1,661
Views
50
CrossRef citations to date
0
Altmetric
Mini Review

What’s to like about the prion-like hypothesis for the spreading of aggregated α-synuclein in Parkinson disease?

, &
Pages 92-97 | Received 08 Aug 2012, Accepted 28 Jan 2013, Published online: 01 Jan 2013

Abstract

α-Synuclein is a key protein in Parkinson disease. Not only is it the major protein component of Lewy bodies, but it is implicated in several cellular processes that are disrupted in Parkinson disease. Misfolded α-synuclein has also been shown to spread from cell-to-cell and, in a prion-like fashion, trigger aggregation of α-synuclein in the recipient cell. In this mini-review we explore the evidence that misfolded α-synuclein underlies the spread of pathology in Parkinson disease and discuss why it should be considered a prion-like protein.

Introduction

Parkinson disease (PD) is the second most prevalent neurodegenerative disease, and the most common synucleinopathy. Synucleinopathies feature aggregated α-synuclein (α-syn) in intracellular inclusion bodies, which are termed Lewy bodies (LB) or Lewy neurites (LN) depending on their location. They are the classical neuropathological hallmark of PD and were first described by Friedrich Lewy a century ago.Citation1 It is not clear why LB and LN form, or what impact these inclusions have on cell function. Proteomic analysis reveals they are comprised of greater than 100 different proteins,Citation2 the major protein being α-syn. Since the discoveries that α-syn was the major protein component of Lewy aggregates,Citation3 and that point mutations and genetic variation in the α-syn gene can cause rare forms of dominantly inherited PD, it has been a major focus for PD researchers. More recently, research on this little understood protein has taken an additional direction with the discovery that not only is α-syn the major protein component of LB and LN, but that intercellular exchange of the misfolded form might actually play a role in spreading α-syn pathology from cell-to-cell.

α-Syn is a 140 amino acid protein of predominantly presynaptic localization in neurons, although it is ubiquitously expressed.Citation2,Citation4 The protein is comprised of 3 domains, (1) an N-terminal lipid binding α-helix, (2) a non-amyloidß component (NAC) domain and (3) an unstructured C-terminus. All three regions are important for the misfolding of α-syn, a process critical for the induction of synucleinopathies. α-Syn is primarily a natively unfolded cytosolic protein, however via its N-terminal α-helix, it does bind to membranes, upon which it adopts an α-helical structure.Citation5 It is also on the membrane that α-syn can misfold and begin to form aggregates.Citation6 When misfolding occurs, the random coil of the NAC region forms β-sheets, leading to protofibril and fibril formation.Citation7 The C-terminus plays a role in inhibiting this fibril formation, but is also home to several phosphorylation sites, of which hyperphosphorylation at S129 (pS129) is associated with α-syn pathology.Citation8

α-syn and neurodegeneration

The link between α-syn and PD is strong with three missense mutations in the α-syn gene (PARK1/SNCA) causing autosomal dominant PD.Citation9-Citation11 Multiplications of SNCACitation12,Citation13 lead to parkinsonian symptoms and genetic variations in the non-coding regions of the gene also increase an individual’s susceptibility to PD.Citation14 α-Syn levels also increase with age,Citation15,Citation16 which correlates with the increased incidence of PD in the aged.Citation17

The direct link between α-syn pathology and PD pathology, including death of dopaminergic neurons, is not entirely clear, with some even suggesting α-syn pathology in the form of LB and LN is neuroprotective.Citation18,Citation19 Despite this, several studies have shown that misfolded α-syn has multiple detrimental effects on a number of cellular processes that could lead to neurodegeneration. Disruptions to these processes are also associated with normal aging, and also impact on the function and homeostasis of α-syn. This raises the question which is the chicken and which is the egg? Do cellular dysfunctions that have been associated with normal aging, e.g., oxidative stress, result in corruption of α-syn? Alternatively, higher cytoplasmic levels of α-syn associated with normal aging might increase the likelihood that other unknown, stochastic events that trigger α-syn misfolding take place. It is also possible that clearance of small quantities of misfolded proteins, e.g., α-syn, is impaired in aged cells leading to the seeding or large aggregates. Whatever the answer, it is clear there is a dynamic interplay between α-syn and many cellular processes and that this protein is likely to play a crucial role in PD pathogenesis.

Perhaps the concept of age-related cellular dysfunction is most important when discussing why α-syn pathology does not spread equally well to all brain regions, and it could explain why some cells are affected by α-syn pathology whereas neighboring cells are sometimes completely unaffected. Thus, cells already challenged by i.e., already high levels of oxidative stress, which has been suggested to apply to substantia nigra dopaminergic neurons, are likely to be more susceptible to a seeding mechanism following uptake of misfolded α-syn.

