Abstract
Protein inclusions made up primarily of misfolded α-synuclein (α-Syn) are the hallmark of a set of disorders known as synucleionopathies, most notably Parkinson’s disease (PD). It is becoming increasingly appreciated that α-Syn misfolding can spread to anatomically connected regions in a prion-like manner. The protein aggregates that ensue are correlated with neurodegeneration in the various yet select neuronal populations that are affected. Recent advances have begun to shed light on the spreading and toxicity mechanisms that may be occurring in PD. Several key emerging themes are arising from this work suggesting that α-Syn mediated neurodegeneration is due to a combination of relative α-Syn expression level, connectivity to affected brain regions, and intrinsic vulnerability to pathology.
Keywords: alpha-synuclein, Parkinson’s disease, protein misfolding, neurodegeneration, selective vulnerability
1. Introduction
Parkinson’s disease (PD) is the most common motor disorder and second most common neurodegenerative disorder after Alzheimer’s disease, affecting an estimated 7 million people worldwide [1, 2]. As age is a significant risk factor, PD prevalence is expected to rise sharply as the average life expectancy among the general population continues to increase [3] Despite its immense social and economic impact, our knowledge regarding the etiology of this condition remains incomplete and no effective treatments which modify the course of disease are presently available.
The most prominent feature in the brains of PD patients is the selective loss of dopaminergic (DA) pigmented neurons within the substantia nigra (SN) and to a lesser extent those residing in the ventral tegmental and retrorubral areas. Significant degeneration is also observed in other nuclei and neurotransmitter systems including the dorsal raphe (serotonergic), locus coeruleus (noradrenergic), nucleus basalis of Meynert (cholinergic) and the dorsal motor nucleus of the vagus nerve [4]. Degeneration in these areas likely contributes to both motor and non-motor symptoms observed in PD, especially hallucinations, depression, and sleep disorders.
For over a century, it has been recognized that neurodegenerative diseases are commonly associated with the accumulation of abnormally folded proteins within or in the vicinity of cells of the central nervous system (CNS) [5]. PD is characterized by the presence of eosinophilic inclusions in the soma of neurons termed Lewy bodies (or Lewy neurites when present in the neurites). Lewy bodies (LBs) represent a complex amalgamation of lipids, neuromelanin, and up to several hundred individual proteins with a key component being alpha-synuclein (α-Syn), an 140 amino-acid protein that is enriched in presynaptic vesicles of vertebrates [6–8]. A highly soluble lipid-binding protein normally thought to regulate synaptic vesicle release through stabilization of SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complexes, α-Syn in Lewy pathology occurs as highly ordered amyloid-type fibrils [6, 9, 10]. In addition to abnormal conformations, α-Syn in PD brains is also commonly post-translationally modified by cleavage, hyperphosphorylation, ubiquitination, nitrosylation, and oxidation [11–16].
Several lines of evidence point to α-Syn playing a key, and possibly causal, role in PD. For example, the genetic studies that led to the discovery of α-Syn as a major component of LBs also revealed several point mutations which result in autosomal dominant PD [17–21]. Duplication or triplication of the wildtype locus that lead to 1.5–2 fold overexpression also give rise to dominantly-inherited PD, while several polymorphisms in the promoter region are reported to elevate the risk of developing disease [22–24].
The significance of α-Syn is further supported by large-scale genome-wide association studies that indicate a clear link between the SNCA locus and PD [25, 26]. Intriguingly, several strong genetic risk factors may have strong interactions with α-Syn. For example, mutations in glucocerebrosidase, which causes the lysosomal storage disorder Gaucher’s disease, also elevates α-Syn levels and increases the risk of PD by up to 30-fold in some populations [27, 28]. Moreover, Lewy pathology has also been reported in multiple forms of familial PD, such as those involving mutations in DJ-1, PINK1, and GBA, although this is less certain for other genetic forms of PD (e.g., LRRK2, Parkin, ATP13A2) [29–34]. Collectively, these data point to α-Syn dysfunction as a key process in PD pathogenesis. Indeed, the observation that cytoplasmic α-Syn inclusions are also characteristic of several other neurodegenerative conditions, most notably multiple system atrophy (MSA) and dementia with Lewy bodies, suggests that α-Syn accumulation is not merely a benign product of the degenerative process [5, 6, 35, 36].
Despite the overwhelming evidence for a central role of α-Syn in PD and other synucleinopathies and the explosion in our knowledge regarding this protein in both health and disease, two fundamental questions still remain unanswered. Firstly, what instigates the neurodegeneration that typifies PD? Secondly, why do selective cell populations undergo degeneration? This review discusses some of the recent advances that illustrate the complex pathobiological relationships between α-Syn misfolding and PD.
2. Prion-like properties of pathologic α-Syn
The distribution of α-Syn inclusions observed in the CNS of patients with PD and other synucleinopathies is highly non-uniform. On the contrary, Braak and colleagues have previously demonstrated that α-Syn inclusions in PD form in a predictable manner during disease progression, allowing categorization into at least six distinct clinicopathological stages in the majority of PD patients. Lewy pathology typically begins in the olfactory bulb and deep brain stem nuclei (stages I and II). This pattern correlates with several known prodromal symptoms of PD including olfactory impairment, autonomic dysfunction, and REM sleep disturbances [37]. Inclusion pathology is then detectable in more rostral regions, most notably midbrain DA neurons in stages III and IV. It is during these stages that the motor symptoms associated with PD become evident. In the ultimate stages (V and VI) pathology begins to be observed in the neocortex and has been correlated with the onset of dementia [38, 39]. While this stereotypic staging system appears to be valid in up to 80% in multiple patient subpopulations, other studies point to a significant portion of patients (up to 50%) that do not follow this classification [40–43].Heterogeneity in the initial location where α-Syn aggregates are first detected appears to be a source of this discrepancy resulting in modified or alternative staging systems that attempt to encompass populations that do not fit the conventional caudo-rostral pattern of spread, such as in dementia with LB [44]. Nonetheless, human studies clearly point to the gradual yet inevitable spread of α-Syn pathology originating from select regions. In conjunction with this evidence, post-mortem examination of fetal mesencephalic neurons transplanted into PD patients show that they develop Lewy pathology in a time-dependent manner. As fetal neurons initially lack any of the pathological processes and were unlikely to contain α-Syn inclusions at the time of transplantation, the appearance of pathology several years in a neurodegenerative environment argues that the initiation factor of pathology had been conferred and affected healthy neurons [45, 46].
In line with this histopathological evidence, more recent work in cell culture and animals models has provided direct indication that α-Syn pathology is transmissible to recipient cells. In particular, it is now clear that brain homogenates enriched in α-Syn pathology can induce Lewy pathology in the CNS of recipient animals. Importantly, the source of this pathology, whether from transgenic mice exhibiting Lewy-like pathology or from MSA or PD patient-derived extracts are capable of initiating pathological formation [47–50]. Indeed, α-Syn itself can propagate pathology as recombinant α-Syn fibrils (or pre-formed fibrils; PFFs) and induce Lewy-like pathology in cultured cells and neurons, as well as in vivo following intracerebral introduction [51–56]. Not only does α-Syn pathology form in these models, but aggregation develops at sites at considerable distances from the injection site in vivo indicating propagation of the misfolding process [53–55].
Interestingly, intramuscular PFF injection also results in CNS pathology in transgenic mice overexpressing α-Syn, suggesting that pathology can transmit via the PNS, although the mechanism responsible remains unclear [57]. Analogous to this, the preponderance of Lewy pathology in deep brain stem nuclei, especially the dorsal motor nucleus of the vagus, seen in early PD is consistent with a spreading process originating in peripheral sites [39]. One location commonly found to harbor α-Syn aggregates in PD patients are gastrointestinal tract neurons, and a recent report suggests that long term risk of PD is significantly reduced following full vagotomy, raising the interesting possibility of this system as potential route of spread [58, 59].
The potential mechanisms by which misfolded α-Syn can initiate and propagate intracellular inclusion formation that is reminiscent of human synucleinopathies have been reviewed in detail elsewhere [5, 60–62] and appears to be predicated on three major components: the generation of misfolded α-Syn species, internalization into a permissive cellular environment/compartment, and engagement with the endogenously expressed α-Syn pool.
2.1 Internalization of extracellular α-Syn
Among neurons, endocytosis appears to be a chief mechanism mediating misfolded α-Syn entry into neurons, although the precise steps and whether other internalization pathways play a role is an active area of research [52, 63]. Blocking endocytosis with a dynamin dominant negative mutant prevents cell to cell transfer of pathology [64]. It is not clear if this is mediated by a selective uptake mechanism, e.g. a specific receptor that binds α-Syn, although α-Syn aggregates appear to have an affinity for heparan sulfate proteoglycans on neuronal cell surface like tau fibrils [65]. In line with these properties, α-Syn internalization has also been described in multiple cell types, and evidence suggests that microglia, astrocytes and oligodendrocytes are capable to internalizing α-Syn [66–70]. Several commonly used cell lines have also been shown to internalize α-Syn [51, 71–73]. This provides evidence as to how glia cytoplasmic inclusions (GCIs) could form in MSA, but it is unusual that these inclusions would be able to form because neither cell type expresses much α-Syn [66, 68]. Another potential mechanism that could spread pathology are nanotunnels between cells which have been shown to be capable of spreading prion protein, but this has yet to be reported for α-Syn [74].
The manner in which α-Syn is released from cells remains unknown as well. α-Syn is found in vesicles released by exocytosis and in several body fluids, such as CSF and plasma [75, 76]. α-Syn secretion continues when ER-Golgi transport is blocked, ruling out conventional exocytosis as the primary mechanism of α-Syn release [75, 77]. Other non-conventional forms of exocytosis are being explored as the mediator of α-Syn spread including exosome release and exophagy, an autophagosome mediated exocytosis [78–81].
2.2 Multiple neuronal populations are permissive to pathological α-Syn propagation
Mice, rats as well as non-human primates have been found to develop pathology and nigrostriatal neurodegeneration after injection of PD patient brain lysate into the striatum further illustrating that pathology and DA neuron toxicity are transmissible [48, 56]. Targeting PFFs to separate CNS nuclei in WT mice also gives rise to distinct spreading patterns for each injection site [82]. Thus, it is clear that multiple cell populations in the CNS are permissive to seeding with exogenous misfolded α-Syn species. This data also indicates that pathology spreads in an anatomically dependent manner. Furthermore,in vivo data suggests that pathology spreads in a retrograde manner to synaptically connected brain regions, and this is unlikely to be mediated by cell death [53, 83, 84]. Pathology has also been observed to be trafficked in vitro both towards and away from the soma [85, 86]. The terminology of retrograde vs. anterograde should be used cautiously as pathology propagation mechanisms are ill-defined and may not conform to traditionally defined modes of axonal transport. Moreover, it is possible that different species of α-Syn (e.g., internalized misfolded vs. converted endogenous species) may undergo different modes of trafficking/transport (i.e., propagation may be a separate but simultaneous event), although pathology is capable of propagating both towards and away from the soma in cultured neurons [61].
The range of conformational species capable of inducing pathology remains a topic of debate. Indeed, aggregates generated under different conditions or isolated from different synucleinopathies have been reported to induce distinct phenotypes, leading to the notion that multiple strains exist. When α-Syn is fibrilized in vitro under stringent conditions, distinct α-Syn fibrils with different biochemical and biophysical properties can be generated. These different fibril strains are able to cause different phenotypes in vivo as well [67, 87]. In addition, α-Syn fibrils formed after serial passaging have been found to gain novel properties and co-aggregate other aggregation prone proteins such as tau, which is implicated in Alzheimer’s and other disorders [88]. This raises the possibility that PD and other synucleinopathies may be caused by different strains of fibrillized α-Syn in humans, although this is not yet definitive.
3. Toxicity mediated by α-Syn misfolding
It is unclear if LB formation precedes cell loss, but the appearance of α-syn aggregates within a neuronal population is highly correlated with the onset of neuronal loss [38, 89, 90]. In PD patients, cell loss in the SNis also highly correlated to worsening of symptoms, and disease progression is associated with changes in network activity in several brain regions including the subthalamic nucleus and globus pallidus [91, 92]. However, the degree to which these two variables correlate and their causality is still contentious.
