabresi, P. Motor complications in Parkinson’s disease: Striatal molecular and electrophysiological mechanisms of dyskinesias. Mov. Disord. 2018, 33, 867–876.
  5. 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.
  6. 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.
  7. Kramer, M.L.; Schulz-Schaeffer, W.J. Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J. Neurosci. 2007, 27, 1405–1410.
  8. Guardia-Laguarta, C.; Area-Gomez, E.; Rub, C.; Liu, Y.; Magrane, J.; Becker, D.; Voos, W.; Schon, E.A.; Przedborski, S. Alpha-synuclein is localized to mitochondria-associated ER membranes. J. Neurosci. 2014, 34, 249–259.
  9. Cali, T.; Ottolini, D.; Negro, A.; Brini, M. Alpha-synuclein controls mitochondrial calcium homeostasis by enhancing endoplasmic reticulum-mitochondria interactions. J. Biol. Chem. 2012, 287, 17914–17929.
  10. Guardia-Laguarta, C.; Area-Gomez, E.; Schon, E.A.; Przedborski, S. Novel subcellular localization for alpha-synuclein: Possible functional consequences. Front. Neuroanat. 2015, 9, 17.
  11. Cali, T.; Ottolini, D.; Vicario, M.; Catoni, C.; Vallese, F.; Cieri, D.; Barazzuol, L.; Brini, M. SplitGFP technology reveals dose-dependent ER-mitochondria interface modulation by alpha-Synuclein A53T and A30P mutants. Cells 2019, 8, 1072.
  12. Hijaz, B.A.; Volpicelli-Daley, L.A. Initiation and propagation of alpha-synuclein aggregation in the nervous system. Mol. Neurodegener. 2020, 15, 19.
  13. Pinho, R.; Paiva, I.; Jercic, K.G.; Fonseca-Ornelas, L.; Gerhardt, E.; Fahlbusch, C.; Garcia-Esparcia, P.; Kerimoglu, C.; Pavlou, M.A.S.; Villar-Pique, A.; et al. Nuclear localization and phosphorylation modulate pathological effects of alpha-synuclein. Hum. Mol. Genet. 2019, 28, 31–50.
  14. Desplats, P.; Spencer, B.; Coffee, E.; Patel, P.; Michael, S.; Patrick, C.; Adame, A.; Rockenstein, E.; Masliah, E. Alpha-synuclein sequesters Dnmt1 from the nucleus: A novel mechanism for epigenetic alterations in Lewy body diseases. J. Biol. Chem. 2011, 286, 9031–9037.
  15. Savyon, M.; Engelender, S. SUMOylation in alpha-synuclein homeostasis and pathology. Front. Aging Neurosci. 2020, 12, 167.
  16. Mavroeidi, P.; Xilouri, M. Neurons and glia Interplay in α-synucleinopathies. Int. J. Mol. Sci. 2021, 22, 4994.
  17. Longhena, F.; Faustini, G.; Missale, C.; Pizzi, M.; Spano, P.; Bellucci, A. The contribution of alpha-synuclein spreading to Parkinson’s disease synaptopathy. Neural Plast. 2017, 2017, 5012129.
  18. Imbriani, P.; Schirinzi, T.; Meringolo, M.; Mercuri, N.B.; Pisani, A. Centrality of early synaptopathy in Parkinson’s disease. Front. Neurol. 2018, 9, 103.
  19. Del Tredici, K.; Braak, H. Review: Sporadic Parkinson’s disease: Development and distribution of alpha-synuclein pathology. Neuropathol. Appl. Neurobiol. 2016, 42, 33–50.
  20. Bidinosti, M.; Shimshek, D.R.; Mollenhauer, B.; Marcellin, D.; Schweizer, T.; Lotz, G.P.; Schlossmacher, M.G.; Weiss, A. Novel one-step immunoassays to quantify alpha-synuclein: Applications for biomarker development and high-throughput screening. J. Biol. Chem. 2012, 287, 33691–33705.
  21. El-Agnaf, O.M.; Salem, S.A.; Paleologou, K.E.; Curran, M.D.; Gibson, M.J.; Court, J.A.; Schlossmacher, M.G.; Allsop, D. Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006, 20, 419–425.