Oxidative stress

Oxidative stress is prevalent in the Parkinsonian brain. This in turn results in damage to lipids, proteins and DNA (especially mitochondrial DNA).Citation20 Dopaminergic neurons in particular are vulnerable to oxidative stress as dopamine itself can undergo oxidation, thus generating reactive oxygen species (ROS).Citation21 Dopamine is usually sequestered into synaptic vesicles soon after synthesis where it is protected from oxidation.Citation21 One suggested function of α-syn is in the regulation of vesicular uptake and turnover of dopamine at the synapse,Citation22 and overexpression of α-syn has been shown to cause synaptic dysfunction leading to alterations in neurotransmitter release.Citation23,Citation24 With uptake of the neurotransmitter perturbed due to non-functional α-syn, cytosolic, and thus oxidation prone, dopamine accumulates.Citation25 Dopamine metabolites have also been suggested to promote α-syn aggregationCitation26 and dopamine-α-syn adducts can stabilize potentially toxic α-syn protofibrils, thus preventing the conversion of protofibrils to fibrils.Citation27

The relationship between α-syn misfolding and oxidative stress is likely to be a self-perpetuating one; α-syn aggregates in an environment rich in ROS, which in turn promotes further ROS production. Thus, evidence suggests that α-syn aggregation promotes generation of ROS.Citation28 Specifically, oligomeric species of α-syn induce higher levels of ROS production than monomeric and fibrillar species.Citation29 Further, α-syn preferentially aggregates in the presence of both cytochrome C and hydrogen peroxide (both sources of oxidative stress) to form dimers and insoluble aggregates which are likely precursors of LB.Citation30 In line with this, LBs contain modified α-syn in the form of nitrated and oxidized species.Citation31

Mitochondrial dysfunction

The notion that PD and mitochondrial dysfunction are connected has been around for over two decades.Citation20 PD patients exhibit decreased activity of complex I of the electron transport chainCitation32 and mitochondrial DNA polymorphisms and haplotypes modulate risk for the disease.Citation33,Citation34 In addition, several genes encoding mitochondrial proteins are linked to inherited forms of PD.Citation35

In this context, it is particularly interesting that α-syn interacts directly with mitochondria and may indeed contain a mitochondrial targeting signal.Citation36 Two possible functions have been attributed to mitochondrial α-syn, i.e., regulation of mitochondrial dynamicsCitation37 and maintenance of mitochondrial calcium homeostasis.Citation38 Overexpression of α-syn leads to mitochondrial dysfunction by interfering with complex ICitation39 and fragmenting the mitochondrial network.Citation40 Misfolded α-syn also accumulates within both mitochondrial membranes,Citation36 leading to disruption of ATP synthesis and disruption of the mitochondrial membrane potential.Citation41 Mitochondrial dysfunction has also been shown to destabilize the microtubule network, in turn leading to α-syn oligomerization.Citation42

Neuroinflammation

The role of microglia and astrocytes, the resident immune cells of the brain, in PD is not clear. Both are activated not just in PD but also in several other neurodegenerative diseases. The activation of these cells, as part of a neuroinflammatory response, obviously implicates the immune system in PD but it does not clarify whether neuroinflammation is a primary event or a secondary consequence in PD pathogenesis (see reviewCitation43). In some genetic forms of PD, a primary role of the immune system seems likely. Thus, mutations in two immune-related genes DJ-1Citation44 and LRRK2Citation45 cause inherited forms of PD. In other forms of PD α-syn might promote an immune response in at least one of two ways. Either α-syn secreted into the extracellular space could stimulate microglia directly,Citation4 or by causing neuronal death, misfolded α-syn might indirectly cause microglial activation.Citation46

Overexpression of α-syn, both the normal and mutant forms, stimulates microglia, resulting in the release of pro-inflammatory cytokines, nitric acid, complements and ROS, promoting further inflammation and neurodegeneration.Citation47,Citation48 In transgenic mice, microglia activation is related spatially and temporally to α-syn overexpression.Citation49 Post-translational modifications of α-syn are also associated with activated microglia.Citation50 While the nuances of the interrelationship between α-syn and neuroinflammation are yet to be clarified, it is clear that the two are dynamically linked.

Autophagy

Under control conditions, α-syn in its native form is degraded by chaperone mediated autophagy (CMA).Citation51 However mutant forms, including post-translationally modified α-syn, block this process, leading to an increase in cytosolic α-syn inclusions.Citation52 Mutant α-syn also blocks receptors of CMA, thus preventing other CMA targets from binding.Citation53 When α-syn is overexpressed, the expression of the CMA receptor protein LAMP-2A is also increased, however if the increased levels of α-syn result in the formation of oligomers, then CMA is unable to degrade these.Citation51,Citation54 α-Syn pathology is associated with an upregulation in autophagic activity in transgenic mice expressing mutant α-syn.Citation55 With CMA blocked, α-syn can be degraded though macroautophagy (commonly referred to as autophagy). This pathway also has the potential to be detrimental to the cell resulting in autophagic cell death.Citation52 Overexpression of α-syn, however, can also impair autophagy;Citation56 although some studies indicate that enhancing autophagy can have a protective effect.Citation57

α-Syn and autophagy can also be linked to mitochondrial dysfunction. Two genes essential to mitophagy (autophagy of mitochondria), PARKIN and PINK1, are linked to PD. Overexpression of α-syn, in particular A53T mutant, results in an increase in mitophagy. In these cells there is a drastic reduction in the number and size of mitochondria, a process for which PARKIN has been found to be essential.Citation58

The prion-like hypothesis

While Braak and colleagues proposed that Lewy pathology spreads throughout the brain and this spread correlates with the disease stage,Citation59 they were uncertain what the spreading agent might be and speculated that it, for example, is a virus. It was not until autopsies were performed on a small number of PD patients who had undergone neural transplantation more than a decade before death that the prion-like hypothesis gained momentum. These particular patients had received embryonic neuronal cell transplants in the striatum. At autopsy it was discovered that the young transplanted neurons were positive for cytosolic α-syn and contained LB,Citation60,Citation61 a surprise given the young age of the neurons and that there was no reason to believe that the graft tissue donors were afflicted by PD.