The sequence of events is somewhat clearer in experimental models and neurons treated with α-Syn PFFs is that PFF-injected mice exhibit pathology formation before the onset of cell loss suggesting that pathological species lead to neurodegeneration [52, 53, 93]. Nonetheless, there is considerable debate as to whether inclusions such as LBs act as a protective sink for toxic α-Syn intermediates or that they lead to sequestration of the physiological α-Syn pool, resulting in the loss of normal α-Syn function. Thus, an important question is whether neuroprotection or toxicity (or neither) is enhanced in cells that contain the pathological aggregates. To address this issue, Unni and colleagues used multi-photon imaging to monitor α-Syn pathology development and long term neuronal viability after intracerebral α-Syn PFF injection in mice expressing an α-Syn-GFP fusion protein [94]. As predicted, these animals developed inclusions of α-Syn-GFP that matured and condensed over time. By following the α-Syn-GFP aggregates within individual neurons over time, the authors found that the aggregate-bearing neurons were selectively lost. Their results also showed that while affected neurons were lost over a range of time after inclusion formation, neighboring neurons were consistently preserved, suggesting the lack of a widespread toxic process involving other cells.
Collectively, these data provide several interesting clues into the toxicity of α-Syn aggregation and the cause of neurodegeneration. First, they suggest that α-Syn inclusion formation is necessary for toxicity and that cell death correlates with inclusion maturation. Although it is clear that neurons containing pathology are the ones that are vulnerable, this raises the question of whether fibrillar forms of α-Syn found in pathological aggregates is necessary for neurodegeneration in the susceptible population. Moreover, synaptic and trafficking impairments are detectable soon after pathological seeding when inclusions are confined to axons [95]. Lastly, they suggest that cell death following inclusion formation is likely a primarily cell-autonomous process. α-Syn inclusion induced cell death is also apparent in primary neuronal culture, where microglial responses are limited [52]. Although, this does not preclude the possibility of microglia activating after the onset of neurodegeneration further enhancing cell death.
3.1 A protein of many (ascribed) functions: implications of loss of function
α-Syn is the most studied member of a three member protein family comprised of α, β, and γ synuclein [7, 96]. α-Syn is an 140-amino acid protein comprised of three distinct domains. An amphipathic N-terminal portion containing seven 11-amino acid repeats in thought to interact with membranes by adopting an alpha helical structure when bound [97]. The function of the acidic and proline-rich C-terminal region is unknown, but the presence of several putative phosphorylation and post translational sites points to involvement in regulating α-Syn activity and/or localization [54, 98]. Biophysical studies also suggest that the C-terminus also interacts with the hydrophobic region that forms the core of the α-Syn molecule, preventing its self-assembly into the beta-sheet rich amyloid fibrils in LBs and GCIs [99, 100].
Although it is found in a number of tissues, α-Syn is most highly expressed in both peripheral and central neurons [101]. At the intracellular level, the highest levels of α-Syn are colocalized to pre-synaptic terminals, consistent with its purported involvement in regulating synaptic vesicle cycling and endocytosis [101–104]. α-Syn has been shown to facilitate and stabilize SNARE complex formation, and overexpression of WT α-syn in cultured neurons leads to decreased exocytosis in both hippocampal and DA neurons [105, 106]. Interestingly, apart from modulation in DA release and recycling pool homeostasis, mice lacking α-Syn show no noticeable phenotype, possibly owing to redundancy of the other synucleins or other compensatory mechanisms [107–110]. Indeed, mice lacking α-, β-, and γ-syn show age-related neurodegeneration and endocytosis abnormalities [111, 112]. Acute knockdown of α-Syn by means of adeno-associated virus expressed RNAi also promotes degeneration in SN neurons [113].
In addition, α-Syn has been reported to be localized to the cytosol, nucleus, the mitochondria associated membrane of the ER, and the inner membrane of mitochondria, suggesting that it may be actively trafficked between different compartments [114, 115]. Additional studies have found that α-syn can affect nuclear protein function [116–118]. α-Syn’s molecular weight is below the nuclear pore cut off allowing α-syn to diffuse from the general cytosolic population into the nucleus without a prototypical NLS. Other putative functions for α-syn involve mitochondrial maintenance, consistent with reports that α-Syn binds to mitochondria, and perturbation of α-syn levels can impair mitochondrial function [119–122]. Nonetheless, many of these functions ascribed to α-Syn remain too poorly defined, and whether their loss contributes to toxicity remains to be elucidated.
3.2 Misfolded α-Syn as toxic species
In addition to the prototypical 12–15 nm wide amyloid fibrils found in LBs, multiple molecular species of various sizes have been reported, including short fibril-like intermediates (protofibrils) and spherical or annular oligomers ranging from ~25 to several hundred α-Syn molecules [123, 124]. Toxic oligomers contain a β-pleated sheet structure and are much smaller compared to aggregates and are variable in size. Oligomers are believed to be on the pathway between α-Syn monomer and fibrils, but off-pathway oligomers have been described to cause toxicity [125, 126]. It is also possible that as α-Syn becomes fibrillar and forms inclusions, the concentration of intermediates (i.e. oligomers) may increase which could increase toxicity [127]. Toxicity or mechanism thereof has been controversial, although a common theme is the disruption of membranes. Winner et al. showed that α-Syn mutants that preferentially formed oligomers resulted in enhanced DA neuron loss when overexpressed in rat midbrain, and α-syn oligomers are capable of forming pores in membranes and cause cell toxicity and death due to increased calcium permeability [128–131].
The topic of oligomers as the toxic species is still contentious because membrane permeabilization may not be specific to oligomers and may only be transient, which would suggest that what is actually being observed is α-syn transiently binding to membranes [132, 133]. Moreover, the unstable/transient nature of oligomeric α-Syn has also precluded their isolation and in-depth characterization. The recent work into oligomer and fibril toxicity suggests a gain of toxic function mechanism, but this does not exclude the possibility of loss of α-syn function further increasing toxicity. These two options need not be mutually exclusive, and this remains an active area of debate.
The α-Syn aggregation and cell loss seen after seeding pathology with α-syn PFFs is highly dependent upon the production of α-Syn within neurons. Importantly, neither cell loss nor the induction of pathology occurs in α-Syn−/− mice or primary neurons [52, 53]. We have also observed that as primary neurons mature they develop pathology at a much faster rate compared to younger neurons, which is most likely due to primary neurons producing more α-syn as they age in culture [52]. Interestingly, it has been noted that the GABAergic neurons are resistant to α-Syn aggregation in primary hippocampal cultures after PFF addition, due to their lower levels of α-Syn expression compared to glutamatergic neurons despite comparable capacity to internalize α-Syn [134]. Similarly, M83 hemizygous mice expressing the A53T α-Syn mutant injected intracerebrally with PFFs show delayed onset of symptoms compared to injected homozygous mice [47, 49]. It is worth noting that symptom-free survival in M83 hemizygous mice was nearly double that of the homozygous counterparts, but their rate of decline was similar suggesting that the increased α-syn expression in the M83 homozygous mice increased the rate of nucleation and not neurodegeneration. Supporting this is the observation that patients with α-Syn triplications develop disease at an earlier age compared to patients with α-Syn duplications [23].
4. Origins of misfolded α-Syn
Despite the expanding literature on cell-to-cell transfer of α-Syn, much less is known about how misfolded α-Syn species initially arise. The ubiquitous and robust expression of α-Syn in neurons makes them the most probable location for this to occur. Impairments in protein degradation and quality control machinery directly increase intracellular α-Syn concentration, augmenting the rate of both cell autonomous α-Syn aggregate formation and the risk of seeding by exogenous α-Syn.
However, the localization of α-Syn may be as important as its quantity. Most groups report that α-Syn is mostly a natively unfolded monomer within the cell using both denaturing and non-denaturing conditions and that α-Syn is only weakly associated with synaptosomal and mitochondrial fractions [112, 135–137]. This contrasts with reports that α-Syn exists as a membrane bound multimer and that stable tetramers can be isolated under non-denaturing conditions [138–140]. Given the affinity of α-Syn for phospholipids and high curvature membranes, this discrepancy could reflect interactions between α-Syn and membranes that are highly transient. Indeed, cross-linking appears to stabilize a multimeric membrane bound pool and SNCA mutations known to cause familial PD cluster within a small stretch of the N-terminal region, suggesting that interactions with membranes is vital [138–143]. These mutations, as well as directed mutations in the imperfect repeats preceding the core hydrophobic domain, reduce membrane binding through disruption of α-Syn multimers [144, 145].
Increased α-Syn dissociation is thought to result in the accumulation of unfolded species in the cytoplasmic compartment shifting the α-Syn equilibrium towards the formation of oligomeric and/or fibrillar species. Although several familial PD α-Syn mutants show accelerated in vitro aggregation kinetics (e.g., E46K, H50Q, A53T), fibrillization in others are reduced (A30P, G51D), indicating that both increased levels of unstructured α-Syn and aggregation propensity factor in the generation of misfolded fibrillar species [146–149]. In line with this, artificial α-Syn mutants with compromised membrane binding also preferably segregate into the soluble unbound pool. Importantly, overexpression of these mutants in vivo greatly enhances toxicity in SN DA neurons, although it remains to be seen whether wildtype α-Syn also undergoes dissociation into a more aggregation-prone state at levels that support misfolding [150].
Although modest accumulation is observed in the brains of transgenic animals overexpressing α-Syn, the dramatic increase in pathological α-Syn following introduction of either PD brain extracts or recombinant α-Syn fibrils suggests that normal α-Syn located within membranes are vulnerable to conversion by pathologic species. This accelerated conversion is also apparent in models of α-Syn seeding using non-transgenic rodents as well as non-human primates indicating that fibrillar, and possibly oligomeric intermediates, are capable of destabilizing native α-Syn [48, 52, 54, 56].
5. Pathophysiology of α-Syn misfolding
Similar to the role α-Syn oligomers play in disease, considerable debate exists regarding the cellular stressors arising from α-Syn misfolding that contribute to neurodegeneration. Nonetheless, work from both genetic and fibril-injection based models of synucleinopathies have provided fresh insights to the knowledge derived from more traditional, mainly neurotoxin (e.g., MPTP, rotenone, and 6-OHDA) dependent models. A particularly interesting observation is that α-Syn appears to modulate the toxicity in these latter models. For example, mice lacking α-Syn expression show relative resistance to MPTP induced degeneration of midbrain DA neurons, further highlighting a role for α-Syn in degeneration and that these approaches may intersect at the stressor pathways they activate [151]. Several different cell stressors have been implicated by both neurotoxin based PD models and genetic models to cause neurodegeneration. Among these are ER stress and the unfolded protein response (UPR), oxidative stress and mitochondrial dysregulation, calcium homeostasis dysregulation, and neuroinflammation.
5.1 Endoplasmic reticulum stress
ER stress and activation of the unfolded protein response have been observed in various cell culture and mouse models of PD where α-Syn aggregation is present, including transgenic mice expressing human α-Syn with the A53T mutation [152, 153]. In both the MPTP and 6-OHDA models of PD upregulation of markers such as CHOP and ATF4 also point to ER stress. Genetic deletion of CHOP can prevent neurodegeneration in the 6-OHDA model, although this same effect did not occur in the MPTP model [154]. In addition, treatment with salubrinal, an inhibitor of ER stress, can alleviate motor symptoms and cause neuroprotection [152]. A downstream consequence of ATF4 and CHOP activation is the increase of Trib3, and knockdown of this pro-apoptotic product reduces ER stress and attenuates neuron loss in both neurotoxin and PFF based models [155].
In PD post mortem patient samples the presence of several key markers, including phosphorylated PERK are also consistent with ER stress and UPR activation in the face of increased misfolded α-Syn [156]. Several genetic mutations that cause familial PD are known to affect protein degradation and trafficking. Chief among these are the GBA, VPS35, ATP13A2, and LRRK2 mutations which affect either lysosomal function or ER trafficking, and there is evidence that VPS35 may interact with α-Syn [34, 157–159]. ER-Golgi trafficking dysfunction has also been noted in the PFF model in multiple cell types [51, 95] while overexpression of Rab1, a regulator of ER-Golgi trafficking in yeast, or its homologs leads to rescue from cell death in PD models [160]. Induced pluripotent stem cells derived from PD patients with genetic mutations also develop ER stress which is alleviated by the ubiquitin ligase Nedd4, further implicating proteosomal/ER stress in PD, and enhanced Nedd4 activity in attenuating this dysfunction [161, 162]. It is unknown if ER stress and UPR activation are a cause or effect in PD pathogenesis, although the evidence presented suggests both.