  22. Pacheco, C.R.; Morales, C.N.; Ramirez, A.E.; Munoz, F.J.; Gallegos, S.S.; Caviedes, P.A.; Aguayo, L.G.; Opazo, C.M. Extracellular alpha-synuclein alters synaptic transmission in brain neurons by perforating the neuronal plasma membrane. J. Neurochem. 2015, 132, 731–741.
  23. Lashuel, H.A.; Overk, C.R.; Oueslati, A.; Masliah, E. The many faces of alpha-synuclein: From structure and toxicity to therapeutic target. Nat. Rev. Neurosci. 2013, 14, 38–48.
  24. Ingelsson, M. Alpha-synuclein oligomers-neurotoxic molecules in Parkinson’s disease and other Lewy body disorders. Front. Neurosci. 2016, 10, 408.
  25. Costa, C.; Sgobio, C.; Siliquini, S.; Tozzi, A.; Tantucci, M.; Ghiglieri, V.; Di Filippo, M.; Pendolino, V.; de Iure, A.; Marti, M.; et al. Mechanisms underlying the impairment of hippocampal long-term potentiation and memory in experimental Parkinson’s disease. Brain 2012, 135, 1884–1899.
  26. Diogenes, M.J.; Dias, R.B.; Rombo, D.M.; Vicente Miranda, H.; Maiolino, F.; Guerreiro, P.; Nasstrom, T.; Franquelim, H.G.; Oliveira, L.M.; Castanho, M.A.; et al. Extracellular alpha-synuclein oligomers modulate synaptic transmission and impair LTP via NMDA-receptor activation. J. Neurosci. 2012, 32, 11750–11762.
  27. Durante, V.; de Iure, A.; Loffredo, V.; Vaikath, N.; De Risi, M.; Paciotti, S.; Quiroga-Varela, A.; Chiasserini, D.; Mellone, M.; Mazzocchetti, P.; et al. Alpha-synuclein targets GluN2A NMDA receptor subunit causing striatal synaptic dysfunction and visuospatial memory alteration. Brain 2019, 142, 1365–1385.
  28. Tozzi, A.; de Iure, A.; Bagetta, V.; Tantucci, M.; Durante, V.; Quiroga-Varela, A.; Costa, C.; Di Filippo, M.; Ghiglieri, V.; Latagliata, E.C.; et al. Alpha-synuclein produces early behavioral alterations via striatal cholinergic synaptic dysfunction by interacting with GluN2D N-methyl-D-aspartate receptor subunit. Biol. Psychiatry 2016, 79, 402–414.
  29. Gelders, G.; Baekelandt, V.; Van der Perren, A. Linking neuroinflammation and neurodegeneration in Parkinson’s disease. J. Immunol. Res. 2018, 2018, 4784268.
  30. Krashia, P.; Cordella, A.; Nobili, A.; La Barbera, L.; Federici, M.; Leuti, A.; Campanelli, F.; Natale, G.; Marino, G.; Calabrese, V.; et al. Blunting neuroinflammation with resolvin D1 prevents early pathology in a rat model of Parkinson’s disease. Nat. Commun. 2019, 10, 3945.
  31. Sanchez-Guajardo, V.; Febbraro, F.; Kirik, D.; Romero-Ramos, M. Microglia acquire distinct activation profiles depending on the degree of alpha-synuclein neuropathology in a rAAV based model of Parkinson’s disease. PLoS ONE 2010, 5, e8784.
  32. Theodore, S.; Cao, S.; McLean, P.J.; Standaert, D.G. Targeted overexpression of human alpha-synuclein triggers microglial activation and an adaptive immune response in a mouse model of Parkinson disease. J. Neuropathol. Exp. Neurol. 2008, 67, 1149–1158.
  33. Watson, M.B.; Richter, F.; Lee, S.K.; Gabby, L.; Wu, J.; Masliah, E.; Effros, R.B.; Chesselet, M.F. Regionally-specific microglial activation in young mice over-expressing human wildtype alpha-synuclein. Exp. Neurol. 2012, 237, 318–334.