The initial observation of α-syn pathology in the grafted neurons sparked a discussion of several possible explanations.Citation62 Oxidative stress, inflammation and excitotoxicity all present in the parkinsonian brain can, to varying degrees, explain the increase in α-syn levels and the presence of α-syn pathology.Citation62 It also led to speculation that the α-syn present originated in neighboring brain areas which were positive for LB pathology.Citation60 Possibly, all the factors mentioned above are responsible for the appearance of α-syn pathology in the graft, with the aged brain environment responsible for the increased cytosolic α-syn but the transfer of oligomeric α-syn seeds responsible for the formation of LB.Citation63

Subsequent studies looking at cell-to-cell transfer have shown that α-syn is capable of transfer between cells and some of the mechanisms behind this spread have been elucidated (see ReviewCitation64). Of course, for a protein to qualify as prion-like, it must also corrupt the native form of endogenous protein in the recipient cell. In the case of α-syn, this corruption is believed to occur with the transferred (misfolded) protein acting as seed, initiating the aggregation of the endogenous protein.

Transfer

Braak and colleagues proposed that the initial site of α-syn pathology is either the olfactory bulb or the enteric nervous system, or that both are involved early on.Citation65 While numerous autopsies lend weight to this hypothesis, modeling the spread under experimental conditions is only just starting to yield results. In experiments analogous to the human transplants described above, host derived α-syn has been identified in grafted neurons in the brain of rodents.Citation66,Citation67 In the case of the experiment of Angot et al.,Citation66 the transferred protein recruited α-syn in the recipient cell. Similar experiments have also been performed using aged brains from PD symptomatic mice, with pathology spreading from the site of injection throughout the brain.Citation68,Citation69 Spread of pathology through the enteric system has also been observed in A53T transgenic mice.Citation70 Human brain tissue extracts from a dementia with Lewy Body patient were injected into the gastric wall of these mice and over the course of four months, α-syn aggregates accumulated in the myenteric neurons.

Perhaps the best indication that α-syn pathology does spread will come from studies using human tissue. While the initial experiments that led to this hypothesis came from autopsy, the nature of this technique means longitudinal studies are impossible. With the suggestion that the gut may be a site where α-syn aggregation is initially triggered, it means that examining gut biopsies from people who later develop PD may be a viable tool to address this hypothesis. Indeed, α-syn aggregates were identified in colonic biopsies from PD patients that had undergone colonic biopsy 2–5 y before the onset of PD symptoms, by contrast, α-syn aggregates were absent in biopsies from control individuals who did not develop PD.Citation71 Taken together these intriguing findings suggest that α-syn pathology might start in the enteric nervous system before it spreads to the central nervous system and the substantia nigra.

The focus on intercellular protein transfer has recently been on prion-like proteins, such as α-syn, amyloid-β (Αβ), Tau, SOD-1 and prion. However, it is worthwhile to remember that other proteins that are not associated with disease can also undergo intercellular transport. Proteins can be exchanged between neighboring neurons at the synapse,Citation72 and for many years it has been known that, e.g., proteins used to trace neuronal pathways can be transported transynaptically.Citation73 The precise mechanism of transport is not known, and several possible explanations have been entertained. For example, exosomes can carry biological material, be it protein, mRNA or miRNA, over large distances. This is believed to be important for cellular communication, particularly by the immune system, although most cells studied to date secrete exosomes. Presently, somewhere in the order of 11,000 proteins have been identified in exosomes.Citation74 Another example of a possible mode of intercellular transport of proteins are tunneling nanotubes, which are able to exchange whole organelles and cytosolic proteins between cells.Citation75

Seeding

A number of groups have observed spreading of α-syn,Citation64 but observing any subsequent seeding has proved more of a challenge. The reason for this could be in the number of factors required for this seeding event to take place, with these processes difficult to model experimentally. Despite this, a number of instances have appeared in the literature recently. While some groups have seen evidence to suggest seeding is occurring, it was the use of pre-formed fibrils in vitro where the seeding effect of α-syn was perhaps best demonstrated.Citation76,Citation77 Not only did incubation of neurons with the pre-formed fibrils lead to aggregation of endogenous α-syn, but it had drastic consequences to the cell with wide spread synaptic dysfunction.Citation77 The same group showed similar results in vivo where the pre-formed fibrils were injected into the striatum and cortex of asymptomatic α-syn transgenic mice.Citation68 Ninety days post injection, α-syn pathology had spread throughout the brain, while mice injected with vehicle remained free of α-syn pathology, indicating that the presence of α-syn fibrils is enough to trigger and spread α-syn pathology. Perhaps even more significantly, when pre-formed fibrils were injected into wildtype mice, aggregated α-syn was again observed as shown previously for the transgenic mice. The key difference being in these mice that by 180 d post-injection, significant PD pathology, including dopaminergic neurodegeneration was observed correlating with the appearance of aggregated α-syn.Citation78

While the experiments performed with pre-formed fibrils were conclusive, it has been debated why it is necessary to use recombinant protein to induce these effects. The debate is similar to the Aβ and the Alzheimer field, where the outcome is highly dependent on the specific protocols used to aggregate the protein prior to intracerebral injection.Citation79 The advantage of using recombinant protein is that the seeding process can be controlled carefully in the test tube before the intracerebral injection is made, which does not apply to a crude mix such as a brain homogenate. However, as long as recombinant protein is used to initiate seeding and spread of pathology debate will remain, therefore future experiments need to address if similar results are obtained using seeds originating from a cell and not a test tube.