5.2. Oxidative Stress
As with ER stress, it is unclear if oxidative stress represents a cause and/or effect in PD pathogenesis. Compared to controls, brain tissue from PD patients contain altered levels of antioxidant proteins, such as GPX4 and glutathione, suggesting that this mechanism may play a role in disease [163]. In addition, mutations in Parkin and PINK1, which maintain mitochondrial health, cause autosomal recessive forms of PD [164–166]. Even though knockout of these genes in mice do not result in α-Syn aggregation, increased mitochondrial stress has been observed in these mice indicating that they play a role in maintaining neuronal health [167, 168]. Interestingly, α-Syn null mice are also protected from DA neuron loss following treatment with the mitochondrial complex I inhibitor MPTP suggesting that α-Syn mediates toxicity [169]. Consistent with this, cultured DA neurons exposed to α-Syn fibrils show elevations in both mitochondrial and cytoplasmic oxidative stress [170]. More recently, α-Syn has been shown to be localized with mitochondrial associated ER, suggesting α-Syn may have a function in mitochondrial homeostasis and disruption of this could contribute to ER stress and mitochondrial dysfunction and providing a link to PD pathogenesis and neurodegeneration of DA neurons [114].
5.3 Calcium buffering and vulnerability within DA subpopulations
Calcium ions (Ca++) are used extensively for cell signaling but imbalances can also be a potential source of toxicity [171]. Furthermore, it is known that multiple neuronal populations affected in PD show a combination of low Ca++ buffering capacity and broad action potentials, leaving them vulnerable to increased cytoplasmic Ca++ levels [172, 173]. In particular, SN neurons utilize Cav1.3 Ca++ channels for their rhythmic pacemaking activity, exposing them to higher calcium conductance relative to neurons in the nearby ventral tegmental area (VTA) [174]. Blockers that prevent Cav1.3 mediated Ca++ entry protect from MPTP but this is not clear for α-Syn PFF models [175, 176]. Curiously, Ca++ toxicity in DA neurons also appears to be α-Syn-dependent [177].
Because the ER represents a significant reservoir for intracellular Ca++, it is possible that ER stress could be a source of Ca++ toxicity in PD. For example disruptions in calcium homeostasis has been shown in midbrain neurons derived from induced pluripotent stem cells from patients bearing GBA mutations [178]. Higher Ca++ conductance has been linked to increased oxidative stress [179]. It is possible that a vicious cycle involving a challenge of ER Ca++ storage capacity and the reliance on voltage gated Cav1.3 channels leads to elevated Ca++ levels that trigger the demise of SN, and possibly other, neurons.
5.4 Neuroinflammation
It has been suggested that neuroinflammation from activated microglia may increase neuronal toxicity. It is known that microglia are activated in areas of neurodegeneration in both post mortem PD tissue and in vivo mouse models of PD [49, 55, 180, 181]. PD patients develop increased pro-inflammatory cytokines in their CSF, such as TNF-α and IL-1β [182, 183]. Oligomeric α-Syn has been shown to bind TLR2, β1-integrin, and CD11b, which can activate microglia [68–70]. It is currently debated whether microglia in PD are actively inducing neurodegeneration, have a more benign role of cleaning the cellular debris left from dying cells, or possibly both. It has been shown that those neurons that develop pathology eventually are lost where the surrounding neurons are spared in PFF models suggesting that cell death is cell autonomous due to the pathological aggregate and not a reactive extracellular mileu [94].
6. Neuron-specific vulnerability
If α-Syn misfolding represents a driving pathogenic event, as suggested by clinical and experimental evidence, then the selective pattern of degeneration that characterizes PD would suggest that certain cell subpopulations possess an enhanced susceptibility to acquire or develop α-Syn pathology along with elevated vulnerability to the induced toxicity (Figure 1). SN DA neurons provide a prototypic example of how these factors might converge in a susceptible population. Firstly, DA neurons carry enormous metabolic demands due to their high-frequency autonomous firing resulting in greater production of oxidative stress and increased calcium entry into the cell for the release of synaptic vesicles [171, 179, 184]. Unlike VTA neurons and other populations that express calcium binding proteins (e.g. calbindin), SN neurons, particularly in the ventral tier, show a paucity of these buffers [174]. This, together with their susceptibility to elevated calcium levels due partly to a dependence on Cav1.3 channels for their pacemaking activity furthers their exquisite sensitivity to perturbations in oxidative respiration (e.g., complex 1 inhibition). In line with the view that DA neurons are especially sensitive to mitochondrial impairment, mutations in mitochondrial DNA or PINK1 and Parkin (two crucial components in mitophagy) are also associated with PD. The bioenergetic demands of DA neurons are further pushed by the need to maintain elaborate and high density of connections with target neurons. It is likely that perturbations in the energy balance disrupt DA neuron arbors leading to the loss of synapses, and in turn make it susceptible to loss of trophic support that likely helps sustain the neurons [185, 186].
Figure 1. Spread and effect of α-Syn pathology.
Proposed sequence of pathological α-Syn transmission. Pathology is initially transmitted between neurons and propagates primarily in a retrograde manner. Resistant populations of neurons (top row) are either capable of degrading the seeds without forming pathology or have increased capacity to survive and function with pathological inclusions. Vulnerable populations (middle row) exhibit α-Syn aggregates that grow and mature over time. These aggregates are capable of disrupting normal neuronal function (yellow-green neurons) by oxidative stress, ER stress, or other potential stress mechanisms. Diseased neurons containing inclusions may be capable of further pathological seeding. After a certain level of cell stress is reached, the diseased neurons undergo cell death (yellow-green neuron with dashed outline).
Given the recent body of evidence suggesting pathological α-Syn transmission, these neurons are at high risk of being exposed to seeding species due to their large number of synapses and poorly myelinated axons. The high levels of α-Syn enriched in processes (especially synaptic terminals) further increases this risk. Consistent with this, over 90% of PD α-Syn pathology is present as Lewy neurites, rather than LBs in the soma [187]. Axons also represent the initial sites for the accumulation of hyperphosphorylated α-Syn inclusions in cultured hippocampal neurons suggesting that synaptic connections represent a general “hot spot” for pathology in other types of neurons [52].
It has also been argued that elevated Ca++ levels may also increase the rate of endocytic and secretory events, major routes hypothesized for pathologic α-Syn entry and exit, respectively, and making neurons prone to seeding aggregation. Possibly exacerbating this is the observation that DA neurons exhibit robust α-Syn expression which increases with age in the SN of both humans and non-human primates, a phenomenon that is not observed in other DA neuronal populations that are relatively spared in PD, such as the VTA [188].
Intriguingly, many of these above properties also apply to a long list of neuron subpopulations selectively affected in PD including those in the locus coeruleus, raphe nucleus, dorsal motor nucleus of the vagus, pedunculopontine nucleus, nucleus basalis of Meynert, and enteric neurons in the large intestine. Another commonality between several of them is their monoaminergic neurotransmitter phenotype. In particular, catecholamines are highly reactive and would predispose cells that contain high levels of them to dysfunction or death due to oxidative stress [184, 189]. Cells that contain catecholamine neurotransmitters also can contain neuromelanin (NM) which is believed to be a protective mechanism preventing excess oxidative stress due to catecholamine production [190]. NM is a sign of oxidative stress and can sequester metal ions like iron which can contribute to the Fenton reaction and lipid peroxidation [191]. Supporting this is an age associated increase in nigral NM content, and humans, the only species known to develop PD, tend to develop much larger stores of NM compared to non-human primates [192]. More recently, it has been shown that catecholaminergic neurons are predisposed to MHC I induction compared to other neuronal populations, and that cell death could occur in the presence of the appropriate Cytotoxic T cells making this population prone to neuronal loss [193].
7. Conclusion
The past several years have witnessed a dramatic expansion in our knowledge regarding the role of α-Syn in the pathogenesis of PD and related proteinopathies. Recent work clearly suggests that rather than a single dominant factor, the causes of α-Syn pathology is multifaceted. Furthermore, these processes interact with inherent vulnerabilities of certain neurons to give rise to the observed degeneration and symptoms. These factors vary across neuron populations in the central and peripheral nervous systems, and likely underlie some of the variability between the synucleinopathies. Pathological α-Syn behaves as a prion-like agent, capable of templating the conversion of intracellular α-Syn and providing a route for the propagation of the pathological process into unaffected neurons. The rate of pathology induction is partially dependent on the total amount of α-Syn, which can vary across neuronal populations. Following this is the onset of neuronal dysfunction and then neurodegeneration of those neurons that contain sufficiently large pathological aggregates (Figure 1). Understanding these areas and how they interact will be key to developing disease modifying therapeutics to treat these patients.