  34. McGeer, P.L.; Itagaki, S.; Boyes, B.E.; McGeer, E.G. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology 1988, 38, 1285–1291.
  35. Gagliano, S.A.; Pouget, J.G.; Hardy, J.; Knight, J.; Barnes, M.R.; Ryten, M.; Weale, M.E. Genomics implicates adaptive and innate immunity in Alzheimer’s and Parkinson’s diseases. Ann. Clin. Transl. Neurol. 2016, 3, 924–933.
  36. Hamza, T.H.; Zabetian, C.P.; Tenesa, A.; Laederach, A.; Montimurro, J.; Yearout, D.; Kay, D.M.; Doheny, K.F.; Paschall, J.; Pugh, E.; et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat. Genet. 2010, 42, 781–785.
  37. Kam, T.I.; Hinkle, J.T.; Dawson, T.M.; Dawson, V.L. Microglia and astrocyte dysfunction in Parkinson’s disease. Neurobiol. Dis. 2020, 144, 105028.
  38. Pierce, S.; Coetzee, G.A. Parkinson’s disease-associated genetic variation is linked to quantitative expression of inflammatory genes. PLoS ONE 2017, 12, e0175882.
  39. Hoffmann, A.; Ettle, B.; Bruno, A.; Kulinich, A.; Hoffmann, A.C.; von Wittgenstein, J.; Winkler, J.; Xiang, W.; Schlachetzki, J.C.M. Alpha-synuclein activates BV2 microglia dependent on its aggregation state. Biochem. Biophys. Res. Commun. 2016, 479, 881–886.
  40. Park, J.Y.; Paik, S.R.; Jou, I.; Park, S.M. Microglial phagocytosis is enhanced by monomeric alpha-synuclein, not aggregated alpha-synuclein: Implications for Parkinson’s disease. Glia 2008, 56, 1215–1223.
  41. 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.
  42. Bruck, D.; Wenning, G.K.; Stefanova, N.; Fellner, L. Glia and alpha-synuclein in neurodegeneration: A complex interaction. Neurobiol. Dis. 2016, 85, 262–274.
  43. Iwai, A.; Masliah, E.; Yoshimoto, M.; Ge, N.; Flanagan, L.; de Silva, H.A.; Kittel, A.; Saitoh, T. 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.
  44. Norris, E.H.; Giasson, B.I.; Lee, V.M. Alpha-synuclein: Normal function and role in neurodegenerative diseases. Curr Top. Dev. Biol. 2004, 60, 17–54.
  45. 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.
  46. Edwards, T.L.; Scott, W.K.; Almonte, C.; Burt, A.; Powell, E.H.; Beecham, G.W.; Wang, L.; Zuchner, S.; Konidari, I.; Wang, G.; et al. Genome-wide association study confirms SNPs in SNCA and the MAPT region as common risk factors for Parkinson disease. Ann. Hum. Genet. 2010, 74, 97–109.
  47. 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.
  48. Ross, O.A.; Braithwaite, A.T.; Skipper, L.M.; Kachergus, J.; Hulihan, M.M.; Middleton, F.A.; Nishioka, K.; Fuchs, J.; Gasser, T.; Maraganore, D.M.; et al. Genomic investigation of alpha-synuclein multiplication and parkinsonism. Ann. Neurol. 2008, 63, 743–750.
  49. Schonhoff, A.M.; Williams, G.P.; Wallen, Z.D.; Standaert, D.G.; Harms, A.S. Innate and adaptive immune responses in Parkinson’s disease. Prog. Brain Res. 2020, 252, 169–216.
  50. Singleton, A.B.; Farrer, M.; Johnson, J.; Singleton, A.; Hague, S.; Kachergus, J.; Hulihan, M.; Peuralinna, T.; Dutra, A.; Nussbaum, R.; et al. Alpha-synuclein locus triplication causes Parkinson’s disease. Science 2003, 302, 841.
  51. 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.
  52. Winslow, A.R.; Chen, C.W.; Corrochano, S.; Acevedo-Arozena, A.; Gordon, D.E.; Peden, A.A.; Lichtenberg, M.; Menzies, F.M.; Ravikumar, B.; Imarisio, S.; et al. Alpha-synuclein impairs macroautophagy: Implications for Parkinson’s disease. J. Cell Biol. 2010, 190, 1023–1037.