Why a prion-like hypothesis makes sense

Why is α-syn described as prion-like and not prion? Certainly, as has been outlined here, misfolded α-syn could be responsible for the cellular transmission of PD pathology, although the underlying mechanisms remain elusive. On the other hand, the pathobiology of prion itself is relatively poorly understood. It is not known how it causes damage to its host cell, what form of prion oligomers are toxic, nor is its native function known. What we do know is that it is capable of cell-to-cell transmission and that it is a change in conformation from the normal cellular form (PrPC) to the toxic form (PrPSc) that causes toxicity. It is assumed that PrPSc acts as a seed for PrPC aggregation, a process that has been described as akin to that of crystal formation.Citation80 While a seeding, or nucleation process has also been described for α-syn, it has yet to be studied in the same level of detail as PrPSc and further work is needed to clarify whether nucleation does indeed occur. Perhaps the most striking difference between PrPSc and α-syn however is in its transmission. While biologically it seems that α-syn in its toxic amyloid form is transmissible from one cell to another,Citation68,Citation70 PrPSc is the only protein that has been shown to be transmissible at the organism level, i.e., from one individual to another. There is currently no evidence that misfolded α-syn can be transmitted from one individual to another. This lack of transmissibility might be coupled to the fact that α-syn has not been shown to be capable of self-replication. Thus with current knowledge, it is clear that α-syn is not a prion protein.

What the experiments highlighted above suggest though is that α-syn can act as a prion-like protein and the dynamic interplay that exists between misfolded α-syn and cellular dysfunction. Of course this interplay on its own could explain neurodegeneration in the PD brain.Citation4 However, it does not fully explain the slow degeneration associated with the disease. There is little debate that PD is a disease that spreads. Pre-motor symptoms can appear decades before the on-set of motor-symptoms,Citation81 and while not every PD patient exactly fits the staging scheme described by Braak,Citation82 the degeneration observed in the majority of cases suggests a slow, progressive spread of neuropathology. The question remains, what causes this spread? And why are some neurons more vulnerable than others?

We know that transferred α-syn can recruit host α-syn, potentially depleting it from its site of normal function e.g., the synapse, which in turn can lead to a reduction in synaptic function, the consequences of which were discussed briefly above. If the transferred protein is misfolded, and consequently causes more protein to misfold, then it is possible α-syn acquires a “novel” function, that of a protein complex that binds and disrupts membranes, again causing stress to the cell. We also know that α-syn levels increase with age. As with all proteins, sometimes it misfolds. As our cells contain more and more α-syn, the proportion of misfolded α-syn will increase, causing greater burden to the cellular machinery tasked with dealing with misfolded protein.

While LBs or LNs can appear in most, if not all, cells of the brain, certain cells appear more vulnerable to degeneration than others.Citation83 The reason for this is likely to lie in the cells ability to handle toxic protein i.e., misfolded α-syn. A cell with a high energy requirement will be less able to cope with an influx of toxic protein, likewise a cell which is rich in ROS is likely to provide the right environment for further α-syn aggregation and subsequent spread of pathology. The spread of pathology is also a very slow process, suggesting that the cell can cope for a certain period of time and only when a threshold is reached does sufficient degeneration occur.

Is α-synuclein a prion-like protein?

There are three factors that have been described for α-syn that are common to other prion-like proteins. (1) α-Syn has different conformations. Under physiological conditions it is predominantly unstructured or α-helical (possibly organized in a tetramer) in structure. The pathological form of α-syn consists of oligomers and fibrils rich in β-sheets. (2) α-Syn transfers from cell-to-cell. Whether or not it transfers to a higher degree than other proteins is unknown, but it is clear that aggregated α-syn can transfer between cells. (3) Aggregated α-syn has been shown to propagate in vivo. Direct catalysis of misfolding of endogenous α-syn has not been observed in these instances, but a time dependent increase in aggregation is clear.

The trigger that leads to misfolding and aggregation of α-syn remains unknown. In a number of rare cases, mutations in the α-syn gene alone may be enough, but in the majority of cases a number of factors are likely to contribute. What is becoming clear however, whatever the trigger, misfolded α-syn is the likely bullet, spreading synuclein pathology throughout the brain.

Acknowledgments

Our work is supported by Michael J. Fox Foundation for Parkinson Research; Swedish Brain Foundation; Swedish Parkinson Foundation; Swedish Research Council, including the Linnaeus grant Bagadilico; European Research Area Network of European Funding for Neuroscience Research Program MIPROTRAN; Human Frontier Science Program; Swedish Brain Power; and a European Research Council Advanced Award. All authors are active in the Strong Research Environment Multipark (multidisciplinary research in Parkinson disease at Lund University).