Acknowledgements
This work was supported by grant NS088322 from the National Institute of Neurological Disorders and Stroke and the Michael J Fox Foundation. EL is supported by a training grant from the National Institutes on Aging (T32-AG000255 – 16).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.de Lau M, Breteler MM. Epidemiology of Parkinson’s disease. Lancet Neurol. 2006;5:525–535. doi: 10.1016/S1474-4422(06)70471-9. [DOI] [PubMed] [Google Scholar]
- 2.Van Den Eeden SK, Tanner CM, Bernstein AL, Fross RD, Leimpeter A, Bloch DA, Nelson LM. Incidence of Parkinson’s disease: variation by age, gender, and race/ethnicity. AmJEpidemiol. 2003;157:1015–1022. doi: 10.1093/aje/kwg068. [DOI] [PubMed] [Google Scholar]
- 3.Reeve A, Simcox E, Turnbull D. Ageing and Parkinson’s disease: why is advancing age the biggest risk factor? Ageing research reviews. 2014;14:19–30. doi: 10.1016/j.arr.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Dauer W, Przedborski S. Parkinson’s disease: mechanisms and models. Neuron. 2003;39:889–909. doi: 10.1016/s0896-6273(03)00568-3. [DOI] [PubMed] [Google Scholar]
- 5.Goedert M, Spillantini MG, Del Tredici K, Braak H. 100 years of Lewy pathology. Nature reviews Neurology. 2013;9:13–24. doi: 10.1038/nrneurol.2012.242. [DOI] [PubMed] [Google Scholar]
- 6.Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–840. doi: 10.1038/42166. [DOI] [PubMed] [Google Scholar]
- 7.Maroteaux L, Scheller RH. The rat brain synucleins; family of proteins transiently associated with neuronal membrane. Brain research Molecular brain research. 1991;11:335–343. doi: 10.1016/0169-328x(91)90043-w. [DOI] [PubMed] [Google Scholar]
- 8.Jellinger KA. Neuropathological spectrum of synucleinopathies. Movement disorders : official journal of the Movement Disorder Society. 2003;18(Suppl 6):S2–S12. doi: 10.1002/mds.10557. [DOI] [PubMed] [Google Scholar]
- 9.Wakabayashi K, Matsumoto K, Takayama K, Yoshimoto M, Takahashi H. NACP, a presynaptic protein, immunoreactivity in Lewy bodies in Parkinson’s disease. Neuroscience letters. 1997;239:45–48. doi: 10.1016/s0304-3940(97)00891-4. [DOI] [PubMed] [Google Scholar]
- 10.Hashimoto M, Masliah E. Alpha-synuclein in Lewy body disease and Alzheimer’s disease. Brain Pathol. 1999;9:707–720. doi: 10.1111/j.1750-3639.1999.tb00552.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Li W, West N, Colla E, Pletnikova O, Troncoso JC, Marsh L, Dawson TM, Jakala P, Hartmann T, Price DL, Lee MK. Aggregation promoting C-terminal truncation of alpha-synuclein is a normal cellular process and is enhanced by the familial Parkinson’s disease-linked mutations. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:2162–2167. doi: 10.1073/pnas.0406976102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Anderson JP, Walker DE, Goldstein JM, de Laat R, Banducci K, Caccavello RJ, Barbour R, Huang J, Kling K, Lee M, Diep L, Keim PS, Shen X, Chataway T, Schlossmacher MG, Seubert P, Schenk D, Sinha S, Gai WP, Chilcote TJ. Phosphorylation of Ser-129 is the dominant pathological modification of alpha-synuclein in familial and sporadic Lewy body disease. The Journal of biological chemistry. 2006;281:29739–29752. doi: 10.1074/jbc.M600933200. [DOI] [PubMed] [Google Scholar]
- 13.Fujiwara H, Hasegawa M, Dohmae N, Kawashima A, Masliah E, Goldberg MS, Shen J, Takio K, Iwatsubo T. alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nature cell biology. 2002;4:160–164. doi: 10.1038/ncb748. [DOI] [PubMed] [Google Scholar]
- 14.Hasegawa M, Fujiwara H, Nonaka T, Wakabayashi K, Takahashi H, Lee VM, Trojanowski JQ, Mann D, Iwatsubo T. Phosphorylated alpha-synuclein is ubiquitinated in alpha-synucleinopathy lesions. The Journal of biological chemistry. 2002;277:49071–49076. doi: 10.1074/jbc.M208046200. [DOI] [PubMed] [Google Scholar]
- 15.Takeda A, Mallory M, Sundsmo M, Honer W, Hansen L, Masliah E. Abnormal accumulation of NACP/alpha-synuclein in neurodegenerative disorders. The American journal of pathology. 1998;152:367–372. [PMC free article] [PubMed] [Google Scholar]
- 16.Giasson BI, Duda JE, Murray IV, Chen Q, Souza JM, Hurtig HI, Ischiropoulos H, Trojanowski JQ, Lee VM. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science. 2000;290:985–989. doi: 10.1126/science.290.5493.985. [DOI] [PubMed] [Google Scholar]
- 17.Polymeropoulos MH, Higgins JJ, Golbe LI, Johnson WG, Ide SE, Di Iorio G, Sanges G, Stenroos ES, Pho LT, Schaffer AA, Lazzarini AM, Nussbaum RL, Duvoisin RC. Mapping of a gene for Parkinson’s disease to chromosome 4q21-q23. Science. 1996;274:1197–1199. doi: 10.1126/science.274.5290.1197. [DOI] [PubMed] [Google Scholar]
- 18.Kruger R, Kuhn W, Muller T, Woitalla D, Graeber M, Kosel S, Przuntek H, Epplen JT, Schols L, Riess O. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nature genetics. 1998;18:106–108. doi: 10.1038/ng0298-106. [DOI] [PubMed] [Google Scholar]
- 19.Zarranz JJ, Alegre J, Gomez-Esteban JC, Lezcano E, Ros R, Ampuero I, Vidal L, Hoenicka J, Rodriguez O, Atares B, Llorens V, Gomez Tortosa E, del Ser T, Munoz DG, de Yebenes JG. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Annals of neurology. 2004;55:164–173. doi: 10.1002/ana.10795. [DOI] [PubMed] [Google Scholar]
- 20.Appel-Cresswell S, Vilarino-Guell C, Encarnacion M, Sherman H, Yu I, Shah B, Weir D, Thompson C, Szu-Tu C, Trinh J, Aasly JO, Rajput A, Rajput AH, Jon Stoessl A, Farrer MJ. Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson’s disease. Movement disorders : official journal of the Movement Disorder Society. 2013;28:811–813. doi: 10.1002/mds.25421. [DOI] [PubMed] [Google Scholar]
- 21.Lesage S, Anheim M, Letournel F, Bousset L, Honore A, Rozas N, Pieri L, Madiona K, Durr A, Melki R, Verny C, Brice A. G51D alpha-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Annals of neurology. 2013;73:459–471. doi: 10.1002/ana.23894. [DOI] [PubMed] [Google Scholar]
- 22.Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nussbaum R, Lincoln S, Crawley A, Hanson M, Maraganore D, Adler C, Cookson MR, Muenter M, Baptista M, Miller D, Blancato J, Hardy J, Gwinn-Hardy K. alpha-Synuclein locus triplication causes Parkinson’s disease. Science. 2003;302:841. doi: 10.1126/science.1090278. [DOI] [PubMed] [Google Scholar]
- 23.Chartier-Harlin MC, Kachergus J, Roumier C, Mouroux V, Douay X, Lincoln S, Levecque C, Larvor L, Andrieux J, Hulihan M, Waucquier N, Defebvre L, Amouyel P, Farrer M, Destee A. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet. 2004;364:1167–1169. doi: 10.1016/S0140-6736(04)17103-1. [DOI] [PubMed] [Google Scholar]
- 24.Maraganore DM, de Andrade M, Elbaz A, Farrer MJ, Ioannidis JP, Kruger R, Rocca WA, Schneider NK, Lesnick TG, Lincoln SJ, Hulihan MM, Aasly JO, Ashizawa T, Chartier-Harlin MC, Checkoway H, Ferrarese C, Hadjigeorgiou G, Hattori N, Kawakami H, Lambert JC, Lynch T, Mellick GD, Papapetropoulos S, Parsian A, Quattrone A, Riess O, Tan EK, Van Broeckhoven C. Collaborative analysis of alpha-synuclein gene promoter variability and Parkinson disease. Jama. 2006;296:661–670. doi: 10.1001/jama.296.6.661. [DOI] [PubMed] [Google Scholar]
- 25.Nalls MA, Plagnol V, Hernandez DG, Sharma M, Sheerin UM, Saad M, Simon-Sanchez J, Schulte C, Lesage S, Sveinbjornsdottir S, Stefansson K, Martinez M, Hardy J, Heutink P, Brice A, Gasser T, Singleton AB, Wood NW. Imputation of sequence variants for identification of genetic risks for Parkinson’s disease: a meta-analysis of genome-wide association studies. Lancet. 2011;377:641–649. doi: 10.1016/S0140-6736(10)62345-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Simon-Sanchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, Paisan-Ruiz C, Lichtner P, Scholz SW, Hernandez DG, Kruger R, Federoff M, Klein C, Goate A, Perlmutter J, Bonin M, Nalls MA, Illig T, Gieger C, Houlden H, Steffens M, Okun MS, Racette BA, Cookson MR, Foote KD, Fernandez HH, Traynor BJ, Schreiber S, Arepalli S, Zonozi R, Gwinn K, van der Brug M, Lopez G, Chanock SJ, Schatzkin A, Park Y, Hollenbeck A, Gao J, Huang X, Wood NW, Lorenz D, Deuschl G, Chen H, Riess O, Hardy JA, Singleton AB, Gasser T. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nature genetics. 2009;41:1308–1312. doi: 10.1038/ng.487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mazzulli JR, Xu YH, Sun Y, Knight AL, McLean PJ, Caldwell GA, Sidransky E, Grabowski GA, Krainc D. Gaucher disease glucocerebrosidase and alpha-synuclein form a bidirectional pathogenic loop in synucleinopathies. Cell. 2011;146:37–52. doi: 10.1016/j.cell.2011.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Gan-Or Z, Giladi N, Orr-Urtreger A. Differential phenotype in Parkinson’s disease patients with severe versus mild GBA mutations. Brain : a journal of neurology. 2009;132:e125. doi: 10.1093/brain/awp161. [DOI] [PubMed] [Google Scholar]
- 29.Bandopadhyay R, Kingsbury AE, Cookson MR, Reid AR, Evans IM, Hope AD, Pittman AM, Lashley T, Canet-Aviles R, Miller DW, McLendon C, Strand C, Leonard AJ, Abou-Sleiman PM, Healy DG, Ariga H, Wood NW, de Silva R, Revesz T, Hardy JA, Lees AJ. The expression of DJ-1 (PARK7) in normal human CNS and idiopathic Parkinson’s disease. Brain : a journal of neurology. 2004;127:420–430. doi: 10.1093/brain/awh054. [DOI] [PubMed] [Google Scholar]
- 30.Samaranch L, Lorenzo-Betancor O, Arbelo JM, Ferrer I, Lorenzo E, Irigoyen J, Pastor MA, Marrero C, Isla C, Herrera-Henriquez J, Pastor P. PINK1-linked parkinsonism is associated with Lewy body pathology. Brain : a journal of neurology. 2010;133:1128–1142. doi: 10.1093/brain/awq051. [DOI] [PubMed] [Google Scholar]
- 31.Neumann J, Bras J, Deas E, O’Sullivan SS, Parkkinen L, Lachmann RH, Li A, Holton J, Guerreiro R, Paudel R, Segarane B, Singleton A, Lees A, Hardy J, Houlden H, Revesz T, Wood NW. Glucocerebrosidase mutations in clinical and pathologically proven Parkinson’s disease. Brain : a journal of neurology. 2009;132:1783–1794. doi: 10.1093/brain/awp044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kalia LV, Lang AE, Hazrati LN, Fujioka S, Wszolek ZK, Dickson DW, Ross OA, Van Deerlin VM, Trojanowski JQ, Hurtig HI, Alcalay RN, Marder KS, Clark LN, Gaig C, Tolosa E, Ruiz-Martinez J, Marti-Masso JF, Ferrer I, Lopez de Munain A, Goldman SM, Schule B, Langston JW, Aasly JO, Giordana MT, Bonifati V, Puschmann A, Canesi M, Pezzoli G, Maues De Paula A, Hasegawa K, Duyckaerts C, Brice A, Stoessl AJ, Marras C. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA neurology. 2015;72:100–105. doi: 10.1001/jamaneurol.2014.2704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Doherty KM, Silveira-Moriyama L, Parkkinen L, Healy DG, Farrell M, Mencacci NE, Ahmed Z, Brett FM, Hardy J, Quinn N, Counihan TJ, Lynch T, Fox ZV, Revesz T, Lees AJ, Holton JL. Parkin disease: a clinicopathologic entity? JAMA neurology. 2013;70:571–579. doi: 10.1001/jamaneurol.2013.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ramirez A, Heimbach A, Grundemann J, Stiller B, Hampshire D, Cid LP, Goebel I, Mubaidin AF, Wriekat AL, Roeper J, Al-Din A, Hillmer AM, Karsak M, Liss B, Woods CG, Behrens MI, Kubisch C. Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nature genetics. 2006;38:1184–1191. doi: 10.1038/ng1884. [DOI] [PubMed] [Google Scholar]
- 35.Gai WP, Power JH, Blumbergs PC, Blessing WW. Multiple-system atrophy: a new alpha-synuclein disease? Lancet. 1998;352:547–548. doi: 10.1016/s0140-6736(05)79256-4. [DOI] [PubMed] [Google Scholar]
- 36.Braak H, Rub U, Jansen Steur EN, Del Tredici K, de Vos RA. Cognitive status correlates with neuropathologic stage in Parkinson disease. Neurology. 2005;64:1404–1410. doi: 10.1212/01.WNL.0000158422.41380.82. [DOI] [PubMed] [Google Scholar]