  53. Xilouri, M.; Stefanis, L. Autophagic pathways in Parkinson disease and related disorders. Expert Rev. Mol. Med. 2011, 13, e8.
  54. Liu, C.Y.; Wang, X.; Liu, C.; Zhang, H.L. Pharmacological targeting of microglial activation: New therapeutic approach. Front. Cell Neurosci. 2019, 13, 514.
  55. Theillet, F.X.; Binolfi, A.; Bekei, B.; Martorana, A.; Rose, H.M.; Stuiver, M.; Verzini, S.; Lorenz, D.; van Rossum, M.; Goldfarb, D.; et al. Structural disorder of monomeric alpha-synuclein persists in mammalian cells. Nature 2016, 530, 45–50.
  56. Tanik, S.A.; Schultheiss, C.E.; Volpicelli-Daley, L.A.; Brunden, K.R.; Lee, V.M. Lewy body-like alpha-synuclein aggregates resist degradation and impair macroautophagy. J. Biol. Chem. 2013, 288, 15194–15210.
  57. Giasson, B.I.; Duda, J.E.; Murray, I.V.; Chen, Q.; Souza, J.M.; Hurtig, H.I.; Ischiropoulos, H.; Trojanowski, J.Q.; Lee, V.M. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science 2000, 290, 985–989.
  58. Tofaris, G.K.; Razzaq, A.; Ghetti, B.; Lilley, K.S.; Spillantini, M.G. Ubiquitination of alpha-synuclein in Lewy bodies is a pathological event not associated with impairment of proteasome function. J. Biol. Chem. 2003, 278, 44405–44411.
  59. El-Agnaf, O.M.; Salem, S.A.; Paleologou, K.E.; Cooper, L.J.; Fullwood, N.J.; Gibson, M.J.; Curran, M.D.; Court, J.A.; Mann, D.M.; Ikeda, S.; et al. Alpha-synuclein implicated in Parkinson’s disease is present in extracellular biological fluids, including human plasma. FASEB J. 2003, 17, 1945–1947.
  60. Emmanouilidou, E.; Melachroinou, K.; Roumeliotis, T.; Garbis, S.D.; Ntzouni, M.; Margaritis, L.H.; Stefanis, L.; Vekrellis, K. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J. Neurosci. 2010, 30, 6838–6851.
  61. Jang, A.; Lee, H.J.; Suk, J.E.; Jung, J.W.; Kim, K.P.; Lee, S.J. Non-classical exocytosis of alpha-synuclein is sensitive to folding states and promoted under stress conditions. J. Neurochem. 2010, 113, 1263–1274.
  62. Lee, H.J.; Bae, E.J.; Lee, S.J. Extracellular alpha-synuclein—A novel and crucial factor in Lewy body diseases. Nat. Rev. Neurol. 2014, 10, 92–98.
  63. Lee, S.J.; Lim, H.S.; Masliah, E.; Lee, H.J. Protein aggregate spreading in neurodegenerative diseases: Problems and perspectives. Neurosci. Res. 2011, 70, 339–348.
  64. Pacheco, C.; Aguayo, L.G.; Opazo, C. An extracellular mechanism that can explain the neurotoxic effects of alpha-synuclein aggregates in the brain. Front. Physiol. 2012, 3, 297.
  65. Schirinzi, T.; Madeo, G.; Martella, G.; Maltese, M.; Picconi, B.; Calabresi, P.; Pisani, A. Early synaptic dysfunction in Parkinson’s disease: Insights from animal models. Mov. Disord. 2016, 31, 802–813.
  66. Kim, C.; Ho, D.H.; Suk, J.E.; You, S.; Michael, S.; Kang, J.; Joong Lee, S.; Masliah, E.; Hwang, D.; Lee, H.J.; et al. Neuron-released oligomeric alpha-synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nat. Commun. 2013, 4, 1562.
  67. Couch, Y.; Alvarez-Erviti, L.; Sibson, N.R.; Wood, M.J.; Anthony, D.C. The acute inflammatory response to intranigral alpha-synuclein differs significantly from intranigral lipopolysaccharide and is exacerbated by peripheral inflammation. J. Neuroinflamm. 2011, 8, 166.