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • Lewy L. Paralysis agitans. In: I. Pathologische Anatomie. Lewandowsky’s Handbuch der Neurologie, 3 Band.
  • Leverenz JB, Umar I, Wang Q, Montine TJ, McMillan PJ, Tsuang DW, et al. Proteomic identification of novel proteins in cortical lewy bodies. Brain Pathol 2007; 17:139 - 45; http://dx.doi.org/10.1111/j.1750-3639.2007.00048.x; PMID: 17388944
  • Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature 1997; 388:839 - 40; http://dx.doi.org/10.1038/42166; PMID: 9278044
  • Jellinger KA. The role of α-synuclein in neurodegeneration — An update. Transl Neurosci. 2012; 3:75 - 122; http://dx.doi.org/10.2478/s13380-012-0013-1
  • Jo E, McLaurin J, Yip CM, St George-Hyslop P, Fraser PE. alpha-Synuclein membrane interactions and lipid specificity. J Biol Chem 2000; 275:34328 - 34; http://dx.doi.org/10.1074/jbc.M004345200; PMID: 10915790
  • Dikiy I, Eliezer D. Folding and misfolding of alpha-synuclein on membranes. Biochim Biophys Acta 2012; 1818:1013 - 8; http://dx.doi.org/10.1016/j.bbamem.2011.09.008; PMID: 21945884
  • Serpell LC, Berriman J, Jakes R, Goedert M, Crowther RA. Fiber diffraction of synthetic alpha-synuclein filaments shows amyloid-like cross-beta conformation. Proc Natl Acad Sci U S A 2000; 97:4897 - 902; http://dx.doi.org/10.1073/pnas.97.9.4897; PMID: 10781096
  • Fujiwara H, Hasegawa M, Dohmae N, Kawashima A, Masliah E, Goldberg MS, et al. alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nat Cell Biol 2002; 4:160 - 4; PMID: 11813001
  • 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 - 7; http://dx.doi.org/10.1126/science.276.5321.2045; PMID: 9197268
  • 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 - 8; http://dx.doi.org/10.1038/ng0298-106; PMID: 9462735
  • 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 - 73; http://dx.doi.org/10.1002/ana.10795; PMID: 14755719
  • Chartier-Harlin M-C, 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 - 9; http://dx.doi.org/10.1016/S0140-6736(04)17103-1; PMID: 15451224
  • 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; http://dx.doi.org/10.1126/science.1090278; PMID: 14593171
  • Fuchs J, Tichopad A, Golub Y, Munz M, Schweitzer KJ, Wolf B, et al. Genetic variability in the SNCA gene influences alpha-synuclein levels in the blood and brain. FASEB J 2008; 22:1327 - 34; http://dx.doi.org/10.1096/fj.07-9348com; PMID: 18162487
  • Chu Y, Kordower JH. Age-associated increases of α-synuclein in monkeys and humans are associated with nigrostriatal dopamine depletion: Is this the target for Parkinson’s disease?. Neurobiol Dis 2007; 25:134 - 49; http://dx.doi.org/10.1016/j.nbd.2006.08.021; PMID: 17055279
  • Li W, Lesuisse C, Xu Y, Troncoso JC, Price DL, Lee MK. Stabilization of alpha-synuclein protein with aging and familial parkinson’s disease-linked A53T mutation. J Neurosci 2004; 24:7400 - 9; http://dx.doi.org/10.1523/JNEUROSCI.1370-04.2004; PMID: 15317865
  • Collier TJ, Kanaan NM, Kordower JH. Ageing as a primary risk factor for Parkinson’s disease: evidence from studies of non-human primates. Nat Rev Neurosci 2011; 12:359 - 66; http://dx.doi.org/10.1038/nrn3039; PMID: 21587290
  • Olanow CW, Perl DP, DeMartino GN, McNaught KSP. Lewy-body formation is an aggresome-related process: a hypothesis. Lancet Neurol 2004; 3:496 - 503; http://dx.doi.org/10.1016/S1474-4422(04)00827-0; PMID: 15261611
  • Tanaka M, Kim YM, Lee G, Junn E, Iwatsubo T, Mouradian MM. Aggresomes formed by alpha-synuclein and synphilin-1 are cytoprotective. J Biol Chem 2004; 279:4625 - 31; http://dx.doi.org/10.1074/jbc.M310994200; PMID: 14627698
  • Schapira AH, Jenner P. Etiology and pathogenesis of Parkinson’s disease. Mov Disord 2011; 26:1049 - 55; http://dx.doi.org/10.1002/mds.23732; PMID: 21626550
  • Lotharius J, Brundin P. Pathogenesis of Parkinson’s disease: dopamine, vesicles and α-synuclein. Nat Rev Neurosci 2002; 3:932 - 42; http://dx.doi.org/10.1038/nrn983; PMID: 12461550
  • Wersinger C, Sidhu A. Attenuation of dopamine transporter activity by alpha-synuclein. Neurosci Lett 2003; 340:189 - 92; http://dx.doi.org/10.1016/S0304-3940(03)00097-1; PMID: 12672538
  • Larsen KE, Schmitz Y, Troyer MD, Mosharov E, Dietrich P, Quazi AZ, et al. Alpha-synuclein overexpression in PC12 and chromaffin cells impairs catecholamine release by interfering with a late step in exocytosis. J Neurosci 2006; 26:11915 - 22; http://dx.doi.org/10.1523/JNEUROSCI.3821-06.2006; PMID: 17108165
  • Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK, et al. Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron 2010; 65:66 - 79; http://dx.doi.org/10.1016/j.neuron.2009.12.023; PMID: 20152114
  • Lotharius J, O’Malley KL. The parkinsonism-inducing drug 1-methyl-4-phenylpyridinium triggers intracellular dopamine oxidation. A novel mechanism of toxicity. J Biol Chem 2000; 275:38581 - 8; http://dx.doi.org/10.1074/jbc.M005385200; PMID: 10969076
  • Lee H-J, Baek SM, Ho D-H, Suk J-E, Cho E-D, Lee S-J. Dopamine promotes formation and secretion of non-fibrillar alpha-synuclein oligomers. Exp Mol Med 2011; 43:216 - 22; http://dx.doi.org/10.3858/emm.2011.43.4.026; PMID: 21415592
  • Conway KA, Rochet JC, Bieganski RM, Lansbury PT Jr.. Kinetic stabilization of the alpha-synuclein protofibril by a dopamine-alpha-synuclein adduct. Science 2001; 294:1346 - 9; http://dx.doi.org/10.1126/science.1063522; PMID: 11701929
  • Hsu LJ, Sagara Y, Arroyo A, Rockenstein E, Sisk A, Mallory M, et al. alpha-synuclein promotes mitochondrial deficit and oxidative stress. Am J Pathol 2000; 157:401 - 10; http://dx.doi.org/10.1016/S0002-9440(10)64553-1; PMID: 10934145
  • Cremades N, Cohen SIA, Deas E, Abramov AY, Chen AY, Orte A, et al. Direct observation of the interconversion of normal and toxic forms of α-synuclein. Cell 2012; 149:1048 - 59; http://dx.doi.org/10.1016/j.cell.2012.03.037; PMID: 22632969
  • Hashimoto M, Takeda A, Hsu LJ, Takenouchi T, Masliah E. Role of cytochrome c as a stimulator of alpha-synuclein aggregation in Lewy body disease. J Biol Chem 1999; 274:28849 - 52; http://dx.doi.org/10.1074/jbc.274.41.28849; PMID: 10506125
  • Giasson BI, Duda JE, Murray IV, Chen Q, Souza JM, Hurtig HI, et al. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science 2000; 290:985 - 9; http://dx.doi.org/10.1126/science.290.5493.985; PMID: 11062131
  • Schapira AH, Cooper JM, Dexter D, Clark JB, Jenner P, Marsden CD. Mitochondrial complex I deficiency in Parkinson’s disease. J Neurochem 1990; 54:823 - 7; http://dx.doi.org/10.1111/j.1471-4159.1990.tb02325.x; PMID: 2154550
  • Autere J, Moilanen JS, Finnilä S, Soininen H, Mannermaa A, Hartikainen P, et al. Mitochondrial DNA polymorphisms as risk factors for Parkinson’s disease and Parkinson’s disease dementia. Hum Genet 2004; 115:29 - 35; http://dx.doi.org/10.1007/s00439-004-1123-9; PMID: 15108120
  • Pyle A, Foltynie T, Tiangyou W, Lambert C, Keers SM, Allcock LM, et al. Mitochondrial DNA haplogroup cluster UKJT reduces the risk of PD. Ann Neurol 2005; 57:564 - 7; http://dx.doi.org/10.1002/ana.20417; PMID: 15786469
  • Valente EM, Arena G, Torosantucci L, Gelmetti V. Molecular pathways in sporadic PD. Parkinsonism Relat Disord 2012; 18:Suppl 1 S71 - 3; http://dx.doi.org/10.1016/S1353-8020(11)70023-2; PMID: 22166460
  • Devi L, Raghavendran V, Prabhu BM, Avadhani NG, Anandatheerthavarada HK. 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 - 100; http://dx.doi.org/10.1074/jbc.M710012200; PMID: 18245082
  • Xie W, Chung KKK. Alpha-synuclein impairs normal dynamics of mitochondria in cell and animal models of Parkinson’s disease. J Neurochem 2012; 122:404 - 14; http://dx.doi.org/10.1111/j.1471-4159.2012.07769.x; PMID: 22537068
  • Calì T, Ottolini D, Negro A, Brini M. α-Synuclein controls mitochondrial calcium homeostasis by enhancing endoplasmic reticulum-mitochondria interactions. J Biol Chem 2012; 287:17914 - 29; http://dx.doi.org/10.1074/jbc.M111.302794; PMID: 22453917
  • Chinta SJ, Mallajosyula JK, Rane A, Andersen JK. Mitochondrial α-synuclein accumulation impairs complex I function in dopaminergic neurons and results in increased mitophagy in vivo. Neurosci Lett 2010; 486:235 - 9; http://dx.doi.org/10.1016/j.neulet.2010.09.061; PMID: 20887775
  • Nakamura K, Nemani VM, Azarbal F, Skibinski G, Levy JM, Egami K, et al. Direct membrane association drives mitochondrial fission by the Parkinson disease-associated protein alpha-synuclein. J Biol Chem 2011; 286:20710 - 26; http://dx.doi.org/10.1074/jbc.M110.213538; PMID: 21489994
  • Kamp F, Exner N, Lutz AK, Wender N, Hegermann J, Brunner B, et al. Inhibition of mitochondrial fusion by α-synuclein is rescued by PINK1, Parkin and DJ-1. EMBO J 2010; 29:3571 - 89; http://dx.doi.org/10.1038/emboj.2010.223; PMID: 20842103
  • Esteves AR, Arduíno DM, Swerdlow RH, Oliveira CR, Cardoso SM. Oxidative stress involvement in alpha-synuclein oligomerization in Parkinson’s disease cybrids. Antioxid Redox Signal 2009; 11:439 - 48; http://dx.doi.org/10.1089/ars.2008.2247; PMID: 18717628