- 37.Lee HM, Koh SB. Many Faces of Parkinson’s Disease: Non-Motor Symptoms of Parkinson’s Disease. Journal of movement disorders. 2015;8:92–97. doi: 10.14802/jmd.15003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiology of aging. 2003;24:197–211. doi: 10.1016/s0197-4580(02)00065-9. [DOI] [PubMed] [Google Scholar]
- 39.Braak H, Rub U, Gai WP, Del Tredici K. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm. 2003;110:517–536. doi: 10.1007/s00702-002-0808-2. [DOI] [PubMed] [Google Scholar]
- 40.Dickson DW, Uchikado H, Fujishiro H, Tsuboi Y. Evidence in favor of Braak staging of Parkinson’s disease. Movement disorders : official journal of the Movement Disorder Society. 2010;25(Suppl 1):S78–S82. doi: 10.1002/mds.22637. [DOI] [PubMed] [Google Scholar]
- 41.Halliday G, Hely M, Reid W, Morris J. The progression of pathology in longitudinally followed patients with Parkinson’s disease. Acta neuropathologica. 2008;115:409–415. doi: 10.1007/s00401-008-0344-8. [DOI] [PubMed] [Google Scholar]
- 42.Jellinger KA. A critical reappraisal of current staging of Lewy-related pathology in human brain. Acta neuropathologica. 2008;116:1–16. doi: 10.1007/s00401-008-0406-y. [DOI] [PubMed] [Google Scholar]
- 43.Kalaitzakis ME, Graeber MB, Gentleman SM, Pearce RK. The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson’s disease: a critical analysis of alpha-synuclein staging. Neuropathology and applied neurobiology. 2008;34:284–295. doi: 10.1111/j.1365-2990.2007.00923.x. [DOI] [PubMed] [Google Scholar]
- 44.Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J, Henry-Watson J, Sasse J, Boyer S, Shirohi S, Brooks R, Eschbacher J, White CL, 3rd, Akiyama H, Caviness J, Shill HA, Connor DJ, Sabbagh MN, Walker DG. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta neuropathologica. 2009;117:613–634. doi: 10.1007/s00401-009-0538-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson’s disease. Nature medicine. 2008;14:504–506. doi: 10.1038/nm1747. [DOI] [PubMed] [Google Scholar]
- 46.Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, Lashley T, Quinn NP, Rehncrona S, Bjorklund A, Widner H, Revesz T, Lindvall O, Brundin P. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nature medicine. 2008;14:501–503. doi: 10.1038/nm1746. [DOI] [PubMed] [Google Scholar]
- 47.Luk KC, Kehm VM, Zhang B, O’Brien P, Trojanowski JQ, Lee VM. Intracerebral inoculation of pathological alpha-synuclein initiates a rapidly progressive neurodegenerative alpha-synucleinopathy in mice. The Journal of experimental medicine. 2012;209:975–986. doi: 10.1084/jem.20112457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Recasens A, Dehay B, Bove J, Carballo-Carbajal I, Dovero S, Perez-Villalba A, Fernagut PO, Blesa J, Parent A, Perier C, Farinas I, Obeso JA, Bezard E, Vila M. Lewy body extracts from Parkinson disease brains trigger alpha-synuclein pathology and neurodegeneration in mice and monkeys. Annals of neurology. 2014;75:351–362. doi: 10.1002/ana.24066. [DOI] [PubMed] [Google Scholar]
- 49.Watts JC, Giles K, Oehler A, Middleton L, Dexter DT, Gentleman SM, DeArmond SJ, Prusiner SB. Transmission of multiple system atrophy prions to transgenic mice. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:19555–195560. doi: 10.1073/pnas.1318268110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mougenot AL, Bencsik A, Nicot S, Vulin J, Morignat E, Verchere J, Betemps D, Lakhdar L, Legastelois S, Baron TG. Transmission of prion strains in a transgenic mouse model overexpressing human A53T mutated alpha-synuclein. Journal of neuropathology and experimental neurology. 2011;70:377–385. doi: 10.1097/NEN.0b013e318217d95f. [DOI] [PubMed] [Google Scholar]
- 51.Luk KC, Song C, O’Brien P, Stieber A, Branch JR, Brunden KR, Trojanowski JQ, Lee VM. Exogenous alpha-synuclein fibrils seed the formation of Lewy body-like intracellular inclusions in cultured cells. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:20051–20056. doi: 10.1073/pnas.0908005106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, Meaney DF, Trojanowski JQ, Lee VM. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron. 2011;72:57–71. doi: 10.1016/j.neuron.2011.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Trojanowski JQ, Lee VM. Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science. 2012;338:949–953. doi: 10.1126/science.1227157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Masuda-Suzukake M, Nonaka T, Hosokawa M, Oikawa T, Arai T, Akiyama H, Mann DM, Hasegawa M. Prion-like spreading of pathological alpha-synuclein in brain. Brain : a journal of neurology. 2013;136:1128–1138. doi: 10.1093/brain/awt037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Sacino AN, Brooks M, McKinney AB, Thomas MA, Shaw G, Golde TE, Giasson BI. Brain injection of alpha-synuclein induces multiple proteinopathies, gliosis, and a neuronal injury marker. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2014;34:12368–12378. doi: 10.1523/JNEUROSCI.2102-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Paumier KL, Luk KC, Manfredsson FP, Kanaan NM, Lipton JW, Collier TJ, Steece-Collier K, Kemp CJ, Celano S, Schulz E, Sandoval IM, Fleming S, Dirr E, Polinski NK, Trojanowski JQ, Lee VM, Sortwell CE. Intrastriatal injection of pre-formed mouse alpha-synuclein fibrils into rats triggers alpha-synuclein pathology and bilateral nigrostriatal degeneration. Neurobiology of disease. 2015;82:185–199. doi: 10.1016/j.nbd.2015.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sacino AN, Brooks M, Thomas MA, McKinney AB, Lee S, Regenhardt RW, McGarvey NH, Ayers JI, Notterpek L, Borchelt DR, Golde TE, Giasson BI. Intramuscular injection of alpha-synuclein induces CNS alpha-synuclein pathology and a rapid-onset motor phenotype in transgenic mice. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:10732–10737. doi: 10.1073/pnas.1321785111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Braak H, de Vos RA, Bohl J, Del Tredici K. Gastric alpha-synuclein immunoreactive inclusions in Meissner’s and Auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neuroscience letters. 2006;396:67–72. doi: 10.1016/j.neulet.2005.11.012. [DOI] [PubMed] [Google Scholar]
- 59.Svensson E, Horvath-Puho E, Thomsen RW, Djurhuus JC, Pedersen L, Borghammer P, Sorensen HT. Vagotomy and Subsequent Risk of Parkinson’s Disease. Annals of neurology. 2015 doi: 10.1002/ana.24448. [DOI] [PubMed] [Google Scholar]
- 60.Dunning CJ, George S, Brundin P. What’s to like about the prion-like hypothesis for the spreading of aggregated alpha-synuclein in Parkinson disease? Prion. 2013;7:92–97. doi: 10.4161/pri.23806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chu Y, Kordower JH. The prion hypothesis of Parkinson’s disease. Current neurology and neuroscience reports. 2015;15:28. doi: 10.1007/s11910-015-0549-x. [DOI] [PubMed] [Google Scholar]
- 62.Guo JL, Lee VM. Cell-to-cell transmission of pathogenic proteins in neurodegenerative diseases. Nature medicine. 2014;20:130–138. doi: 10.1038/nm.3457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hansen C, Angot E, Bergstrom AL, Steiner JA, Pieri L, Paul G, Outeiro TF, Melki R, Kallunki P, Fog K, Li JY, Brundin P. alpha-Synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. The Journal of clinical investigation. 2011;121:715–725. doi: 10.1172/JCI43366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, Spencer B, Masliah E, Lee SJ. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:13010–13015. doi: 10.1073/pnas.0903691106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Holmes BB, DeVos SL, Kfoury N, Li M, Jacks R, Yanamandra K, Ouidja MO, Brodsky FM, Marasa J, Bagchi DP, Kotzbauer PT, Miller TM, Papy-Garcia D, Diamond MI. Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:E3138–E3147. doi: 10.1073/pnas.1301440110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Reyes JF, Rey NL, Bousset L, Melki R, Brundin P, Angot E. Alpha-synuclein transfers from neurons to oligodendrocytes. Glia. 2014;62:387–398. doi: 10.1002/glia.22611. [DOI] [PubMed] [Google Scholar]
- 67.Peelaerts W, Bousset L, Van der Perren A, Moskalyuk A, Pulizzi R, Giugliano M, Van den Haute C, Melki R, Baekelandt V. alpha-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015;522:340–344. doi: 10.1038/nature14547. [DOI] [PubMed] [Google Scholar]
- 68.Kim C, Ho DH, Suk JE, You S, Michael S, Kang J, Joong Lee S, Masliah E, Hwang D, Lee HJ, Lee SJ. Neuron-released oligomeric alpha-synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nature communications. 2013;4:1562. doi: 10.1038/ncomms2534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kim C, Cho ED, Kim HK, You S, Lee HJ, Hwang D, Lee SJ. beta1-integrin-dependent migration of microglia in response to neuron-released alpha-synuclein. Experimental & molecular medicine. 2014;46:e91. doi: 10.1038/emm.2014.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wang S, Chu CH, Stewart T, Ginghina C, Wang Y, Nie H, Guo M, Wilson B, Hong JS, Zhang J. alpha-Synuclein, a chemoattractant, directs microglial migration via H2O2-dependent Lyn phosphorylation. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:E1926–E1935. doi: 10.1073/pnas.1417883112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Chai YJ, Kim D, Park J, Zhao H, Lee SJ, Chang S. The secreted oligomeric form of alpha-synuclein affects multiple steps of membrane trafficking. FEBS letters. 2013;587:452–459. doi: 10.1016/j.febslet.2013.01.008. [DOI] [PubMed] [Google Scholar]
- 72.Bae EJ, Yang NY, Song M, Lee CS, Lee JS, Jung BC, Lee HJ, Kim S, Masliah E, Sardi SP, Lee SJ. Glucocerebrosidase depletion enhances cell-to-cell transmission of alpha-synuclein. Nature communications. 2014;5:4755. doi: 10.1038/ncomms5755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Games D, Valera E, Spencer B, Rockenstein E, Mante M, Adame A, Patrick C, Ubhi K, Nuber S, Sacayon P, Zago W, Seubert P, Barbour R, Schenk D, Masliah E. Reducing C-terminal-truncated alpha-synuclein by immunotherapy attenuates neurodegeneration and propagation in Parkinson’s disease-like models. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2014;34:9441–9454. doi: 10.1523/JNEUROSCI.5314-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Gousset K, Schiff E, Langevin C, Marijanovic Z, Caputo A, Browman DT, Chenouard N, de Chaumont F, Martino A, Enninga J, Olivo-Marin JC, Mannel D, Zurzolo C. Prions hijack tunnelling nanotubes for intercellular spread. Nature cell biology. 2009;11:328–336. doi: 10.1038/ncb1841. [DOI] [PubMed] [Google Scholar]
- 75.Lee HJ, Patel S, Lee SJ. Intravesicular localization and exocytosis of alpha-synuclein and its aggregates. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2005;25:6016–6024. doi: 10.1523/JNEUROSCI.0692-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.El-Agnaf OM, Salem SA, Paleologou KE, Cooper LJ, Fullwood NJ, Gibson MJ, Curran MD, Court JA, Mann DM, Ikeda S, Cookson MR, Hardy J, Allsop D. Alpha-synuclein implicated in Parkinson’s disease is present in extracellular biological fluids, including human plasma. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2003;17:1945–1947. doi: 10.1096/fj.03-0098fje. [DOI] [PubMed] [Google Scholar]
- 77.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. Journal of neurochemistry. 2010;113:1263–1274. doi: 10.1111/j.1471-4159.2010.06695.x. [DOI] [PubMed] [Google Scholar]