  68. Harms, A.S.; Delic, V.; Thome, A.D.; Bryant, N.; Liu, Z.; Chandra, S.; Jurkuvenaite, A.; West, A.B. Alpha-synuclein fibrils recruit peripheral immune cells in the rat brain prior to neurodegeneration. Acta Neuropathol. Commun. 2017, 5, 85.
  69. Masuda-Suzukake, M.; Nonaka, T.; Hosokawa, M.; Kubo, M.; Shimozawa, A.; Akiyama, H.; Hasegawa, M. Pathological alpha-synuclein propagates through neural networks. Acta Neuropathol. Commun. 2014, 2, 88.
  70. Sznejder-Pacholek, A.; Joniec-Maciejak, I.; Wawer, A.; Ciesielska, A.; Mirowska-Guzel, D. The effect of alpha-synuclein on gliosis and IL-1alpha, TNFalpha, IFNgamma, TGFbeta expression in murine brain. Pharm. Rep. 2017, 69, 242–251.
  71. Wilms, H.; Rosenstiel, P.; Romero-Ramos, M.; Arlt, A.; Schafer, H.; Seegert, D.; Kahle, P.J.; Odoy, S.; Claasen, J.H.; Holzknecht, C.; et al. Suppression of MAP kinases inhibits microglial activation and attenuates neuronal cell death induced by alpha-synuclein protofibrils. Int. J. Immunopathol. Pharm. 2009, 22, 897–909.
  72. Pajares, M.; Rojo, A.I.; Manda, G.; Bosca, L.; Cuadrado, A. Inflammation in Parkinson’s disease: Mechanisms and therapeutic implications. Cells 2020, 9, 1687.
  73. Chen, H.; Jacobs, E.; Schwarzschild, M.A.; McCullough, M.L.; Calle, E.E.; Thun, M.J.; Ascherio, A. Nonsteroidal antiinflammatory drug use and the risk for Parkinson’s disease. Ann. Neurol. 2005, 58, 963–967.
  74. Bornebroek, M.; de Lau, L.M.; Haag, M.D.; Koudstaal, P.J.; Hofman, A.; Stricker, B.H.; Breteler, M.M. Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease. Neuroepidemiology 2007, 28, 193–196.
  75. Manthripragada, A.D.; Schernhammer, E.S.; Qiu, J.; Friis, S.; Wermuth, L.; Olsen, J.H.; Ritz, B. Non-steroidal anti-inflammatory drug use and the risk of Parkinson’s disease. Neuroepidemiology 2011, 36, 155–161.
  76. Gagne, J.J.; Power, M.C. Anti-inflammatory drugs and risk of Parkinson disease: A meta-analysis. Neurology 2010, 74, 995–1002.
  77. Hernan, M.A.; Logroscino, G.; Garcia Rodriguez, L.A. Nonsteroidal anti-inflammatory drugs and the incidence of Parkinson disease. Neurology 2006, 66, 1097–1099.
  78. Wahner, A.D.; Bronstein, J.M.; Bordelon, Y.M.; Ritz, B. Nonsteroidal anti-inflammatory drugs may protect against Parkinson disease. Neurology 2007, 69, 1836–1842.
  79. Sardi, S.P.; Cedarbaum, J.M.; Brundin, P. Targeted therapies for Parkinson’s disease: From genetics to the clinic. Mov. Disord. 2018, 33, 684–696.
  80. Zella, M.A.S.; Metzdorf, J.; Ostendorf, F.; Maass, F.; Muhlack, S.; Gold, R.; Haghikia, A.; Tonges, L. Novel immunotherapeutic approaches to target alpha-synuclein and related neuroinflammation in Parkinson’s disease. Cells 2019, 8, 105.
  81. Antonini, A.; Bravi, D.; Sandre, M.; Bubacco, L. Immunization therapies for Parkinson’s disease: State of the art and considerations for future clinical trials. Expert Opin. Investig. Drugs 2020, 29, 685–695.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 586
Revision: 1 time (View History)
Update Date: 29 Jun 2021
1000/1000