  • Lema Tomé CM, Tyson T, Rey NL, Grathwohl S, Britschgi M, Brundin P. Inflammation and α-Synuclein’s Prion-like Behavior in Parkinson’s Disease-Is There a Link?. Mol Neurobiol 2012; http://dx.doi.org/10.1007/s12035-012-8267-8; PMID: 22544647
  • Abou-Sleiman PM, Healy DG, Quinn N, Lees AJ, Wood NW. The role of pathogenic DJ-1 mutations in Parkinson’s disease. Ann Neurol 2003; 54:283 - 6; http://dx.doi.org/10.1002/ana.10675; PMID: 12953260
  • Greggio E, Civiero L, Bisaglia M, Bubacco L. Parkinson’s disease and immune system: is the culprit LRRKing in the periphery?. J Neuroinflammation 2012; 9:94; http://dx.doi.org/10.1186/1742-2094-9-94; PMID: 22594666
  • Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, et al. Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson’s disease. FASEB J 2005; 19:533 - 42; http://dx.doi.org/10.1096/fj.04-2751com; PMID: 15791003
  • Theodore S, Cao S, McLean PJ, Standaert DG. 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 - 58; http://dx.doi.org/10.1097/NEN.0b013e31818e5e99; PMID: 19018246
  • Rojanathammanee L, Murphy EJ, Combs CK. Expression of mutant alpha-synuclein modulates microglial phenotype in vitro. J Neuroinflammation 2011; 8:44; http://dx.doi.org/10.1186/1742-2094-8-44; PMID: 21554732
  • Watson MB, Richter F, Lee SK, Gabby L, Wu J, Masliah E, et al. Regionally-specific microglial activation in young mice over-expressing human wildtype alpha-synuclein. Exp Neurol 2012; 237:318 - 34; http://dx.doi.org/10.1016/j.expneurol.2012.06.025; PMID: 22750327
  • Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ, Lee VMY. Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. J Neurosci 2008; 28:7687 - 98; http://dx.doi.org/10.1523/JNEUROSCI.0143-07.2008; PMID: 18650345
  • Vogiatzi T, Xilouri M, Vekrellis K, Stefanis L. Wild type alpha-synuclein is degraded by chaperone-mediated autophagy and macroautophagy in neuronal cells. J Biol Chem 2008; 283:23542 - 56; http://dx.doi.org/10.1074/jbc.M801992200; PMID: 18566453
  • Lynch-Day MA, Mao K, Wang K, Zhao M, Klionsky DJ. The role of autophagy in Parkinson’s disease. Cold Spring Harb Perspect Med 2012; 2:a009357; PMID: 22474616
  • Martinez-Vicente M, Talloczy Z, Kaushik S, Massey AC, Mazzulli J, Mosharov EV, et al. Dopamine-modified alpha-synuclein blocks chaperone-mediated autophagy. J Clin Invest 2008; 118:777 - 88; PMID: 18172548
  • Xilouri M, Vogiatzi T, Vekrellis K, Park D, Stefanis L. Abberant α-synuclein confers toxicity to neurons in part through inhibition of chaperone-mediated autophagy. PLoS One 2009; 4:e5515; http://dx.doi.org/10.1371/journal.pone.0005515; PMID: 19436756
  • Yu WH, Dorado B, Figueroa HY, Wang L, Planel E, Cookson MR, et al. Metabolic activity determines efficacy of macroautophagic clearance of pathological oligomeric α-synuclein. Am J Pathol 2009; 175:736 - 47; http://dx.doi.org/10.2353/ajpath.2009.080928; PMID: 19628769
  • Winslow AR, Chen C-W, Corrochano S, Acevedo-Arozena A, Gordon DE, Peden AA, et al. α-Synuclein impairs macroautophagy: implications for Parkinson’s disease. J Cell Biol 2010; 190:1023 - 37; http://dx.doi.org/10.1083/jcb.201003122; PMID: 20855506
  • Spencer B, Potkar R, Trejo M, Rockenstein E, Patrick C, Gindi R, et al. Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in α-synuclein models of Parkinson’s and Lewy body diseases. J Neurosci 2009; 29:13578 - 88; http://dx.doi.org/10.1523/JNEUROSCI.4390-09.2009; PMID: 19864570
  • Choubey V, Safiulina D, Vaarmann A, Cagalinec M, Wareski P, Kuum M, et al. Mutant A53T alpha-synuclein induces neuronal death by increasing mitochondrial autophagy. J Biol Chem 2011; 286:10814 - 24; http://dx.doi.org/10.1074/jbc.M110.132514; PMID: 21252228
  • Braak H, Del Tredici K, Bratzke H, Hamm-Clement J, Sandmann-Keil D, Rüb U. Staging of the intracerebral inclusion body pathology associated with idiopathic Parkinson’s disease (preclinical and clinical stages). J Neurol 2002; 249:Suppl 3 III - , 1-5; http://dx.doi.org/10.1007/s00415-002-1301-4; PMID: 12528692
  • Li J-Y, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med 2008; 14:501 - 3; http://dx.doi.org/10.1038/nm1746; PMID: 18391963
  • Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson’s disease. Nat Med 2008; 14:504 - 6; http://dx.doi.org/10.1038/nm1747; PMID: 18391962
  • Brundin P, Li J-Y, Holton JL, Lindvall O, Revesz T. Research in motion: the enigma of Parkinson’s disease pathology spread. Nat Rev Neurosci 2008; 9:741 - 5; http://dx.doi.org/10.1038/nrn2477; PMID: 18769444