- 78.Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis SD, Ntzouni M, Margaritis LH, Stefanis L, Vekrellis K. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2010;30:6838–6851. doi: 10.1523/JNEUROSCI.5699-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kunadt M, Eckermann K, Stuendl A, Gong J, Russo B, Strauss K, Rai S, Kugler S, Falomir Lockhart L, Schwalbe M, Krumova P, Oliveira LM, Bahr M, Mobius W, Levin J, Giese A, Kruse N, Mollenhauer B, Geiss-Friedlander R, Ludolph AC, Freischmidt A, Feiler MS, Danzer KM, Zweckstetter M, Jovin TM, Simons M, Weishaupt JH, Schneider A. Extracellular vesicle sorting of alpha-Synuclein is regulated by sumoylation. Acta neuropathologica. 2015;129:695–713. doi: 10.1007/s00401-015-1408-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hasegawa T, Konno M, Baba T, Sugeno N, Kikuchi A, Kobayashi M, Miura E, Tanaka N, Tamai K, Furukawa K, Arai H, Mori F, Wakabayashi K, Aoki M, Itoyama Y, Takeda A. The AAA-ATPase VPS4 regulates extracellular secretion and lysosomal targeting of alpha-synuclein. PloS one. 2011;6:e29460. doi: 10.1371/journal.pone.0029460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Webb JL, Ravikumar B, Atkins J, Skepper JN, Rubinsztein DC. Alpha-Synuclein is degraded by both autophagy and the proteasome. The Journal of biological chemistry. 2003;278:25009–25013. doi: 10.1074/jbc.M300227200. [DOI] [PubMed] [Google Scholar]
- 82.Masuda-Suzukake M, Nonaka T, Hosokawa M, Kubo M, Shimozawa A, Akiyama H, Hasegawa M. Pathological alpha-synuclein propagates through neural networks. Acta neuropathologica communications. 2014;2:88. doi: 10.1186/s40478-014-0088-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Ulusoy A, Musgrove RE, Rusconi R, Klinkenberg M, Helwig M, Schneider A, Di Monte DA. Neuron-to-neuron alpha-synuclein propagation in vivo is independent of neuronal injury. Acta neuropathologica communications. 2015;3:13. doi: 10.1186/s40478-015-0198-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Brettschneider J, Del Tredici K, Lee VM, Trojanowski JQ. Spreading of pathology in neurodegenerative diseases: a focus on human studies. Nature reviews Neuroscience. 2015;16:109–120. doi: 10.1038/nrn3887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Freundt EC, Maynard N, Clancy EK, Roy S, Bousset L, Sourigues Y, Covert M, Melki R, Kirkegaard K, Brahic M. Neuron-to-neuron transmission of alpha-synuclein fibrils through axonal transport. Ann Neurol. 2012;72:517–524. doi: 10.1002/ana.23747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Reyes JF, Olsson TT, Lamberts JT, Devine MJ, Kunath T, Brundin P. A cell culture model for monitoring alpha-synuclein cell-to-cell transfer. Neurobiology of disease. 2015;77:266–275. doi: 10.1016/j.nbd.2014.07.003. [DOI] [PubMed] [Google Scholar]
- 87.Bousset L, Pieri L, Ruiz-Arlandis G, Gath J, Jensen PH, Habenstein B, Madiona K, Olieric V, Bockmann A, Meier BH, Melki R. Structural and functional characterization of two alpha-synuclein strains. Nature communications. 2013;4:2575. doi: 10.1038/ncomms3575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Guo JL, Covell DJ, Daniels JP, Iba M, Stieber A, Zhang B, Riddle DM, Kwong LK, Xu Y, Trojanowski JQ, Lee VM. Distinct alpha-synuclein strains differentially promote tau inclusions in neurons. Cell. 2013;154:103–117. doi: 10.1016/j.cell.2013.05.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Dijkstra AA, Voorn P, Berendse HW, Groenewegen HJ, Rozemuller AJ, van de Berg WD. Stage-dependent nigral neuronal loss in incidental Lewy body and Parkinson’s disease. Movement disorders : official journal of the Movement Disorder Society. 2014;29:1244–1251. doi: 10.1002/mds.25952. [DOI] [PubMed] [Google Scholar]
- 90.Mori F, Nishie M, Kakita A, Yoshimoto M, Takahashi H, Wakabayashi K. Relationship among alpha-synuclein accumulation, dopamine synthesis, and neurodegeneration in Parkinson disease substantia nigra. Journal of neuropathology and experimental neurology. 2006;65:808–815. doi: 10.1097/01.jnen.0000230520.47768.1a. [DOI] [PubMed] [Google Scholar]
- 91.Huang C, Tang C, Feigin A, Lesser M, Ma Y, Pourfar M, Dhawan V, Eidelberg D. Changes in network activity with the progression of Parkinson’s disease. Brain : a journal of neurology. 2007;130:1834–1846. doi: 10.1093/brain/awm086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Greffard S, Verny M, Bonnet AM, Beinis JY, Gallinari C, Meaume S, Piette F, Hauw JJ, Duyckaerts C. Motor score of the Unified Parkinson Disease Rating Scale as a good predictor of Lewy body-associated neuronal loss in the substantia nigra. Archives of neurology. 2006;63:584–588. doi: 10.1001/archneur.63.4.584. [DOI] [PubMed] [Google Scholar]
- 93.Mahul-Mellier AL, Vercruysse F, Maco B, Ait-Bouziad N, De Roo M, Muller D, Lashuel HA. Fibril growth and seeding capacity play key roles in alpha-synuclein-mediated apoptotic cell death. Cell death and differentiation. 2015 doi: 10.1038/cdd.2015.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Osterberg VR, Spinelli KJ, Weston LJ, Luk KC, Woltjer RL, Unni VK. Progressive aggregation of alpha-synuclein and selective degeneration of lewy inclusion-bearing neurons in a mouse model of parkinsonism. Cell reports. 2015;10:1252–1260. doi: 10.1016/j.celrep.2015.01.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Volpicelli-Daley LA, Gamble KL, Schultheiss CE, Riddle DM, West AB, Lee VM. Formation of alpha-synuclein Lewy neurite-like aggregates in axons impedes the transport of distinct endosomes. Molecular biology of the cell. 2014;25:4010–4023. doi: 10.1091/mbc.E14-02-0741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Bendor JT, Logan TP, Edwards RH. The function of alpha-synuclein. Neuron. 2013;79:1044–1066. doi: 10.1016/j.neuron.2013.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Davidson WS, Jonas A, Clayton DF, George JM. Stabilization of alpha-synuclein secondary structure upon binding to synthetic membranes. The Journal of biological chemistry. 1998;273:9443–9449. doi: 10.1074/jbc.273.16.9443. [DOI] [PubMed] [Google Scholar]
- 98.Oueslati A, Fournier M, Lashuel HA. Role of post-translational modifications in modulating the structure, function and toxicity of alpha-synuclein: implications for Parkinson’s disease pathogenesis and therapies. Progress in brain research. 2010;183:115–145. doi: 10.1016/S0079-6123(10)83007-9. [DOI] [PubMed] [Google Scholar]
- 99.Bertoncini CW, Jung YS, Fernandez CO, Hoyer W, Griesinger C, Jovin TM, Zweckstetter M. Release of long-range tertiary interactions potentiates aggregation of natively unstructured alpha-synuclein. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:1430–1435. doi: 10.1073/pnas.0407146102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Waxman EA, Mazzulli JR, Giasson BI. Characterization of hydrophobic residue requirements for alpha-synuclein fibrillization. Biochemistry. 2009;48:9427–9436. doi: 10.1021/bi900539p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Iwai A, Masliah E, Yoshimoto M, Ge N, Flanagan L, de Silva HA, 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. doi: 10.1016/0896-6273(95)90302-x. [DOI] [PubMed] [Google Scholar]
- 102.Maroteaux L, Campanelli JT, Scheller RH. Synuclein: a neuron-specific protein localized to the nucleus and presynaptic nerve terminal. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1988;8:2804–2815. doi: 10.1523/JNEUROSCI.08-08-02804.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ben Gedalya T, Loeb V, Israeli E, Altschuler Y, Selkoe DJ, Sharon R. Alpha-synuclein and polyunsaturated fatty acids promote clathrin-mediated endocytosis and synaptic vesicle recycling. Traffic. 2009;10:218–234. doi: 10.1111/j.1600-0854.2008.00853.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Chandra S, Gallardo G, Fernandez-Chacon R, Schluter OM, Sudhof TC. Alpha-synuclein cooperates with CSPalpha in preventing neurodegeneration. Cell. 2005;123:383–396. doi: 10.1016/j.cell.2005.09.028. [DOI] [PubMed] [Google Scholar]
- 105.Burre J, Sharma M, Tsetsenis T, Buchman V, Etherton MR, Sudhof TC. Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science. 2010;329:1663–1667. doi: 10.1126/science.1195227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK, Chaudhry FA, Nicoll RA, Edwards RH. Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron. 2010;65:66–79. doi: 10.1016/j.neuron.2009.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Abeliovich A, Schmitz Y, Farinas I, Choi-Lundberg D, Ho WH, Castillo PE, Shinsky N, Verdugo JM, Armanini M, Ryan A, Hynes M, Phillips H, Sulzer D, Rosenthal A. Mice lacking alpha-synuclein display functional deficits in the nigrostriatal dopamine system. Neuron. 2000;25:239–252. doi: 10.1016/s0896-6273(00)80886-7. [DOI] [PubMed] [Google Scholar]
- 108.Yavich L, Tanila H, Vepsalainen S, Jakala P. Role of alpha-synuclein in presynaptic dopamine recruitment. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2004;24:11165–11170. doi: 10.1523/JNEUROSCI.2559-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Yavich L, Jakala P, Tanila H. Abnormal compartmentalization of norepinephrine in mouse dentate gyrus in alpha-synuclein knockout and A30P transgenic mice. Journal of neurochemistry. 2006;99:724–732. doi: 10.1111/j.1471-4159.2006.04098.x. [DOI] [PubMed] [Google Scholar]
- 110.Scott D, Roy S. alpha-Synuclein inhibits intersynaptic vesicle mobility and maintains recycling-pool homeostasis. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2012;32:10129–10135. doi: 10.1523/JNEUROSCI.0535-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Greten-Harrison B, Polydoro M, Morimoto-Tomita M, Diao L, Williams AM, Nie EH, Makani S, Tian N, Castillo PE, Buchman VL, Chandra SS. alphabetagamma-Synuclein triple knockout mice reveal age-dependent neuronal dysfunction. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:19573–19578. doi: 10.1073/pnas.1005005107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Vargas KJ, Makani S, Davis T, Westphal CH, Castillo PE, Chandra SS. Synucleins regulate the kinetics of synaptic vesicle endocytosis. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2014;34:9364–9376. doi: 10.1523/JNEUROSCI.4787-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Gorbatyuk OS, Li S, Nash K, Gorbatyuk M, Lewin AS, Sullivan LF, Mandel RJ, Chen W, Meyers C, Manfredsson FP, Muzyczka N. In vivo RNAi-mediated alpha-synuclein silencing induces nigrostriatal degeneration. Molecular therapy : the journal of the American Society of Gene Therapy. 2010;18:1450–1457. doi: 10.1038/mt.2010.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Guardia-Laguarta C, Area-Gomez E, Rub C, Liu Y, Magrane J, Becker D, Voos W, Schon EA, Przedborski S. alpha-Synuclein is localized to mitochondria-associated ER membranes. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2014;34:249–259. doi: 10.1523/JNEUROSCI.2507-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Nakamura K, Nemani VM, Azarbal F, Skibinski G, Levy JM, Egami K, Munishkina L, Zhang J, Gardner B, Wakabayashi J, Sesaki H, Cheng Y, Finkbeiner S, Nussbaum RL, Masliah E, Edwards RH. Direct membrane association drives mitochondrial fission by the Parkinson disease-associated protein alpha-synuclein. The Journal of biological chemistry. 2011;286:20710–20726. doi: 10.1074/jbc.M110.213538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Goers J, Manning-Bog AB, McCormack AL, Millett IS, Doniach S, Di Monte DA, Uversky VN, Fink AL. Nuclear localization of alpha-synuclein and its interaction with histones. Biochemistry. 2003;42:8465–8471. doi: 10.1021/bi0341152. [DOI] [PubMed] [Google Scholar]
- 117.Kontopoulos E, Parvin JD, Feany MB. Alpha-synuclein acts in the nucleus to inhibit histone acetylation and promote neurotoxicity. Human molecular genetics. 2006;15:3012–3023. doi: 10.1093/hmg/ddl243. [DOI] [PubMed] [Google Scholar]
- 118.Specht CG, Tigaret CM, Rast GF, Thalhammer A, Rudhard Y, Schoepfer R. Subcellular localisation of recombinant alpha- and gamma-synuclein. Molecular and cellular neurosciences. 2005;28:326–334. doi: 10.1016/j.mcn.2004.09.017. [DOI] [PubMed] [Google Scholar]