  • Brundin P, Kordower JH. Neuropathology in transplants in Parkinson’s disease: Implications for disease pathogenesis and the future of cell therapy. Waxman S, Stein DG, Swaab D, Fields H. Elsevier BV, 2012. 217-237.
  • Dunning CJR, Reyes JF, Steiner JA, Brundin P. Can Parkinson’s disease pathology be propagated from one neuron to another?. Prog Neurobiol 2012; 97:205 - 19; http://dx.doi.org/10.1016/j.pneurobio.2011.11.003; PMID: 22115849
  • Hawkes CH, Del Tredici K, Braak H. Parkinson’s disease: a dual-hit hypothesis. Neuropathol Appl Neurobiol 2007; 33:599 - 614; http://dx.doi.org/10.1111/j.1365-2990.2007.00874.x; PMID: 17961138
  • Angot E, Steiner JA, Lema Tomé CM, Ekström P, Mattsson B, Björklund A, et al. Alpha-synuclein cell-to-cell transfer and seeding in grafted dopaminergic neurons in vivo. PLoS One 2012; 7:e39465; http://dx.doi.org/10.1371/journal.pone.0039465; PMID: 22737239
  • Kordower JH, Dodiya HB, Kordower AM, Terpstra B, Paumier K, Madhavan L, et al. Transfer of host-derived α synuclein to grafted dopaminergic neurons in rat. Neurobiol Dis 2011; 43:552 - 7; http://dx.doi.org/10.1016/j.nbd.2011.05.001; PMID: 21600984
  • Luk KC, Kehm VM, Zhang B, O’Brien P, Trojanowski JQ, Lee VM-Y. Intracerebral inoculation of pathological α-synuclein initiates a rapidly progressive neurodegenerative α-synucleinopathy in mice. J Exp Med 2012; 209:975 - 86; http://dx.doi.org/10.1084/jem.20112457; PMID: 22508839
  • Mougenot A-L, Nicot S, Bencsik A, Morignat E, Verchère J, Lakhdar L, et al. Prion-like acceleration of a synucleinopathy in a transgenic mouse model. Neurobiol Aging 2012; 33:2225 - 8; http://dx.doi.org/10.1016/j.neurobiolaging.2011.06.022; PMID: 21813214
  • Lee H-J, Suk J-E, Lee K-W, Park S-H, Blumbergs PC, Gai W-P, et al. Transmission of Synucleinopathies in the Enteric Nervous System of A53T Alpha-Synuclein Transgenic Mice. Exp Neurobiol 2011; 20:181 - 8; http://dx.doi.org/10.5607/en.2011.20.4.181; PMID: 22355263
  • Shannon KM, Keshavarzian A, Mutlu E, Dodiya HB, Daian D, Jaglin JA, et al. Alpha-synuclein in colonic submucosa in early untreated Parkinson’s disease. Mov Disord 2012; 27:709 - 15; http://dx.doi.org/10.1002/mds.23838; PMID: 21766334
  • Chanut F. Your synapse or mine? Promiscuous protein exchanges between neighboring synapses. PLoS Biol 2006; 4:e297; http://dx.doi.org/10.1371/journal.pbio.0040297; PMID: 20076633
  • Broadwell RD, Balin BJ. Endocytic and exocytic pathways of the neuronal secretory process and trans-synaptic transfer of wheat germ agglutinin-horseradish peroxidase in vivo. J Comp Neurol 1985; 242:632 - 50; http://dx.doi.org/10.1002/cne.902420410; PMID: 2418083
  • Mathivanan S, Fahner CJ, Reid GE, Simpson RJ. ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic Acids Res 2012; 40:Database issue D1241 - 4; http://dx.doi.org/10.1093/nar/gkr828; PMID: 21989406
  • Marzo L, Gousset K, Zurzolo C. Multifaceted roles of tunneling nanotubes in intercellular communication. Front Physiol 2012; 3:72; http://dx.doi.org/10.3389/fphys.2012.00072; PMID: 22514537
  • Luk KC, Song C, O’Brien P, Stieber A, Branch JR, Brunden KR, et al. Exogenous alpha-synuclein fibrils seed the formation of Lewy body-like intracellular inclusions in cultured cells. Proc Natl Acad Sci U S A 2009; 106:20051 - 6; PMID: 19892735
  • 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; http://dx.doi.org/10.1016/j.neuron.2011.08.033; PMID: 21982369
  • Luk KC, Kehm VM, 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 - 53; http://dx.doi.org/10.1126/science.1227157; PMID: 23161999
  • Editorial. State of aggregation. Nature Publishing Group 2011; 14:399
  • Aguzzi A, Sigurdson C, Heikenwaelder M. Molecular mechanisms of prion pathogenesis. Annu Rev Pathol 2008; 3:11 - 40; http://dx.doi.org/10.1146/annurev.pathmechdis.3.121806.154326; PMID: 18233951
  • Savica R, Rocca WA, Ahlskog JE. When does Parkinson disease start?. Arch Neurol 2010; 67:798 - 801; http://dx.doi.org/10.1001/archneurol.2010.135; PMID: 20625084
  • Jellinger KA. A critical evaluation of current staging of alpha-synuclein pathology in Lewy body disorders. Biochim Biophys Acta 2009; 1792:730 - 40; http://dx.doi.org/10.1016/j.bbadis.2008.07.006; PMID: 18718530
  • Double KL, Reyes S, Werry EL, Halliday GM. Selective cell death in neurodegeneration: why are some neurons spared in vulnerable regions?. Prog Neurobiol 2010; 92:316 - 29; http://dx.doi.org/10.1016/j.pneurobio.2010.06.001; PMID: 20541584