- 119.Li WW, Yang R, Guo JC, Ren HM, Zha XL, Cheng JS, Cai DF. Localization of alpha-synuclein to mitochondria within midbrain of mice. Neuroreport. 2007;18:1543–1546. doi: 10.1097/WNR.0b013e3282f03db4. [DOI] [PubMed] [Google Scholar]
- 120.Ellis CE, Murphy EJ, Mitchell DC, Golovko MY, Scaglia F, Barcelo-Coblijn GC, Nussbaum RL. Mitochondrial lipid abnormality and electron transport chain impairment in mice lacking alpha-synuclein. Molecular and cellular biology. 2005;25:10190–10201. doi: 10.1128/MCB.25.22.10190-10201.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Martin LJ, Pan Y, Price AC, Sterling W, Copeland NG, Jenkins NA, Price DL, Lee MK. Parkinson’s disease alpha-synuclein transgenic mice develop neuronal mitochondrial degeneration and cell death. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2006;26:41–50. doi: 10.1523/JNEUROSCI.4308-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Narendra D, Walker JE, Youle R. Mitochondrial quality control mediated by PINK1 and Parkin: links to parkinsonism. Cold Spring Harbor perspectives in biology. 2012:4. doi: 10.1101/cshperspect.a011338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Conway KA, Rochet JC, Bieganski RM, Lansbury PT., Jr Kinetic stabilization of the alpha-synuclein protofibril by a dopamine-alpha-synuclein adduct. Science. 2001;294:1346–1349. doi: 10.1126/science.1063522. [DOI] [PubMed] [Google Scholar]
- 124.Norris EH, Giasson BI, Hodara R, Xu S, Trojanowski JQ, Ischiropoulos H, Lee VM. Reversible inhibition of alpha-synuclein fibrillization by dopaminochrome-mediated conformational alterations. The Journal of biological chemistry. 2005;280:21212–21219. doi: 10.1074/jbc.M412621200. [DOI] [PubMed] [Google Scholar]
- 125.Chen SW, Drakulic S, Deas E, Ouberai M, Aprile FA, Arranz R, Ness S, Roodveldt C, Guilliams T, De-Genst EJ, Klenerman D, Wood NW, Knowles TP, Alfonso C, Rivas G, Abramov AY, Valpuesta JM, Dobson CM, Cremades N. Structural characterization of toxic oligomers that are kinetically trapped during alpha-synuclein fibril formation. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:E1994–E2003. doi: 10.1073/pnas.1421204112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lorenzen N, Nielsen SB, Buell AK, Kaspersen JD, Arosio P, Vad BS, Paslawski W, Christiansen G, Valnickova-Hansen Z, Andreasen M, Enghild JJ, Pedersen JS, Dobson CM, Knowles TP, Otzen DE. The role of stable alpha-synuclein oligomers in the molecular events underlying amyloid formation. Journal of the American Chemical Society. 2014;136:3859–3868. doi: 10.1021/ja411577t. [DOI] [PubMed] [Google Scholar]
- 127.Cremades N, Cohen SI, Deas E, Abramov AY, Chen AY, Orte A, Sandal M, Clarke RW, Dunne P, Aprile FA, Bertoncini CW, Wood NW, Knowles TP, Dobson CM, Klenerman D. Direct observation of the interconversion of normal and toxic forms of alpha-synuclein. Cell. 2012;149:1048–1059. doi: 10.1016/j.cell.2012.03.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Butterfield SM, Lashuel HA. Amyloidogenic protein-membrane interactions: mechanistic insight from model systems. Angew Chem Int Ed Engl. 2010;49:5628–5654. doi: 10.1002/anie.200906670. [DOI] [PubMed] [Google Scholar]
- 129.Winner B, Jappelli R, Maji SK, Desplats PA, Boyer L, Aigner S, Hetzer C, Loher T, Vilar M, Campioni S, Tzitzilonis C, Soragni A, Jessberger S, Mira H, Consiglio A, Pham E, Masliah E, Gage FH, Riek R. In vivo demonstration that alpha-synuclein oligomers are toxic. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:4194–4199. doi: 10.1073/pnas.1100976108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Conway KA, Lee SJ, Rochet JC, Ding TT, Williamson RE, Lansbury PT., Jr Acceleration of oligomerization, not fibrillization, is a shared property of both alpha-synuclein mutations linked to early-onset Parkinson’s disease: implications for pathogenesis and therapy. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:571–576. doi: 10.1073/pnas.97.2.571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Campioni S, Mannini B, Zampagni M, Pensalfini A, Parrini C, Evangelisti E, Relini A, Stefani M, Dobson CM, Cecchi C, Chiti F. A causative link between the structure of aberrant protein oligomers and their toxicity. Nature chemical biology. 2010;6:140–147. doi: 10.1038/nchembio.283. [DOI] [PubMed] [Google Scholar]
- 132.van Rooijen BD, Claessens MM, Subramaniam V. Lipid bilayer disruption by oligomeric alpha-synuclein depends on bilayer charge and accessibility of the hydrophobic core. Biochimica et biophysica acta. 2009;1788:1271–1278. doi: 10.1016/j.bbamem.2009.03.010. [DOI] [PubMed] [Google Scholar]
- 133.Roberts HL, Brown DR. Seeking a Mechanism for the Toxicity of Oligomeric alpha-Synuclein. Biomolecules. 2015;5:282–305. doi: 10.3390/biom5020282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Taguchi K, Watanabe Y, Tsujimura A, Tatebe H, Miyata S, Tokuda T, Mizuno T, Tanaka M. Differential expression of alpha-synuclein in hippocampal neurons. PloS one. 2014;9:e89327. doi: 10.1371/journal.pone.0089327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Fortin DL, Troyer MD, Nakamura K, Kubo S, Anthony MD, Edwards RH. Lipid rafts mediate the synaptic localization of alpha-synuclein. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2004;24:6715–6723. doi: 10.1523/JNEUROSCI.1594-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Kahle PJ, Neumann M, Ozmen L, Muller V, Jacobsen H, Schindzielorz A, Okochi M, Leimer U, van Der Putten H, Probst A, Kremmer E, Kretzschmar HA, Haass C. Subcellular localization of wild-type and Parkinson’s disease-associated mutant alpha -synuclein in human and transgenic mouse brain. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2000;20:6365–6373. doi: 10.1523/JNEUROSCI.20-17-06365.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Fauvet B, Mbefo MK, Fares MB, Desobry C, Michael S, Ardah MT, Tsika E, Coune P, Prudent M, Lion N, Eliezer D, Moore DJ, Schneider B, Aebischer P, El-Agnaf OM, Masliah E, Lashuel HA. alpha-Synuclein in central nervous system and from erythrocytes, mammalian cells, and Escherichia coli exists predominantly as disordered monomer. The Journal of biological chemistry. 2012;287:15345–15364. doi: 10.1074/jbc.M111.318949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Bartels T, Choi JG, Selkoe DJ. alpha-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation. Nature. 2011;477:107–110. doi: 10.1038/nature10324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Wang W, Perovic I, Chittuluru J, Kaganovich A, Nguyen LT, Liao J, Auclair JR, Johnson D, Landeru A, Simorellis AK, Ju S, Cookson MR, Asturias FJ, Agar JN, Webb BN, Kang C, Ringe D, Petsko GA, Pochapsky TC, Hoang QQ. A soluble alpha-synuclein construct forms a dynamic tetramer. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:17797–17802. doi: 10.1073/pnas.1113260108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Dettmer U, Newman AJ, Soldner F, Luth ES, Kim NC, von Saucken VE, Sanderson JB, Jaenisch R, Bartels T, Selkoe D. Parkinson-causing alpha-synuclein missense mutations shift native tetramers to monomers as a mechanism for disease initiation. Nature communications. 2015;6:7314. doi: 10.1038/ncomms8314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Zhu M, Fink AL. Lipid binding inhibits alpha-synuclein fibril formation. The Journal of biological chemistry. 2003;278:16873–16877. doi: 10.1074/jbc.M210136200. [DOI] [PubMed] [Google Scholar]
- 142.Jensen MB, Bhatia VK, Jao CC, Rasmussen JE, Pedersen SL, Jensen KJ, Langen R, Stamou D. Membrane curvature sensing by amphipathic helices: a single liposome study using alpha-synuclein and annexin B12. The Journal of biological chemistry. 2011;286:42603–42614. doi: 10.1074/jbc.M111.271130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Middleton ER, Rhoades E. Effects of curvature and composition on alpha-synuclein binding to lipid vesicles. Biophysical journal. 2010;99:2279–2288. doi: 10.1016/j.bpj.2010.07.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Westphal CH, Chandra SS. Monomeric synucleins generate membrane curvature. The Journal of biological chemistry. 2013;288:1829–1840. doi: 10.1074/jbc.M112.418871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Burre J, Sharma M, Sudhof TC. alpha-Synuclein assembles into higher-order multimers upon membrane binding to promote SNARE complex formation. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:E4274–E4283. doi: 10.1073/pnas.1416598111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Conway KA, Lee SJ, Rochet JC, Ding TT, Harper JD, Williamson RE, Lansbury PT., Jr Accelerated oligomerization by Parkinson’s disease linked alpha-synuclein mutants. Annals of the New York Academy of Sciences. 2000;920:42–45. doi: 10.1111/j.1749-6632.2000.tb06903.x. [DOI] [PubMed] [Google Scholar]
- 147.Greenbaum EA, Graves CL, Mishizen-Eberz AJ, Lupoli MA, Lynch DR, Englander SW, Axelsen PH, Giasson BI. The E46K mutation in alpha-synuclein increases amyloid fibril formation. The Journal of biological chemistry. 2005;280:7800–7807. doi: 10.1074/jbc.M411638200. [DOI] [PubMed] [Google Scholar]
- 148.Khalaf O, Fauvet B, Oueslati A, Dikiy I, Mahul-Mellier AL, Ruggeri FS, Mbefo MK, Vercruysse F, Dietler G, Lee SJ, Eliezer D, Lashuel HA. The H50Q mutation enhances alpha-synuclein aggregation, secretion, and toxicity. The Journal of biological chemistry. 2014;289:21856–21876. doi: 10.1074/jbc.M114.553297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Rutherford NJ, Moore BD, Golde TE, Giasson BI. Divergent effects of the H50Q and G51D SNCA mutations on the aggregation of alpha-synuclein. Journal of neurochemistry. 2014;131:859–867. doi: 10.1111/jnc.12806. [DOI] [PubMed] [Google Scholar]
- 150.Burre J, Sharma M, Sudhof TC. Definition of a molecular pathway mediating alpha-synuclein neurotoxicity. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2015;35:5221–5232. doi: 10.1523/JNEUROSCI.4650-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Dauer W, Kholodilov N, Vila M, Trillat AC, Goodchild R, Larsen KE, Staal R, Tieu K, Schmitz Y, Yuan CA, Rocha M, Jackson-Lewis V, Hersch S, Sulzer D, Przedborski S, Burke R, Hen R. Resistance of alpha -synuclein null mice to the parkinsonian neurotoxin MPTP. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:14524–14529. doi: 10.1073/pnas.172514599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Colla E, Coune P, Liu Y, Pletnikova O, Troncoso JC, Iwatsubo T, Schneider BL, Lee MK. Endoplasmic reticulum stress is important for the manifestations of alpha-synucleinopathy in vivo. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2012;32:3306–3320. doi: 10.1523/JNEUROSCI.5367-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Mercado G, Valdes P, Hetz C. An ERcentric view of Parkinson’s disease. Trends in molecular medicine. 2013;19:165–175. doi: 10.1016/j.molmed.2012.12.005. [DOI] [PubMed] [Google Scholar]
- 154.Silva RM, Ries V, Oo TF, Yarygina O, Jackson-Lewis V, Ryu EJ, Lu PD, Marciniak SJ, Ron D, Przedborski S, Kholodilov N, Greene LA, Burke RE. CHOP/GADD153 is a mediator of apoptotic death in substantia nigra dopamine neurons in an in vivo neurotoxin model of parkinsonism. Journal of neurochemistry. 2005;95:974–986. doi: 10.1111/j.1471-4159.2005.03428.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Aime P, Sun X, Zareen N, Rao A, Berman Z, Volpicelli-Daley L, Bernd P, Crary JF, Levy OA, Greene LA. Trib3 Is Elevated in Parkinson’s Disease and Mediates Death in Parkinson’s Disease Models. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2015;35:10731–10749. doi: 10.1523/JNEUROSCI.0614-15.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Hoozemans JJ, van Haastert ES, Eikelenboom P, de Vos RA, Rozemuller JM, Scheper W. Activation of the unfolded protein response in Parkinson’s disease. Biochemical and biophysical research communications. 2007;354:707–711. doi: 10.1016/j.bbrc.2007.01.043. [DOI] [PubMed] [Google Scholar]
- 157.Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, Stoessl AJ, Pfeiffer RF, Patenge N, Carbajal IC, Vieregge P, Asmus F, Muller-Myhsok B, Dickson DW, Meitinger T, Strom TM, Wszolek ZK, Gasser T. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44:601–607. doi: 10.1016/j.neuron.2004.11.005. [DOI] [PubMed] [Google Scholar]
- 158.Aharon-Peretz J, Rosenbaum H, Gershoni-Baruch R. Mutations in the glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. The New England journal of medicine. 2004;351:1972–1977. doi: 10.1056/NEJMoa033277. [DOI] [PubMed] [Google Scholar]
- 159.Zimprich A, Benet-Pages A, Struhal W, Graf E, Eck SH, Offman MN, Haubenberger D, Spielberger S, Schulte EC, Lichtner P, Rossle SC, Klopp N, Wolf E, Seppi K, Pirker W, Presslauer S, Mollenhauer B, Katzenschlager R, Foki T, Hotzy C, Reinthaler E, Harutyunyan A, Kralovics R, Peters A, Zimprich F, Brucke T, Poewe W, Auff E, Trenkwalder C, Rost B, Ransmayr G, Winkelmann J, Meitinger T, Strom TM. A mutation in VPS35, encoding a subunit of the retromer complex, causes late-onset Parkinson disease. American journal of human genetics. 2011;89:168–175. doi: 10.1016/j.ajhg.2011.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ, Bhullar B, Liu K, Xu K, Strathearn KE, Liu F, Cao S, Caldwell KA, Caldwell GA, Marsischky G, Kolodner RD, Labaer J, Rochet JC, Bonini NM, Lindquist S. Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson’s models. Science. 2006;313:324–328. doi: 10.1126/science.1129462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Chung CY, Khurana V, Auluck PK, Tardiff DF, Mazzulli JR, Soldner F, Baru V, Lou Y, Freyzon Y, Cho S, Mungenast AE, Muffat J, Mitalipova M, Pluth MD, Jui NT, Schule B, Lippard SJ, Tsai LH, Krainc D, Buchwald SL, Jaenisch R, Lindquist S. Identification and rescue of alpha-synuclein toxicity in Parkinson patient-derived neurons. Science. 2013;342:983–987. doi: 10.1126/science.1245296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Tardiff DF, Jui NT, Khurana V, Tambe MA, Thompson ML, Chung CY, Kamadurai HB, Kim HT, Lancaster AK, Caldwell KA, Caldwell GA, Rochet JC, Buchwald SL, Lindquist S. Yeast reveal a “druggable” Rsp5/Nedd4 network that ameliorates alpha-synuclein toxicity in neurons. Science. 2013;342:979–983. doi: 10.1126/science.1245321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Bellinger FP, Bellinger MT, Seale LA, Takemoto AS, Raman AV, Miki T, Manning-Bog AB, Berry MJ, White LR, Ross GW. Glutathione Peroxidase 4 is associated with Neuromelanin in Substantia Nigra and Dystrophic Axons in Putamen of Parkinson’s brain. Molecular neurodegeneration. 2011;6:8. doi: 10.1186/1750-1326-6-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. The Journal of cell biology. 2008;183:795–803. doi: 10.1083/jcb.200809125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998;392:605–608. doi: 10.1038/33416. [DOI] [PubMed] [Google Scholar]
- 166.Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio AR, Healy DG, Albanese A, Nussbaum R, Gonzalez-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapiccola B, Auburger G, Wood NW. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science. 2004;304:1158–1160. doi: 10.1126/science.1096284. [DOI] [PubMed] [Google Scholar]
- 167.Poole AC, Thomas RE, Andrews LA, McBride HM, Whitworth AJ, Pallanck LJ. The PINK1/Parkin pathway regulates mitochondrial morphology. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:1638–1643. doi: 10.1073/pnas.0709336105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Kim RH, Smith PD, Aleyasin H, Hayley S, Mount MP, Pownall S, Wakeham A, You-Ten AJ, Kalia SK, Horne P, Westaway D, Lozano AM, Anisman H, Park DS, Mak TW. Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:5215–5220. doi: 10.1073/pnas.0501282102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Dauer W, Kholodilov N, Vila M, Trillat AC, Goodchild R, Larsen KE, Staal R, Tieu K, Schmitz Y, Yuan CA, Rocha M, Jackson-Lewis V, Hersch S, Sulzer D, Przedborski S, Burke R, Hen R. Resistance of alpha -synuclein null mice to the parkinsonian neurotoxin MPTP. ProcNatlAcadSciUS A. 2002;99:14524–14529. doi: 10.1073/pnas.172514599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Dryanovski DI, Guzman JN, Xie Z, Galteri DJ, Volpicelli-Daley LA, Lee VM, Miller RJ, Schumacker PT, Surmeier DJ. Calcium entry and alpha-synuclein inclusions elevate dendritic mitochondrial oxidant stress in dopaminergic neurons. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2013;33:10154–10164. doi: 10.1523/JNEUROSCI.5311-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Surmeier DJ, Guzman JN, Sanchez-Padilla J, Goldberg JA. The origins of oxidant stress in Parkinson’s disease and therapeutic strategies. Antioxid Redox Signal. 2011;14:1289–1301. doi: 10.1089/ars.2010.3521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Foehring RC, Zhang XF, Lee JC, Callaway JC. Endogenous calcium buffering capacity of substantia nigral dopamine neurons. Journal of neurophysiology. 2009;102:2326–2333. doi: 10.1152/jn.00038.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Mosharov E, Larsen K, Kanter E, Phillips K, Wilson K, Schmitz Y, Krantz D, Kobayashi K, Edwards R, Sulzer D. Interplay between cytosolic dopamine, calcium, and alpha-synuclein causes selective death of substantia nigra neurons. Neuron. 2009;62:218–229. doi: 10.1016/j.neuron.2009.01.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Chan CS, Guzman JN, Ilijic E, Mercer JN, Rick C, Tkatch T, Meredith GE, Surmeier DJ. ‘Rejuvenation’ protects neurons in mouse models of Parkinson’s disease. Nature. 2007;447:1081–1086. doi: 10.1038/nature05865. [DOI] [PubMed] [Google Scholar]
- 175.Kupsch A, Sautter J, Schwarz J, Riederer P, Gerlach M, Oertel WH. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity in non-human primates is antagonized by pretreatment with nimodipine at the nigral, but not at the striatal level. Brain research. 1996;741:185–196. doi: 10.1016/s0006-8993(96)00917-1. [DOI] [PubMed] [Google Scholar]
- 176.Kupsch A, Gerlach M, Pupeter SC, Sautter J, Dirr A, Arnold G, Opitz W, Przuntek H, Riederer P, Oertel WH. Pretreatment with nimodipine prevents MPTP-induced neurotoxicity at the nigral, but not at the striatal level in mice. Neuroreport. 1995;6:621–625. doi: 10.1097/00001756-199503000-00009. [DOI] [PubMed] [Google Scholar]
- 177.Mosharov EV, Larsen KE, Kanter E, Phillips KA, Wilson K, Schmitz Y, Krantz DE, Kobayashi K, Edwards RH, Sulzer D. Interplay between cytosolic dopamine, calcium, and alpha-synuclein causes selective death of substantia nigra neurons. Neuron. 2009;62:218–229. doi: 10.1016/j.neuron.2009.01.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Schondorf DC, Aureli M, McAllister FE, Hindley CJ, Mayer F, Schmid B, Sardi SP, Valsecchi M, Hoffmann S, Schwarz LK, Hedrich U, Berg D, Shihabuddin LS, Hu J, Pruszak J, Gygi SP, Sonnino S, Gasser T, Deleidi M. iPSC-derived neurons from GBA1-associated Parkinson’s disease patients show autophagic defects and impaired calcium homeostasis. Nature communications. 2014;5:4028. doi: 10.1038/ncomms5028. [DOI] [PubMed] [Google Scholar]
- 179.Guzman JN, Sanchez-Padilla J, Wokosin D, Kondapalli J, Ilijic E, Schumacker PT, Surmeier DJ. Oxidant stress evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. Nature. 2010;468:696–700. doi: 10.1038/nature09536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38:1285–1291. doi: 10.1212/wnl.38.8.1285. [DOI] [PubMed] [Google Scholar]
- 181.Sanchez-Guajardo V, Tentillier N, Romero-Ramos M. The relation between alpha-synuclein and microglia in Parkinson’s disease: Recent developments. Neuroscience. 2015 doi: 10.1016/j.neuroscience.2015.02.008. [DOI] [PubMed] [Google Scholar]
- 182.Dobbs RJ, Charlett A, Purkiss AG, Dobbs SM, Weller C, Peterson DW. Association of circulating TNF-alpha and IL-6 with ageing and parkinsonism. Acta neurologica Scandinavica. 1999;100:34–41. doi: 10.1111/j.1600-0404.1999.tb00721.x. [DOI] [PubMed] [Google Scholar]
- 183.Mogi M, Harada M, Kondo T, Riederer P, Inagaki H, Minami M, Nagatsu T. Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients. Neuroscience letters. 1994;180:147–150. doi: 10.1016/0304-3940(94)90508-8. [DOI] [PubMed] [Google Scholar]
- 184.Sulzer D, Surmeier DJ. Neuronal vulnerability, pathogenesis, and Parkinson’s disease. Movement disorders : official journal of the Movement Disorder Society. 2013;28:41–50. doi: 10.1002/mds.25095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Matsuda W, Furuta T, Nakamura KC, Hioki H, Fujiyama F, Arai R, Kaneko T. Single nigrostriatal dopaminergic neurons form widely spread and highly dense axonal arborizations in the neostriatum. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2009;29:444–453. doi: 10.1523/JNEUROSCI.4029-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Bolam JP, Pissadaki EK. Living on the edge with too many mouths to feed: why dopamine neurons die. Movement disorders : official journal of the Movement Disorder Society. 2012;27:1478–1483. doi: 10.1002/mds.25135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Kramer ML, Behrens C, Schulz-Schaeffer WJ. Selective detection, quantification, and subcellular location of alpha-synuclein aggregates with a protein aggregate filtration assay. BioTechniques. 2008;44:403–411. doi: 10.2144/000112691. [DOI] [PubMed] [Google Scholar]
- 188.Chu Y, Kordower JH. Age-associated increases of alpha-synuclein in monkeys and humans are associated with nigrostriatal dopamine depletion: Is this the target for Parkinson’s disease? Neurobiology of disease. 2007;25:134–149. doi: 10.1016/j.nbd.2006.08.021. [DOI] [PubMed] [Google Scholar]
- 189.Chen L, Ding Y, Cagniard B, Van Laar AD, Mortimer A, Chi W, Hastings TG, Kang UJ, Zhuang X. Unregulated cytosolic dopamine causes neurodegeneration associated with oxidative stress in mice. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2008;28:425–433. doi: 10.1523/JNEUROSCI.3602-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Sulzer D, Mosharov E, Talloczy Z, Zucca FA, Simon JD, Zecca L. Neuronal pigmented autophagic vacuoles: lipofuscin, neuromelanin, and ceroid as macroautophagic responses during aging and disease. Journal of neurochemistry. 2008;106:24–36. doi: 10.1111/j.1471-4159.2008.05385.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Sulzer D, Bogulavsky J, Larsen KE, Behr G, Karatekin E, Kleinman MH, Turro N, Krantz D, Edwards RH, Greene LA, Zecca L. Neuromelanin biosynthesis is driven by excess cytosolic catecholamines not accumulated by synaptic vesicles. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:11869–11874. doi: 10.1073/pnas.97.22.11869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Fedorow H, Tribl F, Halliday G, Gerlach M, Riederer P, Double KL. Neuromelanin in human dopamine neurons: comparison with peripheral melanins and relevance to Parkinson’s disease. Progress in neurobiology. 2005;75:109–124. doi: 10.1016/j.pneurobio.2005.02.001. [DOI] [PubMed] [Google Scholar]
- 193.Cebrian C, Zucca FA, Mauri P, Steinbeck JA, Studer L, Scherzer CR, Kanter E, Budhu S, Mandelbaum J, Vonsattel JP, Zecca L, Loike JD, Sulzer D. MHC-I expression renders catecholaminergic neurons susceptible to T-cell-mediated degeneration. Nature communications. 2014;5:3633. doi: 10.1038/ncomms4633. [DOI] [PMC free article] [PubMed] [Google Scholar]